351
|
Jakobsen RB, Østrup E, Zhang X, Mikkelsen TS, Brinchmann JE. Analysis of the effects of five factors relevant to in vitro chondrogenesis of human mesenchymal stem cells using factorial design and high throughput mRNA-profiling. PLoS One 2014; 9:e96615. [PMID: 24816923 PMCID: PMC4015996 DOI: 10.1371/journal.pone.0096615] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 04/09/2014] [Indexed: 02/07/2023] Open
Abstract
The in vitro process of chondrogenic differentiation of mesenchymal stem cells for tissue engineering has been shown to require three-dimensional culture along with the addition of differentiation factors to the culture medium. In general, this leads to a phenotype lacking some of the cardinal features of native articular chondrocytes and their extracellular matrix. The factors used vary, but regularly include members of the transforming growth factor β superfamily and dexamethasone, sometimes in conjunction with fibroblast growth factor 2 and insulin-like growth factor 1, however the use of soluble factors to induce chondrogenesis has largely been studied on a single factor basis. In the present study we combined a factorial quality-by-design experiment with high-throughput mRNA profiling of a customized chondrogenesis related gene set as a tool to study in vitro chondrogenesis of human bone marrow derived mesenchymal stem cells in alginate. 48 different conditions of transforming growth factor β 1, 2 and 3, bone morphogenetic protein 2, 4 and 6, dexamethasone, insulin-like growth factor 1, fibroblast growth factor 2 and cell seeding density were included in the experiment. The analysis revealed that the best of the tested differentiation cocktails included transforming growth factor β 1 and dexamethasone. Dexamethasone acted in synergy with transforming growth factor β 1 by increasing many chondrogenic markers while directly downregulating expression of the pro-osteogenic gene osteocalcin. However, all factors beneficial to the expression of desirable hyaline cartilage markers also induced undesirable molecules, indicating that perfect chondrogenic differentiation is not achievable with the current differentiation protocols.
Collapse
Affiliation(s)
- Rune B. Jakobsen
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Biochemistry, Institute of Basic Medical Sciences, The Medical Faculty, University of Oslo, Oslo, Norway
| | - Esben Østrup
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, Norway
| | - Xiaolan Zhang
- Broad Institute, Cambridge, Massachusetts, United States of America
| | - Tarjei S. Mikkelsen
- Broad Institute, Cambridge, Massachusetts, United States of America
- Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Jan E. Brinchmann
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Biochemistry, Institute of Basic Medical Sciences, The Medical Faculty, University of Oslo, Oslo, Norway
- Institute of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- * E-mail:
| |
Collapse
|
352
|
Li S, Oreffo ROC, Sengers BG, Tare RS. The effect of oxygen tension on human articular chondrocyte matrix synthesis: integration of experimental and computational approaches. Biotechnol Bioeng 2014; 111:1876-85. [PMID: 24668194 PMCID: PMC4284020 DOI: 10.1002/bit.25241] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 03/01/2014] [Accepted: 03/19/2014] [Indexed: 02/06/2023]
Abstract
Significant oxygen gradients occur within tissue engineered cartilaginous constructs. Although oxygen tension is an important limiting parameter in the development of new cartilage matrix, its precise role in matrix formation by chondrocytes remains controversial, primarily due to discrepancies in the experimental setup applied in different studies. In this study, the specific effects of oxygen tension on the synthesis of cartilaginous matrix by human articular chondrocytes were studied using a combined experimental-computational approach in a “scaffold-free” 3D pellet culture model. Key parameters including cellular oxygen uptake rate were determined experimentally and used in conjunction with a mathematical model to estimate oxygen tension profiles in 21-day cartilaginous pellets. A threshold oxygen tension (pO2 ≈ 8% atmospheric pressure) for human articular chondrocytes was estimated from these inferred oxygen profiles and histological analysis of pellet sections. Human articular chondrocytes that experienced oxygen tension below this threshold demonstrated enhanced proteoglycan deposition. Conversely, oxygen tension higher than the threshold favored collagen synthesis. This study has demonstrated a close relationship between oxygen tension and matrix synthesis by human articular chondrocytes in a “scaffold-free” 3D pellet culture model, providing valuable insight into the understanding and optimization of cartilage bioengineering approaches. Biotechnol. Bioeng. 2014;111: 1876–1885.
Collapse
Affiliation(s)
- S Li
- Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, Hampshire, UK
| | | | | | | |
Collapse
|
353
|
Pievani A, Scagliotti V, Russo FM, Azario I, Rambaldi B, Sacchetti B, Marzorati S, Erba E, Giudici G, Riminucci M, Biondi A, Vergani P, Serafini M. Comparative analysis of multilineage properties of mesenchymal stromal cells derived from fetal sources shows an advantage of mesenchymal stromal cells isolated from cord blood in chondrogenic differentiation potential. Cytotherapy 2014; 16:893-905. [PMID: 24794181 PMCID: PMC4062948 DOI: 10.1016/j.jcyt.2014.02.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 01/29/2014] [Accepted: 02/09/2014] [Indexed: 12/20/2022]
Abstract
Background aims Cord blood (CB) and amniotic fluid (AF) could represent new and attractive mesenchymal stromal cell (MSC) sources, but their potential therapeutic applications are still limited by lack of standardized protocols for isolation and differentiation. In particular, chondrogenic differentiation has never been deeply investigated. Methods MSCs were obtained from CB and AF samples collected during cesarean sections at term and compared for their biological and differentiation properties, with particular interest in cartilage differentiation, in which quantitative real-time polymerase chain reaction and immunohistochemical analyses were performed to evaluate the expression of type 2 collagen, type 10 collagen, SRY-box9 and aggrecan. Results We were able to isolate MSCs from 12 of 30 (40%) and 5 of 20 (25%) CB and AF units, respectively. Fluorescence in situ hybridization analysis indicated the fetal origin of isolated MSC strains. Both populations expressed mesenchymal but not endothelial and hematopoietic markers, even though we observed a lower expression of human leukocyte antigen (HLA) I in CB-MSCs. No differences in proliferation rate and cell cycle analysis could be detected. After osteogenic induction, both populations showed matrix mineralization and typical marker expression. Under chondrogenic conditions, pellets derived from CB-MSCs, in contrast with AF-MSCs pellets, were significantly larger, showed cartilage-like morphology and resulted positive for chondrocyte-associated markers, such as type 2 collagen, type 10 collagen, SRY-box9 and aggrecan. Conclusions Our results show that CB-MSCs and AF-MSCs collected at term differ from each other in their biological and differentiation properties. In particular, only CB-MSCs showed a clear chondrogenic potential and thus could represent an ideal candidate for cartilage-tissue engineering.
Collapse
Affiliation(s)
- Alice Pievani
- Dulbecco Telethon Institute at Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy; Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Valeria Scagliotti
- Dulbecco Telethon Institute at Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy; Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | | | - Isabella Azario
- Dulbecco Telethon Institute at Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy; Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Benedetta Rambaldi
- Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | | | - Simona Marzorati
- Dulbecco Telethon Institute at Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy; Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Eugenio Erba
- Department of Oncology, Flow Cytometry Unit, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Giovanni Giudici
- Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | - Andrea Biondi
- Pediatric Department, University of Milano-Bicocca, Fondazione MBBM/San Gerardo Hospital, Monza, Italy
| | - Patrizia Vergani
- Department of Obstetrics and Gynecology, University of Milano-Bicocca, Monza, Italy
| | - Marta Serafini
- Dulbecco Telethon Institute at Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy; Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy.
| |
Collapse
|
354
|
Large, stratified, and mechanically functional human cartilage grown in vitro by mesenchymal condensation. Proc Natl Acad Sci U S A 2014; 111:6940-5. [PMID: 24778247 DOI: 10.1073/pnas.1324050111] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The efforts to grow mechanically functional cartilage from human mesenchymal stem cells have not been successful. We report that clinically sized pieces of human cartilage with physiologic stratification and biomechanics can be grown in vitro by recapitulating some aspects of the developmental process of mesenchymal condensation. By exposure to transforming growth factor-β, mesenchymal stem cells were induced to condense into cellular bodies, undergo chondrogenic differentiation, and form cartilagenous tissue, in a process designed to mimic mesenchymal condensation leading into chondrogenesis. We discovered that the condensed mesenchymal cell bodies (CMBs) formed in vitro set an outer boundary after 5 d of culture, as indicated by the expression of mesenchymal condensation genes and deposition of tenascin. Before setting of boundaries, the CMBs could be fused into homogenous cellular aggregates giving rise to well-differentiated and mechanically functional cartilage. We used the mesenchymal condensation and fusion of CMBs to grow centimeter-sized, anatomically shaped pieces of human articular cartilage over 5 wk of culture. For the first time to our knowledge biomechanical properties of cartilage derived from human mesenchymal cells were comparable to native cartilage, with the Young's modulus of >800 kPa and equilibrium friction coeffcient of <0.3. We also demonstrate that CMBs have capability to form mechanically strong cartilage-cartilage interface in an in vitro cartilage defect model. The CMBs, which acted as "lego-like" blocks of neocartilage, were capable of assembling into human cartilage with physiologic-like structure and mechanical properties.
Collapse
|
355
|
Skog M, Muhonen V, Nystedt J, Narcisi R, Kontturi LS, Urtti A, Korhonen M, van Osch GJVM, Kiviranta I. Xeno-free chondrogenesis of bone marrow mesenchymal stromal cells: towards clinical-grade chondrocyte production. Cytotechnology 2014; 67:905-19. [PMID: 24718835 DOI: 10.1007/s10616-014-9721-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 03/20/2014] [Indexed: 01/02/2023] Open
Abstract
Current cell-based cartilage therapies relay on articular cartilage-derived autologous chondrocytes as a cell source, which possesses disadvantages, such as, donor site morbidity and dedifferentiation of chondrocytes during in vitro expansion. Due to these and other limitations, novel cell sources and production strategies are needed. Bone marrow-derived mesenchymal stromal cells (BM-MSCs) are a fascinating alternative, but they are not spontaneously capable of producing hyaline cartilage-like repair tissue in vivo. In vitro pre-differentiation of BM-MSCs could be used to produce chondrocytes for clinical applications. However, clinically compatible defined and xeno-free differentiation protocol is lacking. Hence, this study aimed to develop such chondrogenic differentiation medium for human BM-MSCs. We assessed the feasibility of the medium using three human BM-MSCs donors and validated the method by comparing BM-MSCs to three other cell types holding potential for articular cartilage repair. The effectiveness of the method was compared to conventional serum-free and commercially available chondrogenic differentiation media. The results show that the defined xeno-free differentiation medium is at least as efficient as conventionally used serum-free chondrogenic medium and performed significantly better on all cell types tested compared to the commercially available chondrogenic medium.
Collapse
Affiliation(s)
- Maria Skog
- Department of Surgery, Institute of Clinical Medicine, University of Helsinki, Biomedicum Helsinki, Haartmaninkatu 8, 00290, Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
356
|
Potier E, Rivron NC, Van Blitterswijk CA, Ito K. Micro-aggregates do not influence bone marrow stromal cell chondrogenesis. J Tissue Eng Regen Med 2014; 10:1021-1032. [PMID: 24700552 DOI: 10.1002/term.1887] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 11/04/2013] [Accepted: 02/24/2014] [Indexed: 12/31/2022]
Abstract
Although bone marrow stromal cells (BMSCs) appear promising for cartilage repair, current clinical results are suboptimal and the success of BMSC-based therapies relies on a number of methodological improvements, among which is better understanding and control of their differentiation pathways. We investigated here the role of the cellular environment (paracrine vs juxtacrine signalling) in the chondrogenic differentiation of BMSCs. Bovine BMSCs were encapsulated in alginate beads, as dispersed cells or as small micro-aggregates, to create different paracrine and juxtacrine signalling conditions. BMSCs were then cultured for 21 days with TGFβ3 added for 0, 7 or 21 days. Chondrogenic differentiation was assessed at the gene (type II and X collagens, aggrecan, TGFβ, sp7) and matrix (biochemical assays and histology) levels. The results showed that micro-aggregates had no beneficial effects over dispersed cells: matrix production was similar, whereas chondrogenic marker gene expression was lower for the micro-aggregates, under all TGFβ conditions tested. This weakened chondrogenic differentiation might be explained by a different cytoskeleton organization at day 0 in the micro-aggregates. Copyright © 2014 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- E Potier
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - N C Rivron
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - C A Van Blitterswijk
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - K Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
357
|
Mesenchymal stem cells, neural lineage potential, heparan sulfate proteoglycans and the matrix. Dev Biol 2014; 388:1-10. [DOI: 10.1016/j.ydbio.2014.01.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/08/2014] [Accepted: 01/30/2014] [Indexed: 12/23/2022]
|
358
|
Ansboro S, Hayes JS, Barron V, Browne S, Howard L, Greiser U, Lalor P, Shannon F, Barry FP, Pandit A, Murphy JM. A chondromimetic microsphere for in situ spatially controlled chondrogenic differentiation of human mesenchymal stem cells. J Control Release 2014; 179:42-51. [DOI: 10.1016/j.jconrel.2014.01.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 01/19/2014] [Accepted: 01/22/2014] [Indexed: 12/20/2022]
|
359
|
Trujillo NA, Popat KC. Increased Adipogenic and Decreased Chondrogenic Differentiation of Adipose Derived Stem Cells on Nanowire Surfaces. MATERIALS 2014; 7:2605-2630. [PMID: 28788586 PMCID: PMC5453350 DOI: 10.3390/ma7042605] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 12/14/2022]
Abstract
Despite many advances in tissue engineering, there are still significant challenges associated with restructuring, repairing, or replacing damaged tissue in the body. Currently, a major obstacle has been trying to develop a scaffold for cartilage tissue engineering that provides the correct mechanical properties to endure the loads associated with articular joints as well as promote cell-scaffold interactions to aid in extracellular matrix deposition. In addition, adipogenic tissue engineering is widely growing due to an increased need for more innovative reconstructive therapies following adipose tissue traumas and cosmetic surgeries. Recently, lipoaspirate tissue has been identified as a viable alternative source for mesenchymal stem cells because it contains a supportive stroma that can easily be isolated. Adipose derived stem cells (ADSCs) can differentiate into a variety of mesodermal lineages including the adipogenic and chondrogenic phenotypes. Biodegradable polymeric scaffolds have been shown to be a promising alternative and stem cells have been widely used to evaluate the compatibility, viability, and bioactivity of these materials. Polycaprolactone is a bioresorbable polymer, which has been widely used for biomedical and tissue engineering applications. The fundamental concept behind successful synthetic tissue-engineered scaffolds is to promote progenitor cell migration, adhesion, proliferation, and induce differentiation, extracellular matrix synthesis, and finally integration with host tissue. In this study, we investigated the adhesion, proliferation, and chondrogenic and adipogenic differentiation of ADSCs on nanowire surfaces. A solvent-free gravimetric template technique was used to fabricate polycaprolactone nanowires surfaces. The results indicated that during the growth period i.e., initial 7 days of culture, the nanowire surfaces (NW) supported adhesion and proliferation of the cells that had elongated morphologies. However, cell on surfaces without nanowires had non-elongated morphologies. Further, immunofluorescence imaging of marker proteins showed that the nanowires surfaces did not appear to support chondrogenic differentiation whereas supported adipogenic differentiation of ADSCs.
Collapse
Affiliation(s)
- Nathan A Trujillo
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA.
| | - Ketul C Popat
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
360
|
Archambeault DR, Yao HHC. Loss of smad4 in Sertoli and Leydig cells leads to testicular dysgenesis and hemorrhagic tumor formation in mice. Biol Reprod 2014; 90:62. [PMID: 24501173 DOI: 10.1095/biolreprod.113.111393] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
As the central component of canonical TGFbeta superfamily signaling, SMAD4 is a critical regulator of organ development, patterning, tumorigenesis, and many other biological processes. Because numerous TGFbeta superfamily ligands are expressed in developing testes, there may exist specific requirements for SMAD4 in individual testicular cell types. Previously, we reported that expansion of the fetal testis cords requires expression of SMAD4 by the Sertoli cell lineage. To further uncover the role of Smad4 in murine testes, we produced conditional knockout mice lacking Smad4 in either Leydig cells or in both Sertoli and Leydig cells simultaneously. Loss of Smad4 concomitantly in Sertoli and Leydig cells led to underdevelopment of the testis cords during fetal life and mild testicular dysgenesis in young adulthood (decreased testis size, partially dysgenic seminiferous tubules, and low sperm production). When the Sertoli/Leydig cell Smad4 conditional knockout mice aged (56- to 62-wk old), the testis phenotypes became exacerbated with the appearance of hemorrhagic tumors, Leydig cell adenomas, and a complete loss of spermatogenesis. In contrast, loss of Smad4 in Leydig cells alone did not appreciably alter fetal and adult testis development. Our findings support a cell type-specific requirement of Smad4 in testis development and suppression of testicular tumors.
Collapse
|
361
|
Chen J, Li C, Wang S. Periodic heat shock accelerated the chondrogenic differentiation of human mesenchymal stem cells in pellet culture. PLoS One 2014; 9:e91561. [PMID: 24632670 PMCID: PMC3954764 DOI: 10.1371/journal.pone.0091561] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/12/2014] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is one of diseases that seriously affect elderly people's quality of life. Human mesenchymal stem cells (hMSCs) offer a potential promise for the joint repair in OA patients. However, chondrogenic differentiation from hMSCs in vitro takes a long time (∼6 weeks) and differentiated cells are still not as functionally mature as primary isolated chondrocytes, though chemical stimulations and mechanical loading have been intensively studied to enhance the hMSC differentiation. On the other hand, thermal stimulations of hMSC chondrogenesis have not been well explored. In this study, the direct effects of mild heat shock (HS) on the differentiation of hMSCs into chondrocytes in 3D pellet culture were investigated. Periodic HS at 41°C for 1 hr significantly increased sulfated glycosaminoglycan in 3D pellet culture at Day 10 of chondrogenesis. Immunohistochemical and Western Blot analyses revealed an increased expression of collagen type II and aggrecan in heat-shocked pellets than non heat-shocked pellets on Day 17 of chondrogenesis. In addition, HS also upregulated the expression of collagen type I and X as well as heat shock protein 70 on Day 17 and 24 of differentiation. These results demonstrate that HS accelerated the chondrogenic differentiation of hMSCs and induced an early maturation of chondrocytes differentiated from hMSCs. The results of this study will guide the design of future protocols using thermal treatments to facilitate cartilage regeneration with human mesenchymal stem cells.
Collapse
Affiliation(s)
- Jing Chen
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
| | - Chenghai Li
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
| | - Sihong Wang
- Department of Biomedical Engineering, The City College of the City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
362
|
Spina J, Warnock J, Duesterdieck-Zellmer K, Baltzer W, Ott J, Bay B. Comparison of growth factor treatments on the fibrochondrogenic potential of canine fibroblast-like synoviocytes for meniscal tissue engineering. Vet Surg 2014; 43:750-60. [PMID: 24617289 DOI: 10.1111/j.1532-950x.2014.12170.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 06/01/2013] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To determine the in vitro effects of differing growth factor treatments on the fibrochondrogenic potential of fibroblast-like synoviocytes from cruciate ligament deficient femorotibial joints of dogs. STUDY DESIGN In vitro study. SAMPLE POPULATION Synoviocytes from dogs (n = 8) with naturally occurring cruciate ligament insufficiency. METHODS Synoviocytes were cultured in monolayer and synthesized into tensioned synoviocyte bioscaffolds (TSB) suspended in media containing TGF-β3, or FGF-2, TGF-β1, and IGF-I. The 1,9-dimethylmethylene blue (DMMB) assay and toluidine blue stain assessed glycosaminoglycan content; hydroxyproline assay, and collagen I and II immunohistochemistry assessed collagen content. Biomechanical properties were determined by materials testing/force-deformation curves. RESULTS All tissue cultures formed tensioned fibrous tissue-like constructs. Mean tissue cellularity and cellular viability was significantly greater in the triple growth factor-treated TSB by 0.09% and 44%, respectively. Percentage collagen content, and relative gene expression for collagen I, II, and aggrecan was not significantly different between groups. Median percentage of GAG content was significantly greater in triple growth factor-treated TSB by 1.6%. Biomechanical properties were not different in compression. Triple growth factor-treated TSB were significantly stronger in toughness, peak load to failure, and stiffness in tension. CONCLUSIONS TGF-β3 cultured bioscaffolds failed to outperform triple growth factor-treated TSB. Architectural extracellular matrix (ECM) organization and cellularity likely explained the differences between groups. TGF-β3 alone cannot be recommended at this time for in vitro formation of autologous fibrocartilage bioscaffolds for meniscal deficiency.
Collapse
Affiliation(s)
- Jason Spina
- Department of Clinical Sciences, College of Veterinary Medicine, Corvallis, Oregon
| | | | | | | | | | | |
Collapse
|
363
|
Scaffold-mediated lentiviral transduction for functional tissue engineering of cartilage. Proc Natl Acad Sci U S A 2014; 111:E798-806. [PMID: 24550481 DOI: 10.1073/pnas.1321744111] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The ability to develop tissue constructs with matrix composition and biomechanical properties that promote rapid tissue repair or regeneration remains an enduring challenge in musculoskeletal engineering. Current approaches require extensive cell manipulation ex vivo, using exogenous growth factors to drive tissue-specific differentiation, matrix accumulation, and mechanical properties, thus limiting their potential clinical utility. The ability to induce and maintain differentiation of stem cells in situ could bypass these steps and enhance the success of engineering approaches for tissue regeneration. The goal of this study was to generate a self-contained bioactive scaffold capable of mediating stem cell differentiation and formation of a cartilaginous extracellular matrix (ECM) using a lentivirus-based method. We first showed that poly-L-lysine could immobilize lentivirus to poly(ε-caprolactone) films and facilitate human mesenchymal stem cell (hMSC) transduction. We then demonstrated that scaffold-mediated gene delivery of transforming growth factor β3 (TGF-β3), using a 3D woven poly(ε-caprolactone) scaffold, induced robust cartilaginous ECM formation by hMSCs. Chondrogenesis induced by scaffold-mediated gene delivery was as effective as traditional differentiation protocols involving medium supplementation with TGF-β3, as assessed by gene expression, biochemical, and biomechanical analyses. Using lentiviral vectors immobilized on a biomechanically functional scaffold, we have developed a system to achieve sustained transgene expression and ECM formation by hMSCs. This method opens new avenues in the development of bioactive implants that circumvent the need for ex vivo tissue generation by enabling the long-term goal of in situ tissue engineering.
Collapse
|
364
|
Mhanna R, Öztürk E, Vallmajo-Martin Q, Millan C, Müller M, Zenobi-Wong M. GFOGER-modified MMP-sensitive polyethylene glycol hydrogels induce chondrogenic differentiation of human mesenchymal stem cells. Tissue Eng Part A 2014; 20:1165-74. [PMID: 24134736 DOI: 10.1089/ten.tea.2013.0519] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The cellular microenvironment plays a crucial role in directing proliferation and differentiation of stem cells. Cells interact with their microenvironment via integrins that recognize certain peptide sequences of extracellular matrix proteins. This receptor-ligand binding has profound impact on cell fate. Interactions of human bone marrow mesenchymal stem cells (hMSCs) with the triple helical collagen mimetic, GPC(GPP)5-GFOGER-(GPP)5GPC-NH2, and the fibronectin adhesion peptide, RGD, were studied in degradable or nondegradable polyethylene glycol (PEG) gels formed by Michael-type addition chemistry. Proliferation, cytoskeletal morphology, and chondrogenic differentiation of encapsulated hMSCs were evaluated. The hMSCs adopted a highly spread morphology within the GFOGER-modified gels, whereas RGD induced a star-like spreading of the cells. hMSCs within GFOGER-modified degradable gels had a high proliferation rate compared with cells in peptide-free gels (p=0.017). Gene expression of type II collagen was highest in GFOGER-modified degradable gels after 21 days. Peptide incorporation increased GAG production in degradable gels after 7 and 21 days and GFOGER-modified degradable hydrogels had on average the highest GAG content, a finding that was confirmed by Alcian blue staining. In conclusion, the GFOGER peptide enhances proliferation in degradable PEG gels and provides a better chondrogenic microenvironment compared with the RGD peptide.
Collapse
Affiliation(s)
- Rami Mhanna
- Cartilage Engineering+Regeneration Laboratory , ETH Zürich, Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
365
|
Osawa A, Harner CD, Gharaibeh B, Matsumoto T, Mifune Y, Kopf S, Ingham SJM, Schreiber V, Usas A, Huard J. The use of blood vessel-derived stem cells for meniscal regeneration and repair. Med Sci Sports Exerc 2014; 45:813-23. [PMID: 23247715 DOI: 10.1249/mss.0b013e31827d1e06] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Surgical repairs of tears in the vascular region of the meniscus usually heal better than repairs performed in the avascular region; thus, we hypothesized that this region might possess a richer supply of vascular-derived stem cells than the avascular region. METHODS In this study, we analyzed 6 menisci extracted from aborted human fetuses and 12 human lateral menisci extracted from adult human subjects undergoing total knee arthroplasty. Menisci were immunostained for CD34 (a stem cell marker) and CD146 (a pericyte marker) in situ, whereas other menisci were dissected into two regions (peripheral and inner) and used to isolate meniscus-derived cells by flow cytometry. Cell populations expressing CD34 and CD146 were tested for their multilineage differentiation potentials, including chondrogenic, osteogenic, and adipogenic lineages. Fetal peripheral meniscus cells were transplanted by intracapsular injection into the knee joints of an athymic rat meniscal tear model. Rat menisci were extracted and histologically evaluated after 4 wk posttransplantation. RESULTS Immunohistochemistry and flow cytometric analyses demonstrated that a higher number of CD34- and CD146-positive cells were found in the peripheral region compared with the inner region. The CD34- and CD146-positive cells isolated from the vascular region of both fetal and adult menisci demonstrated multilineage differentiation capacities and were more potent than cells isolated from the inner (avascular) region. Fetal CD34- and CD146-positive cells transplanted into the athymic rat knee joint were recruited into the meniscal tear sites and contributed to meniscus repair. CONCLUSIONS The vascularized region of the meniscus contains more stem cells than the avascular region. These meniscal-derived stem cells were multipotent and contributed to meniscal regeneration.
Collapse
Affiliation(s)
- Aki Osawa
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
366
|
Daley ELH, Alford AI, Miller JD, Goldstein SA. Phenotypic differences in white-tailed deer antlerogenic progenitor cells and marrow-derived mesenchymal stromal cells. Tissue Eng Part A 2014; 20:1416-25. [PMID: 24313802 DOI: 10.1089/ten.tea.2013.0420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Deer antlers are bony appendages that are annually cast and rapidly regrown in a seasonal process coupled to the reproductive cycle. Due to the uniqueness of this process among mammals, we reasoned that a fundamental characterization of antler progenitor cell behavior may provide insights that could lead to improved strategies for promoting bone repair. In this study, we investigated whether white-tailed deer antlerogenic progenitor cells (APC) conform to basic criteria defining mesenchymal stromal cells (MSC). In addition, we tested the effects of the artificial glucocorticoid dexamethasone (DEX) on osteogenic and chondrogenic differentiation as well as the degree of apoptosis during the latter. Comparisons were made to animal-matched marrow-derived MSC. APC and MSC generated similar numbers of colonies. APC cultures expanded less rapidly overall but experienced population recovery at later time points. In contrast to MSC, APC did not display adipogenic in vitro differentiation capacity. Under osteogenic culture conditions, APC and MSC exhibited different patterns of alkaline phosphatase activity over time. DEX increased APC alkaline phosphatase activity only initially but consistently led to decreased activity in MSC. APC and MSC in osteogenic culture underwent different time and DEX-dependent patterns of mineralization, yet APC and MSC achieved similar levels of mineral accrual in an ectopic ossicle model. During chondrogenic differentiation, APC exhibited high levels of apoptosis without a reduction in cell density. DEX decreased proteoglycan production and increased apoptosis in chondrogenic APC cultures but had the opposite effects in MSC. Our results suggest that APC and MSC proliferation and differentiation differ in their dependence on time, factors, and milieu. Antler tip APC may be more lineage-restricted osteo/chondroprogenitors with distinctly different responses to apoptotic and glucocorticoid stimuli.
Collapse
Affiliation(s)
- Ethan L H Daley
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | | | | | | |
Collapse
|
367
|
Orth P, Rey-Rico A, Venkatesan JK, Madry H, Cucchiarini M. Current perspectives in stem cell research for knee cartilage repair. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2014; 7:1-17. [PMID: 24520197 PMCID: PMC3897321 DOI: 10.2147/sccaa.s42880] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protocols based on the delivery of stem cells are currently applied in patients, showing encouraging results for the treatment of articular cartilage lesions (focal defects, osteoarthritis). Yet, restoration of a fully functional cartilage surface (native structural organization and mechanical functions) especially in the knee joint has not been reported to date, showing the need for improved designs of clinical trials. Various sources of progenitor cells are now available, originating from adult tissues but also from embryonic or reprogrammed tissues, most of which have already been evaluated for their chondrogenic potential in culture and for their reparative properties in vivo upon implantation in relevant animal models of cartilage lesions. Nevertheless, particular attention will be needed regarding their safe clinical use and their potential to form a cartilaginous repair tissue of proper quality and functionality in the patient. Possible improvements may reside in the use of biological supplements in accordance with regulations, while some challenges remain in establishing standardized, effective procedures in the clinics.
Collapse
Affiliation(s)
- Patrick Orth
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany
| | - Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Jagadeesh K Venkatesan
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Henning Madry
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg, Germany ; Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
368
|
Davey GC, Patil SB, O'Loughlin A, O'Brien T. Mesenchymal stem cell-based treatment for microvascular and secondary complications of diabetes mellitus. Front Endocrinol (Lausanne) 2014; 5:86. [PMID: 24936198 PMCID: PMC4047679 DOI: 10.3389/fendo.2014.00086] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022] Open
Abstract
The worldwide increase in the prevalence of Diabetes mellitus (DM) has highlighted the need for increased research efforts into treatment options for both the disease itself and its associated complications. In recent years, mesenchymal stromal cells (MSCs) have been highlighted as a new emerging regenerative therapy due to their multipotency but also due to their paracrine secretion of angiogenic factors, cytokines, and immunomodulatory substances. This review focuses on the potential use of MSCs as a regenerative medicine in microvascular and secondary complications of DM and will discuss the challenges and future prospects of MSCs as a regenerative therapy in this field. MSCs are believed to have an important role in tissue repair. Evidence in recent years has demonstrated that MSCs have potent immunomodulatory functions resulting in active suppression of various components of the host immune response. MSCs may also have glucose lowering properties providing another attractive and unique feature of this therapeutic approach. Through a combination of the above characteristics, MSCs have been shown to exert beneficial effects in pre-clinical models of diabetic complications prompting initial clinical studies in diabetic wound healing and nephropathy. Challenges that remain in the clinical translation of MSC therapy include issues of MSC heterogeneity, optimal mode of cell delivery, homing of these cells to tissues of interest with high efficiency, clinically meaningful engraftment, and challenges with cell manufacture. An issue of added importance is whether an autologous or allogeneic approach will be used. In summary, MSC administration has significant potential in the treatment of diabetic microvascular and secondary complications but challenges remain in terms of engraftment, persistence, tissue targeting, and cell manufacture.
Collapse
Affiliation(s)
- Grace C Davey
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland
| | - Swapnil B Patil
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland
| | - Aonghus O'Loughlin
- Department of Medicine, Galway University Hospital (GUH) , Galway , Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI) and Biosciences Building, National University of Ireland , Galway , Ireland ; Department of Medicine, Galway University Hospital (GUH) , Galway , Ireland
| |
Collapse
|
369
|
Carroll SF, Buckley CT, Kelly DJ. Cyclic hydrostatic pressure promotes a stable cartilage phenotype and enhances the functional development of cartilaginous grafts engineered using multipotent stromal cells isolated from bone marrow and infrapatellar fat pad. J Biomech 2013; 47:2115-21. [PMID: 24377681 DOI: 10.1016/j.jbiomech.2013.12.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/02/2013] [Accepted: 12/06/2013] [Indexed: 01/14/2023]
Abstract
The objective of this study was to investigate how joint specific biomechanical loading influences the functional development and phenotypic stability of cartilage grafts engineered in vitro using stem/progenitor cells isolated from different source tissues. Porcine bone marrow derived multipotent stromal cells (BMSCs) and infrapatellar fat pad derived multipotent stromal cells (FPSCs) were seeded in agarose hydrogels and cultured in chondrogenic medium, while simultaneously subjected to 10MPa of cyclic hydrostatic pressure (HP). To mimic the endochondral phenotype observed in vivo with cartilaginous tissues engineered using BMSCs, the culture media was additionally supplemented with hypertrophic factors, while the loss of phenotype observed in vivo with FPSCs was induced by withdrawing transforming growth factor (TGF)-β3 from the media. The application of HP was found to enhance the functional development of cartilaginous tissues engineered using both BMSCs and FPSCs. In addition, HP was found to suppress calcification of tissues engineered using BMSCs cultured in chondrogenic conditions and acted to maintain a chondrogenic phenotype in cartilaginous grafts engineered using FPSCs. The results of this study point to the importance of in vivo specific mechanical cues for determining the terminal phenotype of chondrogenically primed multipotent stromal cells. Furthermore, demonstrating that stem or progenitor cells will appropriately differentiate in response to such biophysical cues might also be considered as an additional functional assay for evaluating their therapeutic potential.
Collapse
Affiliation(s)
- S F Carroll
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - C T Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
| | - D J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and BioEngineering Research (AMBER) Centre, Trinity College Dublin, Ireland.
| |
Collapse
|
370
|
Lee JM, Kim JD, Oh EJ, Oh SH, Lee JH, Im GI. PD98059-impregnated functional PLGA scaffold for direct tissue engineering promotes chondrogenesis and prevents hypertrophy from mesenchymal stem cells. Tissue Eng Part A 2013; 20:982-91. [PMID: 24188591 DOI: 10.1089/ten.tea.2013.0290] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In cartilage tissue engineering from mesenchymal stem cells, it is important to suppress hypertrophy to produce a neocartilage with stable phenotypes of hyaline articular cartilage (AC). The aim of this study was to develop and test the usefulness of functional chondrogenic scaffolds that serve the purpose of hypertrophy suppression. PD98059-impregnated poly(lactic-co-glycolic acid) (PLGA) scaffold is fabricated and compared with transforming growth factor (TGF)-β2-immobilized scaffold. The PD98059 is continuously released from the scaffolds over 140 days in contrast to the rapid release in TGF-β2-immobilized scaffold. The in vitro culture results show that the PD98059-impregated scaffold is more effective in suppressing hypertrophy than the TGF-β2-immobilized scaffold while both scaffolds enhance chondrogenesis from human mesenchymal stem cells. After 10 weeks of in vivo implantation in rabbits, the osteochondral defects is successfully repaired in both PD98059-impregnated and TGF-β2-immobilized scaffold seeded with rabbit mesenchymal stem cells when evaluated grossly and microscopically. However, type X collagen is not observed from regenerated cartilage in PD98059-impregnated scaffold, whereas it is detected around chondrocytes in the TGF-β2-impregnated scaffolds. In addition, the PD98059-impregnated scaffold has better reconstitution of the subchondral plate. These results suggest that the use of the PD98059-impregnated scaffold leads to AC regeneration of better quality and prevents hypertrophy when implanted in the osteochondral defects.
Collapse
Affiliation(s)
- Jong Min Lee
- 1 Department of Orthopaedics, Dongguk University Ilsan Hospital , Goyang, Republic of Korea
| | | | | | | | | | | |
Collapse
|
371
|
Brady K, Dickinson SC, Guillot PV, Polak J, Blom AW, Kafienah W, Hollander AP. Human fetal and adult bone marrow-derived mesenchymal stem cells use different signaling pathways for the initiation of chondrogenesis. Stem Cells Dev 2013; 23:541-54. [PMID: 24172175 DOI: 10.1089/scd.2013.0301] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cartilage injuries and osteoarthritis are leading causes of disability in developed countries. The regeneration of damaged articular cartilage using cell transplantation or tissue engineering holds much promise but requires the identification of an appropriate cell source with a high proliferative propensity and consistent chondrogenic capacity. Human fetal mesenchymal stem cells (MSCs) have been isolated from a range of perinatal tissues, including first-trimester bone marrow, and have demonstrated enhanced expansion and differentiation potential. However, their ability to form mature chondrocytes for use in cartilage tissue engineering has not been clearly established. Here, we compare the chondrogenic potential of human MSCs isolated from fetal and adult bone marrow and show distinct differences in their responsiveness to specific growth factors. Transforming growth factor beta 3 (TGFβ3) induced chondrogenesis in adult but not fetal MSCs. In contrast, bone morphogenetic protein 2 (BMP2) induced chondrogenesis in fetal but not adult MSCs. When fetal MSCs co-stimulated with BMP2 and TGFβ3 were used for cartilage tissue engineering, they generated tissue with type II collagen and proteoglycan content comparable to adult MSCs treated with TGFβ3 alone. Investigation of the TGFβ/BMP signaling pathway showed that TGFβ3 induced phosphorylation of SMAD3 in adult but not fetal MSCs. These findings demonstrate that the initiation of chondrogenesis is modulated by distinct signaling mechanisms in fetal and adult MSCs. This study establishes the feasibility of using fetal MSCs in cartilage repair applications and proposes their potential as an in vitro system for modeling chondrogenic differentiation and skeletal development studies.
Collapse
Affiliation(s)
- Kyla Brady
- 1 Faculty of Medical and Veterinary Sciences, School of Cellular and Molecular Medicine, University of Bristol , Bristol, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
372
|
Kuroda Y, Dezawa M. Mesenchymal stem cells and their subpopulation, pluripotent muse cells, in basic research and regenerative medicine. Anat Rec (Hoboken) 2013; 297:98-110. [PMID: 24293378 DOI: 10.1002/ar.22798] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have gained a great deal of attention for regenerative medicine because they can be obtained from easy accessible mesenchymal tissues, such as bone marrow, adipose tissue, and the umbilical cord, and have trophic and immunosuppressive effects to protect tissues. The most outstanding property of MSCs is their potential for differentiation into cells of all three germ layers. MSCs belong to the mesodermal lineage, but they are known to cross boundaries from mesodermal to ectodermal and endodermal lineages, and differentiate into a variety of cell types both in vitro and in vivo. Such behavior is exceptional for tissue stem cells. As observed with hematopoietic and neural stem cells, tissue stem cells usually generate cells that belong to the tissue in which they reside, and do not show triploblastic differentiation. However, the scientific basis for the broad multipotent differentiation of MSCs still remains an enigma. This review summarizes the properties of MSCs from representative mesenchymal tissues, including bone marrow, adipose tissue, and the umbilical cord, to demonstrate their similarities and differences. Finally, we introduce a novel type of pluripotent stem cell, multilineage-differentiating stress-enduring (Muse) cells, a small subpopulation of MSCs, which can explain the broad spectrum of differentiation ability in MSCs.
Collapse
Affiliation(s)
- Yasumasa Kuroda
- Department of Anatomy and Anthropology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | |
Collapse
|
373
|
Isolation and characterization of mesenchymal stem cells derived from fetal bovine liver. Cell Tissue Bank 2013; 15:439-50. [DOI: 10.1007/s10561-013-9410-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 11/15/2013] [Indexed: 01/22/2023]
|
374
|
The degradation of chondrogenic pellets using cocultures of synovial fibroblasts and U937 cells. Biomaterials 2013; 35:1185-91. [PMID: 24225084 DOI: 10.1016/j.biomaterials.2013.10.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 10/19/2013] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) of the knee is often characterized by joint space narrowing on X-ray, knee pain, and a loss of joint function through progressive cartilage degradation and intermittent synovial inflammation. The objective of this work was to develop an in vitro model in a clinically relevant system. Normal human synovial fibroblasts were cultured with U937 cells for 3 days then combined with a chondrogenic stem cell pellet for another 4 days. This culture system mimicked many of the aspects of early stage OA including production of cytokines and degradative enzymes, MMP-1 and MMP-3, resulting in a conditioned medium profile similar to OA synovial fluid. This catabolic environment resulted in the release of glycosaminoglycan (GAG) from the pellet. In a similar manner to early stage OA, the pellet had increased aggrecan and collagen II expression. All of these effects are hallmarks of early stage OA. This relatively simple tissue model containing a 3D cartilage component interacting with synoviocytes and macrophages could be useful to understand early causes and progression of OA. It can be scaled easily thus useful for high throughput screening of disease modifying drugs in a clinically relevant system.
Collapse
|
375
|
Leferink AM, Hendrikson WJ, Rouwkema J, Karperien M, van Blitterswijk CA, Moroni L. Increased cell seeding efficiency in bioplotted three-dimensional PEOT/PBT scaffolds. J Tissue Eng Regen Med 2013; 10:679-89. [PMID: 24668928 DOI: 10.1002/term.1842] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 08/20/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022]
Abstract
In regenerative medicine studies, cell seeding efficiency is not only optimized by changing the chemistry of the biomaterials used as cell culture substrates, but also by altering scaffold geometry, culture and seeding conditions. In this study, the importance of seeding parameters, such as initial cell number, seeding volume, seeding concentration and seeding condition is shown. Human mesenchymal stem cells (hMSCs) were seeded into cylindrically shaped 4 × 3 mm polymeric scaffolds, fabricated by fused deposition modelling. The initial cell number ranged from 5 × 10(4) to 8 × 10(5) cells, in volumes varying from 50 µl to 400 µl. To study the effect of seeding conditions, a dynamic system, by means of an agitation plate, was compared with static culture for both scaffolds placed in a well plate or in a confined agarose moulded well. Cell seeding efficiency decreased when seeded with high initial cell numbers, whereas 2 × 10(5) cells seemed to be an optimal initial cell number in the scaffolds used here. The influence of seeding volume was shown to be dependent on the initial cell number used. By optimizing seeding parameters for each specific culture system, a more efficient use of donor cells can be achieved. Copyright © 2013 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- A M Leferink
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - W J Hendrikson
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - J Rouwkema
- Laboratory of Biomechanical Engineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - M Karperien
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands.,Department of Developmental Bioengineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - C A van Blitterswijk
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| | - L Moroni
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands
| |
Collapse
|
376
|
Ma S, Xie N, Li W, Yuan B, Shi Y, Wang Y. Immunobiology of mesenchymal stem cells. Cell Death Differ 2013; 21:216-25. [PMID: 24185619 PMCID: PMC3890955 DOI: 10.1038/cdd.2013.158] [Citation(s) in RCA: 570] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be isolated from almost all tissues and effectively expanded in vitro. Although their true in situ properties and biological functions remain to be elucidated, these in vitro expanded cells have been shown to possess potential to differentiate into specific cell lineages. It is speculated that MSCs in situ have important roles in tissue cellular homeostasis by replacing dead or dysfunctional cells. Recent studies have demonstrated that in vitro expanded MSCs of various origins have great capacity to modulate immune responses and change the progression of different inflammatory diseases. As tissue injuries are often accompanied by inflammation, inflammatory factors may provide cues to mobilize MSCs to tissue sites with damage. Before carrying out tissue repair functions, MSCs first prepare the microenvironment by modulating inflammatory processes and releasing various growth factors in response to the inflammation status. In this review, we focus on the crosstalk between MSCs and immune responses and their potential clinical applications, especially in inflammatory diseases.
Collapse
Affiliation(s)
- S Ma
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - N Xie
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - W Li
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| | - B Yuan
- National Institutes for Food and Drug Control, No. 2 Tiantan Xili, Beijing 100050, China
| | - Y Shi
- 1] Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China [2] Child Health Institute of New Jersey, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, New Jersey 08901, USA
| | - Y Wang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences of Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, 225 South Chongqing Road, Shanghai 200025, China
| |
Collapse
|
377
|
Kang SH, Chung YG, Oh IH, Kim YS, Min KO, Chung JY. Bone regeneration potential of allogeneic or autogeneic mesenchymal stem cells loaded onto cancellous bone granules in a rabbit radial defect model. Cell Tissue Res 2013; 355:81-8. [PMID: 24169864 DOI: 10.1007/s00441-013-1738-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 09/25/2013] [Indexed: 10/26/2022]
Abstract
For developing a clinically effective bone regeneration strategy, we compare the bone regeneration potential of cultured allogeneic bone marrow-derived mesenchymal stem cells (BM-MSCs) and of autologous BM-MSCs loaded onto allogeneic cancellous bone granule scaffolds. A critical-sized segmental bone defect was made at the mid-shaft of both radiuses in 19 New Zealand White rabbits (NWRs). In the experimental group, allogeneic BM-MSCs loaded onto small-sized allogeneic cancellous bone granules (300~700 um in diameter) were implanted in one side of a bone defect. In the control group, autologous BM-MSCs loaded onto allogeneic cancellous granules were grafted in the other side. Bone regeneration was assessed by radiographic evaluation at 4, 8, 12 and 16 weeks post-implantation and by micro-computed tomography (micro-CT) and histological evaluation at 8 and 16 weeks. The experimental groups showed lower bone quantity indices (BQIs) than the control groups at 12 and 16 weeks (p < 0.05), although no significant difference was observed at 4 and 8 weeks (p > 0.05). Micro-CT analysis revealed that both groups had similar mean total bone volume and other parameters including trabecular thickness, number and separation at either 8 or 16 weeks. Only bone surface area revealed less area in the experimental group at 16 weeks. Histological evaluation of 8-week and 16-week specimens showed similar biologic processes of new bone formation and maturation. There was no inflammatory reaction indicating an adverse immune response in both allogeneic and autologous MSC groups. In conclusion, allogeneic BM-MSCs loaded onto allogeneic cancellous bone granules had comparable bone regeneration potential to autologous BM-MSCs in a rabbit radial defect model.
Collapse
Affiliation(s)
- Soo-Hwan Kang
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | | | | | | | | | | |
Collapse
|
378
|
Nagel S, Rohr F, Weber C, Kier J, Siemers F, Kruse C, Danner S, Brandenburger M, Matthiessen AE. Multipotent nestin-positive stem cells reside in the stroma of human eccrine and apocrine sweat glands and can be propagated robustly in vitro. PLoS One 2013; 8:e78365. [PMID: 24205211 PMCID: PMC3813437 DOI: 10.1371/journal.pone.0078365] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/18/2013] [Indexed: 01/24/2023] Open
Abstract
Human skin harbours multiple different stem cell populations. In contrast to the relatively well-characterized niches of epidermal and hair follicle stem cells, the localization and niches of stem cells in other human skin compartments are as yet insufficiently investigated. Previously, we had shown in a pilot study that human sweat gland stroma contains Nestin-positive stem cells. Isolated sweat gland stroma-derived stem cells (SGSCs) proliferated in vitro and expressed Nestin in 80% of the cells. In this study, we were able to determine the precise localization of Nestin-positive cells in both eccrine and apocrine sweat glands of human axillary skin. We established a reproducible isolation procedure and characterized the spontaneous, long-lasting multipotent differentiation capacity of SGSCs. Thereby, a pronounced ectodermal differentiation was observed. Moreover, the secretion of prominent cytokines demonstrated the immunological potential of SGSCs. The comparison to human adult epidermal stem cells (EpiSCs) and bone marrow stem cells (BMSCs) revealed differences in protein expression and differentiation capacity. Furthermore, we found a coexpression of the stem cell markers Nestin and Iα6 within SGSCs and human sweat gland stroma. In conclusion the initial results of the pilot study were confirmed, indicating that human sweat glands are a new source of unique stem cells with multilineage differentiation potential, high proliferation capacity and remarkable self renewal. With regard to the easy accessibility of skin tissue biopsies, an autologous application of SGSCs in clinical therapies appears promising.
Collapse
Affiliation(s)
- Sabine Nagel
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Franziska Rohr
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Caroline Weber
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Janina Kier
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Frank Siemers
- Department of Plastic and Hand Surgery, University of Lübeck, Lübeck, Germany
| | - Charli Kruse
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | - Sandra Danner
- Fraunhofer Research Institution for Marine Biotechnology, Lübeck, Germany
| | | | | |
Collapse
|
379
|
Olee T, Grogan SP, Lotz MK, Colwell CW, D'Lima DD, Snyder EY. Repair of cartilage defects in arthritic tissue with differentiated human embryonic stem cells. Tissue Eng Part A 2013; 20:683-92. [PMID: 24028447 DOI: 10.1089/ten.tea.2012.0751] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chondrocytes have been generated in vitro from a range of progenitor cell types and by a number of strategies. However, achieving reconstitution of actual physiologically relevant, appropriately-laminated cartilage in situ that would be applicable to conditions, such as arthritis and cartilage degeneration remains elusive. This lack of success is multifactorial and includes limited cell source, decreased proliferation rate of mature chondrocytes, lack of maintenance of phenotype, reduced matrix synthesis, and poor integration with host tissue. We report an efficient approach for deriving mesenchymal chondroprogenitor cells from human embryonic stem cells. These cells generated tissue containing cartilage-specific matrix proteins that integrated in situ in a partial-thickness defect in ex vivo articular cartilage harvested from human arthritic joints. Given that stem cells provide a virtually inexhaustible supply of starting material and that our technique is easily scalable, cartilaginous tissue primed and grafted in this manner could be suitable for clinical translation.
Collapse
Affiliation(s)
- Tsaiwei Olee
- 1 Shiley Center for Orthopaedic Research and Education at Scripps Health , La Jolla, California
| | | | | | | | | | | |
Collapse
|
380
|
Zhang C, Zhai W, Xie Y, Chen Q, Zhu W, Sun X. Mesenchymal stem cells derived from breast cancer tissue promote the proliferation and migration of the MCF-7 cell line in vitro.. Oncol Lett 2013; 6:1577-1582. [PMID: 24260049 PMCID: PMC3833946 DOI: 10.3892/ol.2013.1619] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 09/25/2013] [Indexed: 01/11/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are critical in promoting cancer progression, including tumor growth and metastasis. MSCs, as a subpopulation of cells found in the tumor microenvironment, have been isolated from several tumor tissues, but have not been isolated from breast cancer tissue to date. Therefore, the purpose of this study was to isolate MSCs from primary human breast cancer tissue, and to study the effect of breast cancer MSCs (BC-MSCs) on the proliferation and migration of the MCF-7 cell line in vitro. MSCs were isolated and identified from primary breast cancer tissue obtained from 9 patients. The MCF-7 cell line was treated with 10 and 20% breast cancer-associated MSC (BC-MSC)-conditioned medium (CM) for 10–48 h, and changes in proliferation and migration were observed. Furthermore, we investigated the migration of 10 and 20% CM concentrations on MCF-7 through a scratch wound assay and a transwell migration assay. We successfully isolated and identified MSCs from primary breast cancer tissues. BC-MSCs showed characteristics similar to those of bone marrow MSCs, and possessed the capability of multipotential differentiation into osteoblasts and adipocytes. The results of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay showed that 10 and 20% CM concentrations increased the proliferation of MCF-7 cells to different levels. The results also revealed a greater increase in different levels compared with the control group. In conclusion, MSCs were confirmed to exist in human breast cancer tissues, and BC-MSCs may promote the proliferation and migration of breast cancer cells.
Collapse
Affiliation(s)
- Chunfu Zhang
- The Second People's Hospital of Kunshan, Kunshan, Jiangsu 215300, P.R. China ; School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | | | | | | | | | | |
Collapse
|
381
|
Omes C, Fassina L, Van Vlierberghe S, Magenes G, Dubruel P, Vaghi P, Reguzzoni M, Riva F. A case of successful interaction between cells derived from human ovarian follicular liquid and gelatin cryogel for biotech and medical applications. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2013:6240-3. [PMID: 24111166 DOI: 10.1109/embc.2013.6610979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Significant research efforts have been undertaken in the last decade to develop specific cell-based therapies and, in particular, adult multipotent mesenchymal stem cells (MSCs) hold great promise toward such regenerative strategies. Bio-materials have been widely used in reconstructive bone surgery to heal critical-size bone defects due to trauma, tumor resection, and tissue degeneration. In particular, gelatin cryogel scaffolds are promising new biomaterials owing to their biocompatibility. There is an increasing demand for MSC-based regenerative approaches in the musculoskeletal system. Combining stem cells with biomaterial scaffolds provides a promising strategy for tissue engineering. Our previous studies showed the possibility to obtain MSCs from the human ovarian follicular liquid (FL) that is usually wasted during in vitro fertilization (IVF). In this study, we tested the ability of these FL cells to grow on gelatin cryogel in comparison with MSCs derived from human bone marrow. Samples and controls were analyzed with confocal and scanning electron microscopes. Results demonstrated that FL cells could grow on the biomaterial not only on the top but also in the layers below till 60 µm of deepness. Data suggested that the observed cells were mesenchymal since positive for vimentin and CD-44, typical MSC markers. Successful growth of putative MSCs derived from follicular liquid on 3D gelatin cryogel opens potential developments in biotech and medical applications.
Collapse
|
382
|
de Vries-van Melle ML, Narcisi R, Kops N, Koevoet WJLM, Bos PK, Murphy JM, Verhaar JAN, van der Kraan PM, van Osch GJVM. Chondrogenesis of mesenchymal stem cells in an osteochondral environment is mediated by the subchondral bone. Tissue Eng Part A 2013; 20:23-33. [PMID: 23980750 DOI: 10.1089/ten.tea.2013.0080] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In articular cartilage repair, cells that will be responsible for the formation of repair tissue are often exposed to an osteochondral environment. To study cartilage repair mechanisms in vitro, we have recently developed a bovine osteochondral biopsy culture model in which cartilage defects can be simulated reproducibly. Using this model, we now aimed at studying the chondrogenic potential of human bone marrow-derived mesenchymal stem cells (hBMSCs) in an osteochondral environment. In contrast to standard in vitro chondrogenesis, it was found that supplementing transforming growth factor beta (TGFβ) to culture medium was not required to induce chondrogenesis of hBMSCs in an osteochondral environment. hBMSC culture in defects created in osteochondral biopsies or in bone-only biopsies resulted in comparable levels of cartilage-related gene expression, whereas culture in cartilage-only biopsies did not induce chondrogenesis. Subcutaneous implantation in nude mice of osteochondral biopsies containing hBMSCs in osteochondral defects resulted in the formation of more cartilaginous tissue than hBMSCs in chondral defects. The subchondral bone secreted TGFβ; however, the observed results could not be attributed to TGFβ, as either capturing TGFβ with an antibody or blocking the canonical TGFβ signaling pathway did not result in significant changes in cartilage-related gene expression of hBMSCs in the osteochondral culture model. Inhibition of BMP signaling did not prevent chondrogenesis. In conclusion, we demonstrate that chondrogenesis of hBMSCs is induced by factors secreted from the bone. We have strong indications that this is not solely mediated by members of the TGFβ family but other, yet unknown, factors originating from the subchondral bone appeared to play a key role.
Collapse
|
383
|
Tay CY, Koh CG, Tan NS, Leong DT, Tan LP. Mechanoregulation of stem cell fate via micro-/nano-scale manipulation for regenerative medicine. Nanomedicine (Lond) 2013; 8:623-38. [PMID: 23560412 DOI: 10.2217/nnm.13.31] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Recent developments in the field of mechanobiology have renewed the call for a better understanding of the role of mechanical forces as potent regulators and indicators of stem cell fate. Although it is well established that mechanical forces play a crucial role in guiding tissue development, little is known about how submicroscopic biomechanical forces can influence key stem cell behaviors. This review will detail the use of micro-/nano-technologies that are advancing our current understanding of stem cell mechanobiology, and mechanoregulation of stem cell fate using engineered surface topographies and small-scale patterning techniques. The involvement of focal adhesions and the cytoskeleton systems as a common biophysical impetus through which these mechanical signals are transduced via distinct signaling pathways will also be discussed. These insights are envisioned to provide the basis for the rational design of future biocompatible materials and may inspire alternative drug-free therapeutic strategies to manage diseased sites via biomechanical management.
Collapse
Affiliation(s)
- Chor Yong Tay
- Department of Chemical & Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, 117576, Singapore
| | | | | | | | | |
Collapse
|
384
|
Cheng A, Hardingham TE, Kimber SJ. Generating cartilage repair from pluripotent stem cells. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:257-66. [PMID: 23957872 DOI: 10.1089/ten.teb.2012.0757] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The treatment of degeneration and injury of articular cartilage has been very challenging for scientists and surgeons. As an avascular and hypocellular tissue, cartilage has a very limited capacity for self-repair. Chondrocytes are the only cell type in cartilage, in which they are surrounded by the extracellular matrix that they secrete and assemble. Autologous chondrocyte implantation for cartilage defects has achieved good results, but the limited resources and complexity of the procedure have hindered wider application. Stem cells form an alternative to chondrocytes as a source of chondrogenic cells due to their ability to proliferate extensively while retaining the potential for differentiation. Adult stem cells such as mesenchymal stem cells have been differentiated into chondrocytes, but the limitations in their proliferative ability and the heterogeneous cell population hinder their adoption as a prime alternative source for generating chondrocytes. Human embryonic stem cells (hESCs) are attractive as candidates for cell replacement therapy because of their unlimited self-renewal and ability for differentiation into mesodermal derivatives as well as other lineages. In this review, we focus on current protocols for chondrogenic differentiation of ESCs, in particular the chemically defined culture system developed in our lab that could potentially be adapted for clinical application.
Collapse
Affiliation(s)
- Aixin Cheng
- 1 North West Embryonic Stem Cell Centre, Faculty of Life Science, University of Manchester , Manchester, United Kingdom
| | | | | |
Collapse
|
385
|
Karl A, Olbrich N, Pfeifer C, Berner A, Zellner J, Kujat R, Angele P, Nerlich M, Mueller MB. Thyroid hormone-induced hypertrophy in mesenchymal stem cell chondrogenesis is mediated by bone morphogenetic protein-4. Tissue Eng Part A 2013; 20:178-88. [PMID: 23937304 DOI: 10.1089/ten.tea.2013.0023] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chondrogenic differentiating mesenchymal stem cells (MSCs) express markers of hypertrophic growth plate chondrocytes. As hypertrophic cartilage undergoes ossification, this is a concern for the application of MSCs in articular cartilage tissue engineering. To identify mechanisms that elicit this phenomenon, we used an in vitro hypertrophy model of chondrifying MSCs for differential gene expression analysis and functional experiments with the focus on bone morphogenetic protein (BMP) signaling. Hypertrophy was induced in chondrogenic MSC pellet cultures by transforming growth factor β (TGFβ) and dexamethasone withdrawal and addition of triiodothyronine. Differential gene expression analysis of BMPs and their receptors was performed. Based on these results, the in vitro hypertrophy model was used to investigate the effect of recombinant BMP4 and the BMP inhibitor Noggin. The enhancement of hypertrophy could be shown clearly by an increased cell size, alkaline phosphatase activity, and collagen type X deposition. Upon induction of hypertrophy, BMP4 and the BMP receptor 1B were upregulated. Addition of BMP4 further enhanced hypertrophy in the absence, but not in the presence of TGFβ and dexamethasone. Thyroid hormone induced hypertrophy by upregulation of BMP4 and this induced enhancement of hypertrophy could be blocked by the BMP antagonist Noggin. BMP signaling is an important modulator of the late differentiation stages in MSC chondrogenesis and the thyroid hormone induces this pathway. As cartilage tissue engineering constructs will be exposed to this factor in vivo, this study provides important insight into the biology of MSC-based cartilage. Furthermore, the possibility to engineer hypertrophic cartilage may be helpful for critical bone defect repair.
Collapse
Affiliation(s)
- Alexandra Karl
- Department of Trauma Surgery, University of Regensburg Medical Center , Regensburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
386
|
Tan SL, Ahmad TS, Selvaratnam L, Kamarul T. Isolation, characterization and the multi-lineage differentiation potential of rabbit bone marrow-derived mesenchymal stem cells. J Anat 2013; 222:437-50. [PMID: 23510053 DOI: 10.1111/joa.12032] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2013] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are recognized by their plastic adherent ability, fibroblastic-like appearance, expression of specific surface protein markers, and are defined by their ability to undergo multi-lineage differentiation. Although rabbit bone marrow-derived MSCs (rbMSCs) have been used extensively in previous studies especially in translational research, these cells have neither been defined morphologically and ultrastructurally, nor been compared with their counterparts in humans in their multi-lineage differentiation ability. A study was therefore conducted to define the morphology, surface marker proteins, ultrastructure and multi-lineage differentiation ability of rbMSCs. Herein, the primary rbMSC cultures of three adult New Zealand white rabbits (at least 4 months old) were used for three independent experiments. rbMSCs were isolated using the gradient-centrifugation method, an established technique for human MSCs (hMSCs) isolation. Cells were characterized by phase contrast microscopy observation, transmission electron microscopy analysis, reverse transcriptase-polymerase chain reaction (PCR) analysis, immunocytochemistry staining, flow cytometry, alamarBlue(®) assay, histological staining and quantitative (q)PCR analysis. The isolated plastic adherent cells were in fibroblastic spindle-shape and possessed eccentric, irregular-shaped nuclei as well as rich inner cytoplasmic zones similar to that of hMSCs. The rbMSCs expressed CD29, CD44, CD73, CD81, CD90 and CD166, but were negative (or dim positive) for CD34, CD45, CD117 and HLD-DR. Despite having similar morphology and phenotypic expression, rbMSCs possessed significantly larger cell size but had a lower proliferation rate as compared with hMSCs. Using established protocols to differentiate hMSCs, rbMSCs underwent osteogenic, adipogenic and chondrogenic differentiation. Interestingly, differentiated rbMSCs demonstrated higher levels of osteogenic (Runx2) and chondrogenic (Sox9) gene expressions than that of hMSCs (P < 0.05). There was, however, no difference in the adipogenic (Pparγ) expressions between these cell types (P > 0.05). rbMSCs possess similar morphological characteristics to hMSCs, but have a higher potential for osteogenic and chondrogenic differentiation, despite having a lower cell proliferation rate than hMSCs. The characteristics reported here may be used as a comprehensive set of criteria to define or characterize rbMSCs.
Collapse
Affiliation(s)
- Sik-Loo Tan
- Tissue Engineering Group, National Orthopaedics Centre of Excellence in Research & Learning, Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
387
|
Amable PR, Teixeira MVT, Carias RBV, Granjeiro JM, Borojevic R. Identification of appropriate reference genes for human mesenchymal cells during expansion and differentiation. PLoS One 2013; 8:e73792. [PMID: 24023904 PMCID: PMC3759474 DOI: 10.1371/journal.pone.0073792] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/31/2013] [Indexed: 02/06/2023] Open
Abstract
Background Quantitative real time polymerase chain reaction (qPCR) is an extremely powerful technique for monitoring gene expression. The quantity of the messenger ribonucleic acids (mRNA) of interest should be normalized using a reference gene, in order to avoid unreliable results originated by the obtained RNA quality and quantity, manipulation errors and inhibitory contaminants. A reference gene is any gene that is stably and consistently expressed under the conditions being studied. Completely false data can be generated if a reference gene is not chosen adequately. Results In the present study, we compared expression levels of five putative reference genes (HPRT1, ACTB, GAPDH, RPL13A and B2M) in primary cultures of four different human cells: mesenchymal stromal cells obtained from bone marrow, adipose tissue or umbilical cord Whartońs Jelly, and dermal fibroblasts, under different expansion and differentiation conditions. We observed that reference genes are not the same for different cells under the same culture conditions. Conclusion Most stable reference genes under our experimental conditions were: RPL13A for adipose tissue- and Whartońs Jelly-derived mesenchymal stromal cells, and HPRT1 for bone marrow-derived mesenchymal stromal cells and dermal fibroblasts. ACTB was the most unstable gene when evaluating adipose tissue- and Whartońs Jelly-derived mesenchymal stromal cells, whilst GAPDH and B2M were the most unstable genes for bone marrow-derived mesenchymal stromal cells and dermal fibroblasts, respectively.
Collapse
Affiliation(s)
| | | | | | - José Mauro Granjeiro
- Bioengineering, National Institute of Metrology, Quality and Technology, Rio de Janeiro, Brazil
| | - Radovan Borojevic
- Excellion Biomedical Services, Petrópolis, Rio de Janeiro, Brazil
- Bioengineering, National Institute of Metrology, Quality and Technology, Rio de Janeiro, Brazil
| |
Collapse
|
388
|
Sampson S, Botto-van Bemden A, Aufiero D. Autologous bone marrow concentrate: review and application of a novel intra-articular orthobiologic for cartilage disease. PHYSICIAN SPORTSMED 2013; 41:7-18. [PMID: 24113698 DOI: 10.3810/psm.2013.09.2022] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Younger adults, aged < 65 years, increasingly present to their physicians with advanced cartilage disease or post-traumatic osteoarthritis. A number of treatments exist for lessening patient pain and improving patient function. However, many patients are becoming aware of the potential of regenerative therapies and are now seeking solutions to the impaired biology underlying their conditions rather than addressing only their symptoms. Patients do not want to merely lessen their symptoms temporarily with a surgical procedure that replaces damaged tissue, but instead seek correction and repair of the underlying biology to regenerate damaged tissue and alleviate their symptoms altogether. Current therapies for patients with cartilage disease or osteoarthritis range from non-surgical intra-articular injections with biologics, such as hyaluronic acid (HA), to total joint arthroplasty for advanced stages of disease. Total joint arthroplasty is a successful procedure for patients aged > 65 years; however, the limited long-term durability of implanted prostheses decreases the preference of using such methods in more active patients aged < 65 years. The potential of cell-based orthobiologic injection therapies (pertaining to therapeutic injectables that aim to restore the biologic environment and/or structural components of diseased or damaged musculoskeletal tissue) is of tremendous interest for younger, more active patients, and is even more appealing in that such therapy can be delivered at point-of-care in the clinic during an office visit. Notably, the exponential rate of progress in biotechnology has allowed for immediate application of myriad novel therapies prior to clear evidence of benefit from randomized clinical trials. Orthobiologic intra-articular injection therapies include HA and platelet-rich plasma (PRP). We report on current, available findings for a third-generation intra-articular orthobiologic injectable therapy for cartilage disease, bone marrow concentrate (BMC). Bone marrow concentrate contains mesenchymal stem cells (MSCs), hematopoetic stem cells, platelets (containing growth factors), and cytokines. The anti-inflammatory and immunomodulatory properties of bone marrow stem cells (BMSCs) can facilitate regeneration of tissue. Additionally, BMSCs enhance the quality of cartilage repair by increasing aggrecan content and tissue firmness. Following bone marrow aspiration (BMA), BMC is easily prepared using centrifugation, and is available for a same-day procedure with minimal manipulation of cells, thus complying with US Food and Drug Association (FDA) restrictions. To date, there are no published randomized controlled trials on the efficacy of use of autologous BMC intra-articular injections performed as a same-day in-office procedure for treating patients with cartilage disease; however, several publications have reported the ease of use of this method, its strong safety profile, and the fundamental science suggesting great therapeutic potential.
Collapse
Affiliation(s)
- Steven Sampson
- Clinical Instructor of Medicine, David Geffen School of Medicine at UCLA; Clinical Assistant Professor, Western University of Health Sciences; Adjunct Assistant Professor, Touro University; Founder, The Orthohealing Center and The Orthobiologic Institute (TOBI), Los Angeles, CA.
| | | | | |
Collapse
|
389
|
Xue R, Li JYS, Yeh Y, Yang L, Chien S. Effects of matrix elasticity and cell density on human mesenchymal stem cells differentiation. J Orthop Res 2013; 31:1360-5. [PMID: 23606500 DOI: 10.1002/jor.22374] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Accepted: 03/26/2013] [Indexed: 02/04/2023]
Abstract
Human mesenchymal stem cells (hMSCs) can differentiate into various cell types, including osteogenic and chondrogenic cells. The matrix elasticity and cell seeding density are important factors in hMSCs differentiation. We cultured hMSCs at different seeding densities on polyacrylamide hydrogels with different stiffness corresponding to Young's moduli of 1.6 ± 0.3 and 40 ± 3.6 kPa. The promotion of osteogenic marker expression by hard gel is overridden by a high seeding density. Cell seeding density, however, did not influence the chondrogenic marker expressions induced by soft gel. These findings suggest that interplays between cell-matrix and cell-cell interactions contribute to hMSCs differentiation. The promotion of osteogenic differentiation on hard matrix was shown to be mediated through the Ras pathway. Inhibition of Ras (RasN17) significantly decreased ERK, Smad1/5/8 and AKT activation, and osteogenic markers expression. However, constitutively active Ras (RasV12) had little effect on osteogenic marker expression, suggesting that the Ras pathways are necessary but not sufficient for osteogenesis. Taken together, our results indicate that matrix elasticity and cell density are important microenvironmental cues driving hMSCs proliferation and differentiation.
Collapse
Affiliation(s)
- Ruyue Xue
- 111 Project Laboratory of Biomechanics and Tissue Repair, Bioengineering College, Chongqing University, Chongqing, 400044, PR China
| | | | | | | | | |
Collapse
|
390
|
Liu W, Wei Y, Zhang X, Xu M, Yang X, Deng X. Lower extent but similar rhythm of osteogenic behavior in hBMSCs cultured on nanofibrous scaffolds versus induced with osteogenic supplement. ACS NANO 2013; 7:6928-6938. [PMID: 23906375 DOI: 10.1021/nn402118s] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Nanotopographic cues from biomaterials exert powerful effects on the osteogenic differentiation of mesenchymal stem cells because of their niche-mimicking features. However, the biological mechanisms underlying cell lineage determination by surface nanotopography have not been clearly elucidated. Here, we explored the osteogenic behavior of human bone marrow mesenchymal stem cells (hBMSCs) on poly-l-lactide nanofibers with different orientations and monitored the dynamic changes in global gene expression triggered by topographical cues. RT-PCR analysis of osteogenic marker genes and ALP activity assays demonstrated that hBMSCs cultured on random nanofibers showed enhanced osteogenic-specific fate compared with those on aligned nanofibers. Microarray analysis demonstrated a similar temporal change in gene expression patterns between hBMSCs cultured on random nanofibers and those induced with an osteogenic supplement (OS). However, the extent of osteogenic differentiation on the fibrous scaffold was much lower than that driven by chemical OS. In-depth pathway analysis revealed that focal adhesion kinase, TGF-β, Wnt, and MAPK pathways were involved in the activation of osteogenic differentiation in hBMSCs on random nanofibers. These findings suggested that a lower extent but similar rhythm of dynamic cellular behavior was induced on random nanofibers when compared with the OS condition and that mechanotransduction could trigger nonspecific and multilevel responses in hBMSCs. This study provides insight into the regulation of osteogenesis directed by substratum surfaces.
Collapse
Affiliation(s)
- Wentao Liu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, People's Republic of China
| | | | | | | | | | | |
Collapse
|
391
|
Song N, Armstrong AD, Li F, Ouyang H, Niyibizi C. Multipotent mesenchymal stem cells from human subacromial bursa: potential for cell based tendon tissue engineering. Tissue Eng Part A 2013; 20:239-49. [PMID: 23865619 DOI: 10.1089/ten.tea.2013.0197] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rotator cuff injuries are a common clinical problem either as a result of overuse or aging. Biological approaches to tendon repair that involve use of scaffolding materials or cell-based approaches are currently being investigated. The cell-based approaches are focused on applying multipotent mesenchymal stem cells (MSCs) mostly harvested from bone marrow. In the present study, we focused on characterizing cells harvested from tissues associated with rotator cuff tendons based on an assumption that these cells would be more appropriate for tendon repair. We isolated MSCs from bursa tissue associated with rotator cuff tendons and characterized them for multilineage differentiation in vitro and in vivo. Human bursa was obtained from patients undergoing rotator cuff surgery and cells within were isolated using collagenase and dispase digestion. The cells isolated from the tissues were characterized for osteoblastic, adipogenic, chondrogenic, and tenogenic differentiation in vitro and in vivo. The results showed that the cells isolated from bursa tissue exhibited MSCs characteristics as evidenced by the expression of putative cell surface markers attributed to MSCs. The cells exhibited high proliferative capacity and differentiated toward cells of mesenchymal lineages with high efficiency. Bursa-derived cells expressed markers of tenocytes when treated with bone morphogenetic protein-12 (BMP-12) and assumed aligned morphology in culture. Bursa cells pretreated with BMP-12 and seeded in ceramic scaffolds formed extensive bone, as well as tendon-like tissue in vivo. Bone formation was demonstrated by histological analysis and immunofluorescence for DMP-1 in tissue sections made from the scaffolds seeded with the cells. Tendon-like tissue formed in vivo consisted of parallel collagen fibres typical of tendon tissues. Bursa-derived cells also formed a fibrocartilagenous tissue in the ceramic scaffolds. Taken together, the results demonstrate a new source of MSCs with a high potential for application in tendon repair.
Collapse
Affiliation(s)
- Na Song
- 1 College of Animal Sciences, Jilin University , Changchun, China
| | | | | | | | | |
Collapse
|
392
|
Sritanaudomchai H, Kitiyanant Y, Tong-ngam P, Thonabulsombat C, White KL, Kusamran T. Enhanced chondrogenesis through specific growth factors in a buffalo embryonic stem cell model. Cell Biol Int 2013; 37:1246-58. [PMID: 23852953 DOI: 10.1002/cbin.10153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 07/08/2013] [Indexed: 11/10/2022]
Abstract
Chondrogenic differentiation of embryonic stem cells (ESCs) via embryoid bodies (EBs) is an established model to investigate chondrogenesis signaling pathways and molecular mechanisms in vitro. Our aim has been to improve upon the number of differentiated cells needed for the in vitro development of functional cartilage. Chondrogenic differentiation of buffalo ESCs was modulated by bone morphogenetic protein 2 (BMP-2), fibroblast growth factor 10 (FGF-10), transforming growth factor-beta1 (TGF-β1 ) individually and their combination. ESCs differentiation into chondrocytes was characterized by the appearance of Alcian blue-stained nodules and the expression of cartilage-associated genes (RT-PCR) and protein (immunocytochemistry). BMP-2 or FGF-10 treatment enhanced chondrogenic differentiation, whereas TGF-β1 treatment inhibited buffalo ESC-derived chondrogenesis. The combination of BMP-2 and FGF-10 was the most effective treatment. This treatment resulted in a higher number of Alcian blue-positive nodules by 15.2-fold, expression of the mesenchymal cell marker scleraxis gene by 3.25-fold, and the cartilage matrix protein collagen II gene and protein 1.9- and 7-fold, respectively, compared to the untreated control group. Chondrogenesis was also recapitulated from mesenchymal and chondrogenic progenitor cells, resulting in the establishment of mature chondrocytes. Thus, buffalo ESCs can be successfully triggered in vitro to differentiate into chondrocyte-like cells by specific growth factors, which may provide a novel in vitro model for further investigation of the regulatory mechanism(s) involved.
Collapse
Affiliation(s)
- Hathaitip Sritanaudomchai
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | | | | | | | | | |
Collapse
|
393
|
Madry H, Rey-Rico A, Venkatesan JK, Johnstone B, Cucchiarini M. Transforming growth factor Beta-releasing scaffolds for cartilage tissue engineering. TISSUE ENGINEERING PART B-REVIEWS 2013; 20:106-25. [PMID: 23815376 DOI: 10.1089/ten.teb.2013.0271] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The maintenance of a critical threshold concentration of transforming growth factor beta (TGF-β) for a given period of time is crucial for the onset and maintenance of chondrogenesis. Thus, the development of scaffolds that provide temporal and/or spatial control of TGF-β bioavailability has appeal as a mechanism to induce the chondrogenesis of stem cells in vitro and in vivo for articular cartilage repair. In the past decade, many types of scaffolds have been designed to advance this goal: hydrogels based on polysaccharides, hyaluronic acid, and alginate; protein-based hydrogels such as fibrin, gelatin, and collagens; biopolymeric gels and synthetic polymers; and solid and hybrid composite (hydrogel/solid) scaffolds. In this study, we review the progress in developing strategies to deliver TGF-β from scaffolds with the aim of enhancing chondrogenesis. In the future, such scaffolds could prove critical for tissue engineering cartilage, both in vitro and in vivo.
Collapse
Affiliation(s)
- Henning Madry
- 1 Center of Experimental Orthopaedics, Saarland University , Homburg, Germany
| | | | | | | | | |
Collapse
|
394
|
Izal I, Aranda P, Sanz-Ramos P, Ripalda P, Mora G, Granero-Moltó F, Deplaine H, Gómez-Ribelles JL, Ferrer GG, Acosta V, Ochoa I, García-Aznar JM, Andreu EJ, Monleón-Pradas M, Doblaré M, Prósper F. Culture of human bone marrow-derived mesenchymal stem cells on of poly(L-lactic acid) scaffolds: potential application for the tissue engineering of cartilage. Knee Surg Sports Traumatol Arthrosc 2013; 21:1737-50. [PMID: 22864678 DOI: 10.1007/s00167-012-2148-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Accepted: 07/17/2012] [Indexed: 01/09/2023]
Abstract
PURPOSE Due to the attractive properties of poly(L-lactic acid) (PLLA) for tissue engineering, the aim was to determine the growth and differentiation capacity of mesenchymal stromal cells (MSCs) in PLLA scaffolds and their potential use in the treatment of cartilage diseases. METHODS MSCs were cultured in PLLA films and thin porous membranes to study adherence and proliferation. Permeability and porosity were determined for the different scaffolds employed. The optimal conditions for cell seeding were first determined, as well as cell density and distribution inside the PLLA. Scaffolds were then maintained in expansion or chondrogenic differentiation media for 21 days. Apoptosis, proliferation and chondrogenic differentiation was assessed after 21 days in culture by immunohistochemistry. Mechanical characteristics of scaffolds were determined before and after cell seeding. RESULTS MSCs uniformly adhered to PLLA films as well as to porous membranes. Proliferation was detected only in monolayers of pure PLLA, but was no longer detected after 10 days. Mechanical characterization of PLLA scaffolds showed differences in the apparent compression elastic modulus for the two sizes used. After determining high efficiencies of seeding, the production of extracellular matrix (ECM) was determined and contained aggrecan and collagens type I and X. ECM produced by the cells induced a twofold increase in the apparent elastic modulus of the composite. CONCLUSIONS Biocompatible PLLA scaffolds have been developed that can be efficiently loaded with MSCs. The scaffold supports chondrogenic differentiation and ECM deposition that improves the mechanics of the scaffold. Although this improvement does not met the expectations of a hyaline-like cartilage ECM, in part due to the lack of a mechanical stimulation, their potential use in the treatment of cartilage pathologies encourages to improve the mechanical component.
Collapse
Affiliation(s)
- Iñigo Izal
- Laboratory for Orthopaedic Research, University of Navarra, Irunlarrea, 1, 31080, Pamplona, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
395
|
Watts AE, Ackerman-Yost JC, Nixon AJ. A comparison of three-dimensional culture systems to evaluate in vitro chondrogenesis of equine bone marrow-derived mesenchymal stem cells. Tissue Eng Part A 2013; 19:2275-83. [PMID: 23725547 DOI: 10.1089/ten.tea.2012.0479] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To compare in vitro three-dimensional (3D) culture systems that model chondrogenesis of bone marrow-derived mesenchymal stem cells (MSCs). METHODS MSCs from five horses 2-3 years of age were consolidated in fibrin 0.3% alginate, 1.2% alginate, 2.5×10(5) cell pellets, 5×10(5) cell pellets, and 2% agarose, and maintained in chondrogenic medium with supplemental TGF-β1 for 4 weeks. Pellets and media were tested at days 1, 14, and 28 for gene expression of markers of chondrogenic maturation and hypertrophy (ACAN, COL2B, COL10, SOX9, 18S), and evaluated by histology (hematoxylin and eosin, Toluidine Blue) and immunohistochemistry (collagen type II and X). RESULTS alginate, fibrin alginate (FA), and both pellet culture systems resulted in chondrogenic transformation. Adequate RNA was not obtained from agarose cultures at any time point. There was increased COL2B, ACAN, and SOX9 expression on day 14 from both pellet culture systems. On day 28, increased expression of COL2B was maintained in 5×10(5) cell pellets and there was no difference in ACAN and SOX9 between FA and both pellet cultures. COL10 expression was significantly lower in FA cultures on day 28. Collagen type II was abundantly formed in all culture systems except alginate and collagen type X was least in FA hydrogels. CONCLUSION equine MSCs respond to 3D culture in FA blended hydrogel and both pellet culture systems with chondrogenic induction. For prevention of terminal differentiation and hypertrophy, FA culture may be superior to pellet culture systems.
Collapse
Affiliation(s)
- Ashlee E Watts
- Comparative Orthopaedics Laboratory, Department of Clinical Sciences, Cornell University, Ithaca, NY 14853, USA
| | | | | |
Collapse
|
396
|
Li C, Bi W, Gong Y, Ding X, Guo X, Sun J, Cui L, Yu Y. Transforming growth factor-beta1 inhibits tissue engineering cartilage absorption via inducing the generation of regulatory T cells. J Tissue Eng Regen Med 2013; 10:E113-20. [PMID: 23868873 DOI: 10.1002/term.1777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 01/31/2013] [Accepted: 04/22/2013] [Indexed: 01/26/2023]
Affiliation(s)
- Chichi Li
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Wei Bi
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Yiming Gong
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Xiaojun Ding
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Xuehua Guo
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Jian Sun
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| | - Lei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People's Hospital; Shanghai Jiao Tong University School of Medicine; 639 Zhi Zao Ju Road Shanghai 200011 PR China
| | - Youcheng Yu
- Department of Stomatology; Zhongshan Hospital of Fudan University; 180 Fenglin Road Shanghai 200032 PR China
| |
Collapse
|
397
|
Human umbilical cord blood-derived mesenchymal stem cells in the cultured rabbit intervertebral disc: a novel cell source for disc repair. Am J Phys Med Rehabil 2013; 92:420-9. [PMID: 23598901 DOI: 10.1097/phm.0b013e31825f148a] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Back pain associated with symptomatic disc degeneration is a common clinical condition. Intervertebral disc (IVD) cell apoptosis and senescence increase with aging and degeneration. Repopulating the IVD with cells that could produce and maintain extracellular matrix would be an alternative therapy to surgery. The objective of this study was to determine the potential of human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) as a novel cell source for disc repair. In this study, we intended to confirm the potential for hUCB-MSCs to differentiate and display a chondrocyte-like phenotype after culturing in micromass and after injection into the rabbit IVD explant culture. We also wanted to confirm hUCB-MSC survival after transplantation into the IVD explant culture. DESIGN This study consisted of micromass cultures and in vitro rabbit IVD explant cultures to assess hUCB-MSC survival and differentiation to display chondrocyte-like phenotype. First, hUCB-MSCs were cultured in micromass and stained with Alcian blue dye. Second, to confirm cell survival, hUCB-MSCs were labeled with an infrared dye and a fluorescent dye before injection into whole rabbit IVD explants (host). IVD explants were then cultured for 4 wks. Cell survival was confirmed by two independent techniques: an imaging system detecting the infrared dye at the organ level and fluorescence microscopy detecting fluorescent dye at the cellular level. Cell viability was assessed by staining the explant with CellTracker green, a membrane-permeant tracer specific for live cells. Human type II collagen gene expression (from the graft) was assessed by polymerase chain reaction. RESULTS We have shown that hUCB-MSCs cultured in micromass are stained blue with Alcian blue dye, which suggests that proteoglycan-rich extracellular matrix is produced. In the cultured rabbit IVD explants, hUCB-MSCs survived for at least 4 wks and expressed the human type II collagen gene, suggesting that the injected hUCB-MSCs are differentiating into a chondrocyte-like lineage. CONCLUSIONS This study demonstrates the abiity of hUBC-MSCs to survive and assume a chondrocyte-like phenotype when injected into the rabbit IVD. These data support the potential for hUBC-MSCs as a cell source for disc repair. Further measures of the host response to the injection and studies in animal models are needed before trials in humans.
Collapse
|
398
|
Mentink A, Hulsman M, Groen N, Licht R, Dechering KJ, van der Stok J, Alves HA, Dhert WJ, van Someren EP, Reinders MJ, van Blitterswijk CA, de Boer J. Predicting the therapeutic efficacy of MSC in bone tissue engineering using the molecular marker CADM1. Biomaterials 2013; 34:4592-601. [DOI: 10.1016/j.biomaterials.2013.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/01/2013] [Indexed: 12/17/2022]
|
399
|
Wee AS, Lim CK, Merican AM, Ahmad TS, Kamarul T. Total cell pooling in vitro: an effective isolation method for bone marrow-derived multipotent stromal cells. In Vitro Cell Dev Biol Anim 2013; 49:424-32. [PMID: 23708918 DOI: 10.1007/s11626-013-9626-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/18/2013] [Indexed: 12/18/2022]
Abstract
In vitro cellular proliferation and the ability to undergo multilineage differentiation make bone marrow-derived multipotent stromal cells (MSCs) potentially useful for clinical applications. Several methods have been described to isolate a homogenous bone marrow-derived MSCs population; however, none has been proven most effective, mainly due to their effects on proliferation and differentiation capability of the isolated cells. It is hypothesized that our newly established total cell pooling method may provide a better alternative as compared to the standard isolation method (density gradient centrifugation method). For the total cell pooling method, MSCs were isolated from rabbit bone marrow and were subsequently cultured in the growth medium without further separation as in the standard isolation method. The total cell pooling method was 65 min faster than the standard isolation method in completing cell isolation. Nevertheless, both methods did not differ significantly in the number of primary viable cells and population doubling time in the cultures (p > 0.05). The isolated cells from both methods expressed CD29 and CD44 markers, but not CD45 markers. Furthermore, they displayed multilineage differentiation characteristics of chondroblasts, osteoblasts, and adipocytes. In conclusion, both methods provide similar efficiency in the isolation of rabbit bone marrow-derived MSCs; however, the total cell pooling method is technically simpler and more cost effective than the standard isolation method.
Collapse
Affiliation(s)
- Ai-Sze Wee
- Tissue Engineering Group (TEG), National Orthopaedic Centre for Research and Learning (NOCERAL), Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | | | | | | | | |
Collapse
|
400
|
Moroni L, Fornasari PM. Human mesenchymal stem cells: a bank perspective on the isolation, characterization and potential of alternative sources for the regeneration of musculoskeletal tissues. J Cell Physiol 2013; 228:680-7. [PMID: 22949310 DOI: 10.1002/jcp.24223] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 08/27/2012] [Indexed: 01/14/2023]
Abstract
The continuous discovery of human mesenchymal stem cells (hMSCs) in different tissues is stirring up a tremendous interest as a cell source for regenerative medicine therapies. Historically, hMSCs have been always considered a sub-population of mononuclear cells present in the bone marrow (BM). Although BM-hMSCs are still nowadays considered as the most promising mesenchymal stem cell population to reach the clinics due to their capacity to differentiate into multiple tissues, hMSCs derived from other adult and fetal tissues have also demonstrated to possess similar differentiation capacities. Furthermore, different reports have highlighted a higher recurrence of hMSCs in some of these tissues as compared to BM. This offer a fascinating panorama for cell banking, since the creation of a stem cell factory could be envisioned where hMSCs are stocked and used for ad hoc clinical applications. In this review, we summarize the main findings and state of the art in hMSCs isolation, characterization, and differentiation from alternative tissue sources and we attempt to compare their potency for musculoskeletal regeneration.
Collapse
Affiliation(s)
- Lorenzo Moroni
- Muscoloskeletal Tissue Bank, Rizzoli Orthopaedic Institute, Bologna, Italy.
| | | |
Collapse
|