1
|
Kahraman E, Vasconcelos D, Ribeiro B, Monteiro AC, Mastromatteo E, Bortolin A, Couto M, Boschis L, Lamghari M, Neto E. Deciphering cartilage neuro-immune interactions and innervation profile through 3D engineered osteoarthritic micropathophysiological system. Mater Today Bio 2025; 31:101491. [PMID: 39896288 PMCID: PMC11786692 DOI: 10.1016/j.mtbio.2025.101491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/15/2024] [Accepted: 01/12/2025] [Indexed: 02/04/2025] Open
Abstract
Osteoarthritis (OA) is an inflammatory musculoskeletal disorder that results in cartilage breakdown and alterations in the surrounding tissue microenvironment. Imbalances caused by inflammation and catabolic processes potentiate pathological nerves and blood vessels outgrowth toward damaged areas leading to pain in the patients. Yet, the precise mechanisms leading the nerve sprouting into the aneural cartilaginous tissue remain elusive. In this work, we aim to recapitulate in vitro the hallmarks of OA pathophysiology, including the sensory innervation profile, and provide a sensitive and reliable analytical tool to monitor the in vitro disease progression at microscale. Leveraging the use of patient-derived cells and bioengineering cutting-edge technologies, we engineered cartilage-like microtissues composed of primary human chondrocytes encapsulated in gelatin methacrylate hydrogel. Engineered constructs patterned inside microfluidic devices show the expression of cartilage markers, namely collagen type II, aggrecan, SOX-9 and glycosaminoglycans. Upon pro-inflammatory triggering, using primary human pro-inflammatory macrophage secretome, hallmarks of OA are recapitulated namely catabolic processes of human chondrocytes and the sensory innervation profile, supported by gene expression and functional assays. To monitor the OA micropathological system, a highly sensitive technology - EliChip™ - is presented to quantitively assess the molecular signature of cytokines and growth factors (interleukin 6 and nerve growth factor) produced from a single microfluidic chip. Herein, we report a miniaturized pathophysiological model and analytical tool to foster the neuro-immune interactions playing a role in cartilage-related disorders.
Collapse
Affiliation(s)
- Emine Kahraman
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Daniela Vasconcelos
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Beatriz Ribeiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Ana Carolina Monteiro
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Enzo Mastromatteo
- Trustech Innovation Technology, Via Baraggino, 76, 10034, Chivasso, Torino, Italy
| | - Andrea Bortolin
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
| | - Marina Couto
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Rua Jorge de Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Laura Boschis
- Trustech Innovation Technology, Via Baraggino, 76, 10034, Chivasso, Torino, Italy
| | - Meriem Lamghari
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| | - Estrela Neto
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135, Porto, Portugal
| |
Collapse
|
2
|
Dong DL, Jin GZ. Targeting Chondrocyte Hypertrophy as Strategies for the Treatment of Osteoarthritis. Bioengineering (Basel) 2025; 12:77. [PMID: 39851351 PMCID: PMC11760869 DOI: 10.3390/bioengineering12010077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by pain and functional impairment, which severely impacts the quality of life of middle-aged and elderly individuals. During normal bone development, chondrocyte hypertrophy is a natural physiological process. However, in the progression of OA, chondrocyte hypertrophy becomes one of its key pathological features. Although there is no definitive evidence to date confirming that chondrocyte hypertrophy is the direct cause of OA, substantial experimental data indicate that it plays an important role in the disease's pathogenesis. In this review, we first explore the mechanisms underlying chondrocyte hypertrophy in OA and offer new insights. We then propose strategies for inhibiting chondrocyte hypertrophy from the perspectives of targeting signaling pathways and tissue engineering, ultimately envisioning the future prospects of OA treatment.
Collapse
Affiliation(s)
- Da-Long Dong
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
- Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Guang-Zhen Jin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea;
| |
Collapse
|
3
|
Palma C, Piazza S, Visone R, Ringom R, Björklund U, Bermejo Gómez A, Rasponi M, Occhetta P. An Advanced Mechanically Active Osteoarthritis-on-Chip Model to Test Injectable Therapeutic Formulations: The SYN321 Case Study. Adv Healthc Mater 2024; 13:e2401187. [PMID: 39318108 DOI: 10.1002/adhm.202401187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/10/2024] [Indexed: 09/26/2024]
Abstract
Current treatments for osteoarthritis (OA) often fail to address the underlying pathophysiology and may have systemic side effects, particularly associated with long-term use of non-steroidal anti-inflammatory drugs (NSAIDs). Thus, researchers are currently directing their efforts toward innovative polymer-drug combinations, such as mixtures of hyaluronic acid viscoelastic hydrogels and NSAIDs like diclofenac, to ensure sustained release of the NSAID within the joint following intra-articular injection. However, the progress of novel injectable therapies for OA is hindered by the absence of preclinical models that accurately represent the pathology of the disease. The uBeat® MultiCompress platform is here presented as a novel approach for studying anti-OA injectable therapeutics on human mechanically-damaged OA cartilage microtissues, in a physiologically relevant environment. This platform can accommodate injectable therapeutic formulations and is successfully tested with SYN321, a novel diclofenac-sodium hyaluronate conjugate under development as a treatment for knee OA. Results indicate the platform's effectiveness in evaluating therapeutic potential, showing downregulation of inflammatory markers and reduction in matrix degradation in OA cartilage micro-tissues treated with SYN321. The uBeat® MultiCompress platform thus represents a valuable tool for OA research, offering a bridge between traditional in vitro studies and potential clinical applications, with implications for future drug discovery.
Collapse
Affiliation(s)
- Cecilia Palma
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Stefano Piazza
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| | - Roberta Visone
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| | - Rune Ringom
- Recipharm OT Chemistry AB, Virdings allé 18, Uppsala, 754 50, Sweden
| | - Ulf Björklund
- UB-consulting AB, Trädgårdsgatan 7A, Uppsala, 753 09, Sweden
| | | | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
| | - Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Via Ponzio 34/5, Milan, 20133, Italy
- BiomimX Srl, Viale Decumano 41, MIND - Milano Innovation District, Milan, 20157, Italy
| |
Collapse
|
4
|
Rosochowicz MA, Lach MS, Richter M, Jagiełło I, Suchorska WM, Trzeciak T. The iPSC secretome is beneficial for in vitro propagation of primary osteoarthritic chondrocytes cell lines. Biochem Biophys Res Commun 2024; 730:150392. [PMID: 39003867 DOI: 10.1016/j.bbrc.2024.150392] [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: 05/08/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND One of the obstacles to autologous chondrocyte implantation (ACI) is obtaining a large quantity of chondrocytes without depletion of their properties. The conditioned medium (CM) from different subpopulations of stem cells (mesenchymal stromal cells (MSC) or induced pluripotent stem cells (iPSC)) could be a gamechanger. MSCs' potential is related to the donor's health and age, which could be omitted when, as a source, iPSCs are used. There is a lack of data regarding their use in the chondrocyte culture expansion. Thus, we wanted to verify whether iPSC-CM could be beneficial for the cell culture of primary chondrocyte cells. METHODS We added the iPSC-CMs from GPCCi001-A and ND 41658*H cells to the culture of primary chondrocyte cell lines isolated from OA patients (n = 6) for other two passages. The composition of the CM was evaluated using Luminex technology. Then, we analysed the senescence, proliferation rate and using flow cytometry: viability, distribution of cell cycle phases, production of reactive oxygen species (ROS) and double-strand breaks. The cartilage-related markers were evaluated using Western blot and immunofluorescence. Additionally, a three-dimensional cell culture was used to determine the potential to form cartilage particles. RESULTS iPSC-CM increased proliferation and diminished cell ROS production and senescence. CM influenced the cartilage-related protein expression and promoted the growth of cartilage particles. The cell exposed to CM did not lose the ECM proteins, suggesting the chondroprotective effect for prolonged culture time. CONCLUSION Our preliminary results suggest a beneficial effect on maintaining chondrocyte biology during in vitro expansion.
Collapse
Affiliation(s)
- Monika A Rosochowicz
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland; Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland.
| | - Michał S Lach
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland; Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Magdalena Richter
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| | - Inga Jagiełło
- Department of Tumour Pathology, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland
| | - Wiktoria M Suchorska
- Radiobiology Laboratory, Greater Poland Cancer Centre, Garbary 15 Street, 61-866, Poznan, Poland; Department of Electroradiology, Poznan University of Medical Sciences, Garbary 15 Street, 61-866, Poznan, Poland
| | - Tomasz Trzeciak
- Department of Orthopaedics and Traumatology, Poznan University of Medical Sciences, 28 Czerwca 1956r. 135/147 Street, 61-545, Poznan, Poland
| |
Collapse
|
5
|
Dönges L, Damle A, Mainardi A, Bock T, Schönenberger M, Martin I, Barbero A. Engineered human osteoarthritic cartilage organoids. Biomaterials 2024; 308:122549. [PMID: 38554643 DOI: 10.1016/j.biomaterials.2024.122549] [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: 10/06/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
The availability of human cell-based models capturing molecular processes of cartilage degeneration can facilitate development of disease-modifying therapies for osteoarthritis [1], a currently unmet clinical need. Here, by imposing specific inflammatory challenges upon mesenchymal stromal cells at a defined stage of chondrogenesis, we engineered a human organotypic model which recapitulates main OA pathological traits such as chondrocyte hypertrophy, cartilage matrix mineralization, enhanced catabolism and mechanical stiffening. To exemplify the utility of the model, we exposed the engineered OA cartilage organoids to factors known to attenuate pathological features, including IL-1Ra, and carried out mass spectrometry-based proteomics. We identified that IL-1Ra strongly reduced production of the transcription factor CCAAT/enhancer-binding protein beta [2] and demonstrated that inhibition of the C/EBPβ-activating kinases could revert the degradative processes. Human OA cartilage organoids thus represent a relevant tool towards the discovery of new molecular drivers of cartilage degeneration and the assessment of therapeutics targeting associated pathways.
Collapse
Affiliation(s)
- Laura Dönges
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Atharva Damle
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Andrea Mainardi
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum University of Basel, 4056, Basel, Switzerland
| | - Monica Schönenberger
- Nano Imaging Lab, Swiss Nanoscience Institute, University of Basel, 4056, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland.
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| |
Collapse
|
6
|
Gardner OFW, Zhang Y, Khan IM. BMP9 is a potent inducer of chondrogenesis, volumetric expansion and collagen type II accumulation in bovine auricular cartilage chondroprogenitors. PLoS One 2023; 18:e0294761. [PMID: 37992123 PMCID: PMC10664884 DOI: 10.1371/journal.pone.0294761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023] Open
Abstract
Reconstruction of the outer ear currently requires harvesting of cartilage from the posterior of the auricle or ribs leading to pain and donor site morbidity. An alternative source for auricular reconstruction is in vitro tissue engineered cartilage using stem/progenitor cells. Several candidate cell-types have been studied with tissue-specific auricular cartilage progenitor cells (AuCPC) of particular interest. Whilst chondrogenic differentiation of competent stem cells using growth factor TGFβ1 produces cartilage this tissue is frequently fibrocartilaginous and lacks the morphological features of hyaline cartilage. Recent work has shown that growth factor BMP9 is a potent chondrogenic and morphogenetic factor for articular cartilage progenitor cells, and we hypothesised that this property extends to cartilage-derived progenitors from other tissues. In this study we show monoclonal populations of AuCPCs from immature and mature bovine cartilage cultured with BMP9 produced cartilage pellets have 3-5-fold greater surface area in sections than those grown with TGFβ1. Increased volumetric growth using BMP9 was due to greater sGAG deposition in immature pellets and significantly greater collagen accumulation in both immature and mature progenitor pellets. Polarised light microscopy and immunohistochemical analyses revealed that the organisation of collagen fibrils within pellets is an important factor in the growth of pellets. Additionally, chondrocytes in BMP9 stimulated cell pellets had larger lacunae and were more evenly dispersed throughout the extracellular matrix. Interestingly, BMP9 tended to normalise the response of immature AuCPC monoclonal cell lines to differentiation cues whereas cells exhibited more variation under TGFβ1. In conclusion, BMP9 appears to be a potent inducer of chondrogenesis and volumetric growth for AuCPCs a property that can be exploited for tissue engineering strategies for reconstructive surgery though with the caveat of negligible elastin production following 21-day treatment with either growth factor.
Collapse
Affiliation(s)
- Oliver F. W. Gardner
- Stem Cells & Regenerative Medicine, Great Ormond Street Institute of Child Health, University College London, England, United Kingdom
| | - Yadan Zhang
- Faculty of Medicine, Health & Life Science, Swansea University Medical School, Wales, United Kingdom
| | - Ilyas M. Khan
- Faculty of Medicine, Health & Life Science, Swansea University Medical School, Wales, United Kingdom
| |
Collapse
|
7
|
Riegger J, Brenner RE. Increase of cell surface vimentin is associated with vimentin network disruption and subsequent stress-induced premature senescence in human chondrocytes. eLife 2023; 12:e91453. [PMID: 37855367 PMCID: PMC10622146 DOI: 10.7554/elife.91453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 10/04/2023] [Indexed: 10/20/2023] Open
Abstract
Accumulation of dysfunctional chondrocytes has detrimental consequences on the cartilagehomeostasis and is thus thought to play a crucial role during the pathogenesis of osteoarthritis(OA). However, the underlying mechanisms of phenotypical alteration in chondrocytes areincompletely understood. Here, we provide evidence that disruption of the intracellularvimentin network and consequent phenotypical alteration in human chondrocytes results in anexternalization of the intermediate filament. The presence of the so-called cell surfacevimentin (CSV) on chondrocytes was associated with the severity of tissue degeneration inclinical OA samples and was enhanced after mechanical injury of cartilage tissue. By meansof a doxorubicine-based in vitro model of stress-induced premature senescence (SIPS), wecould confirm the connection between cellular senescence and amount of CSV. AlthoughsiRNA-mediated silencing of CDKN2A clearly reduced the senescent phenotype as well asCSV levels of human chondrocytes, cellular senescence could not be completely reversed.Interestingly, knockdown of vimentin resulted in a SIPS-like phenotype and consequentlyincreased CSV. Therefore, we concluded that the integrity of the intracellular vimentinnetwork is crucial to maintain cellular function in chondrocytes. This assumption could beconfirmed by chemically- induced collapse of the vimentin network, which resulted in cellularstress and enhanced CSV expression. Regarding its biological function, CSV was found to beassociated with enhanced chondrocyte adhesion and plasticity. While osteogenic capacitiesseemed to be enhanced in chondrocytes expressing high levels of CSV, the chondrogenicpotential was clearly compromised. Overall, our study reinforces the importance of thevimentin network in maintenance of the chondrogenic phenotype and introduces CSV as anovel membrane-bound marker of dysfunctional chondrocytes.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| | - Rolf E Brenner
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, University of UlmUlmGermany
| |
Collapse
|
8
|
Zhang C, Wang G, An Y. Achieving Nasal Septal Cartilage In Situ Regeneration: Focus on Cartilage Progenitor Cells. Biomolecules 2023; 13:1302. [PMID: 37759702 PMCID: PMC10527213 DOI: 10.3390/biom13091302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/10/2023] [Accepted: 08/17/2023] [Indexed: 09/29/2023] Open
Abstract
The nasal septal cartilage plays an important role in preventing the collapse of the nasal bones and maintaining the appearance of the nose. In the context of inherent difficulties regarding septal cartilage repair and the shortage of cartilage graft resources for regeneration, tissue engineering, especially the in situ strategy based on scaffolds, has become a new prospect and become one of the most promising approaches. Given that it is difficult for chondrocytes to achieve directional migration and secrete matrix components to participate in tissue repair after cartilage injury, cartilage progenitor cells (CPCs), with great migratory ability and stem cell characteristics, have caught the attention of researchers and brought hope for nasal septal cartilage in situ regeneration. In this review, we first summarized the distribution, characteristics, isolation, and culture methods of nasal septal CPCs. Subsequently, we described the roles of migratory CPCs in cartilage regeneration. Finally, we reviewed the existing studies on CPCs-based cartilage tissue engineering and summarized the strategies for promoting the migration and chondrogenesis of CPCs so as to provide ideas for achieving nasal septal cartilage in situ regeneration.
Collapse
Affiliation(s)
| | | | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, 49 North Garden Road, Haidian District, Beijing 100191, China; (C.Z.)
| |
Collapse
|
9
|
Muthu S, Korpershoek JV, Novais EJ, Tawy GF, Hollander AP, Martin I. Failure of cartilage regeneration: emerging hypotheses and related therapeutic strategies. Nat Rev Rheumatol 2023; 19:403-416. [PMID: 37296196 DOI: 10.1038/s41584-023-00979-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2023] [Indexed: 06/12/2023]
Abstract
Osteoarthritis (OA) is a disabling condition that affects billions of people worldwide and places a considerable burden on patients and on society owing to its prevalence and economic cost. As cartilage injuries are generally associated with the progressive onset of OA, robustly effective approaches for cartilage regeneration are necessary. Despite extensive research, technical development and clinical experimentation, no current surgery-based, material-based, cell-based or drug-based treatment can reliably restore the structure and function of hyaline cartilage. This paucity of effective treatment is partly caused by a lack of fundamental understanding of why articular cartilage fails to spontaneously regenerate. Thus, research studies that investigate the mechanisms behind the cartilage regeneration processes and the failure of these processes are critical to instruct decisions about patient treatment or to support the development of next-generation therapies for cartilage repair and OA prevention. This Review provides a synoptic and structured analysis of the current hypotheses about failure in cartilage regeneration, and the accompanying therapeutic strategies to overcome these hurdles, including some current or potential approaches to OA therapy.
Collapse
Affiliation(s)
- Sathish Muthu
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, New Delhi, India
- Department of Biotechnology, Faculty of Engineering, Karpagam Academy of Higher Education, Coimbatore, India
| | - Jasmijn V Korpershoek
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, Netherlands
| | - Emanuel J Novais
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Gwenllian F Tawy
- Division of Cell Matrix Biology & Regenerative Medicine, University of Manchester, Manchester, UK
| | - Anthony P Hollander
- Institute of Lifecourse and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
10
|
Chen Y, Liao G, Ma T, Li L, Yang J, Shen B, Lu Y, Si H. YY1/miR-140-5p/Jagged1/Notch axis mediates cartilage progenitor/stem cells fate reprogramming in knee osteoarthritis. Int Immunopharmacol 2023; 121:110438. [PMID: 37295026 DOI: 10.1016/j.intimp.2023.110438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/11/2023]
Abstract
Osteoarthritis is a multifactorial disease characterized by cartilage degeneration, while cartilage progenitor/stem cells (CPCs) are responsible for endogenous cartilage repair. However, the relevant regulatory mechanisms of CPCs fate reprogramming in OA are rarely reported. Recently, we observed fate disorders in OA CPCs and found that microRNA-140-5p (miR-140-5p) protects CPCs from fate changes in OA. This study further mechanistically investigated the upstream regulator and downstream effectors of miR-140-5p in OA CPCs fate reprogramming. As a result, luciferase reporter assay and validation assays revealed that miR-140-5p targets Jagged1 and inhibits Notch signaling in human CPCs, and the loss-/gain-of-function experiments and rescue assays discovered that miR-140-5p improves OA CPCs fate, but this effect can be counteracted by Jagged1. Moreover, increased transcription factor Ying Yang 1 (YY1) was associated with OA progression, and YY1 could disturb CPCs fate via transcriptionally repressing miR-140-5p and enhancing the Jagged1/Notch signaling. Finally, the relevant changes and mechanisms of YY1, miR-140-5p, and Jagged1/Notch signaling in OA CPCs fate reprogramming were validated in rats. Conclusively, this study identified a novel YY1/miR-140-5p/Jagged1/Notch signaling axis that mediates OA CPCs fate reprogramming, wherein YY1 and Jagged1/Notch signaling exhibits an OA-stimulative role, and miR-140-5p plays an OA-protective effect, providing attractive targets for OA therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guangneng Liao
- Experimental Animal Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Ma
- Department of Operating Room of Anesthesia Surgery Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lan Li
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Yang
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Shen
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yanrong Lu
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haibo Si
- Department of Orthopedic Surgery & Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
11
|
Chen Y, Huang H, Zhong W, Li L, Lu Y, Si HB. miR-140-5p protects cartilage progenitor/stem cells from fate changes in knee osteoarthritis. Int Immunopharmacol 2023; 114:109576. [PMID: 36527878 DOI: 10.1016/j.intimp.2022.109576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022]
Abstract
Cartilage progenitor/stem cells (CPCs) are promising seed cells for cartilage regeneration, but their fate changes and regulatory mechanisms in osteoarthritis (OA) pathogenesis remain unclear. This study aimed to investigate the role and potential mechanism of the microRNA-140-5p (miR-140-5p), whose protective role in knee OA has been confirmed by our previous studies, in OA CPCs fate reprogramming. Firstly, the normal and OA CPCs were isolated, and the fate indicators, miR-140-5p, Jagged1, and Notch signals were detected and analyzed. Then, the effect of miR-140-5p and the Notch pathway on CPCs fate reprogramming and miR-140-5p on Jagged1/Notch signaling was investigated in IL-1β-induced chondrocytes in vitro. Finally, the effect of miR-140-5p on OA CPCs fate reprogramming and the potential mechanisms were validated in OA rats. As a result, CPCs percentage was increased in the mild OA cartilage-derived total chondrocytes while decreased in the advanced OA group. Significant fate changes (including reduced cell viability, migration, chondrogenesis, and increased apoptosis), increased Jagged1 and Notch signals, and reduced miR-140-5p were observed in OA CPCs and associated with OA progression. IL-1β induced OA-like changes in CPCs fate, which could be exacerbated by miR-140-5p inhibitor while alleviated by DAPT (a specific Notch inhibitor) and miR-140-5p mimic. Finally, the in vitro phenomenal and mechanistic findings were validated in OA rats. Overall, miR-140-5p protects CPCs from fate changes via inhibiting Jagged1/Notch signaling in knee OA, providing attractive targets for OA therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Hua Huang
- Department of Orthopedics, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Wen Zhong
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lan Li
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yanrong Lu
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Hai-Bo Si
- Department of Orthopedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
12
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
13
|
Li S, Co CM, Izuagbe S, Hong Y, Liao J, Borrelli J, Tang L. Biomolecules-releasing click chemistry-based bioadhesives for repairing acetabular labrum tears. J Orthop Res 2022; 40:2646-2655. [PMID: 35112388 DOI: 10.1002/jor.25290] [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: 04/01/2021] [Revised: 12/07/2021] [Accepted: 01/30/2022] [Indexed: 02/04/2023]
Abstract
Currently, there are no effective clinical or experimental treatments to fully restore the function of the torn acetabular labrum. To fill the gap, here, we report the finding of progenitor cells in labral tissue, which can be recruited and stimulated to repair torn acetabular labral tissue. This study aimed to develop a biomolecule releasing bioadhesive which can speed up labral tissue healing by eliciting autologous labral progenitor cellular responses. A click chemistry-based bioadhesive, capable of releasing biomolecules, was synthesized to exert ~3× adhesion strength compared with fibrin glue. Via the release of platelet-derived growth factor (PDGF), the adhesive was shown to actively recruit and stimulate the proliferation of labral progenitor cells to the tear sites and within the adhesive. Finally, the ability of this biomolecules-releasing adhesive designed to promote labral tissue regeneration was evaluated using discarded human acetabular labrum tissue compared with surgical suture ex vivo. Histological analysis shows that PDGF-releasing bioadhesive yielded significantly more labrum cell responses and extracellular matrix protein (proteoglycan and collagen) production at the tear tissue site than surgical suture controls. The results confirm that the new PDGF-releasing bioadhesive can activate the responses of autologous labral progenitor cells to significantly improve labral tissue regeneration. Clinical significance: These PDGF-releasing bioadhesives may serve as a new and effective tool for repairing and regenerating acetabular labrum tears.
Collapse
Affiliation(s)
- Shuxin Li
- Department of Research & Development, Progenitec Inc., Arlington, Texas, USA
| | - Cynthia M Co
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Samira Izuagbe
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Joseph Borrelli
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Liping Tang
- Department of Research & Development, Progenitec Inc., Arlington, Texas, USA.,Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
14
|
Stavely R, Hotta R, Picard N, Rahman AA, Pan W, Bhave S, Omer M, Ho WLN, Guyer RA, Goldstein AM. Schwann cells in the subcutaneous adipose tissue have neurogenic potential and can be used for regenerative therapies. Sci Transl Med 2022; 14:eabl8753. [PMID: 35613280 PMCID: PMC9745588 DOI: 10.1126/scitranslmed.abl8753] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stem cell therapies for nervous system disorders are hindered by a lack of accessible autologous sources of neural stem cells (NSCs). In this study, neural crest-derived Schwann cells are found to populate nerve fiber bundles (NFBs) residing in mouse and human subcutaneous adipose tissue (SAT). NFBs containing Schwann cells were harvested from mouse and human SAT and cultured in vitro. During in vitro culture, SAT-derived Schwann cells remodeled NFBs to form neurospheres and exhibited neurogenic differentiation potential. Transcriptional profiling determined that the acquisition of these NSC properties can be attributed to dedifferentiation processes in cultured Schwann cells. The emerging population of cells were termed SAT-NSCs because of their considerably distinct gene expression profile, cell markers, and differentiation potential compared to endogenous Schwann cells existing in vivo. SAT-NSCs successfully engrafted to the gastrointestinal tract of mice, migrated longitudinally and circumferentially within the muscularis, differentiated into neurons and glia, and exhibited neurochemical coding and calcium signaling properties consistent with an enteric neuronal phenotype. These cells rescued functional deficits associated with colonic aganglionosis and gastroparesis, indicating their therapeutic potential as a cell therapy for gastrointestinal dysmotility. SAT can be harvested easily and offers unprecedented accessibility for the derivation of autologous NSCs from adult tissues. Evidence from this study indicates that SAT-NSCs are not derived from mesenchymal stem cells and instead originate from Schwann cells within NFBs. Our data describe efficient isolation procedures for mouse and human SAT-NSCs and suggest that these cells have potential for therapeutic applications in gastrointestinal motility disorders.
Collapse
|
15
|
Gu J, Wang B, Wang T, Zhang N, Liu H, Gui J, Lu Y. Effects of Cartilage Progenitor Cells, Bone Marrow Mesenchymal Stem Cells and Chondrocytes on Cartilage Repair as Seed Cells: An in vitro Study. Drug Des Devel Ther 2022; 16:1217-1230. [PMID: 35509492 PMCID: PMC9059879 DOI: 10.2147/dddt.s356936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Jiaxiang Gu
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Bin Wang
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Tianliang Wang
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Naichen Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Hongjun Liu
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
| | - Jianchao Gui
- Department of Orthopedics, Nanjing Medical University Affiliated Nanjing First Hospital, Nanjing, People’s Republic of China
| | - Yiming Lu
- Clinical Medical College, Yangzhou University, Yangzhou, People’s Republic of China
- Department of Foot and Hand Surgery, Northern Jiangsu People’s Hospital, Yangzhou, People’s Republic of China
- Correspondence: Yiming Lu, Email
| |
Collapse
|
16
|
Lan X, Liang Y, Vyhlidal M, Erkut EJN, Kunze M, Mulet-Sierra A, Osswald M, Ansari K, Seikaly H, Boluk Y, Adesida AB. In vitro maturation and in vivo stability of bioprinted human nasal cartilage. J Tissue Eng 2022; 13:20417314221086368. [PMID: 35599742 PMCID: PMC9122109 DOI: 10.1177/20417314221086368] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 02/23/2022] [Indexed: 11/25/2022] Open
Abstract
The removal of skin cancer lesions on the nose often results in the loss of nasal
cartilage. The cartilage loss is either surgically replaced with autologous
cartilage or synthetic grafts. However, these replacement options come with
donor-site morbidity and resorption issues. 3-dimensional (3D) bioprinting
technology offers the opportunity to engineer anatomical-shaped autologous nasal
cartilage grafts. The 3D bioprinted cartilage grafts need to embody a
mechanically competent extracellular matrix (ECM) to allow for surgical suturing
and resistance to contraction during scar tissue formation. We investigated the
effect of culture period on ECM formation and mechanical properties of 3D
bioprinted constructs of human nasal chondrocytes (hNC)-laden type I collagen
hydrogel in vitro and in vivo. Tissue-engineered nasal cartilage constructs
developed from hNC culture in clinically approved collagen type I and type III
semi-permeable membrane scaffold served as control. The resulting 3D bioprinted
engineered nasal cartilage constructs were comparable or better than the
controls both in vitro and in vivo. This study demonstrates that 3D bioprinted
constructs of engineered nasal cartilage are feasible options in nasal cartilage
reconstructive surgeries.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Margaret Vyhlidal
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Esra JN Erkut
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Martin Osswald
- Institute for Reconstructive Sciences in Medicine, Misericordia Community Hospital, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Khalid Ansari
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Hadi Seikaly
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yaman Boluk
- Department of Civil and Environmental Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Adetola B Adesida
- Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
17
|
Scialla S, Gullotta F, Izzo D, Palazzo B, Scalera F, Martin I, Sannino A, Gervaso F. Genipin-crosslinked collagen scaffolds inducing chondrogenesis: a mechanical and biological characterization. J Biomed Mater Res A 2022; 110:1372-1385. [PMID: 35262240 DOI: 10.1002/jbm.a.37379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 12/14/2022]
Abstract
Articular cartilage degeneration is still an unsolved issue owing to its weak repairing capabilities, which usually result in fibrocartilage tissue formation. This fibrous tissue lacks of structural and bio-mechanical properties, degrading over time. Currently, arthroscopic techniques and autologous transplantation are the most used clinical procedures. However, rather than restoring cartilage integrity, these methods only postpone further cartilage deterioration. Therefore, tissue engineering strategies aimed at selecting scaffolds that remarkably support the chondrogenic differentiation of human mesenchymal stem cells (hMSCs) could represent a promising solution, but they are still challenging for researchers. In this study, the influence of two different genipin (Gp) crosslinking routes on collagen (Coll)-based scaffolds in terms of hMSCs chondrogenic differentiation and biomechanical performances was investigated. Three-dimensional (3D) porous Coll scaffolds were fabricated by freeze-drying techniques and were crosslinked with Gp following a "two-step" and an in "bulk" procedure, in order to increase the physico-mechanical stability of the structure. Chondrogenic differentiation efficacy of hMSCs and biomechanical behavior under compression forces through unconfined stress-strain tests were assessed. Coll/Gp scaffolds revealed an isotropic and highly homogeneous pore distribution along with an increase in the stiffness, also supported by the increase in the Coll denaturation temperature (Td = 57-63°C) and a significant amount of Coll and GAG deposition during the 3 weeks of chondrogenic culture. In particular, the presence of Gp in "bulk" led to a more uniform and homogenous chondral-like matrix deposition by hMSCs if compared to the results obtained from the Gp "two-step" functionalization procedure.
Collapse
Affiliation(s)
- Stefania Scialla
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,Institute of Polymers, Composites and Biomaterials - National Research Council, Naples, Italy
| | - Fabiana Gullotta
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Daniela Izzo
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Barbara Palazzo
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,ENEA, Division for Sustainable Materials - Research Centre of Brindisi, Brindisi, Italy
| | - Francesca Scalera
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,CNR Nanotec - Institute of Nanotechnology, Lecce, Italy
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Lecce, Italy
| | - Francesca Gervaso
- Department of Engineering for Innovation, University of Salento, Lecce, Italy.,CNR Nanotec - Institute of Nanotechnology, Lecce, Italy
| |
Collapse
|
18
|
The clinical potential of articular cartilage-derived progenitor cells: a systematic review. NPJ Regen Med 2022; 7:2. [PMID: 35013329 PMCID: PMC8748760 DOI: 10.1038/s41536-021-00203-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 11/30/2021] [Indexed: 01/09/2023] Open
Abstract
Over the past two decades, evidence has emerged for the existence of a distinct population of endogenous progenitor cells in adult articular cartilage, predominantly referred to as articular cartilage-derived progenitor cells (ACPCs). This progenitor population can be isolated from articular cartilage of a broad range of species, including human, equine, and bovine cartilage. In vitro, ACPCs possess mesenchymal stromal cell (MSC)-like characteristics, such as colony forming potential, extensive proliferation, and multilineage potential. Contrary to bone marrow-derived MSCs, ACPCs exhibit no signs of hypertrophic differentiation and therefore hold potential for cartilage repair. As no unique cell marker or marker set has been established to specifically identify ACPCs, isolation and characterization protocols vary greatly. This systematic review summarizes the state-of-the-art research on this promising cell type for use in cartilage repair therapies. It provides an overview of the available literature on endogenous progenitor cells in adult articular cartilage and specifically compares identification of these cell populations in healthy and osteoarthritic (OA) cartilage, isolation procedures, in vitro characterization, and advantages over other cell types used for cartilage repair. The methods for the systematic review were prospectively registered in PROSPERO (CRD42020184775).
Collapse
|
19
|
Guerrero J, Dasen B, Frismantiene A, Pigeot S, Ismail T, Schaefer DJ, Philippova M, Resink TJ, Martin I, Scherberich A. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:213-229. [PMID: 35259280 PMCID: PMC8929526 DOI: 10.1093/stcltm/szab021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 10/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cells of the stromal vascular fraction (SVF) of human adipose tissue have the capacity to generate osteogenic grafts with intrinsic vasculogenic properties. However, cultured adipose-derived stromal cells (ASCs), even after minimal monolayer expansion, lose osteogenic capacity in vivo. Communication between endothelial and stromal/mesenchymal cell lineages has been suggested to improve bone formation and vascularization by engineered tissues. Here, we investigated the specific role of a subpopulation of SVF cells positive for T-cadherin (T-cad), a putative endothelial marker. We found that maintenance during monolayer expansion of a T-cad-positive cell population, composed of endothelial lineage cells (ECs), is mandatory to preserve the osteogenic capacity of SVF cells in vivo and strongly supports their vasculogenic properties. Depletion of T-cad-positive cells from the SVF totally impaired bone formation in vivo and strongly reduced vascularization by SVF cells in association with decreased VEGF and Adiponectin expression. The osteogenic potential of T-cad-depleted SVF cells was fully rescued by co-culture with ECs from a human umbilical vein (HUVECs), constitutively expressing T-cad. Ectopic expression of T-cad in ASCs stimulated mineralization in vitro but failed to rescue osteogenic potential in vivo, indicating that the endothelial nature of the T-cad-positive cells is the key factor for induction of osteogenesis in engineered grafts based on SVF cells. This study demonstrates that crosstalk between stromal and T-cad expressing endothelial cells within adipose tissue critically regulates osteogenesis, with VEGF and adiponectin as associated molecular mediators.
Collapse
Affiliation(s)
- Julien Guerrero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Boris Dasen
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Agne Frismantiene
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sebastien Pigeot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Tarek Ismail
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Maria Philippova
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Therese J Resink
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Arnaud Scherberich
- Corresponding author: Arnaud Scherberich, Department of Biomedicine, Hebelstrasse 20, University Hospital Basel, 4031 Basel, Switzerland. Tel: +41 061 328 73 75;
| |
Collapse
|
20
|
Peng Y, Li J, Lin H, Tian S, Liu S, Pu F, Zhao L, Ma K, Qing X, Shao Z. Endogenous repair theory enriches construction strategies for orthopaedic biomaterials: a narrative review. BIOMATERIALS TRANSLATIONAL 2021; 2:343-360. [PMID: 35837417 PMCID: PMC9255795 DOI: 10.12336/biomatertransl.2021.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
The development of tissue engineering has led to new strategies for mitigating clinical problems; however, the design of the tissue engineering materials remains a challenge. The limited sources and inadequate function, potential risk of microbial or pathogen contamination, and high cost of cell expansion impair the efficacy and limit the application of exogenous cells in tissue engineering. However, endogenous cells in native tissues have been reported to be capable of spontaneous repair of the damaged tissue. These cells exhibit remarkable plasticity, and thus can differentiate or be reprogrammed to alter their phenotype and function after stimulation. After a comprehensive review, we found that the plasticity of these cells plays a major role in establishing the cell source in the mechanism involved in tissue regeneration. Tissue engineering materials that focus on assisting and promoting the natural self-repair function of endogenous cells may break through the limitations of exogenous seed cells and further expand the applications of tissue engineering materials in tissue repair. This review discusses the effects of endogenous cells, especially stem cells, on injured tissue repairing, and highlights the potential utilisation of endogenous repair in orthopaedic biomaterial constructions for bone, cartilage, and intervertebral disc regeneration.
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jinye Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Feifei Pu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lei Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
21
|
Acevedo L, Iselin L, Berkelaar MHM, Salzmann GM, Wolf F, Feliciano S, Vogel N, Pagenstert G, Martin I, Pelttari K, Barbero A, Arnold MP. Comparison of Human Articular Cartilage Tissue and Chondrocytes Isolated from Peripheral versus Central Regions of Traumatic Lesions. Cartilage 2021; 13:68S-81S. [PMID: 32959685 PMCID: PMC8804865 DOI: 10.1177/1947603520958154] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE Cellular and molecular events occurring in cartilage regions close to injury are poorly investigated, but can possibly compromise the outcome of cell-based cartilage repair. In this study, key functional properties were assessed for cartilage biopsies collected from the central part of traumatic joint lesions (central) and from regions surrounding the defect (peripheral). These properties were then correlated with the quality of the initial cartilage biopsy and the inflammatory state of the joint. DESIGN Cartilage samples were collected from knee joints of 42 patients with traumatic knee injuries and analyzed for cell phenotype (by reverse transcriptas-polymerase chain reaction), histological quality, cellularity, cell viability, proliferation capacity, and post-expansion chondrogenic capacity of chondrocytes (in pellet culture). Synovium was also harvested and analyzed for the expression of inflammatory cytokines. RESULTS Cartilage quality and post-expansion chondrogenic capacity were higher in peripheral versus central samples. Differences between these 2 parameters were more pronounced in joints with high inflammatory features characterized by >100-fold difference in the mRNA levels of IL6 and IL8 in the corresponding synovium. Peripheral chondrocytes isolated from good- versus bad-quality biopsies expressed higher levels of collagen II/I and aggrecan/versican and lower levels of MMP13 and ADAMTS5. They also exhibited reduced proliferation and enhanced cartilage-forming capacity. CONCLUSIONS Chondrocytes at the periphery of traumatic lesions better maintain properties of healthy cartilage compared to those isolated from the center, even when derived from bad-quality tissues harvested from highly inflamed joints. Future studies are necessary to investigate the change of functional properties of peripheral chondrocytes over time.
Collapse
Affiliation(s)
- Lina Acevedo
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Lukas Iselin
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland,Department of Orthopedics and
Traumatology, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Majoska H. M. Berkelaar
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | | | - Francine Wolf
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Sandra Feliciano
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Nicole Vogel
- Practice LEONARDO, Hirslanden Clinic
Birshof, Münchenstein, Switzerland
| | - Geert Pagenstert
- Department of Surgery, University
Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University
Hospital Basel, University of Basel, Basel, Switzerland,Andrea Barbero, Department of Biomedicine,
University Hospital Basel, University of Basel, Basel 4031, Switzerland.
| | - Markus P. Arnold
- Practice LEONARDO, Hirslanden Clinic
Birshof, Münchenstein, Switzerland
| |
Collapse
|
22
|
Philippe V, Laurent A, Abdel-Sayed P, Hirt-Burri N, Ann Applegate L, Martin R. Human Platelet Lysate as an Alternative to Autologous Serum for Human Chondrocyte Clinical Use. Cartilage 2021; 13:509S-518S. [PMID: 34330164 PMCID: PMC8808884 DOI: 10.1177/19476035211035433] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE A pivotal aspect of cartilage tissue engineering resides in cell culture medium supplementation, in view of maximizing in vitro cell proliferation and preserving cellular functionality. Autologous human serum (aHS) is commonly used as an inducive supplement for safe human articular chondrocyte (HAC) proliferation prior to clinical implantation. However, practical clinical use of aHS is hindered by constraining manufacturing requirements and quality assurance-driven downstream processing. The present study investigated potential alternative use of commercial human platelet lysate (hPL) supplements in HAC manufacturing workflows related to clinical therapeutic pathways. DESIGN Differential effects of hPL, aHS, and fetal bovine serum were assessed on primary cultured HAC parameters (viability, proliferative rates, and morphology) in 2-dimensional in vitro systems. A 3-dimensional HAC pellet model served for postexpansion assessment of cellular functionality, by visualizing proteoglycan production (Alcian blue staining), and by using qRT-PCR relative quantification of chondrogenic marker (SOX9, COL2-A1, and ACAN) genetic expression. RESULTS We found that monolayer HAC culture with hPL or aHS supplements presented similar characteristics (elongated cell morphology and nearly identical growth kinetics). Chondrogenic activity appeared as conserved in HACs expanded with human or bovine supplements, wherein histologic analysis indicated a progressive sGAG accumulation and SOX9, COL2-A1, ACAN gene expression was upregulated in 3-dimensional HAC pellet models. CONCLUSION This study therefore supports the use of hPL as a functional equivalent and alternative to aHS for cultured HAC batch preparation, with the potential to effectively alleviate pressure on clinical and manufacturing bottlenecks in cell therapy approaches for cartilage regeneration.
Collapse
Affiliation(s)
- Virginie Philippe
- Service of Orthopaedic Surgery and
Traumatology, Lausanne University Hospital, University of Lausanne,
Switzerland,Regenerative Therapy Unit, Lausanne
University Hospital, University of Lausanne, Switzerland,Virginie Philippe, Service of Orthopaedic
Surgery and Traumatology, Lausanne University Hospital, Pierre-Decker 4,
Lausanne, CH-1011, Switzerland. Email
| | - Alexis Laurent
- Regenerative Therapy Unit, Lausanne
University Hospital, University of Lausanne, Switzerland
| | - Philippe Abdel-Sayed
- Regenerative Therapy Unit, Lausanne
University Hospital, University of Lausanne, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne
University Hospital, University of Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne
University Hospital, University of Lausanne, Switzerland
| | - Robin Martin
- Service of Orthopaedic Surgery and
Traumatology, Lausanne University Hospital, University of Lausanne,
Switzerland
| |
Collapse
|
23
|
Rikkers M, Korpershoek J, Levato R, Malda J, Vonk L. Progenitor Cells in Healthy and Osteoarthritic Human Cartilage Have Extensive Culture Expansion Capacity while Retaining Chondrogenic Properties. Cartilage 2021; 13:129S-142S. [PMID: 34802263 PMCID: PMC8804833 DOI: 10.1177/19476035211059600] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Articular cartilage-derived progenitor cells (ACPCs) are a potential new cell source for cartilage repair. This study aims to characterize endogenous ACPCs from healthy and osteoarthritic (OA) cartilage, evaluate their potential for cartilage regeneration, and compare this to cartilage formation by chondrocytes. DESIGN ACPCs were isolated from full-thickness healthy and OA human cartilage and separated from the total cell population by clonal growth after differential adhesion to fibronectin. ACPCs were characterized by growth kinetics, multilineage differentiation, and surface marker expression. Chondrogenic redifferentiation of ACPCs was compared with chondrocytes in pellet cultures. Pellets were assessed for cartilage-like matrix production by (immuno)histochemistry, quantitative analyses for glycosaminoglycans and DNA content, and expression of chondrogenic and hypertrophic genes. RESULTS Healthy and OA ACPCs were successfully differentiated toward the adipogenic and chondrogenic lineage, but failed to produce calcified matrix when exposed to osteogenic induction media. Both ACPC populations met the criteria for cell surface marker expression of mesenchymal stromal cells (MSCs). Healthy ACPCs cultured in pellets deposited extracellular matrix containing proteoglycans and type II collagen, devoid of type I collagen. Gene expression of hypertrophic marker type X collagen was lower in healthy ACPC pellets compared with OA pellets. CONCLUSIONS This study provides further insight into the ACPC population in healthy and OA human articular cartilage. ACPCs show similarities to MSCs, yet do not produce calcified matrix under well-established osteogenic culture conditions. Due to extensive proliferative potential and chondrogenic capacity, ACPCs show potential for cartilage regeneration and possibly for clinical application, as a promising alternative to MSCs or chondrocytes.
Collapse
Affiliation(s)
- M. Rikkers
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - J.V. Korpershoek
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - R. Levato
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,Department of Clinical Sciences,
Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - J. Malda
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,Department of Clinical Sciences,
Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - L.A. Vonk
- Department of Orthopaedics, University
Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands,CO.DON AG, Teltow, Germany,L.A. Vonk, Department of Orthopaedics,
University Medical Center Utrecht, Utrecht University, P.O. Box 85500, 3508 GA
Utrecht, The Netherlands.
| |
Collapse
|
24
|
Lindberg GCJ, Cui X, Durham M, Veenendaal L, Schon BS, Hooper GJ, Lim KS, Woodfield TBF. Probing Multicellular Tissue Fusion of Cocultured Spheroids-A 3D-Bioassembly Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2103320. [PMID: 34632729 PMCID: PMC8596109 DOI: 10.1002/advs.202103320] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Indexed: 05/02/2023]
Abstract
While decades of research have enriched the knowledge of how to grow cells into mature tissues, little is yet known about the next phase: fusing of these engineered tissues into larger functional structures. The specific effect of multicellular interfaces on tissue fusion remains largely unexplored. Here, a facile 3D-bioassembly platform is introduced to primarily study fusion of cartilage-cartilage interfaces using spheroids formed from human mesenchymal stromal cells (hMSCs) and articular chondrocytes (hACs). 3D-bioassembly of two adjacent hMSCs spheroids displays coordinated migration and noteworthy matrix deposition while the interface between two hAC tissues lacks both cells and type-II collagen. Cocultures contribute to increased phenotypic stability in the fusion region while close initial contact between hMSCs and hACs (mixed) yields superior hyaline differentiation over more distant, indirect cocultures. This reduced ability of potent hMSCs to fuse with mature hAC tissue further underlines the major clinical challenge that is integration. Together, this data offer the first proof of an in vitro 3D-model to reliably study lateral fusion mechanisms between multicellular spheroids and mature cartilage tissues. Ultimately, this high-throughput 3D-bioassembly model provides a bridge between understanding cellular differentiation and tissue fusion and offers the potential to probe fundamental biological mechanisms that underpin organogenesis.
Collapse
Affiliation(s)
- Gabriella C. J. Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Mitchell Durham
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Laura Veenendaal
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Benjamin S. Schon
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Gary J. Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Khoon S. Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| | - Tim B. F. Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) GroupDepartment of Orthopaedic SurgeryUniversity of Otago Christchurch2 Riccarton AvenueChristchurch8011New Zealand
| |
Collapse
|
25
|
Linde PE, Puttlitz CM, Kisiday JD. Adult ovine connective tissue cells resemble mesenchymal stromal cells in their propensity for extensive ex vivo expansion. Connect Tissue Res 2021; 62:671-680. [PMID: 33153311 DOI: 10.1080/03008207.2020.1847099] [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] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: Expanded, human connective tissue cells can adopt mesenchymal stromal cell (MSC) properties that are favorable for applications in regenerative medicine. Sheep are used as a large animal model for cell therapies, although for preclinical testing it is important to establish whether ovine cells resemble humans in their tendency to adopt MSC properties. The objective of this study was to investigate whether cells from five ovine connective tissues are MSC-like in their propensity for extensive expansion and immunophenotype.Materials and Methods: Monolayer cultures were established with cells from annulus fibrosus, cartilage, meniscus, tendon, and nucleus pulposus. Bone marrow MSCs were evaluated as a control. Cultures were seeded at 500 cells/cm2, and subcultured every 5 days up to day 20. Flow cytometry was used to evaluate expression of cluster of differentiation (CD) molecules associated with MSCs (29, 44, 166). Colony formation was evaluated using time-lapse imaging of individual cells.Results: By day 20, cumulative population doublings ranged between 22 (chondrocytes) and 27 (MSCs). All cells uniformly expressed CD44 and 73. Expression of CD166 for MSCs was 98-99%, and ranged between 64 and 97% for the other cell types. Time-lapse imaging demonstrated that 58-94% of the cells colonized as indicated by 3 population doublings within 52 hours.Conclusions: Cells from ovine connective tissues resembled MSCs in their propensity for sustained, colony-forming growth and expression of CD molecules. These data supports the potential for preclinical testing of MSC-like connective tissue cells in sheep.
Collapse
Affiliation(s)
- Peter E Linde
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - Christian M Puttlitz
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, Colorado, USA
| | - John D Kisiday
- Orthopaedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
26
|
Donahue RP, Nordberg RC, Bielajew BJ, Hu JC, Athanasiou KA. The effect of neonatal, juvenile, and adult donors on rejuvenated neocartilage functional properties. Tissue Eng Part A 2021; 28:383-393. [PMID: 34605665 PMCID: PMC9131355 DOI: 10.1089/ten.tea.2021.0167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cartilage does not naturally heal, and cartilage lesions from trauma and wear-and-tear can lead to eventual osteoarthritis. To address long-term repair, tissue engineering of functional biologic implants to treat cartilage lesions is desirable, but the development of such implants is hindered by several limitations including 1) donor tissue scarcity due to the presence of diseased tissues in joints, 2) dedifferentiation of chondrocytes during expansion, and 3) differences in functional output of cells dependent on donor age. Toward overcoming these challenges, 1) costal cartilage has been explored as a donor tissue, and 2) methods have been developed to rejuvenate the chondrogenic phenotype of passaged chondrocytes for generating self-assembled neocartilage. However, it remains unclear how the rejuvenation processes are influenced by donor age, and, thus, how to develop strategies that specifically target age-related differences. Using histological, biochemical, proteomic, and mechanical assays, this study sought to determine the differences among neocartilage generated from neonatal, juvenile, and adult donors using the Yucatan minipig, a clinically relevant large animal model. Based on the literature, a relatively young adult population of animals was chosen due to a reduction in functional output of human articular chondrocytes after 40 years of age. After isolation, costal chondrocytes were expanded, rejuvenated, and self-assembled, and the neocartilages were assessed. The aggregate modulus values of neonatal constructs were at least 1.65-fold of those from the juvenile or adult constructs. Poisson's ratio also significantly differed among all groups, with neonatal constructs exhibiting values 49% higher than adult constructs. Surprisingly, other functional properties such as tensile modulus and GAG content did not significantly differ among groups. Total collagen content was slightly elevated in the adult constructs when compared to neonatal and juvenile constructs. A more nuanced view via bottom-up mass spectrometry showed that Col2a1 protein was not significantly different among groups, but content of several other collagen subtypes (i.e., Col1a1, Col9a1, Col11a2, and Col12a1) was modulated by donor age. For example, Col12a1 in adult constructs was found to be 102.9% higher than neonatal-derived constructs. Despite these differences, this study shows that different aged donors can be used to generate neocartilages of similar functional properties.
Collapse
Affiliation(s)
- Ryan P Donahue
- University of California, Irvine, Biomedical Engineering, Irvine, California, United States;
| | - Rachel C Nordberg
- University of California, Irvine, Biomedical Engineering, Irvine, California, United States;
| | - Benjamin J Bielajew
- University of California, Irvine, Biomedical Engineering, Irvine, California, United States;
| | - Jerry C Hu
- University of California, Irvine, Biomedical Engineering, Irvine, California, United States;
| | - Kyriacos A Athanasiou
- University of California, Irvine, Biomedical Engineering, Irvine, California, United States;
| |
Collapse
|
27
|
Urlić I, Ivković A. Cell Sources for Cartilage Repair-Biological and Clinical Perspective. Cells 2021; 10:cells10092496. [PMID: 34572145 PMCID: PMC8468484 DOI: 10.3390/cells10092496] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/04/2023] Open
Abstract
Cell-based therapy represents a promising treatment strategy for cartilage defects. Alone or in combination with scaffolds/biological signals, these strategies open many new avenues for cartilage tissue engineering. However, the choice of the optimal cell source is not that straightforward. Currently, various types of differentiated cells (articular and nasal chondrocytes) and stem cells (mesenchymal stem cells, induced pluripotent stem cells) are being researched to objectively assess their merits and disadvantages with respect to the ability to repair damaged articular cartilage. In this paper, we focus on the different cell types used in cartilage treatment, first from a biological scientist’s perspective and then from a clinician’s standpoint. We compare and analyze the advantages and disadvantages of these cell types and offer a potential outlook for future research and clinical application.
Collapse
Affiliation(s)
- Inga Urlić
- Department of Biology, Faculty of Science, University of Zagreb, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| | - Alan Ivković
- Department of Orthopaedic Surgery, University Hospital Sveti Duh, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Clinical Medicine, University of Applied Health Sciences, 10000 Zagreb, Croatia
- Correspondence: (I.U.); (A.I.)
| |
Collapse
|
28
|
Acevedo Rua L, Mumme M, Manferdini C, Darwiche S, Khalil A, Hilpert M, Buchner DA, Lisignoli G, Occhetta P, von Rechenberg B, Haug M, Schaefer DJ, Jakob M, Caplan A, Martin I, Barbero A, Pelttari K. Engineered nasal cartilage for the repair of osteoarthritic knee cartilage defects. Sci Transl Med 2021; 13:eaaz4499. [PMID: 34516821 DOI: 10.1126/scitranslmed.aaz4499] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Lina Acevedo Rua
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcus Mumme
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Salim Darwiche
- Musculoskeletal Research Unit MSRU, Equine Department, University of Zurich, 8057 Zürich, Switzerland
| | - Ahmad Khalil
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Morgane Hilpert
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106 , USA
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Paola Occhetta
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Brigitte von Rechenberg
- Competence Center for Applied Biotechnology and Molecular Medicine CABMM, University of Zurich, 8057 Zürich, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Dirk J Schaefer
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Arnold Caplan
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14-16, 4123 Allschwil, Switzerland Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland
| |
Collapse
|
29
|
Korpershoek JV, Rikkers M, de Windt TS, Tryfonidou MA, Saris DBF, Vonk LA. Selection of Highly Proliferative and Multipotent Meniscus Progenitors through Differential Adhesion to Fibronectin: A Novel Approach in Meniscus Tissue Engineering. Int J Mol Sci 2021; 22:ijms22168614. [PMID: 34445320 PMCID: PMC8395239 DOI: 10.3390/ijms22168614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 12/26/2022] Open
Abstract
Meniscus injuries can be highly debilitating and lead to knee osteoarthritis. Progenitor cells from the meniscus could be a superior cell type for meniscus repair and tissue-engineering. The purpose of this study is to characterize meniscus progenitor cells isolated by differential adhesion to fibronectin (FN-prog). Human osteoarthritic menisci were digested, and FN-prog were selected by differential adhesion to fibronectin. Multilineage differentiation, population doubling time, colony formation, and MSC surface markers were assessed in the FN-prog and the total meniscus population (Men). Colony formation was compared between outer and inner zone meniscus digest. Chondrogenic pellet cultures were performed for redifferentiation. FN-prog demonstrated multipotency. The outer zone FN-prog formed more colonies than the inner zone FN-prog. FN-prog displayed more colony formation and a higher proliferation rate than Men. FN-prog redifferentiated in pellet culture and mostly adhered to the MSC surface marker profile, except for HLA-DR receptor expression. This is the first study that demonstrates differential adhesion to fibronectin for the isolation of a progenitor-like population from the meniscus. The high proliferation rates and ability to form meniscus extracellular matrix upon redifferentiation, together with the broad availability of osteoarthritis meniscus tissue, make FN-prog a promising cell type for clinical translation in meniscus tissue-engineering.
Collapse
Affiliation(s)
- Jasmijn V. Korpershoek
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.V.K.); (M.R.); (T.S.d.W.); (D.B.F.S.)
| | - Margot Rikkers
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.V.K.); (M.R.); (T.S.d.W.); (D.B.F.S.)
| | - Tommy S. de Windt
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.V.K.); (M.R.); (T.S.d.W.); (D.B.F.S.)
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands;
| | - Daniel B. F. Saris
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.V.K.); (M.R.); (T.S.d.W.); (D.B.F.S.)
- Department of Orthopedic Surgery and Sports Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Reconstructive medicine, University of Twente, 7522 NB Enschede, The Netherlands
| | - Lucienne A. Vonk
- Department of Orthopedics, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; (J.V.K.); (M.R.); (T.S.d.W.); (D.B.F.S.)
- Correspondence: ; Tel.: +49-0-3328-4346-25
| |
Collapse
|
30
|
Brose TZ, Kubosch EJ, Schmal H, Stoddart MJ, Armiento AR. Crosstalk Between Mesenchymal Stromal Cells and Chondrocytes: The Hidden Therapeutic Potential for Cartilage Regeneration. Stem Cell Rev Rep 2021; 17:1647-1665. [PMID: 33954877 DOI: 10.1007/s12015-021-10170-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Cartilage injuries following trauma create a puzzling clinical scenario. The finite reparative potential of articular cartilage is well known, and injuries are associated with an increased risk of osteoarthritis. Cell-based therapies have spotlighted chondrocytes and mesenchymal stromal cells (MSCs) as the functional unit of articular cartilage and the progenitor cells, respectively. The available clinical treatments cannot reproduce the biomechanical properties of articular cartilage and call for continuous investigations into alternative approaches. Co-cultures of chondrocytes and MSCs are an attractive in vitro system to step closer to the in vivo multicellular environment's complexity. Research on the mechanisms of interaction between both cell types will reveal essential cues to understand cartilage regeneration. This review describes the latest discoveries on these interactions, along with advantages and main challenges in vitro and in vivo. The successful clinical translation of in vitro studies requires establishing rigorous standards and clinically relevant research models and an organ-targeting therapeutic strategy.
Collapse
Affiliation(s)
- Teresa Z Brose
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Eva J Kubosch
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Hagen Schmal
- Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.,Department of Orthopaedics and Trauma Surgery, Medical Centre, Faculty of Medicine, Albert Ludwigs University of Freiburg, Hugstetterstrasse 55, 79106, Freiburg, Germany
| | - Angela R Armiento
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.
| |
Collapse
|
31
|
Duverney C, Abbasi H, Berkelaar M, Pelttari K, Cattin PC, Barbero A, Zam A, Rauter G. Sterile Tissue Ablation Using Laser Light—System Design, Experimental Validation, and Outlook on Clinical Applicability. J Med Device 2021; 15. [DOI: 10.1115/1.4049396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
Abstract
Preparation of biological samples for further processing or analysis is generally performed manually by means of standard mechanical tools such as scalpels or biopsy punches. While this approach is uncomplicated and swift, it entails constraints such as low, operator-dependent cutting accuracy and reproducibility. Tissue segments surrounding the cut may further suffer mechanical and thermal damage due to shear forces and friction between tool and sample. These hindrances affect procedures both in the laboratory environment as well as within clinical settings. A system has been developed leveraging robotic positioning and laser light for precise, controlled, and contactless tissue ablation, and providing a concise and intuitive graphical user interface. Additionally, sterility of the process is demonstrated, a paramount element for clinical application. The proposed process does not require sterilization of the robotic components or the lasers, easing a prospective integration into existing workflows. In the context of this work, mainly cartilage repair surgery is targeted. The proposed system allows for highly accurate and reproducible shaping of the cartilage lesion area as well as its corresponding engineered cartilage graft, possibly leading to better and faster integration at the defect site. Promising results could be obtained in a first test series with human cartilage samples, validating the functionality of the preparation system and the feasibility of the sterility concept.
Collapse
Affiliation(s)
- Cédric Duverney
- Bio-Inspired RObots for MEDicine-Laboratory (BIROMED-Lab), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, Basel-Landschaft 4123, Switzerland
| | - Hamed Abbasi
- Biomedical Laser and Optics Group (BLOG), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, Basel-Landschaft 4123, Switzerland
| | - Majoska Berkelaar
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, Basel, Basel-Stadt 4031, Switzerland
| | - Karoliina Pelttari
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, Basel, Basel-Stadt 4031, Switzerland
| | - Philippe C. Cattin
- Center for medical Image Analysis and Navigation (CIAN), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, Basel-Landschaft 4123, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University of Basel and University Hospital Basel, Hebelstrasse 20, Basel, Basel-Stadt 4031, Switzerland
| | - Azhar Zam
- Biomedical Laser and Optics Group (BLOG), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, Basel-Landschaft 4123, Switzerland
| | - Georg Rauter
- Bio-Inspired RObots for MEDicine-Laboratory (BIROMED-Lab), Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, Basel-Landschaft 4123, Switzerland
| |
Collapse
|
32
|
Bianchi VJ, Parsons M, Backstein D, Kandel RA. Endoglin Level Is Critical for Cartilage Tissue Formation In Vitro by Passaged Human Chondrocytes. Tissue Eng Part A 2021; 27:1140-1150. [PMID: 33323019 DOI: 10.1089/ten.tea.2020.0120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor beta (TGFβ) signaling is required for in vitro chondrogenesis. In animal models of osteoarthritis (OA), TGFβ receptor alterations are detected in chondrocytes in severe OA cartilage. It is not known whether such changes are dependent on the grade of human OA and if they affect chondrogenesis. Thus, the purpose of this study was to determine if human OA chondrocytes obtained from low-grade or high-grade disease could form cartilage tissue and to assess the role of the co-receptors, endoglin (ENG) and TGFβ receptor 3 (TGFBRIII), in the regulation of this tissue generation in vitro. We hypothesized that the grade of OA disease would not affect the ability of cells to form cartilage tissue and that the TGFβ co-receptor, ENG, would be critical to regulating tissue formation. Chondrocytes isolated from low-grade OA or high-grade OA human articular cartilage (AC) were analyzed directly (P0) or passaged in monolayer to P2. Expression of the primary TGFβ receptor ALK5, and the co-receptors ENG and TGFβRIII, was assessed by image flow cytometry. To assess the ability to form cartilaginous tissue, cells were placed in three-dimensional culture at high density and cultured in chondrogenic media containing TGFβ3. ENG knockdown was used to determine its role in regulating tissue formation. Overall, grade-specific differences in expression of ALK5, ENG, and TGFβRIII in primary or passaged chondrocytes were not detected; however, ENG expression increased significantly after passaging. Despite the presence of ALK5, P0 cells did not form cartilaginous tissue. In contrast, P2 cells derived from low-grade and high-grade OA AC formed hyaline-like cartilaginous tissues of similar quality. Knockdown of ENG in P2 cells inhibited cartilaginous tissue formation compared to controls indicating that the level of ENG protein expression is critical for in vitro chondrogenesis by passaged articular chondrocytes. This study demonstrates that it is not the grade of OA, but the levels of ENG in the presence of ALK5 that influences the ability of human passaged articular chondrocytes to form cartilaginous tissue in vitro in 3D culture. This has implications for cartilage repair therapies. Impact statement These findings are important clinically, given the limited availability of osteoarthritis (OA) cartilage tissue. Being able to use cells from all grades of OA will increase our ability to obtain sufficient cells for cartilage repair. In addition, it is possible that endoglin (ENG) levels, in the presence of ALK5 expression, may be suitable to use as biomarkers to identify cells able to produce cartilage.
Collapse
Affiliation(s)
- Vanessa J Bianchi
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | | | - David Backstein
- Division of Orthopaedic Surgery, Mount Sinai Hospital, Toronto, Canada
| | - Rita A Kandel
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| |
Collapse
|
33
|
Mou TC, Feng JY. Research advances in cartilage stem cells markers and induced differentiation. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:108-114. [PMID: 33723946 DOI: 10.7518/hxkq.2021.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cartilage stem cells (CSCs) are cells that self-proliferate, have surface antigen expression, and have multidirectional differentiation potential in the articular cartilage. CSCs, as an ideal source of stem cells, has a good application prospect in stem cell therapy. This article reviews the CSCs markers, cartilage differentiation signaling pathway, and clinical treatment of osteoarthritis.
Collapse
Affiliation(s)
- Ting-Chen Mou
- Dept. of Stomatological, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Jian-Ying Feng
- College of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
34
|
Kwon H, Brown WE, O'Leary SA, Hu JC, Athanasiou KA. Rejuvenation of extensively passaged human chondrocytes to engineer functional articular cartilage. Biofabrication 2021; 13. [PMID: 33418542 DOI: 10.1088/1758-5090/abd9d9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/08/2021] [Indexed: 11/11/2022]
Abstract
Human articular chondrocytes (hACs) are scarce and lose their chondrogenic potential during monolayer passaging, impeding their therapeutic use. This study investigated i) the translatability of conservative chondrogenic passaging and aggregate rejuvenation on restoring chondrogenic properties of hACs passaged up to P9; and ii) the efficacy of a combined treatment of TGF-β1 (T), chondroitinase-ABC (C), and lysyl oxidase-like 2 (L), collectively termed TCL, on engineering functional human neocartilage via the self-assembling process, as a function of passage number up to P11. Here, we show that aggregate rejuvenation enhanced glycosaminoglycan (GAG) content and type II collagen staining at all passages and yielded human neocartilage with chondrogenic phenotype present up to P7. Addition of TCL extended the chondrogenic phenotype to P11 and significantly enhanced GAG content and type II collagen staining at all passages. Human neocartilage derived from high passages, treated with TCL, displayed mechanical properties that were on par with or greater than those derived from low passages. Conservative chondrogenic passaging and aggregate rejuvenation may be a viable new strategy 1) to address the perennial problem of chondrocyte scarcity and 2) to successfully rejuvenate the chondrogenic phenotype of extensively passaged cells (up to P11). Furthermore, tissue engineering human neocartilage via self-assembly in conjunction with TCL treatment advances the clinical use of extensively passaged human chondrocytes for cartilage repair.
Collapse
Affiliation(s)
- Heenam Kwon
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Wendy E Brown
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Siobhan A O'Leary
- Align Technology Inc, 2820 Orchard Pkwy, San Jose, California, 95134, UNITED STATES
| | - Jerry C Hu
- University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| | - Kyriacos A Athanasiou
- Biomedical Engineering, University of California Irvine, University of California Irvine, Irvine, California, 92697, UNITED STATES
| |
Collapse
|
35
|
Krueger S, Riess A, Jonitz-Heincke A, Weizel A, Seyfarth A, Seitz H, Bader R. Establishment of a New Device for Electrical Stimulation of Non-Degenerative Cartilage Cells In Vitro. Int J Mol Sci 2021; 22:ijms22010394. [PMID: 33401406 PMCID: PMC7794805 DOI: 10.3390/ijms22010394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/17/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
In cell-based therapies for cartilage lesions, the main problem is still the formation of fibrous cartilage, caused by underlying de-differentiation processes ex vivo. Biophysical stimulation is a promising approach to optimize cell-based procedures and to adapt them more closely to physiological conditions. The occurrence of mechano-electrical transduction phenomena within cartilage tissue is physiological and based on streaming and diffusion potentials. The application of exogenous electric fields can be used to mimic endogenous fields and, thus, support the differentiation of chondrocytes in vitro. For this purpose, we have developed a new device for electrical stimulation of chondrocytes, which operates on the basis of capacitive coupling of alternating electric fields. The reusable and sterilizable stimulation device allows the simultaneous use of 12 cavities with independently applicable fields using only one main supply. The first parameter settings for the stimulation of human non-degenerative chondrocytes, seeded on collagen type I elastin-based scaffolds, were derived from numerical electric field simulations. Our first results suggest that applied alternating electric fields induce chondrogenic re-differentiation at the gene and especially at the protein level of human de-differentiated chondrocytes in a frequency-dependent manner. In future studies, further parameter optimizations will be performed to improve the differentiation capacity of human cartilage cells.
Collapse
Affiliation(s)
- Simone Krueger
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Alexander Riess
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
- Correspondence: (S.K.); (A.R.)
| | - Anika Jonitz-Heincke
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Alina Weizel
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Anika Seyfarth
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
| | - Hermann Seitz
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, 18051 Rostock, Germany;
| | - Rainer Bader
- Biomechanics and Implant Technology Research Laboratory, Department of Orthopedics, Rostock University Medical Center, 18057 Rostock, Germany; (A.J.-H.); (A.S.); (R.B.)
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany;
| |
Collapse
|
36
|
Otahal A, Kramer K, Kuten-Pella O, Weiss R, Stotter C, Lacza Z, Weber V, Nehrer S, De Luna A. Characterization and Chondroprotective Effects of Extracellular Vesicles From Plasma- and Serum-Based Autologous Blood-Derived Products for Osteoarthritis Therapy. Front Bioeng Biotechnol 2020; 8:584050. [PMID: 33102466 PMCID: PMC7546339 DOI: 10.3389/fbioe.2020.584050] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Autologous blood products gain increasing interest in the field of regenerative medicine as well as in orthopedics, aesthetic surgery, and cosmetics. Currently, citrate-anticoagulated platelet-rich plasma (CPRP) preparations are often applied in osteoarthritis (OA), but more physiological and cell-free alternatives such as hyperacute serum (hypACT) are under development. Besides growth factors, blood products also bring along extracellular vesicles (EVs) packed with signal molecules, which open up a new level of complexity at evaluating the functional spectrum of blood products. Large proportions of EVs originated from platelets in CPRP and hypACT, whereas very low erythrocyte and monocyte-derived EVs were detected via flow cytometry. EV treatment of chondrocytes enhanced the expression of anabolic markers type II collagen, SRY-box transcription factor 9 (SOX9), and aggrecan compared to full blood products, but also the catabolic marker and tissue remodeling factor matrix metalloproteinase 3, whereas hypACT EVs prevented type I collagen expression. CPRP blood product increased SOX9 protein expression, in contrast to hypACT blood product. However, hypACT EVs induced SOX9 protein expression while preventing interleukin-6 secretion. The results indicate that blood EVs are sufficient to induce chondrogenic gene expression changes in OA chondrocytes, while preventing proinflammatory cytokine release compared to full blood product. This highlights the potential of autologous blood-derived EVs as regulators of cartilage extracellular matrix metabolism and inflammation, as well as candidates for new cell-free therapeutic approaches for OA.
Collapse
Affiliation(s)
- Alexander Otahal
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Karina Kramer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Olga Kuten-Pella
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria.,OrthoSera GmbH, Krems an der Donau, Austria
| | - René Weiss
- Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| | - Christoph Stotter
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Zsombor Lacza
- Deptartment Sports Physiology, University of Physical Education, Budapest, Hungary
| | - Viktoria Weber
- Center for Biomedical Technology, Department for Biomedical Research, Danube University Krems, Krems an der Donau, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| | - Andrea De Luna
- Center for Regenerative Medicine, Department for Health Sciences, Medicine and Research, Danube University Krems, Krems an der Donau, Austria
| |
Collapse
|
37
|
Kisiday JD, Liebig BE, Goodrich LR. Adult ovine chondrocytes in expansion culture adopt progenitor cell properties that are favorable for cartilage tissue engineering. J Orthop Res 2020; 38:1996-2005. [PMID: 32222117 PMCID: PMC8442064 DOI: 10.1002/jor.24671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 02/18/2020] [Accepted: 03/06/2020] [Indexed: 02/04/2023]
Abstract
Human chondrocytes in expansion culture can become progenitor-like in their ability to proliferate extensively and secrete neocartilage in chondrogenic culture. Sheep are used as a large animal model for cartilage tissue engineering, although for testing progenitor-like chondrocytes it is important that ovine chondrocytes resemble human in the ability to adopt progenitor properties. Here, we investigate whether ovine chondrocytes can adopt progenitor properties as indicated by rapid proliferation in a colony-forming fashion, and high levels of neocartilage secretion in chondrogenic culture. In conditions known to promote expansion of mesenchymal stromal cells, ovine chondrocytes proliferated through approximately 12 population doublings in 10 days. Time-lapse imaging indicated rapid proliferation in a colony-forming pattern. Expanded ovine chondrocytes that were seeded into agarose and cultured in chondrogenic medium accumulated neocartilage over 2 weeks, to a greater extent than primary chondrocytes. These data confirm that ovine chondrocytes resemble human chondrocytes in their ability to acquire progenitor properties that are important for cartilage tissue engineering. Given the broad interest in using progenitor cells to heal connective tissues, next we compared proliferation and trilineage differentiation of ovine chondrocytes, meniscus cells, and tenocytes. Meniscus cells and tenocytes experienced more than 13 population doublings in 10 days. In chondrogenic culture, cartilage matrix accumulation, and gene expression were largely similar among the cell types. All cell types resisted osteogenesis, while expanded tenocytes and meniscal cells were capable of adipogenesis. While ovine connective tissue cells demonstrated limited lineage plasticity, these data support the potential to promote certain progenitor properties with expansion.
Collapse
Affiliation(s)
- John D. Kisiday
- Department of Clinical Sciences, Orthopaedic Reserch CenterC. Wayne McIlwraith Translational Medicine Institute Fort Collins Colorado
| | - Bethany E. Liebig
- Department of Clinical Sciences, Orthopaedic Reserch CenterC. Wayne McIlwraith Translational Medicine Institute Fort Collins Colorado
| | - Laurie R. Goodrich
- Department of Clinical Sciences, Orthopaedic Reserch CenterC. Wayne McIlwraith Translational Medicine Institute Fort Collins Colorado
| |
Collapse
|
38
|
Influence of Conditioned Media on the Re-Differentiation Capacity of Human Chondrocytes in 3D Spheroid Cultures. J Clin Med 2020; 9:jcm9092798. [PMID: 32872610 PMCID: PMC7564315 DOI: 10.3390/jcm9092798] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/20/2020] [Accepted: 08/28/2020] [Indexed: 12/27/2022] Open
Abstract
A major challenge of cell-based therapy for cartilage lesions is the preservation of the chondrogenic phenotype during ex vivo cell cultivation. In this in vitro study, the chondro-inductive capacity of two different hyaline cartilage-conditioned cell culture media on human chondrocytes in 3D spheroids was determined. Media were conditioned by incubation of 200 mg/mL vital or devitalized cartilage matrix in growth media over 35 days. The media were analyzed for the content of soluble procollagen type (Col) II and glycosaminoglycans (GAGs) as well as released TGF-β1, IGF-1 and IGFBP3. Unconditioned medium served as a negative control while the positive medium control was supplemented with TGF-β1 and IGF-1. Spheroid cultures prepared from human chondrocytes were cultivated at 37 °C, 5% CO2 and 21% O2 in the respective media and controls. After 14 and 35 days, the deposition of ECM components was evaluated by histological analysis. Vital cartilage-conditioned medium contained significantly higher levels of Col II and active TGF-β1 compared to medium conditioned with the devitalized cartilage matrix. Despite these differences, the incubation with vital as well as devitalized cartilage conditioned medium led to similar results in terms of deposition of proteoglycans and collagen type II, which was used as an indicator of re-differentiation of human chondrocytes in spheroid cultures. However, high density 3D cell cultivation showed a positive influence on re-differentiation.
Collapse
|
39
|
Bourebaba L, Röcken M, Marycz K. Osteochondritis dissecans (OCD) in Horses - Molecular Background of its Pathogenesis and Perspectives for Progenitor Stem Cell Therapy. Stem Cell Rev Rep 2020; 15:374-390. [PMID: 30796679 PMCID: PMC6534522 DOI: 10.1007/s12015-019-09875-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Osteochondrosis (osteochondrosis dissecans; OCD) is a disease syndrome of growing cartilage related to different clinical entities such as epiphysitis, subchondral cysts and angular carpal deformities, which occurs in growing animals of all species, including horses. Nowadays, these disorders are affecting increasing numbers of young horses worldwide. As a complex multifactorial disease, OCD is initiated when failure in cartilage canals because of existing ischemia, chondrocyte biogenesis impairment as well as biochemical and genetic disruptions occur. Recently, particular attention have been accorded to the definition of possible relations between OCD and some metabolic disorders; in this way, implication of mitochondrial dysfunctions, endoplasmic reticulum disruptions, oxidative stress or endocrinological affections are among the most considered axes for future researches. As one of the most frequent cause of impaired orthopaedic potential, which may result in a sharp decrease in athletic performances of the affected animals, and lead to the occurrence of complications such as joint fragility and laminitis, OCD remains as one of the primary causes of considerable economic losses in all sections of the equine industry. It would therefore be important to provide more information on the exact pathophysiological mechanism(s) underlying early OC(D) lesions, in order to implement innovative strategies involving the use of progenitor stem cells, which are considered nowadays as a promising approach to regenerative medicine, with the potential to treat numerous orthopaedic disorders, including osteo-degenerative diseases, for prevention and reduction of incidence of the disease, not only in horses, but also in human medicine, as the equine model is already widely accepted by the scientific community and approved by the FDA, for the research and application of cellular therapies in the treatment of human conditions.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Norwida 27B, 50-375, Wrocław, Poland. .,Faculty of Veterinary Medicine, Equine Clinic - Equine Surgery, Justus-Liebig-University, 35392, Gießen, Germany.
| |
Collapse
|
40
|
Nakayama N, Pothiawala A, Lee JY, Matthias N, Umeda K, Ang BK, Huard J, Huang Y, Sun D. Human pluripotent stem cell-derived chondroprogenitors for cartilage tissue engineering. Cell Mol Life Sci 2020; 77:2543-2563. [PMID: 31915836 PMCID: PMC11104892 DOI: 10.1007/s00018-019-03445-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/24/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
The cartilage of joints, such as meniscus and articular cartilage, is normally long lasting (i.e., permanent). However, once damaged, especially in large animals and humans, joint cartilage is not spontaneously repaired. Compensating the lack of repair activity by supplying cartilage-(re)forming cells, such as chondrocytes or mesenchymal stromal cells, or by transplanting a piece of normal cartilage, has been the basis of therapy for biological restoration of damaged joint cartilage. Unfortunately, current biological therapies face problems on a number of fronts. The joint cartilage is generated de novo from a specialized cell type, termed a 'joint progenitor' or 'interzone cell' during embryogenesis. Therefore, embryonic chondroprogenitors that mimic the property of joint progenitors might be the best type of cell for regenerating joint cartilage in the adult. Pluripotent stem cells (PSCs) are expected to differentiate in culture into any somatic cell type through processes that mimic embryogenesis, making human (h)PSCs a promising source of embryonic chondroprogenitors. The major research goals toward the clinical application of PSCs in joint cartilage regeneration are to (1) efficiently generate lineage-specific chondroprogenitors from hPSCs, (2) expand the chondroprogenitors to the number needed for therapy without loss of their chondrogenic activity, and (3) direct the in vivo or in vitro differentiation of the chondroprogenitors to articular or meniscal (i.e., permanent) chondrocytes rather than growth plate (i.e., transient) chondrocytes. This review is aimed at providing the current state of research toward meeting these goals. We also include our recent achievement of successful generation of "permanent-like" cartilage from long-term expandable, hPSC-derived ectomesenchymal chondroprogenitors.
Collapse
Affiliation(s)
- Naoki Nakayama
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA.
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA.
| | - Azim Pothiawala
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - John Y Lee
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Nadine Matthias
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
| | - Katsutsugu Umeda
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Department of Pediatrics, Kyoto University School of Medicine, Kyoto, Japan
| | - Bryan K Ang
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston Medical School, 1825 Pressler St., Houston, TX, 77030, USA
- Weil Cornell Medicine, New York, NY, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, The University of Texas Health Science Center at Houston Medical School, Houston, TX, USA
- Steadman Philippon Research Institute, Vail, CO, USA
| | - Yun Huang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| | - Deqiang Sun
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX, USA
| |
Collapse
|
41
|
Lehoczky G, Wolf F, Mumme M, Gehmert S, Miot S, Haug M, Jakob M, Martin I, Barbero A. Intra-individual comparison of human nasal chondrocytes and debrided knee chondrocytes: Relevance for engineering autologous cartilage grafts. Clin Hemorheol Microcirc 2020; 74:67-78. [PMID: 31743993 DOI: 10.3233/ch-199236] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Implantation of autologous chondrocytes for cartilage repair requires harvesting of undamaged cartilage, implying an additional joint arthroscopy surgery and further damage to the articular surface. As alternative possible cell sources, in this study we assessed the proliferation and chondrogenic capacity of debrided Knee Chondrocytes (dKC) and Nasal Chondrocytes (NC) collected from the same patients. METHODS Matched NC and dKC pairs from 13 patients enrolled in two clinical studies (NCT01605201 and NCT026739059) were expanded in monolayer and then chondro-differentiated in 3D collagenous scaffolds in medium with or without Transforming Growth Factor beta 1 (TGFβ1). Cell proliferation and amount of cartilage matrix production by these two cell types were assessed. RESULTS dKC exhibited an inferior proliferation rate than NC, and a lower capacity to chondro-differentiate. Resulting dKC-grafts contained lower amounts of cartilage specific matrix components glycosaminoglycans and type II collagen. The cartilage forming capacity of dKC did not significantly correlate with specific clinical parameters and was only partially improved by medium supplemention with TGFβ1. CONCLUSIONS dKC exhibit a reproducibly poor capacity to engineer cartilage grafts. Our in vitro data suggest that NC would be a better suitable cell source for the generation of autologous cartilage grafts.
Collapse
Affiliation(s)
- Gyözö Lehoczky
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Francine Wolf
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marcus Mumme
- Department of Surgery, University Hospital of Basel, Basel, Switzerland.,Department of Orthopaedics, University Children's Hospital Basel, Basel, Switzerland
| | - Sebastian Gehmert
- Department of Orthopaedics, University Children's Hospital Basel, Basel, Switzerland
| | - Sylvie Miot
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Haug
- Department of Surgery, University Hospital of Basel, Basel, Switzerland
| | - Marcel Jakob
- Department of Surgery, University Hospital of Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
42
|
Bi R, Yin Q, Mei J, Chen K, Luo X, Fan Y, Zhu S. Identification of human temporomandibular joint fibrocartilage stem cells with distinct chondrogenic capacity. Osteoarthritis Cartilage 2020; 28:842-852. [PMID: 32147536 DOI: 10.1016/j.joca.2020.02.835] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/08/2020] [Accepted: 02/26/2020] [Indexed: 02/05/2023]
Abstract
OBJECTIVE This study was aimed to identify the residence of human fibrocartilage stem cells (hFCSCs), characterize their stem cell properties and investigate the functional mechanisms which regulate fibrocartilage stem cells (FCSCs) toward chondrogenic differentiation during cartilage homeostasis and repairing. METHODS Cytological characteristics of hFCSCs and human orofacial mesenchymal stem cells (hOFMSCs) were analyzed. Chondrogenic potential of hFCSCs was compared with hOFMSCs both in vitro and in vivo. Regulatory role of SOX9 during FCSCs chondrogenesis was studied by shRNA interference in vitro, and by GFP+ FCSCs treatment in rat condylar cartilage defect model. SOX9 expression was also examined in temporomandibular joint osteoarthritis (TMJOA) patients' cartilage surface. RESULTS hFCSCs exhibited typical mesenchymal stem cell characteristics, with significantly stronger chondrogenic capability compared to hOFMSCs. Moreover, hFCSCs showed remarkably increased expression of SOX9. During cartilage pellet culture, there was stronger SOX9 expression in hFCSCs than hOFMSCs. SOX9 shRNA interference downregulated chondrogenic capability of hFCSCs in vitro, as well as disrupting migration and chondrogenic differentiation of GFP+ FCSCs toward mature chondrocytes in rat condylar cartilage defect. Of note, SOX9 expression was also found suppressed in the condylar superficial zone of TMJOA patients. CONCLUSION We found the existence of FCSCs in human TMJ cartilage, and characterized their distinct stem cell features. SOX9 is essential for hFCSCs chondrogenic differentiation, and a comprehensive understanding of the regulatory role of SOX9 in hFCSCs would be important for exploring potential intervention strategy of condylar cartilage degradation during TMJ disorders.
Collapse
Affiliation(s)
- R Bi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Q Yin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - J Mei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - K Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - X Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Y Fan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - S Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthognathic and TMJ Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
43
|
Grandi FC, Baskar R, Smeriglio P, Murkherjee S, Indelli PF, Amanatullah DF, Goodman S, Chu C, Bendall S, Bhutani N. Single-cell mass cytometry reveals cross-talk between inflammation-dampening and inflammation-amplifying cells in osteoarthritic cartilage. SCIENCE ADVANCES 2020; 6:eaay5352. [PMID: 32201724 PMCID: PMC7069698 DOI: 10.1126/sciadv.aay5352] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/17/2019] [Indexed: 05/06/2023]
Abstract
Aging or injury leads to degradation of the cartilage matrix and the development of osteoarthritis (OA). Because of a paucity of single-cell studies of OA cartilage, little is known about the interpatient variability in its cellular composition and, more importantly, about the cell subpopulations that drive the disease. Here, we profiled healthy and OA cartilage samples using mass cytometry to establish a single-cell atlas, revealing distinct chondrocyte progenitor and inflammation-modulating subpopulations. These rare populations include an inflammation-amplifying (Inf-A) population, marked by interleukin-1 receptor 1 and tumor necrosis factor receptor II, whose inhibition decreased inflammation, and an inflammation-dampening (Inf-D) population, marked by CD24, which is resistant to inflammation. We devised a pharmacological strategy targeting Inf-A and Inf-D cells that significantly decreased inflammation in OA chondrocytes. Using our atlas, we stratified patients with OA in three groups that are distinguished by the relative proportions of inflammatory to regenerative cells, making it possible to devise precision therapeutic approaches.
Collapse
Affiliation(s)
- Fiorella Carla Grandi
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Reema Baskar
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Piera Smeriglio
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Shravani Murkherjee
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | | | - Derek F. Amanatullah
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Stuart Goodman
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Constance Chu
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
- Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304, USA
| | - Sean Bendall
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA 94303, USA
| | - Nidhi Bhutani
- Department of Orthopedic Surgery, School of Medicine, Stanford University, Stanford, CA 94303, USA
- Corresponding author.
| |
Collapse
|
44
|
D'Costa S, Rich MJ, Diekman BO. Engineered Cartilage from Human Chondrocytes with Homozygous Knockout of Cell Cycle Inhibitor p21. Tissue Eng Part A 2019; 26:441-449. [PMID: 31642391 DOI: 10.1089/ten.tea.2019.0214] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Osteoarthritis (OA) is a highly prevalent disease with limited treatment options. The search for disease-modifying OA therapies would benefit from a more comprehensive knowledge of the genetic variants that contribute to chondrocyte dysfunction and the barriers to cartilage regeneration. One goal of this study was to establish a system for producing engineered cartilage tissue from genetically defined primary human chondrocytes through genome editing and single-cell expansion. This process was utilized to investigate the functional effect of biallelic knockout of the cell cycle inhibitor p21. The use of ribonucleoprotein (RNP) CRISPR/Cas9 complexes targeting two sites in the coding region of p21 resulted in a high frequency (16%) of colonies with homozygous p21 knockout. Chondrogenic pellet cultures from expanded chondrocytes with complete loss of p21 produced more glycosaminoglycans (GAG) and maintained a higher cell number. Single-cell-derived colonies retained the potential for robust matrix production after expansion, allowing for analysis of colony variability from the same population of targeted cells. The effect of enhanced cartilage matrix production in p21 knockout chondrocytes persisted when matrix production from individual colonies was analyzed. Chondrocytes had lower levels of p21 protein with further expansion, and the difference in GAG production with p21 knockout was strongest at early passages. These results support previous findings that implicate p21 as a barrier to cartilage matrix production and regenerative capacity. Furthermore, this work establishes the use of genome-edited human chondrocytes as a promising approach for engineered tissue models containing user-defined gene knockouts and other genetic variants for investigation of OA pathogenesis. Impact Statement This work provides two important advances to the field of tissue engineering. One is the demonstration that engineered cartilage tissue can be produced from genetically defined populations of primary human chondrocytes. While CRISPR/Cas-9 genome editing has been extensively used in cell lines that divide indefinitely, this work extends the technique to an engineered tissue model system to support investigation of genetic changes that affect cartilage production. A second contribution is the finding that chondrocytes with p21 knockout synthesized more cartilage matrix tissue than unedited controls. This supports the continued investigation of p21 as a potential barrier to effective cartilage regeneration.
Collapse
Affiliation(s)
- Susan D'Costa
- Thurston Arthritis Research Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Matthew J Rich
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina and North Carolina State University, Raleigh, North Carolina
| | - Brian O Diekman
- Thurston Arthritis Research Center, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina and North Carolina State University, Raleigh, North Carolina.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
45
|
Charlier E, Deroyer C, Ciregia F, Malaise O, Neuville S, Plener Z, Malaise M, de Seny D. Chondrocyte dedifferentiation and osteoarthritis (OA). Biochem Pharmacol 2019; 165:49-65. [PMID: 30853397 DOI: 10.1016/j.bcp.2019.02.036] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 02/28/2019] [Indexed: 02/08/2023]
|
46
|
Zheng H, Liu J, Tycksen E, Nunley R, McAlinden A. MicroRNA-181a/b-1 over-expression enhances osteogenesis by modulating PTEN/PI3K/AKT signaling and mitochondrial metabolism. Bone 2019; 123:92-102. [PMID: 30898695 PMCID: PMC6491221 DOI: 10.1016/j.bone.2019.03.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/13/2019] [Accepted: 03/16/2019] [Indexed: 12/14/2022]
Abstract
MicroRNAs are small non-coding RNAs that play important roles in many cellular processes including proliferation, metabolism and differentiation. They function by binding to specific regions within the 3'UTR of target mRNAs resulting in suppression of protein synthesis and modulation of potentially many cellular pathways. We previously showed that miRNA expression levels differed between cells from distinct regions of developing human embryonic long bones. Specifically, we found that miR-181a-1 was significantly more highly expressed in hypertrophic chondrocytes compared to proliferating differentiated or progenitor chondrocytes, suggesting a potential role in regulating chondrocyte hypertrophy and/or endochondral bone formation. The goal of this study was to determine how miR-181a-1 together with its clustered miRNA, miR-181b-1, regulates osteogenesis. We show that over-expression of the miR-181a/b-1 cluster enhanced osteogenesis and that cellular pathways associated with protein synthesis and mitochondrial metabolism were significantly up-regulated. Metabolic assays revealed that the oxygen consumption rate and ATP-linked respiration were increased by miR-181a/b-1. To further decipher a potential mechanism causing these metabolic changes, we showed that PTEN (phosphatase and tensin homolog) levels were suppressed following miR-181a/b-1 over-expression, and that PI3K/AKT signaling was subsequently increased. Over-expression of PTEN was found to attenuate the enhancing effects of miR-181a/b-1, providing further evidence that miR-181a/b-1 regulates the PTEN/PI3K/AKT axis to enhance osteogenic differentiation and mitochondrial metabolism. These findings have important implications for the design of miR-181a/b targeting strategies to treat bone conditions such as fractures or heterotopic ossification.
Collapse
Affiliation(s)
- Hongjun Zheng
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Jin Liu
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Eric Tycksen
- Genome Technology Access Center, Washington University School of Medicine, St Louis, MO, United States of America.
| | - Ryan Nunley
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America.
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, United States of America; Department of Cell Biology, Washington University School of Medicine, St. Louis, MO, United States of America; Shriners Hospital for Children - St Louis, St Louis, MO, United States of America.
| |
Collapse
|
47
|
Si H, Liang M, Cheng J, Shen B. [Effects of cartilage progenitor cells and microRNA-140 on repair of osteoarthritic cartilage injury]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2019; 33:650-658. [PMID: 31090363 PMCID: PMC8337193 DOI: 10.7507/1002-1892.201806060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 03/12/2019] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To summarize the effect of cartilage progenitor cells (CPCs) and microRNA-140 (miR-140) on the repair of osteoarthritic cartilage injury, and analyze their clinical prospects. METHODS The recent researches regarding the CPCs, miR-140, and repair of cartilage in osteoarthritis (OA) disease were extensively reviewed and summarized. RESULTS CPCs possess the characteristics of self-proliferation, expression of stem cell markers, and multi-lineage differentiation potential, and their chondrogenic ability is superior to other tissues-derived mesenchymal stem cells. CPCs are closely related to the development of OA, but the autonomic activation and chondrogenic ability of CPCs around the osteoarthritic cartilage lesion cannot meet the requirements of complete cartilage repair. miR-140 specifically express in cartilage, and has the potential to activate CPCs by inhibiting key molecules of Notch signaling pathway and enhance its chondrogenic ability, thus promoting the repair of osteoarthritic cartilage injury. Intra-articular delivery of drugs is one of the main methods of OA treatment, although intra-articular injection of miR-140 has a significant inhibitory effect on cartilage degeneration in rats, it also exhibit some limitations such as non-targeted aggregation, low bioavailability, and rapid clearance. So it is a good application prospect to construct a carrier with good safety, cartilage targeting, and high-efficiency for miR-140 based on articular cartilage characteristics. In addition, CPCs are mainly dispersed in the cartilage surface, while OA cartilage injury also begins from this layer, it is therefore essential to emphasize early intervention of OA. CONCLUSION miR-140 has the potential to activate CPCs and promote the repair of cartilage injury in early OA, and it is of great clinical significance to further explore the role of miR-140 in OA etiology and to develop new OA treatment strategies based on miR-140.
Collapse
Affiliation(s)
- Haibo Si
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Mingwei Liang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Jingqiu Cheng
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P.R.China
| | - Bin Shen
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu Sichuan, 610041,
| |
Collapse
|
48
|
Human Diseased Articular Cartilage Contains a Mesenchymal Stem Cell-Like Population of Chondroprogenitors with Strong Immunomodulatory Responses. J Clin Med 2019; 8:jcm8040423. [PMID: 30925656 PMCID: PMC6517884 DOI: 10.3390/jcm8040423] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023] Open
Abstract
Background: osteoarthritic human articular cartilage (AC)-derived cartilage cells (CCs) with same-donor bone marrow (BMSCs) and adipose tissue (ASCs)-derived mesenchymal stem cells were compared, in terms of stemness features, and secretory and immunomodulatory responses to inflammation. Methods: proteoglycan 4 (PRG4) presence was evaluated in AC and CCs. MSCs and CCs (n = 8) were cultured (P1 to P4) and characterized for clonogenicity, nanog homeobox (NANOG), and POU class 5 homeobox 1 (POU5F1) expression, immunotypification, and tri-lineage differentiation. Their basal and interleukin-1β (IL-1β)-stimulated expression of matrix metalloproteases (MMPs), tissue inhibitors (TIMPs), release of growth factors, and cytokines were analyzed, along with the immunomodulatory ability of CCs. Results: PRG4 was mainly expressed in the intact AC surface, whereas shifted to the intermediate zone in damaged cartilage and increased its expression in CCs upon culture. All cells exhibited a similar phenotype and stemness maintenance over passages. CCs showed highest chondrogenic ability, no adipogenic potential, a superior basal secretion of growth factors and cytokines, the latter further increased after inflammatory stimulation, and an immunomodulatory behavior. All stimulated cells shared an increased MMP expression without a corresponding TIMP production. Conclusion: based on the observed features, CCs obtained from pathological joints may constitute a potential tissue-specific therapeutic target or agent to improve damaged cartilage healing, especially damage caused by inflammatory/immune mediated conditions.
Collapse
|
49
|
Haeni DL, Lafosse T, Haggerty C, Plath J, Kida Y, Sanchez-Brass M, Wolf F, Calvo E, Müller AM, Barbero A, Lafosse L. Tissue on the Transferred Coracoid Graft After Latarjet Procedure: Histological and Morphological Findings. Am J Sports Med 2019; 47:704-712. [PMID: 30716278 DOI: 10.1177/0363546518819825] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Anterior shoulder instability is a debilitating condition that can require stabilization via a Latarjet procedure. PURPOSE The aim of this study was to characterize the histological composition of the articular-sided surface of the coracoid bone graft after Latarjet procedure. Specific features of cells isolated from the coracoid and graft tissues were assessed. STUDY DESIGN Case series; Level of evidence, 4. METHODS Tissue samples were harvested from 9 consecutive patients undergoing arthroscopic debridement and screw removal after arthroscopic or open Latarjet procedure. Tissues were processed histologically. In 2 patients, the samples were analyzed to assess specific cellular properties. RESULTS Safranin O staining indicated that glenoid tissues contained variable amounts of glycosaminoglycan (GAG) and round chondrocytic cells mainly organized in clusters. Graft tissues contained less GAG and were more cellular but were not organized in clusters and had variable morphological features. An association appeared to exist between the cartilage quality of glenoid tissues and that of the graft tissues. Cells isolated from glenoid and graft tissues exhibited similar proliferation capacity. CONCLUSION The results of our analysis show that cells located at the articular-sided surface of transferred coracoid grafts demonstrate fibrocartilaginous properties and may have the capacity for chondral proliferation. Further studies are needed to confirm this observation and future application.
Collapse
Affiliation(s)
- David L Haeni
- Orthopaedic Department, Children's Hospital, University of Basel, Basel, Switzerland
| | | | | | | | - Yoshikazu Kida
- Pioneer Peek Orthopedics, Palmer, Alaska, USA.,Department of Orthopaedics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | - Françine Wolf
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Emilio Calvo
- Fundacion Jimenez Diaz, Universidad Autonoma de Madrid, Madrid, Spain
| | - Andreas M Müller
- Orthopaedic Department, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andrea Barbero
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | | |
Collapse
|
50
|
Islam A, Fossum V, Hansen AK, Urbarova I, Knutsen G, Martinez-Zubiaurre I. In vitro chondrogenic potency of surplus chondrocytes from autologous transplantation procedures does not predict short-term clinical outcomes. BMC Musculoskelet Disord 2019; 20:19. [PMID: 30630436 PMCID: PMC6329094 DOI: 10.1186/s12891-018-2380-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 12/12/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Autologous chondrocyte implantation (ACI) has been used over the last two decades to treat focal cartilage lesions aiming to delay or prevent the onset of osteoarthritis; however, some patients do not respond adequately to the procedure. A number of biomarkers that can forecast the clinical potency of the cells have been proposed, but evidence for the relationship between in vitro chondrogenic potential and clinical outcomes is missing. In this study, we explored if the ability of cells to make cartilage in vitro correlates with ACI clinical outcomes. Additionally, we evaluated previously proposed chondrogenic biomarkers and searched for new biomarkers in the chondrocyte proteome capable of predicting clinical success or failure after ACI. METHODS The chondrogenic capacity of chondrocytes derived from 14 different donors was defined based on proteoglycans staining and visual histological grading of tissues generated using the pellet culture system. A Lysholm score of 65 two years post-ACI was used as a cut-off to categorise "success" and "failure" clinical groups. A set of predefined biomarkers were investigated in the chondrogenic and clinical outcomes groups using flow cytometry and qPCR. High-throughput proteomics of cell lysates was used to search for putative biomarkers to predict chondrogenesis and clinical outcomes. RESULTS Visual histological grading of pellets categorised donors into "high" and "low" chondrogenic groups. Direct comparison between donor-matched in vitro chondrogenic potential and clinical outcomes revealed no significant associations. Comparative analyses of selected biomarkers revealed that expression of CD106 and TGF-β-receptor-3 was enhanced in the low chondrogenic group, while expression of integrin-α1 and integrin-β1 was significantly upregulated in the high chondrogenic group. Additionally, increased surface expression of CD166 was observed in the clinical success group, while the gene expression of cartilage oligomeric matrix protein was downregulated. High throughput proteomics revealed no differentially expressed proteins from success and failure clinical groups, whereas seven proteins including prolyl-4-hydroxylase 1 were differentially expressed when comparing chondrogenic groups. CONCLUSION In our limited material, we found no correlation between in vitro cartilage-forming capacity and clinical outcomes, and argue on the limitations of using the chondrogenic potential of cells or markers for chondrogenesis as predictors of clinical outcomes.
Collapse
Affiliation(s)
- Ashraful Islam
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.
| | - Vegard Fossum
- Department of Orthopaedic Surgery, University Hospital of Northern Norway, Tromsø, Norway
| | - Ann Kristin Hansen
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway.,Department of Orthopaedic Surgery, University Hospital of Northern Norway, Tromsø, Norway
| | - Ilona Urbarova
- Department of Medical Biology, Tromsø University Proteomics Platform, UiT The Arctic University of Norway, Tromsø, Norway
| | - Gunnar Knutsen
- Department of Orthopaedic Surgery, University Hospital of Northern Norway, Tromsø, Norway
| | | |
Collapse
|