151
|
Pei M, He F. Extracellular matrix deposited by synovium-derived stem cells delays replicative senescent chondrocyte dedifferentiation and enhances redifferentiation. J Cell Physiol 2012; 227:2163-74. [PMID: 21792932 DOI: 10.1002/jcp.22950] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The aim of this study was to assess the effect of extracellular matrix (ECM) deposited by synovium-derived stem cells (SDSCs) on articular chondrocyte expansion and maintenance of differentiation status and redifferentiation capacity. Passage 0 (P0) pig articular chondrocytes were expanded for six passages on plastic flasks (Plastic), SDSC-derived ECM (ECM), or substrate switching from either Plastic to ECM (PtoE) or ECM to Plastic (EtoP). Cell morphology, gene expression profiles, and immunophenotypes at each passage were used to characterize differentiation status of expanded cells. Chondrocytes at P0, P2, and P6 were assessed for redifferentiation capacity in a pellet culture system treated with either TGF-β1- or serum-containing medium for 14 days, using histology, immunohistochemistry, biochemistry, Western blot, and real-time PCR. We found that ECM not only greatly enhanced chondrocyte expansion but also delayed dedifferentiation of expanded chondrocytes. Intriguingly, compared to a dramatic decrease in CD90+/CD105+ cells and CD90+ cells, CD105+ cells dramatically increased when chondrocytes were plated on Plastic; on the contrary, ECM expansion dramatically increased CD90+ cells and delayed the decrease of CD90+/CD105+ cells. Interestingly, expanded chondrocytes on ECM also acquired a strong redifferentiation capacity, particularly in the pellets treated with TGF-β1. In conclusion, the ratio of CD90 to CD105 may serve as a marker indicative of proliferation and redifferentiation capacity of dedifferentiated chondrocytes. ECM deposited by SDSCs provides a tissue-specific three-dimensional microenvironment for ex vivo expansion of articular chondrocytes while retaining redifferentiation capacity, suggesting that ECM may provide a novel approach for autologous chondrocyte-based cartilage repair.
Collapse
Affiliation(s)
- Ming Pei
- Department of Orthopaedics, West Virginia University, Morgantown, West Virginia 26506-9196, USA.
| | | |
Collapse
|
152
|
Mahjoub M, Sassi N, Driss M, Laadhar L, Allouche M, Hamdoun M, Romdhane KB, Sellami S, Makni S. Expression patterns of Notch receptors and their ligands in human osteoarthritic and healthy articular cartilage. Tissue Cell 2012; 44:182-94. [PMID: 22455903 DOI: 10.1016/j.tice.2012.03.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2011] [Revised: 03/02/2012] [Accepted: 03/02/2012] [Indexed: 11/17/2022]
Abstract
Notch pathway plays a pivotal role in cell fate determination. There is much interest surrounding its therapeutic potential, in osteoarthritis, but the expression profile of Notch-related molecules, as well as their relation with cartilage pathological parameters, remains unclear. The purpose of our study is to analyze the expression pattern of Notch family members, type II and type I collagen, in normal (healthy) and osteoarthritic human knee cartilage. Osteoarthritic cartilages were obtained from 3 patients undergoing a total knee replacement. Macroscopically normal cartilage was dissected from 3 human knees at the time of autopsy or surgery. Immunohistochemical staining was performed using Notch1,2,3 and 4, Delta, Jagged, type II collagen and type I collagen antibodies. In healthy cartilage, type II collagen was abundantly expressed while type I was absent. This latter increased proportionally to the osteoarthritic grade. Type II collagen expression remained intense in osteoarthritic cartilage. In healthy cartilage as well as in cartilage with minor lesions, Notch family member's proteins were not or just weakly expressed at the surface and in the cells. However, Notch molecules were over-expressed in osteoarthritic cartilage compared to healthy one. This expression pattern was different according to the cartilage zone and the severity of OA. Our data suggest that Notch signaling is activated in osteoarthritic cartilage, compared to healthy cartilage, with a much more abundant expression in the most damaged areas.
Collapse
Affiliation(s)
- M Mahjoub
- Immuno-Rheumatology Research Laboratory, Rheumatology Department, La Rabta Hospital, University of Tunis-El Manar, 1007 Tunis, Tunisia
| | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Hypoxia mediated isolation and expansion enhances the chondrogenic capacity of bone marrow mesenchymal stromal cells. Stem Cell Res Ther 2012; 3:9. [PMID: 22385573 PMCID: PMC3392769 DOI: 10.1186/scrt100] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/18/2011] [Accepted: 03/02/2012] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION The capacity of bone marrow mesenchymal stromal cells (BMSCs) to be induced into chondrocytes has drawn much attention for cell-based cartilage repair. BMSCs represent a small proportion of cells of the bone marrow stromal compartment and, thus, culture expansion is a necessity for therapeutic use. However, there is no consensus on how BMSCs should be isolated nor expanded to maximize their chondrogenic potential. During embryonic development pluripotent stem cells differentiate into chondrocytes and form cartilage in a hypoxic microenvironment. METHODS Freshly harvested human BMSCs were isolated and expanded from the aspirates of six donors, under either hypoxic conditions (3% O2) or normoxic conditions (21% O2). A colony-forming unit fibroblastic (Cfu-f) assay was used to determine the number of cell colonies developed from each donor. BMSCs at passage 2 (P2) were characterized by flow cytometry for the phenotypic expression of cell surface markers on mesenchymal stem cells. BMSCs at P2 were subsequently cultured in vitro as three-dimensional cell pellets in a defined serum-free chondrogenic medium under normoxic and hypoxic conditions. Chondrogenic differentiation of the BMSCs was characterized by biochemical and histological methods and by quantitative gene-expression analysis. RESULTS After 14 days of culture, the number of BMSC colonies developed under hypoxia was generally higher (8% to 38% depending on donor) than under normoxia. BMSCs were positive for the cell surface markers CD13, CD29, CD44, CD73, CD90, CD105 and CD151, and negative for CD34. Regardless of the oxygen tension during pellet culture, hypoxia-expanded BMSC pellets underwent a more robust chondrogenesis than normoxia-expanded BMSC pellets after three weeks of culture, as judged by increased glycosaminoglycan synthesis and Safranin O staining, along with increased mRNA expression of aggrecan, collagen II and Sox9. Hypoxic conditions enhanced the mRNA expression of hypoxia inducible factor-2 alpha (HIF-2α) but suppressed the mRNA expression of collagen X in BMSC pellet cultures regardless of the oxygen tension during BMSC isolation and propagation. CONCLUSIONS Taken together, our data demonstrate that isolation and expansion of BMSCs under hypoxic conditions augments the chondrogenic potential of BMSCs. This suggests that hypoxia-mediated isolation and expansion of BMSCs may improve clinical applications of BMSCs for cartilage repair.
Collapse
|
154
|
Egli RJ, Wernike E, Grad S, Luginbühl R. Physiological cartilage tissue engineering effect of oxygen and biomechanics. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 289:37-87. [PMID: 21749898 DOI: 10.1016/b978-0-12-386039-2.00002-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In vitro engineering of cartilaginous tissues has been studied for many years, and tissue-engineered constructs are sought to be used clinically for treating articular cartilage defects. Even though there is a plethora of studies and data available, no breakthroughs have been achieved yet that allow for implanting in vivo cultured articular cartilaginous tissues in patients. A review of contributions to cartilage tissue engineering over the past decades emphasizes that most of the studies were performed under environmental conditions neglecting the physiological situation. This is specifically pronounced in the use of bioreactor systems which neither allow for application of near physiomechanical stimulations nor for controlling a hypoxic environment as it is experienced in synovial joints. It is suspected that the negligence of these important parameters has slowed down progress and prevented major breakthroughs in the field. This review focuses on the main aspects of cartilage tissue engineering with emphasis on the relation and understanding of employing physiological conditions.
Collapse
|
155
|
Kužmová E, Kotrba R, Rolf HJ, Bartoš L, Wiese KG, Schulz J, Bubenik GA. Factors affecting the number of STRO-1+ stem cells derived from regenerating antler and pedicle cells of red and fallow deer. ANIMAL PRODUCTION SCIENCE 2012. [DOI: 10.1071/an12012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Mesenchymal stem cells positive to surface antigen STRO-1 were isolated from regenerating antlers of red deer (Cervus elaphus) and fallow deer (Dama dama) using a magnetic-activated cell sorting (MACS) method. In this study we analysed factors potentially affecting the number of STRO-1+ cells in the cell cultures. With regard to the STRO-1 antigen, we evaluated data from 188 MACS separation procedures of cell cultures cultivated in Dulbecco’s Modified Eagle Medium and 10% fetal calf serum of four fallow deer males (130 procedures) and four red deer males (58 procedures). The analysed factors were the sampling site of the antler or the pedicle, cell passage and type of the cell culture (mixed or STRO-1 negative cell cultures). The percentage of obtained STRO-1+ cells varied greatly from 0.4 to 38.9% for fallow deer and from 1.8 to 16.5% for red deer. We have not found any significant influence of the sampling site. The passage and the type of culture were significant factors for both fallow and red deer cells. The highest numbers of STRO-1+ cells were obtained from the second passage from both fallow and red deer cell cultures (24.6 and 5.5%, respectively). Our experiment revealed that we can maximise the number of STRO-1+ cells in the cultures by manipulating the cultivation factors.
Collapse
|
156
|
Acharya C, Adesida A, Zajac P, Mumme M, Riesle J, Martin I, Barbero A. Enhanced chondrocyte proliferation and mesenchymal stromal cells chondrogenesis in coculture pellets mediate improved cartilage formation. J Cell Physiol 2011; 227:88-97. [PMID: 22025108 DOI: 10.1002/jcp.22706] [Citation(s) in RCA: 187] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In this study, we aimed at investigating the interactions between primary chondrocytes and mesenchymal stem/stromal cells (MSC) accounting for improved chondrogenesis in coculture systems. Expanded MSC from human bone marrow (BM-MSC) or adipose tissue (AT-MSC) were cultured in pellets alone (monoculture) or with primary human chondrocytes from articular (AC) or nasal (NC) cartilage (coculture). In order to determine the reached cell number and phenotype, selected pellets were generated by combining: (i) human BM-MSC with bovine AC, (ii) BM-MSC from HLA-A2+ with AC from HLA-A2- donors, or (iii) human green fluorescent protein transduced BM-MSC with AC. Human BM-MSC and AC were also cultured separately in transwells. Resulting tissues and/or isolated cells were assessed immunohistologically, biochemically, cytofluorimetrically, and by RT-PCR. Coculture of NC or AC (25%) with BM-MSC or AT-MSC (75%) in pellets resulted in up to 1.6-fold higher glycosaminoglycan content than what would be expected based on the relative percentages of the different cell types. This effect was not observed in the transwell model. BM-MSC decreased in number (about fivefold) over time and, if cocultured with chondrocytes, increased type II collagen and decreased type X collagen expression. Instead, AC increased in number (4.2-fold) if cocultured with BM-MSC and maintained a differentiated phenotype. Chondro-induction in MSC-chondrocyte coculture is a robust process mediated by two concomitant effects: MSC-induced chondrocyte proliferation and chondrocyte-enhanced MSC chondrogenesis. The identified interactions between progenitor and mature cell populations may lead to the efficient use of freshly harvested chondrocytes for ex vivo cartilage engineering or in situ cartilage repair.
Collapse
|
157
|
Musumeci G, Lo Furno D, Loreto C, Giuffrida R, Caggia S, Leonardi R, Cardile V. Mesenchymal stem cells from adipose tissue which have been differentiated into chondrocytes in three-dimensional culture express lubricin. Exp Biol Med (Maywood) 2011; 236:1333-1341. [PMID: 22036733 DOI: 10.1258/ebm.2011.011183] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The present study focused on the isolation, cultivation and characterization of human mesenchymal stem cells (MSCs) from adipose tissue and on their differentiation into chondrocytes through the NH ChondroDiff medium. The main aim was to investigate some markers of biomechanical quality of cartilage, such as lubricin, and collagen type I and II. Little is known, in fact, about the ability of chondrocytes from human MSCs of adipose tissue to generate lubricin in three-dimensional (3D) culture. Lubricin, a 227.5-kDa mucinous glycoprotein, is known to play an important role in articular joint physiology, and the loss of accumulation of lubricin is thought to play a role in the pathology of osteoarthritis. Adipose tissue is an alternative source for the isolation of multipotent MSCs, which allows them to be obtained by a less invasive method and in larger quantities than from other sources. These cells can be isolated from cosmetic liposuctions in large numbers and easily grown under standard tissue culture conditions. 3D chondrocytes were assessed by histology (hematoxylin and eosin) and histochemistry (Alcian blue and Safranin-O/fast green staining). Collagen type I, II and lubricin expression was determined through immunohistochemistry and Western blot. The results showed that, compared with control cartilage and monolayer chondrocytes showing just collagen type I, chondrocytes from MSCs (CD44-, CD90- and CD105- positive; CD45-, CD14- and CD34-negative) of adipose tissue grown in nodules were able to express lubricin, and collagen type I and II, indicative of hyaline cartilage formation. Based on the function of lubricin in the joint cavity and disease and as a potential therapeutic agent, our results suggest that MSCs from adipose tissue are a promising cell source for tissue engineering of cartilage. Our results suggest that chondrocyte nodules producing lubricin could be a novel biotherapeutic approach for the treatment of cartilage abnormalities.
Collapse
Affiliation(s)
- Giuseppe Musumeci
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
158
|
do Amaral RJFC, Pedrosa CDSG, Kochem MCL, Silva KRD, Aniceto M, Claudio-da-Silva C, Borojevic R, Baptista LS. Isolation of human nasoseptal chondrogenic cells: a promise for cartilage engineering. Stem Cell Res 2011; 8:292-9. [PMID: 22099383 DOI: 10.1016/j.scr.2011.09.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 08/03/2011] [Accepted: 09/27/2011] [Indexed: 12/20/2022] Open
Abstract
In cartilaginous tissues, perichondrium cambium layer may be the source of new cartilage. Human nasal septal perichondrium is considered to be a homogeneous structure in which some authors do not recognize the perichondrium internal zone or the cambium layer as a layer distinct from adjacent cartilage surface. In the present study, we isolated a chondrogenic cell population from human nasal septal cartilage surface zone. Nasoseptal chondrogenic cells were positive for surface markers described for mesenchymal stem cells, with exception of CD146, a perivascular cell marker, which is consistent with their avascular niche in cartilage. Although only Sox-9 was constitutively expressed, they also revealed osteogenic and chondrogenic, but not adipogenic, potentials in vitro, suggesting a more restricted lineage potential compared to mesenchymal stem cells. Interestingly, even in absence of chondrogenic growth factors in the pellet culture system, nasoseptal chondrogenic cells had a capacity to synthesize sulfated glycosaminoglycans, large amounts of collagen type II and to a lesser extent collagen type I. The spontaneous chondrogenic potential of this population of cells indicates that they may be a possible source for cartilage tissue engineering. Besides, the pellet culture system using nasoseptal chondrogenic cells may also be a model for studies of chondrogenesis.
Collapse
Affiliation(s)
- Ronaldo J F C do Amaral
- Programa de Pós-graduação em Ciências Morfológicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
159
|
McCarthy HE, Bara JJ, Brakspear K, Singhrao SK, Archer CW. The comparison of equine articular cartilage progenitor cells and bone marrow-derived stromal cells as potential cell sources for cartilage repair in the horse. Vet J 2011; 192:345-51. [PMID: 21968294 DOI: 10.1016/j.tvjl.2011.08.036] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 08/22/2011] [Accepted: 08/31/2011] [Indexed: 02/07/2023]
Abstract
A chondrocyte progenitor population isolated from the surface zone of articular cartilage presents a promising cell source for cell-based cartilage repair. In this study, equine articular cartilage progenitor cells (ACPCs) and equine bone marrow-derived stromal cells (BMSCs) were compared as potential cell sources for repair. Clonally derived BMSCs and ACPCs demonstrated expression of the cell fate selector gene, Notch-1, and the putative stem cell markers STRO-1, CD90 and CD166. Chondrogenic induction revealed positive labelling for collagen type II and aggrecan. Collagen type X was not detected in ACPC pellets but was observed in all BMSC pellets. In addition, it was observed that BMSCs labelled for Runx2 and matrilin-1 antibodies, whereas ACPC labelling was significantly less or absent. For both cell types, osteogenic induction revealed positive von Kossa staining in addition to positive labelling for osteocalcin. Adipogenic induction revealed a positive result via oil red O staining in both cell types. ACPCs and BMSCs have demonstrated functional equivalence in their multipotent differentiation capacity. Chondrogenic induction of BMSCs resulted in a hypertrophic cartilage (endochondral) phenotype, which can limit cartilage repair as the tissue can undergo mineralisation. ACPCs may therefore be considered superior to BMSCs in producing cartilage capable of functional repair.
Collapse
Affiliation(s)
- Helen E McCarthy
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK.
| | | | | | | | | |
Collapse
|
160
|
Tan AR, Dong EY, Andry JP, Bulinski JC, Ateshian GA, Hung CT. Coculture of engineered cartilage with primary chondrocytes induces expedited growth. Clin Orthop Relat Res 2011; 469:2735-43. [PMID: 21267800 PMCID: PMC3171525 DOI: 10.1007/s11999-011-1772-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Soluble factors released from chondrocytes can both enhance and induce chondrocyte-like behavior in cocultured dedifferentiated cells. The ability to similarly prime and modulate biosynthetic activity of differentiated cells encapsulated in a three-dimensional environment is unknown. QUESTIONS/PURPOSES To understand the effect of coculture on engineered cartilage, we posed three hypotheses: (1) coculturing with a monolayer of chondrocytes ("chondrocyte feeder layer") expedites and increases engineered tissue growth; (2) expedited growth arises from paracrine effects; and (3) these effects are dependent on the specific morphology and expression of the two-dimensional feeder cells. METHODS In three separate studies, chondrocyte-laden hydrogels were cocultured with chondrocyte feeder layers. Mechanical properties and biochemical content were quantified to evaluate tissue properties. Histology and immunohistochemistry stains were observed to visualize each constituent's distribution and organization. RESULTS Coculture with a chondrocyte feeder layer led to stiffer tissue constructs (Young's modulus and dynamic modulus) with greater amounts of glycosaminoglycan and collagen. This was dependent on paracrine signaling between the two populations of cells and was directly modulated by the rounded morphology and expression of the feeder cell monolayer. CONCLUSIONS These findings suggest a potential need to prime and modulate tissues before implantation and present novel strategies for enhancing medium formulations using soluble factors released by feeder cells. CLINICAL RELEVANCE Determining the soluble factors present in the coculture system can enhance a chondrogenic medium formulation for improved growth of cartilage substitutes. The feeder layer strategy described here may also be used to prime donor cartilage allografts before implantation to increase their success in vivo.
Collapse
Affiliation(s)
- Andrea R. Tan
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027 USA
| | - Elizabeth Y. Dong
- Department of Biological Sciences, Columbia University, New York, NY USA
| | - James P. Andry
- Department of Orthopaedic Surgery, St Luke’s-Roosevelt Hospital Center, New York, NY USA
| | - J. Chloë Bulinski
- Department of Biological Sciences, Columbia University, New York, NY USA
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027 USA
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, 351 Engineering Terrace, 1210 Amsterdam Avenue, New York, NY 10027 USA
| |
Collapse
|
161
|
Santoro R, Krause C, Martin I, Wendt D. On-line monitoring of oxygen as a non-destructive method to quantify cells in engineered 3D tissue constructs. J Tissue Eng Regen Med 2011; 6:696-701. [PMID: 21932277 DOI: 10.1002/term.473] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2011] [Revised: 06/06/2011] [Accepted: 07/05/2011] [Indexed: 11/05/2022]
Abstract
Regulatory guidelines have established the importance of introducing quantitative quality controls during the production and/or at the time of release of cellular grafts for clinical applications. In this study we aimed to determine whether on-line measurements of oxygen can be used as a non-destructive method to estimate the number of chondrocytes within an engineered cartilage graft. Human chondrocytes were seeded and cultured in a perfusion bioreactor, and oxygen levels in the culture medium were continuously monitored at the inlet and outlet of the bioreactor chamber throughout the culture period. We found that the drop in oxygen across the perfused construct was linearly correlated with the number of cells per construct (R(2) = 0.82, p < 0.0001). The method was valid for a wide range of cell numbers, including cell densities currently used in the manufacture of cartilage grafts for clinical applications. Given that few or no non-destructive assays that quantitatively characterize an engineered construct currently exist, this non-invasive method could represent a relevant instrument in regulatory compliant manufacturing of engineered grafts meeting specific quality criteria.
Collapse
Affiliation(s)
- R Santoro
- Departments of Surgery and of Biomedicine, University Hospital Basel, Switzerland
| | - C Krause
- PreSens Precision Sensing GmbH, Regensburg, Germany
| | - I Martin
- Departments of Surgery and of Biomedicine, University Hospital Basel, Switzerland
| | - D Wendt
- Departments of Surgery and of Biomedicine, University Hospital Basel, Switzerland
| |
Collapse
|
162
|
Borzacchiello A, Gloria A, Mayol L, Dickinson S, Miot S, Martin I, Ambrosio L. Natural/synthetic porous scaffold designs and properties for fibro-cartilaginous tissue engineering. J BIOACT COMPAT POL 2011. [DOI: 10.1177/0883911511420149] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The goal of this study was to produce and characterize the scaffolds by combining the advantages of both natural and synthetic polymers for engineering fibro-cartilaginous tissues. Porous three-dimensional composite scaffolds were produced based on glycosaminoglycans and hyaluronic acid (HYAFF11) reinforced with polycaprolactone. The mechanical properties of scaffolds were evaluated as a function of time and compared with those of scaffolds seeded with human chondrocytes (constructs) and cultured in vitro up to 6 weeks. The composite scaffolds had a porosity of 68% with interconnected macropores with average pore sizes of 200 μm, an equilibrium swelling of 350%, and a predominant elastic behavior, typical of a macromolecular gel. The composite constructs maintained chondrocyte phenotype and degraded with the deposition of macromolecules synthesized by the cells. The scaffold presented mechanical properties and the ability to dissipate energy similar to the fibro-cartilaginous tissue.
Collapse
Affiliation(s)
- A. Borzacchiello
- Institute of Composite and Biomedical Materials-C.N.R and Interdisciplinary Research Centre on Biomaterials-University of Naples “Federico II” Piazzale Tecchio 80, 80125 Naples, Italy,
| | - A. Gloria
- Institute of Composite and Biomedical Materials-C.N.R and Interdisciplinary Research Centre on Biomaterials-University of Naples “Federico II” Piazzale Tecchio 80, 80125 Naples, Italy
| | - L. Mayol
- School of Biotechnological Sciences, Department of Pharmaceutical and Toxicological Chemistry, University of Naples, Federico , Via D. Montesano 49, 80131 Naples, Italy
| | - Sally Dickinson
- Department of Cellular & Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| | - S. Miot
- Departments of Surgery and of Biomedicine, University Hospital Basel, Switzerland
| | - I. Martin
- Departments of Surgery and of Biomedicine, University Hospital Basel, Switzerland
| | - L. Ambrosio
- Institute of Composite and Biomedical Materials-C.N.R and Interdisciplinary Research Centre on Biomaterials-University of Naples “Federico II” Piazzale Tecchio 80, 80125 Naples, Italy
| |
Collapse
|
163
|
Sampat SR, O'Connell GD, Fong JV, Alegre-Aguarón E, Ateshian GA, Hung CT. Growth factor priming of synovium-derived stem cells for cartilage tissue engineering. Tissue Eng Part A 2011; 17:2259-65. [PMID: 21542714 PMCID: PMC3161099 DOI: 10.1089/ten.tea.2011.0155] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 05/03/2011] [Indexed: 11/12/2022] Open
Abstract
This study investigated the potential use of synovium-derived stem cells (SDSCs) as a cell source for cartilage tissue engineering. Harvested SDSCs from juvenile bovine synovium were expanded in culture in the presence (primed) or absence (unprimed) of growth factors (1 ng/mL transforming growth factor-β(1), 10 ng/mL platelet-derived growth factor-ββ, and 5 ng/mL basic fibroblast growth factor-2) and subsequently seeded into clinically relevant agarose hydrogel scaffolds. Constructs seeded with growth factor-primed SDSCs that received an additional transient application of transforming growth factor-β(3) for the first 21 days (release) exhibited significantly better mechanical and biochemical properties compared to constructs that received sustained growth factor stimulation over the entire culture period (continuous). In particular, the release group exhibited a Young's modulus (267±96 kPa) approaching native immature bovine cartilage levels, with corresponding glycosaminoglycan content (5.19±1.45%ww) similar to native values, within 7 weeks of culture. These findings suggest that SDSCs may serve as a cell source for cartilage tissue engineering applications.
Collapse
Affiliation(s)
- Sonal R Sampat
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | | | | | | | | | | |
Collapse
|
164
|
Sá-Lima H, Tuzlakoglu K, Mano JF, Reis RL. Thermoresponsive poly(N-isopropylacrylamide)-g-methylcellulose hydrogel as a three-dimensional extracellular matrix for cartilage-engineered applications. J Biomed Mater Res A 2011; 98:596-603. [DOI: 10.1002/jbm.a.33140] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Revised: 04/04/2011] [Accepted: 04/05/2011] [Indexed: 11/10/2022]
|
165
|
Saleh FA, Genever PG. Turning round: multipotent stromal cells, a three-dimensional revolution? Cytotherapy 2011; 13:903-12. [PMID: 21696237 DOI: 10.3109/14653249.2011.586998] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mesenchymal stromal cells (MSC) can be isolated from adult tissues and induced to differentiate into skeletal cells, such as osteoblasts, chondrocytes and adipocytes. Consequently, ex vivo MSC are valuable systems for studying the mechanisms that control tissue-context lineage commitment and may offer broad therapeutic applications in the orthopedic theater and beyond. To date, most of these studies have used MSC grown on two-dimensional (2-D) plastic surfaces. The use of three-dimensional (3-D) in vitro growth techniques for MSC may accelerate these areas of research by providing a more representative 'in vivo-like' environment, where cells interact with each other and their cellular products, rather than a plastic surface. We introduce some of the techniques used for 3-D in vitro cultures and how they relate to the MSC field. We will present evidence of how MSC grown as 3-D spheroids not only permits appropriate MSC-like behavior, but appears to promote their stem-cell attributes and therapeutic benefit in applications ranging from regenerative medicine to anti-inflammatory treatments and cancer therapy. 3-D culture techniques also allow de/reconstruction of the specialized in vivo niche of the tissue-resident stem cell where microenvironmental influences can be recognized.
Collapse
Affiliation(s)
- Fatima A Saleh
- Department of Biology (Area 9), University of York, York, UK
| | | |
Collapse
|
166
|
Feng J, Mantesso A, De Bari C, Nishiyama A, Sharpe PT. Dual origin of mesenchymal stem cells contributing to organ growth and repair. Proc Natl Acad Sci U S A 2011; 108:6503-8. [PMID: 21464310 PMCID: PMC3081015 DOI: 10.1073/pnas.1015449108] [Citation(s) in RCA: 306] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
In many adult tissues, mesenchymal stem cells (MSCs) are closely associated with perivascular niches and coexpress many markers in common with pericytes. The ability of pericytes to act as MSCs, however, remains controversial. By using genetic lineage tracing, we show that some pericytes differentiate into specialized tooth mesenchyme-derived cells--odontoblasts--during tooth growth and in response to damage in vivo. As the pericyte-derived mesenchymal cell contribution to odontoblast differentiation does not account for all cell differentiation, we identify an additional source of cells with MSC-like properties that are stimulated to migrate toward areas of tissue damage and differentiate into odontoblasts. Thus, although pericytes are capable of acting as a source of MSCs and differentiating into cells of mesenchymal origin, they do so alongside other MSCs of a nonpericyte origin. This study identifies a dual origin of MSCs in a single tissue and suggests that the pericyte contribution to MSC-derived mesenchymal cells in any given tissue is variable and possibly dependent on the extent of the vascularity.
Collapse
Affiliation(s)
- Jifan Feng
- Department of Craniofacial Development and Comprehensive Biomedical Research Centre, Dental Institute, Kings College London, London SE1 9RT, United Kingdom
| | - Andrea Mantesso
- Department of Craniofacial Development and Comprehensive Biomedical Research Centre, Dental Institute, Kings College London, London SE1 9RT, United Kingdom
- Department of Oral Pathology, Dental Institute, University of Sao Paulo, CEP 05508-900, Sao Paulo, Brazil
| | - Cosimo De Bari
- Division of Applied Medicine, University of Aberdeen, Aberdeen AB25 22D, United Kingdom; and
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269-4243
| | - Paul T. Sharpe
- Department of Craniofacial Development and Comprehensive Biomedical Research Centre, Dental Institute, Kings College London, London SE1 9RT, United Kingdom
| |
Collapse
|
167
|
Pretzel D, Linss S, Rochler S, Endres M, Kaps C, Alsalameh S, Kinne RW. Relative percentage and zonal distribution of mesenchymal progenitor cells in human osteoarthritic and normal cartilage. Arthritis Res Ther 2011; 13:R64. [PMID: 21496249 PMCID: PMC3132059 DOI: 10.1186/ar3320] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 02/15/2011] [Accepted: 04/15/2011] [Indexed: 01/22/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSC) are highly attractive for use in cartilage regeneration. To date, MSC are usually recruited from subchondral bone marrow using microfracture. Recent data suggest that isolated cells from adult human articular cartilage, which express the combination of the cell-surface markers CD105 and CD166, are multi-potent mesenchymal progenitor cells (MPC) with characteristics similar to MSC. MPC within the cartilage matrix, the target of tissue regeneration, may provide the basis for in situ regeneration of focal cartilage defects. However, there is only limited information concerning the presence/abundance of CD105+/CD166+ MPC in human articular cartilage. The present study therefore assessed the relative percentage and particularly the zonal distribution of cartilage MPC using the markers CD105/CD166. Methods Specimens of human osteoarthritic (OA; n = 11) and normal (n = 3) cartilage were used for either cell isolation or immunohistochemistry. Due to low numbers, isolated cells were expanded for 2 weeks and then analyzed by flow cytometry (FACS) or immunofluorescence in chamber slides for the expression of CD105 and CD166. Following immunomagnetic separation of CD166+/- OA cells, multi-lineage differentiation assays were performed. Also, the zonal distribution of CD166+ cells within the matrix of OA and normal cartilage was analyzed by immunohistochemistry. Results FACS analysis showed that 16.7 ± 2.1% (mean ± SEM) of OA and 15.3 ± 2.3 of normal chondrocytes (n.s.) were CD105+/CD166+ and thus carried the established MPC marker combination. Similarly, 13.2% ± 0.9% and 11.7 ± 2.1 of CD105+/CD166+cells, respectively, were identified by immunofluorescence in adherent OA and normal chondrocytes. The CD166+ enriched OA cells showed a stronger induction of the chondrogenic phenotype in differentiation assays than the CD166+ depleted cell population, underlining the chondrogenic potential of the MPC. Strikingly, CD166+ cells in OA and normal articular cartilage sections (22.1 ± 1.7% and 23.6% ± 1.4%, respectively; n.s.) were almost exclusively located in the superficial and middle zone. Conclusions The present results underline the suitability of CD166 as a biomarker to identify and, in particular, localize and/or enrich resident MPC with a high chondrogenic potential in human articular cartilage. The percentage of MPC in both OA and normal cartilage is substantially higher than previously reported, suggesting a yet unexplored reserve capacity for regeneration.
Collapse
Affiliation(s)
- David Pretzel
- Experimental Rheumatology Unit, Department of Orthopedics, University Hospital Jena, Klosterlausnitzer Str, 81, Eisenberg, D-07607, Germany.
| | | | | | | | | | | | | |
Collapse
|
168
|
Sassi N, Laadhar L, Driss M, Kallel-Sellami M, Sellami S, Makni S. The role of the Notch pathway in healthy and osteoarthritic articular cartilage: from experimental models to ex vivo studies. Arthritis Res Ther 2011; 13:208. [PMID: 21457519 PMCID: PMC3132010 DOI: 10.1186/ar3255] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Osteoarthritis is the most prevalent form of arthritis in the world. With the progressive ageing of the population, it is becoming a major public health problem. The involvement of certain signaling pathways, such as the Notch pathway, during cartilage pathology has been reported. In this review, we report on studies that investigated the expression pattern of the Notch family members in articular cartilage and the eventual involvement of this pathway in the modulation of the physiology and pathology of chondrocytes. Temporal and/or spatial modulation of this signaling pathway may help these cells to synthesize a new functional extracellular matrix and restore the functional properties of the articular cartilage.
Collapse
Affiliation(s)
- Nadia Sassi
- Osteoarthritis-osteoporosis Research Laboratory, Rheumatology Department, LaRabta Hospital, 1007 Tunis, Tunisia.
| | | | | | | | | | | |
Collapse
|
169
|
Francioli SE, Candrian C, Martin K, Heberer M, Martin I, Barbero A. Effect of three-dimensional expansion and cell seeding density on the cartilage-forming capacity of human articular chondrocytes in type II collagen sponges. J Biomed Mater Res A 2011; 95:924-31. [PMID: 20845491 DOI: 10.1002/jbm.a.32917] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chondrocytes for tissue engineering strategies are typically expanded in monolayer (2D), leading to cell dedifferentiation but allowing to generate large cell numbers for seeding into scaffolds. Direct chondrocyte culture in scaffolds, instead, may support better maintenance of the differentiated phenotype but reduce the extent of proliferation and thus the resulting cell density. This study investigates whether the quality of cartilaginous tissues generated in vitro by human articular chondrocytes (HAC) on type II collagen sponges is enhanced (1) by direct expansion on the scaffolds (3D), as compared with standard 2D, or (2) by increasing cell seeding density, which in turn requires extensive 2D expansion. Three-dimensional expansion of HAC on the scaffolds, as compared with 2D expansion for the same number of doublings, better maintained the chondrocytic phenotype of the expanded cells (13.7-fold higher levels of type II collagen mRNA) but did not enhance their accumulation of glycosaminoglycan (GAG) following chondrogenic culture. Instead, increasing the HAC seeding density in the scaffolds (from 25 × 10(3) to 66 × 10(3) cells/mm(3)) significantly improved chondrogenesis (up to 3.3-fold higher GAG accumulation and up to 9.3-fold higher type II collagen mRNA), even if seeded cells had to be expanded and dedifferentiated more extensively in 2D to reach the required cell numbers. This study indicates that, under the specific conditions tested, a high-seeding density of HAC in 3D scaffolds is more critical for the generation of cartilaginous constructs than the stage of cell differentiation reached following expansion.
Collapse
Affiliation(s)
- Silvia E Francioli
- Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
170
|
Koo J, Kim KI, Min BH, Lee GM. Controlling medium osmolality improves the expansion of human articular chondrocytes in serum-free media. Tissue Eng Part C Methods 2011; 16:957-63. [PMID: 20001269 DOI: 10.1089/ten.tec.2009.0525] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To investigate the effects of medium osmolality on the expansion of human articular chondrocytes (HACs) with serum-free media (SFM), proprietary SFM of various osmolalities (290, 320, 350, 400, and 450 mOsm/kg), supplemented with components known to enhance chondrocyte growth, were constructed by the adjustment of NaCl concentration. It was found that HACs obtained better expansion in SFM at osmolalites lower than the average osmolality (400 mOsm/kg) of human articular cartilage in vivo. SFM at 290, 320, and 350 mOsm/kg showed similar growth, attaining up to a 1.55-fold increase in the proliferation rate compared with SFM at 400 mOsm/kg. Increasing SFM osmolality to 450 mOsm/kg resulted in a proliferation rate of 0.65-fold lower than at 400 mOsm/kg. Chondrogenic capacity was also examined via three-dimensional pellet cultures in a chondrogenic medium with HACs expanded in 320 and 400 mOsm/kg SFM. Biochemical, histological, and immunohistochemical analyses revealed similar glycosaminoglycan and collagen type II contents in both groups. Taken together, these results show that the expansion of HACs in SF cultures can be improved by adjusting the medium osmolality to be within the range of 290-350 mOsm/kg and that controlling medium osmolality during monolayer cultures does not deter the tissue-forming capability of the cells.
Collapse
Affiliation(s)
- Jane Koo
- Department of Biological Sciences, KAIST, Yuseong-gu, Daejeon, Republic of Korea
| | | | | | | |
Collapse
|
171
|
van Buul GM, van Osch GJVM. Musculoskeletal Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
172
|
Lovati AB, Corradetti B, Lange Consiglio A, Recordati C, Bonacina E, Bizzaro D, Cremonesi F. Comparison of equine bone marrow-, umbilical cord matrix and amniotic fluid-derived progenitor cells. Vet Res Commun 2010; 35:103-21. [PMID: 21193959 DOI: 10.1007/s11259-010-9457-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2010] [Indexed: 01/08/2023]
Abstract
The aim of the study was to compare in vitro the stemness features of horse progenitor cells derived from bone marrow (BM-MSCs), amniotic fluid (AF-MSCs) and umbilical cord matrix (EUC-MSCs). It has been suggested that there may be a stem cell population within both umbilical cord matrix and amniotic fluid. However, little knowledge exists about the characteristics of these progenitor cells within these sources in the equine species. This study wanted to investigate an alternative and non-invasive stem cell source for the equine tissue engineering and to learn more about the properties of these cells for future cell banking. Bone marrow, umbilical cord and amniotic fluid samples were harvested from different horses. Cells were analyzed for proliferation, immunocytochemical, stem cell gene expression and multilineage plasticity. BM- and AF-MSCs took similar time to reach confluence and showed comparable plating efficiency. All cell lines expressed identical stem cell markers and capability to differentiate towards osteogenic lineage. Almost all cell lines differentiated into the adipogenic lineage as demonstrated by cytochemical staining, even if no adipose gene expression was detectable for AF-MSCs. AF- and EUC-MSCs showed a limited chondrogenic differentiation compared with BM-MSCs as demonstrated by histological and biochemical analyses. These findings suggest that AF-MSCs appeared to be a readily obtainable and highly proliferative cell line from an uninvasive source that may represent a good model system for stem cell biology. More studies are needed to investigate their multilineage potential. EUC-MSCs need to be further investigated regarding their particular behavior in vitro represented by spheroid formation.
Collapse
Affiliation(s)
- Arianna Barbara Lovati
- Department of Veterinary Clinical Science, Equine Reproduction Unit, Università degli Studi di Milano, Strada dell'Università 6, Località Polledra, 26900 Lodi, Italy.
| | | | | | | | | | | | | |
Collapse
|
173
|
Grote MJ, Palumberi V, Wagner B, Barbero A, Martin I. Dynamic formation of oriented patches in chondrocyte cell cultures. J Math Biol 2010; 63:757-77. [DOI: 10.1007/s00285-010-0390-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 11/23/2010] [Indexed: 12/01/2022]
|
174
|
Boeuf S, Richter W. Chondrogenesis of mesenchymal stem cells: role of tissue source and inducing factors. Stem Cell Res Ther 2010; 1:31. [PMID: 20959030 PMCID: PMC2983444 DOI: 10.1186/scrt31] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are an attractive cell source for cell therapy in cartilage. Although their therapeutic potential is clear, the requirements and conditions for effective induction of chondrogenesis in MSCs and for the production of a stable cartilaginous tissue by these cells are far from being understood. Different sources of MSCs have been considered for cartilage tissue engineering, mainly based on criteria of availability, as for adipose tissue, or of proximity to cartilage and the joint environment in vivo, as for bone marrow and synovial tissues. Focussing on human MSCs, this review will provide an overview of studies featuring comparative analysis of the chondrogenic differentiation of MSCs from different sources. In particular, it will examine the influence of the cells' origin on the requirements for the induction of chondrogenesis and on the phenotype achieved by the cells after differentiation.
Collapse
Affiliation(s)
- Stephane Boeuf
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | | |
Collapse
|
175
|
Stojkovska J, Bugarski B, Obradovic B. Evaluation of alginate hydrogels under in vivo-like bioreactor conditions for cartilage tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2010; 21:2869-2879. [PMID: 20717710 DOI: 10.1007/s10856-010-4135-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 07/17/2010] [Indexed: 05/29/2023]
Abstract
Alginate hydrogels in forms of discs and packed beds of microbeads (~800 μm) were tested in a novel bioreactor at 10% strain using two regimes: at a loading rate of 337.5 μm/s and at sequential increments of 50 μm displacement every 30 min. Compressive strength increased with the increase in alginate concentration (1.5 vs. 2% w/w) and the content of guluronic residues (38.5 vs. 67%). Packed beds of microbeads exhibited significantly higher (~1.5-3.4 fold) compression moduli than the respective discs indicating the effects of gel form and entrapped water. Short-term cultivation of microbeads with immobilized bovine calf chondrocytes (1.5% w/w, 33 × 10(6) cells/ml) under biomimetic conditions (dynamic compression: 1 h on/1 h off, 0.42 Hz, 10% strain) resulted in cell proliferation and bed compaction, so that the compression modulus slightly increased. Thus, the novel bioreactor demonstrated advantages in evaluation of biomaterial properties and cell-biomaterial interactions under in vivo-like settings.
Collapse
Affiliation(s)
- Jasmina Stojkovska
- Department of Chemical Engineering, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | | | | |
Collapse
|
176
|
Kasashima Y, Ueno T, Tomita A, Goodship AE, Smith RKW. Optimisation of bone marrow aspiration from the equine sternum for the safe recovery of mesenchymal stem cells. Equine Vet J 2010; 43:288-94. [PMID: 21492205 DOI: 10.1111/j.2042-3306.2010.00215.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
REASONS FOR PERFORMING STUDY Mesenchymal stem cell (MSC) therapy for orthopaedic disease is being used with increasing frequency; there is a need to define a safe, reliable and effective technique for the recovery of MSCs from the sternum of the horse. OBJECTIVES To describe an optimised safe technique for obtaining bone marrow-derived MSCs from the sternum of the Thoroughbred horse. METHODS The anatomical relationship of the sternum with the heart and internal anatomy was demonstrated in cadavers. Sternal anatomy was evaluated ultrasonographically and after midline sectioning. Sternebrae were examined histologically after aspiration to determine the effect of needle insertion. The quality of the aspirate was evaluated as the number of colony-forming units from sequential and separately aspirated 5 ml aliquots and assessed for their multipotency using trilineage differentiation. RESULTS The optimal safe location for the needle was the 5th sternebra because it had a safe dorsoventral thickness and was cranial to the apex of the heart. This sternebra could be reliably identified ultrasonographically. Aspirates could also be obtained from the 4th and 6th sternebrae, although the former is between the front limbs and the latter closer to the heart. Minimal disruption of the internal bony architecture was seen after needle insertion through the thin outer cortex and the first 5 ml aliquot contained the greatest number of colony-forming units of mesenchymal stem cells with trilineage capabilities. CONCLUSIONS Accurate placement of a Jamshidi needle into the medullary cavity of the 4th-6th individual sternebrae is facilitated by the use of ultrasonography and enables aspiration of bone marrow reliably with minimal damage to the sternum and risk to the horse. POTENTIAL CLINICAL RELEVANCE Sternal marrow aspiration as described is a safe and reliable technique to obtain MSCs for orthopaedic cell-based therapies.
Collapse
Affiliation(s)
- Y Kasashima
- Clinical Science and Pathobiology Division, Equine Research Institute, Japan Racing Association, Tokami-cho, Japan.
| | | | | | | | | |
Collapse
|
177
|
Candrian C, Miot S, Wolf F, Bonacina E, Dickinson S, Wirz D, Jakob M, Valderrabano V, Barbero A, Martin I. Are ankle chondrocytes from damaged fragments a suitable cell source for cartilage repair? Osteoarthritis Cartilage 2010; 18:1067-76. [PMID: 20434576 DOI: 10.1016/j.joca.2010.04.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 04/16/2010] [Accepted: 04/21/2010] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To characterize the post-expansion cartilage-forming capacity of chondrocytes harvested from detached fragments of osteochondral lesions (OCLs) of ankle joints (Damaged Ankle Cartilage Fragments, DACF), with normal ankle cartilage (NAC) as control. DESIGN DACF were obtained from six patients (mean age: 35 years) with symptomatic OCLs of the talus, while NAC were from 10 autopsies (mean age: 55 years). Isolated chondrocytes were expanded for two passages and then cultured in pellets for 14 days or onto HYAFF-11 meshes (FAB, Italy) for up to 28 days. Resulting tissues were assessed histologically, biochemically [glycosaminoglycan (GAG), DNA and type II collagen (CII)] and biomechanically. RESULTS As compared to NAC, DACF contained significantly lower amounts of DNA (3.0-fold), GAG (5.3-fold) and CII (1.5-fold) and higher amounts of type I collagen (6.2-fold). Following 14 days of culture in pellets, DACF-chondrocytes generated tissues less intensely stained for Safranin-O and CII, with significantly lower GAG contents (2.8-fold). After 28 days of culture onto HYAFF((R))-11, tissues generated by DACF-chondrocytes were less intensely stained for Safranin-O and CII, contained significantly lower amounts of GAG (1.9-fold) and CII (1.4-fold) and had lower equilibrium (1.7-fold) and dynamic pulsatile modulus (3.3-fold) than NAC-chondrocytes. CONCLUSION We demonstrated that DACF-chondrocytes have inferior cartilage-forming capacity as compared to NAC-chondrocytes, possibly resulting from environmental changes associated with trauma/disease. The study opens some reservations on the use of DACF-derived cells for the repair of ankle cartilage defects, especially in the context of tissue engineering-based approaches.
Collapse
Affiliation(s)
- C Candrian
- Department of Biomedicine, University Hospital Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Henderson JH, Ginley NM, Caplan AI, Niyibizi C, Dennis JE. Low oxygen tension during incubation periods of chondrocyte expansion is sufficient to enhance postexpansion chondrogenesis. Tissue Eng Part A 2010; 16:1585-93. [PMID: 19958052 DOI: 10.1089/ten.tea.2009.0411] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To determine whether low oxygen (O(2)) tension during expansion affects the matrix density, as well as quantity, of cartilage formed, and to determine whether application of low O(2) tension during incubation periods alone is sufficient to modulate chondrogenic expression, rabbit chondrocytes expanded at either 21% O(2) or 5% O(2) were analyzed for glycosaminoglycan (GAG) and DNA content, total collagen, and gene expression during expansion and postexpansion aggregate cultures. When cultured as aggregates at 21% O(2), chondrocytes expanded at 5% O(2) produced cartilage aggregates that contained more total GAG, GAG per wet weight, GAG per DNA, and total collagen than chondrocytes expanded at 21% O(2). Less of an effect on GAG and collagen content was observed when aggregate culture was performed at 5% O(2). Real-time polymerase chain reaction analysis of COL2A1 expression showed upregulated levels of type IIA (an early marker) and IIB (a late marker) during expansion and elevated levels of type IIB during aggregate culture in chondrocytes expanded in low O(2). The application of low O(2) tension during incubation periods of chondrocyte expansion enhances the ultimate cartilage matrix density and quantity, and this enhancement can be achieved through the use of an O(2) control incubator.
Collapse
Affiliation(s)
- James H Henderson
- Department of Biology (Skeletal Research Center), Case Western Reserve University, Cleveland, OH, USA.
| | | | | | | | | |
Collapse
|
179
|
Kurose R, Ichinohe S, Tajima G, Horiuchi S, Kurose A, Sawai T, Shimamura T. Characterization of human synovial fluid cells of 26 patients with osteoarthritis knee for cartilage repair therapy. Int J Rheum Dis 2010; 13:68-74. [PMID: 20374387 DOI: 10.1111/j.1756-185x.2009.01456.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
AIM To investigate the possibility of chondrogenic differentiation and cartilage repair of synovial fluid cells of osteoarthritis (OA) knee. METHODS Synovial fluids from 26 patients with OA knee were aspirated from each knee joint and cultured in vitro. The morphology of cultured synovial fluid cells, cell proliferation rate, the phenotype, and chondrogenic differentiation were analyzed in in vitro. Also, human autologous synovial fluid cells were transplanted to OA cartilage, and the cells were traced in ex vivo. RESULTS In 19 of 26 materials, the cells proliferated satisfactorily. The cell proliferation in six materials was very slow and one material contaminated. Culture-expanded synovial fluid cells had a fibroblastic morphology and the phenotype was negative for CD10, CD14, and CD45, and positive for CD13, CD44, and CD105. Pellet culture of synovial fluid cells showed chondrogenic differentiation. In the ex vivo study, autologous transplanted synovial fluid cells were observed in repaired or enhanced regenerative cartilage areas and showed a tendency to infiltrate the original degenerative cartilage of OA. CONCLUSIONS This study proved the possibility of chondrogenic differentiation of synovial fluid cells of OA knee joints despite the pathologic environment within a diseased joint. Synovial fluid cells were actually heterogeneous cells but they showed chondrogenic differentiation, similar to that of bone marrow-derived mesenchymal progenitor cells (BMMPCs). The Ex vivo study suggested that synovial fluid cells had a tendency to adhere to OA degenerative cartilage in humans.
Collapse
Affiliation(s)
- Rie Kurose
- Department of Orthopaedic Surgery, Iwate Medical University, Morioka, Japan.
| | | | | | | | | | | | | |
Collapse
|
180
|
Sassi N, Laadhar L, Mahjoub M, Driss M, Zitouni M, Benromdhane K, Makni S, Sellami S. Expression of Notch family members in cultured murine articular chondrocytes. Biotech Histochem 2010; 84:313-20. [PMID: 19562571 DOI: 10.3109/10520290903054382] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The Notch family is involved in cell differentiation during embryogenesis. Osteoarthritic chondrocytes undergo morphological and biochemical changes leading to the de-differentiation process. In the study reported here, we were interested in the involvement of the Notch pathway in murine articular chondrocyte de-differentiation. Articular chondrocytes were subjected to several cell culture passages and treated with or without a Notch inhibitor, N-[N-(3, 5-difluorophenacetyl-L-alanyl)]-S-phenylglycine t-Butyl Ester (DAPT). Chondrocyte morphology was studied using optical microscopy. Immunocytochemistry and immunoblot were performed to study the expression of collagens and Notch family members. Without DAPT treatment, chondrocyte de-differentiation resulted in fibroblast-like morphology. This was confirmed by immunocytochemical staining and immunoblot analysis, which showed an increase in collagen type I (col I) and a decrease in collagen type II (col II) expression. With DAPT treatment, de-differentiation was delayed. Immunocytochemistry and immunoblot analysis showed during the first passages inhibition of col II expression, which then was re-instituted during the last passage, suggesting chondrocyte re-differentiation. In the study reported here, we showed that inhibition of the Notch receptor not only delayed the de-differentiation process, but also chondrocyte re-differentiation, which confirms the involvement of the Notch pathway in chondrocyte de-differentiation.
Collapse
Affiliation(s)
- N Sassi
- Osteoarthritis-Osteoporosis Research Laboratory, Department of Rheumatology, LaRabta Hospital, Tunis, Tunisia.
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Abstract
Over the past 40 years there have been giant steps forward in our understanding of cellular and molecular biology that have given us the framework by which to understand tissue organization and tissue function on a range of scales. However, although the progress has been great, the more we have discovered, the more we are aware of what we don't yet know. In this article, I would like to flag up some issues of cartilage biology, function and pathology where we still have significant ignorance. As scientists we all provide contributions to add to the greater understanding of science and progress is on a broad front, but gaps are left where particular difficulty is encountered and in life sciences it is no different. Progress is fast where new knowledge and techniques pave the way, but where study is complex and relevant techniques poorly developed the gaps are left behind. In cartilage research and matrix biology, the gaps can particularly be seen at interfaces between disciplines and where technology development has lagged behind and in the particular challenges of understanding how molecular properties can explain tissue macro properties.
Collapse
Affiliation(s)
- Timothy E Hardingham
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.
| |
Collapse
|
182
|
Quintin A, Schizas C, Scaletta C, Jaccoud S, Applegate LA, Pioletti DP. Plasticity of fetal cartilaginous cells. Cell Transplant 2010; 19:1349-57. [PMID: 20447338 DOI: 10.3727/096368910x506854] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Tissue-specific stem cells found in adult tissues can participate in the repair process following injury. However, adult tissues, such as articular cartilage and intervertebral disc, have low regeneration capacity, whereas fetal tissues, such as articular cartilage, show high regeneration ability. The presence of fetal stem cells in fetal cartilaginous tissues and their involvement in the regeneration of fetal cartilage is unknown. The aim of the study was to assess the chondrogenic differentiation and the plasticity of fetal cartilaginous cells. We compared the TGF-β3-induced chondrogenic differentiation of human fetal cells isolated from spine and cartilage tissues to that of human bone marrow stromal cells (BMSC). Stem cell surface markers and adipogenic and osteogenic plasticity of the two fetal cell types were also assessed. TGF-β3 stimulation of fetal cells cultured in high cell density led to the production of aggrecan, type I and II collagens, and variable levels of type X collagen. Although fetal cells showed the same pattern of surface stem cell markers as BMSCs, both type of fetal cells had lower adipogenic and osteogenic differentiation capacity than BMSCs. Fetal cells from femoral head showed higher adipogenic differentiation than fetal cells from spine. These results show that fetal cells are already differentiated cells and may be a good compromise between stem cells and adult tissue cells for a cell-based therapy.
Collapse
Affiliation(s)
- Aurelie Quintin
- Cellular Therapy Unit, Department of Musculoskeletal Medicine, University Hospital Center and University of Lausanne, CHUV-UNIL, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
183
|
Rui YF, Lui PPY, Li G, Fu SC, Lee YW, Chan KM. Isolation and characterization of multipotent rat tendon-derived stem cells. Tissue Eng Part A 2010; 16:1549-1558. [PMID: 20001227 DOI: 10.1089/ten.tea.2009.0529] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Stem cells have recently been isolated from humans and mice but not from rat tendon tissue. This study reports the isolation and characterization of stem cells from rat tendon. Nucleated cells isolated from rat flexor tendon tissues after collagenase digestion were plated at a low cell density to allow the selective proliferation of tendon-derived stem cells. About 1-2% of the cells isolated under this optimized culturing condition showed clonogenicity, high proliferative potential at low seeding density, and osteogenic, chondrogenic, and adipogenic multidifferentiation potential. These cells were CD44(+), CD90(+), CD34(-), and CD31(-). Although they shared some common properties with mesenchymal stem cells, they also exhibited their unique characteristics by expressing tenogenic and chondrogenic markers. There was expression of tenogenic markers, including alpha-smooth muscle actin, tenascin C, and tenomodulin, but not collagen type I at passage 0 (P0) and P3. Expression of a chondrogenic marker, aggrecan, was observed at P0 and P3, whereas expression of collagen type II was observed in few cells only at P3. The successful isolation of tendon-derived stem cells under the optimized growth and differentiation conditions was useful for future stem-cell-based tissue regenerative studies as well as studies on their roles in tendon physiology, healing, and disorders using the rat model.
Collapse
Affiliation(s)
- Yun-Feng Rui
- Department of Orthopaedics and Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
184
|
Bader A, Macchiarini P. Moving towards in situ tracheal regeneration: the bionic tissue engineered transplantation approach. J Cell Mol Med 2010; 14:1877-89. [PMID: 20406329 PMCID: PMC3823270 DOI: 10.1111/j.1582-4934.2010.01073.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
In June 2008, the world's first whole tissue-engineered organ - the windpipe - was successfully transplanted into a 31-year-old lady, and about 18 months following surgery she is leading a near normal life without immunosuppression. This outcome has been achieved by employing three groundbreaking technologies of regenerative medicine: (i) a donor trachea first decellularized using a detergent (without denaturing the collagenous matrix), (ii) the two main autologous tracheal cells, namely mesenchymal stem cell derived cartilage-like cells and epithelial respiratory cells and (iii) a specifically designed bioreactor that reseed, before implantation, the in vitro pre-expanded and pre-differentiated autologous cells on the desired surfaces of the decellularized matrix. Given the long-term safety, efficacy and efforts using such a conventional approach and the potential advantages of regenerative implants to make them available for anyone, we have investigated a novel alternative concept how to fully avoid in vitro cell replication, expansion and differentiation, use the human native site as micro-niche, potentiate the human body's site-specific response by adding boosting, permissive and recruitment impulses in full respect of sociological and regulatory prerequisites. This tissue-engineered approach and ongoing research in airway transplantation is reviewed and presented here.
Collapse
Affiliation(s)
- Augustinus Bader
- Centre for Biotechnology and Biomedicine, Department of Applied Stem Cell Biology and Cell Techniques, University of Leipzig, Leipzig, Germany
| | | |
Collapse
|
185
|
Taylor SE, Smith RKW, Clegg PD. Mesenchymal stem cell therapy in equine musculoskeletal disease: scientific fact or clinical fiction? Equine Vet J 2010; 39:172-80. [PMID: 17378447 DOI: 10.2746/042516407x180868] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The goal in the therapeutic use of mesenchymal stem cells (MSCs) in musculoskeletal disease is to harness the regenerative nature of these cells focussing on their potential to grow new tissues and organs to replace damaged or diseased tissue. Laboratory isolation of MSCs is now well established and has recently been demonstrated for equine MSCs. Stem cell science has attracted considerable interest in both the scientific and clinical communities because of its potential to regenerate tissues. Research into the use of MSCs in tissue regeneration in general reflects human medical needs, however, the nature, prevalence and prognosis of superficial digital flexor tendonitis has put equine veterinary science at the forefront of tendon regeneration research. Much has been investigated and learnt but it must be appreciated that in spite of this, the field is still relatively young and both communities must prepare themselves for considerable time and effort to develop the technology into a highly efficient treatments. The promise of functional tissue engineering to replace old parts with new fully justifies the interest. At present, however, it is important to balance the understanding of our current limitations with a desire to progress the technology.
Collapse
Affiliation(s)
- S E Taylor
- Department of Veterinary Clinical Science, University of Liverpool, Leahurst, Chester High Road, Neston, Cheshire CH64 7TE, UK
| | | | | |
Collapse
|
186
|
Cartilage engineering from mesenchymal stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2010; 123:163-200. [PMID: 20535603 DOI: 10.1007/10_2010_67] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mesenchymal progenitor cells known as multipotent mesenchymal stromal cells or mesenchymal stem cells (MSC) have been isolated from various tissues. Since they are able to differentiate along the mesenchymal lineages of cartilage and bone, they are regarded as promising sources for the treatment of skeletal defects. Tissue regeneration in the adult organism and in vitro engineering of tissues is hypothesized to follow the principles of embryogenesis. The embryonic development of the skeleton has been studied extensively with respect to the regulatory mechanisms governing morphogenesis, differentiation, and tissue formation. Various concepts have been designed for engineering tissues in vitro based on these developmental principles, most of them involving regulatory molecules such as growth factors or cytokines known to be the key regulators in developmental processes. Growth factors most commonly used for in vitro cultivation of cartilage tissue belong to the fibroblast growth factor (FGF) family, the transforming growth factor-beta (TGF-β) super-family, and the insulin-like growth factor (IGF) family. In this chapter, in vivo actions of members of these growth factors described in the literature are compared with in vitro concepts of cartilage engineering making use of these growth factors.
Collapse
|
187
|
Koelling S, Miosge N. Stem cell therapy for cartilage regeneration in osteoarthritis. Expert Opin Biol Ther 2009; 9:1399-405. [PMID: 19793003 DOI: 10.1517/14712590903246370] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Enhancing the regeneration potential of hyaline cartilage tissue remains a great challenge. During embryonic development, some of the cells of the inner cell mass will turn into the mesoderm. This will be the founder of the mesenchymal cells in connective tissues of adult life, such as bone, tendon, muscle, and cartilage. Some of these embryonic mesenchymal cells are believed not to differentiate, but reside in each of the tissues. These are now collectively described as adult mesenchymal stem cells, which are thought to be capable of repairing injured tissue. We will briefly summarize the current knowledge about stem cell-related cells in cartilage tissue and carefully discuss the potential of the cell population we described recently as a starting-point for a regenerative therapy for osteoarthritis. We found that repair tissue from human articular cartilage during the late stages of osteoarthritis harbors a unique progenitor cell population, termed chondrogenic progenitor cells (CPC). These exhibit stem cell characteristics combined with a high chondrogenic potential. They offer new insights into the biology of progenitor cells and may be relevant in the development of novel therapeutic approaches for a cell-based therapy for late stages of osteoarthritis.
Collapse
Affiliation(s)
- Sebastian Koelling
- Georg August University, Tissue Regeneration Work Group, Department of Prosthodontics, Abteilung Prothetik im Zentrum ZMK, Robert-Koch-Str. 40, Goettingen, D-37075, Germany
| | | |
Collapse
|
188
|
Giovannini S, Diaz-Romero J, Aigner T, Mainil-Varlet P, Nesic D. Population doublings and percentage of S100-positive cells as predictors of in vitro chondrogenicity of expanded human articular chondrocytes. J Cell Physiol 2009; 222:411-20. [PMID: 19890919 DOI: 10.1002/jcp.21965] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aim of this study was to investigate the interconnection between the processes of proliferation, dedifferentiation, and intrinsic redifferentiation (chondrogenic) capacities of human articular chondrocyte (HAC), and to identify markers linking HAC dedifferentiation status with their chondrogenic potential. Cumulative population doublings (PD) of HAC expanded in monolayer culture were determined, and a threshold range of 3.57-4.19 PD was identified as indicative of HAC loss of intrinsic chondrogenic capacity in pellets incubated without added chondrogenic factors. While several specific gene and surface markers defined early HAC dedifferentiation process, no clear correlation with the loss of intrinsic chondrogenic potential could be established. CD90 expression during HAC monolayer culture revealed two subpopulations, with sorted CD90-negative cells showing lower proliferative capacity and higher chondrogenic potential compared to CD90-positive cells. Although these data further validated PD as critical for in vitro chondrogenesis, due to the early shift in expression, CD90 could not be considered for predicting chondrogenic potential of HAC expanded for several weeks. In contrast, an excellent mathematically modeled correlation was established between PD and the decline of HAC expressing the intracellular marker S100, providing a direct link between the number of cell divisions and dedifferentiation/loss of intrinsic chondrogenic capacity. Based on the dynamics of S100-positive HAC during expansion, we propose asymmetric cell division as a potential mechanism of HAC dedifferentiation, and S100 as a marker to assess chondrogenicity of HAC during expansion, of potential value for cell-based cartilage repair treatments.
Collapse
|
189
|
Abstract
Mesenchymal stem cells (MSCs) resident in bone marrow are one of the most studied and clinically important populations of adult stem cells. Cells with, similar properties to these MSCs have been described in several different tooth tissues and the potential ease with which these dental MSCs could be obtained from patients has prompted great interest in these cells as a source of MSCs for cell-based therapeutics. In this review we address the current state of knowledge regarding these cells, their properties, origins, locations, functions and potential uses in tooth tissue engineering and repair. We discuss some of the key controversies and outstanding issues, not least of which whether dental stem cells actually exist.
Collapse
Affiliation(s)
- Andrea Mantesso
- Guy's Hospital, Dental Institute, Kings College London, Department of Craniofacial Development, London SE1 9RT, UK
| | | |
Collapse
|
190
|
Ho STB, Yang Z, Hui HPJ, Oh KWS, Choo BHA, Lee EH. A serum free approach towards the conservation of chondrogenic phenotype during in vitro cell expansion. Growth Factors 2009; 27:321-33. [PMID: 19626506 DOI: 10.1080/08977190903137595] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Functionally viable chondrocytes in sufficient quantity is crucial for the success of matrix associated autologous chondrocyte implantation. This is difficult with conventional methods as chondrocytes dedifferentiate during 2D expansion with the loss of their chondrogenic phenotype. Moreover, established protocols are dependent on the use of serum which is not without its drawbacks. This study sought to address the issue by evaluating the feasibility of serum free, growth factors supplemented chondrocyte media with extracellular matrix (ECM) coatings. DESIGN Passage 2 human chondrocytes were cultured in serum supplemented media or serum free media with collagen I or fibronectin coatings. Cell attachment and proliferation were assessed in these conditions. The cells were redifferentiated via pellet cultures for 7 and 14 days before being subjected to histological and gene expression analysis. RESULTS The serum-free, growth factor cocktail supplemented with ECM coating improved long-term chondrocyte proliferation with enhanced basal Sox 9 expression. Upon induction, the redifferentiated chondrocytes expressed aggrecan and collagen II especially so for the cells plated on collagen coated surfaces. The chondrocytic phenotype was better conserved under the serum free conditions but the loss of the hyaline cartilage characteristics was not completely halted given the expression of collagen I. These essential cartilage markers were, however, reduced or absented for cells expanded with serum. Moreover, serum cultures displayed a higher tendency of undergoing hypertrophy given the stronger collagen X gene expression. CONCLUSION The advocated technique promoted cell expansion with respect to conventional serum supplemented cultures while reducing the loss of the chondrogenic phenotype. This demonstrates the feasibility and potential of the novel concomitant use of serum free media and ECM coatings in the expansion of chondrocytes for cartilage regenerative applications.
Collapse
Affiliation(s)
- Saey Tuan Barnabas Ho
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medical, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
191
|
Bocelli-Tyndall C, Bracci L, Schaeren S, Feder-Mengus C, Barbero A, Tyndall A, Spagnoli GC. Human bone marrow mesenchymal stem cells and chondrocytes promote and/or suppress the in vitro proliferation of lymphocytes stimulated by interleukins 2, 7 and 15. Ann Rheum Dis 2009; 68:1352-9. [PMID: 18647856 DOI: 10.1136/ard.2008.094003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES To investigate whether human bone marrow-derived mesenchymal stem cells (BM-MSCs) and articular chondrocytes (ACs) affect the in vitro proliferation of T lymphocytes and peripheral blood mononuclear cells (PBMCs) driven by the homeostatic interleukin (IL)2, IL7 and IL15 cytokines binding to the common cytokine receptor gamma-chain (gamma(c)) in the absence of T cell receptor (TCR) triggering. METHODS PBMCs, total T cells and T cell subsets (CD4+ and CD8+) were stimulated with IL2, IL7 or IL15 and exposed to cultured BM-MSCs and ACs at varying cell:cell ratio either in contact or in transwell conditions. Lymphocyte proliferation was measured by (3)H-thymidine uptake or by flow cytometry of carboxyfluorescein succinimidyl ester (CFSE)-labelled lymphocytes. RESULTS MSCs and ACs enhanced and inhibited lymphocyte proliferation depending on the extent of lymphocyte baseline proliferation and on the MSC/AC to lymphocyte ratio. Enhancement was significant on poorly proliferating lymphocytes and mostly at lower MSC/AC to lymphocyte ratio. Suppression occurred only on actively proliferating lymphocytes and at high MSC/AC to lymphocyte ratio. Neither enhancement nor inhibition required cell-cell contact. CONCLUSIONS There is a dichotomous effect of MSCs/ACs on lymphocytes proliferating in response to the homeostatic IL2, IL7 and IL15 cytokines likely to be encountered in homeostatic and autoimmune inflammatory conditions. The effect is determined by baseline lymphocyte proliferation, cell:cell ratio and is dependent on soluble factor(s). This should be taken into account when planning cellular therapy for autoimmune disease (AD) using stromal-derived cells such as MSCs.
Collapse
Affiliation(s)
- C Bocelli-Tyndall
- Institute of Surgical Research and Hospital Management and Department of Biomedicine, University Hospital Basel, Basel, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
192
|
Abstract
The view of articular cartilage as a non-regeneration organ has been challenged in recent years. The articular cartilage consists of distinct zones with different cellular and molecular phenotypes, and the superficial zone has been hypothesized to harbour stem cells. Furthermore, the articular cartilage demonstrates a distinct pattern regarding stem cell markers (that is, Notch-1, Stro-1, and vascular cell adhesion molecule-1). These results, in combination with the positive identification of side population cells in articular cartilage, give additional support for the hypothesis that articular cartilage has residing stem cells with a potential regenerative capacity where the controlling mechanism could be future biomarkers or drug targets or both.
Collapse
|
193
|
Koelling S, Kruegel J, Irmer M, Path JR, Sadowski B, Miro X, Miosge N. Migratory chondrogenic progenitor cells from repair tissue during the later stages of human osteoarthritis. Cell Stem Cell 2009; 4:324-35. [PMID: 19341622 DOI: 10.1016/j.stem.2009.01.015] [Citation(s) in RCA: 301] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 01/02/2009] [Accepted: 01/22/2009] [Indexed: 12/29/2022]
Abstract
The regeneration of diseased hyaline cartilage continues to be a great challenge, mainly because degeneration--caused either by major injury or by age-related processes--can overextend the tissue's self-renewal capacity. We show that repair tissue from human articular cartilage during the late stages of osteoarthritis harbors a unique progenitor cell population, termed chondrogenic progenitor cells (CPCs). These exhibit stem cell characteristics such as clonogenicity, multipotency, and migratory activity. The isolated CPCs, which exhibit a high chondrogenic potential, were shown to populate diseased tissue ex vivo. Moreover, downregulation of the osteogenic transcription factor runx-2 enhanced the expression of the chondrogenic transcription factor sox-9. This, in turn, increased the matrix synthesis potential of the CPCs without altering their migratory capacity. Our results offer new insights into the biology of progenitor cells in the context of diseased cartilage tissue. Our work may be relevant in the development of novel therapeutics for the later stages of osteoarthritis.
Collapse
Affiliation(s)
- Sebastian Koelling
- Tissue Regeneration Group, Medical Faculty, Department of Prosthodontics, Georg August University, 37075 Goettingen, Germany
| | | | | | | | | | | | | |
Collapse
|
194
|
Karlsson C, Thornemo M, Henriksson HB, Lindahl A. Identification of a stem cell niche in the zone of Ranvier within the knee joint. J Anat 2009; 215:355-63. [PMID: 19563472 DOI: 10.1111/j.1469-7580.2009.01115.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A superficial lesion of the articular cartilage does not spontaneously self-repair and has been suggested to be partly due to lack of progenitor cells within the joint that can reach the site of injury. To study whether progenitor cells are present within the joint, 3-month-old New Zealand white rabbits were exposed to bromodeoxyuridine (BrdU) for 12 consecutive days and were then sacrificed 4, 6, 10, 14, 28 and 56 days after the first BrdU administration. Presence of BrdU and localization of progenitor markers were detected using immunohistochemistry. After 10 days of BrdU exposure, BrdU-positive cells, i.e. proliferating cells, were abundantly detected in the epiphyseal plate, the perichondrial groove of Ranvier, and in all zones of the articular cartilage. After a wash-out period, BrdU-positive cells were still present, i.e. those considered to be progenitor cells, in these regions of the knee except for the proliferative zone of the epiphyseal plate. Cells in the perichondrial groove of Ranvier were further positive for several markers associated with progenitor cells and stem cell niches, including Stro-1, Jagged1, and BMPr1a. Our results demonstrate that a small population of progenitor cells is present in the perichondrial groove of Ranvier as well as within the articular cartilage in the knee. The perichondrial groove of Ranvier also demonstrates the properties of a stem cell niche.
Collapse
Affiliation(s)
- Camilla Karlsson
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska University Hospital, Gothenburg University, S-413 45 Gothenburg, Sweden.
| | | | | | | |
Collapse
|
195
|
Schulze-Tanzil G. Activation and dedifferentiation of chondrocytes: implications in cartilage injury and repair. Ann Anat 2009; 191:325-38. [PMID: 19541465 DOI: 10.1016/j.aanat.2009.05.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2009] [Revised: 05/19/2009] [Accepted: 05/20/2009] [Indexed: 01/12/2023]
Abstract
Cartilage injury remains a major challenge in orthopedic surgery due to the fact that articular cartilage has only a limited capacity for intrinsic healing. Cartilage impaction is followed by a post-traumatic inflammatory response. Chondrocytes and synoviocytes are activated to produce inflammatory mediators and degradative enzymes which can induce a progradient cartilage self-destruction finally leading to secondary osteoarthritis (OA). However, an anti-inflammatory compensatory response is also detectable in cartilage by up-regulation of anti-inflammatory cytokines, probably a temporary attempt by chondrocytes to restore cartilage homeostasis. Matrix-assisted autologous chondrocyte transplantation (MACT) is a suitable technique for improving the rate of repair of larger articular cartilage defects. For MACT, autologous chondrocytes were isolated from a cartilage biopsy of a non-load bearing joint area. This technique requires sufficient expansion of differentiated autologous chondrocytes, which were then seeded on suitable biodegradable three-dimensional (3D) matrices to preform an extracellular cartilage matrix (ECM) before implantation into the defect. Cell expansion is accompanied by chondrocyte dedifferentiation, whereby substantial changes occur at multiple levels of chondrocyte synthetic profiles: including the ECM, cell surface receptors and cytoskeletal proteins. Since these dedifferentiated chondrocytes produce a non-specific mechanically inferior ECM, they are not suitable for MACT. 3D cultures are means of inducing and maintaining chondrocyte (re)differentiation and to preform ECM. The combination of MACT with anabolic growth factors and anti-inflammatory strategies using anti-inflammatory mediators might be useful for stabilizing the differentiated chondrocyte phenotype, to support neocartilage formation and inhibit post-traumatic cartilage inflammation and hence, the development of secondary OA.
Collapse
Affiliation(s)
- Gundula Schulze-Tanzil
- Department of Trauma and Reconstructive Surgery, Charité-University of Medicine, Campus Benjamin Franklin, FEM, Krahmerstrasse 6-10, Berlin, Germany
| |
Collapse
|
196
|
Grogan SP, Miyaki S, Asahara H, D'Lima DD, Lotz MK. Mesenchymal progenitor cell markers in human articular cartilage: normal distribution and changes in osteoarthritis. Arthritis Res Ther 2009; 11:R85. [PMID: 19500336 PMCID: PMC2714136 DOI: 10.1186/ar2719] [Citation(s) in RCA: 187] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 05/07/2009] [Accepted: 06/05/2009] [Indexed: 12/20/2022] Open
Abstract
Introduction Recent findings suggest that articular cartilage contains mesenchymal progenitor cells. The aim of this study was to examine the distribution of stem cell markers (Notch-1, Stro-1 and VCAM-1) and of molecules that modulate progenitor differentiation (Notch-1 and Sox9) in normal adult human articular cartilage and in osteoarthritis (OA) cartilage. Methods Expression of the markers was analyzed by immunohistochemistry (IHC) and flow cytometry. Hoechst 33342 dye was used to identify and sort the cartilage side population (SP). Multilineage differentiation assays including chondrogenesis, osteogenesis and adipogenesis were performed on SP and non-SP (NSP) cells. Results A surprisingly high number (>45%) of cells were positive for Notch-1, Stro-1 and VCAM-1 throughout normal cartilage. Expression of these markers was higher in the superficial zone (SZ) of normal cartilage as compared to the middle zone (MZ) and deep zone (DZ). Non-fibrillated OA cartilage SZ showed reduced Notch-1 and Sox9 staining frequency, while Notch-1, Stro-1 and VCAM-1 positive cells were increased in the MZ. Most cells in OA clusters were positive for each molecule tested. The frequency of SP cells in cartilage was 0.14 ± 0.05% and no difference was found between normal and OA. SP cells displayed chondrogenic and osteogenic but not adipogenic differentiation potential. Conclusions These results show a surprisingly high number of cells that express putative progenitor cell markers in human cartilage. In contrast, the percentage of SP cells is much lower and within the range of expected stem cell frequency. Thus, markers such as Notch-1, Stro-1 or VCAM-1 may not be useful to identify progenitors in cartilage. Instead, their increased expression in OA cartilage implicates involvement in the abnormal cell activation and differentiation process characteristic of OA.
Collapse
Affiliation(s)
- Shawn P Grogan
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | | | | | |
Collapse
|
197
|
Klein TJ, Malda J, Sah RL, Hutmacher DW. Tissue engineering of articular cartilage with biomimetic zones. TISSUE ENGINEERING. PART B, REVIEWS 2009; 15:143-57. [PMID: 19203206 PMCID: PMC3121783 DOI: 10.1089/ten.teb.2008.0563] [Citation(s) in RCA: 208] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2008] [Accepted: 02/07/2009] [Indexed: 12/20/2022]
Abstract
Articular cartilage damage is a persistent and increasing problem with the aging population, and treatments to achieve biological repair or restoration remain a challenge. Cartilage tissue engineering approaches have been investigated for over 20 years, but have yet to achieve the consistency and effectiveness for widespread clinical use. One of the potential reasons for this is that the engineered tissues do not have or establish the normal zonal organization of cells and extracellular matrix that appears critical for normal tissue function. A number of approaches are being taken currently to engineer tissue that more closely mimics the organization of native articular cartilage. This review focuses on the zonal organization of native articular cartilage, strategies being used to develop such organization, the reorganization that occurs after culture or implantation, and future prospects for the tissue engineering of articular cartilage with biomimetic zones.
Collapse
Affiliation(s)
- Travis J. Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Jos Malda
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert L. Sah
- Department of Bioengineering, University of California–San Diego, La Jolla, California
| | - Dietmar W. Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
198
|
van Osch GJVM, Brittberg M, Dennis JE, Bastiaansen-Jenniskens YM, Erben RG, Konttinen YT, Luyten FP. Cartilage repair: past and future--lessons for regenerative medicine. J Cell Mol Med 2009; 13:792-810. [PMID: 19453519 PMCID: PMC3823400 DOI: 10.1111/j.1582-4934.2009.00789.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Since the first cell therapeutic study to repair articular cartilage defects in the knee in 1994, several clinical studies have been reported. An overview of the results of clinical studies did not conclusively show improvement over conventional methods, mainly because few studies reach level I of evidence for effects on middle or long term. However, these explorative trials have provided valuable information about study design, mechanisms of repair and clinical outcome and have revealed that much is still unknown and further improvements are required. Furthermore, cellular and molecular studies using new technologies such as cell tracking, gene arrays and proteomics have provided more insight in the cell biology and mechanisms of joint surface regeneration. Besides articular cartilage, cartilage of other anatomical locations as well as progenitor cells are now considered as alternative cell sources. Growth Factor research has revealed some information on optimal conditions to support cartilage repair. Thus, there is hope for improvement. In order to obtain more robust and reproducible results, more detailed information is needed on many aspects including the fate of the cells, choice of cell type and culture parameters. As for the clinical aspects, it becomes clear that careful selection of patient groups is an important input parameter that should be optimized for each application. In addition, the study outcome parameters should be improved. Although reduced pain and improved function are, from the patient's perspective, the most important outcomes, there is a need for more structure/tissue-related outcome measures. Ideally, criteria and/or markers to identify patients at risk and responders to treatment are the ultimate goal for these more sophisticated regenerative approaches in joint surface repair in particular, and regenerative medicine in general.
Collapse
Affiliation(s)
- Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| | | | | | | | | | | | | |
Collapse
|
199
|
Bian L, Angione S, Ng K, Lima E, Williams D, Mao D, Ateshian G, Hung C. Influence of decreasing nutrient path length on the development of engineered cartilage. Osteoarthritis Cartilage 2009; 17:677-85. [PMID: 19022685 PMCID: PMC3387279 DOI: 10.1016/j.joca.2008.10.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2008] [Accepted: 10/02/2008] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Chondrocyte-seeded agarose constructs of 4mm diameter (2.34 mm thickness) develop spatially inhomogeneous material properties with stiffer outer edges and a softer central core suggesting nutrient diffusion limitations to the central construct region [Guilak F, Sah RL, Setton LA. Physical regulation of cartilage metabolism. In: Mow VC, Hayes WC, Eds. Basic Orthopaedic Biomechanics, Philadelphia 1997;179-207.]. The effects of reducing construct thickness and creating channels running through the depth of the thick constructs were examined. METHODS In Study 1, the properties of engineered cartilage of 0.78 mm (thin) or 2.34 mm (thick) thickness were compared. In Study 2, a single nutrient channel (1 mm diameter) was created in the middle of each thick construct. In Study 3, the effects of channels on larger 10 mm diameter, thick constructs were examined. RESULTS Thin constructs developed superior mechanical and biochemical properties than thick constructs. The channeled constructs developed significantly higher mechanical properties vs control channel-free constructs while exhibiting similar glycosaminoglycan (GAG) and collagen content. Collagen staining suggested that channels resulted in a more uniform fibrillar network. Improvements in constructs of 10 mm diameter were similarly observed. CONCLUSIONS This study demonstrated that more homogeneous tissue-engineered cartilage constructs with improved mechanical properties can be achieved by reducing their thickness or incorporating macroscopic nutrient channels. Our data further suggests that these macroscopic channels remain open long enough to promote this enhanced tissue development while exhibiting the potential to refill with cell elaborated matrix with additional culture time. Together with reports that <3 mm defects in cartilage heal in vivo and that irregular holes are associated with clinically used osteochondral graft procedures, we anticipate that a strategy of incorporating macroscopic channels may aid the development of clinically relevant engineered cartilage with functional properties.
Collapse
Affiliation(s)
- L. Bian
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - S.L. Angione
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - K.W. Ng
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - E.G. Lima
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - D.Y. Williams
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - D.Q. Mao
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - G.A. Ateshian
- Musculoskeletal Biomechanics Laboratory, Department of Mechanical Engineering, Columbia, University, New York, NY 10027, USA
| | - C.T. Hung
- Cellular Engineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
200
|
[Regenerative potential of human adult precursor cells: cell therapy--an option for treating cartilage defects?]. Z Rheumatol 2009; 68:234-8. [PMID: 19384550 DOI: 10.1007/s00393-008-0396-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cell-based therapeutical approaches are already in clinical use and are attracting growing interest for the treatment of joint defects. Mesenchymal stem and precursor cells (MSC) cover a wide range of properties that are useful for the regeneration process of bone and cartilage defects. The following article is an overview of the regenerative potential of MSC and discusses how the properties of these cells can be used for the development of new strategies in regenerative medicine.
Collapse
|