Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Synoviocyte Derived-Extracellular Matrix Enhances Human Articular Chondrocyte Proliferation and Maintains Re-Differentiation Capacity at Both Low and Atmospheric Oxygen Tensions

  • Thomas J. Kean ,

    thomas.kean@bcm.edu

    Current address: Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, United States of America

    Affiliations Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, United States of America, Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States of America, Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States of America

  • James E. Dennis

    Current address: Orthopedic Surgery, Baylor College of Medicine, Houston, Texas, United States of America

    Affiliations Department of Orthopaedics, Case Western Reserve University, Cleveland, OH, United States of America, Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States of America, Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States of America

Correction

14 Sep 2015: Kean TJ, Dennis JE (2015) Correction: Synoviocyte Derived-Extracellular Matrix Enhances Human Articular Chondrocyte Proliferation and Maintains Re-Differentiation Capacity at Both Low and Atmospheric Oxygen Tensions. PLOS ONE 10(9): e0138409. https://doi.org/10.1371/journal.pone.0138409 View correction

Abstract

Background

Current tissue engineering methods are insufficient for total joint resurfacing, and chondrocytes undergo de-differentiation when expanded on tissue culture plastic. De-differentiated chondrocytes show poor re-differentiation in culture, giving reduced glycosaminoglycan (GAG) and collagen matrix accumulation. To address this, porcine synoviocyte-derived extracellular matrix and low (5%) oxygen tension were assessed for their ability to enhance human articular chondrocyte expansion and maintain re-differentiation potential.

Methods

Porcine synoviocyte matrices were devitalized using 3 non-detergent methods. These devitalized synoviocyte matrices were compared against tissue culture plastic for their ability to support human chondrocyte expansion. Expansion was further compared at both low (5%), and atmospheric (20%) oxygen tension on all surfaces. Expanded cells then underwent chondrogenic re-differentiation in aggregate culture at both low and atmospheric oxygen tension. Aggregates were assessed for their GAG and collagen content both biochemically and histologically.

Results

Human chondrocytes expanded twice as fast on devitalized synoviocyte matrix vs. tissue culture plastic, and cells retained their re-differentiation capacity for twice the number of population doublings. There was no significant difference in growth rate between low and atmospheric oxygen tension. There was significantly less collagen type I, collagen type II, aggrecan and more MMP13 expression in cells expanded on synoviocyte matrix vs. tissue culture plastic. There were also significant effects due to oxygen tension on gene expression, wherein there was greater collagen type I, collagen type II, SOX9 and less MMP13 expression on tissue culture plastic compared to synoviocyte matrix. There was a significant increase in GAG, but not collagen, accumulation in chondrocyte aggregates re-differentiated at low oxygen tension over that achieved in atmospheric oxygen conditions.

Conclusions

Synoviocyte-derived matrix supports enhanced expansion of human chondrocytes such that the chondrocytes are maintained in a state from which they can re-differentiate into a cartilage phenotype after significantly more population doublings. Also, low oxygen tension supports GAG, but not collagen, accumulation. These findings are a step towards the production of a more functional, tissue engineered cartilage.

Introduction

Human articular cartilage has long been known to have a poor repair capacity [1]. Cartilage consists of a largely avascular, hypocellular, collagen and glycosaminoglycan (GAG) matrix [2]. Current orthopedic clinical practice attempts to relieve pain, aid repair or delay damage using non-steroidal anti-inflammatories, steroid injections, or hyaluronic acid injections. For large lesions, or those that do not respond to these treatments, surgeons have several options, including: microfracture, mosaicplasty [3], autologous chondrocyte implantation (ACI) [4], matrix assisted autologous chondrocyte implantation (MACI) [5], or, finally, total joint replacement. There are advantages and disadvantages to each technique. Microfracture and mosaicplasty have the advantage of being performed in a single surgery, but are only applicable to small lesions, and microfracture often results in a fibrous repair [5]. Both ACI and MACI are autologous cell transplantation techniques, which have the disadvantage of requiring two surgical procedures; cartilage biopsies are taken from low load-bearing regions, grown in culture and then implanted. However, ACI is a generally accepted technique, with good long-term outcomes when assessed by patient scoring systems [6, 7]. In an objective assessment of chondrogenesis, biopsies of 406 ACI patients showed hyaline cartilage in only 14.9% of patients, mixed hyaline and fibrous cartilage in 27.5%, fibrocartilage in 47.7% and fibrous repair in the remaining 9.9% [8]; similar results have been reported elsewhere [9]. This lack of hyaline cartilage formation could be due to the limited ability of human chondrocytes to expand on tissue culture (TC) plastic and their progressive loss of chondrogenic phenotype [1012] and decreased capacity to re-differentiate [13]. Without the formation of hyaline cartilage that is able to provide mechanical support during joint function, the long-term prognosis for these repaired joints is poor. In addition, joints with even larger lesions are unlikely to see any clinical benefit from these procedures. As such, it is currently unfeasible for a human total joint to be resurfaced with autologous chondrocytes without improved expansion conditions that preserve chondrogenic potential.

Synoviocyte matrix has previously been shown to provide an environment that enhanced the growth and reduced the dedifferentiation of porcine articular chondrocytes [14], but this has not been shown for human chondrocytes. Previous work on synoviocyte matrices has used detergent as a decellularization method [14, 15]. However, detergent decellularization removes some extracellular matrix and disrupts collagen [16], and it is also difficult to completely remove detergents, which can have detrimental effects on cell viability [17]. This report describes the assessment of three non-detergent methods of devitalization [16], including a novel ethanol-based devitalization, of synoviocyte matrix for the expansion of human chondrocytes.

Low O2 (5%) tension during expansion culture has been reported to trend towards enhanced growth rate in rabbit chondrocytes [18], but to have no significant effect on human articular chondrocyte growth [19]. When cells are re-differentiated in low O2, chondrogenesis has been elevated in rabbit [18], bovine [20], and human [19, 21] articular chondrocyte cultures. However, it has also been shown to have negative effects on chondrogenesis in bovine articular chondrogenesis [22] and highly donor dependent results with human bone marrow MSCs [23]. Due to the conflicting results found in the study of chondrocyte expansion under low or atmospheric oxygen tensions (Table 1) [24], we considered oxygen tension to be an important parameter to investigate. Therefore, low O2 conditions were tested in conjunction with chondrocyte expansion on synoviocyte matrices and their subsequent re-differentiation in aggregate culture.

thumbnail
Table 1. Effect of reduced oxygen tension on in vitro human chondrogenesis.

https://doi.org/10.1371/journal.pone.0129961.t001

The present study looked at two factors vital to improved cartilage tissue engineering: 1) increased cell expansion; and 2) maintenance of chondrogenic potential. It was hypothesized that devitalized synoviocyte matrix would both increase the expansion of human chondrocytes and preserve their chondrogenic potential. Initial studies determined that detergent-based devitalization methods, even at low concentrations or after extended washes, caused the majority of the synoviocyte cell matrix to detach, and did not improve human chondrocyte expansion. To address these problems, three alternative synoviocyte extraction methods were assessed: freeze/thaw; freeze/thaw plus sonication; dry ice-cooled ethanol. All three showed high retention of the devitalized synoviocyte matrix and supported human chondrocyte growth. These three methods were compared to identify the one which produced the greatest population expansion while maintaining differentiation potential. It was also hypothesized that low O2 tension would support chondrocyte growth and re-differentiation. It was further hypothesized that the combination of the synoviocyte matrix and low O2 tension would have an additive effect on cell growth and re-differentiation. Re-differentiation was assessed by GAG and collagen content biochemically, and by GAG and type II collagen staining histologically. Re-differentiation into hyaline- like articular cartilage is defined by GAG and collagen accumulation, specifically type II collagen.

In summary, it was hypothesized that synoviocyte matrix expansion would support human chondrocyte chondrogenesis over more population doublings (PDs); that low O2 tension would support chondrogenesis; and that the combination of the synoviocyte matrix expansion and low O2 tension would have a cumulative effect on chondrogenesis.

Materials and Methods

Ethics Statement

Human chondrocytes were obtained from discarded tissue from total joint replacement surgeries with written informed consent under an IRB approved protocol: IRB # IRB08-00104 of The Metro Health System, Cleveland, OH. Synovial tissue was obtained from humanely euthanized sows after their use in surgical training (unpublished, not connected with this research). The surgical training lab was conducted under an IACUC approved protocol. Pigs were used in a terminal procedure and euthanized by lethal injection of Euthasol (pentobarbital sodium and phenytoin sodium) per protocol at the end of the lab, but prior to tissue harvesting (Case Western Reserve University and University of Washington).

Synoviocyte isolation, culture and devitalization methods

Synoviocytes were isolated and cultured much as previously described [15]. Briefly, synovial membrane tissue was isolated from the knees of ~6-month-old sows within 2 h postmortem using sterile surgical technique (n = 5). The isolated membrane was then minced and digested with bovine testicular hyaluronidase (Sigma; 660 Units/ml in DMEM-LG) for 15 min at 37°C, centrifuged (500 RCF, 5 min) and the supernatant discarded. The tissue was then digested with trypsin/EDTA (Gibco; 0.25% (w/v)) for 30 min at 37°C, centrifuged (500 RCF, 5 min) and supernatant discarded before a final digestion in type II collagenase (Worthington Biochemical Corp.; 583 Units/ml) for 3 h at 37°C. The resulting solution was passed through a 70 μm filter and centrifuged (690 RCF; 10 min). Cells were counted and flasks seeded at 3,000 cells/cm2 and cultured in DMEM-LG, 10% (v/v) FBS, Pen/Strep (100 Units/100 μg/ml) until confluent. At the end of primary culture (P0) and second passage (P2) stocks were made by controlled rate freezing in FBS containing 10% (v/v) DMSO and stored under liquid nitrogen. For the production of synoviocyte matrix-coated flasks, P2 cells were thawed from frozen stocks and seeded on TC flasks (Corning) at 6,000 cells/cm2. They were grown in DMEM-LG + FBS (10% v/v) until 80–90% confluence (~7 days), and then switched to DMEM-LG + FBS (10% v/v) containing 50 μM ascorbate-2-phosphate for an additional 7 days. Fully confluent flasks were rinsed with PBS, the PBS removed and quick frozen on dry ice-cooled ethanol (190 proof). Flasks were either stored at -80°C for later devitalization or immediately devitalized by one of three methods: 1) 3 cycles of thawing/freezing (F-SCM); 2) 3 cycles of thawing/freezing followed by 1 min of sonication (S-SCM); 3) addition of dry ice cooled ethanol (200 proof) onto the synoviocyte matrix surface (30 s) on dry ice-cooled ethanol (E-SCM). The ethanol extract was discarded and the flask dried in a laminar flow sterile environment for approximately 30 min. The flasks were rinsed twice with PBS and then used for the culture of human chondrocytes.

Human chondrocyte expansion

Human chondrocytes were thawed from the end of P0 stocks, rinsed in expansion medium (DMEM-LG with 10% v/v FBS [human MSC selected lot [24]] containing penicillin/streptomycin [Invitrogen]), centrifuged (500 RCF, 5 min), resuspended in expansion medium, and a trypan blue-stained aliquot was counted in a hemacytometer. Cells were seeded into T175 TC plastic flasks (Corning) at 1 x 106 cells/flask at both low (5%) and atmospheric (20%) O2. At confluence (end of P1), flasks were trypsinized (0.25% w/v trypsin/EDTA), the trypsin neutralized with FBS, centrifuged (500 RCF, 5 min), trypan blue stained, counted and split between synoviocyte matrix-coated or uncoated flasks under the same O2 tension as the prior expansion at 6,000 cells/cm2 (Fig 1). At confluence (end of P2, P3, P4), flasks were trypsinized, viable cells counted using trypan blue and passaged to a new flask under the same conditions and aggregate cultures made (Fig 1). PDs were calculated from the point the cells were split between each condition using the formula PD = (log10 (harvested cell count)—log10 (seeded cell count)) x 3.32 and sequential passages summed to give cumulative population doublings [25].

thumbnail
Fig 1. Experimental culture conditions overview.

Human articular chondrocytes were thawed from frozen stocks and initially expanded on TC plastic before being split between each of the surfaces (TC plastic, F-SCM, S-SCM and E-SCM) and expanded for 3 passages with aggregates made at each passage. All steps were conducted at either (20%) atmospheric or low (5%) O2 tension.

https://doi.org/10.1371/journal.pone.0129961.g001

qPCR chondrocyte gene expression analysis

qPCR experiments were conducted in accordance with the minimum information for publication of quantitative real-time pcr experiment (MIQE) guidelines[26]. Human chondrocytes (7 donors) were thawed from end of P0 stocks, and expanded as described above except on T75 flasks and only comparing TC plastic with E-SCM (Fig 1). Flasks were harvested at day 6 (P1), day 11 (P2), day 16 (P3) and day 21 (P4) during the logarithmic growth phase with 1 flask being extracted for RNA analysis with TRK Lysis buffer (E.Z.N.A. Tissue RNA Kit, Omega Bio-Tek) and the lysate frozen on dry ice and stored (-80°C, 1–6 weeks). The other flask was trypsinized, counted and seeded into a new flask. Total RNA was isolated from the lysates after thawing on ice using column purification (Direct-zol RNA mini-prep, Zymo Research), as per the manufacturer’s instructions with on-column DNA digest (DNase I, Zymo Research). RNA purity was analyzed by 260 nm / 280 nm ratio (Tecan Nanoquant) and degradation assessed using an Agilent Bioanalyzer (Genomic and RNA Profiling Core at Baylor College of Medicine). cDNA was synthesized from 500 ng of RNA using a Superscript III First Strand RT-PCR kit with oligo(dT)20 priming, according to the manufacturer’s instructions (Invitrogen). Quantitative real-time PCR was performed on a LightCycler Instrument (Roche) (2 μl of cDNA at a 1:8 dilution; with 16.5 μl of a mix containing 9.6 μl water (Roche), 2 μl of forward and reverse primers (50 nM), 2 μl Master mix (LightCycler FastStart DNA Master SYBR Green I, Roche), 2.4 μl MgCl2 (25 mM, Roche) with 0.4 μl of Uracil-DNA Glycosylase [Roche]). Cycling parameters were: 95°C for 10s then 45 cycles of 95°C 10s, 60°C 7s, 72°C 20s, followed by melt curve analysis. Seven reference genes (Ribosomal 18S, Glyceraldehyde 3-phosphate dehydrogenase, Beta-2 microglobulin, Hydroxymethylbilane synthase, Beta-actin, Tata-box binding protein and Hypoxanthine Phosphoribosyltransferase 1 (HPRT) were assessed for stability on a subset of samples. HPRT was determined to be the most stable across all conditions and therefore used as the internal control (reference gene) to normalize gene expression (primer sequences used are shown in S1 Table). All quantification cycles (Cq) were within the linear portion of log dilution analyses and >2 cycles higher than the negative control, melting temperatures were also consistent (S2 Table). Data for the comparison of expansion on TC plastic or E-SCM is presented as the expression of the target gene vs HPRT relative to the P1 sample on TC plastic. In the comparison of the effect of oxygen tension, each donor’s normalized gene expression at low oxygen tension was compared to its expression at atmospheric oxygen tension.

Human chondrocyte re-differentiation—Aggregate cultures

Differentiation media contained DMEM-HG, ascorbate-2-phosphate (120 μM; Wako Pure Chemicals), dexamethasone (100 nM; Sigma), ITS + Premix (1% v/v; BD Biosciences), TGFβ1 (10 ng/ml; Peprotech), and sodium pyruvate (1 mM; Invitrogen) [27]. Trypsinized cells were resuspended in differentiation media and aggregate cultures made using 250,000 cells/aggregate (200 μl) seeded in each well of a polypropylene v-bottom 96-well plate. Plates were then centrifuged (600 RCF, 5 min), and placed in an incubator at the same O2 tension as that in which they were expanded. Media was changed on aggregates after 1 day, and every other day after that until harvest at day 21. At harvest, aggregates were weighed and either frozen (-80°C) for biochemical assays or fixed in neutral buffered formalin for histology.

Biochemical analyses

Aggregates were thawed and papain digested (200 μl of papain digest buffer: 25 μg/ml papain (Sigma) in aqueous 2 mM cysteine (Sigma); 50 mM sodium phosphate (Sigma); 2 mM EDTA (Sigma); adjusted to pH 6.5 with 1 M HCl/NaOH) for 3 h at 65°C with vortexing every 30 min. Digests were then split between two tubes for either DNA/GAG assay [28] or hydroxyproline assay [2932]. For the DNA/GAG assay, papain digestion was arrested by the addition of 200 μl of 0.1 M NaOH, incubated at RT for 30 min. This solution was then neutralized with 200 μl phosphate buffer (100 mM NaHPO4 (Sigma) pH 7.2; 0.1 M HCl). The DNA content was analyzed in triplicate against a standard curve produced with calf thymus DNA (Sigma) diluted in neutralized papain digest buffer using Hoechst dye (Hoechst 33258; Sigma; 0.667 μg/ml in 0.2 M phosphate buffer, pH 8). Fluorescence was read on a Fusion plate reader (Excitation 365 nm, Emission 460 nm; Packard Biosciences) using black 96-well plates (Greiner). GAG content was assessed from the same neutralized solution against a standard curve generated using chondroitin sulfate (Seikagaku) diluted in neutralized papain buffer. Colorimetric assays using Safranin-O were performed as previously described [28], and absorbance read at 536 nm (Optimax microplate reader, Molecular Devices). For the hydroxyproline assay, modifications to previously-described methods were made [2932]. Briefly, 1 ml of HCl (6 M) was added to the digest and the sample hydrolyzed by incubation at 100–110°C overnight. Samples were then dried by incubation at 65–70°C for 1–2 days. Dried samples were then solubilized in 100 μl of 18 megaohm water (Nanopure, Thermo Scientific) and 20 μl transferred to a 96-well clear plate in triplicate. A standard curve of hydroxyproline diluted in papain buffer was also added. To these, 20 μl of 0.15 M CuSO4 and 20 μl of 2.5 M NaOH was added and incubated at 50°C for 5 min, followed by the addition of 20 μl of 6% (v/v) H2O2 and a further incubation at 50°C for 10 min. Plates were allowed to cool to RT before addition of 80 μl of 1.5 M H2SO4, then 40 μl of Ehrlich’s reagent (10% (w/v) p-dimethyl-amino-benzaldehyde (Sigma) in 60% (v/v) isopropanol, 18.2% (v/v) perchloric acid, 21.8% (v/v) megaohm water) and incubation at 70°C for 16 min. Following this, plates were allowed to cool to RT before analysis of absorbance (505 nm, Optimax microplate reader, Molecular Devices).

Histological assessment

Aggregates, fixed in neutral buffered formalin (Azer Scientific), were paraffin-embedded and sectioned (5 μm). Sections were stained for GAG: 6 min in 0.1% (w/v) Safranin-O solution containing 1% (v/v) acetic acid and counterstained for 2 min in 1% (w/v) Fast Green containing 7% (v/v) acetic acid. For immunohistochemistry, unstained sections were de-paraffinized, outlined with a mini pap pen (Diagnostic Biosystems) and covered with pronase (1 mg/ml (Sigma) in PBS containing 5 mM CaCl2) and incubated at RT for 10 min. Sections were then rinsed with PBS, blocked with PBS/BSA 1% (w/v) (10 min RT), incubated with primary antibody overnight at 4°C (type II collagen: 1/500 in PBS/BSA 1% (w/v) [mouse monoclonal IgG1, raised against chicken collagen type II; II-II6B3 cell culture supernatant, DSHB]; type X collagen: 1/500 in PBS/BSA 1% (w/v) [mouse monoclonal, a kind gift from Dr. Gary Gibson, Henry Ford Hospital]; elastin: 1/400 in PBS/BSA 1% (w/v) [mouse monoclonal clone 10B8 raised against chick tropoelastin, MAB2505; Chemicon]). Following this they were rinsed with PBS and incubated for 1 h at RT with secondary antibody (Horse anti-mouse, 1/2000 in PBS/BSA 1% (w/v); BA-2000, Vector Labs), rinsed with PBS then incubated with streptavidin-conjugated horseradish peroxidase for 45 min at RT (1/400 in PBS/BSA 1% (w/v), SNN1004, Gibco), rinsed with PBS then incubated with peroxidase substrate (VIP substrate, Vectashield) for 10 min at RT. Slides were rinsed in distilled water and counterstained with Fast Green (0.0125% (w/v) in 24.25% (v/v) ethanol, 0.875% (v/v) acetic acid; Alfa Aesar) before rinsing in distilled water, and dehydration with ethanol then xylene, before drying and mounting (Permount, Fisher Scientific). Images were taken on a DME Upright brightfield microscope (Leica) at RT using a 10x 0.25 NA lens, using Leica Acquisition Software EZ (v1.7.0) with a Leica EC3 camera and background corrected and tiled using GNU Image Manipulation Program (GIMP 2.8.6).

Statistical analyses

Comparisons between regression curves were made using a method equivalent to an Analysis of Covariance (ANCOVA, GraphPad Prism V.6.02). Comparisons of growth rates were made using one-way repeated measures analysis of variance (ANOVA) with Sidak’s correction for multiple comparisons (GraphPad Prism V.6.02). Matrix accumulation results were compared with repeated measures two-way ANOVA with Holm-Sidak’s correction for multiple comparisons (GraphPad Prism V.6.02). Relative gene expression values were compared between surfaces at each passage with a ratio paired t-test (GraphPad Prism V.6.02) using Bonferroni correction to indicate significance (i.e. p < 0.05/3). For comparison between oxygen tensions, ratio paired t-tests were used (GraphPad Prism V.6.02) using Bonferroni correction to indicate significance. Comparisons of gene expression with the control (P1 TC plastic expansion) were performed with Linear trend post-hoc analysis (GraphPad Prism V.6.0.2). Values quoted in the text are mean ± S.D.

Results

Expansion

We found significantly enhanced growth of human chondrocytes on all synoviocyte matrix-coated flasks compared to cells on TC plastic alone (uncoated) at both atmospheric (20%) O2 (Fig 2A) and low (5%) O2 (Fig 2B) conditions, regardless of the method used to devitalize the matrix. Although none of the growth curves on devitalized matrix were significantly different from each other, they were all significantly different from those on uncoated plastic (p < 0.01). The growth rate on matrix-coated flasks was approximately double that on uncoated flasks (Fig 3A and 3B) with all devitalized synoviocyte matrix-coated flasks yielding significantly greater cell numbers than uncoated flasks (p < 0.002). Devitalization using the dry ice-cooled ethanol method (E-SCM) resulted in the most consistent surface, with no detachment of the cell layer, giving a significant increase in growth rate at atmospheric O2 over that found with the other methods (F-SCM & S-SCM; Fig 3A). At low O2, this increase in growth rate over the two other methods was less pronounced and only statistically significant between the E-SCM and S-SCM groups (Fig 3B). Within each expansion surface, oxygen tension did not have a significant effect on growth rate (Figures A and B in S1 Fig). Control comparisons between human chondrocyte growth on TC plastic, and expansion on TC plastic coated with gelatin, collagen type I, or fibronectin showed no apparent morphological or proliferative benefit from the coatings (S2 Fig).

thumbnail
Fig 2. Human chondrocyte expansion on plastic and devitalized synoviocyte matrix.

Cumulative population doublings; 5 independent experiments from 4 donors with 3 passages in each condition. A) treatment comparison at atmospheric O2. B) treatment comparison at low O2. Linear regressions from matrix expanded cells were significantly different compared to uncoated controls (p < 0.01).

https://doi.org/10.1371/journal.pone.0129961.g002

thumbnail
Fig 3. Population doubling rate on plastic and devitalized synoviocyte matrix.

The population doubling rate from 5 independent experiments from 4 donors with 3 passages in each condition. A) treatment comparison at atmospheric O2. B) treatment comparison at low O2. Conditions which do not share the same letter were significantly different (p < 0.002).

https://doi.org/10.1371/journal.pone.0129961.g003

Gene expression

RNA isolated from monolayer culture had high 260/280 nm ratio (2.03 ± 0.02) and good yield (377 ± 147 ng/μl giving 18.8 ± 7.4 μg total). RNA integrity numbers (RIN) also showed intact RNA (9.04 ± 0.83). Reference gene analysis showed HPRT to be the most stable across the conditions (S1 Table) and was similar for passage 1–3 with increased expression at P4 (Figure B in S3 Fig), and a linear trend was evident within each condition against population doublings (Figure A in S3 Fig). Further analysis showed that, within each passage, there was no significant difference in HPRT expression between the groups tested (Figures C-F in S3 Fig). Relative gene expression was therefore tested within passages, and comparison between passages was only made up to P3. Expansion on E-SCM significantly decreased collagen type I expression at both atmospheric and low oxygen tensions (Fig 4A). Collagen type I expression increased with passage on TC plastic (p = 0.0003 at Atm O2, p = 0.0010 at Low O2) with no significant trend on E-SCM. Type II collagen expression was also significantly lower when cultured on matrix at both oxygen tensions (Fig 4B). Type II collagen also showed a trend toward increased expression with passage on TC plastic (p = 0.0116 at Atm O2, p = 0.0075 at Low O2) with no significant trend on E-SCM. Type X collagen was unaffected by culture on synoviocyte matrix at either oxygen tension with no trend across passages (Figure A in S4 Fig). Aggrecan expression was significantly lower in chondrocytes cultured on synoviocyte matrix at both oxygen tensions (Fig 4C) with a negative linear trend on E-SCM at Low O2 (p < 0.0001). A statistically significant effect on SOX9 expression was only evident at P4 with an increase in expression on E-SCM over TC plastic (Figure B in S4 Fig). There was no significant effect on matrillin-1 expression under either oxygen tension (Figure C in S4 Fig) but the samples did appear to separate into two distinct levels of response at both oxygen tensions. MMP13 was significantly increased by expansion on E-SCM at both oxygen tensions (Fig 4D) with a negative linear trend on TC plastic at both oxygen tensions (p = 0.0049 at Atm O2, p = 0.0239 at Low O2) and no trend on E-SCM.

thumbnail
Fig 4. Effect of expansion culture surface on gene expression.

Human chondrocytes from 7 donors were expanded on TC plastic or E-SCM at either atmospheric or low O2. Each point represents a donor, and lines connect the condition. A) Fold change of COL1A1 relative to P1 TC plastic expansion. B) Fold change of COL2A1 relative to P1 TC plastic expansion. C) Fold change of ACAN relative to P1 TC plastic expansion. D) Fold change of ACAN relative to P1 TC plastic expansion. p values and significance are indicated above comparisons.

https://doi.org/10.1371/journal.pone.0129961.g004

Examining effect of oxygen tension on gene expression, there was a significant increase in collagen type I expression on TC plastic at P1 and P3 but not on E-SCM (Fig 5A). A statistically significant increase in collagen type II expression was observed only at P1 on TC plastic, with later passages showing a trend towards increased expression on TC plastic but not on E-SCM (Fig 5B). Oxygen tension had no effect on collagen type X expression under any condition (Figure A in S5 Fig). At first passage on TC plastic, there was a significant increase in aggrecan expression at low O2 with trends evident at other passages, but not on E-SCM (Figure B in S5 Fig). SOX9 expression was significantly increased under low oxygen tension on TC plastic, but was not significantly different on E-SCM (Fig 5C). When plotted against population doublings (Figure A in S6 Fig), SOX9 showed a strong negative linear trend with population doubling on TC plastic (r2 = 0.23, p = 0.0001 at Atm O2; r2 = 0.52, p = 0.0002 at Low O2), while the negative slope was minimal on cultures on E-SCM. Oxygen tension had no significant effect on matrillin-1 expression (Figure C in S5 Fig). There was a significant decrease in MMP13 expression at lower oxygen tension at P3 on TC plastic, with trends apparent in other groups (Fig 5D). MMP13 expression showed a significant negative trend with increasing passage on TC plastic (Figure B in S6 Fig; r2 = 0.45, p = 0.0008 at Atm O2, r2 = 0.41, p = 0.0017 at Low O2).

thumbnail
Fig 5. Effect of oxygen tension during expansion on gene expression.

Human chondrocytes from 7 donors were expanded on TC plastic or E-SCM at either atmospheric or low O2. Each point represents a donor, and lines connect the condition. A) HPRT normalized COL1A1 expression. B) HPRT normalized COL2A1 expression. C) HPRT normalized SOX9 expression. D) HPRT normalized MMP13 expression. p values and significance are indicated above comparisons.

https://doi.org/10.1371/journal.pone.0129961.g005

Re-differentiation

At atmospheric O2, cells that were expanded on devitalized matrix accumulated as much (F-SCM, E-SCM) or more (S-SCM) GAG than cells expanded on TC plastic alone, both in terms of total GAG and GAG/DNA (Fig 6). At low O2, there was no significant difference in GAG accumulation between the groups. A comparison between low and atmospheric O2 showed significantly more GAG accumulation at low O2 in all conditions (Fig 6). Regression analysis of the GAG/DNA content vs. PDs indicated that there was no significant difference in the slope of the lines for uncoated or coated flasks, but that there was a significant difference in the intercept, indicating that, for the same GAG content, cells expanded on devitalized matrix would have undergone a greater number of cell divisions while still producing the equivalent amount of GAG per DNA (Figures D and I in S7 Fig).

thumbnail
Fig 6. Glycosaminoglycan accumulation of differentially expanded human chondrocytes.

Human chondrocyte aggregates from 4 donors were analyzed from 3 passages on TC plastic and SCM devitalized by 3 methods; aggregates were cultured at atmospheric (open symbols) and low (closed symbols) O2 tension. Each point represents the average of the aggregates assessed for that experiment (n = 1–3); beside these are the overall mean of all 3 passages for the 4 donors ± S.D., n = 12.

https://doi.org/10.1371/journal.pone.0129961.g006

Cells which were expanded on E-SCM accumulated significantly more hydroxyproline (HP), corresponding to collagen, at both atmospheric and low O2 (Fig 7). Regression analysis of HP/DNA content vs. PDs indicated that coated flasks had similar slopes to uncoated flasks (or rates of loss of collagen content) but a higher intercept, indicating that at similar PDs, cells expanded on devitalized matrix would have greater collagen content (Figures E and J in S7 Fig).

thumbnail
Fig 7. Hydroxyproline accumulation of differentially expanded human chondrocytes.

Human chondrocyte aggregates from 4 donors were analyzed from 3 passages on uncoated flasks and synoviocyte matrix devitalized by 3 methods; aggregates were cultured at atmospheric (open symbols) and low (closed symbols) O2 tension. Each point represents the average of the aggregates assessed for that experiment (n = 1–3); mean and standard deviations are shown beside the individual data points, n = 12; * significantly different vs. uncoated at low O2 tension; there were no significant differences between hydroxyproline accumulation at different O2 tensions within the same culture conditions.

https://doi.org/10.1371/journal.pone.0129961.g007

Histologically, aggregates that were expanded on devitalized synoviocyte matrices showed Safranin-O staining that correlated qualitatively with the quantitative results seen by the GAG assay. Synoviocyte matrix-expanded cells in aggregates stained positively for GAG to a similar or greater extent despite having undergone significantly more PDs (Fig 8A). Immunohistochemistry for collagen type II showed similar or greater levels of staining in aggregates expanded on devitalized matrix, which was even more pronounced at low O2 (Fig 8B). Immunohistochemistry for secondary antibody alone, collagen type X and elastin were negative, and rabbit and human controls were positive (S8 Fig).

thumbnail
Fig 8. Histological assessment.

A) Glycosaminoglycan accumulation in aggregates—Human chondrocyte aggregates from all three passages on the different substrates were fixed, embedded and sectioned then stained with Safranin-O/Fast Green for analysis. Representative images from one donor/set of experiments are shown. All images are at the same magnification (100x). Numbers in the bottom left corner for each image indicate the cumulated population doublings for those cells. B) Type II collagen immunohistochemistry—Human chondrocyte aggregates stained with anti-type II collagen antibody from the first passage (P1) are shown. Representative images from one donor/experiment are shown. All images are at the same magnification (100x). Numbers in the bottom left corner for each image indicate the cumulated population doublings for those cells.

https://doi.org/10.1371/journal.pone.0129961.g008

Discussion

This study showed a significant improvement in human chondrocyte expansion with all 3 devitalized synoviocyte matrices compared to standard TC plates. In addition, low O2 promoted greater chondrogenesis during differentiation culture for both TC plastic- and synoviocyte matrix-expanded chondrocytes. The synoviocyte matrix-expanded chondrocytes, having undergone significantly more PDs, had levels of chondrogenesis similar to those for cells expanded on TC plastic.

This study also showed, in agreement with other studies, that expansion conditions can have a dramatic effect on chondrogenic capacity [18, 3335]. Growth factor supplementation during expansion (basic fibroblast growth factor [bFGF]), epidermal growth factor, platelet derived growth factor, TGFβ1) has been investigated for its effect on human chondrocyte expansion [33, 35]. Both studies showed greater re-differentiation after bFGF-supplemented expansion, but only one achieved an increased growth rate [33]. It is likely that the devitalized matrix, in addition to providing a 3D surface that is amenable to chondrocyte growth, contains a mix of growth factors which will be investigated in future studies. It is interesting to note that co-culture experiments of synoviocytes and chondrocytes have shown a decrease in chondrogenesis [36, 37]. This was true for both bovine cartilage/synovium [37] and osteoarthritic human cartilage/synovium [36], so whatever detrimental effects living synoviocytes had on the chondrocytes was not present in the extracted matrix.

Some decellularization methods include a DNase treatment in the processing of the tissue [38]; this was not included in our protocol as DNase treatment after devitalization had no beneficial effect on the proliferation rate of the cells. Also, DNases are naturally present in FBS used in the expansion media. In our study, the synoviocyte-matrix is not acting as a conduit for transplantation as is the case with many decellularized tissues, so residual nuclear material is a diminished concern. To ensure synoviocytes were devitalized, serum-containing media was incubated on the devitalized matrix, and no growth or nuclei were discernible using Hoechst (3 μg/ml in PBS with Mg and Ca, 33342; Life Technologies) staining after 6 days (S7 Fig).

We concur with Pei and He [14] in their description of the synoviocyte matrix as “a tissue-specific three-dimensional microenvironment for ex vivo expansion of articular chondrocytes”. Similar expansion and differentiation results were achieved with human chondrocytes in our study as those previously described with porcine chondrocytes [14]. While Pei et al. have found detergent-based decellularization methods acceptable for porcine chondrocytes [14], porcine synovial stem cells [15], and human mesenchymal stem cells (MSCs) [39], we did not find that to be the case for human chondrocytes. This may have been due to residual detergent [17], the extensive loss of porcine synoviocyte matrix components, or from inherent differences between osteoarthritic human chondrocytes and healthy porcine chondrocytes. While the chondrocytes in this study were harvested from macroscopically normal areas, all donors were undergoing total joint replacement, so these cells represent a realistic sampling of the kinds of cells that would be used for ACI. Of the devitalization methods tested, dry ice cooled Ethanol-based devitalization proved to be the most consistent and facile method as it was rapid and showed little loss of the cell matrix layer. TC plastic expansion is known to de-differentiate chondrocytes, their ability to redifferentiate diminishing with increased passage number. In this study, we observed that the cells on synoviocyte matrix-coated plastic still lost the ability to re-differentiate, but the curve has been shifted to the right to give greater PDs (5–10 depending on the metric), with the same amount of GAG and collagen. For example, in the TC plastic vs. E-SCM samples at atmospheric O2, at 5 μg GAG/DNA the TC plastic expanded sample has undergone 3.3 PDs while the E-SCM expanded sample has gone through 10.5 PDs. From an average Carticel biopsy of 180,000 to 455,000 cells [4], TC plastic flasks could yield 1.8–4.4 x 106 cells with chondrogenic potential, whereas E-SCM flasks could yield 2.6–6.4 x 108 cells with equivalent chondrogenic properties.

The effect of the matrix support on gene transcription has given some interesting insights into chondrocyte biology. While many mature chondrocyte genes were downregulated during expansion on synoviocyte matrix, they clearly rebound during differentiation. This highlights an important point: Chondrocyte expansion is a separate function from differentiation, and seeking to induce the expression of mature chondrocyte genes during expansion might be a misstep. These studies clearly showed that the culture surface has a dramatic effect on gene expression, and a relationship between the surface and oxygen tension was also evident. The diminished changes with passage observed between the control P1 condition and increased passages on E-SCM support the hypothesis that the chondrocytes cultured on E-SCM are maintained in a state from which they can redifferentiate more readily.

The effect of oxygen tension on gene expression during expansion was more pronounced at earlier passages and often abrogated by E-SCM expansion. This dulled effect is unlikely to be due to the increased proliferation rate causing hypoxia. Although the cells would have increased oxygen consumption, the diffusion rate of oxygen is high in vitro [40]. An increase in collagen type II expression was observed with increasing passage along with a concurrent decrease in SOX9 expression; this was most evident on TC plastic, which presents an interesting dichotomy. Aigner et al. [41] found that SOX9 did not correlate with COL2A1 expression in adult human articular chondrocytes, which was proposed to be due to post-translational events/subcellular localization and could also be valid reasoning in our studies. In contrast to the study by Lin et al. [42], we found increased levels of COL2A1 and COL1A1 and no effect on ACAN with increasing passage. This disparity could be due to the use of confluent cultures or the use of GAPDH as a reference gene. Ma et al. also report decreased COL2A1 expression with increased passage, but no data is reported for ACAN or COL1A1 [11]. This difference could also be due to expansion conditions as Ma et al. cultured cells at a high density (25,000 cells/cm2) and did not report population doublings. Our choice of timing was made with the effect of culture confluence in mind, such that cells were harvested before confluence was achieved on either TC plastic or E-SCM, ensuring the cells were in as similar a proliferative stage as possible. It has been suggested that the relative expression of type II collagen/type I collagen is critical in terms of articular chondrogenesis [43]. Surprisingly, in our study, a relatively high expression of collagen type II was maintained throughout the expansion on both TC plastic and E-SCM and was even seen to increase compared to P1 on TC plastic. A similarly high expression of type II collagen was observed when cells were cultured on cartilage matrix [44]. A comparison of the normalized data demonstrates that a high COL2A1/COL1A1 ratio was maintained throughout expansion culture (Fig 5A and 5B).

Although oxygen tension has long been realized to be important in chondrogenesis [45], its mechanism and chondrogenic outcomes are not clear [46] (Table 1). In contrast to Egli et al.[47], Qu et al. [22] and Schrobback et al.[19], but in agreement with Markway et al.[48] and Strobel et al.[49], re-differentiation of chondrocytes under low O2 increased GAG accumulation within the aggregates. No significant increase in collagen content was found in cells cultured and re-differentiated at low O2, which is in contrast to the results of Strobel et al.[49] but in agreement with Markway et al.[48]. However, an increase in collagen deposition in aggregates from cells that had been cultured on E-SCM was found. This, in addition to the collagen type II staining, could point towards an enhancement of chondrogenic capacity which was lost through culture on TC plastic. It is clear that the cell type, species, expansion conditions, time in culture and differentiation conditions are all important. Concentrating on only human cells, the overall impression is that low oxygen tension is beneficial in one or more outcome measures, especially for articular chondrogenesis (Table 1). However, some groups have seen negative effects of low oxygen tension, with others observing no significant effects, on one or more outcome measures. It is clear that more work is needed to investigate this important parameter in tissue culture.

In contrast to MSC chondrogenesis, where type X collagen and hypertrophy are commonly reported [27, 50], we observed no type X collagen protein expression in any of our chondrocyte cultures and very low relative gene expression. This is supportive of the use of chondrocytes as the cell of choice for tissue engineering of articular cartilage.

Donor and passage number were significant sources of variability within this study in terms of both gene expression and matrix synthesis. Large donor variability has been described by others [23, 51], with over a 1,000-fold difference in collagen gene expression at the end of first passage, which was not related to age or gender. Despite this, synoviocyte matrix coated flasks gave significantly more cells with all donors. Ideally, we would also have been able to compare between aggregates made from cells expanded on the different surfaces at the same number of PDs. A close approximation to this can be seen in the Safranin-O histology (Fig 6A) where, at P1, E-SCM-expanded cells at atmospheric O2 have undergone 5.7 PDs and, at P2, TC plastic-expanded cells have undergone 5.5 PDs, and there is significantly greater Safranin-O staining for the E-SCM-expanded cells. The lack of GAG staining in the later passage supports the work presented by Giovanni et al., in which a 3.6–4.2 PD limit for chondrogenic capacity of human chondrocytes is suggested [13]. However, expansion on synoviocyte matrix seems to extend this limit to upwards of 8 PDs.

These data clearly indicate that, relative to TC plastic, synoviocyte-derived matrix supports enhanced expansion of human-derived chondrocytes, such that the chondrocytes are maintained in a state from which they are able to re-differentiate into a cartilage phenotype after significantly more PDs.

Supporting Information

S1 Table. Assessment of reference genes for qPCR.

https://doi.org/10.1371/journal.pone.0129961.s001

(DOCX)

S1 Fig. Effect of oxygen tension on human articular chondrocyte growth.

A) Comparison of population doubling rate between each surface at atmospheric (open symbols) and low (5%) oxygen tension (closed symbols). B) qPCR expansion data, recapitulating the increased expansion rate and lack of effect of oxygen on human chondrocyte expansion.

https://doi.org/10.1371/journal.pone.0129961.s003

(TIF)

S2 Fig. Common tissue culture coated surface comparison.

Analysis of growth and morphology on common surface coatings. Chondrocytes were seeded on each of the surfaces and tracked for 7 days. All images are at the same magnification (100x).

https://doi.org/10.1371/journal.pone.0129961.s004

(TIF)

S3 Fig. Reference gene analysis.

HPRT was assessed across all conditions for its stability. A) Regression curve analysis vs. population doublings in each condition. B) Quantification cycle (Cq) assessment by passage, P4 was significantly different to all other passages (p < 0.0001, ANOVA). C) Comparison of Cq at P1 in each condition. D) Comparison of Cq at P2 in each condition. E) Comparison of Cq at P3 in each condition. F) Comparison of Cq at P4 in each condition. No significant differences were found for C-F.

https://doi.org/10.1371/journal.pone.0129961.s005

(TIF)

S4 Fig. Effect of expansion surface on the expression of COL10A1, SOX9 and MATN1.

Human chondrocytes from 7 donors were expanded on TC plastic or E-SCM at either atmospheric or low O2. Each point represents a donor and lines connect the condition. A) Fold change of COL10A1 relative to P1 TC plastic expansion. B) Fold change of SOX9 relative to P1 TC plastic expansion. C) Fold change of MATN1 relative to P1 TC plastic expansion. p values and significance are indicated above comparisons.

https://doi.org/10.1371/journal.pone.0129961.s006

(TIF)

S5 Fig. Effect of oxygen tension during expansion on COL10A1, ACAN and MATN1.

Human chondrocytes from 7 donors were expanded on TC plastic or E-SCM at either atmospheric or low O2. Each point represents a donor and lines connect the condition. A) HPRT normalized COL10A1 expression. B) HPRT normalized ACAN expression. C) HPRT normalized MATN1 expression. p values and significance are indicated above comparisons.

https://doi.org/10.1371/journal.pone.0129961.s007

(TIF)

S6 Fig. Correlation of SOX9 and MMP13 with cumulative population doublings.

A) Correlations of SOX9 expression with cumulative population doublings (P1–P3) in each condition were made. B) Correlations of MMP13 expression with cumulative population doublings (P1–P3) in each condition were made.

https://doi.org/10.1371/journal.pone.0129961.s008

(TIF)

S7 Fig. Regression analysis for synoviocyte-matrix expanded chondrocytes.

Regression analyses of biochemical measures against population doublings. Regressions were made combining all data from all 3 donors (n ≥ 23). A-E Atmospheric oxygen tension, F-J Low (5%) oxygen tension; A, F) Wet weight vs. population doublings; B,G) Total GAG (per aggregate) vs. population doublings; C,H) Total HP (per aggregate) vs. population doublings; D, I) Normalized GAG (GAG/DNA) vs. population doublings; E,J) Normalized HP (HP/DNA) vs. population doublings.

https://doi.org/10.1371/journal.pone.0129961.s009

(TIF)

S8 Fig. Immunohistochemistry controls.

Representative sections of control tissues: rabbit (Rb) articular and auricular cartilage and human (Hu) articular cartilage. Representative sections from aggregates produced from cells expanded at atmospheric O2 on tissue culture plastic (uncoated) or E-SCM are also shown.

https://doi.org/10.1371/journal.pone.0129961.s010

(PPTX)

S9 Fig. Growth media catalysed DNA degradation.

Synoviocyte-derived matrix was incubated with DMEM/FBS or PBS for 6 days; wells were then stained with Hoechst and imaged for nuclear staining (Fluorescence).

https://doi.org/10.1371/journal.pone.0129961.s011

(TIF)

Acknowledgments

The authors would like to thank Dr. Donald Lennon for the human MSC FBS selection, Amad Awadallah for sectioning and Safranin-O staining and the Benaroya Research Institute Matrix Biology Program for their support and discussion. We are very grateful to Terry Bertin for his advice, help and expertise in qPCR and the laboratory of Dr. Brendan Lee for the use of their equipment. Experiments were conducted at Case Western Reserve University, the Benaroya Research Institute, and Baylor College of Medicine. Funding was provided through NIH grants AR053622 and DE015322 to J.E.D. The II-II6B3 monoclonal antibody developed by T.F. Linsenmayer was obtained from the Developmental Studies Hybridoma Bank, created by the NICHD of the NIH and maintained at The University of Iowa, Department of Biology, Iowa City, IA 52242. This project was supported by the Genomic and RNA Profiling Core at Baylor College of Medicine and the expert assistance of the core director, Dr. Lisa D. White, Ph.D.

Author Contributions

Conceived and designed the experiments: TJK JED. Performed the experiments: TJK. Analyzed the data: TJK. Contributed reagents/materials/analysis tools: JED. Wrote the paper: TJK.

References

  1. 1. Hunter W. Of the Structure and Diseases of Articulating Cartilages. Philosophical Transactions of the Royal Society of London. 1743;42:514–21.
  2. 2. Hayes AJ, Tudor D, Nowell MA, Caterson B, Hughes CE. Chondroitin sulfate sulfation motifs as putative biomarkers for isolation of articular cartilage progenitor cells. J Histochem Cytochem. 2008;56(2):125–38. Epub 2007/10/17. doi: jhc.7A7320.2007 [pii] pmid:17938280; PubMed Central PMCID: PMC2324172.
  3. 3. Gudas R, Kalesinskas RJ, Kimtys V, Stankevicius E, Toliusis V, Bernotavicius G, et al. A prospective randomized clinical study of mosaic osteochondral autologous transplantation versus microfracture for the treatment of osteochondral defects in the knee joint in young athletes. Arthroscopy. 2005;21(9):1066–75. Epub 2005/09/21. doi: S0749-8063(05)00918-7 [pii] pmid:16171631.
  4. 4. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med. 1994;331(14):889–95. Epub 1994/10/06. pmid:8078550.
  5. 5. Kon E, Filardo G, Di Matteo B, Perdisa F, Marcacci M. Matrix assisted autologous chondrocyte transplantation for cartilage treatment: A systematic review. Bone Joint Res. 2013;2(2):18–25. Epub 2013/04/24. pmid:23610698; PubMed Central PMCID: PMC3626217.
  6. 6. Beris AE, Lykissas MG, Kostas-Agnantis I, Manoudis GN. Treatment of full-thickness chondral defects of the knee with autologous chondrocyte implantation: a functional evaluation with long-term follow-up. Am J Sports Med. 2012;40(3):562–7. Epub 2011/12/06. pmid:22138109.
  7. 7. Peterson L, Vasiliadis HS, Brittberg M, Lindahl A. Autologous chondrocyte implantation: a long-term follow-up. Am J Sports Med. 2010;38(6):1117–24. Epub 2010/02/26. pmid:20181804.
  8. 8. Shekkeris A, Perera J, Bentley G, Flanagan A, Miles J, Carrington R, et al. Histological Results of 406 Biopsies Following ACI/MACI Procedures for Osteochondral Defects in the Knee. Journal of Bone & Joint Surgery, British Volume. 2012;94-B(SUPP XXXVI):12.
  9. 9. Gikas PD, Bayliss L, Bentley G, Briggs TW. An overview of autologous chondrocyte implantation. J Bone Joint Surg Br. 2009;91(8):997–1006. Epub 2009/08/05. pmid:19651824.
  10. 10. Dell'Accio F, De Bari C, Luyten FP. Molecular markers predictive of the capacity of expanded human articular chondrocytes to form stable cartilage in vivo. Arthritis Rheum. 2001;44(7):1608–19. Epub 2001/07/24. pmid:11465712.
  11. 11. Ma B, Leijten JC, Wu L, Kip M, van Blitterswijk CA, Post JN, et al. Gene expression profiling of dedifferentiated human articular chondrocytes in monolayer culture. Osteoarthritis Cartilage. 2013;21(4):599–603. pmid:23376013.
  12. 12. Schnabel M, Marlovits S, Eckhoff G, Fichtel I, Gotzen L, Vecsei V, et al. Dedifferentiation-associated changes in morphology and gene expression in primary human articular chondrocytes in cell culture. Osteoarthritis Cartilage. 2002;10(1):62–70. Epub 2002/02/14. pmid:11795984.
  13. 13. 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. 2010;222(2):411–20. Epub 2009/11/06. pmid:19890919.
  14. 14. 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(5):2163–74. Epub 2011/07/28. pmid:21792932; PubMed Central PMCID: PMC3265606.
  15. 15. He F, Chen X, Pei M. Reconstruction of an in vitro tissue-specific microenvironment to rejuvenate synovium-derived stem cells for cartilage tissue engineering. Tissue Eng Part A. 2009;15(12):3809–21. Epub 2009/06/24. pmid:19545204.
  16. 16. Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization processes. Biomaterials. 2011;32(12):3233–43. Epub 2011/02/08. pmid:21296410; PubMed Central PMCID: PMC3084613.
  17. 17. Cebotari S, Tudorache I, Jaekel T, Hilfiker A, Dorfman S, Ternes W, et al. Detergent decellularization of heart valves for tissue engineering: toxicological effects of residual detergents on human endothelial cells. Artif Organs. 2010;34(3):206–10. Epub 2010/05/08. pmid:20447045.
  18. 18. 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(5):1585–93. Epub 2009/12/05. pmid:19958052; PubMed Central PMCID: PMC2952116.
  19. 19. Schrobback K, Klein TJ, Crawford R, Upton Z, Malda J, Leavesley DI. Effects of oxygen and culture system on in vitro propagation and redifferentiation of osteoarthritic human articular chondrocytes. Cell Tissue Res. 2012;347(3):649–63. Epub 2011/06/04. pmid:21638206.
  20. 20. Coyle CH, Izzo NJ, Chu CR. Sustained hypoxia enhances chondrocyte matrix synthesis. J Orthop Res. 2009;27(6):793–9. Epub 2008/12/04. pmid:19051248.
  21. 21. Babur BK, Ghanavi P, Levett P, Lott WB, Klein T, Cooper-White JJ, et al. The interplay between chondrocyte redifferentiation pellet size and oxygen concentration. PLoS One. 2013;8(3):e58865. Epub 2013/04/05. pmid:23554943; PubMed Central PMCID: PMC3598946.
  22. 22. Qu C, Lindeberg H, Ylarinne JH, Lammi MJ. Five percent oxygen tension is not beneficial for neocartilage formation in scaffold-free cell cultures. Cell Tissue Res. 2012;348(1):109–17. pmid:22392735.
  23. 23. Gawlitta D, van Rijen MH, Schrijver EJ, Alblas J, Dhert WJ. Hypoxia impedes hypertrophic chondrogenesis of human multipotent stromal cells. Tissue Eng Part A. 2012;18(19–20):1957–66. pmid:22563686.
  24. 24. Lennon D, Haynesworth S, Bruder S, Jaiswal N, Caplan A. Human and animal mesenchymal progenitor cells from bone marrow: Identification of serum for optimal selection and proliferation. In Vitro Cellular & Developmental Biology—Animal. 1996;32(10):602–11.
  25. 25. Kim TK, Sharma B, Williams CG, Ruffner MA, Malik A, McFarland EG, et al. Experimental model for cartilage tissue engineering to regenerate the zonal organization of articular cartilage. Osteoarthritis Cartilage. 2003;11(9):653–64. Epub 2003/09/05. doi: S1063458403001201 [pii]. pmid:12954236.
  26. 26. Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, et al. The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin Chem. 2009;55(4):611–22. Epub 2009/02/28. doi: clinchem.2008.112797 [pii] pmid:19246619.
  27. 27. Johnstone B, Hering TM, Caplan AI, Goldberg VM, Yoo JU. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res. 1998;238(1):265–72. Epub 1998/02/11. doi: S0014-4827(97)93858-1 [pii] pmid:9457080.
  28. 28. Carrino DA, Arias JL, Caplan AI. A spectrophotometric modification of a sensitive densitometric Safranin O assay for glycosaminoglycans. Biochem Int. 1991;24(3):485–95. Epub 1991/06/01. pmid:1772427.
  29. 29. Ham KD, Oegema TR, Loeser RF, Carlson CS. Effects of long-term estrogen replacement therapy on articular cartilage IGFBP-2, IGFBP-3, collagen and proteoglycan levels in ovariectomized cynomolgus monkeys. Osteoarthritis Cartilage. 2004;12(2):160–8. Epub 2004/01/16. doi: S1063458403002140 [pii]. pmid:14723875.
  30. 30. Ignat’eva N, Danilov N, Averkiev S, Obrezkova M, Lunin V, Sobol’ E. Determination of hydroxyproline in tissues and the evaluation of the collagen content of the tissues. Journal of Analytical Chemistry. 2007;62(1):51–7.
  31. 31. Whitney GA, Mera H, Weidenbecher M, Awadallah A, Mansour JM, Dennis JE. Methods for producing scaffold-free engineered cartilage sheets from auricular and articular chondrocyte cell sources and attachment to porous tantalum. Biores Open Access. 2012;1(4):157–65. Epub 2013/03/22. pmid:23514898; PubMed Central PMCID: PMC3559237.
  32. 32. Woessner JF Jr. The determination of hydroxyproline in tissue and protein samples containing small proportions of this imino acid. Arch Biochem Biophys. 1961;93:440–7. Epub 1961/05/01. pmid:13786180.
  33. 33. Jakob M, Demarteau O, Schafer D, Hintermann B, Dick W, Heberer M, et al. Specific growth factors during the expansion and redifferentiation of adult human articular chondrocytes enhance chondrogenesis and cartilaginous tissue formation in vitro. J Cell Biochem. 2001;81(2):368–77. Epub 2001/03/10. pmid:11241676.
  34. 34. van Osch GJ, Mandl EW, Marijnissen WJ, van der Veen SW, Verwoerd-Verhoef HL, Verhaar JA. Growth factors in cartilage tissue engineering. Biorheology. 2002;39(1–2):215–20. Epub 2002/06/26. pmid:12082284.
  35. 35. Yang KG, Saris DB, Geuze RE, Helm YJ, Rijen MH, Verbout AJ, et al. Impact of expansion and redifferentiation conditions on chondrogenic capacity of cultured chondrocytes. Tissue Eng. 2006;12(9):2435–47. Epub 2006/09/26. pmid:16995777.
  36. 36. Beekhuizen M, Bastiaansen-Jenniskens YM, Koevoet W, Saris DB, Dhert WJ, Creemers LB, et al. Osteoarthritic synovial tissue inhibition of proteoglycan production in human osteoarthritic knee cartilage: establishment and characterization of a long-term cartilage-synovium coculture. Arthritis Rheum. 2011;63(7):1918–27. Epub 2011/03/26. pmid:21437876.
  37. 37. Steinhagen J, Bruns J, Niggemeyer O, Fuerst M, Ruther W, Schunke M, et al. Perfusion culture system: Synovial fibroblasts modulate articular chondrocyte matrix synthesis in vitro. Tissue Cell. 2010;42(3):151–7. Epub 2010/04/30. pmid:20427066.
  38. 38. Gilbert TW, Sellaro TL, Badylak SF. Decellularization of tissues and organs. Biomaterials. 2006;27(19):3675–83. Epub 2006/03/08. doi: S0142-9612(06)00168-2 [pii] pmid:16519932.
  39. 39. Pei M, He F, Kish VL. Expansion on extracellular matrix deposited by human bone marrow stromal cells facilitates stem cell proliferation and tissue-specific lineage potential. Tissue Eng Part A. 2011;17(23–24):3067–76. Epub 2011/07/12. pmid:21740327; PubMed Central PMCID: PMC3226057.
  40. 40. Androjna C, Gatica JE, Belovich JM, Derwin KA. Oxygen diffusion through natural extracellular matrices: implications for estimating "critical thickness" values in tendon tissue engineering. Tissue Eng Part A. 2008;14(4):559–69. pmid:18377199.
  41. 41. Aigner T, Gebhard PM, Schmid E, Bau B, Harley V, Poschl E. SOX9 expression does not correlate with type II collagen expression in adult articular chondrocytes. Matrix Biol. 2003;22(4):363–72. pmid:12935820.
  42. 42. Lin Z, Fitzgerald JB, Xu J, Willers C, Wood D, Grodzinsky AJ, et al. Gene expression profiles of human chondrocytes during passaged monolayer cultivation. J Orthop Res. 2008;26(9):1230–7. pmid:18404652.
  43. 43. Albrecht C, Tichy B, Nurnberger S, Hosiner S, Zak L, Aldrian S, et al. Gene expression and cell differentiation in matrix-associated chondrocyte transplantation grafts: a comparative study. Osteoarthritis Cartilage. 2011;19(10):1219–27. pmid:21820068.
  44. 44. Shahdadfar A, Loken S, Dahl JA, Tunheim SH, Collas P, Reinholt FP, et al. Persistence of collagen type II synthesis and secretion in rapidly proliferating human articular chondrocytes in vitro. Tissue Eng Part A. 2008;14(12):1999–2007. Epub 2008/07/19. pmid:18636947.
  45. 45. Bassett CA, Herrmann I. Influence of oxygen concentration and mechanical factors on differentiation of connective tissues in vitro. Nature. 1961;190:460–1. pmid:13687643.
  46. 46. Malda J, Martens DE, Tramper J, van Blitterswijk CA, Riesle J. Cartilage tissue engineering: controversy in the effect of oxygen. Crit Rev Biotechnol. 2003;23(3):175–94. Epub 2004/01/28. pmid:14743989.
  47. 47. Egli RJ, Bastian JD, Ganz R, Hofstetter W, Leunig M. Hypoxic expansion promotes the chondrogenic potential of articular chondrocytes. J Orthop Res. 2008;26(7):977–85. Epub 2008/02/28. pmid:18302236.
  48. 48. Markway BD, Cho H, Johnstone B. Hypoxia promotes redifferentiation and suppresses markers of hypertrophy and degeneration in both healthy and osteoarthritic chondrocytes. Arthritis Res Ther. 2013;15(4):R92. pmid:23965235; PubMed Central PMCID: PMC3979022.
  49. 49. Strobel S, Loparic M, Wendt D, Schenk AD, Candrian C, Lindberg RL, et al. Anabolic and catabolic responses of human articular chondrocytes to varying oxygen percentages. Arthritis Res Ther. 2010;12(2):R34. Epub 2010/03/03. doi: ar2942 [pii] pmid:20193091; PubMed Central PMCID: PMC2888180.
  50. 50. Hellingman CA, Koevoet W, van Osch GJ. Can one generate stable hyaline cartilage from adult mesenchymal stem cells? A developmental approach. J Tissue Eng Regen Med. 2012;6(10):e1–e11. Epub 2011/11/23. pmid:22106029.
  51. 51. Grunder T, Gaissmaier C, Fritz J, Stoop R, Hortschansky P, Mollenhauer J, et al. Bone morphogenetic protein (BMP)-2 enhances the expression of type II collagen and aggrecan in chondrocytes embedded in alginate beads. Osteoarthritis Cartilage. 2004;12(7):559–67. Epub 2004/06/29. pmid:15219571.
  52. 52. Collins JA, Moots RJ, Winstanley R, Clegg PD, Milner PI. Oxygen and pH-sensitivity of human osteoarthritic chondrocytes in 3-D alginate bead culture system. Osteoarthritis Cartilage. 2013;21(11):1790–8. pmid:23850530; PubMed Central PMCID: PMC3807787.
  53. 53. Murphy CL, Polak JM. Control of human articular chondrocyte differentiation by reduced oxygen tension. J Cell Physiol. 2004;199(3):451–9. pmid:15095292.
  54. 54. Das RH, van Osch GJ, Kreukniet M, Oostra J, Weinans H, Jahr H. Effects of individual control of pH and hypoxia in chondrocyte culture. J Orthop Res. 2010;28(4):537–45. Epub 2009/10/09. pmid:19813243.
  55. 55. Duval E, Leclercq S, Elissalde JM, Demoor M, Galera P, Boumediene K. Hypoxia-inducible factor 1alpha inhibits the fibroblast-like markers type I and type III collagen during hypoxia-induced chondrocyte redifferentiation: hypoxia not only induces type II collagen and aggrecan, but it also inhibits type I and type III collagen in the hypoxia-inducible factor 1alpha-dependent redifferentiation of chondrocytes. Arthritis Rheum. 2009;60(10):3038–48. pmid:19790048.
  56. 56. Dare EV, Griffith M, Poitras P, Wang T, Dervin GF, Giulivi A, et al. Fibrin sealants from fresh or fresh/frozen plasma as scaffolds for in vitro articular cartilage regeneration. Tissue Eng Part A. 2009;15(8):2285–97. pmid:19226200.
  57. 57. Foldager CB, Nielsen AB, Munir S, Ulrich-Vinther M, Soballe K, Bunger C, et al. Combined 3D and hypoxic culture improves cartilage-specific gene expression in human chondrocytes. Acta Orthop. 2011;82(2):234–40. pmid:21434761; PubMed Central PMCID: PMC3235297.
  58. 58. Thoms BL, Murphy CL. Inhibition of hypoxia-inducible factor-targeting prolyl hydroxylase domain-containing protein 2 (PHD2) enhances matrix synthesis by human chondrocytes. J Biol Chem. 2010;285(27):20472–80. pmid:20404338; PubMed Central PMCID: PMC2898297.
  59. 59. Thoms BL, Dudek KA, Lafont JE, Murphy CL. Hypoxia promotes the production and inhibits the destruction of human articular cartilage. Arthritis Rheum. 2013;65(5):1302–12. pmid:23334958.
  60. 60. Legendre F, Ollitrault D, Hervieu M, Bauge C, Maneix L, Goux D, et al. Enhanced hyaline cartilage matrix synthesis in collagen sponge scaffolds by using siRNA to stabilize chondrocytes phenotype cultured with bone morphogenetic protein-2 under hypoxia. Tissue Eng Part C Methods. 2013;19(7):550–67. pmid:23270543.
  61. 61. Duval E, Bauge C, Andriamanalijaona R, Benateau H, Leclercq S, Dutoit S, et al. Molecular mechanism of hypoxia-induced chondrogenesis and its application in in vivo cartilage tissue engineering. Biomaterials. 2012;33(26):6042–51. pmid:22677190.
  62. 62. Malda J, van Blitterswijk CA, van Geffen M, Martens DE, Tramper J, Riesle J. Low oxygen tension stimulates the redifferentiation of dedifferentiated adult human nasal chondrocytes. Osteoarthritis Cartilage. 2004;12(4):306–13. pmid:15023382.