Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

TISSUE ENGINEERING

Volume 12, Number 9, 2006


# Mary Ann Liebert, Inc.

Chondrogenic Differentiation of Human Embryonic


Stem Cell–Derived Cells in Arginine-Glycine-Aspartate–
Modified Hydrogels

NATHANIEL S. HWANG, B.S., SHYNI VARGHESE, Ph.D., ZIJUN ZHANG, M.D.,


and JENNIFER ELISSEEFF, Ph.D.

ABSTRACT

Human embryonic stem cells (hESCs) have the potential to self-renew and generate multiple cell types,
producing critical building blocks for tissue engineering and regenerative medicine applications. Here,
we describe the efficient derivation and chondrogenic differentiation of mesenchymal-like cells
from hESCs. These cells exhibit mesenchymal stem cell (MSC) surface markers, including CD29,
CD44, CD105, and platelet-derived growth factor receptor-a. Under appropriate growth conditions, the
hESC-derived cells proliferated without phenotypic changes and maintained MSC surface markers.
The chondrogenic capacity of the cells was studied in pellet culture and after encapsulation in
poly(ethylene glycol)-diacrylate (PEGDA) hydrogels with exogenous extracellular proteins or arginine-
glycine-aspartate (RGD)-modified PEGDA hydrogels. The hESC-derived cells exhibited growth factor–
dependent matrix production in pellet culture but did not produce tissue characteristic of cartilage
morphology. In PEGDA hydrogels containing exogenous hyaluronic acid or type I collagen, no significant
cell growth or matrix production was observed. In contrast, when these cells were encapsulated in RGD-
modified poly(ethylene glycol)hydrogels, neocartilage with basophilic extracellular matrix deposition was
observed within 3 weeks of culture, producing cartilage-specific gene up-regulation and extracellular
matrix production. Our results indicate that precursor cells characteristic of a MSC population can be
cultured from differentiating hESCs through embryoid bodies, thus holding great promise for a po-
tentially unlimited source of cells for cartilage tissue engineering.

INTRODUCTION limited and decreases with age.2 Deleterious phenotypic


changes in the chondrocyte hinder the in vitro expansion of a

B ECAUSE OF THE INTRINSIC BIOLOGY OF CARTILAGE TISSUES,


such as limited blood supply and lack of self-
regeneration capacity, treatment of cartilage lesion remains
clinically useful number of chondrocytes from a small
biopsy specimen, which may itself be diseased. Primary
isolated chondrocytes have the tendency to de-differentiate
an intractable problem.1 Cell-based therapies and tissue in ex vivo culture.3
engineering may provide a solution for cartilage re- Human embryonic stem cells (hESCs), derived from the
construction. However, limited proliferative capacity of dif- inner cell mass of the blastocyst, may have significant po-
ferentiated chondrocytes and bone marrow-derived tential to affect cartilage tissue engineering.4 These cells are
mesenchymal stem cells (MSCs) pose a major challenge to being studied to understand early development and to for-
providing adequate cell numbers for transplantation therapy. mulate cell- and tissue-regeneration therapies.5 The obvious
The self-renewal and proliferative capacity of MSCs is advantage of using embryonic stem cells (ESCs) for cartilage

Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.

2695
2696 HWANG ET AL.

regeneration is that they are immortal and could potentially plemented with 10% fetal bovine serum (Hyclone, Logan,
provide unlimited numbers of differentiated chondrocytes UT), 2 mM L-glutamine (Gibco), 100 U/mL penicillin, and
and chondro-progenitor cells for transplantation.6, 7 Re- 100 mg/mL streptomycin (Gibco). Human MSCs (Cambrex,
cently, ESCs were cultured on 3-dimensional (3D) scaffolds East Rutherford, NJ) were expanded in MSC growth med-
for tissue engineering.8–10 However, application of ESCs to ium up to P5 and morphologically compared with ESC-
tissue engineering still faces the difficulty of appropriately derived cells.
differentiating the stem cells to the desired lineage in a
controlled and homogenous fashion.
Identification of appropriate biomaterials that support
Pellet culture
cellular attachment, proliferation, and—most importantly in For chondrogenic pellet cultures, 3105 cells (P5) were
the case of hESCs—differentiation is also critical for tissue collected in 1-mL Eppendorf tubes and centrifuged at
engineering and cellular therapy. The extracellular micro- 150g for 5 min. Serum-free chondrogenic medium was pre-
environment plays a significant role in controlling cellular pared with DMEM (Gibco) containing 2 mM L-glutamine;
behavior.11–13 Currently, a number of scaffolding materials, 100 nM dexamethasone; 50 mg/mL ascorbic acid phosphate
including synthetic and naturally derived biomaterials, have (Wako, Neuss, Germany); 1 mM sodium pyruvate; 40 mg/
been used in tissue engineering.14–16 Scaffold biomaterials mL proline; 1% insulin, transferrin, and selenious acid
have been incorporated with specific peptides or naturally (Collaborative Biomedical Products, Bedford, MA); and
occurring biological proteins to influence cell function.17–22 10 ng/mL transforming growth factor (TGF)-b1 or 25 ng/
Hydrogels are 3D networks of hydrophilic polymers that im- mL bone morphogenetic protein (BMP)-2 (Research Di-
bibe a large quantity of water, as well as biological fluids.23 agnostics, Inc., Flanders, NJ).
Poly(ethylene glycol) (PEG)-based hydrogels are ideal for
cartilage tissue engineering scaffolds because of their water
content, elasticity, biocompatibility, and ability to permit Photoencapsulation
diffusion of nutrients and bioactive molecules.15,24 hESC-derived cells (P5*P7) were encapsulated into 4
Recent studies have indicated osteogenic and chondro- types of hydrogels. The primary hydrogel solution was pre-
genic lineage differentiation of hESCs.25–27 However, pro- pared by mixing 10% (w/v) poly(ethylene glycol)-diacrylate
ducing discreet, well-developed cartilaginous structures on a (PEGDA; Nektar, Huntsville, AL) in sterile phosphate-
3D artificial scaffold remains a challenge. Here, we present buffered saline (PBS) with penicillin (100 U/mL) and strep-
culture conditions for the derivation of mesenchymal-like tomycin (100 mg/mL) (Gibco). Hydrogels with exogenous
cells from embryoid bodies (EBs) and demonstrate specific ECM components were prepared by mixing 50/50 volume of
cell–extracellular matrix (ECM) interactions, in the form of 20% (w/v) PEGDA solution with type I collagen solution
arginine-glycine-aspartate (RGD)-integrin binding, are re- (0.2% w/v in PBS) or hyaluronic acid (HA) solution (5 mg/
quired for enhanced chondrogenic commitment and carti- mL in PBS). RGD-modified PEG-based hydrogels were
laginous tissue formation. Given limited availability of cells prepared according to procedures previously described.20–22
for cartilage tissue–replacement therapy, this work shows Briefly, tyrosine-arginine-glycine-aspartate-serine (YRGDS)
promise for potential use of hESCs for cartilage tissue– was added to acryloyl-PEG-N-hydroxysuccinimide (3400 MW;
engineering applications. Nektar) in 50 mM TRIS buffer (pH 8.2) for 2 h at room tem-
perature. The product was lyophilized, and a final concentra-
tion of polymer solution containing 2.5 mM RGD was prepared
by mixing PEGDA and YRGDS-PEG-zcrylate in sterile PBS.
MATERIALS AND METHODS The hESC-derived cells were gently mixed with the polymer
solution at a concentration of 2107 cells/mL. The photo-in-
hESC culture and EB formation itiator, Igracure 2959, was added to the cell–hydrogel solu-
The BG02 cell line was obtained from Bresagen (Athens, tion and mixed thoroughly to make a final concentration of
GA) and cultured according to the manufacturer’s instruc- 0.05% (w/v). Photopolymerization of 100-mL cell–polymer–
tions.28 For EB formation, ESC cultures were dissociated photo-initiator suspension was conducted with 365-nm light
into small clumps by incubating at 378C for 30 min with (4.5 mW/cm2) for 5 min. The constructs were then cultured
1 mg/mL collagenase IV (Gibco, Gaithersburg, MD). The at 378C with 5% carbon dioxide in 2.5 mL chondrogenic
small ESC clumps were pelleted, resuspended in hESC differentiation medium containing TGF-b1 (10 ng/mL) for
medium without basic fibroblast growth factor (bFGF), and 3 weeks.
cultured in Petri dishes for 10 days with a medium change
every other day. The EBs were transferred to gelatin-coated
(0.1% w/v) plates. Upon 70% confluence, the cells were
FACS analysis
further subcultured at an initial cell density of 2104 cells/ hMSCs (P5) and hESC-derived cells (4 passages) were
cm2 for 4 passages in MSC growth medium consisting of harvested and resuspended to approximately 3105 cells in
Dulbecco’s modified Eagle medium (DMEM, Gibco) sup- 50 mL PBS containing 0.1% bovine serum albumin. Cell
CHONDROGENIC DIFFERENTIATION OF hES-DERIVED CELLS 2697

samples were separately labeled with fluorescein iso- with 5% normal goat serum in PBS for 30 min and in-
thiocyanate (FITC)-conjugated rat anti-human antigen CD45 cubated with rabbit polyclonal antibodies against types I, II,
(Becton Dickinson, San Jose, CA), phycoerythrin-conjugated and X collagen (Research Diagnostics, Inc.) with 1:100,
rat anti-human CD 29, CD45, CD73, and platelet-derived 1:00, and 1:40 dilutions, respectively. Sections were in-
growth factor receptor (PDGFR)-a (Becton Dickinson) on cubated with FITC- or Texas Red–conjugated goat anti-
ice for 30 min. Cells were also incubated with purified mouse rabbit secondary antibodies (all 1:100 dilutions) for 1 h
anti-human CD105 (Becton Dickinson) and then stained with ( Jackson ImmunoResearch Laboratory, West Grove, PA).
FITC-conjugated goat anti-mouse IgM ( Jackson Im-
munoResearch Laboratories, West Grove, PA). At least Reverse transcriptase polymerase
15,000 events were collected from each run of flow cyto-
chain reaction
metry. Data were analyzed using the FACSCalibur cytometer
and CellQuest software (Becton Dickinson). Total ribonucleic acid was extracted with Trizol and
reverse-transcribed into complementary DNA using Super-
Script First-Strand Synthesis System (Invitrogen, Carlsbad,
Swelling ratio CA). Reverse transcriptase polymerase chain reactions (RT-
Acellular control hydrogels and hydrogels with exogen- PCRs) were performed for the following genes: aggrecan (F-
ous type I collagen (0.1% w/v) or HA (2.5 mg/mL) were tgaggagggctggaacaagtacc, R-ggaggtggtaattgcagggaaca, 608C;
prepared (n ¼ 4). The wet weight was measured for swel- 305bp), Type I collagen (F-tgacgagaccaagaactg, R-ccatc-
ling after incubating the hydrogels up to 21 days in PBS at caaaccactgaaacc, 608C; 600bp), Type II collagen (F-
378C. The swelling ratio was measured by comparing the tttcccaggtcaagatggtc, R- cttcagcacctgtctcacca, 608C 377bp),
wet weight and lyophilized dried weight of hydrogels. and link protein (F-gctctgtgcaatatcccatc, R-cccacttttgcaatct-
gagc, 608C; 232bp), with b-actin (F-tggcaccacaccttcta-
caatgagc, R-gcacagcttctccttaatgtc) as a reference.
Biochemical assays
Constructs were lyophilized and digested in papain ac- Statistical analysis
cording to Kim et al.,29 with the exception that digestion
buffer was adjusted to pH 6.3. Lyophilized constructs were Data are expressed as mean  standard deviation (SD).
crushed with a tissue grinder (pellet pestle mixer, Kimble/ Statistical significance was determined using single-factor
Kontes, Vineland, NJ), and then incubated with 1 mL 0.2- analysis of variance with p < 0.05 or p < 0.01.
mM papain digestion buffer (100 mM phosphate buffer,
10 mM cysteine, 10 mM ethylenediaminetetraacetic acid,
pH 6.3) containing 125 mg/mL papain type III (Worthington RESULTS
Biomedical, Lakewood, NJ) at 608C for 16 h. Deoxyr-
ibonucleic acid (DNA) assay was performed as previously Derivation of mesenchymal-like cells from hESCs
described.30 Proteoglycan content was determined accord- BG02 cells were induced to differentiate through embry-
ing to chondroitin sulfate using dimethylmethylene blue oid body formation followed by culture on gelatin-coated
spectrophotometric assay at A525, as previously described.31 plates. Initially, the cultures contained a heterogeneous po-
Chondroitin sulfate C (Sigma-Aldrich, St. Louis, MO) dis- pulation of cells that became more homogeneous with sub-
solved in de-ionized water was used to generate the standard sequent passages (4 passages). Morphologically, the hESC-
curve. The hydroxyproline content of insoluble collagen was derived cells have a similar morphology to fibroblast and
determined according to the method of Stegemann and mesenchymal-like cells (Fig. 1). Flow cytometry analysis
Stalder.32 After a portion of the papain-digested sample was confirmed that the hESC-derived cells expressed hMSC
acid hydrolyzed, it was neutralized and oxidized with surface markers (Fig. 2), including CD29, CD44, CD105,
chloramine-T/Ehrlich solutions, using 0.1 as the ratio of and PDGFR-a. However, the hMSC marker CD73 was not
hydroxyproline to collagen. present. CD45, a negative marker for hMSC, was not ex-
pressed in the hESC-derived cells.
Histology and immunostaining
Growth factor response of hESC-derived
Pellets and hydrogel constructs were fixed in 4% paraf-
ormaldehyde, dehydrated in serial isopropanol dilutions,
cells in pellet culture
and paraffin embedded. Hydrogels and pellets were cut into To evaluate the chondrogenic differentiation capacity of
5-mm sections and stained with hematoxylin and eosin, the cells, hESC-derived cells were induced to differentiate in
Safranin-O/fast green, and Masson’s trichrome. For im- a standard pellet culture system with subsequent exposure to
munostaining, the hydrogel sections were de-paraffinized BMP-2 or TGF-b1. After 3 weeks of culture, TGF-b1
with xylenes and hydrated with serial concentration of treatment induced a greater increase in weight and matrix
100%, 95%, and 90% ethanol. Sections were then blocked production per cell than BMP-2 treatment (Fig. 3A, B).
2698 HWANG ET AL.

FIG. 1. Inverted light microscopy images of human mesenchymal stem cells (hMSCs) and Human embryonic stem cell (hESC)-
derived mesenchymal cells. hESC-derived mesenchymal cells (A) were morphologically comparable with hMSCs (B). hMSCs plated at
an initial cell density of 5103 cells/cm2 were expanded up to 5 passages. Embryoid bodies (Ebs) were formed from hESCs using liquid
suspension methods for 10 days and plated onto gelatin-coated tissue culture plates. After 15 days of monolayer culture, EBs were
enzymatically dissociated and subsequently passaged at initial cell density of 20104 cells/cm2 and expanded up to 4 passages.

Pellets exposed to TGF-b1 for 2 weeks and subsequently maintained viability throughout 3 weeks of culture in
exposed to BMP-2 for 1 week were equivalent in size and chondrogenic medium containing TGF-b1 (data not shown).
matrix production rate as pellets treated with TGF-b1 for all Cell numbers measured according to DNA content of the
3 weeks of culture. Even though the cells in pellet culture constructs and matrix glycosaminoglycan (GAG) content
responded to TGF-b1 by increasing their size, chondrogenic indicated that individual components of the cartilage ECM,
capacity of hESC-derived mesenchymal cells was limited HA, or collagen I, did not influence the growth or GAG
in pellet culture. Chondrogenic differentiation, evaluated synthesis (Fig. 4B, C). hESC-derived mesenchymal cells
using Safranin-O staining, indicates lack of cartilage-related exposed to type I collagen and HA resulted in 20% and
proteoglycans (Fig. 3C). 43% less GAG accumulation, respectively, in the hydrogels
than cells encapsulated in the control PEGDA hydrogel
without any ECM components. However, the environment
Requirement of scaffold and ECM
created in the RGD-modified hydrogel significantly influ-
microenvironment in chondrogenic
enced the behavior of hESC-derived mesenchymal cells
differentiation of hESC-derived cells (Fig. 4C), resulting in 7% w/v of GAG accumulation, more
We subsequently evaluated whether addition of ECM than a 200% increase in GAG content than in the control
components known to induce chondrogenic responses PEGDA hydrogel constructs.
would promote the differentiation of hESC-derived me- Histochemical and immunostaining analysis for pro-
senchymal cell into the chondrogenic lineage. Exogenous teoglycans and collagen types I, II, and X performed after
matrix proteins or specific peptide sequences known to 21 days demonstrated significant differences between
induce differentiation can be simply introduced into hy- scaffoldings used to differentiate hESC-derived mesench-
drogels using covalent or noncovalent mechanisms.20,33 ymal cells. Safranin-O staining indicated that RGD-
Microenvironments for hESCs-derived mesenchymal cell modified hydrogels induced the hESC-derived cells to
differentiation were created in PEGDA photo-polymerizing produce basophilic ECM deposition containing proteogly-
hydrogels by adding exogenous type I collagen (0.1 mg/ cans characteristic of neocartilage, whereas no significant
mL) and HA (2.4 mg/mL) and covalently incorporating Safranin-O staining was observed in the other hydrogel
RGD peptides (2.5 mM) into PEGDA hydrogels (Fig. 4A). constructs (Fig. 6). Cells in the RGD-modified hydrogels
Amount of exogenous type I collagen and HA introduced had a consistent morphology, with a spherical shape typical
into the hydrogel was decided based on similar swelling of chondrocytes, and produced intense and well-defined
ratios of hydrogels. Control acellular hydrogels with exo- staining with type II collagen antibody in the periphery of
genous HA and collagen show that these ECM components the cells. Proliferating cells were present in lacunae, typical
are retained in the hydrogels for the duration of the ex- of cartilaginous tissue. Cells in the PEGDA hydrogel pro-
periments (Fig. 5). hESC-derived mesenchymal cells were duced minimal staining of cartilage-specific proteins,
encapsulated in photo-polymerizing hydrogels and cultured whereas exogenous type I collagen or HA resulted in less-
for 3 weeks in chondrogenic medium containing TGF-b1. intense staining of collagen types II and I and minimal
Cells survived the hydrogel encapsulation process and detection of type I collagen in PEGDA–collagen gels. Type
CHONDROGENIC DIFFERENTIATION OF hES-DERIVED CELLS 2699

FIG. 2. (A) Flow cytometry analysis of surface markers on human embryonic stem cell–derived mesenchymal cells. (B) Flow cytometry
analysis of surface markers on adult-derived human mesenchymal stem cells. PDGFR, platelet-derived growth factor receptor.

X collagen was weakly detected in all of the hydrogel thesis activities, were observed in cells in RGD-modified
constructs. hydrogels than in control hydrogels (Fig. 8). RGD signaling
was not sufficient to induce chondrogenic response alone
without exogenous TGF-b1 (data not shown). Reduced Sa-
RGD-modified PEG-based hydrogels induced
franin-O staining was observed in the center of the hydrogel,
adhesive and TGF-b1-dependent chondrogenic
where TGF-b1 diffusion was limited (Fig. 8C, F).
differentiation To validate differentiation, RT-PCR was performed to
After 7 days of culture, filopodia-like structures anchoring evaluate gene expression of chondrogenic markers (Fig. 9).
the cells to the scaffold was evidence of interaction between The time-course analysis of cartilage-related gene expression
integrin receptors of differentiating hESC-derived cells and further validated that the chondrogenic tissue lineage pro-
RGD-containing PEG hydrogels. Cells in RGD-modified gression of hESC-derived mesenchymal cells in RGD-
hydrogels were more than twice the size of cells in non-RGD modified PEG hydrogels was greater than that of control
hydrogels (Fig. 7). After 3 weeks of culture, significantly PEGDA hydrogels. At day 7, hESC-derived mesenchymal
greater rates of GAG production, as well as collagen syn- cells in PEGDA and RGD-modified PEG did not express
2700 HWANG ET AL.

FIG. 3. Chondrogenic differentiation capacities of human embryonic stem cell mesenchymal cells were limited in pellet culture.
2105 cells were collected in 1-mL Eppendorf tubes and sedimented at 150g for 5 min. After 3 weeks of culture, initial treatment of
transforming growth factor (TGF)-b1 induced a greater increase in weight and matrix/cells than bone morphogenetic protein (BMP)-2-
treated pellets (A, B). However, chondrogenic differentiation, evaluated using Safranin-O staining, indicated no cartilaginous tissues (C)
(1) BMP-2 (25 ng/mL, 2 weeks) followed by TGF-b1 (10 ng/mL) treatment (1 week). (2) TGF-b1 treatment (2 weeks) followed by
BMP-2 (1 week) (3) TGF-b1 (3 weeks) *p < 0.05, **p < 0.01). DNA, deoxyribonucleic acid. Color images available online at
www.liebertpub.com/ten.

chondrocyte-specific genes, such as type II collagen, RGD-modified hydrogels was significantly upregulated.
aggrecan, and link proteins. Type I collagen, which is ex- Aggrecan and link protein expression were also upregulated
pressed in early mesenchymal cells, was expressed through- in the RGD-modified hydrogels, confirming the immuno-
out the culture. At day 21, type II collagen expression in staining, as well as the biochemical analysis.

FIG. 4. Gross image of an acellular poly(ethylene oxide)-diacrylate (PEGDA) photo-polymerizing hydrogel (A). Chondrogenic
differentiation capacities of embryonic stem cell–derived mesenchymal cells were investigated by adding extracellular matrix com-
ponents to hydrogels, as well as modifying the hydrogels with arginine-glycine-aspartate (RGD) peptides. Glycosaminoglycan (GAG)
production significantly increased in RGD-modified hydrogels (B). GAG amount was quantified using dimethylmethylene blue assay
and normalized to the dry weight of the construct for comparison. The deoxyribonucleic acid (DNA) content of the constructs was
normalized by dry weight for comparison (C) 1. Poly(ethylene glycol)-diacrylate (PEGDA). 2. PEGDA-collagen type I (0.1% w/v); 3.
PEGDA–hyaluronic acid (2.5 mg/mL). 4. PEGDA-RGD (2.5 mM). (* p < 0.05, ** p < 0.01).
CHONDROGENIC DIFFERENTIATION OF hES-DERIVED CELLS 2701

FIG. 5. (A) Swelling ratio of acellular poly(ethylene glycol)-diacrylate (PEGDA), PEGDA-collagen (Col), and PEGDA–hyaluronic
acid (HA) hydrogels. Comparable swelling ratio was observed in different hydrogels for the duration of the experiment, indicating that
exogenous extracellular matrix components are retained in the gel after 3 weeks of culture (n ¼ 4). (B) Acellular hydrogels containing
exogenous type I collagen (0.1% w/v) were incubated in phosphate-buffered saline up to 21 days, and collagen content was quantified
by measuring hydroxyproline content and normalized to the dry weight (n ¼ 4).

DISCUSSION wide differentiation potential.4,9 It has been proposed that


ESC differentiation through the formation of EBs mimics
hESCs, derived from the inner cell mass of the blas- the early developmental stages during embryogenesis, there-
tocyst, are a promising cell source for tissue regeneration by providing a window of opportunity to detect and pos-
applications because of their strong proliferative ability and sibly isolate tissue-restricted progenitor cells.34 Recent

FIG. 6. Addition of type I collagen (0.1% w/v) and hyaluronic acid (HA) (2.5 mg/mL) into poly(ethylene glycol)-diacrylate (PEGDA)
hydrogel resulted in no significant Safranin-O stained area, whereas covalent incorporation of arginine-glycine-aspartate (RGD) into
PEGDA resulted in chondrogenic differentiation, as evidenced by positive Safranin-O staining, and immunostaining for types I, II, and
X collagen. (A, E, I, M) PEGDA; (B, F, J, N) PEGDA-collagen type I (0.1% w/v); (C, G, K, O) PEGDA-HA (2.5 mg/mL); (D, H, L, P)
PEGDA-RGD (2.5 mm). Color images available online at www.liebertpub.com/ten.
2702 HWANG ET AL.

culture have been frequently used for efficient chondrogen-


esis of MSCs or chondro-progenitor cells.37 N-cadherin-
dependent cell–cell interactions leading to mesenchymal
condensation have been found to be distinct markers of
chondrogenesis.38 TGF-b and BMP act distinctly in terms
of their chondrogenic potential during mesenchymal con-
densation and limb cartilage development, TGF-b having an
early positive role in promoting chondrogenesis and BMP
having an early negative and late positive role during
chondrogenesis.7,39 Our results indicate that the hESC-de-
rived cells had the greatest response to TGF-b1. Initial ex-
posure of pellets to BMP for 2 weeks followed by 1 week of
TGF-b1 resulted in a smaller increase in matrix production
than with pellets that were treated for 2 weeks with TGF-b1
followed by 1 week of BMP-2 or pellets exposed to TGF-b1
for all 3 weeks.
A significant number of cells expressed PDGFR-a, a
marker indicating cells that have chondrogenic potential
upon exposure to TGF-b1.7 Even though growth factor–
dependent matrix production was observed, histological
analysis indicated that the chondrogenic capacity of these
cells was limited, perhaps requiring matrix-stimulated
FIG. 7. Morphological analysis of cells encapsulated in poly chondrogenic response. Safranin-O staining for sulfated
(ethylene glycol)-diacrylate (PEGDA) hydrogels (A) and glycosaminoglycans showed no indication of cartilaginous
arginine-glycine-aspartate (RGD)-modified poly(ethylene glycol) matrix deposition. RT-PCR confirmed an absence of chon-
(PEG) hydrogel (B) after 7 days of encapsulation. Cell-matrix drocyte gene expression (data not shown). Lack of chon-
interaction is evident in RGD-modified PEG hydrogels (arrow), drogenesis could be related to the short culture time of 3
resulting in larger cell diameter (C). Bar ¼ 15 mm (**p < 0.01).
weeks, which may be insufficient to induce a chondrogenic
Color images available online at www.liebertpub.com/ten.
response of these cells. Also, the lack of CD73 expression,
which it has been shown is expressed in adult-derived MSCs,
isolation of mesenchymal-like cells exhibiting CD73 from may have contributed to the lack in chondrogenesis in pellet
ESCs provides evidence of lineage-restricted cells from culture. Alternatively, lack of bioactive signals in the cell-
ESCs. However, this method requires long term co-culture matrix microenvironment may have resulted in failure to
with murine OP9 cells, which might pose difficulties when initiate chondrogenic differentiation.
applying to therapeutic applications, and only a small number Various cues from the surrounding microenvironment
of the co-cultured hESCs exhibited CD73.27 A large generally control differentiation of stem cells.11,40 The me-
number of fibroblastic cells with multipotent differentiation chanisms involved in ECM-dependent phenotypic cell dif-
capacity have been derived from differentiating EBs and ferentiation have been increasingly studied in recent years.41
pushed toward adipogenesis and osteogenesis, but these Briefly, ECM proteins, through specific peptide motifs, in-
cells showed limited chondrogenic potential.35 In this study, teract with integrins, a family of heterodimeric transmem-
we demonstrate the derivation methods for mesenchymal- brane protein.42,43 This interaction changes the integrin
like cells from differentiating embryoid bodies and their conformation, resulting in the activation of intracellular
therapeutic potential for cartilage tissue engineering on an pathways that are involved in a number of cell responses. To
artificial 3D scaffold system. create a microenvironment for specific matrix–cell interac-
Our results indicate that hESC-derived mesenchymal tions in 3D culture, we encapsulated the hESC-derived
cells exhibit the mesodermal progenitor cell marker, mesenchymal cells in hydrogels with different ECM com-
PDGFR-a, as well as MSC markers CD29, CD44, and ponents known to induce chondrogenic responses in chondro-
CD105. hESC-derived mesenchymal cells also exhibited progenitor cells. Microenvironments created by exogenous
fibroblastic phenotype that was comparable with that of type I collagen and HA have been shown to be beneficial for
human MSCs, but they did not express CD73. Tissue en- viability and chondrogenic differentiation of MSCs.44–47
gineering of human cartilage is a complex process, because Hydrogels modified with an RGD-containing peptide se-
the functional development of chondro-progenitors and their quence have been shown to promote cell multiplication and
differentiation from chondrocytes require that regulatory facilitate cell adhesion and spreading.19,20,48 We introduced
signals be temporally and spatially displayed.36 The 3D exogenous proteins such as type I collagen (0.1% w/v) and
cellular interactions in the form of pellet or micro-mass HA (2.5 mg/mL), as well as covalently linking RGD
CHONDROGENIC DIFFERENTIATION OF hES-DERIVED CELLS 2703

FIG. 8. Basophilic extracellular matrix (ECM) deposition characteristic of neocartilage from human embryonic stem cell–derived
mesenchymal cells was promoted using arginine-glycine-aspartate (RGD) and transforming growth factor (TGF)-b1. (A) Hematoxylin
and eosin (H&E) staining and (D) Safranin-O staining of cells encapsulated in poly(ethylene glycol)-diacrylate (PEGDA). No significant
matrix production and significant Safranin-O staining were observed in PEGDA alone. Cells encapsulated in RGD-modified poly
(ethylene glycol) showed TGF-b1 dependent chondrogenic response. Regions of hydrogel near the periphery showed strong matrix
production (B, E), whereas toward the center of the hydrogel, where diffusion of TGF-b1 is limited, less matrix was produced around
the cell (arrow) (C, F). RGD incorporation enhanced the secretion and accumulation of sulfated glycosaminoglycans (GAGs) and
collagen. GAG amount was quantified using dimethylmethylene blue assay and normalized to the total deoxyribonucleic acid (DNA) of
the construct (G). Collagen was quantified by measuring hydroxyproline content and normalized to the total DNA of the construct for
comparison (H). (*p < 0.05, **p < 0.01). Color images available online at www.liebertpub.com/ten.

(2.5 mM) motifs to the hydrogel macromers to create a simplified mimics of natural ECM, lacking the essential
specific cell-matrix microenvironment for differentiation. natural temporal and spatial complexity, this RGD-mod-
Immunostaining for specific chondrocyte matrix proteins ified synthetic biomaterial was sufficient to promote
and Safranin-O staining indicated the varying degree of chondrocyte-specific differentiation and morphogenesis and
matrix production induced by the microenvironments. Cells cartilage tissue development. RT-PCR analysis indicated
encapsulated with exogenous type I collagen, which has that RGD–cell interactions induced the cells to express full
non-RGD integrin-binding sites, and HA, which interacts chondrocyte markers, including type II collagen and link
with CD44, resulted in an inhibitory effect on GAG protein. Within 3 weeks of culture, basophilic ECM de-
synthesis and accumulation. The optimal differentiation, as position was observed in RGD-modified PEG hydrogels.
well as matrix production, was observed in the RGD- Immunostaining for type I, II, and X collagen and Safranin-
modified hydrogels, resulting in 7% w/v of GAG accu- O staining for negatively charged proteoglycans indi-
mulation in 3 weeks of culture, which is higher than the cated that these ECM proteins are mainly localized around
3.5% w/v GAG value reported by Williams et al. with the cells, having islands of ECM and showing limited
MSCs cultured in similar conditions for 6 weeks.49 Cells diffusivity within the hydrogel. Controlling ECM deposi-
encapsulated in RGD-modified hydrogel were larger than tion and tissue formation by incorporating biodegradable
cells encapsulated in PEGDA hydrogels and formed ex- components into the hydrogel, as well as having longer
tensions to interact with the surrounding environment. incubation period, would improve the quality of tissue
Although RGD-modified PEG hydrogels represent over- formation.
2704 HWANG ET AL.

may serve to support further efforts toward characterizing


cell–ECM and cell growth factor roles in controlling and
eliciting differentiation.

ACKNOWLEDGMENTS
This study was supported by the Johns Hopkins Uni-
versity ( JHU)-Technion Joint Program and the Whitaker
Foundation. The authors are grateful to Ms. Yoojin An
(University of Pennsylvania) and Ms. Angela Ferran ( JHU)
for their critical review and technical assistance.

REFERENCES
1. Buckwalter, J.A., and Mankin, H.J. Articular cartilage: de-
generation and osteoarthritis, repair, regeneration, and trans-
FIG. 9. Reverse transcriptase polymerase chain reaction analy- plantation. Instr Course Lect 47, 487, 1998.
sis indicated distinct chondrogenic response of human embryonic 2. Fehrer, C., and Lepperdinger, G. Mesenchymal stem cell
stem cell–derived mesenchymal cells in arginine-glycine-aspartate aging. Exp Gerontol 41, 1, 2005.
(RGD)-modified poly(ethylene glycol) (PEG) hydrogel. At day 7, 3. Schnabel, M., Marlovitz, S., Eckhoff, G., Fichtel I., Gotzen,
no chondrocyte specific genes were detected in either culture L., Vecsei, V., and Schlegel, J. Dedifferentiation-associated
system (Columns 1 and 3). After 21 days of culture, cells en- changes in morphology and gene expression in primary hu-
capsulated in poly(ethylene glycol)-diacrylate (PEGDA) hydrogel man articular chondrocytes in cell culture. Osteoarthr Cartil
minimally expressed aggrecan and type II collagen, with no de- 10, 62, 2002.
tectable level of expression of link protein (Column 3). Cells 4. Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz,
encapsulated in RGD-modified PEG hydrogels resulted in signi- M.A., Swiergiel, J.J., Marshall, V.S., and Jones, J.M. Em-
ficant upregulation of aggrecan, type II collagen, and link protein bryonic stem cell lines derived from human blastocysts.
(Column 4). 1. D7-PEGDA, 2. D21-PEGDA, 3. D7-PEGDA-RGD, Science 282, 1145, 1998.
4. D21-PEGDA-RGD. 5. Levenberg, S., and Langer, R. Advances in tissue engineering.
Curr Top Dev Biol 61, 113, 2004.
6. Hegert, C., Kramer, J., Hargus, G., Muller, J., Guan, K.,
Wobus, A.M., Miller, P.K., and Rohwedel, J. Differentiation
Recent evidence indicates that RGD interaction with av plasticity of chondrocytes derived from mouse embryonic
and b1 integrin subunits enhanced TGF-b1 secretion, and stem cells. J Cell Sci 115, 4617, 2002.
RGD-dependent integrin activation has been linked to 7. Nakayama, N., Duryea, D., Manoukian, R., Chow, G., and
modulation of TGF-b1 actions.50 RGD binding and acti- Han, C.Y. Macroscopic cartilage formation with embryonic
vation of integrins increase the tyrosine kinase-dependent stem-cell-derived mesodermal progenitor cells. J Cell Sci
116, 2015, 2003.
phosphorylation of human mesangial cell proteins, as well
8. Liu, H., and Roy, K. Biomimetic three-dimensional cultures
as integrin link kinase activity, which in turn converge significantly increase hematopoietic differentiation efficacy of
with TGF-b1 signaling pathways to regulate multiple embryonic stem cells. Tissue Eng 11, 319, 2005.
transcription factors.51 Indeed, RGD interaction alone 9. Levenberg, S., Huang, N.F., Lavik, E., Rogers, A.B., Itskovitz-
was not sufficient to induce chondrogenic response of Eldor, J., and Langer, R. Differentiation of human embryonic
hESC-derived mesenchymal cells, and we observed limited stem cells on three-dimensional polymer scaffolds. Proc Natl
matrix production of hESC-derived mesenchymal cells Acad Sci U S A 100, 12741, 2003.
in the center of the hydrogels due to limited diffusion 10. Levenberg, S., Burdick, J.A., Kraehenbuehl, T., and Langer,
TGF-b1. R. Neurotrophin-induced differentiation of human embryonic
Our data show that hESCs contain precursors to a MSC stem cells on three-dimensional polymeric scaffolds. Tissue
population and that RGD-specific cell–matrix interactions, Eng 11, 506, 2005.
11. Streuli, C. Extracellular matrix remodelling and cellular dif-
which in turn may approximate the cell environment of
ferentiation. Curr Opin Cell Biol 11, 635, 1999.
condensing mesenchyme in the developing limb in vivo, 12. Cukierman, E., Pankov, R., Stevens, D.R., and Yamada, K.M.
can promote the differentiation of hESC-derived cells into Taking cell-matrix adhesions to the third dimension. Science
chondrocytes and create homogeneous structures of tissue 294, 5547, 2001.
morphologically comparable with cartilage. Such micro- 13. Battista, S., Guamieri, D., Borselli, C., Zeppetelli, S., Bor-
environments for cell differentiation may be useful in tar- zacchiello, A., Mayol, L., Gerbasio, D., Keene, D.R., Am-
geting stem cells for tissue-specific differentiation and brosio, L., and Netti, P.A. The effect of matrix composition of
CHONDROGENIC DIFFERENTIATION OF hES-DERIVED CELLS 2705

3D constructs on embryonic stem cell differentiation. Bio- 32. Stegemann, H. and Stalder, K. Determination of hydro-
materials 26, 6194, 2005. xyproline. Clin Chim Acta 18, 267, 1967.
14. Kim, B.S., and Mooney, D.J. Development of biocompatible 33. Lum, L.Y., Cher, N.L., Williams, C.G., and Elisseeff, J. An
synthetic extracellular matrices for tissue engineering. Trends extracellular matrix extract for tissue-engineered cartilage.
Biotechnol 16, 224, 1998. IEEE Eng Med Biol Mag 22, 71, 2003.
15. Elisseeff, J., McIntosh, W., Anseth, L., Riley, S., Ragan, P., 34. Dvash, T., and Benvenisty, N. Human embryonic stem cells
and Langer, R. Photoencapsulation of chondrocytes in poly as a model for early human development. Best Pract Res Clin
(ethylene oxide)-based semi-interpenetrating networks. J Obstet Gynaecol 18, 929, 2004.
Biomed Mater Res 51, 164, 2000. 35. Xu, C., Jiang, J., Sottile, V., McWhir, J., Lebkowski, J., and
16. Langer, R. Biomaterials in drug delivery and tissue en- Carpenter, M.K. Immortalized fibroblast-like cells derived
gineering: one laboratory’s experience. Acc Chem Res 33, 94, from human embryonic stem cells support undifferentiated
2000. cell growth. Stem Cells 22, 972, 2004.
17. Hubbell, J.A. Biomaterials in tissue engineering. Bio- 36. DeLise, A.M., Fischer, L., and Tuan, R.S. Cellular interac-
technology (NY) 13, 565, 1995. tions and signaling in cartilage development. Osteoarthr Cartil
18. Shin, H., Jo, S., and Mikos, A.G. Biomimetic materials for 8, 309, 2000.
tissue engineering. Biomaterials 24, 4353, 2003. 37. Mackay, A.M., Beck, S.C., Murphy, J.M., Barry, F.P.,
19. Alsberg, E., Anderson, K.W., Albeiruti, A., Rowley, J.A., and Chickester, C.O., and Pittenger, M.F. Chondrogenic differ-
Mooney, D.J. Engineering growing tissues. Proc Natl Acad entiation of cultured human mesenchymal stem cells from
Sci U S A 99, 12025, 2002. marrow. Tissue Eng 4, 415, 1998.
20. Yang, F., Williams, C.G., Wang, D.A., Lee, H., Manson, 38. Tuli, R., Tuli, S., Nadi, S., Huang, X., Manner, P.A., Hozack,
P.N., and Elisseeff, J. The effect of incorporating RGD ad- W.J., Danielson, K.G., Hall, D.J., and Tuan, R.S. Trans-
hesive peptide in polyethylene glycol diacrylate hydrogel on forming growth factor-beta-mediated chondrogenesis of hu-
osteogenesis of bone marrow stromal cells. Biomaterials 26, man mesenchymal progenitor cells involves N-cadherin and
5991, 2005. mitogen-activated protein kinase and Wnt signaling cross-
21. Burdick, J.A., and Anseth, K.S. Photoencapsulation of os- talk. J Biol Chem 278, 41227, 2003.
teoblasts in injectable RGD-modified PEG hydrogels for bone 39. Yoon, B.S. and Lyons, K.M. Multiple functions of BMPs in
tissue engineering. Biomaterials 23, 4315, 2002. chondrogenesis. J Cell Biochem 93, 93, 2004.
22. DeLong, S.A., Gobin, A.S., and West, J.L. Covalent im- 40. Spradling, A., Drummond-Barbosa, D., and Kai, T. Stem cells
mobilization of RGDS on hydrogel surfaces to direct cell find their niche. Nature 414, 98, 2001.
alignment and migration. J Control Release 109, 139, 2005. 41. Grashoff, C., Aszodi, A., Sakai, T., Hunziker, E.B., and
23. Hoffman, A.S. Hydrogels for biomedical applications. Adv Fassler, R. Integrin-linked kinase regulates chondrocyte shape
Drug Deliv Rev 54, 3, 2002. and proliferation. EMBO Rep 4, 428, 2003.
24. Bryant, S.J., Nuttelman, C.R., and Anseth, K.S. The effects of 42. Martin, K.H., Slack, J.K., Boerner, S.A., Martin, C.C., and
crosslinking density on cartilage formation in photo- Parsons, J.T. Integrin connections map: to infinity and be-
crosslinkable hydrogels. Biomed Sci Instrum 35, 309, 1999. yond. Science 296, 1652, 2002.
25. Bielby, R.C., Boccaccini, A.R., Polak, J.M., and Buttery, L.D. 43. Giancotti, F.G., and Ruoslahti, E. Integrin signaling. Science
In vitro differentiation and in vivo mineralization of osteo- 285, 1028, 1999.
genic cells derived from human embryonic stem cells. Tissue 44. Meinel, L., Hofmann, S., Karageorgiou, V., Zichner, L.,
Eng 10, 1518, 2004. Langer, R., Kaplan, D., and Vunjak-Novakovic, G. En-
26. Sottile, V., Thomson, A., and McWhir, J. In vitro osteogenic gineering cartilage-like tissue using human mesenchymal
differentiation of human ES cells. Cloning Stem Cells 5, 149, stem cells and silk protein scaffolds. Biotechnol Bioeng 88,
2003. 379, 2004.
27. Barberi, T., Willis, L.M., Socci, N.D., and Studer, L. Deri- 45. Batorsky, A., Liao, J., Lund, A.W., Plopper, G.E., and Ste-
vation of multipotent mesenchymal precursors from human gemann, J.P. Encapsulation of adult human mesenchymal
embryonic stem cells. PLoS Med 2, 161, 2005. stem cells within collagen-agarose microenvironments. Bio-
28. Zeng, X., Mirua, T., Luo, Y., Bhattacharya, B., Condie, B., technol Bioeng 92, 492, 2005.
Chen, J., Ginis, I., Lyons, I., Mejido, J., Puri, R.K., Rao, M.S., 46. Hegewald, A.A., Ringe, J., Bartel, J., Kruger, I., Notter, M.,
and Freed, W.J. Properties of pluripotent human embryonic Barnewitz, D., Kaps, C., and Sittinger, M. Hyaluronic acid
stem cells BG01 and BG02. Stem Cells 22, 292, 2004. and autologous synovial fluid induce chondrogenic differ-
29. Kim, M.S., Hwang, N.S., Lee, J., Kim, T.K., Leong, K., entiation of equine mesenchymal stem cells: a preliminary
Shamblott, M.J., Gearhart, J., and Elisseeff, J. Musculoske- study. Tissue Cell 36, 431, 2004.
letal differentiation of cells derived from human embryonic 47. Miralles, G., Baudoin, R., Dumas, D., Baptiste, D., Hubert, P.,
germ cells. Stem Cells 23, 113, 2005. Stoltz, J.F., Dellacherie, E., Mainard, D., Netter, P., Netter, P.,
30. Kim, Y.J., Sah, R.L., Doong, J.Y., and Grodzinsky, A.J. and Payan, E. Sodium alginate sponges with or without so-
Fluorometric assay of DNA in cartilage explants using dium hyaluronate: in vitro engineering of cartilage. J Biomed
Hoechst 33258. Anal Biochem 174, 168, 1988. Mater Res 57, 268, 2001.
31. Farndale, R.W., Buttle, D.J., and Barrett, A.J. Improved 48. Mann, B.K., Tsai, A.T., Scott-Burden, T., and West,
quantitation and discrimination of sulphated glycosaminogly- J.L. Modification of surfaces with cell adhesion peptides
cans by use of dimethylmethylene blue. Biochim Biophys Acta alters extracellular matrix deposition. Biomaterials 20, 2281,
883, 173, 1986. 1999.
2706 HWANG ET AL.

49. Williams, C.G., Kim, T.K., Toboas, A., Malik, A., Manson, Address reprint requests to:
P., and Elisseeff, J. In vitro chondrogenesis of bone marrow- Jennifer Elisseeff, Ph.D.
derived mesenchymal stem cells in a photopolymerizing hy- Department of Biomedical Engineering
drogel. Tissue Eng 9, 679, 2003. Johns Hopkins University School of Medicine
50. Ortega-Velazquez, R., Diez-Marques, M.L., Ruiz-Torres, Clark Hall 106
M.P., Gonzalez-Rubio, M., Rodriguez-Puyol, M., and Ro-
3400 North Charles Street
driguez-Puyol, D. Arg-Gly-Asp-Ser (RGDS) peptide stimu-
Baltimore, MD 21218
lates transforming growth factor beta1 transcription and
secretion through integrin activation. Faseb J 17, 1529, 2003.
51. Dedhar, S., Williams, B., and Hannigan, G. Integrin-linked E-mail: jhe@bme.jhu.edu
kinase (ILK): a regulator of integrin and growth-factor sig-
nalling. Trends Cell Biol 9, 319, 1999.

You might also like