1
|
Jin B, Su G, Zhou X, Xu L, Wang W, Zhou T, Tan Y, Wang S, Li G. Basic Fibroblast Growth Factor Supports the Function of Limbal Niche Cells via the Wnt/β-Catenin Pathway. J Ocul Pharmacol Ther 2024. [PMID: 39083404 DOI: 10.1089/jop.2024.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Purpose: To test the effects and underlying mechanisms of basic fibroblast growth factor (bFGF) on the limbal niche cell (LNC) function ex vivo. Methods: By using different concentrations of bFGF (0, 4, 8, 12, and 16 ng/mL) and fibroblast growth factor receptor (FGFR) inhibitors, the effects of bFGF on LNC proliferation, expression of stem cell markers, and transcription levels of the β-catenin were investigated. Single-cell RNA sequencing (scRNA-seq) was used to analyze the action and mechanisms of FGFR subtypes and the Wnt/β-catenin pathway during LNC culture. An mature corneal epithelial cell (MCEC)/LNC three-dimensional model was constructed to verify whether bFGF activates the Wnt/β-catenin pathway in LNC by inhibiting FGFR or β-catenin targets. Results: scRNA-seq showed that FGFR1 is the main receptor in LNC, along with the molecules in the Wnt pathway, including WNT2, FZD7, LRP5, LRP6, and β-catenin. The 12 ng/mL bFGF treatment group showed higher LNC proliferation rate and transcription levels of OCT4, SOX2, NANOG, and β-catenin than any other groups (P < 0.001). In the MCEC/LNC co-culture model, MCEC/LNC treated with 12 ng/mL bFGF promoted the aggregation of the spheres than other groups, associated with increased transcription levels of P63α, WNT2, β-catenin, and a decreased transcription level of CK12 (P < 0.001). Wnt/β-catenin inhibitor LF3 treatment reversed the abovementioned effect of bFGF. Conclusions: bFGF could maintain and promote the stemness of LNC via the FGFR1/Wnt2/FZD7/LRP6 axis in a concentration-dependent manner.
Collapse
Affiliation(s)
- Bihui Jin
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanyu Su
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongyao Tan
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusheng Wang
- Department of Cell and Molecular Biology & Ophthalmology, Tulane University, New Orleans, Louisiana, USA
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Burdis R, Gallostra XB, Kelly DJ. Temporal Enzymatic Treatment to Enhance the Remodeling of Multiple Cartilage Microtissues into a Structurally Organized Tissue. Adv Healthc Mater 2024; 13:e2300174. [PMID: 37858935 PMCID: PMC11468768 DOI: 10.1002/adhm.202300174] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 08/29/2023] [Indexed: 10/21/2023]
Abstract
Scaffold-free tissue engineering aims to recapitulate key aspects of normal developmental processes to generate biomimetic grafts. Although functional cartilaginous tissues are engineered using such approaches, considerable challenges remain. Herein, the benefits of engineering cartilage via the fusion of multiple cartilage microtissues compared to using (millions of) individual cells to generate a cartilaginous graft are demonstrated. Key advantages include the generation of a richer extracellular matrix, more hyaline-like cartilage phenotype, and superior shape fidelity. A major drawback of aggregate engineering is that individual microtissues do not completely (re)model and remnants of their initial architectures remain throughout the macrotissue. To address this, a temporal enzymatic (chondroitinase-ABC) treatment is implemented to accelerate structural (re)modeling and shown to support robust fusion between adjacent microtissues, enhance microtissue (re)modeling, and enable the development of a more biomimetic tissue with a zonally organized collagen network. Additionally, enzymatic treatment is shown to modulate matrix composition, tissue phenotype, and to a lesser extent, tissue mechanics. This work demonstrates that microtissue self-organization is an effective method for engineering scaled-up cartilage grafts with a predefined geometry and near-native levels of matrix accumulation. Importantly, key limitations associated with using biological building blocks can be alleviated by temporal enzymatic treatment during graft development.
Collapse
Affiliation(s)
- Ross Burdis
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
| | - Xavier Barceló Gallostra
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
| | - Daniel J. Kelly
- Trinity Centre for Biomedical EngineeringTrinity Biomedical Sciences InstituteTrinity College DublinDublinD02 PN40Ireland
- Department of MechanicalManufacturing and Biomedical EngineeringSchool of EngineeringTrinity College DublinDublinD02 PN40Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College DublinDublinD02 PN40Ireland
- Department of Anatomy and Regenerative MedicineRoyal College of Surgeons in IrelandDublinD02 YN77Ireland
| |
Collapse
|
3
|
Ohnishi T, Homan K, Fukushima A, Ukeba D, Iwasaki N, Sudo H. A Review: Methodologies to Promote the Differentiation of Mesenchymal Stem Cells for the Regeneration of Intervertebral Disc Cells Following Intervertebral Disc Degeneration. Cells 2023; 12:2161. [PMID: 37681893 PMCID: PMC10486900 DOI: 10.3390/cells12172161] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/24/2023] [Accepted: 08/26/2023] [Indexed: 09/09/2023] Open
Abstract
Intervertebral disc (IVD) degeneration (IDD), a highly prevalent pathological condition worldwide, is widely associated with back pain. Treatments available compensate for the impaired function of the degenerated IVD but typically have incomplete resolutions because of their adverse complications. Therefore, fundamental regenerative treatments need exploration. Mesenchymal stem cell (MSC) therapy has been recognized as a mainstream research objective by the World Health Organization and was consequently studied by various research groups. Implanted MSCs exert anti-inflammatory, anti-apoptotic, and anti-pyroptotic effects and promote extracellular component production, as well as differentiation into IVD cells themselves. Hence, the ultimate goal of MSC therapy is to recover IVD cells and consequently regenerate the extracellular matrix of degenerated IVDs. Notably, in addition to MSC implantation, healthy nucleus pulposus (NP) cells (NPCs) have been implanted to regenerate NP, which is currently undergoing clinical trials. NPC-derived exosomes have been investigated for their ability to differentiate MSCs from NPC-like phenotypes. A stable and economical source of IVD cells may include allogeneic MSCs from the cell bank for differentiation into IVD cells. Therefore, multiple alternative therapeutic options should be considered if a refined protocol for the differentiation of MSCs into IVD cells is established. In this study, we comprehensively reviewed the molecules, scaffolds, and environmental factors that facilitate the differentiation of MSCs into IVD cells for regenerative therapies for IDD.
Collapse
Affiliation(s)
- Takashi Ohnishi
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Kentaro Homan
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Akira Fukushima
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Daisuke Ukeba
- Department of Orthopedic Surgery, Hokkaido University Hospital, Sapporo 060-8648, Japan;
| | - Norimasa Iwasaki
- Department of Orthopedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan; (T.O.); (K.H.); (A.F.); (N.I.)
| | - Hideki Sudo
- Department of Advanced Medicine for Spine and Spinal Cord Disorders, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
4
|
Burdis R, Kronemberger GS, Kelly DJ. Engineering High-Quality Cartilage Microtissues Using Hydrocortisone Functionalized Microwells. Tissue Eng Part C Methods 2023; 29:121-133. [PMID: 36719783 DOI: 10.1089/ten.tec.2022.0181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Engineering clinically relevant musculoskeletal tissues at a human scale is a considerable challenge. Developmentally inspired scaffold-free approaches for engineering cartilage tissues have shown great promise in recent years, enabling the generation of highly biomimetic tissues. Despite the relative success of these approaches, the absence of a supporting scaffold or hydrogel creates challenges in the development of large-scale tissues. Combining numerous scaled-down tissue units (herein termed microtissues) into a larger macrotissue represents a promising strategy to address this challenge. The overall success of such approaches, however, relies on the development of strategies which support the robust and consistent chondrogenic differentiation of clinically relevant cell sources such as mesenchymal stem/stromal cells (MSCs) within microwell arrays to biofabricate numerous microtissues rich in cartilage-specific extracellular matrix components. In this article, we first describe a simple method to manufacture cartilage microtissues at various scales using novel microwell array stamps. This system allows the rapid and reliable generation of cartilage microtissues and can be used as a platform to study microtissue phenotype and development. Based on the unexpected discovery that Endothelial Growth Medium (EGM) enhanced MSC aggregation and chondrogenic capacity within the microwell arrays, this work also sought to identify soluble factors within the media capable of supporting robust differentiation using heterogeneous MSC populations. Hydrocortisone was found to be the key factor within EGM that enhanced the chondrogenic capacity of MSCs within these microwell arrays. This strategy represents a promising means of generating large numbers of high-quality, scaffold-free cartilage microtissues for diverse biofabrication applications. Impact statement This study addresses a key challenge facing emerging modular biofabrication strategies that use microtissues as biological building blocks. Namely, achieving the necessary robust and consistent differentiation of clinically relevant cell sources, for example, mesenchymal stem/stromal cells (MSCs), and the accumulation of sufficient tissue-specific extracellular matrix (ECM) to engineer tissue of scale. We achieved this by establishing hydrocortisone as a simple and potent method for improving MSC chondrogenesis, resulting in the biofabrication of high-quality (ECM rich) cartilage microtissues. These findings could enable the generation of more scalable engineered cartilage by ensuring the formation of high-quality microtissue building blocks generated using heterogeneous MSC populations.
Collapse
Affiliation(s)
- Ross Burdis
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Gabriela S Kronemberger
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin, Ireland.,Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
5
|
Schol J, Sakai D, Warita T, Nukaga T, Sako K, Wangler S, Tamagawa S, Zeiter S, Alini M, Grad S. Homing of vertebral-delivered mesenchymal stromal cells for degenerative intervertebral discs repair - an in vivo proof-of-concept study. JOR Spine 2023; 6:e1228. [PMID: 36994461 PMCID: PMC10041374 DOI: 10.1002/jsp2.1228] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/04/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
Introduction Cell transplantation shows promising results for intervertebral disc (IVD) repair, however, contemporary strategies present concerns regarding needle puncture damage, cell retention, and straining the limited nutrient availability. Mesenchymal stromal cell (MSC) homing is a natural mechanism of long-distance cellular migration to sites of damage and regeneration. Previous ex vivo studies have confirmed the potential of MSC to migrate over the endplate and enhance IVD-matrix production. In this study, we aimed to exploit this mechanism to engender IVD repair in a rat disc degeneration model. Methods Female Sprague Dawley rats were subjected to coccygeal disc degeneration through nucleus pulposus (NP) aspiration. In part 1; MSC or saline was transplanted into the vertebrae neighboring healthy or degenerative IVD subjected to irradiation or left untouched, and the ability to maintain the IVD integrity for 2 and 4 weeks was assessed by disc height index (DHI) and histology. For part 2, ubiquitously GFP expressing MSC were transplanted either intradiscally or vertebrally, and regenerative outcomes were compared at days 1, 5, and 14 post-transplantation. Moreover, the homing potential from vertebrae to IVD of the GFP+ MSC was assessed through cryosection mediated immunohistochemistry. Results Part 1 of the study revealed significantly improved maintenance of DHI for IVD vertebrally receiving MSC. Moreover, histological observations revealed a trend of IVD integrity maintenance. Part 2 of the study highlighted the enhanced DHI and matrix integrity for discs receiving MSC vertebrally compared with intradiscal injection. Moreover, GFP rates highlighted MSC migration and integration in the IVD at similar rates as the intradiscally treated cohort. Conclusion Vertebrally transplanted MSC had a beneficial effect on the degenerative cascade in their neighboring IVD, and thus potentially present an alternative administration strategy. Further investigation will be needed to determine the long-term effects, elucidate the role of cellular homing versus paracrine signaling, and validate our observations on a large animal model.
Collapse
Affiliation(s)
- Jordy Schol
- Department of Orthopaedic SurgeryTokai University School of MedicineIseharaJapan
- Research Center for Regenerative MedicineTokai University School of MedicineIseharaJapan
| | - Daisuke Sakai
- Department of Orthopaedic SurgeryTokai University School of MedicineIseharaJapan
| | - Takayuki Warita
- Research Center for Regenerative MedicineTokai University School of MedicineIseharaJapan
- TUNZ Pharma Co. Ltd.OsakaJapan
| | - Tadashi Nukaga
- Department of Orthopaedic SurgeryTokai University School of MedicineIseharaJapan
| | - Kosuke Sako
- Department of Orthopaedic SurgeryTokai University School of MedicineIseharaJapan
| | - Sebastian Wangler
- AO Research Institute DavosDavosSwitzerland
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University HospitalUniversity of BernBernSwitzerland
| | - Shota Tamagawa
- Department of Orthopaedic SurgeryTokai University School of MedicineIseharaJapan
- Department of Medicine for Orthopaedics and Motor OrganJuntendo University Graduate School of MedicineTokyoJapan
| | | | | | - Sibylle Grad
- AO Research Institute DavosDavosSwitzerland
- ETH Zürich, Institute for BiomechanicsZürichSwitzerland
| |
Collapse
|
6
|
Gutierrez RA, Fonseca VC, Darling EM. Chondrogenesis of Adipose-Derived Stem Cells Using an Arrayed Spheroid Format. Cell Mol Bioeng 2022; 15:587-597. [PMID: 36531862 PMCID: PMC9751248 DOI: 10.1007/s12195-022-00746-8] [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: 07/14/2022] [Accepted: 10/08/2022] [Indexed: 11/03/2022] Open
Abstract
Objective The chondrogenic response of adipose-derived stem cells (ASCs) is often assessed using 3D micromass protocols that use upwards of hundreds of thousands of cells. Scaling these systems up for high-throughput testing is technically challenging and wasteful given the necessary cell numbers and reagent volumes. However, adopting microscale spheroid cultures for this purpose shows promise. Spheroid systems work with only thousands of cells and microliters of medium. Methods Molded agarose microwells were fabricated using 2% w/v molten agarose and then equilibrated in medium prior to introducing cells. ASCs were seeded at 50, 500, 5k cells/microwell; 5k, 50k, cells/well plate; and 50k and 250k cells/15 mL centrifuge tube to compare chondrogenic responses across spheroid and micromass sizes. Cells were cultured in control or chondrogenic induction media. ASCs coalesced into spheroids/pellets and were cultured at 37 °C and 5% CO2 for 21 days with media changes every other day. Results All culture conditions supported growth of ASCs and formation of viable cell spheroids/micromasses. More robust growth was observed in chondrogenic conditions. Sulfated glycosaminoglycans and collagen II, molecules characteristics of chondrogenesis, were prevalent in both 5000-cell spheroids and 250,000-cell micromasses. Deposition of collagen I, characteristic of fibrocartilage, was more prevalent in the large micromasses than small spheroids. Conclusions Chondrogenic differentiation was consistently induced using high-throughput spheroid formats, particularly when seeding at cell densities of 5000 cells/spheroid. This opens possibilities for highly arrayed experiments investigating tissue repair and remodeling during or after exposure to drugs, toxins, or other chemicals. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-022-00746-8.
Collapse
Affiliation(s)
- Robert A. Gutierrez
- Center for Biomedical Engineering, Brown University, Box G-B397, Providence, RI 02912 USA
| | - Vera C. Fonseca
- Department of Pathology and Laboratory Medicine, Providence, USA
| | - Eric M. Darling
- Center for Biomedical Engineering, Brown University, Box G-B397, Providence, RI 02912 USA
- Department of Pathology and Laboratory Medicine, Providence, USA
- School of Engineering, Brown University, Providence, USA
- Department of Orthopaedics, Brown University, Providence, USA
| |
Collapse
|
7
|
Spatial patterning of phenotypically distinct microtissues to engineer osteochondral grafts for biological joint resurfacing. Biomaterials 2022; 289:121750. [DOI: 10.1016/j.biomaterials.2022.121750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 08/16/2022] [Indexed: 02/04/2023]
|
8
|
Effects of chondrogenic priming duration on mechanoregulation of engineered cartilage. J Biomech 2021; 125:110580. [PMID: 34198021 DOI: 10.1016/j.jbiomech.2021.110580] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/21/2022]
Abstract
Chondrocyte maturation during cartilage development occurs under diverse and dynamic mechanical environments. Mechanical stimulation through bioreactor culture may mimic these conditions to direct cartilage tissue engineering in vitro. Mechanical cues can promote chondrocyte homeostasis or hypertrophy and mineralization, depending potentially on the timing of load application. Here, we tested the effects of chondrogenic priming duration on the response of engineered human cartilage constructs to dynamic mechanical compression. We cultured human bone marrow stromal cells (hMSCs) in fibrin hydrogels under chondrogenic priming conditions for periods of 0, 2, 4, or 6 weeks prior to two weeks of either static culture or dynamic compression. We measured construct mechanical properties, cartilage matrix composition, and gene expression. Dynamic compression increased the equilibrium and dynamic modulus of the engineered tissue, depending on the duration of chondrogenic priming. For priming times of 2 weeks or greater, dynamic compression enhanced COL2A1 and AGGRECAN mRNA expression at the end of the loading period, but did not alter total collagen or glycosaminoglycan matrix deposition. Load initiation at priming times of 4 weeks or less repressed transient osteogenic signaling (RUNX2, OPN) and expression of CYR61, a YAP/TAZ-TEAD-target gene. No suppression of osteogenic gene expression was observed if loading was initiated after 6 weeks of in vitro priming, when mechanical stimulation was observed to increase the expression of type X collagen. Taken together, these data demonstrate that the duration of in vitro chondrogenic priming regulates the cell response to dynamic mechanical compression and suggests that early loading may preserve chondrocyte homeostasis while delayed loading may support cartilage maturation.
Collapse
|
9
|
Burdis R, Kelly DJ. Biofabrication and bioprinting using cellular aggregates, microtissues and organoids for the engineering of musculoskeletal tissues. Acta Biomater 2021; 126:1-14. [PMID: 33711529 DOI: 10.1016/j.actbio.2021.03.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/18/2022]
Abstract
The modest clinical impact of musculoskeletal tissue engineering (TE) can be attributed, at least in part, to a failure to recapitulate the structure, composition and functional properties of the target tissue. This has motivated increased interest in developmentally inspired TE strategies, which seek to recapitulate key events that occur during embryonic and post-natal development, as a means of generating truly biomimetic grafts to replace or regenerate damaged tissues and organs. Such TE strategies can be substantially enabled by emerging biofabrication and bioprinting strategies, and in particular the use of cellular aggregates, microtissues and organoids as 'building blocks' for the development of larger tissues and/or organ precursors. Here, the application of such biological building blocks for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. The importance of first scaling-down to later scale-up will be discussed, as this is viewed as a key component of engineering functional grafts using cellular aggregates or microtissues. In the context of engineering anatomically accurate tissues of scale suitable for tissue engineering and regenerative medicine applications, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues fuse and self-organise will be reviewed. Throughout the paper, we will highlight some of the key challenges facing this emerging field. STATEMENT OF SIGNIFICANCE: The field of bioprinting has grown substantially in recent years, but despite the hype and excitement it has generated, there are relatively few examples of bioprinting strategies producing implants with superior regenerative potential to that achievable with more traditional tissue engineering approaches. This paper provides an up-to-date review of emerging biofabrication and bioprinting strategies which use cellular aggregates and microtissues as 'building blocks' for the development of larger musculoskeletal tissues and/or organ precursors - a field of research that can potentially enable functional regeneration of damaged and diseased tissues. The application of cellular aggregates and microtissues for the engineering of musculoskeletal tissues, from vascularised bone to zonally organised articular cartilage, will be reviewed. In the context of engineering anatomically accurate tissues of scale, novel bioprinting modalities and their application in controlling the process by which cellular aggregates or microtissues self-organise is addressed, as well as key challenges facing this emerging field.
Collapse
|
10
|
Witte K, de Andrés MC, Wells J, Dalby MJ, Salmeron-Sanchez M, Oreffo ROC. Chondrobags: A high throughput alginate-fibronectin micromass platform for in vitro human cartilage formation. Biofabrication 2020; 12:045034. [DOI: 10.1088/1758-5090/abb653] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
11
|
Volleman TNE, Schol J, Morita K, Sakai D, Watanabe M. Wnt3a and wnt5a as Potential Chondrogenic Stimulators for Nucleus Pulposus Cell Induction: A Comprehensive Review. Neurospine 2020; 17:19-35. [PMID: 32252152 PMCID: PMC7136098 DOI: 10.14245/ns.2040040.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/18/2020] [Indexed: 12/20/2022] Open
Abstract
Low back pain remains a highly prevalent pathology engendering a tremendous socioeconomic burden. Low back pain is generally associated with intervertebral disc (IVD) degeneration, a process involving the deterioration of nucleus pulpous (NP) cells and IVD matrix. Scientific interest has directed efforts to restoring cell numbers as a strategy to enable IVD regeneration. Currently, mesenchymal stromal cells (MSCs) are being explored as cell therapy agents, due to their easy accessibility and differentiation potential. For enhancement of MSCs, growth factor supplementation is commonly applied to induce differentiation towards a chondrogenic (NP) cell phenotype. The wnt signaling pathways play a crucial role in chondrogenesis, nonetheless, literature appears to present controversies with regard to wnt3a and wnt5a for the induction of NP cells, chondrocytes, and MSCs. This review aims to summarize the reporting on wnt3a/wnt5a mediated NP cell differentiation, and to elucidate the mechanisms involved in wnt3a and wnt5a mediated chondrogenesis for potential application as cell therapy supplements for IVD regeneration. Our review suggests that wnt3a, subsequently replaced with a chondrogenic stimulating growth factor, can enhance the chondrogenic potential of MSCs in vitro. Contrariwise, wnt5a is suggested to play a role in maintaining cell potency of differentiated NP or chondrogenic cells.
Collapse
Affiliation(s)
- Tibo Nico Emmie Volleman
- Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jordy Schol
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Kosuke Morita
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
12
|
Li YY, Lam KL, Chen AD, Zhang W, Chan BP. Collagen microencapsulation recapitulates mesenchymal condensation and potentiates chondrogenesis of human mesenchymal stem cells – A matrix-driven in vitro model of early skeletogenesis. Biomaterials 2019; 213:119210. [DOI: 10.1016/j.biomaterials.2019.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 04/28/2019] [Accepted: 05/10/2019] [Indexed: 01/01/2023]
|
13
|
Liu F, Huang X, Luo Z, He J, Haider F, Song C, Peng L, Chen T, Wu B. Hypoxia-Activated PI3K/Akt Inhibits Oxidative Stress via the Regulation of Reactive Oxygen Species in Human Dental Pulp Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6595189. [PMID: 30728888 PMCID: PMC6343138 DOI: 10.1155/2019/6595189] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 09/19/2018] [Accepted: 10/30/2018] [Indexed: 01/02/2023]
Abstract
In order to use stem cells as a resource for tissue regeneration, it is necessary to induce expansion in vitro. However, during culture, stem cells often lose functional properties and become senescent. Increasing evidence indicates that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in vitro. The purpose of the current study was to test the hypothesis that hypoxic preconditioning with physiological oxygen concentration can maintain the functional properties of stem cells in culture by reducing oxidative stress. In vitro studies were performed in primary human dental pulp cells (hDPCs). Reduced levels of oxidative stress and increased cellular "stemness" in response to physiological hypoxia were dependent upon the expression of reactive oxygen species (ROS). Subsequently, RNA-sequencing analysis revealed the increased expression of phosphoinositide 3-kinase (PI3K)/Akt signaling in culture, a pathway which regulates oxidative stress. Furthermore, we found evidence that PI3K/Akt signaling might affect intracellular ROS production by negatively regulating expression of the downstream protein Forkhead Box Protein O1 (FOXO1) and Caspase 3. Collectively, our data show that the PI3K/Akt pathway is activated in response to hypoxia and inhibits oxidative stress in a ROS-dependent manner. This study identified redox-mediated hypoxic preconditioning regulatory mechanisms that may be significant for tissue regeneration.
Collapse
Affiliation(s)
- Fei Liu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- College of Stomatology, Southern Medical University, Guangzhou 510515, China
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Xin Huang
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhenhua Luo
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | - Jingjun He
- International Medical Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Farhan Haider
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ci Song
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ling Peng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Ting Chen
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Buling Wu
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
14
|
Park MJ, Lee J, Byeon JS, Jeong DU, Gu NY, Cho IS, Cha SH. Effects of three-dimensional spheroid culture on equine mesenchymal stem cell plasticity. Vet Res Commun 2018; 42:171-181. [PMID: 29721754 DOI: 10.1007/s11259-018-9720-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 04/02/2018] [Indexed: 01/01/2023]
Abstract
Mesenchymal stem cells (MSCs) are useful candidates for tissue engineering and cell therapy fields. We optimize culture conditions of equine adipose tissue-derived MSCs (eAD-MSCs) for treatment of horse fractures. To investigate enhancing properties of three-dimensional (3D) culture system in eAD-MSCs, we performed various sized spheroid formation and determined changes in gene expression levels to obtain different sized spheroid for cell therapy. eAD-MSCs were successfully isolated from horse tailhead. Using hanging drop method, spheroid formation was generated for three days. Quantitative real-time PCR was performed to analyze gene expression. As results, expression levels of pluripotent markers were increased depending on spheroid size and the production of PGE2 was increased in spheroid formation compared to that in monolayer. Ki-67 showed a remarkable increase in the spheroid formed with 2.0 × 105 cells/drop as compared to that in the monolayer. Expression levels of angiogenesis-inducing factors such as VEGF, IL-6, IL-8, and IL-18 were significantly increased in spheroid formation compared to those in the monolayer. Expression levels of bone morphogenesis-inducing factors such as Cox-2 and TGF-β1 were also significantly increased in spheroid formation compared to those in the monolayer. Expression levels of osteocyte-specific markers such as RUNX2, osteocalcin, and differentiation potential were also significantly increased in spheroid formation compared to those in the monolayer. Therefore, spheroid formation of eAD-MSCs through the hanging drop method can increases the expression of angiogenesis-inducing and bone morphogenesis-inducing factors under optimal culture conditions.
Collapse
Affiliation(s)
- Mi Jeong Park
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jienny Lee
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Jeong Su Byeon
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Da-Un Jeong
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Na-Yeon Gu
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - In-Soo Cho
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea
| | - Sang-Ho Cha
- Viral Disease Research Division, Animal and Plant Quarantine Agency, 177, Hyeoksin 8-ro, Gimcheon-si, Gyeongsangbuk-do, 39660, Republic of Korea.
| |
Collapse
|
15
|
Developmentally inspired programming of adult human mesenchymal stromal cells toward stable chondrogenesis. Proc Natl Acad Sci U S A 2018; 115:4625-4630. [PMID: 29666250 DOI: 10.1073/pnas.1720658115] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is generally accepted that adult human bone marrow-derived mesenchymal stromal cells (hMSCs) are default committed toward osteogenesis. Even when induced to chondrogenesis, hMSCs typically form hypertrophic cartilage that undergoes endochondral ossification. Because embryonic mesenchyme is obviously competent to generate phenotypically stable cartilage, it is questioned whether there is a correspondence between mesenchymal progenitor compartments during development and in adulthood. Here we tested whether forcing specific early events of articular cartilage development can program hMSC fate toward stable chondrogenesis. Inspired by recent findings that spatial restriction of bone morphogenetic protein (BMP) signaling guides embryonic progenitors toward articular cartilage formation, we hypothesized that selective inhibition of BMP drives the phenotypic stability of hMSC-derived chondrocytes. Two BMP type I receptor-biased kinase inhibitors were screened in a microfluidic platform for their time- and dose-dependent effect on hMSC chondrogenesis. The different receptor selectivity profile of tested compounds allowed demonstration that transient blockade of both ALK2 and ALK3 receptors, while permissive to hMSC cartilage formation, is necessary and sufficient to maintain a stable chondrocyte phenotype. Remarkably, even upon compound removal, hMSCs were no longer competent to undergo hypertrophy in vitro and endochondral ossification in vivo, indicating the onset of a constitutive change. Our findings demonstrate that adult hMSCs effectively share properties of embryonic mesenchyme in the formation of transient but also of stable cartilage. This opens potential pharmacological strategies to articular cartilage regeneration and more broadly indicates the relevance of developmentally inspired protocols to control the fate of adult progenitor cell systems.
Collapse
|
16
|
Khimchenko A, Schulz G, Thalmann P, Müller B. Implementation of a double-grating interferometer for phase-contrast computed tomography in a conventional system nanotom ® m. APL Bioeng 2018; 2:016106. [PMID: 31069291 PMCID: PMC6481705 DOI: 10.1063/1.5022184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 12/26/2017] [Indexed: 02/07/2023] Open
Abstract
Visualizing the internal architecture of large soft tissue specimens within the laboratory environment in a label-free manner is challenging, as the conventional absorption-contrast tomography yields a poor contrast. In this communication, we present the integration of an X-ray double-grating interferometer (XDGI) into an advanced, commercially available micro computed tomography system nanotom® m with a transmission X-ray source and a micrometer-sized focal spot. The performance of the interferometer is demonstrated by comparing the registered three-dimensional images of a human knee joint sample in phase- and conventional absorption-contrast modes. XDGI provides enough contrast (1.094 ± 0.152) to identify the cartilage layer, which is not recognized in the conventional mode (0.287 ± 0.003). Consequently, the two modes are complementary, as the present XDGI set-up only reaches a spatial resolution of (73 ± 6) μm, whereas the true micrometer resolution in the absorption-contrast mode has been proven. By providing complimentary information, XDGI is especially a supportive quantitative method for imaging soft tissues and visualizing weak X-ray absorbing species in the direct neighborhood of stronger absorbing components at the microscopic level.
Collapse
Affiliation(s)
- Anna Khimchenko
- Biomaterials Science Center, University of Basel, 4123 Allschwil, Switzerland
| | - Georg Schulz
- Biomaterials Science Center, University of Basel, 4123 Allschwil, Switzerland
| | - Peter Thalmann
- Biomaterials Science Center, University of Basel, 4123 Allschwil, Switzerland
| | - Bert Müller
- Biomaterials Science Center, University of Basel, 4123 Allschwil, Switzerland
| |
Collapse
|
17
|
Marklein RA, Lam J, Guvendiren M, Sung KE, Bauer SR. Functionally-Relevant Morphological Profiling: A Tool to Assess Cellular Heterogeneity. Trends Biotechnol 2018; 36:105-118. [DOI: 10.1016/j.tibtech.2017.10.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 12/16/2022]
|
18
|
Somoza RA, Correa D, Labat I, Sternberg H, Forrest ME, Khalil AM, West MD, Tesar P, Caplan AI. Transcriptome-Wide Analyses of Human Neonatal Articular Cartilage and Human Mesenchymal Stem Cell-Derived Cartilage Provide a New Molecular Target for Evaluating Engineered Cartilage. Tissue Eng Part A 2017; 24:335-350. [PMID: 28602122 DOI: 10.1089/ten.tea.2016.0559] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cellular differentiation comprises a progressive, multistep program that drives cells to fabricate a tissue with specific and site distinctive structural and functional properties. Cartilage constitutes one of the potential differentiation lineages that mesenchymal stem cells (MSCs) can follow under the guidance of specific bioactive agents. Single agents such as transforming growth factor beta (TGF-β) and bone morphogenetic protein 2 in unchanging culture conditions have been historically used to induce in vitro chondrogenic differentiation of MSCs. Despite the expression of traditional chondrogenic biomarkers such as type II collagen and aggrecan, the resulting tissue represents a transient cartilage rather than an in vivo articular cartilage (AC), differing significantly in structure, chemical composition, cellular phenotypes, and mechanical properties. Moreover, there have been no comprehensive, multicomponent parameters to define high-quality and functional engineered hyaline AC. To address these issues, we have taken an innovative approach based on the molecular interrogation of human neonatal articular cartilage (hNAC), dissected from the knees of 1-month-old cadaveric specimens. Subsequently, we compared hNAC-specific transcriptional regulatory elements and differentially expressed genes with adult human bone marrow (hBM) MSC-derived three-dimensional cartilage structures formed in vitro. Using microarray analysis, the transcriptome of hNAC was found to be globally distinct from the transient, cartilage-like tissue formed by hBM-MSCs in vitro. Specifically, over 500 genes that are highly expressed in hNAC were not expressed at any time point during in vitro human MSC chondrogenesis. The analysis also showed that the differences were less variant during the initial stages (first 7 days) of the in vitro chondrogenic differentiation program. These observations suggest that the endochondral fate of hBM-MSC-derived cartilage may be rerouted at earlier stages of the TGF-β-stimulated chondrogenic differentiation program. Based on these analyses, several key molecular differences (transcription factors and coded cartilage-related proteins) were identified in hNAC that will be useful as molecular inductors and identifiers of the in vivo AC phenotype. Our findings provide a new gold standard of a molecularly defined AC phenotype that will serve as a platform to generate novel approaches for AC tissue engineering.
Collapse
Affiliation(s)
- Rodrigo A Somoza
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio.,2 CWRU Center for Multimodal Evaluation of Engineered Cartilage, Cleveland, Ohio
| | - Diego Correa
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio.,3 Division of Sports Medicine, Department of Orthopaedics, Diabetes Research Institute and Cell Transplant Center, University of Miami , Miller School of Medicine, Miami, Florida
| | | | | | - Megan E Forrest
- 5 Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| | - Ahmad M Khalil
- 2 CWRU Center for Multimodal Evaluation of Engineered Cartilage, Cleveland, Ohio.,5 Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| | | | - Paul Tesar
- 5 Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| | - Arnold I Caplan
- 1 Department of Biology, Skeletal Research Center, Case Western Reserve University , Cleveland, Ohio.,2 CWRU Center for Multimodal Evaluation of Engineered Cartilage, Cleveland, Ohio
| |
Collapse
|
19
|
Pei M. Environmental preconditioning rejuvenates adult stem cells' proliferation and chondrogenic potential. Biomaterials 2016; 117:10-23. [PMID: 27923196 DOI: 10.1016/j.biomaterials.2016.11.049] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 11/24/2016] [Indexed: 12/13/2022]
Abstract
Adult stem cells are a promising cell source for cartilage regeneration. Unfortunately, due to donor age and ex vivo expansion, stem cell senescence becomes a huge hurdle for these cells to be used clinically. Increasing evidence indicates that environmental preconditioning is a powerful approach in promoting stem cells' ability to resist a harsh environment post-engraftment, such as hypoxia and inflammation. However, few reports organize and evaluate the literature regarding the rejuvenation effect of environmental preconditioning on stem cell proliferation and chondrogenic differentiation capacity, which are important variables for stem cell based tissue regeneration. This report aims to identify several critical environmental factors such as oxygen concentration, growth factors, and extracellular matrix and to discuss their preconditioning influence on stem cells' rejuvenation including proliferation and chondrogenic potential as well as underlying molecular mechanisms. We believe that environmental preconditioning based rejuvenation is a simpler and safer strategy to program pre-engraftment stem cells for better survival and enhanced proliferation and differentiation capacity without the undesired effects of some treatments, such as genetic manipulation.
Collapse
Affiliation(s)
- Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA; Exercise Physiology, West Virginia University, Morgantown, WV, USA; Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
20
|
Li Z, Ba R, Wang Z, Wei J, Zhao Y, Wu W. Angiogenic Potential of Human Bone Marrow-Derived Mesenchymal Stem Cells in Chondrocyte Brick-Enriched Constructs Promoted Stable Regeneration of Craniofacial Cartilage. Stem Cells Transl Med 2016; 6:601-612. [PMID: 28191761 PMCID: PMC5442805 DOI: 10.5966/sctm.2016-0050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/15/2016] [Indexed: 12/12/2022] Open
Abstract
Craniofacial deformities caused by congenital defects or trauma remain challenges for clinicians, whereas current surgical interventions present limited therapeutic outcomes. Injection of bone marrow‐derived mesenchymal stem cells (BMSCs) into the defect is highly desirable because such a procedure is microinvasive and grafts are more flexible to fill the lesions. However, preventing hypertrophic transition and morphological contraction remain significant challenges. We have developed an “all host derived” cell transplantation system composed of chondrocyte brick (CB)‐enriched platelet‐rich plasma (P) gel and BMSCs (B). Without exogenous biomaterials or growth factors, such grafts regenerate cartilage efficiently and present great clinical promise. In immunodeficient mice, we compared performance of BMSCs and BMSCs lacking angiogenic potential in CB‐B‐P constructs and followed the cartilage maturation process by histology, immunostaining, micro‐computed tomography, and protein analysis. We determined that angiogenesis occurred quickly inside rudimentary cartilage derived from CB‐B‐P constructs after implantation, which improved tissue survival, tissue growth, and production of chondrogenic signals from chondrocytes. In contrast, silencing angiogenic potential of BMSCs led to poor chondrogenesis accompanied by necrosis. Chondrocyte bricks merged rapidly with angiogenesis, which constituted an enclosed chondrogenic niche and effectively inhibited runt‐related transcription factor‐2‐dependent hypertrophic transition of BMSCs as well as endochondral ossification; progressive chondrogenic differentiation of BMSCs resulted in vascularization regression, thus favoring persistent chondrogenesis and effectively augmenting nasal cartilage. In conclusion, these findings provided a novel, efficient approach to regenerating cartilage tissues in vivo. Chondrocyte bricks mixed with P provide transient vascularization and a persistently chondrogenic microenvironment for BMSCs; this provides a mini‐invasive approach for craniofacial cartilage reconstruction. Stem Cells Translational Medicine2017;6:601–612
Collapse
Affiliation(s)
- Zhiye Li
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
- Department of Prosthodontics, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Ruikai Ba
- Department of Prosthodontics, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Zhifa Wang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jianhua Wei
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yimin Zhao
- Department of Prosthodontics, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Wei Wu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, and Shaanxi Key Laboratory of Stomatology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| |
Collapse
|
21
|
Fernández-Pernas P, Fafián-Labora J, Lesende-Rodriguez I, Mateos J, De la Fuente A, Fuentes I, De Toro Santos J, Blanco García F, Arufe MC. 3, 3', 5-triiodo-L-thyronine Increases In Vitro Chondrogenesis of Mesenchymal Stem Cells From Human Umbilical Cord Stroma Through SRC2. J Cell Biochem 2016; 117:2097-108. [PMID: 26869487 DOI: 10.1002/jcb.25515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/09/2016] [Indexed: 02/05/2023]
Abstract
Our group focuses on the study of mesenchymal stem cells (MSCs) from human umbilical cord stroma or Warthońs jelly and their directed differentiation toward chondrocyte-like cells capable of regenerating damaged cartilage when transplanted into an injured joint. This study aimed to determine whether lactogenic hormone prolactin (PRL) or 3, 3', 5-triiodo-L-thyronine (T3), the active thyroid hormone, modulates chondrogenesis in our in vitro model of directed chondrogenic differentiation, and whether Wnt signalling is involved in this modulation. MSCs from human umbilical cord stroma underwent directed differentiation toward chondrocyte-like cells by spheroid formation. The addition of T3 to the chondrogenic medium increased the expression of genes linked to chondrogenesis like collagen type 2, integrin alpha 10 beta 1, and Sox9 measured by quantitative real time polymerase chain reaction (qRT-PCR) analysis. Levels of collagen type 2 and aggrecane analyzed by immunohistochemistry, and staining by Safranin O were increased after 14 days in spheroid culture with T3 compared to those without T3 or only with PRL. B-catenin, Frizzled, and GSK-3β gene expressions were significantly higher in spheroids cultured with chondrogenic medium (CM) plus T3 compared to CM alone after 14 days in culture. The increase of chondrogenic differentiation was inhibited when the cells were treated with T3 plus ML151, an inhibitor of the T3 steroid receptor. This work demonstrates, for first time, that T3 promotes differentiation towards chondrocytes-like cells in our in vitro model, that this differentiation is mediated by steroid receptor co-activator 2 (SRC2) and does not induce hypertrophy. J. Cell. Biochem. 117: 2097-2108, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Pablo Fernández-Pernas
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Juan Fafián-Labora
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Iván Lesende-Rodriguez
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Jesús Mateos
- Grupo de Proteómica-PBR2-ProteoRed/ISCIII-Servicio de Reumatologia, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, España
| | - Alexandre De la Fuente
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Isaac Fuentes
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Javier De Toro Santos
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| | - Fco Blanco García
- Grupo de Proteómica-PBR2-ProteoRed/ISCIII-Servicio de Reumatologia, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, España
| | - María C Arufe
- Grupo de Terapia Celular y Medicina Regenerativa (TCMR-CHUAC), CIBER-BBN/ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), SERGAS, Departamento de Medicina, Facultade de Oza, Universidade da Coruña (UDC), As Xubias, 15006, A Coruña, Spain
| |
Collapse
|
22
|
Sridharan B, Lin SM, Hwu AT, Laflin AD, Detamore MS. Stem Cells in Aggregate Form to Enhance Chondrogenesis in Hydrogels. PLoS One 2015; 10:e0141479. [PMID: 26719986 PMCID: PMC4697858 DOI: 10.1371/journal.pone.0141479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/08/2015] [Indexed: 12/22/2022] Open
Abstract
There are a variety of exciting hydrogel technologies being explored for cartilage regenerative medicine. Our overall goal is to explore whether using stem cells in an aggregate form may be advantageous in these applications. 3D stem cell aggregates hold great promise as they may recapitulate the in vivo skeletal tissue condensation, a property that is not typically observed in 2D culture. We considered two different stem cell sources, human umbilical cord Wharton’s jelly cells (hWJCs, currently being used in clinical trials) and rat bone marrow-derived mesenchymal stem cells (rBMSCs). The objective of the current study was to compare the influence of cell phenotype, aggregate size, and aggregate number on chondrogenic differentiation in a generic hydrogel (agarose) platform. Despite being differing cell sources, both rBMSC and hWJC aggregates were consistent in outperforming cell suspension control groups in biosynthesis and chondrogenesis. Higher cell density impacted biosynthesis favorably, and the number of aggregates positively influenced chondrogenesis. Therefore, we recommend that investigators employing hydrogels consider using cells in an aggregate form for enhanced chondrogenic performance.
Collapse
Affiliation(s)
- BanuPriya Sridharan
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
| | - Staphany M. Lin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Alexander T. Hwu
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Amy D. Laflin
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - Michael S. Detamore
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, United States of America
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
23
|
Janeczek AA, Tare RS, Scarpa E, Moreno-Jimenez I, Rowland CA, Jenner D, Newman TA, Oreffo ROC, Evans ND. Transient Canonical Wnt Stimulation Enriches Human Bone Marrow Mononuclear Cell Isolates for Osteoprogenitors. Stem Cells 2015; 34:418-30. [PMID: 26573091 PMCID: PMC4981914 DOI: 10.1002/stem.2241] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/01/2015] [Indexed: 12/27/2022]
Abstract
Activation of the canonical Wnt signaling pathway is an attractive anabolic therapeutic strategy for bone. Emerging data suggest that activation of the Wnt signaling pathway promotes bone mineral accrual in osteoporotic patients. The effect of Wnt stimulation in fracture healing is less clear as Wnt signaling has both stimulatory and inhibitory effects on osteogenesis. Here, we tested the hypothesis that transient Wnt stimulation promotes the expansion and osteogenesis of a Wnt‐responsive stem cell population present in human bone marrow. Bone marrow mononuclear cells (BMMNCs) were isolated from patients undergoing hip arthroplasty and exposed to Wnt3A protein. The effect of Wnt pathway stimulation was determined by measuring the frequency of stem cells within the BMMNC populations by fluorescence‐activated cell sorting and colony forming unit fibroblast (CFU‐F) assays, before determining their osteogenic capacity in in vitro differentiation experiments. We found that putative skeletal stem cells in BMMNC isolates exhibited elevated Wnt pathway activity compared with the population as whole. Wnt stimulation resulted in an increase in the frequency of skeletal stem cells marked by the STRO‐1bright/Glycophorin A− phenotype. Osteogenesis was elevated in stromal cell populations arising from BMMNCs transiently stimulated by Wnt3A protein, but sustained stimulation inhibited osteogenesis in a concentration‐dependent manner. These results demonstrate that Wnt stimulation could be used as a therapeutic approach by transient targeting of stem cell populations during early fracture healing, but that inappropriate stimulation may prevent osteogenesis. Stem Cells2016;34:418–430
Collapse
Affiliation(s)
- Agnieszka A Janeczek
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Rahul S Tare
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Edoardo Scarpa
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Ines Moreno-Jimenez
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Caroline A Rowland
- Microbiology group, Chemical, Biological and Radiological Division, Salisbury, United Kingdom
| | - Dominic Jenner
- Microbiology group, Chemical, Biological and Radiological Division, Salisbury, United Kingdom
| | - Tracey A Newman
- Clinical and Experimental Sciences, Faculty of Medicine, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Richard O C Oreffo
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Nicholas D Evans
- Centre for Human Development, Stem Cells and Regeneration, University of Southampton, Southampton, United Kingdom
- Bone and Joint Research Group, Human Development and Health Academic Unit, Institute for Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
24
|
Oral Presentations. Regen Med 2015. [DOI: 10.2217/rme.15.72] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
25
|
Herklotz M, Prewitz MC, Bidan CM, Dunlop JW, Fratzl P, Werner C. Availability of extracellular matrix biopolymers and differentiation state of human mesenchymal stem cells determine tissue-like growth in vitro. Biomaterials 2015; 60:121-9. [DOI: 10.1016/j.biomaterials.2015.04.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 04/19/2015] [Accepted: 04/30/2015] [Indexed: 12/12/2022]
|
26
|
Klumpers DD, Mooney DJ, Smit TH. From Skeletal Development to Tissue Engineering: Lessons from the Micromass Assay. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:427-37. [PMID: 25946390 DOI: 10.1089/ten.teb.2014.0704] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Damage and degeneration of the skeletal elements due to disease, trauma, and aging lead to a significant health and economical burden. To reduce this burden, skeletal tissue engineering strategies aim to regenerate functional bone and cartilage in the adult body. However, challenges still exist. Such challenges involve the identification of the external cues that determine differentiation, how to control chondrocyte hypertrophy, and how to achieve specific tissue patterns and boundaries. To address these issues, it could be insightful to look at skeletal development, a robust morphogenetic process that takes place during embryonic development and is commonly modeled in vitro by the micromass assay. In this review, we investigate what the tissue engineering field can learn from this assay. By comparing embryonic skeletal precursor cells from different anatomic locations and developmental stages in micromass, the external cues that guide lineage commitment can be identified. The signaling pathways regulating chondrocyte hypertrophy, and the cues required for tissue patterning, can be elucidated by combining the micromass assay with genetic, molecular, and engineering tools. The lessons from the micromass assay are limited by two major differences between developmental and regenerative skeletogenesis: cell type and scale. We highlight an important difference between embryonic and adult skeletal progenitor cells, in that adult progenitors are not able to form mesenchymal condensations spontaneously. Also, the mechanisms of tissue patterning need to be adjusted to the larger tissue engineering constructs. In conclusion, mechanistic insights of skeletal tissue generation gained from the micromass model could lead to improved tissue engineering strategies and constructs.
Collapse
Affiliation(s)
- Darinka D Klumpers
- 1 School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts.,2 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts.,3 Department of Orthopedic Surgery, VU University Medical Centre MOVE Research Institute , Amsterdam, The Netherlands
| | - David J Mooney
- 1 School of Engineering and Applied Sciences, Harvard University , Cambridge, Massachusetts.,2 Wyss Institute for Biologically Inspired Engineering, Harvard University , Boston, Massachusetts
| | - Theo H Smit
- 3 Department of Orthopedic Surgery, VU University Medical Centre MOVE Research Institute , Amsterdam, The Netherlands
| |
Collapse
|
27
|
Occhetta P, Centola M, Tonnarelli B, Redaelli A, Martin I, Rasponi M. High-Throughput Microfluidic Platform for 3D Cultures of Mesenchymal Stem Cells, Towards Engineering Developmental Processes. Sci Rep 2015; 5:10288. [PMID: 25983217 PMCID: PMC4650750 DOI: 10.1038/srep10288] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 04/09/2015] [Indexed: 01/16/2023] Open
Abstract
The development of in vitro models to screen the effect of different concentrations, combinations and temporal sequences of morpho-regulatory factors on stem/progenitor cells is crucial to investigate and possibly recapitulate developmental processes with adult cells. Here, we designed and validated a microfluidic platform to (i) allow cellular condensation, (ii) culture 3D micromasses of human bone marrow-derived mesenchymal stromal cells (hBM-MSCs) under continuous flow perfusion, and (ii) deliver defined concentrations of morphogens to specific culture units. Condensation of hBM-MSCs was obtained within 3 hours, generating micromasses in uniform sizes (56.2 ± 3.9 μm). As compared to traditional macromass pellet cultures, exposure to morphogens involved in the first phases of embryonic limb development (i.e. Wnt and FGF pathways) yielded more uniform cell response throughout the 3D structures of perfused micromasses (PMMs), and a 34-fold higher percentage of proliferating cells at day 7. The use of a logarithmic serial dilution generator allowed to identify an unexpected concentration of TGFβ3 (0.1 ng/ml) permissive to hBM-MSCs proliferation and inductive to chondrogenesis. This proof-of-principle study supports the described microfluidic system as a tool to investigate processes involved in mesenchymal progenitor cells differentiation, towards a 'developmental engineering' approach for skeletal tissue regeneration.
Collapse
Affiliation(s)
- Paola Occhetta
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Matteo Centola
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Beatrice Tonnarelli
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Ivan Martin
- Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| |
Collapse
|
28
|
The effect of growth-mimicking continuous strain on the early stages of skeletal development in micromass culture. PLoS One 2015; 10:e0124948. [PMID: 25915898 PMCID: PMC4411057 DOI: 10.1371/journal.pone.0124948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/19/2015] [Indexed: 11/19/2022] Open
Abstract
Embryonic skeletogenesis involves proliferation, condensation and subsequent chondrogenic differentiation of mesenchymal precursor cells, and the strains and stresses inherent to these processes have been hypothesized to influence skeletal development. The aim of this study was to determine the effect of growth-mimicking strain on the process of early skeletal development in vitro. To this end, we applied continuous uniaxial strain to embryonic skeletal precursor cells in micromass culture. Strain was applied at different times of culture to specifically address the effect of mechanical loading on the sequential stages of cellular proliferation, condensation and differentiation. We found that growth-mimicking strain at all three times did not affect proliferation or chondrogenic differentiation under the tested conditions. However, the timing of the applied strain did play a role in the density of mesenchymal condensations. This finding suggests that a mechanically dynamic environment, and specifically strain, can influence skeletal patterning. The growth-mimicking micromass model presented here may be a useful tool for further studies into the role of mechanical loading in early skeletal development.
Collapse
|
29
|
Bhattacharjee M, Coburn J, Centola M, Murab S, Barbero A, Kaplan DL, Martin I, Ghosh S. Tissue engineering strategies to study cartilage development, degeneration and regeneration. Adv Drug Deliv Rev 2015; 84:107-22. [PMID: 25174307 DOI: 10.1016/j.addr.2014.08.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 08/01/2014] [Accepted: 08/20/2014] [Indexed: 01/09/2023]
Abstract
Cartilage tissue engineering has primarily focused on the generation of grafts to repair cartilage defects due to traumatic injury and disease. However engineered cartilage tissues have also a strong scientific value as advanced 3D culture models. Here we first describe key aspects of embryonic chondrogenesis and possible cell sources/culture systems for in vitro cartilage generation. We then review how a tissue engineering approach has been and could be further exploited to investigate different aspects of cartilage development and degeneration. The generated knowledge is expected to inform new cartilage regeneration strategies, beyond a classical tissue engineering paradigm.
Collapse
|
30
|
Stoddart MJ, Bara J, Alini M. Cells and secretome--towards endogenous cell re-activation for cartilage repair. Adv Drug Deliv Rev 2015; 84:135-45. [PMID: 25174306 DOI: 10.1016/j.addr.2014.08.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/26/2014] [Accepted: 08/20/2014] [Indexed: 01/01/2023]
Abstract
Regenerative medicine approaches to cartilage tissue repair have mainly been concerned with the implantation of a scaffold material containing monolayer expanded cells into the defect, with the aim to differentiate the cells into chondrocytes. While this may be a valid approach, the secretome of the implanted cells and its effects on the endogenous resident cells, is gaining in interest. This review aims to summarize the knowledge on the secretome of mesenchymal stem cells, including knowledge from other tissues, in order to indicate how these mechanisms may be of value in repairing articular cartilage defects. Potential therapies and their effects on the repair of articular cartilage defects will be discussed, with a focus on the transition from classical cell therapy to the implantation of cell free matrices releasing specific cytokines.
Collapse
|
31
|
Cell-bricks based injectable niche guided persistent ectopic chondrogenesis of bone marrow-derived mesenchymal stem cells and enabled nasal augmentation. Stem Cell Res Ther 2015; 6:16. [PMID: 25886527 PMCID: PMC4396784 DOI: 10.1186/s13287-015-0006-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/24/2014] [Accepted: 02/09/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Developing cartilage constructs with injectability, appropriate matrix composition and persistent cartilaginous phenotype remains an enduring challenge in cartilage repair. Bone marrow derived mesenchymal stem cells (BMSCs) have chondrogenic potential. Current approaches to drive their chondrogenic differentiation require extensive cell manipulation ex vivo and using exogenous growth factors. However, preventing hypertrophic transition of BMSCs in vivo and maintaining persistent chondrogenesis remain bottlenecks in clinical application. This study aimed to develop completely biological, injectable constructs to generate cartilage by co-transplanting chondrocyte and BMSCs. Methods We fabricated fragmented chondrocyte macroaggregate (cell bricks) and mixed them with platelet rich plasma (PRP); BMSCs were mixed into the above constructs, allowed to clot and then subcutaneously injected into nude mice. Gross morphology observation, histological and immunohistochemical assay, immunofluorescence assay, biochemical analysis and gene expression analysis were used to compare the properties of BMSC-cell bricks-PRP complex with BMSC in PRP or BMSC/chondrocytes in PRP. Results The constructs of BMSCs-cell bricks-PRP that were subcutaneously injected resulted in persistent chondrogenesis with appropriate morphology, adequate central nutritional perfusion without central necrosis or ossification, and further augmented nasal dorsum without obvious contraction and deformation. Conclusions We concluded that cell bricks-enriched PRP clotting provides an autologous substance derived niche for chondrogenic differentiation of BMSCs in vivo, which suggests that such an injectable, completely biological system is a suitable stem cell carrier for micro-invasive cartilage repair.
Collapse
|
32
|
Su X, Zuo W, Wu Z, Chen J, Wu N, Ma P, Xia Z, Jiang C, Ye Z, Liu S, Liu J, Zhou G, Wan C, Qiu G. CD146 as a new marker for an increased chondroprogenitor cell sub-population in the later stages of osteoarthritis. J Orthop Res 2015; 33:84-91. [PMID: 25266708 DOI: 10.1002/jor.22731] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 08/18/2014] [Indexed: 02/04/2023]
Abstract
Cartilage-derived mesenchymal stem cells (MSCs) have been isolated with different methods. In this study lateral and medial femoral condyles were respectively collected from patients with late-stage osteoarthritis during the total knee arthroplasty. After digestion of the cartilage tissues with type II collagenase and analysis by fluorescence-activated cell sorting (FACS) with CD146, a chondroprogenitor cell sub-population were isolated and purified. The expression of other MSC-associated markers in the CD146+ chondroprogenitors was analyzed by flow cytometry. Multi-lineage differentiation capacity of CD146+ chondroprogenitors was compared with that of unsorted chondrocytes and adipose-derived MSCs (ADMSCs). Higher percentage of CD146+ chondroprogenitors isolated from the medial femoral condyles was observed than that from the lateral. CD146+ chondroprogenitors expressed high levels of MSC-specific surface antigens, and showed higher chondrogenesis capacity than ADMSCs and unsorted chondrocytes in a 3D cell pellet culture model. Thus CD146 might be a new cell surface marker for cartilage progenitor cell population in the late-stage osteoarthritis.
Collapse
Affiliation(s)
- Xinlin Su
- Department of Orthopaedics Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Centola M, Tonnarelli B, Hendriks J, van den Doel M, Feliciano S, Papadimitropoulos A, Piccinini E, Geurts J, Martin I, Barbero A. An improved cartilage digestion method for research and clinical applications. Tissue Eng Part C Methods 2014; 21:394-403. [PMID: 25226356 DOI: 10.1089/ten.tec.2014.0393] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Enzymatic isolation of chondrocytes from a cartilage biopsy is the first step to establish in vitro models of chondrogenesis or to generate cell-based grafts for cartilage repair. Such process is based on manually operated procedures and typically results in yields lower than 20% of the total available cells. In this study, we hypothesized that, as compared to conventionally used protocols, the enzymatic digestion of human articular cartilage in the presence of ascorbic acid 2-phosphate (AscA2P) or of sodium chloride (NaCl), in combination with the use of a perfusion bioreactor system, leads to a higher and more reproducible yield of cell populations with high proliferation and chondrogenic capacity. The addition of AscA2P within the enzymatic digestion medium did not significantly increase the cell yield, but resulted in a significant decrease of the intradonor variability in cell yield (-17.8% ± 10.7%, p = 0.0247) and in a significant increase of the proliferation rate of the isolated chondrocytes (+19.0% ± 1.4%, p < 0.05) with respect to the control group. The addition of NaCl during cartilage digestion did not modulate cell yield. When the cartilage digestion was further performed under direct perfusion flow, beneficial synergistic effects were achieved, with an overall increase of 34.7% ± 6.8% (p < 0.001) in the cell yield and an average decrease of 57.8% ± 11.2% (p < 0.01) in the coefficient of variation with respect to the control group. Importantly, by implementing this strategy it was possible to retrieve clonal subpopulations more efficiently capable of undergoing chondrogenesis, both in vitro and in vivo. Our findings bear relevance for the preparation of human chondrocytes for laboratory investigations, and in the perspective of efficient and streamlined manufacturing of cell/tissue grafts for articular cartilage repair.
Collapse
Affiliation(s)
- Matteo Centola
- 1 Departments of Surgery and of Biomedicine, University Hospital Basel, University of Basel , Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Moghadam FH, Tayebi T, Dehghan M, Eslami G, Nadri H, Moradi A, Vahedian-Ardakani H, Barzegar K. Differentiation of bone marrow mesenchymal stem cells into chondrocytes after short term culture in alkaline medium. Int J Hematol Oncol Stem Cell Res 2014; 8:12-9. [PMID: 25774263 PMCID: PMC4345293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 03/23/2014] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (MSCs) are one of the undifferentiated multipotential cell sources of human body. MSCs have the capacity to form a variety of cell types, especially chondrocytes and osteocytes. Learning about responses of MSCs to external milieu and chemical factors such as pH could recommend new approaches for preparation of suitable scaffolds for bone and cartilage tissue engineering. In present study, the effect of alkaline medium on chondrogenic and osteogenic differentiation of rat MSCs was evaluated. METHODS MSCs were harvested from bone marrow of animals and then the response of passage1 and 2 of MSCs (P1 MSCs & P2 MSCs) to the culture in alkaline medium (pH: 8) was evaluated. Cytochemical and immunocytochemical staining were performed to distinguish chondrocytes and osteocytes. Real-time PCR was performed to evaluate the type II collagen and osteopontin mRNA levels. RESULTS Staining for type II collagen, a chondrocytic specific marker, revealed that after one-week culture in alkaline medium, a considerable amount of P1 MSCs had shown chondrocytic morphology. By prolonging the culture period up to 4 weeks, osteogenic cells with expanded matrix and mineralized areas around them were appeared. Results of real-time PCR showed that P1 MSCs after one week culture in alkaline medium expressed highest rate of type II collagen and osteopontin mRNA among all groups. CONCLUSION This study demonstrated that alkaline medium is a potent chondrogenic differentiation inducer for MSCs in their first passage.
Collapse
Affiliation(s)
- Farshad Homayouni Moghadam
- Department of Physiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran,Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Tahereh Tayebi
- Department of Physiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maryam Dehghan
- Neurobiomedical Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Gilda Eslami
- Department of Medical Parasitology and Mycology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamid Nadri
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Alireza Moradi
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hassanali Vahedian-Ardakani
- Department of Internal Medicine, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Kazem Barzegar
- English Language Department, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
35
|
Klumpers DD, Mao AS, Smit TH, Mooney DJ. Linear patterning of mesenchymal condensations is modulated by geometric constraints. J R Soc Interface 2014; 11:20140215. [PMID: 24718453 DOI: 10.1098/rsif.2014.0215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The development of the vertebral column starts with the formation of a linear array of mesenchymal condensations, forming the blueprint for the eventual alternating pattern of bone and cartilage. Despite growing insight into the molecular mechanisms of morphogenesis, the impact of the physical aspects of the environment is not well understood. We hypothesized that geometric boundary conditions may play a pivotal role in the linear patterning of condensations, as neighbouring tissues provide physical constraints to the cell population. To study the process of condensation and the patterning thereof under tightly controlled geometric constraints, we developed a novel in vitro model that combines micropatterning with the established micromass assay. The spacing and alignment of condensations changed with the width of the cell adhesive patterns, a phenomenon that could not be explained by cell availability alone. Moreover, the extent of chondrogenic commitment was increased on substrates with tighter geometric constraints. When the in vivo pattern of condensations was investigated in the developing vertebral column of chicken embryos, the measurements closely fit into the quantitative relation between geometric constraints and inter-condensation distance found in vitro. Together, these findings suggest a potential role of geometric constraints in skeletal patterning in a cellular process of self-organization.
Collapse
Affiliation(s)
- Darinka D Klumpers
- School of Engineering and Applied Sciences, Harvard University, , 29 Oxford St., Cambridge, MA 02138, USA
| | | | | | | |
Collapse
|
36
|
Abstract
With few exceptions, tissue regeneration strategies based on the conventional combination of cells, scaffolding materials, and soluble factors (tissue engineering) have introduced a rather limited clinical impact. While it is being recognized that the nonconvincing benefits of engineered grafts require more fundamental knowledge on mechanisms of action and potency factors, the attempt to mimic and recapitulate developmental events has inspired an evolution of the paradigm. In the context of skeletal regeneration, a "developmental engineering" approach has been advocated to generate intermediate grafts (i.e., hypertrophic cartilage templates) which, as suggested by limb developmental biology, are capable of autonomous spatial and temporal evolution into fully functional bone organs. However, limited consideration has been given to the fact that the recipient site within adult organisms may not be compatible with well-established developmental processes. This can be due to the possibly restricted function of resident progenitors, to the critical mechanical and physical boundary conditions of mature organs, or to the strong role of inflammatory signals and immune cells at repair sites. We thus propose that predictable, orderly, and durable tissue regeneration should be based on a "developmental RE-engineering" paradigm, with the challenge to instruct the execution of developmental programs in the context of an adult system.
Collapse
|