You are on page 1of 12

ARTHRITIS & RHEUMATISM

Vol. 44, No. 7, July 2001, pp 1608–1619


© 2001, American College of Rheumatology
Published by Wiley-Liss, Inc.

Molecular Markers Predictive of the Capacity of


Expanded Human Articular Chondrocytes to
Form Stable Cartilage In Vivo

Francesco Dell’Accio, Cosimo De Bari, and Frank P. Luyten

Objective. To establish a model and associated receptor–like kinase 1 messenger RNA. Anchorage-
molecular markers for monitoring the capacity of in independent growth and the reexpression of COL2A1 in
vitro–expanded chondrocytes to generate stable carti- agarose culture were insufficient to predict cartilage
lage in vivo. formation in vivo.
Methods. Adult human articular chondrocytes Conclusion. AHAC have a finite capacity to form
(AHAC) were prepared by collagenase digestion of stable cartilage in vivo; this capacity is lost throughout
samples obtained postmortem and were expanded in passaging and can be monitored using a nude mouse
monolayer. Upon passaging, aliquots of chondrocyte model and associated molecular markers. This cartilage-
suspensions were either injected intramuscularly into forming ability in vivo may be pivotal for successful
nude mice, cultured in agarose, or used for gene expres- cell-based joint surface defect repair protocols.
sion analysis. Cartilage formation in vivo was docu-
mented by histology, histochemistry, immunofluores- Joint destruction represents the outcome of most
cence for type II collagen, and proteoglycan analysis by inflammatory and degenerative rheumatic diseases and
35
S-sulfate incorporation and molecular sieve chroma- leads to severe disability. Damage of the joint surface
tography of the radiolabeled macromolecules. In situ often results in end-stage osteoarthritis, requiring surgi-
hybridization for species-specific genomic repeats was cal intervention and total joint replacement. Its high
used to discriminate human-derived from mouse- prevalence, heavy impact on working capacity, and the
derived cells. Gene expression dynamics were analyzed high costs and morbidity associated with current thera-
by semiquantitative reverse transcription–polymerase peutic approaches make joint disease a major social
chain reaction. issue.
Results. Intramuscular injection of freshly iso- Recent developments in basic science and clinical
lated AHAC into nude mice resulted in stable cartilage rheumatology have achieved a substantial understanding
implants that were resistant to mineralization, vascular of the mechanisms leading to cartilage breakdown and
invasion, and replacement by bone. In vitro expansion of considerable control of the progression of the damage to
AHAC resulted in the loss of in vivo cartilage formation. the joint surface. In contrast, the range of interventions
This capacity was positively associated with the expres- for the repair of joint surface defects (JSD) is often
sion of fibroblast growth factor receptor 3, bone mor- limited to prosthetic replacement of end-stage joints.
phogenetic protein 2, and ␣1(II) collagen (COL2A1), Modern tissue engineering strategies offer an opportu-
and its loss was marked by the up-regulation of activin nity for biologic repair of JSD (1,2). Autologous chon-
drocyte transplantation (ACT) represents a paradigm in
Supported by FWO grant G.0192.99. biologic repair for JSD (3,4). In the original description,
Francesco Dell’Accio, MD, Cosimo De Bari, MD, Frank P. autologous chondrocytes were obtained arthroscopically
Luyten, MD, PhD: University Hospitals Katholieke Universiteit Leu- via cartilage biopsy from a minor weight-bearing area of
ven, Leuven, Belgium.
Address correspondence and reprint requests to Frank P. the joint and were expanded in vitro in the presence of
Luyten, MD, PhD, Laboratory for Skeletal Development and Joint autologous serum. In a subsequent open knee surgery
Disorders, Onderwijs & Navorsing, Herestraat 49, 3000 Leuven, procedure, expanded chondrocytes were injected into
Belgium.
Submitted for publication November 3, 2000; accepted in the JSD under a periosteal flap sutured to the cartilage
revised form March 14, 2001. to seal the JSD (3).
1608
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1609

Conceptual problems inherent to the technique necessary, but are not necessarily sufficient for the
are the origin of the cells and the expansion in vitro. The formation of cartilage tissue in vivo, starting from a
cartilage biopsy, although taken from a non–weight- suspension of isolated chondrocytes. These assays, in-
bearing area of the joint, is itself an additional injury to deed, have never been shown to predict the capacity to
the joint surface. Moreover, in vitro expansion of chon- form stable cartilage in vivo. An assay that directly
drocytes is known to result in a phenotypic derange- measures cartilage formation in vivo is therefore
ment, often referred to as dedifferentiation (5). There is needed.
a possibility that the loss of phenotypic traits in vitro We have standardized an in vivo assay in nude
could reflect a progressive loss of potential to form in mice to measure the potential of adult human articular
vivo stable cartilage that is resistant to vascular invasion, chondrocytes (AHAC) to form stable cartilage in vivo
mineralization, and bone formation, thereby jeopardiz- that is resistant to vascular invasion, dystrophic calcifi-
ing the long-term outcome of ACT. The use of progen- cation, and bone formation. We have identified a set of
itor cells from, for example, bone marrow (6) or perios- molecular markers that reproducibly predicts the out-
teum (7) may potentially circumvent these problems. come of our in vivo assay, independently of donor age.
Progenitor cells can be obtained without inducing This set of molecular markers proved to be more
irreversible damage, and they are extensively expandable accurate than the agarose assay in predicting the in vivo
and phenotypically stable throughout expansion (8,9). cartilage-forming ability, and may therefore be useful in
The use of mesenchymal cells is restricted by the as-yet- assessing the suitability of chondrocyte suspensions for
insufficient knowledge of the long-term stability of the cell-based joint surface repair protocols.
repair tissue and by their tendency to differentiate
toward other cell lineages. Their multilineage potential PATIENTS AND METHODS
may present a risk of heterotopic tissue formation
Cartilage harvest and chondrocyte isolation. Normal
(10,11). articular cartilage was obtained from the femoral condyles of
ACT offers the advantage of using cells that are human donors within 12 hours postmortem. Cartilage was
stably committed to the appropriate phenotype and natu- sliced full thickness, excluding the mineralized cartilage and
rally resistant to vascular invasion, mineralization, and the subchondral bone, and placed in Hanks’ balanced salt
ossification. Therefore, it is crucial to ensure that this solution (HBSS; Life Technologies, Merelbeke, Belgium) sup-
phenotypic stability is retained throughout expansion. To plemented with 2⫻ antibiotic–antimycotic solution (final con-
centrations 200 units/ml of penicillin, 200 ␮g/ml of streptomy-
date, unfortunately, the choice of the appropriate assay to cin, and 0.5 ␮g/ml of amphotericin B; Life Technologies).
measure this property is rather unsatisfactory, being lim- Cartilage slices were minced and washed twice in HBSS
ited to the expression of some extracellular matrix (ECM) supplemented with 2⫻ antibiotic–antimycotic solution for 5
molecules such as type II collagen and to anchorage- minutes at 37°C.
independent growth in agarose culture (5). Chondrocytes were released by overnight digestion at
37°C in 0.2% crude type IV collagenase (Life Technologies) in
Tissue regeneration is thought to recapitulate the high-glucose Dulbecco’s modified Eagle medium (DMEM;
molecular events that take place during embryonic de- Life Technologies) containing 10% fetal bovine serum (FBS;
velopment (12–18). It requires not only cell proliferation BioWhittaker, Verviers, Belgium), and 2⫻ antibiotic–
and deposition of the proper ECM, but also reproduc- antimycotic solution. Cells were washed twice in DMEM
tion of the tissue architecture and patterning (1,19). This supplemented with 10% FBS and 2⫻ antibiotic–antimycotic
solution and then counted. Trypan blue exclusion was used to
appears to be particularly relevant for articular cartilage, adjust for the number of viable cells. Routinely, ⬎95% of the
since the expression of cartilage-specific ECM mole- cells were viable.
cules, such as type II collagen, and anchorage- Chondrocyte expansion and agarose culture. Chon-
independent growth in agarose culture (5) have been drocytes were plated at a density of 5 ⫻ 104 cells/cm2 in growth
used to evaluate the phenotypic stability of expanded medium (DMEM supplemented with 10% FBS and 1⫻
antibiotic–antimycotic solution at final concentrations of 100
chondrocytes for ACT (3). Type II collagen expression units/ml of penicillin, 100 ␮g/ml of streptomycin, and 0.25
reports the differentiation state of the cells at the ␮g/ml of amphotericin B) and cultured at 37°C in a humidified
moment of the test. The agarose assay consists of atmosphere containing 5% CO2. Cells were released 10 days
evaluating the capacity to grow in anchorage- after confluence by trypsin–EDTA (0.25% trypsin, 1 mM
independent conditions and to reexpress cartilage- disodium EDTA; Life Technologies) treatment. Cultures in
LTM agarose were performed as described elsewhere (5).
specific markers in low temperature melting (LTM) Nude mouse assay. Five million viable cells were
agarose in in vitro culture (5). Anchorage-independent resuspended in 50 ␮l of phosphate buffered saline (PBS) and
growth and capacity to differentiate are likely to be injected intramuscularly into the adductor muscles of the
1610 DELL’ACCIO ET AL

Table 1. Sequences of the primers used for polymerase chain reaction and size of the products*
Product
Genes Sense primer Reverse primer size (bp)
COL2A1 5⬘-CTGCTCGTCGCCGCTGTCCTT-3⬘ 5⬘-AAGGGTCCCAGGTTCTCCATC-3⬘ 511
FGFR-3 5⬘-GCTGAAAGACGATGCCACTG-3⬘ 5⬘-AGGACCCCAAAGGACCAGAC-3⬘ 522
BMP-2 5⬘-CAGAGACCCACCCCCAGCA-3⬘ 5⬘-CTGTTTGTGTTTGGCTTGAC-3⬘ 672
ALK-1 5⬘-CGACGGAGGCAGGAGAAGCAG-3⬘ 5⬘-TGAAGTCGCGGTGGGCAATGG-3⬘ 565
␤-actin 5⬘-TGACGGGGTCACCCACACTGTGCCCATCTA-3⬘ 5⬘-CTAGAAGCATTTGCGGTGGACGATGGAGGG-3⬘ 661
Aggrecan 5⬘-GTTGTCATCAGCACCAGCATC-3⬘ 5⬘-ACCACACAGTCCTCTCCAGC-3⬘ 509
Versican 5⬘-AACATTTTTCAGGTGGTGAG-3⬘ 5⬘-GATGCAGAACTTGGAACTAT-3⬘ 304
Biglycan 5⬘-TCCCAGACCTCAAGCTCCTC-3⬘ 5⬘-TGGAGACTTGGACACTTGGG-3⬘ 544
Decorin 5⬘-ATTGTCATAGAACTGGGCAC-3⬘ 5⬘-AAGACTCACACCCGAATAAG-3⬘ 459
Fibromodulin 5⬘-CTGGACGGGAACGAGATCAA-3⬘ 5⬘-TGTGGAGGAAAGTGGAGTGG-3⬘ 577
Lumican 5⬘-ACCTTGAAAACTATTACCTGGAG-3⬘ 5⬘-ATTGAGTTTGATGTCTATTCGTG-3⬘ 407
␤2m 5⬘-GTGTCTGGGTTTCATCAATC-3⬘ 5⬘-GGCAGGCATACTCATCTTTT-3⬘ 163

* COL2A1 ⫽ ␣1(II) collagen; FGFR-3 ⫽ fibroblast growth factor receptor 3; BMP-2 ⫽ bone morphogenetic protein 2; ALK-1 ⫽ activin
receptor–like kinase receptor 1; ␤2m ⫽ ␤2-microglobulin.

thighs of 4–5-week-old female NMRI nu/nu mice. Animals Briefly, explants from adult human knee articular cartilage and
were maintained in isolator cages on an unrestricted diet. All 3 implants from injected AHAC were minced and radiolabeled
the procedures were approved by the local ethics committee. for 6 hours at 37°C in DMEM supplemented with 35S-sulfate at
Mice were killed by cervical dislocation after 3 weeks unless 50 ␮Ci/ml. Radiolabeled tissues were extracted for 48 hours at
otherwise specified. room temperature in 4M guanidine HCl, 0.2% (v/v) Triton
Histochemistry. Formalin-fixed paraffin-embedded X-100, and 50 mM sodium acetate, pH 5.8, containing protease
sections were stained with Alcian blue, pH 2.5, toluidine blue, inhibitors (5 mM benzamidine HCl, 10 mM EDTA, 10 mM
Masson’s trichrome, and Safranin O according to standard N-ethylmaleimide, and 1 mM phenylmethylsulfonyl fluoride).
protocols. The residual material was then extracted with 0.5M sodium
Indirect immunofluorescence. After deparaffinization, hydroxide to determine the efficiency of the extraction procedure.
sections were digested with 0.25 mg/ml of pepsin (Sigma, Unincorporated radiolabel was removed by Sephadex
Bornem, Belgium) in 0.2N HCl for 1 hour at 37°C, postfixed in G-25 gel chromatography (PD-10 columns; Amersham Pharma-
buffered 4% paraformaldehyde for 10 minutes at room tem- cia Biotech, Roosendaal, The Netherlands) equilibrated in 7M
perature, and washed extensively with PBS. Slides were incu- urea, 0.2M NaCl, 0.2% (v/v) Triton X-100, and 50 mM sodium
bated twice in 50 mM ammonium chloride in PBS for 10 acetate, pH 6.0. An aliquot of the radiolabeled macromolecular
minutes, washed in PBS, and blocked with Blocking Reagent fraction was analyzed on a Sephacryl S-500 HR column (1 ⫻ 30
(Roche Molecular Biochemicals, Brussels, Belgium) supple- cm; Amersham Pharmacia Biotech) equilibrated with 4M guani-
mented with 0.2% (volume/volume) Triton X-100 (Bio-Rad, dine HCl, 0.5% (v/v) Triton-X 100 in 50 mM sodium acetate, pH
Nazareth Eke, Belgium). 6. The flow rate was 0.4 ml/minute. Fractions were collected at
Slides were incubated overnight at 4°C with a mouse 1-minute intervals and counted for radioactivity.
anti-human type II collagen antibody (Chemicon, Hofheim, In situ hybridization for species-specific genomic re-
Germany) diluted 1:5, extensively washed with PBS containing peats. In situ hybridization for human-specific ALU repeats
0.2% Triton X-100, and incubated for 1 hour at room temper- was performed as described elsewhere (24), with a few modi-
ature with Cy3-conjugated goat anti-mouse IgG antibody fications. Briefly, sections were deparaffinized and rehydrated.
(Jackson ImmunoResearch, De Pinte, Belgium) diluted 1:200. Matrix digestion was obtained by enzymatic treatment with 1
After 3 washes in PBS–0.2% Triton X-100, slides were coun- mg/ml of pepsin in 0.2N HCl for 10 minutes at 37°C. Sections
terstained with 4⬘,6-diamidino-2-phenylindole (DAPI; ICN, were immediately postfixed in 3% formaldehyde for 10 min-
Asse-Relegem, Belgium) and mounted with Mowiol utes and equilibrated in PBS. Sections were acetylated with
(Calbiochem-Merck Belgolabo, Overijse, Belgium). Red and 0.25% acetic acid containing 0.1M triethanolamine, pH 8, for
blue digital images were acquired by Spot camera and super- 10 minutes, and prehybridized with 50% deionized formamide
imposed using Spot software version 3.0.4 (Diagnostic Instru- containing 4⫻ saline–sodium citrate (SSC) at 37°C for 15
ments, Sterling Heights, MI). minutes. Sections were covered with hybridization buffer (1⫻
Chondrocyte devitalization and irradiation. To obtain Denhardt’s solution, 5% Dextran sulfate, 0.2 mg/ml denatured
nonviable chondrocytes, cell suspensions were snap-frozen and sheared salmon sperm DNA, 4⫻ SSC, and 50% deionized
thawed at 37°C for 3 minutes for 3 consecutive times as formamide) containing 1 ng/␮l of a digoxigenin (DIG)–labeled
described in detail elsewhere (20). To arrest cell division while double-stranded DNA probe specific for human ALU genomic
preserving cell viability, chondrocytes were x-irradiated with a repeats (a kind gift from K. Satomura, First Department of
single radiation dose of 50 Gy (21,22). Oral and Maxillofacial Surgery, University of Tokushima,
35
S-sulfate incorporation and hydrodynamic profile of Tokushima, Japan) and covered with a glass coverslip.
proteoglycans. The analysis of 35S-sulfate incorporation into Denaturation of both the probe and the genomic DNA
macromolecules was performed as described elsewhere (23). template was achieved by heating the slides at 95°C for 45
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1611

seconds. Hybridization was performed at 42°C for 3 hours.


Sections were washed twice for 30 minutes at room tempera-
ture with 2⫻ SSC and 0.1⫻ SSC. Digoxigenin was detected
using a commercially available kit (DIG Nucleic Acid Detec-
tion kit; Roche Molecular Biochemicals) according to the
manufacturer’s protocol.
DIG-labeled mouse-specific L1 probe was obtained as
described previously (25). In situ hybridization for m-specific
L1 repeats was carried out as described above for human ALU
repeats, with a few modifications. Hybridization was allowed
overnight at 42°C, and stringency washes were twice for 30
minutes at room temperature in 1⫻ SSC and once for 30
minutes at 50°C in 0.5⫻ SSC.
Reverse transcription–polymerase chain reaction (RT-
PCR) analysis. Total RNA was isolated using TRIzol reagent
(Life Technologies) according to the manufacturer’s instruc-
tions. Total RNA from agarose cultures was obtained as
described elsewhere (26). Complementary DNA (cDNA) were
obtained by reverse transcription of 1 ␮g of total RNA
(Thermoscript; Life Technologies) using oligo(dT)20 as a
primer. PCR was performed in a 10-␮l volume. The cDNA
were added to the following PCR mixture: 0.5 units of Taq
polymerase (Eurogentec, Seraing, Belgium), 0.2 mM dNTPs,
0.5 ␮M specific primers, and 1.5 mM MgCl2. Negative controls
were either RT without enzyme or PCR without cDNA
template. PCR reactions were carried out in a Perkin Elmer
Thermal Cycler model 9600 (Applied Biosystems, Lennik,
Belgium). The sequences of the primers and the expected sizes
of the amplification products are listed in Table 1.
Marker analysis by PCR was preceded by equalization
of cDNA for the housekeeping gene ␤-actin. Figure 1. Generation of cartilage tissue by freshly isolated adult
human articular chondrocytes after intramuscular injection into nude
mice. a and b, Safranin O staining of adult human articular cartilage
RESULTS harvested from the femoral condyle. c and d, Safranin O staining of a
cartilage implant. b and d, Higher-magnification view of the boxed
Generation of a stable hyaline-like cartilage im- areas shown in a and c, respectively. Compared with adult human
plant in nude mice. The intramuscular injection of 5 ⫻ articular cartilage, the implant is hypercellular. e, Masson’s trichrome
106 freshly isolated AHAC into the posterior compart- staining displays the absence of neoangiogenesis or endochondral bone
ment of the thigh of NMRI nu/nu mice resulted in the formation. f, Immunofluorescence for type II collagen is positive in the
extracellular matrix of the implant. Asterisk indicates adjacent muscle
reproducible (15 of 15) retrieval of a distinct cartilage
tissue. Nuclei are counterstained with 4⬘,6-diamidino-2-phenylindole.
implant as early as 7 days after injection. The implant Bars ⫽ 200 ␮m.
was stable for at least 12 weeks, without signs of vascular
invasion, dystrophic calcification, or bone formation.
Calcium deposits were undetectable by alizarin red performed. Masson’s trichrome as well as hematoxylin
staining at all time points examined, up to 12 weeks and eosin preparations did not reveal the presence of
(data not shown). dense collagen bundles as in fibrocartilage. These over-
Safranin O staining was approximately as strong all characteristics allowed us to consider the cartilage
in the implants (Figures 1c and d) as in normal adult implants to be of hyaline-like nature.
articular cartilage (Figures 1a and b), indicating the To study the specificity of the assay, we injected
presence of sulfated proteoglycans, but the implants human primary skin fibroblasts and primary cells and
were hypercellular and lacked typical columnar organi- cell lines known to have chondrogenic potential. The
zation. Chondrocytes were embedded in typical lacunae. injection of primary human skin fibroblasts (kindly
Masson’s trichrome staining (Figure 1e) revealed no provided by S. Tejpar, Centrum Menselijke Erfelijkheid,
vascular invasion or bone formation. The detection of Katholieke Universiteit Leuven, Leuven, Belgium)
type II collagen protein by immunostaining further failed to produce any detectable implant. The injection
supported the cartilaginous nature of the implant (Fig- of primary adult human periosteal cells resulted in
ure 1f). The ECM was homogeneous with all the stains fibrous tissue formation (data not shown). The injection
1612 DELL’ACCIO ET AL

Figure 2. Hydrodynamic profiles of sulfated proteoglycans and proteoglycan expression profile.


35
S-sulfate incorporation and size fractionation of macromolecules in 3 implants (a) shows the
presence of high molecular weight proteoglycans with the same hydrodynamic size as those in adult
human articular cartilage explants (b). V0 ⫽ void volume; Vt ⫽ total volume. Proteoglycan
expression profile generated by semiquantitative reverse transcription–polymerase chain reaction
analysis with RNA obtained from a cartilage implant (IMP) (c). Confluent primary adult human
articular chondrocytes (P0) were used as positive control. Uninjected mouse muscle (MM) was
used as a negative control. The template cDNA from MM and IMP were equalized for the
expression of ␤-actin using primers that amplify both the mouse and human (mh) ␤-actin cDNA.
Human cDNA from IMP and P0 were normalized for the number of human cells using
human-specific primers (h) for ␤2-microglobulin. All the primers used to detect proteoglycan
cDNA were human-specific.

of cell lines that have in vitro chondrogenic potential, quired, we x-irradiated chondrocytes with a single 50-Gy
such as the mouse teratocarcinoma cell line ATDC5 (27) dose, which blocks proliferation but does not kill the
or the rat calvaria cell lines RCJ (28), RCJ 3.1 C5.18 cells (21,22). Under our conditions, chondrocytes that
(29), and CFK2 (30), did not produce any retrievable had been made unable to proliferate by x-irradiation
implants under the conditions we used. could still generate a cartilage implant in vivo (data not
It is known that cartilage formation can be in- shown).
duced in similar systems by nonviable materials contain- Proteoglycan synthesis and characterization.
ing chondrogenic factors, such as transforming growth High molecular weight sulfated proteoglycans constitute an
factor ␤ (TGF␤) and bone morphogenetic proteins important component of the cartilage matrix (23) and are
(BMPs) (31–33) or nonchondrogenic cells producing critical for the biomechanical properties of articular carti-
those factors (34–36). To address whether, in our sys- lage, such as elasticity and resistance to load and impact
tem, living chondrocytes were required, we implanted, in (37). Incorporation of 35S-sulfate and size fractionation of
parallel and in duplicate, living chondrocytes and chon- macromolecules (Figure 2) showed the presence in the
drocytes that had been lysed by repeated freezing and implants of high molecular weight sulfated proteoglycans
thawing (20). Only the injection of living chondrocytes comparable in hydrodynamic size to human articular car-
resulted in cartilage implants. tilage from an age-matched donor (73 years old). The
To investigate whether cell proliferation was re- lower ratio of high:low hydrodynamic–size sulfated proteo-
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1613

Figure 3. Origin of the implant. To determine the relative contribution of the donor (human) and
host (mouse) tissue to the formation of the cartilage implant, consecutive sections were hybridized
with a, human-specific or b, mouse-specific probes recognizing genomic repeats (ALU and
mouse-specific L1, respectively). c, Superimposition of a and b using artificial colors. The
intermingling of human and mouse cells at the left edge of the implant was due to infiltration of
chondrocytes between the muscle fibers, as shown by the toluidine blue staining in d. Bar ⫽ 200 ␮m.

glycans in the extracts from articular cartilage explants may Contribution of the injected cells to the forma-
be related to the lower extraction efficiency (data not tion of the implant. In our in vivo system, the injected
shown). human cells and the surrounding muscle are free to
To better characterize the proteoglycan expres- interact. Chondrogenesis in the mouse muscle could
sion profile of the implants, we performed RT-PCR potentially be induced by the injected chondrocytes, for
analysis using human-specific primers for aggrecan, ver- example, by sustained secretion of growth factors and
sican, biglycan, decorin, and lumican (Figure 2c). Unin- morphogens (33). We explored the relative contribution
jected mouse muscle was used as the negative control. of donor-derived (human) and host-derived (mouse)
Primers for both human and mouse ␤-actin were used to cells to the formation of the implant by the detection of
normalize cDNA from the implant and from the unin- human-specific and mouse-specific genomic repeats by
jected mouse muscle. Confluent primary AHAC were in situ hybridization.
the positive control. Since the implant may have con- Consecutive sections were hybridized separately
tained a considerable number of mouse cells, cDNA with DNA probes specific for human-specific ALU and
from the implant and from primary human chondrocytes mouse-specific L1 genomic repeats. Images were
were normalized for the expression of the housekeeping aligned, converted to artificial colors, and superimposed.
gene ␤2-microglobulin using human-specific primers. As seen in Figure 3, although some chondrocytes infil-
This set of data confirms the expression of cartilage trated the muscle tissue, there was no significant contri-
proteoglycans at the messenger RNA (mRNA) level. bution of mouse-derived cells to the implant.
The absence of versican transcript suggests that most of Loss of in vivo cartilage-forming capacity on
the high molecular weight peak represents sulfate incor- serial passaging. It is well known that expansion of
poration into aggrecan. chondrocytes in monolayer culture results in the loss of
1614 DELL’ACCIO ET AL

Figure 4. Serial expansion of human adult articular chondrocytes, resulting in the loss of their
capacity to form cartilage in vivo. Samples from 2 independent donors (S1 and S2; ages 86 and 28
years, respectively) were expanded. Upon passaging, aliquots of the cell suspension were injected
into nude mice or were used for gene expression analysis. After 2 population doublings,
chondrocytes could still form mature cartilage tissue, as shown by Alcian blue (a and aⴕ) and
Safranin O (c and cⴕ) staining. After 4 population doublings (solid arrow), the loss of cartilage-
forming ability was heralded by the formation of more immature implants, as shown by Alcian blue
(b and bⴕ) and Safranin O (d and dⴕ) staining. Chondrocytes from further passages (open arrow)
did not form any retrievable implant. The loss of cartilage-forming potential was marked by a
down-regulation of ␣1(II) collagen (COL2A1), fibroblast growth factor receptor 3 (FGFR3), and
bone morphogenetic protein 2 (BMP2) mRNA, while the expression of activin receptor–like kinase
1 (ALK-1) mRNA was up-regulated. FI is freshly isolated chondrocytes. Bar ⫽ 200 ␮m.

their phenotype, as measured by type II collagen expres- sion of molecules that are known to play a role in the
sion and proteoglycan synthesis (5). To investigate formation and maintenance of the cartilage phenotype.
whether this phenotypic alteration was associated with a The molecular analysis included the expression of matrix
loss of the capacity to generate stable cartilage in vivo, molecules and ligands, receptors, and intracellular com-
we tested serially passaged human articular chondro- ponents of signaling pathways (TGF␤ superfamily, fibro-
cytes in our in vivo assay. Primary AHAC lost their in blast growth factor [FGF], parathyroid hormone/
vivo cartilage-forming potential after 2–3 passages in parathyroid hormone–related peptide, and hedgehog
vitro (about 4–6 population doublings). In the samples proteins) (19). This analysis resulted in the selection of a
shown in Figure 4, after 4 population duplications, restricted number of molecular markers, the expression
chondrocytes generated an implant that stained poorly of which tightly and reproducibly correlated with the
with Alcian blue and Safranin O. Chondrocytes from capacity of expanded chondrocytes to form stable carti-
later passages failed to generate any retrievable implant. lage in vivo. The results of the molecular analysis are
Molecular markers that predict the in vivo car- summarized in Figure 4.
tilage forming ability of articular chondrocytes indepen- The expression of ␣1(II) collagen (COL2A1) and
dently of donor age. To identify molecular markers that FGF receptor 3 (FGFR-3) correlated positively with our
are predictive of cartilage-forming capacity in our assay, in vivo assay. These markers were consistently down-
we monitored throughout in vitro expansion the expres- regulated when the cartilage-forming potential of ex-
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1615

Figure 5. Markers predict the capacity of adult human articular chondrocytes to form stable
cartilage in vivo, independently of the donor age. Freshly isolated (FI) and serially passaged
chondrocytes from donors of different ages (range 28–86 years) were challenged in our in vivo
assay throughout expansion. Solid arrows mark the passage when a decline of the maturity of the
implant was first detected. Open arrows mark the first passage from which no implant could be
retrieved. COL2A1 ⫽ ␣1(II) collagen; FGFR3 ⫽ fibroblast growth factor receptor 3; BMP2 ⫽
bone morphogenetic protein 2; ALK-1 ⫽ activin receptor–like kinase 1.

panded chondrocytes was reduced and immature carti- plant was retrieved when AHAC were injected into nude
lage implants were retrieved. The expression of these mice in 2 independent experiments each performed in
markers became undetectable when chondrocytes were duplicate. Interestingly the up-regulation of ALK-1 in
unable to organize a retrievable implant. serially passaged chondrocytes could not be reversed to
In contrast to the steep decrease of FGFR-3 and normal by the agarose culture.
COL2A1 mRNA levels, BMP-2 was strongly expressed
in freshly isolated chondrocytes, decreased throughout
DISCUSSION
passaging, and never completely disappeared within the
passages examined. The expression of activin receptor– ACT is a conceptually attractive therapeutic ap-
like kinase 1 (ALK-1) mRNA, as yet reported to be proach to JSD repair (3). The capacity to organize stable
predominantly expressed by endothelial cells (38), ap- cartilage in vivo is an important requisite for the cell
peared to be a negative marker of chondrocyte stability, population used for JSD repair. Many independent
being steadily up-regulated during expansion. factors can interfere with the in vivo cartilage-forming
Under our conditions, this set of molecular mark- capability of the chondrocyte preparation. These include
ers appeared to be predictive of the in vivo capacity to the age of the donor (39) and the variability of the
generate cartilage tissue reproducibly, independently of autologous serum, both of which affect the growth rate
age. Figure 5 shows the molecular profiles of 3 chondro- and, potentially, the phenotypic stability of chondrocytes
cyte populations from donors of different age (24, 28, in vitro. Indeed, it is well known that in vitro expansion
and 86 years old) throughout in vitro expansion. of articular chondrocytes results in the progressive loss
The agarose assay is not sufficient to predict the of phenotypic traits (5). Consequently, the risk that this
potential of expanded chondrocytes to generate stable process of dedifferentiation may result in a reduced
cartilage in vivo. Anchorage-independent growth and capacity to form stable cartilage in vivo is clearly a
rescue of type II collagen expression in anchorage- concern in cartilage engineering. The importance of this
independent culture (5) have been considered to be issue has been underscored by the variability of the
predictive of the phenotypic stability of chondrocytes repair tissue formed after ACT (4). Importantly, the
(3). To validate the agarose assay as a predictor of the formation of either hyaline-like cartilage tissue or disor-
cartilage-forming potential in our assay, we challenged ganized fibrocartilage seems to affect the clinical out-
AHAC at passage 5 (⬃10 population doublings) with come (4) and is likely to depend on the phenotypic
our in vivo assay and with the agarose assay. stability of the implanted chondrocytes.
Passage 5 AHAC were still able to form colonies To pursue a more consistent and reproducible
(Figure 6a) and to recover to the original levels of outcome of ACT, it would be useful to establish para-
COL2A1 and FGFR-3 mRNA (Figure 6b) when cul- meters that would allow us to monitor the capacity of
tured in LTM agarose. However, no cartilaginous im- expanded chondrocytes to form cartilage in vivo. Ideally,
1616 DELL’ACCIO ET AL

into nude mice. The implants consisted of differentiated


hyaline-like cartilage, as evaluated by histology and
confirmed by polarized light microscopy (data not
shown), containing a significant amount of high molec-
ular weight proteoglycans and type II collagen. The
implant was stable for at least 12 weeks, without signs of
vascular invasion or endochondral bone formation, un-
like in fetal swine chondrocytes, where these phenomena
were evident as early as after 3 weeks (data not shown).
This cartilage-forming ability was progressively lost dur-
ing in vitro expansion. Subsequently, we identified a set
of molecular markers predictive of the outcome of our in
vivo assay. Importantly, we have shown that the
anchorage-independent growth and the rescue of
COL2A1 expression in agarose culture are not sufficient
to predict the cartilage-forming ability in our in vivo
model.
We are aware that the nude mouse assay does not
exactly reproduce the same environment of a JSD in a
human joint such as the knee. Experimental evidence is
still lacking that this, or any other assay, can directly
Figure 6. The agarose assay is not sufficient to predict the cartilage-
predict the outcome of ACT. Ideally, studies in large
forming ability of expanded chondrocytes in our in vivo assay. Freshly
isolated (FI) and serially passaged adult human articular chondrocytes animals would represent the optimum system for vali-
were injected into nude mice and tested in the agarose assay. a, dating chondrocyte expansion procedures for ACT. Un-
Although the cells lost their in vivo cartilage tissue–forming ability fortunately, due to significant anatomic and biologic
after the second passage, they could still grow under anchorage- differences between human cartilage and the articular
independent conditions in agarose after passage 5 (⬃10 population
cartilage of other animals typically used (essentially,
doublings). b, The molecular profile of the same chondrocytes
throughout passaging and after agarose culture [A(10)]. Note that rabbit, dog, sheep, and goat), none of the animal models
while the expression of ␣1(II) collagen (COL2A1), fibroblast growth of ACT available today is close enough to the human to
factor receptor 3 (FGFR3), and bone morphogenetic protein 2 be considered truly representative (40). In addition,
(BMP-2) was rescued in the agarose culture system, the up-regulation some spontaneous tissue repair takes place in these
of activin receptor–like kinase 1 (ALK-1) was not reversed. Arrow
animal models, and the actual contribution of the im-
indicates the first passage from which no implant could be retrieved.
planted cells to the repair tissue is not always clear. On
the other hand, studies in humans cannot be properly
controlled for ethical reasons.
these parameters should be independent of the stage of The possibility of addressing these critical issues
differentiation of the cell population, the age of the relies on the development of an adequate animal model
patient, and any variable in the culture system and cell of ACT where every experimental step is controlled.
handling. So far, anchorage-independent growth and the Our nude mouse model represents a well-validated,
rescue of type II collagen expression in LTM agarose stringent, flexible, and tightly controlled screening assay
culture (5) have been used to validate chondrocyte for addressing specific biologic questions in vivo. The
expansion procedures for ACT (3). Their value for the use of costly and time-consuming animal studies could
specific use in cartilage engineering may be limited, be restricted to a further, preclinical validation step for
however, because none of them necessarily predicts the cell-based JSD repair protocols.
capacity of expanded chondrocytes to form stable carti- The use of the nude mouse model in cartilage
lage tissue in vivo. biology was introduced by Lipman et al (41) and has
In the present study, we standardized and vali- become popular over the last few years in tissue engi-
dated an assay in nude mice that assesses the capacity of neering. In a wide array of technical variants and using
AHAC to organize stable cartilage in vivo. Freshly cells from disparate origins, the nude mouse model has
isolated human articular chondrocytes reproducibly been used to test cell–polymer constructs for cartilage
formed cartilage implants when injected intramuscularly engineering (42) and to optimize culture conditions (43).
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1617

The nude mouse system is a powerful assay and the endothelial marker Flk-1 (49) in late-passage chon-
provides in vivo data, but a careful validation is recom- drocytes, as evaluated by RT-PCR (data not shown).
mended for specific experimental settings. The mouse The identification of a molecular profile of the
tissues are sensitive to signaling molecules that are phenotypically stable chondrocyte opens an array of
produced by the implant or present in the matrix. clinical applications, from quality control for cell expan-
Cartilage and/or bone formation derived from the host sion for ACT to the optimization of culture conditions
(mouse) tissue is induced by implantation of a variety of for specific applications in cartilage tissue engineering.
either mesenchyme- or epithelium-derived cells (32,34– The presence of a negative marker such as ALK-1
36,44–46). The type of tissue obtained may also depend mRNA offers the opportunity of an internal control for
on the site of the injection (32). Importantly, species gene expression analysis. Possible quantitative applica-
specificity of the assay has been described. Injections tions of this principle can be real-time quantitative
into nude mice of either rabbit or dog articular chon- RT-PCR and flow cytometry.
drocytes that have been expanded for 14 days resulted in The rescuing/enhancement of the cartilage phe-
hyaline cartilage implants in the former case, and in notype has been documented in a large number of
fibrous tissue in the latter (41). systems such as the culture in agarose or alginate (5,50),
Therefore, for the specific use as a quality control culture on Matrigel (51), and treatment with dihydrocy-
and a tool for investigating JSD repair protocols, it was tochalasin B (52), retinoic acid (53), and members of the
necessary to standardize and validate the nude mouse TGF␤ superfamily (14,54–56). The possibility that some
assay with the use of human adult articular chondro- of these systems also rescue the potential of late-passage
cytes. We have shown that, under our experimental chondrocytes to form cartilage in vivo is very relevant for
cartilage tissue engineering and is being tested in our
conditions, the implants were constituted by the human
laboratory.
injected cells with no significant contribution from the
Our experimental settings do not address the
mouse host. In addition, cell viability, but not cell
question of the homogeneity of the chondrocyte popu-
proliferation, was required.
lation throughout in vitro expansion. Based on our data,
Use of the nude mouse assay requires time and
however, it is likely that the in vivo potential to generate
skills that are not compatible with a routine, consistent,
cartilage in our assay is related to the predominance of
and reproducible quality control analysis for individual
cells expressing the positive markers over cells express-
patients. To circumvent this problem, we have identified
ing the negative ones. If this assumption proves to be
positive and negative molecular markers that predict the true, the development of specific antibodies to extracel-
outcome of the in vivo assay independently of the age of lular domains of FGFR-3 and ALK-1 would offer the
the donor. COL2A1, FGFR-3, and BMP-2 were posi- opportunity of enriching cell populations with stable
tively associated with the stable phenotype, while the chondrocytes by cell sorting techniques. More consistent
up-regulation of ALK-1 marked the loss of the in vivo and homogeneous cell populations for ACT would prob-
cartilage-forming potential. This set of molecular mark- ably ensure more reproducible clinical outcomes.
ers has proven to be more stringent than the agarose
assay in predicting the capacity to form cartilage in vivo.
ACKNOWLEDGMENTS
In addition, the up-regulation of ALK-1 in serially
passaged chondrocytes could not be reversed to normal The DIG-labeled ALU probe was kindly provided by
Dr. K. Satomura (First Department of Oral and Maxillofacial
by the agarose culture.
Surgery, University of Tokushima, Tokushima, Japan). Nor-
We do not address in this study the molecular mal adult human skin fibroblasts were a gift of Dr. S. Tejpar
mechanisms underlying the phenomena described, espe- (Centrum Meiselijke Erfelijkheid, Katholieke Universiteit
cially since, with the exception of type II collagen (47), Leuven, Leuven, Belgium). Thanks are due to people working
the transcriptional regulation of these genes is not in the Mortuary of the University Hospitals of Leuven, Bel-
gium, for providing the cartilage samples. We thank Drs. M.
completely elucidated. In addition, while the role of Moos, T. Thomas, and P. Tylzanowski for many suggestions
BMP-2 (23) and FGFR-3 (48) in cartilage development and for critically reviewing the manuscript.
and homeostasis has been extensively studied, ALK-1 is
known to predominantly play a role in angiogenesis in REFERENCES
early development (38). ALK-1 up-regulation, in our
1. Luyten FP. A scientific basis for the biologic regeneration of
model, was unlikely to be due to endothelial contamina- synovial joints. Oral Surg Oral Med Oral Pathol Oral Radiol
tion, since it was not associated with detectable levels of Endod 1997;83:167–9.
1618 DELL’ACCIO ET AL

2. American College of Rheumatology Subcommittee on Osteoar- 23. Luyten FP, Yu YM, Yanagishita M, Vukicevic S, Hammonds RG,
thritis Guidelines. Recommendations for the medical management Reddi AH. Natural bovine osteogenin and recombinant human
of osteoarthritis of the hip and knee: 2000 update. Arthritis bone morphogenetic protein-2B are equipotent in the mainte-
Rheum 2000;43:1905–15. nance of proteoglycans in bovine articular cartilage explant cul-
3. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, tures. J Biol Chem 1992;267:3691–5.
Peterson L. Treatment of deep cartilage defects in the knee with 24. Kuznetsov SA, Krebsbach PH, Satomura K, Kerr J, Riminucci M,
autologous chondrocyte transplantation. N Engl J Med 1994;331: Benayahu D, et al. Single-colony derived strains of human marrow
889–95. stromal fibroblasts from bone after transplantation in vivo. J Bone
4. Peterson L, Minas T, Brittberg M, Nilsson A, Sjogren-Jansson E, Miner Res 1997;12:1335–47.
Lindahl A. Two- to 9-year outcome after autologous chondrocyte 25. Hatano H, Tokunaga K, Ogose A, Hotta T, Yamagiwa H, Hayami
transplantation of the knee. Clin Orthop 2000;374:212–34. T, et al. Origin of bone-forming cells in human osteosarcomas
5. Benya PD, Shaffer JD. Dedifferentiated chondrocytes reexpress transplanted into nude mice—which cells produce bone, human or
the differentiated collagen phenotype when cultured in agarose mouse? J Pathol 1998;185:204–11.
gels. Cell 1982;30:215–24. 26. Szuts V, Mollers U, Bittner K, Schurmann G, Muratoglu S, Deak
6. Butnariu-Ephrat M, Robinson D, Mendes DG, Halperin N, Nevo F, et al. Terminal differentiation of chondrocytes is arrested at
Z. Resurfacing of goat articular cartilage by chondrocytes derived distinct stages identified by their expression repertoire of marker
from bone marrow. Clin Orthop 1996;330:234–43. genes. Matrix Biol 1998;17:435–48.
7. Wakitani S, Goto T, Pineda SJ, Young RG, Mansour JM, Caplan 27. Atsumi T, Miwa Y, Kimata K, Ikawa Y. A chondrogenic cell line
AI, et al. Mesenchymal cell-based repair of large, full-thickness derived from a differentiating culture of AT805 teratocarcinoma
defects of articular cartilage. J Bone Joint Surg Am 1994;76: cells. Cell Differ Dev 1990;30:109–16.
579–92. 28. Grigoriadis AE, Heersche JN, Aubin JE. Differentiation of mus-
8. De Bari C, Dell’Accio F, Luyten FP. Human periosteum-derived cle, fat, cartilage, and bone from progenitor cells present in a
cells maintain phenotypic stability and chondrogenic potential bone-derived clonal cell population: effect of dexamethasone.
throughout expansion regardless of donor age. Arthritis Rheum J Cell Biol 1988;106:2139–51.
2001;44:85–95. 29. Lau WF, Tertinegg I, Heersche JN. Effects of retinoic acid on
9. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, cartilage differentiation in a chondrogenic cell line. Teratology
Mosca JD, et al. Multilineage potential of adult human mesenchy- 1993;47:555–63.
mal stem cells. Science 1999;284:143–7. 30. Bernier SM, Desjardins J, Sullivan AK, Goltzman D. Establish-
10. Minas T, Nehrer S. Current concepts in the treatment of articular ment of an osseous cell line from fetal rat calvaria using an
cartilage defects. Orthopedics 1997;20:525–38. immunocytolytic method of cell selection: characterization of the
11. Gilbert JE. Current treatment options for the restoration of cell line and of derived clones. J Cell Physiol 1990;145:274–85.
articular cartilage. Am J Knee Surg 1998;11:42–6. 31. Urist MR. Bone formation by autoinduction. Science 1965;150:
12. Francis-West PH, Abdelfattah A, Chen P, Allen C, Parish J, 893–9.
Ladher R, et al. Mechanisms of GDF-5 action during skeletal 32. Thyberg J, Moskalewski S. Bone formation in cartilage produced
development. Development 1999;126:1305–15. by transplanted epiphyseal chondrocytes. Cell Tissue Res 1979;
13. Tsumaki N, Tanaka K, Arikawa-Hirasawa E, Nakase T, Kimura T, 204:77–94.
Thomas JT, et al. Role of CDMP-1 in skeletal morphogenesis: 33. Chang SC, Hoang B, Thomas JT, Vukicevic S, Luyten FP, Ryba
promotion of mesenchymal cell recruitment and chondrocyte NJ, et al. Cartilage-derived morphogenetic proteins: new members
differentiation. J Cell Biol 1999;144:161–73. of the transforming growth factor-beta superfamily predominantly
14. Erlacher L, Ng C-K, Ullrich R, Krieger S, Luyten FP. Presence of expressed in long bones during human embryonic development.
cartilage-derived morphogenetic proteins in articular cartilage and J Biol Chem 1994;269:28227–34.
enhancement of matrix replacement in vitro. Arthritis Rheum 34. Boyan BD, Swain LD, Schwartz Z, Ramirez V, Carnes DLJ.
1998;41:263–73. Epithelial cell lines that induce bone formation in vivo produce
15. Polinkovsky A, Robin NH, Thomas JT, Irons M, Lynn A, Good- alkaline phosphatase-enriched matrix vesicles in culture. Clin
man FR, et al. Mutations in CDMP1 cause autosomal dominant Orthop 1992;277:266–76.
brachydactyly type C [letter]. Nat Genet 1997;17:18–9. 35. Van Noorden CJ, Jonges GN, Vogels IM, Hoeben KA, van Urk B,
16. Thomas JT, Kilpatrick MW, Lin K, Erlacher L, Lembessis P, Costa Everts V. Ectopic mineralized cartilage formation in human
T, et al. Disruption of human limb morphogenesis by a dominant undifferentiated pancreatic adenocarcinoma explants grown in
negative mutation in CDMP1. Nat Genet 1997;17:58–64. nude mice. Calcif Tissue Int 1995;56:145–53.
17. Wang S, Krinks M, Lin K, Luyten FP, Moos MJ. Frzb, a secreted 36. Ostrowski K, Wlodarski K, Aden D. Heterotopic chondrogenesis
protein expressed in the Spemann organizer, binds and inhibits and osteogenesis induced by transformed cells: use of nude mice as
Wnt-8. Cell 1997;88:757–66. a model system. Somatic Cell Genet 1975;1:391–5.
18. Lin K, Thomas JT, McBride OW, Luyten FP. Assignment of a new 37. Schmidt MB, Mow VC, Chun LE, Eyre DR. Effects of proteogly-
TGF-beta superfamily member, human cartilage-derived morpho- can extraction on the tensile behavior of articular cartilage.
genetic protein-1, to chromosome 20q11.2. Genomics 1996;34: J Orthop Res 1990;8:353–63.
150–1. 38. Oh SP, Seki T, Goss KA, Imamura T, Yi Y, Donahoe PK, et al.
19. Dell’Accio F, De Bari C, Luyten FP. Molecular basis of joint Activin receptor-like kinase 1 modulates transforming growth
development. Jpn J Rheumatol 1999;9:17–29. factor-beta 1 signaling in the regulation of angiogenesis. Proc Natl
20. Boskey AL, Doty SB, Stiner D, Binderman I. Viable cells are a Acad Sci U S A 2000;97:2626–31.
requirement for in vitro cartilage calcification. Calcif Tissue Int 39. Adolphe M, Ronot X, Jaffray P, Hecquet C, Fontagne J, Lechat P.
1996;58:177–85. Effects of donor’s age on growth kinetics of rabbit articular
21. Matsumoto T, Iwasaki K, Sugihara H. Effects of radiation on chondrocytes in culture. Mech Ageing Dev 1983;23:191–8.
chondrocytes in culture. Bone 1994;15:97–100. 40. Hunziker EB. Biologic repair of articular cartilage: defect models
22. Cornelissen M, Thierens H, de Ridder L. Effects of ionizing in experimental animals and matrix requirements. Clin Orthop
radiation on the size distribution of proteoglycan aggregates 1999;367 Suppl:S135–46.
synthesized by chondrocytes in agarose. Scanning Microsc 1993;7: 41. Lipman JM, McDevitt CA, Sokoloff L. Xenografts of articular
1263–7. chondrocytes in the nude mouse. Calcif Tissue Int 1983;35:767–72.
MOLECULAR MARKERS OF STABLE CHONDROCYTES 1619

42. Freed LE, Marquis JC, Nohria A, Emmanual J, Mikos AG, Langer 50. Bonaventure J, Kadhom N, Cohen-Solal L, Ng KH, Bourguignon
R. Neocartilage formation in vitro and in vivo using cells cultured J, Lasselin C, et al. Reexpression of cartilage-specific genes by
on synthetic biodegradable polymers. J Biomed Mater Res 1993; dedifferentiated human articular chondrocytes cultured in alginate
27:11–23. beads. Exp Cell Res 1994;212:97–104.
43. Ohlsson C, Nilsson A, Isaksson OG, Lindahl A. Effect of growth 51. Basic N, Basic V, Bulic K, Grgic M, Kleinman HK, Luyten FP, et
hormone and insulin-like growth factor-I on DNA synthesis and al. TGF-beta and basement membrane Matrigel stimulate the
matrix production in rat epiphyseal chondrocytes in monolayer chondrogenic phenotype in osteoblastic cells derived from fetal rat
culture. J Endocrinol 1992;133:291–300. calvaria. J Bone Miner Res 1996;11:384–91.
44. Tokunaga K, Ogose A, Endo N, Nomura S, Takahashi HE. 52. Benya PD, Brown PD, Padilla SR. Microfilament modification by
Human osteosarcoma (OST) induces mouse reactive bone forma-
dihydrocytochalasin B causes retinoic acid-modulated chondro-
tion in xenograft system. Bone 1996;19:447–54.
cytes to reexpress the differentiated collagen phenotype without a
45. Quarto R, Campanile G, Cancedda R, Dozin B. Modulation of
commitment, proliferation, and differentiation of chondrogenic change in shape. J Cell Biol 1988;106:161–70.
cells in defined culture medium. Endocrinology 1997;138:4966–76. 53. Benya PD, Padilla SR. Modulation of the rabbit chondrocyte
46. Wright GCJ, Miller F, Sokoloff L. Induction of bone xenografts of phenotype by retinoic acid terminates type II collagen synthesis
rabbit growth plate chondrocytes in the nude mouse. Calcif Tissue without inducing type I collagen: the modulated phenotype differs
Int 1985;37:250–6. from that produced by subculture. Dev Biol 1986;118:296–305.
47. Zhou G, Lefebvre V, Zhang Z, Eberspaecher H, de Crombrugghe 54. Chen P, Vukicevic S, Sampath TK, Luyten FP. Osteogenic pro-
B. Three high mobility group-like sequences within a 48-base pair tein-1 promotes growth and maturation of chick sternal chondro-
enhancer of the Col2a1 gene are required for cartilage-specific cytes in serum-free cultures. J Cell Sci 1995;108:105–14.
expression in vivo. J Biol Chem 1998;273:14989–97. 55. Harrison ETJ, Luyten FP, Reddi AH. Osteogenin promotes
48. Rousseau F, Bonaventure J, Legeai-Mallet L, Pelet A, Rozet JM, reexpression of cartilage phenotype by dedifferentiated articular
Maroteaux P, et al. Mutations in the gene encoding fibroblast chondrocytes in serum-free medium. Exp Cell Res 1991;192:
growth factor receptor-3 in achondroplasia. Nature 1994;371: 340–5.
252–4. 56. Harrison ETJ, Luyten FP, Reddi AH. Transforming growth factor-
49. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, beta: its effect on phenotype reexpression by dedifferentiated
Breitman ML, et al. Failure of blood-island formation and vascu- chondrocytes in the presence and absence of osteogenin. In Vitro
logenesis in Flk-1-deficient mice. Nature 1995;376:62–6. Cell Dev Biol 1992;28A:445–8.

You might also like