干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 255879|回复: 236
go

Directing Stem Cell Differentiation into the Chondrogenic Lineage In Vitro [复制链接]

Rank: 7Rank: 7Rank: 7

积分
威望
0  
包包
483  
楼主
发表于 2009-3-5 10:35 |只看该作者 |倒序浏览 |打印
Key Words. Chondrogenesis ? Chondrocytes ? Stem cells ? Differentiation ? In vitro+ }! C0 P5 [/ a, G0 L, G

8 a$ M6 x% M  b) }Correspondence: Tong Cao, D.D.S., Ph.D., Faculty of Dentistry, National University of Singapore, 5 Lower Kent Ridge Road, Singapore 119074. Telephone: 65-6874-4630, ext. 1612; Fax: 65-6774-5701; e-mail: dencaot@nus.edu.sg& ]4 ~6 }" |- A& e3 W4 \3 q
) t5 u8 x- Z+ M4 w+ i# f
ABSTRACT
! q) m+ W, K8 E1 k9 i
* d! n3 b/ S% \8 {7 tTop
6 [% V+ P8 @5 r) ?* y! L' U7 ?4 e0 b0 ?3 q; H- u# n
Abstract
- t% e! R3 [# O+ L1 D( b( G* [* N. ~
Introduction" R2 y4 z3 w+ i3 g# ~0 z. ~
9 L& k3 o3 a3 O* j
Adult Stem Cells versus...
4 K7 s! E2 z+ z, O; _4 S, {4 T+ u' r- M- O
Development of Defined Culture...
: I! z8 ~7 D" H0 G% e* |2 k, j% T- L7 o% S( H% c6 i- f
Coculture and Cell-Conditioned...3 m, I4 V( l# s7 [& E8 X
% m8 S2 |% j! }2 c1 H5 ~
Directing Chondrogenic...
, [- L. i) I6 e) C7 I+ I  l; B% w) |4 o
4 y: [' F8 q! Y2 C& O5 L8 BFuture Perspectives
. Q- i+ B) ]+ [7 ^9 F8 J0 o6 Q2 [6 L. ^- k
Concluding Remarks
% a1 U1 K' W8 M% z
. _! R" G9 Z" [  U2 kReferneces% p# I- d& g9 H( t: z3 }' C
$ Y3 J2 }2 T1 A. M
A major area in regenerative medicine is the application of stem cells in cartilage tissue engineering and reconstructive surgery. This requires well-defined and efficient protocols for directing the differentiation of stem cells into the chondrogenic lineage, followed by their selective purification and proliferation in vitro. The development of such protocols would reduce the likelihood of spontaneous differentiation of stem cells into divergent lineages upon transplantation, as well as reduce the risk of teratoma formation in the case of embryonic stem cells. Additionally, such protocols could provide useful in vitro models for studying chondrogenesis and cartilaginous tissue biology. The development of pharmacokinetic and cytotoxicity/genotoxicity screening tests for cartilage-related biomaterials and drugs could also utilize protocols developed for the chondrogenic differentiation of stem cells. Hence, this review critically examines the various strategies that could be used to direct the differentiation of stem cells into the chondrogenic lineage in vitro.$ W6 h) r' t& u5 F

/ ~, f% x3 A: u8 b4 m  B" L( t- vINTRODUCTION
% x  c$ n: f- {' i& q5 y
4 y- u" h2 X' {1 xTop! }' x% Y  T3 Q$ H6 v
1 ?2 g5 S$ I; V. t; n) }
Abstract
4 u% {' C# V0 I' @0 R
! g9 z, \- k  ^5 d% O- |! PIntroduction
+ ]; d2 x3 j& E0 A
- f$ b% H  C) a* t: S- hAdult Stem Cells versus...
# z0 p  h) U, Y$ C- _( ]- T2 k: C6 a3 S( J+ o6 f+ k' o
Development of Defined Culture...
. q( x% I* ?, Q4 J) o
: ^' Q' A2 W3 \& K/ xCoculture and Cell-Conditioned...
2 Z3 K$ P5 G% V
7 r$ o, j! i" l) ^9 MDirecting Chondrogenic...
1 @8 H' B9 C" W7 V* A
, ?0 F! n( O  z4 }- j8 \# @Future Perspectives
- U8 d# U) y% F9 K. [7 @: F
! w+ G6 G8 O3 P7 u' O) S3 qConcluding Remarks
5 x+ X* g- [! B" a: c! O5 U! E/ h' _% f) s
Referneces
, A* F( i$ A' |1 W" r1 Z) l7 N" G: x6 [+ g( G% W& }$ q1 }
Traumatic injury and age-related degenerative diseases associated with cartilage are major health problems worldwide . Despite recent advances in surgical and nonsurgical interventions, the treatment of cartilage lesions remains an intractable problem . This is attributable in large part to the intrinsic biology of cartilaginous tissue, which limits its capacity to self-regenerate. Because cartilage is nonvascularized and noninnervated, the normal mechanism of tissue repair involving humoral factors and recruitment of stem/progenitor cells to the site of damage does not apply . Moreover, the low cell density  within cartilaginous tissue reduces the likelihood of local chondrocytes contributing to self-regeneration .8 y! I7 }. d5 U/ `3 v* M1 u

* I" g# v# e* j4 b- g# dCell transplantation therapy has shown much promise in augmenting the inadequate repair mechanisms within damaged cartilage . At present, the only U.S. Food and Drug Administration–approved cell-based therapy (CarticelTM ) for cartilage repair in the human clinical model uses mature autologous chondrocytes. However, the limited proliferative capacity of differentiated chondrocytes could pose a major problem in providing adequate cell numbers for transplantation therapy . Furthermore, it was also reported that the proliferative potential of autologous chondrocytes decreases with patient age . This would in turn pose a significant challenge in the treatment of age-related chondrodegenerative diseases (i.e., osteoarthritis). Hence, this review examines adult and embryonic stem (ES) cells as alternative sources of donor cells  for the treatment of cartilage lesions.. |; H! V  i) b# t9 s

4 y, q& p/ n: NStem cells can be distinguished from progenitor cells by their capacity for both self-renewal and multilineage differentiation, whereas progenitor cells are capable only of multi-lineage differentiation without self-renewal . It is this capacity for self-renewal that makes stem cells particularly useful for transplantation medicine, because this in theory could provide an unlimited supply of donor material. Moreover, stem cells and their differentiated derivatives possess much higher proliferative and regenerative potential compared with mature differentiated somatic cells. This in turn is more likely to guarantee adequate regeneration and cell turnover at the transplantation site for an extended period of time, possibly a lifetime.
: \$ V# u' D6 Q$ t  c9 l4 E
1 V3 f. [  d, a- [In attempting to use stem cells for cartilage repair , it is imperative to develop well-defined and efficient protocols for directing stem cell differentiation into the chondrogenic lineage in vitro. This is followed by selective purification and proliferation of the differentiated subpopulation of stem cells before transplantation within the recipient. This will reduce the likelihood of spontaneous differentiation of stem cells into multiple divergent lineages at the transplantation site  other than the chondrogenic lineage. In the case of ES cells, this is particularly important for avoiding teratoma formation  within the transplant recipient. The transplantation of stem cells that are well-differentiated into the chondrogenic lineage is also likely to result in higher engraftment efficiency and better integration within recipient cartilaginous tissues. Moreover, cartilage tissue engineering requires differentiated chondrogenic progenitors or chondrocytes, rather than undifferentiated stem cells, for seeding and attachment onto artificially synthesized matrices.# d! t; k# x) A/ }/ n9 x
6 K" A( }" B; J; U, i
Besides human clinical therapy, the development of such protocols would also provide useful in vitro models for studying chondrogenesis and cartilage development. It is extremely difficult to elucidate the molecular mechanisms and signaling pathways that regulate chondrogenesis in vivo within live animal models. Efficient protocols for directing the chondrogenic differentiation of stem cells in vitro will therefore provide a model that is more amenable to molecular characterization and genetic manipulation. An added advantage is that such protocols may also facilitate the genetic manipulation of stem cells for the delivery of recombinant genes/proteins in cartilage regeneration therapy . The development of pharmacokinetic and cytoxicity/genotoxicity screening tests for cartilage-related biomaterials/drugs may also utilize protocols for the chondrogenic differentiation of human stem cells in vitro. Compared with laboratory testing on animal cell/tissue cultures or live animals, such screening tests based directly on human-derived cells and tissues will be much more clinically relevant, timely, and accurate, as well as more cost-effective. Moreover, the ethical issues surrounding the use of live animals for pharmacokinetic and cytotoxicity/genotoxicity testing will be circumvented through the development of such screening tests.
; }) [5 P' P, s) c7 z& [# l. s
Hence, the purpose of this review is to critically examine the various strategies that could be used to direct the differentiation of stem cells into the chondrogenic lineage in vitro.2 I  P) g* i8 A/ Z8 Q/ V2 X- l' S

* c1 x9 g0 M5 S5 F" G7 O$ `ADULT STEM CELLS VERSUS EMBRYONIC STEM CELLS FOR CARTILAGE REPAIR AND TISSUE ENGINEERING
5 J. U' u' p0 V
! x# a' ~1 y# R' l5 K; l: E* VTop8 L+ a) [& T$ C3 N
5 X9 b0 }5 ?1 R
Abstract. M3 [6 J5 N/ `4 N+ `- E7 t6 y; d
" t  C' f0 C2 C" ]
Introduction
1 e1 T! m, G6 s: W
4 X: |( ^2 l: ]/ N( m' F: mAdult Stem Cells versus...8 g% f2 M0 x* W5 M& h

' |) k2 l8 i2 e4 ]4 RDevelopment of Defined Culture..." ]7 b4 ~% d! e$ `" G! `
) @5 j  M8 P9 s1 h/ c; i) z( L, D
Coculture and Cell-Conditioned...
# |* q; w' ^/ |* _* V; E8 L  K) @# l* l
Directing Chondrogenic...
; l( x$ T/ E& R  X: w0 v" W+ j2 l  B/ I- o) z6 l
Future Perspectives
4 j% w( f9 [7 w
2 [% t& q6 m3 b5 y1 @% zConcluding Remarks, Q0 K/ l: `- g  _1 o1 m/ S+ `

/ G' x* C6 p0 U; XReferneces$ F/ G1 y& I- ~8 s: c
# n& B2 x$ u' \
Stem cells for cartilage repair can be derived from two major sources: ES cells derived from the inner cell mass of blasto-cysts-stage embryos  and mesenchymal stem cells (MSCs) derived from the bone marrow . Additionally, adult stem cells derived from other tissue types, such as the endothelial , epithelial , adipogenic , myogenic , and hematopoietic  lineages, have also been reported to be capable of transdifferentiating into the chondrogenic lineage. However, at this point in time, adult stem cell plasticity and transdifferentiation are highly controversial issues  due to evidence of cell fusion and heterokaryon formation . In any case, evidence from the scientific literature suggests that adult stem cell plasticity and transdifferentiation are relatively rare and sporadic events . Hence, this review will focus only on ES cells  and bone marrow–derived MSCs  for chondrogenic differentiation.  d) z3 }5 V. T1 l; k
, R( ~$ S. a, X( D, A
The most obvious advantage of using ES cells instead of MSCs for cartilage regeneration is that ES cells are immortal and could potentially provide an unlimited supply of differentiated chondrocytes and chondroprogenitor cells for transplantation. In contrast, the self-renewal and proliferative capacity of MSCs is very much limited and seems to decrease with age . This would obviously limit their usefulness in autologous cell transplantation therapy for the treatment of age-related degenerative diseases of cartilage such as osteoarthritis. The putative MSCs from bone marrow is in fact a highly heterogenous population, with only a limited proportion of cells being capable of differentiation into the chondrogenic lineage. If the capacity for continuous self-renewal is taken strictly as the defining criteria for stem cells, then an extremely low proportion (perhaps approximately 1 in 100,000) of bone marrow stromal cells can be considered true stem cells . This would pose a major challenge in the development of isolation and purification protocols. Additionally, MSCs may also contain more genetic abnormalities than ES cells, caused by exposure to metabolic toxins and errors in DNA replication accumulated during the course of a lifetime .$ ]0 k9 a% y& n, d1 ?6 q6 v

) k( Y; M. W& Y2 |* ^In attempting to use ES cells for cartilage repair, the major challenge is to overcome immunological rejection from the transplant recipient unless an isogenic source of ES cells is derived from therapeutic cloning. Although this has recently been achieved in the human model , this is still a remote option, given the technical difficulties of such an approach . A more viable alternative is to create a bank of ES cells with different major histocompatibilty complex (MHC) genotypes for matching with the transplant recipient. It could also be possible to downregulate the antigenicity of ES cells through suppression of MHC gene expression. In contrast, autologous MSCs derived from the patient’s bone marrow would not face any immunological barriers in transplantation. Another major problem is the risk of teratoma formation by ES cells upon transplantation in situ , which is not the case for MSCs. Moreover, it is much easier to direct MSCs into the chondrogenic lineage compared with ES cells. At present, all reported studies on the chondrogenic differentiation of ES cells were based solely on animal models , with no successful differentiation yet being accomplished with human-derived ES cells.- Z4 `* X: x+ t

! X2 ~  h! a# q- X, G) \9 W+ P; N3 jDEVELOPMENT OF DEFINED CULTURE MILIEU FOR DIRECTING THE CHONDROGENIC DIFFERENTIATION OF STEM CELLS IN VITRO! }" x! g, ]0 D& N: }' C8 F
' g+ `5 \+ z: Z6 C7 A  d% l
Top
, \& Z; ?8 R2 O2 N6 Y9 Z# O, T; c! |* e4 \( w* s; {5 \
Abstract
% o1 ]8 l* V' @& [2 y% d: `$ n% j, g" O! {
Introduction) ^! K4 O: X) j1 L5 c
9 D& x% y; D+ E. Z& ]9 F. z
Adult Stem Cells versus...
3 V! F; r* `: Z) k# _5 Q) |! W% x6 c# u
Development of Defined Culture...' I$ V4 p/ ?% O! v% H. O9 z

7 n( _) b- k, y/ O/ W/ PCoculture and Cell-Conditioned...( V# H- g& A# }* K; i4 T) o

& Q' L$ ~3 D7 \% gDirecting Chondrogenic...
( |) R: T4 }, F) y% G
5 o1 w3 T( `- f& _# UFuture Perspectives* e- o# B' R9 ^
( k; M& q% |2 Z# c1 _3 Q5 l
Concluding Remarks
* O% q' _9 k" V0 J# {% O" _6 G! s% A
Referneces# O4 w$ R3 @6 L! a

$ r# o. J* Q5 A8 |6 ~  ]" w. JAdvantages of Defined Culture Milieu# B4 m, i2 n, y1 L5 I

7 v4 L) h- c7 J9 P; C$ u2 nFor clinical applications of stem cell transplantation therapy, it is imperative that in vitro culture protocols should be devoid of animal or human products to avoid potential contamination with pathogens. The avoidance of products of animal or human origin would also reduce variability within the culture milieu and provide a more stringent level of quality control. Moreover, supplemented animal or human proteins may adhere onto the surface of cultured stem cells, which could possibly enhance their antigenicity upon transplantation. Hence, the ideal culture milieu for promoting the chondrogenic differentiation of stem cells in vitro should be chemically defined and either be serum-free or use synthetic serum replacements , with the possible supplementation of specific recombinant cytokines and growth factors (Table 1) if so required.
& }( k3 r" l% j9 r
5 U& @4 }) a0 N( y, M" LTable 1. Cytokines and growth factors that promote chondrogenesis
5 O+ Z4 c5 \" `: v" c7 M8 A- c& e. n( r5 d& k: \1 w
Serum-Free Culture Conditions for Promoting Chondrogenic Differentiation# v, i6 i/ H6 C: U2 e, {
$ ?" B, v% _$ w1 `& q
The major challenge in attempting to culture stem cells under serum-free conditions is that cells generally tend to have a lower mitotic index, become apoptotic, and display poor adhesion in the absence of serum . The removal of serum has been reported to slow down the proliferation rate of MSCs . Nevertheless, the osteochondral potential of MSCs can still be maintained in a chemically defined serum-free medium . Presently, most studies with MSCs use serum-free in vitro culture conditions so as to avoid the arte-factual and pleiotropic effects of serum on the experimental data. However, for optimal ex vivo proliferation of MSCs, the presence of serum is still preferred . With primary chondrocytes, the absence of serum has also been reported to be detrimental to the proliferation rate . In the case of ES cells, serum-free in vitro culture condition for differentiation into the chondrogenic lineage was recently reported by Nakayama et al. .. j- j( W' O& |- y! v6 k

" F% F/ S" b. c& UTo prevent transmission of pathogens, the patient’s own serum could be used for the in vitro culture of stem cells. However, for clinical applications, there are several reasons that make it preferable to eliminate serum from the in vitro culture milieu. First and foremost, the composition of serum is poorly defined, with a considerable degree of interbatch variation, even when obtained from the same patient or manufacturer. This impedes good quality control in the laboratory. Serum is also not completely physiological, because it is essentially a pathological fluid formed in response to blood clotting. Additionally, it may also contain uncharacterized growth and differentiation factors, which may result in the uncontrolled spontaneous differentiation of stem cells into divergent multiple lineages other than the chondrogenic lineage.
% N+ N7 |9 S6 z: g  w( U( ]( ]' D5 ^$ k
A step toward serum-free culture conditions is the development of chemically defined synthetic serum substitutes. At present, there are several such commercially available synthetic serum substitutes . Most notable of these is knockout serum replacement (KSR), which was specifically developed for the maintenance of ES cells in an undifferentiated state within in vitro culture . The exact chemical composition of KSR is not available, because it is a protected trade secret. However, it has been reported to be completely devoid of any undefined growth factors or differentiation-promoting factors . This would be extremely useful for achieving controlled differentiation of stem cells into the chondrogenic lineage in vitro.6 a7 {* P, L5 ?9 a" \
. C6 n  o% r9 R" U( k, b4 j( _- z3 X. y$ w
Use of Exogenous Cytokines and Growth Factors to Promote Chondrogenic Differentiation
) d+ G, e, n5 q% }/ |4 \- F- ~
/ d5 V" U* y, m/ H+ e8 {) Q4 B+ b- M! G  ^The use of exogenous cytokines and growth factors is another step forward in the development of a defined culture milieu for directing the chondrogenic differentiation of stem cells. Indeed, numerous cytokines and growth factors have been implicated in chondrogenesis (Table 1), and many of these display a high degree of functional overlap. Because the process of chondrogenesis is so closely intertwined with osteogenesis, many of the cytokines and growth factors that promote chondrogenic differentiation are also somewhat implicated in osteogenic differentiation . Hence, the challenge is to find an optimized subtle combination of these various cytokines and growth factors that would bias differentiation specifically toward the chondrogenic lineage. An added complication is that many of these cytokines and growth factors exert nonspecific pleiotropic effects on stem cell differentiation, which is most likely attributable to the activation of multiple intracellular signaling pathways by each individual cytokine or growth factor.1 D1 F% v! C: c  l& W) d
/ ~9 I/ c% _3 h/ @% F2 `! k
Among the most potent inducers of chondrogenic differentiation are members of the transforming growth factor beta (TGF-?) family. These consist of more than 40 polypeptide growth factors that share a high degree of homology, in particular the seven conserved residues in their C-terminal region . Besides the various isoforms of TGF-?  and bone morphogenetic protein (BMP) , other members of the TGF-? superfamily include activin , osteogenic protein-1 , and GDF-5 . The effects of cytokines of the TGF-? superfamily on chondrogenic differentiation are transduced through two major intracellular signaling pathways. The first of these involves the SMAD family of signaling molecules , whereas the second pathway involves mitogen-activated protein kinase (MAPK) signaling . Interestingly, both signaling cascades are activated by the same TGF-? receptor complex. This comprises two distinct transmembrane protein components (type I and II receptor) that undergo heterodimerization upon binding to cytokines of the TGF-? superfamily. More recently, there is also evidence of regulatory cross-talk between TGF-?–activated signaling cascades and the Wnt pathway during chondrogenic differentiation . An interesting aspect of the TGF-? superfamily is the modulation of its biological activity through binding interactions with extracellular matrix proteins such as ?-glycan and endoglien . These are thought to alter the presentation of these cytokines to their corresponding receptors on the cell surface.$ S  A! \6 L( ]8 }9 i" j

: t4 E: q, `' w/ H( b4 aAnother family of cytokines that plays an important role in chondrogenesis is comprised of the various isoforms of fibroblast growth factor (FGF). Numerous studies have implicated the FGF family of cytokines in limb bud cartilage formation . The effects of FGF on chondrogenic differentiation are transduced primarily through MAPK signaling . This leads to increased expression of the transcription factor Sox9, which is the master regulator of chondrogenesis. Additionally, there is also evidence of regulatory cross-talk between FGF-activated signaling cascades and the signaling pathways initiated by parathyroid hormone–related peptides and Indian hedgehog .; N" s0 `% \$ {! j( U; T

% P2 W) ]( R0 U3 Y6 _Besides the TGF-? and FGF family of cytokines, insulin-like growth factor-1 (IGF-1)  has also been shown to have potent stimulatory effects on chondrogenic differentiation. It is believed that the chondrogenic-promoting activity of IGF-1 is primarily transduced through a phosphatidyl inositol 3-kinase signaling pathway . Because the combination of TGF-?1 with either IGF-1  or FGF-2  was reported to have a synergistic effect on chondrogenic differentiation in vitro, it is likely that the IGF-1–activated signaling cascade could have regulatory cross-talk with the signaling pathways initiated by the TGF-? and FGF family of cytokines. Other proteins that have also been shown to have a stimulatory effect on chondrogenesis include prolactin , interleukin-1? , Cyr61 , HB-GAM , and growth hormone . All of these are summarized in Table 1.! n* W0 M0 O/ g6 ]9 C; \

7 w( a9 H& c: |# F! D3 X+ nUse of Nonproteinaceous Chemical Compounds to Promote Chondrogenic Differentiation
2 ?! q6 B  F2 V! b4 ]  ?) _) o9 T0 l
- Q* d! f% d$ Q( i3 `3 XIn addition to protein-based cytokines and growth factors, several nonproteinaceous chemical compounds have also been shown to promote chondrogenic differentiation in vitro (Table 2). Such chemicals tend to be less labile, with a longer active half-life in solution, compared with protein-based cytokines and growth factors. This is advantageous for prolonged in vitro cell culture over several days or even weeks. Moreover, unlike proteins that have to be synthesized in living cells and subjected to complex post-translational modifications (i.e., glycosylation, peptide splicing, conformational folding), non–protein-based chemical compounds can be manufactured by chemical reactions in the laboratory and hence are more structurally and chemically defined compared with proteins.  D5 U$ {+ e3 L  Z" N) B
, D  \- s9 l$ T
Table 2. Nonproteinaceous chemical factors that promote chondrogenesis# O! i2 B, R% y
" `6 Z2 \5 T/ v, m: K; i9 f
Among the nonproteinaceous chemical compounds that are known to promote chondrogenic differentiation in vitro (Table 2) are dexamethasone, thyroid hormone, 1,25-dihydroxy vitamin D3, prostaglandin E2, ascorbic acid, ethanol, staurosporine, dibutryl cAMP, concavalin A, vanadate, and FK506. Dexamethasone is a synthetic glucocorticoid that is a potent inducer of chondrogenic differentiation in human-derived mesenchymal stem cells . Thyroid hormones are steroid derivatives of cholesterol metabolism that have also been implicated in chondrogenic differentiation . In an interesting study by Locker et al. , it was reported that the sequential addition of dexamethasone and thyroid hormone (triiodothyronine) under serum-free conditions permitted full chondrogenic differentiation of the pluripotent mesoblastic C1 cell line. By contrast, the addition of exogenous cytokines (IGF-1 and TGF-?), as well as the cell-intrinsic activation of the BMP autocrine signaling pathway, failed to elicit full chondrogenic differentiation . 1,25-dihydroxy vitamin D3, also known as calcitriol, is the active form of vitamin D. It has been reported to stimulate chondrogenesis of embryonic limb bud mesenchymal cells  and promote expression of differentiated chondrocyte function within in vitro culture . Prostaglandin E2 (PE2) is a naturally occurring eicosanoid that is derived from arachidonic acid metabolism. Numerous studies have implicated PE2 in limb cartilage formation  and chondrocyte differentiation . Ascorbic acid, better known as vitamin C, has been shown to stimulate chondrogenic differentiation by promoting 1,25-dihydroxy vitamin D3 synthesis and cartilage matrix production . The teratogenic effects of ethanol on vertebrate development are well-known. It has been demonstrated to have potent stimulatory effects on the chondrogenic differentiation of both embryonic limb  and embryonic facial  mesenchyme cells. Staurosporine, a protein kinase C inhibitor, has also been demonstrated to have stimulatory effects on the chondrogenic differentiation of both embryonic limb  and embryonic facial  mesenchyme cells. Dibutryl cAMP, the cell-permeable form of cAMP, was reported to have a stimulatory effect on embryonic limb bud cartilage formation . Concanavalin A is a member of the lectin family of carbohydrate-based compounds. It has been reported to induce both neural and cartilage tissue formation in amphibian early gastrula ectoderm . Vanadate, an inorganic vanadium salt, has been demonstrated to promote cartilage-matrix proteoglycan synthesis in rabbit coastal chondrocyte cultures . FK506 is an immunosuppressive drug that has been shown to stimulate chondrogenic differentiation of a clonal murine embryonic carcinoma cell line, ATDC5 .
% y2 z- d6 \* d. j  b1 ?6 ~) @; e9 b5 k9 J1 q6 Y
Use of Naturally Occurring and Artificially Synthesized Extracellular Matrix Substratum to Promote Chondrogenic Differentiation
& Q% \0 m- `  @+ i4 L# f) j* h; t5 {' P7 W9 K7 L8 h$ N
The in vitro culture milieu for directing chondrogenic differentiation should also incorporate naturally occurring and artificially synthesized extracellular matrix (ECM) substratum to optimize cell attachment, growth, and differentiation. Indeed, the presence of ECM components would more closely replicate the physiological environment that supports chondrogenesis. In situ, cartilage tissue consists of chondrocytes embedded within an avascular ECM that serves to maintain its structural integrity. This is primarily comprised of collagenous proteins  and proteoglycans  that account for the bulk of its dry weight (60%–90%), together with smaller quantities of noncollagenous proteins  and hyaluronan . Within cartilaginous tissue, the major collagen isoform is collagen type II , whereas the predominant proteoglycan is aggrecan . In addition to its structural role, cartilage ECM plays a physiological role by influencing the immediate microenvironment of the chondrocytes embedded within it. Histological studies have reported extensive remodeling of cartilage ECM during growth and development  as well as under various pathological conditions .
- X2 w! ?0 L% y$ o" f& Q
. R6 X# v% X) ?/ \1 m: [0 G, E3 zHence, the introduction of appropriate ECM substratum within in vitro culture would certainly enhance the directed differentiation of stem cells into the chondrogenic lineage . These can either be based on naturally occurring components of cartilage ECM or may use synthetic materials. Additionally, composite matrix scaffolds of both natural and synthetic materials have also been fabricated.' x2 F+ {; v& g( V4 [

8 X4 d5 X- g# OIt is important to note the various properties of ECM substratum that would be favorable for chondrogenesis. For tissue engineering applications, it is imperative that the synthesized matrix substratum is biocompatible and has no cytotoxic properties. Biodegradability is also another preferred characteristic , although this is not absolutely critical. Culture on 3D matrix scaffolds has been reported to be superior to conventional 2D monolayer culture for maintaining the differentiated phenotype of chondrocytes . Porosity of the ECM substratum is essential for 3D tissue growth. Hence, matrix scaffolds for cartilage tissue engineering are often fabricated as porous foams or granules. At the present moment, there are as yet no reported studies on the use of ECM substratum to promote the chondrogenic differentiation of ES cells. Hence, the following discussion will focus solely on MSC and primary chondrocytes.1 e6 o9 X5 S  H+ `
0 Y% }* L8 K9 B3 R$ Z3 ]
The attachment, proliferation, and subsequent chondrogenic differentiation of MSC and chondrocytes on the ECM substratum are dependent on several interrelated properties, namely chemical composition, electrostatic charge, surface texture/roughness, and geometrical configuration . Obviously, the chemical composition of the ECM substratum is the most critical factor in determining its ability to influence chondrogenesis. These should preferably incorporate naturally occurring constituents of cartilage ECM, so as to provide a more physiological environment for chondrogenic differentiation. Indeed, matrix scaffolds fabricated for cartilage tissue engineering are often based on either collagen , hyaluronan , chondroitin sulfate , mineralized calcium , fibrin , or composites of these materials , all of which are constituents of cartilage ECM. Other naturally occurring materials that are not found in cartilage ECM have also been used. These include gelatin , chitosan , and alginate . Additionally, matrix scaffolds based completely on synthetic materials such as poly (lactic-co-glycolic acid), poly (ethylene glycol), poly (epsilon-caprolactone) have also been fabricated . Nevertheless, it would be preferable to use these as composites with naturally occurring materials. The electrostatic charge on the surface of the ECM substratum is another important factor. The presences of negative charges have been reported to be advantageous for the adhesion and spreading of MSCs on ECM substratum . Furthermore, there is evidence that variations in the electrostatic charge content of the peri-cellular matrix synthesized by chondrocytes have a profound influence on the biomechanical properties of cartilaginous tissue . Surface texture/roughness seems to have differential effects on chondrocyte proliferation, differentiation, and matrix production, depending on the maturational state of the chondrocyte itself . Bhardwaj et al.  reported that geometric configuration of the matrix scaffold, as defined by average pore size, had profound effects on chondrocyte proliferation and matrix synthesis.- j7 f: {' a( p, \- Y2 D+ {

( Q6 w5 S, d5 O( l8 SAs mentioned earlier, collagen type II is the most abundant collagen isoform within cartilage ECM. Indeed, it has been reported that cartilage-specific collagen type II is superior to collagen type I for primary culture of chondrocytes . Although chondrocytes appeared to maintain high proliferative capacity when cultured in collagen type I, there was a gradual loss of differentiated phenotype . To enhance their structural and chondroinductive properties, collagen-based matrices are often fabricated as composites that may incorporate a variety of naturally occurring and synthetic materials such as mineralized calcium phosphate , agarose , proteoglycan aggregates , demineralized bone powder , fibronectin , chondroitin sulfate , hyaluronan , poly-L-lactic acid , and polylactide-coglycolid .
; Z! K" j: w; e9 @  `" l; @8 ^3 _1 D$ P- |! B! G; ]
Another class of bone ECM molecules that is known to play an integral role in chondrogenic differentiation is the glycosaminoglycan (GAG), which is essentially a long-chain sugar molecule. With the exception of hyaluronan, all of GAGs (heparin sulfate, chondroitin sulfate, keratin sulfate, and dermatin sulfate) are sulfated branch-chained molecules that are conjugated to proteins in the form of proteoglycans. Chondroitin sulfate is the major sulfated glycosaminogly can found in cartilage (predominantly conjugated to aggrecan). Indeed, chondroitin sulfate–based matrix scaffolds have been reported to enhance chondrocyte proliferation and maturation . Additionally, hyaluronan-based matrix scaffolds are also widely utilized in cartilage tissue engineering .
/ u6 a1 _# m2 U) L. f/ B
; w! f) Y5 t3 P) e7 }% jAt present, the development of ECM substratum for application in cartilage tissue engineering is progressing rapidly. It is anticipated that more novel types of composite matrix scaffolds incorporating a variety of natural and synthetic materials will be developed in the near future for the controlled differentiation of MSC and even ES cells into the chondrogenic lineage.+ W4 J* j% d1 [" z! ?3 `

. k! P8 |. ~6 vCOCULTURE AND CELL-CONDITIONED MEDIA! E1 G) L5 p; d9 O  u( o! M

/ H& d1 e: L2 ?; E2 F% B) ITop
: I3 H1 Y& V) I4 [" M
" z) E2 N. }9 yAbstract* w$ W* J+ {. T( }

3 F  S7 P: C& R( RIntroduction
. f' j, ~! v! P$ x2 h. \( Z) F' w7 a% w
Adult Stem Cells versus...# I( |" d2 ~% f( ?

- ?9 w3 u$ y) j: W3 _, _' JDevelopment of Defined Culture...
) }& s8 K2 K; h6 `/ s
* P/ r/ x( u& b7 UCoculture and Cell-Conditioned.... i' `" |2 T- Y+ E, e
- e8 E1 }/ }; b( Z
Directing Chondrogenic...
' a* i% v. B! u! J( w. d4 U, b1 W9 i3 X
Future Perspectives2 h8 |4 h! n* F! d

) w+ Y1 n* w  s% GConcluding Remarks
2 S: }' S8 l* E3 K  x$ D; E% v
. \; L( O+ W$ Y: ]5 d$ KReferneces
# ~' @/ \  _! e4 B* U- G7 Z4 k, M& ^& b+ z% ]
Another strategy to direct the chondrogenic differentiation of stem cells is to coculture the stem cells with a different cell population. Differentiation of ES cells into the chondrogenic lineage was enhanced by coculture with limb bud progenitor cells . With mesenchymal cells, coculture with synovial-lining macrophages  and embryonic calvarial cells  was reported to stimulate chondrogenic differentiation. The main advantage of coculture systems is that this allows intimate contact between different cell types, which may lead to a more efficient transduction of molecular signals that induce chondrogenic differentiation. The surface receptors of cocultured cells come into direct physical contact, and the autocrine and paracrine factors secreted by one cell type readily interact with the other cell type.5 k0 \: Q9 \& {

- W& K# H# N4 y4 E/ L( ]/ H) zMore recently, there has been evidence that intimate physical contact may lead to fusion of different cell types in vivo, resulting in the formation of heterokaryons . In fact, cell-fusion phenomenon has been used to explain the ability of adult stem cells to transdifferentiate into cell types that are radically different from their tissue of origin when transplanted in vivo . Nevertheless, there is as yet no evidence that the stimulatory effect of coculture on chondrogenic differentiation  is the result of cell fusion. Coculture of two or more distinct cell populations also carries a strong risk of transmission of pathogens, in particular viruses. This would constitute a major obstacle to the clinical application of coculture for chondrogenic differentiation. In the clinical situation, it would simply not be practical to stimulate chondrogenic differentiation of stem cells through coculture with either an autogenic or donated cell source. Another major shortcoming of coculture is the difficultly in the separation of cocultured cell populations. The highest degree of purity upon separation could be achieved by fluorescence-activated cell sorting (FACS) . However, FACS is skill intensive and requires expensive instrumentation. Magnetic-affinity cell sorting  is much cheaper compared with FACS, but the degree of purity upon separation is much lower. The problem of separating distinct cell populations, as well as the potential problem of cell fusion, may be overcome by keeping cocultured cell populations physically separated through the use of commercially available Transwell inserts . The other alternative is to use filtered cell-conditioned media instead.& `9 I6 q% @/ r8 B
4 k/ M! T- _/ H- C
Indeed, culture media conditioned by embryonic calvarial cells , embryonic limb bud , articular perichondrial cells , and primary chondrocytes  have all been reported to have stimulatory effects on chondrogenesis. This could therefore suggest that intimate cellular contact within coculture might not actually be necessary to achieve stimulation of chondrogenic differentiation. Nevertheless, it is important to note that the use of filtered conditioned media does not alleviate the risk of viral transmission. Also, secreted factors within conditioned media may be labile and hence may not be suitable for prolonged durations of in vitro culture." R$ H" z* j3 l/ q" p! \9 j

8 n4 Y+ r: _# tDIRECTING CHONDROGENIC DIFFERENTIATION THROUGH GENETIC MODULATION8 }# S5 t8 p  y2 l
4 ?5 K- ~, J! L* h
Top
* i+ K7 {, D  x. z: p* D/ h7 f1 V, R- C0 \6 g1 S0 G2 m: r
Abstract( o* l1 J; U( A

: P' q* f4 Z( f. K# X* O' I' pIntroduction
! v5 l( F. b( C# Q3 r4 S! V
) j$ q* E% u0 Y* s' MAdult Stem Cells versus...6 c! T3 N/ e7 `% [! S+ e
& @" q+ |8 v6 W8 z( L2 Q
Development of Defined Culture...% J4 O6 U, K9 i$ e/ X/ U$ H

) N& H6 S/ P3 W7 l% NCoculture and Cell-Conditioned...
0 G6 C+ f" b: x7 u! ~6 N
, w* d! A/ r0 E" {3 [Directing Chondrogenic...
% L( u  u* O1 K/ d8 t/ X4 p; h# [* H% z9 ], Q; k9 M
Future Perspectives
1 b9 z! d" a+ [8 q8 C8 s
5 z/ i( F7 n. k: IConcluding Remarks
. D2 Q: @4 Z8 E4 d, q" t7 s& k! I) K! I6 Q( a+ Y0 N) ?1 l
Referneces# c. y- j; i  D+ V
! b7 X8 W: t% g! c% O, Q
Directing chondrogenic differentiation with exogenous cytokines, growth factors, and ECM substratum or even with coculture and conditioned media would require prolonged durations of in vitro culture. This has two major disadvantages if autologous adult stem cells are to be used for cartilage regeneration. First, this would obviously delay treatment to the patient. Second, there is evidence that prolonged durations of ex vivo culture could somehow alter the immunogenicity of cultured autologous cells, which could in turn lead to immunorejection upon transplantation .
+ w' N  C6 N% X8 m3 D
' K/ u" K! A5 ?' O; ?2 B) i7 ?0 pA novel alternative for directing and controlling the chondrogenic differentiation of stem cells is through genetic modulation, which would obviate prolonged durations of in vitro culture. This could be achieved by transfecting stem cells with recombinant DNA constructs encoding for the expression of certain proteins or growth factors that promote chondrogenesis. Of particular interest are transcription factors implicated in the pathway of chondrogenic differentiation. These control the expression of the entire array of proteins specific to the chondrogenic lineage and include members of the Sox family, Sox5 , Sox6 , Sox8 , and Sox9 , as well as a variety of other cytosolic proteins such as Lc-Maf , CREB-binding protein , Nkx3.2 , AP-2, SP-1 , Pax1, Pax9 , DEC1 , Brachyury , and the Smad family of signaling molecules . Of these various transcription factors, Sox-8 and Sox-9 are thought to be the most downstream regulators of chondrogenesis .4 W2 L' Y3 M/ d; b, I: D
$ j2 F5 q: C: T0 W. X; n3 J0 }
Indeed, recombinant overexpression of Sox9 , DEC1 , and Brachyury  have all been reported to accelerate chondrogenesis. Besides transcription factors, the recombinant expression of several other proteins has also been reported to promote chondrogenic differentiation. These include IGF-1 , TGF-?2 , Midkine , and NCAM . Additionally, it was also reported that recombinant expression of Wnt-3a enhances BMP-2–induced chondrogenesis of murine mesenchymal cells .2 K' l- D& T8 v2 E

. m$ Q& }- t& l- M# ?; WThe disadvantage of directing chondrogenic differentiation through genetic modulation is the potential risk associated with using recombinant DNA technology in human clinical therapy. For example, the constitutive overexpression of any one particular protein or growth factor within transfected stem cells would certainly have unpredictable physiological effects on transplantation in vivo. This problem may be overcome by placing the recombinant expression of the particular protein under the control of switchable promoters, several of which have been developed for expression in eukaryotic systems. Such switchable promoters could be responsive to exogenous chemicals, heat shock, or even light. Of particular interest are light-responsive promoters , because these would avoid the potentially toxic or pleiotropic effects of exogenous chemicals and heat treatment. At present, there are as yet no reported studies on the coupling of chondro-specific genes to light-responsive promoters. Indeed, the creation of such recombinant constructs and their subsequent transfection within stem cells would certainly make an interesting study with potentially useful clinical applications.
& R: b8 I* X( B+ O. U( ?/ W8 r$ d' H' _/ |* O  i0 [
The cellular signaling pathway for chondrogenic differentiation could be given a kick start through temporary expression of chondro-specific transcription factors coupled with light-inducible promoters. After that, it is possible that the pathway for chondrogenic differentiation could carry on independently of the recombinant expression of these transcription factors, because the entire array of chondro-specific genes would have already been activated. The advantage of this approach is that there is no constitutive overexpression of any one particular transcription factor. Also, the natural cellular pathway for chondrogenic differentiation could carry on physiologically switching off the recombinant expression of these transcription factors through removal of light stimulus. Upon transplantation in vivo, it is extremely unlikely that light-inducible promoters would again be activated, because light stimulus would be completely absent in situ.+ y, |9 d. v- M' g

3 ]1 L3 s/ m- e, `4 ?4 d1 p1 c# AGenetically modified stem cells may also run the risk of becoming malignant within the transplanted recipient. Moreover, there are overriding safety concerns with regards to the use of recombinant viral-based vectors in the genetic manipulation of stem cells . It remains uncertain as to whether legislation would ultimately permit the use of genetically modified stem cells for human clinical therapy. At present, the potential detrimental effects of transplanting genetically modified stem cells in vivo are not well-studied. More research needs to be carried out on animal models to address the safety aspects of such an approach.
9 R/ n- C: b; O4 k+ Q0 r+ U/ x' y! j5 t& e0 S+ t4 _, ~9 t
FUTURE PERSPECTIVES
- n5 B; y6 K2 v: K5 y3 n" ]
/ {0 G$ E1 v& _1 t9 e: E# BTop7 F% r. p' S- R
! q! Z/ z( Q8 P" b  W# \
Abstract
, j4 L5 h/ {* B' t3 a
6 Q5 \! {, c$ q1 W8 F! VIntroduction
3 V3 F( P, B$ ?# N
. {# j4 s) J7 Y: d  p- pAdult Stem Cells versus...
# h! j9 T2 p! c! A7 l( Z
4 h6 B3 C+ I) w9 x, R. nDevelopment of Defined Culture...9 S9 Y/ u! R3 Q3 T/ f
- n* R& G+ L0 A( I5 W7 ]
Coculture and Cell-Conditioned...
# z1 }( Z; S: x2 l! c- r, K
/ I* [6 J# N" l5 _5 E( ^Directing Chondrogenic...2 V/ E9 w0 D1 N) b2 Q+ z
) j& z: ]7 v- S' C
Future Perspectives
7 ]: R- p# J% w0 W
2 h' F  }! f- nConcluding Remarks
) ~6 N2 g/ K- i" l' y; H, _1 Y" V" d( m4 B1 t
Referneces
# j# [# `! f% s& |7 |' R, ~8 t! X$ W) y' ]
For therapeutic applications, there seem to be two major strategies for directing stem cell differentiation into the chondrogenic lineage in vitro: through the development of a defined serum-free culture milieu and through genetic modulation with recombinant DNA technology. The use of coculture and conditioned media to promote chondrogenic differentiation would most likely not be useful for clinical applications. However, these could still provide useful in vitro experimental models for further studies in chondrogenesis.
0 F3 K- `- J% ~
, {" `/ |) B% t' Y( QAs mentioned earlier, there are overwhelming safety concerns with regards to the use of genetic modulation to direct stem cells into the chondrogenic lineage. Unless such safety concerns can be allayed, the method of choice to direct the chondrogenic differentiation of stem cells for therapeutic applications would most probably use a defined serum-free culture milieu incorporating various cytokines, growth factors, chemicals, and ECM substratum. Figure 1 summarizes the major components of such a defined culture milieu.
: v- S2 B1 j1 R9 p. B) i1 B, z6 a6 Z6 v* T3 R
Figure 1. Schematic diagram representing the major components of a defined culture milieu for directing the differentiation of stem cells into the chondrogenic lineage. (A): Protein-based cytokines and growth factors; (B): extracellular matrix; (C): nonproteinaceous chemicals; (D): biophysical parameters such as O2 tension and temperature; (E): cell density, which determines the degree of cell-to-cell contact and gap junction–mediated intercellular coupling. Abbreviation: TGF-?, transforming growth factor ?.
7 s5 V' n! D& d0 C' E( [/ Z4 ^& J7 l9 r6 s3 ~' Y7 F3 @# v
To further optimize the culture milieu for chondrogenic differentiation, it may be worthwhile to look at other biophysical parameters in addition to the culture media composition. This would include varying oxygen tension  and temperature within the incubator, as well as the application of physical stimuli in the form of electrical  and electromagnetic  fields, mechanical forces , heat shock , ultrasound , and even laser irradiation . Indeed, evidence from the scientific literature suggests that the pathway of chondrogenic differentiation can be profoundly influenced by variations in such biophysical parameters., l+ f1 ^# z+ c1 Q: A8 |

$ c, H+ V; S* S0 z/ eBecause cellular contact and gap junction–mediated intercellular coupling play an integral role in mesenchymal condensation during the initial stages of chondrogenesis , it is likely that variations in cell density within in vitro culture could profoundly affect the pathway of chondrogenic differentiation. Additionally, it may also be useful to look at cytokines that encourage gap junction formation during chondrogenesis . Nevertheless, it is important to note that at more advanced stages of chondrogenic differentiation, the maturing chondrocytes gradually lose their gap junctional communication  and become surrounded by a layer of pericellular matrix . This would preclude any direct cell-to-cell contact, as well as gap junction–mediated intercellular coupling.
5 u! |- g4 d9 v
& E& n3 Y3 M1 c6 DOther possible techniques to direct chondrogenic differentiation include exposure to cytoplasmic extracts and cybridization. H?kelien and colleagues  managed to reprogram 293 T fibroblasts to express T-cell and neuronal function by exposing permeabilized cells (i.e., with streptolysin-O) to concentrated cytoplasmic extracts of T cells and neuronal progenitors, respectively. However, to date, there are as yet no reported studies on the use of cytoplasmic extracts of chondrocytes or chondroblasts to direct the differentiation of either fibroblasts or stem cells into the chondrogenic lineage. Another novel approach is to fuse stem cells with enucleated cytoplasts derived from chondrocytes or chondroblasts to form cytoplasmic hybrids, or cybrids. Techniques for generating enucleated cytoplasts from differentiated somatic cells have been developed and refined over the course of the past three decades . Briefly, this usually involves treatment of the somatic cells with a microtubulin inhibitor (i.e., cytochalasin), followed by high-speed centrifugation within a layered density gradient (comprised of either Ficoll or Percoll). The enucleated cytoplasts can then be fused with nucleated cells through a variety of different techniques using electrical pulse  or polyethylene glycol  or with Sendai virus . To date, there are no studies that have yet been reported on the use of cybridization to direct chondrogenic differentiation. Nevertheless, previous studies have shown that cybridization could be used to induce teratocarcinoma cells to express myoblast function  as well as direct erythroid  and myeloid  differentiation.# B' l; Q( D5 ~8 S/ A
; n- X* j5 K3 ^% V: `
CONCLUDING REMARKS, n% p% h4 J) p0 W9 }, F

- P6 X! E' @% t* M$ g# RTop
( Y$ [6 D9 o# k5 D# P" a; |0 F" w% p# F: L; Z' {1 V/ D/ Z
Abstract
% ?6 l9 J! M, n. t  n2 A; R1 _2 n: u: g/ L* O
Introduction
% _  i7 J$ Q+ h" o" V2 x4 {) @/ H) G4 C4 L3 R4 _
Adult Stem Cells versus...9 c' P5 N9 _$ C8 h

% a' d# ~! `6 V2 u& T# a  NDevelopment of Defined Culture...1 P" W+ U& w  J& q7 v6 f! o
& f& ^" f. K4 P- z
Coculture and Cell-Conditioned..." u' p3 ~6 a9 K/ |0 D4 `0 O# P
0 T# I% \5 t) g7 e3 ^: ?& K
Directing Chondrogenic...
& _" S: U7 X; {5 N# g1 x8 Q( b. R
( i8 C0 _  W# F  |& RFuture Perspectives
% k0 z) H9 t2 [1 T
$ n7 T) T# O) TConcluding Remarks
5 b* d. H4 q& K/ e+ e3 `+ J% J/ t5 k. n- [6 X8 d- D( O5 I
Referneces
9 a% q  C1 l: T* p% v/ m; s+ \/ j: q
Despite the large number of studies that have recently been carried out on the chondrogenic differentiation of stem cells in vitro, this particular area of research is still in its relative infancy. Chondrogenic differentiation may be additionally enhanced if the various techniques that have so far been discussed are used in combination rather than exclusively by themselves. In the natural milieu, the chondrogenic differentiation of stem cells probably involves multiple signaling pathways. This may be mimicked in vitro by using a combination of these various techniques to achieve a synergistic effect on the differentiation of stem cells into the chondrogenic lineage.
6 ^5 n  G. E: }4 k7 Y3 G8 q" a
2 @' ^* q3 z+ h3 j8 {; W( JHowever, it must be kept in mind that for clinical applications, it is imperative to develop well-defined and efficient in vitro protocols for the chondrogenic differentiation of stem cells that would use chemically defined culture media supplemented with recombinant cytokines and growth factors. This will then provide the stringent levels of safety and quality control that would make the clinical applications of stem cell transplantation therapy realizable. The hope is that this will be achieved in the near future.
  m! M8 Q7 ~1 I$ K1 \2 f
& G3 \, x# y8 V3 W, l1 ?  ?( r2 z5 xREFERNECES
# h$ H  w, _! g6 a( V+ Y# M: M2 }3 ]
Top
+ a- k/ g4 O' q9 K! b+ Q$ A- E: K1 M5 a8 G9 p4 Z: E; }3 G
Abstract
9 {2 j  H8 U& V! `
+ W6 L; r& W* r/ _2 ~4 ?; QIntroduction
0 L9 j6 C* b$ C! m) ^# i: {0 R+ ]% [+ }4 c
Adult Stem Cells versus...
  j7 F6 b  ~( {  Y7 w
8 u' T" `# l0 T8 Q2 T: x$ WDevelopment of Defined Culture...8 a" b* J1 i3 l# h( _- e
+ S8 m# d5 ?" T) k5 x/ w
Coculture and Cell-Conditioned...
: b3 ~. }, ~2 R* c- ~& p$ i0 t3 n( Q8 G  n' s" U
Directing Chondrogenic...
! x0 k2 j: U4 `3 [
  e  E, d$ k) x% B8 ]/ V0 bFuture Perspectives3 q  _2 |. d" A; L0 Y

/ \8 `! l8 }4 Z; l" tConcluding Remarks" _3 {6 a, M4 s8 K- x- t' o% w
! {  j6 x$ \# }- ~  e5 r
Referneces
3 Z& ~3 C% [: Y% O" V" Z: G- Z. }% k% I6 Y( C1 O1 y5 a( M8 R/ z
Buckwalter JA. Articular cartilage injuries. Clin Orthop 2002:21–37.
' u0 ^/ o7 g$ F& ~$ i0 X! X
) ~& x0 G/ h( x: @/ E' X* m, H# [Martin JA, Buckwalter JA. Aging, articular cartilage chondrocyte senescence and osteoarthritis. Biogerontology 2002;3:257–264.6 V: m& L+ \5 m8 q
; q. f9 ^; Z' c+ r9 {
Buckwalter JA, Mankin HJ. Articular cartilage: degeneration and osteoarthritis, repair, regeneration, and transplantation. Instr Course Lect 1998;47:487–504.
0 b9 y) f  `+ u: r: r5 z5 k8 V* k! W$ h
Frenkel SR, Di Cesare PE. Scaffolds for articular cartilage repair. Ann Biomed Eng 2004;32:26–34.5 K/ _- o. F# d* D+ l" W7 g

+ l5 K7 z7 P* y( f, LHardingham T, Tew S, Murdoch A. Tissue engineering: chondrocytes and cartilage. Arthritis Res 2002;4(suppl 3):S63–S68.
% g% Q- t# u3 g$ o+ T- b  N
4 i) g* g& B+ g: U/ wMitchell N, Shepard N. The resurfacing of adult rabbit articular cartilage by multiple perforations through the subchondral bone. J Bone Joint Surg Am 1976;58:230–233.
  A! v: n. T9 z1 f3 t) x* i5 s# t2 }& y' t+ s' e$ j
Mankin H, Mow V, Buckwalter J et al. Form and function of articular cartilage. In: Simon S, ed. Orthopaedic Basic Science. Rosemont, IL: American Academy of Orthopaedic Surgeons, 1994:443–470.$ _; F3 s2 q9 z. Y
; `6 m2 K9 D+ ?  \$ B7 x8 x0 o
Frenkel SR, Clancy RM, Ricci JL et al. Effects of nitric oxide on chondrocyte migration, adhesion, and cytoskeletal assembly. Arthritis Rheum 1996;39:1905–1912.2 A  [+ O% K! }% `

6 W) }% ^, A6 E8 qBrittberg M, Lindahl A, Nilsson A et al. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med 1994;331:889–895.
& ?" u  H6 q( W
: l) N  X( N' jRichardson JB, Caterson B, Evans EH et al. Repair of human articular cartilage after implantation of autologous chondrocytes. J Bone Joint Surg Br 1999;81:1064–1068.) ^6 }( @8 x% w% s! }4 |

6 R( U% o7 S9 H  y+ A* z- P4 rPeterson L, Minas T, Brittberg M et al. Two- to 9-year outcome after autologous chondrocyte transplantation of the knee. Clin Orthop 2000:212–234.7 Y5 ~7 x0 N" L; k
1 e4 j& q! c7 S4 X. E' z) B
Peterson L, Brittberg M, Kiviranta I et al. Autologous chondrocyte transplantation: biomechanics and long-term durability. Am J Sports Med 2002;30:2–12.' q* E  i  T% |# ~
* \. Q& h; M  k
Cancedda R, Dozin B, Giannoni P et al. Tissue engineering and cell therapy of cartilage and bone. Matrix Biol 2003; 22:81–91.! C& p; f+ @2 @& I' J; K' `
7 F0 M5 B) x$ S4 y; Z2 P* f
Dozin B, Malpeli M, Camardella L et al. Response of young, aged and osteoarthritic human articular chondrocytes to inflammatory cytokines: molecular and cellular aspects. Matrix Biol 2002;21:449–459.
+ L8 {6 S4 z# }4 B4 U0 H  g/ w5 Z+ Z( l3 p3 t
Czyz J, Wiese C, Rolletschek A et al. Potential of embryonic and adult stem cells in vitro. Biol Chem 2003;384:1391–1409.: T6 L$ v8 V3 M$ R, X7 i  ~

1 ~- d# Z- c8 j9 D; ]& pWeissman IL. Translating stem and progenitor cell biology to the clinic: barriers and opportunities. Science 2000;287: 1442–1446.
8 N$ R8 W/ ^" t2 l+ ~% A8 y. U% z3 r8 y0 T0 ]- F% k, z% j
Jorgensen C, Noel D, Apparailly F et al. Stem cells for repair of cartilage and bone: the next challenge in osteoarthritis and rheumatoid arthritis. Ann Rheum Dis 2001;60:305–309., x( \& D/ s* ~0 n5 v

6 ~- J+ G# W; Y5 l( Y, t. XMackenzie TC, Flake AW. Multilineage differentiation of human MSC after in utero transplantation. Cytotherapy 2001;3:403–405.4 X* g& K2 k% f  a. g0 y
: }7 W2 V+ b1 t3 \8 ~0 k( B
Trounson A. Human embryonic stem cells: mother of all cell and tissue types. Reprod Biomed Online 2002;4(suppl 1):58–63.
7 Y  W7 ]2 t4 ~; {" V
9 l8 K" V# b! Q& r% K6 k1 Z& v$ WYoo JU, Mandell I, Angele P et al. Chondrogenitor cells and gene therapy. Clin Orthop 2000;(379 suppl):S164–S170.# {! J" `4 n3 K) n; t; M; c9 w1 Q0 q

- x, P$ Q9 u5 Z/ u3 \  sThomson JA, Itskovitz-Eldor J, Shapiro SS et al Embryonic stem cell lines derived from human blastocysts. Science 1998;282:1145–1147.* I8 H: {0 A: L$ _
) S9 `/ l& R3 h
Reubinoff BE, Pera MF, Fong CY et al Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 2000;18:399–404.; A8 @$ t8 M- j( c3 q

4 D% ]# n7 q8 l) N# UYoo JU, Barthel TS, Nishimura K et al. The chondrogenic potential of human bone-marrow-derived mesenchymal progenitor cells. J Bone Joint Surg Am 1998;80:1745–1757.3 \$ k# v/ N4 K2 N# F

) ^" `0 t2 {8 M" p: ], t. \2 V( MSchulick AH, Taylor AJ, Zuo W et al. Overexpression of transforming growth factor beta1 in arterial endothelium causes hyperplasia, apoptosis, and cartilaginous metaplasia. Proc Natl Acad Sci U S A 1998;95:6983–6988.
8 A8 ?% N* x; W9 J  K' {! r. E. z# e' A# o5 d" ]  {
Doherty MJ, Ashton BA, Walsh S et al. Vascular pericytes express osteogenic potential in vitro and in vivo. J Bone Miner Res 1998;13:828–838.
6 Z, R" z- _. f8 A' a; a9 ?# |8 d1 [4 I, w# ^- K7 I3 o4 u
Mizuno S, Glowacki J. Chondroinduction of human dermal fibroblasts by demineralized bone in three-dimensional culture. Exp Cell Res 1996;227:89–97.
% Y3 k' V' n$ j! {9 l  T: `7 q# R& d  N# h( _* f6 `) u/ S
Dragoo JL, Samimi B, Zhu M et al. Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. J Bone Joint Surg Br 2003;85:740–747.8 F! n3 C- c3 r

7 c- K$ W9 X9 K2 s' G% l4 EAwad HA, Quinn Wickham M, Leddy HA et al. Chondrogenic differentiation of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials 2004;25:3211–3222.  W6 Y6 d# s' |* [, f
' {  o4 a( t& e4 B" x# Q& p  x
Huang JI, Zuk PA, Jones NF et al. Chondrogenic potential of multipotential cells from human adipose tissue. Plast Reconstr Surg 2004;113:585–594.
" F8 y: u2 ?0 T7 n5 y7 H) x& Q! w& m7 e8 I2 w, k9 w
Nathanson MA. Transdifferentiation of skeletal muscle into cartilage: transformation or differentiation? Curr Top Dev Biol 1986;20:39–62.% c% Y; E- b9 G6 Z% k/ {9 `
0 c+ G5 J$ g; r, ^6 Z4 K  \* n
Labat ML, Bringuier AF, Seebold-Choqueux C et al. Possible monocytic origin of chondrosarcoma: in vitro transdifferentiation of HLA-DR blood monocytelike cells from a patient with chondrosarcoma, into neofibroblasts and chondrocyte-like cells. Biomed Pharmacother 1997;51:79–93." o' T4 d5 W7 d2 f% X

% A# M+ |  [. F4 ^) G6 pVerfaillie CM. Adult stem cells: assessing the case for pluripotency. Trends Cell Biol 2002;12:502–508.& h4 S! z: s1 A0 s+ I( l
6 u$ B! h0 c7 j; L8 x/ T
Terada N, Hamazaki T, Oka M et al. Bone marrow cells adopt the phenotype of other cells by spontaneous cell fusion. Nature 2002;4:542–545.
5 X0 X$ a9 B& I& `, C, O. G  O% H. p3 p/ _4 e
Ying QL, Nichols J, Evans EP et al. Changing potency by spontaneous fusion. Nature 2002;4:545–548.
4 [3 o6 \. ]0 W/ V; k  [6 o6 ^  B( {$ e% x* d
Goodell MA. Stem-cell "plasticity": befuddled by the muddle. Curr Opin Hematol 2003;10:208–213.1 T; k  S5 n3 p$ [

' L( E  p4 v/ U, n& FKramer J, Hegert C, Guan K et al. Embryonic stem cell-derived chondrogenic differentiation in vitro: activation by BMP-2 and BMP-4. Mech Dev 2000;92:193–205.3 D+ b7 o  f1 O8 d  d, W" W- |

* q$ ?- V" h' [Hegert C, Kramer J, Hargus G et al. Differentiation plasticity of chondrocytes derived from mouse embryonic stem cells. J Cell Sci 2002;115:4617–4628.
# V. B) e6 E' h' z1 n
* A" Y2 M" ~/ W9 z5 RNakayama N, Duryea D, Manoukian R et al. Macroscopic cartilage formation with embryonic stem-cell-derived mesodermal progenitor cells. J Cell Sci 2003;116:2015–2028.
4 L% `4 E: l0 n: Z" R+ r4 X8 Z2 ?5 M* n% |& o! o6 Z
Sui Y, Clarke T, Khillan JS. Limb bud progenitor cells induce differentiation of pluripotent embryonic stem cells into chondrogenic lineage. Differentiation 2003;71:578–585.
$ e! d' P6 X! o1 {! t5 I  A  D8 Z: t% F5 _; s/ K* c' h+ I/ l
Steinert A, Weber M, Dimmler A et al. Chondrogenic differentiation of mesenchymal progenitor cells encapsulated in ultrahigh-viscosity alginate. J Orthop Res 2003;21:1090–1097.
* m. L) ?) Z, W2 ^4 p. e3 W& m9 d7 k6 \* ]# s* p0 Z! F( k5 y# M; D
Tuli R, Tuli S, Nandi S et al. Transforming growth factor-beta-mediated chondrogenesis of human mesenchymal progenitor cells involves N-cadherin and mitogen-activated protein kinase and Wnt signaling cross-talk. J Biol Chem 2003;278:41227–41236.8 x( i) e0 r' Z- ~0 }
& _, O! S5 ]# V5 Y, m: y
McCulloch CA, Strugurescu M, Hughes F et al. Osteogenic progenitor cells in rat bone marrow stromal populations exhibit self-renewal in culture. Blood 1991;77:1906–1911.
" Q. r0 p0 C' k/ [" N/ w; Y9 B! I/ P& w; m+ D; g* b+ `
Quarto R, Thomas D, Liang CT. Bone progenitor cell deficits and the age-associated decline in bone repair capacity. Calcif Tissue Int 1995;56:123–129.6 p  |! a5 j# N' _
# h5 F* B1 n4 a9 [6 c) p
D’Ippolito G, Schiller PC, Ricordi C et al. Age-related osteogenic potential of mesenchymal stromal stem cells from human vertebral bone marrow. J Bone Miner Res 1999;14:1115–1122.
/ n! M* M) [" g9 G7 @6 T* l. B3 _4 {/ S3 W2 \% B* o9 i  X
Quarto R, Campanile G, Cancedda R et al. Modulation of commitment, proliferation, and differentiation of chondrogenic cells in defined culture medium. Endocrinology 1997;138:4966–4976.
, b' P, U; w4 V9 Z! o; ?( u& L7 F+ Y1 K6 q7 N$ ?% H% g
NIH Stem Cell Backgrounder, 2003. Available at: http://stemcells.nih.gov/info/basics/. Accessed October 27, 2004.
; [5 m8 T+ R0 R( h* m% A- n5 u8 ?! Q" t- E
Hwang WS, Ryu YJ, Park JH et al. Evidence of a pluripotent human embryonic stem cell line derived from a cloned blastocyst. Science 2004;303:1669–1674.
- ~8 B% d9 X6 [% q: T8 C* `9 }, s, a; h$ s* Q  K0 i
Lanza RP, Cibelli JB, West MD. Prospects for the use of nuclear transfer in human transplantation. Nat Biotechnol 1999;17:1171–1174.+ ~0 s+ s+ m3 M+ l5 Q

) N4 N5 X9 E7 f: o; U3 k6 o% [Wong M, Tuan RS. Nuserum, a synthetic serum replacement, supports chondrogenesis of embryonic chick limb bud mesenchymal cells in micromass culture. In Vitro Cell Dev Biol Anim 1993;29A:917–922.
0 b( g, i7 T7 q2 ^; f, c
  `2 b; H0 @2 E$ U, g/ j# FGoldsborough MD, Tilkins ML, Price PJ et al. Serum-free culture of murine embryonic stem (ES) cells. Focus 1998;20: 8–12.
, C" h* c! L% Z' k. |9 ~
$ F3 q; p/ B+ |Lennon DP, Haynesworth SE, Young RG et al. A chemically defined medium supports in vitro proliferation and maintains the osteochondral potential of rat marrow-derived mesenchymal stem cells. Exp Cell Res 1995;219:211–222.* K7 H6 S" o( \6 B

- S3 ^3 r2 }+ d. [. I- d% o+ }5 ePochompally RR, Smith JR, Ylostalo J et al. Serum deprivation of human marrow stomal cells (hMSCs) selects for a subpopulation of early progenitor cells with enhanced expression of OCT-4 and other embryonic genes. Blood 2004;103:1647–1652.
3 V6 u: o# V7 [" `4 q2 b4 ^
+ K% s$ j" v2 g5 I6 zSchmitt B, Ringe J, Haupl T et al. BMP2 initiates chondrogenic lineage development of adult human mesenchymal stem cells in high-density culture. Differentiation 2003;71: 567–577.1 K! _( }; t% C& P& F
: i6 w$ y" u& h0 W
Kuznetsov SA, Mankani MH, Robey PG. Effect of serum on human bone marrow stromal cells: ex vivo expansion and in vivo bone formation. Transplantation 2000;70:1780–1787.0 t0 m, _1 H# U- @8 h6 H
2 S* w/ S! r4 K7 A1 y9 [
Freyria AM, Cortial D, Ronziere MC et al. Influence of medium composition, static and stirred conditions on the proliferation of and matrix protein expression of bovine articular chondrocytes cultured in a 3-D collagen scaffold. Biomaterials 2004;25:687–697.
) h: |) J$ {/ u$ H, u* _/ ^$ @7 a/ M2 e8 }& N8 h% m! s0 S- e. Z
Mastrogiacomo M, Cancedda R, Quarto R. Effect of different growth factors on the chondrogenic potential of human bone marrow stromal cells. Osteoarthritis Cartilage 2001;9 (supplA):S36–S40., _0 F- d. z5 O8 I

( V$ e+ _3 X# j2 uShea CM, Edgar CM, Einhorn TA et al. BMP treatment of C3H10T1/2 mesenchymal stem cells induces both chondrogenesis and osteogenesis. J Cell Biochem 2003;90:1112–1127.
/ d& D+ s' G# ^$ I- u) T5 O
6 l: m/ E2 ^% z0 P+ ?% lWozney JM. The bone morphogenetic protein family: multifunctional cellular regulators in the embryo and adult. Eur J Oral Sci 1998;106(suppl 1):160–166.
+ P% T# @; v  ]' L' C# l0 s1 p( [+ B5 n+ _7 P$ g$ f
Iwasaki M, Nakata K, Nakahara H et al. Transforming growth factor-beta 1 stimulates chondrogenesis and inhibits osteogenesis in high density culture of periosteum-derived cells. Endocrinology 1993;132:1603–1608.0 O/ p- T3 N* H, d
! c8 Q- W+ D- ?
Chimal-Monroy J, Diaz de Leon L. Differential effects of transforming growth factors beta 1, beta 2, beta 3 and beta 5 on chondrogenesis in mouse limb bud mesenchymal cells. Int J Dev Biol 1997;41:91–102.
8 ^6 z; L7 o2 t" T4 M9 r$ B4 h
Awad HA, Halvorsen YD, Gimble JM et al. Effects of transforming growth factor beta1 and dexamethasone on the growth and chondrogenic differentiation of adipose-derived stromal cells. Tissue Eng 2003;9:1301–1312.' J( t% e/ z: `. y$ Q
9 d* S. V, c' W- D1 P, Q4 I
Chen P, Carrington JL, Hammonds RG et al. Stimulation of chondrogenesis in limb bud mesoderm cells by recombinant human bone morphogenetic protein 2B (BMP-2B) and modulation by transforming growth factor beta 1 and beta 2. Exp Cell Res 1991;195:509–515.6 e$ B) C7 `2 Z' X

) ~0 }* H) E0 E& J/ @3 LJiang TX, Yi JR, Ying SY et al. Activin enhances chondrogenesis of limb bud cells: stimulation of precartilaginous mesenchymal condensations and expression of NCAM. Dev Biol 1993;155:545–557.
9 s$ I( }9 E2 |  T6 k8 J( \7 C+ m8 c$ \7 y- |9 M
Asahina I, Sampath TK, Nishimura I et al. Human osteogenic protein-1 induces both chondroblastic and osteoblastic differentiation of osteoprogenitor cells derived from newborn rat calvaria. J Cell Biol 1993;123:921–933.
' v5 I+ }6 ?/ q2 y6 M& h4 D6 g9 |/ z& E1 X* r
Spiro RC, Liu L, Heidaran MA et al. Inductive activity of recombinant human growth and differentiation factor-5. Biochem Soc Trans 2000;28:362–328.# ^. {6 C& Y8 A

2 L! R2 G! G& R$ X, xMassague J, Wotton D. Transcriptional control by the TGF-beta/Smad signaling system. EMBO J 2000;19:1745–1754.( X7 L, I5 a2 C; P4 o" a) r

5 _( ?9 ~5 R* Z4 M/ PTuli R, Seghatoleslami MR, Tuli S et al. p38 MAP kinase regulation of AP-2 binding in TGF-beta1-stimulated chondrogenesis of human trabecular bone-derived cells. Ann N Y Acad Sci 2002;961:172–177.
% B# |/ L  ]/ R9 |/ ~7 w4 M% X! m! A7 G, s' }
Zhou S, Eid K, Glowaacki J. Cooperation between TGF-? and Wnt pathways during chondrocyte and adipocyte differentiation of human marrow stromal cells. J Bone Miner Res 2004;19:463–470.
2 X1 M. o: @: n, ~; M$ H( f8 c3 s! t6 q& B
Wang XF, Lin HY, Ng-Eaton E et al. Expression cloning and characterization of the TGF-beta type III receptor. Cell 1991;67:797–805.+ A4 E# j1 [9 s" G
) ~) g+ S) u3 T1 T3 X+ k3 L- x! ~
Lopez-Casillas F, Payne HM, Andres JL et al. Betaglycan can act as a dual modulator of TGF-beta access to signaling receptors: mapping of ligand binding and GAG attachment sites. J Cell Biol 1994;124:557–568.- V' S5 }" @# s, P6 N3 c: Y1 l3 {
4 ^3 j/ n& Y% B1 V5 n( K
Ornitz DM, Marie PJ. FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease. Genes Dev 2002;16:1446–1465.
- ?, i: H1 e' w2 n6 Q- m, _6 ]3 B9 W* }8 K5 {# E. Y
Murakami S, Kan M, McKeehan WL et al. Up-regulation of the chondrogenic Sox9 gene by fibroblast growth factors is mediated by the mitogen-activated protein kinase pathway. Proc Natl Acad Sci U S A 2000;97:1113–1118.
" b  d! O& {- U. c. c) @6 v  M% |3 y7 `0 ?7 Q/ @  b6 }9 q
Naski MC, Ornitz DM. FGF signaling in skeletal development Front Biosci 1998;3:D781–D794.
: g2 y5 w+ _; Y: |3 a" t  X0 L# P$ E9 ^! _* J7 L. F
Chen L, Li C, Qiao W et al. A Ser(365) Cys mutation of fibroblast growth factor receptor 3 in mouse downregulates Ihh/PTHrP signals and causes severe achondroplasia. Hum Mol Genet 2001;10:457–465.- a9 B+ _) }# `, \3 U
2 a; @8 f, Q: T9 j( y
Makower AM, Wroblewski J, Pawlowski A. Effects of IGF-I, rGH, FGF, EGF and NCS on DNA-synthesis, cell proliferation and morphology of chondrocytes isolated from rat rib growth cartilage. Cell Biol Int Rep 1989;13:259–270.
" A: i- [( z  V/ r3 A* _: N* x1 x
1 O: V. @* d: y* C) I9 k- h; dFortier LA, Nixon AJ, Lust G. Phenotypic expression of equine articular chondrocytes grown in three-dimensional cultures supplemented with supraphysiologic concentrations of insulin-like growth factor-1. Am J Vet Res 2002;63:301–305.
0 N$ e5 t3 X9 D/ ~
4 Q9 |8 T- Q' qFrench MM, Rose S, Canseco J et al. Chondrogenic differentiation of adult dermal fibroblasts. Ann Biomed Eng 2004;32:50–56.
; V5 n; G/ k- q& b- L; j0 W2 k+ N* J/ h, g% C& R, |6 x
Hidaka K, Kanematsu T, Takeuchi H et al. Involvement of the phosphoinositide 3-kinase/protein kinase B signaling pathway in insulin/IGF-I-induced chondrogenesis of the mouse embryonal carcinoma-derived cell line ATDC5. Int J Biochem Cell Biol 2001;33:1094–1103.
( I3 _, ^  _9 F3 ^3 _1 D% Q& f0 U# {+ S  S0 j
Oh CD, Chun JS. Signaling mechanisms leading to the regulation of differentiation and apoptosis of articular chondrocytes by insulin-like growth factor-1. J Biol Chem 2003; 278:36563–36571.
/ }6 D+ q9 i1 \+ a) T
3 _3 B9 b; @, WFukumoto T, Sperling JW, Sanyal A et al. Combined effects of insulin-like growth factor-1 and transforming growth factor-beta1 on periosteal mesenchymal cells during chondrogenesis in vitro. Osteoarthritis Cartilage 2003;11:55–64.* l2 b6 z* d# P, }! J5 {

% r; N# a& d; fFrenz DA, Liu W, Williams JD et al. Induction of chondrogenesis: requirement for synergistic interaction of basic fibroblast growth factor and transforming growth factor-beta. Development 1994;120:415–424.) p: |; r1 R- N" P# K  x  ]" f* [

/ Z& h' A5 Z# d' G  R2 C7 J7 m& M0 O9 C$ HOgueta S, Munoz J, Obregon E et al. Prolactin is a component of the human synovial liquid and modulates the growth and chondrogenic differentiation of bone marrow-derived mesenchymal stem cells. Mol Cell Endocrinol 2002;190: 51–63.
/ z) j- O" N% g/ Q/ N4 F
3 F3 \5 _- ?. T+ {. c9 TTonon R, D’Andrea P. Interleukin-1beta increases the functional expression of connexin 43 in articular chondrocytes: evidence for a Ca2 -dependent mechanism. J Bone Miner Res 2000;15:1669–1677.
6 ]8 D5 O5 b0 b0 M9 b: O9 H* H
  ^4 i& t$ Q9 A) t8 b3 nWong M, Kireeva ML, Kolesnikova TV et al. Cyr61, product of a growth factor-inducible immediate-early gene, regulates chondrogenesis in mouse limb bud mesenchymal cells. Dev Biol 1997;192:492–508.8 B2 d/ ~0 Z" v9 J) h0 H5 W2 C

* d! e: l+ ?, V: T0 S7 T, }Dreyfus J, Brunet-de Carvalho N, Duprez D et al. HB-GAM/pleiotrophin but not RIHB/midkine enhances chondrogenesis in micromass culture. Exp Cell Res 1998;241: 171–180.
1 R6 n0 R: K! t6 [2 w. F: B" _* A+ ]% N+ {1 X
Tsukazaki T, Matsumoto T, Enomoto H et al. Growth hormone directly and indirectly stimulates articular chondrocyte cell growth. Osteoarthritis Cartilage 1994;2:259–267.) d: K7 I3 F7 ^+ N

2 g' {% l4 ~" z7 W7 s" xJohnstone B, Hering TM, Caplan AI et al. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp Cell Res 1998;238:265–272.
  I* t2 }3 e, B  [: H. f* g# T9 Q0 E' s) _7 I+ a8 [, w
Mackay AM, Beck SC, Murphy JM et al. Chondrogenic differentiation of cultured human mesenchymal stem cells from marrow. Tissue Eng 1998;4:415–428.
* {( K9 l2 x* T3 R8 K( h! V6 x: L' t$ c9 r9 Y8 @+ g& ?3 L
Wakita R, Izumi T, Itoman M. Thyroid hormone-induced chondrocyte terminal differentiation in rat femur organ culture. Cell Tissue Res 1998;293:357–364.
4 U1 M0 w  ]8 |. }6 h) o- R( [' D/ e; j. M  w
Siebler T, Robson H, Shalet SM et al. Dexamethasone inhibits and thyroid hormone promotes differentiation of mouse chondrogenic ATDC5 cells. Bone 2002;31:457–464.
% E. Z- \1 Z0 [3 h/ D$ Y* w
" L. G* C- A: g' k% XLocker M, Kellermann O, Boucquey M et al. Paracrine and autocrine signals promoting full chondrogenic differentiation of mesoblastic cell line. J Bone Miner Res 2004;19: 100–110.
3 ]4 \$ O% H4 F; u; T8 }4 [; [, D" {* i! ?; Y. G: t
Tsonis PA. 1,25-Dihydroxyvitamin D3 stimulates chondrogenesis of the chick limb bud mesenchymal cells. Dev Biol 1991;143:130–134.$ W) h: i$ B, J% n& N6 M$ ?
' \+ \2 e1 C3 Q+ p. _
Harmand MF, Thomasset M, Rouais F et al. In vitro stimulation of articular chondrocyte differentiated function by 1,25-dihydroxycholecalciferol or 24R,25-dihydroxychole-calciferol. J Cell Physiol 1984;119:359–365.
( S8 v" Y# Q" ?
& U, H" Q! L6 s7 T5 f  T% {Kosher RA, Walker KH. The effect of prostaglandins on in vitro limb cartilage differentiation. Exp Cell Res 1983; 145:145–153.
! p- c- }% W' }4 G, J3 ?" D! r: ?6 b7 r. Y# c! d7 e3 q" X7 s
Biddulph DM, Dozier MM, Capehart AA. Inhibition of prostaglandin synthesis reduces cyclic AMP levels and inhibits chondrogenesis in cultured chick limb mesenchyme. Methods Cell Sci 2000;22:9–16." U* N1 W7 P) z" U7 v2 m$ I$ H

  [) ~1 T/ C6 T3 Z" Z  D: dMiyamoto M, Ito H, Mukai S et al. Simultaneous stimulation of EP2 and EP4 is essential to the effect of prostaglandin E2 in chondrocyte differentiation. Osteoarthritis Cartilage 2003;11:644–652./ |' L0 K/ K6 x

- \# D2 ]9 I  |7 {" [; CFarquharson C, Berry JL, Mawer EB et al. Ascorbic acid-induced chondrocyte terminal differentiation: the role of the extracellular matrix and 1,25-dihydroxyvitamin D. Eur J Cell Biol 1998;76:110–118.
% y' T3 v& F: T: a
" F$ W& }" V4 j# m$ ~. cKulyk WM, Hoffman LM. Ethanol exposure stimulates cartilage differentiation by embryonic limb mesenchyme cells. Exp Cell Res 1996;223:290–300.  Q9 [: h9 j  Q
* j8 S6 G6 r% i8 v; q
Hoffman LM, Kulyk WM. Alcohol promotes in vitro chondrogenesis in embryonic facial mesenchyme. Int J Dev Biol 1999;43:167–174.
0 Z4 R# h% E: O+ z+ `
& a" D( A. v6 Z) _+ u% zKulyk WM. Promotion of embryonic limb cartilage differentiation in vitro by staurosporine, a protein kinase C inhibitor. Dev Biol 1991;146:38–48.' ^, S# `3 L5 {" w# Z4 j
' R- n' X8 F$ g) z
Kulyk WM, Reichert C. Staurosporine, a protein kinase inhibitor, stimulates cartilage differentiation by embryonic facial mesenchyme. J Craniofac Genet Dev Biol 1992; 12:90–97.
6 s: x, T% ~2 F: W* A8 a: K
9 g0 T' {. s3 }: J  J& Q- ?  sRevillion-Carette F, Desbiens X, Meunier L et al. Chondrogenesis in mouse limb buds in vitro: effects of dibutyryl cyclic AMP treatment. Differentiation 1986;33:121–129.
, K4 w& C; |7 U4 ^* x" {5 {6 ~" Z+ e4 E  r1 F# o4 p. U
Mikhailov AT, Gorgolyuk NA. Concanavalin A induces neural tissue and cartilage in amphibian early gastrula ectoderm. Cell Differ 1988;22:145–154.
+ n7 L% H# V6 t: T# t8 y
+ L0 L" O& f+ X) b$ u; xKato Y, Iwamoto M, Koike T et al. Effect of vanadate on cartilage-matrix proteoglycan synthesis in rabbit costal chondrocyte cultures. J Cell Biol 1987;104:311–319.
: f, E" D* Q) P2 ~# F
8 o  ]. f* R/ a( c% X" LNishigaki F, Sakuma S, Ogawa T et al. FK506 induces chondrogenic differentiation of clonal mouse embryonic carcinoma cells, ATDC5. Eur J Pharmacol 2002;437:123–128.
0 g: M! r" Q( u* X- K; V! R
5 c0 \' [& d8 D- _, ~, z6 PMuir H. The chondrocyte, architect of cartilage: biomechanics, structure, function and molecular biology of cartilage matrix macromolecules. Bioessays 1995;17:1039–1048.4 [3 y# |4 s% @: U( `
( G7 R, z8 D, y# e9 ~
Maroudas A, Bayliss MT, Venn MF. Further studies on the composition of human femoral head cartilage. Ann Rheum Dis 1980;39:514–523.0 E& I1 _( s# K1 M( Z+ B1 a5 ^

# ^8 t* a* e) |! j4 xHeinegard D, Oldberg A. Structure and biology of cartilage and bone matrix noncollagenous macromolecules. FASEB J 1989;3:2042–2051.: v4 @3 i% Z4 ~0 D! ], ^! O

2 c! p& ^/ R6 E6 O- }, {Neame PJ, Tapp H, Azizan A. Noncollagenous, nonproteoglycan macromolecules of cartilage. Cell Mol Life Sci 1999;55:1327–1340.% Y% C0 n) i- u& N1 @. W0 p+ f! i

/ Z# e' |- i! D9 D$ ?5 q- EKnudson CB. Hyaluronan and CD44: strategic players for cell-matrix interactions during chondrogenesis and matrix assembly. Birth Defects Res Part C Embryo Today 2003;69: 174–196.
" u5 H* {) l& s  o9 j/ Y9 k
+ s" _/ a9 V/ l2 x6 _- b& C; sEyre D. Collagen of articular cartilage. Arthritis Res 2002; 4:30–35.& }3 U% \) n6 b3 x$ [" H
; f+ Z5 ~# z# Q: {6 h5 S9 S" y' \
Knudson CB, Knudson W. Cartilage proteoglycans. Semin Cell Dev Biol 2001;12:69–78.
( x$ d- G8 B" p# u. H0 t! K( ~( H( P, x: W4 V2 k
Werb Z, Chin JR. Extracellular matrix remodeling during morphogenesis.Ann NYAcad Sci 1998;857:110–118.+ X* B; L  P& Y

0 L* P$ e' {& j+ S4 |# ?Gepstein A, Arbel G, Blumenfeld I et al. Association of metalloproteinases, tissue inhibitors of matrix metalloproteinases, and proteoglycans with development, aging, and osteoarthritis processes in mouse temporomandibular joint. Histochem Cell Biol 2003;120:23–32.+ l9 S' @0 S5 [3 n
% e& W* [, f: j
Krane SM. Elucidation of the potential roles of matrix metalloproteinases in skeletal biology. Arthritis Res Ther 2003;5:2–4.
0 @: y1 A9 m6 p2 y, E* ^$ l2 r% k( E+ ~
van der Kraan PM, Buma P, van Kuppevelt T et al. Interaction of chondrocytes, extracellular matrix and growth factors: relevance for articular cartilage tissue engineering. Osteoarthritis Cartilage 2002;10:631–637.
, G; h, Z9 }& W  C9 ^1 H
0 |9 [* J2 T% q  tFreed LE, Marquis JC, Nohria A et al. Cartilage formation in vitro and in vivo using cells cultured on synthetic biodegradable polymers. J Biomed Mater Res 1993;27:11–23.
6 O5 \8 m6 i! Y0 Q6 m% M; e8 c4 h' ^9 z' U: g
Malda J, van Blitterswijk CA, Grojec M et al. Expansion of bovine chondrocytes on microcarriers enhances redifferentiation. Tissue Eng 2003;9:939–948.+ z8 \8 h( v6 _  g% X4 O/ x

$ _4 `' e# z  n; HBoyan BD, Hummert TW, Dean DD et al. Role of material surfaces in regulating bone and cartilage cell response. Biomaterials 1996;17:137–146." N% S! A% O( ]* w
7 t7 a' C. Y, H0 U6 Y, _- W
Lee CR, Breinan HA, Nehrer S et al. Articular cartilage chondrocytes in type I and type II collagen-GAG matrices exhibit contractile behavior in vitro. Tissue Eng 2000;6: 555–565.
* z5 K9 b9 g1 C9 l8 t9 ~+ ]1 Q- m1 `0 v* j$ W3 X1 q! R3 F4 T# E7 S
Buma P, Pieper JS, van Tienen T et al. Cross-linked type I and type II collagenous matrices for the repair of full-thickness articular cartilage defects: a study in rabbits. Biomaterials 2003;24:3255–3263.
$ K  ]- L  R4 O
. _8 D1 ]1 p( T  U8 K, B1 xCherubino P, Grassi FA, Bulgheroni P et al. Autologous chondrocyte implantation using a bilayer collagen membrane: a preliminary report. J Orthop Surg (Hong Kong) 2003;11:10–15.2 K1 i' X. v/ J: s6 v2 N8 V: z
9 t. s: q# O, M5 p7 ~& o
Grigolo B, Roseti L, Fiorini M et al. Transplantation of chondrocytes seeded on a hyaluronan derivative (hyaff-11) into cartilage defects in rabbits. Biomaterials 2001;22: 2417–2424.' s" Q$ i+ t+ q5 c7 V% u; L, U' r2 O9 Z

- V! J( K: I1 j5 X  k3 wPavesio A, Abatangelo G, Borrione A et al. Hyaluronan-based scaffolds (Hyalograft C) in the treatment of knee cartilage defects: preliminary clinical findings. Novartis Found Symp 2003;249:203–217.. x! W6 x; J0 y3 c1 P: S2 S
4 z$ P5 ]  C0 _- g
Sechriest VF, Miao YJ, Niyibizi C et al. GAG-augmented polysaccharide hydrogel: a novel biocompatible and biodegradable material to support chondrogenesis. J Biomed Mater Res 2000;49:534–541.
" R! f; E/ s+ x+ e4 w- T9 k% i( M- M! J5 ]: i6 S
Li Q, Williams CG, Sun DD et al. Photocrosslinkable polysaccharides based on chondroitin sulfate. J Biomed Mater Res 2004;68A:28–33.
9 ^% T+ W6 g' ]7 G' F  B" L( V
8 O. H! x" O1 OCheung HS. In vitro cartilage formation on porous hydroxyapatite ceramic granules. In Vitro Cell Dev Biol 1985; 21:353–357.
' l+ a# _1 D; c, y1 ^8 ^3 h, Y2 b5 `( F- D) V9 ?5 a  j1 e8 E
Yan M, Dang G. Construction of porous hydroxyapatite (HA) block loaded with cultured chondrocytes. Zhonghua Wai Ke Za Zhi 1999;37:403–405.
4 N; M  b4 L# u" C# N* C: ]$ T3 r8 W7 i
Haisch A, Schultz O, Perka C et al. Tissue engineering of human cartilage tissue for reconstructive surgery using bio-compatible resorbable fibrin gel and polymer carriers. HNO 1996;44:624–629.
' g/ M( l9 A: J& C$ o; K
3 U$ O4 i, |$ G. BHunter CJ, Mouw JK, Levenston ME. Dynamic compression of chondrocyte-seeded fibrin gels: effects on matrix accumulation and mechanical stiffness. Osteoarthritis Cartilage 2004;12:117–130.* x1 W# a6 B+ _

9 K! N# b' o/ D! p  u: ?van Susante JL, Buma P, van Osch GJ et al. Culture of chondrocytes in alginate and collagen carrier gels. Acta Orthop Scand 1995;66:549–556.6 u: ^; ^8 G  \3 G

* }: O( K4 b, M( @3 K7 ?8 V4 u% yChang CH, Liu HC, Lin CC et al. Gelatin-chondroitin-hyaluronan tri-copolymer scaffold for cartilage tissue engineering. Biomaterials 2003;24:4853–4858.. l$ o9 X8 {8 V8 Q4 k' _
7 }+ q; o$ J1 \. }
Ibusuki S, Fujii Y, Iwamoto Y et al. Tissue-engineered cartilage using an injectable and in situ gelable thermoresponsive gelatin: fabrication and in vitro performance. Tissue Eng 2003;9:371–384.
5 Q8 u& K* s7 Z( K2 D8 I* C5 A0 `. w# [  q6 i: ~
Elder SH, Nettles DL, Bumgardner JD. Synthesis and characterization of chitosan scaffolds for cartilage-tissue engineering. Methods Mol Biol 2004;238:41–48.- a$ y( E9 L: I' _% A

4 M+ q- k: g; v/ n. ]* i' ~6 MChang SC, Tobias G, Roy AK et al. Tissue engineering of autologous cartilage for craniofacial reconstruction by injection molding. Plast Reconstr Surg 2003;112:793–799.
) }& l3 W# I) C9 r+ S. r  Z" l0 Q8 s
6 I4 r4 H2 U1 a  m: a8 vKim G, Okumura M, Bosnakovski D et al. Effects of ascorbic acid on proliferation and biological properties of bovine chondrocytes in alginate beads. Jpn J Vet Res 2003;51:83–94.. _* S6 ]  W% H
! U' b* h. T" A& I6 I
Masuda K, Sah RL, Hejna MJ et al. A novel two-step method for the formation of tissue-engineered cartilage by mature bovine chondrocytes: the alginate-recovered-chondrocyte (ARC) method. J Orthop Res 2003;21:139–148.6 f5 a' o* b# m' \
# x$ ^: i8 j3 n8 T& V1 H
Chia SH, Schumacher BL, Klein TJ et al. Tissue-engineered human nasal septal cartilage using the alginate-recovered-chondrocyte method. Laryngoscope 2004;114:38–45.( o0 v/ V8 \7 P" z7 J8 Y

. j, \& z$ M" i* i( PGiurea A, Klein TJ, Chen AC et al. Adhesion of perichondrial cells to a polylactic acid scaffold. J Orthop Res 2003;21:584–589.
& V1 h: b" m4 ]
# q8 }% f9 \( V4 SHomicz MR, Chia SH, Schumacher BL et al. Human septal chondrocyte redifferentiation in alginate, polyglycolic acid scaffold, and monolayer culture. Laryngoscope 2003;113: 25–32.- V. N# D7 S$ d" X4 O
+ t. b1 S! v3 X6 @" F, p
Ma Z, Gao C, Gong Y et al. Paraffin spheres as porogen to fabricate poly(L-lactic acid) scaffolds with improved cyto-compatibility for cartilage tissue engineering. J Biomed Mater Res 2003;67:610–617.8 c7 C  j6 I- ~' E# B3 F! P! \

& x1 j3 C% `8 }; _Moran JM, Pazzano D, Bonassar LJ. Characterization of polylactic acid-polyglycolic acid composites for cartilage tissue engineering. Tissue Eng 2003;9:63–70.
, z) K: k% y+ Q$ Y5 b8 }, C1 _. r, ~
Lee WK, Ichi T, Ooya T et al. Novel poly(ethylene glycol) scaffolds crosslinked by hydrolyzable polyrotaxane for cartilage tissue engineering. J Biomed Mater Res 2003;67: 1087–1092.
5 o( z6 f8 z6 Q4 m1 i: W! O
& g/ h& T7 o2 M# d9 |# H$ b# BLi WJ, Danielson KG, Alexander PG et al. Biological response of chondrocytes cultured in three-dimensional nanofibrous poly(epsilon-caprolactone) scaffolds. J Biomed Mater Res 2003;67A:1105–1114.  H7 C+ c  v" q. E6 n5 `$ g) a

: N9 p0 k, K* r2 d- Q% bEriksson C. Bone morphogenesis and surface charge. Clin Orthop 1976:295–302.
/ x7 z% G4 y" V( G- S5 X$ f
6 `2 e0 U9 a- K9 g5 v$ P  WMaroudas NG. Sulphonated polystyrene as an optimal substratum for the adhesion and spreading of mesenchymal cells in monovalent and divalent saline solutions. J Cell Physiol 1977;90:511–519.
( [$ ^! P- g. q5 y1 C# Y
. P* {3 P2 M% F% E. mVan Damme MP, Sinnaya P, Derry K et al. Temporal changes in charge content of cultured chondrocytes from bovine cartilaginous tissues. Matrix Biol 1977;15:495–502.( z  u7 ^; H( I

# i. j6 _) a9 J! Q: J& nSchwartz Z, Martin JY, Dean DD et al. Effect of titanium surface roughness on chondrocyte proliferation, matrix production, and differentiation depends on the state of cell maturation. J Biomed Mater Res 1996;30:145–155.: q! ]( x0 d% |' b

' x7 U% t; m# \  J, ^Boyan BD, Lincks J, Lohmann CH et al. Effect of surface roughness and composition on costochondral chondrocytes is dependent on cell maturation state. J Orthop Res 1999; 17:446–457., A$ T9 T# t  B6 F# v; U  x' x# d
- ^# v* ~$ p# ], h+ j" w+ p" f. B
Bhardwaj T, Pilliar RM, Grynpas MD et al. Effect of material geometry on cartilagenous tissue formation in vitro. J Biomed Mater Res 2001;57:190–199.
4 ~0 `9 N& ?& M8 r
3 q. k0 Q8 L. I. _van Susante JLC, Pieper J, Buma P et al. Linkage of chon-droitin-sulfate to type I collagen scaffolds stimulates the bioactivity of seeded chondrocytes in vitro. Biomaterials 2001;22:2359–2369.; E: i: e9 D/ Z3 S* A

6 l' l* E7 h! @, `9 r5 `7 mYaylaoglu MB, Yildiz C, Korkusuz F et al. A novel osteochondral implant. Biomaterials 1999;20:1513–1520.
0 {$ f3 Q- i, e% l; \2 B( f. j/ `5 l' c* p9 G$ L/ I6 I
Watt FM, Dudhia J. Prolonged expression of differentiated phenotype by chondrocytes cultured at low density on a composite substrate of collagen and agarose that restricts cell spreading. Differentiation 1988;38:140–147.
( X% b& {1 p  k, G: E6 x4 m1 @$ V$ k' y% h' f
Thomas JT, Grant ME. Cartilage proteoglycan aggregate and fibronectin can modulate the expression of type X collagen by embryonic chick chondrocytes cultured in collagen gels. Biosci Rep 1988;8:163–171.
. [( {* u+ K' W: d  w7 ?) d& }: f; W4 H' T* ^' D) Q+ e9 ]
Mizuno S, Glowacki J. Three-dimensional composite of demineralized bone powder and collagen for in vitro analysis of chondroinduction of human dermal fibroblasts. Biomaterials 1996;17:1819–1825.
* A0 {) {: ?# [9 x6 }. ]4 A2 B- X3 l2 R& S* q( ~
Taguchi T, Ikoma T, Tanaka J. An improved method to prepare hyaluronic acid and type II collagen composite matrices. J Biomed Mater Res 2002;61:330–336.! @- C2 q  v% J0 M' Y3 @/ `
9 K& k8 G+ d4 H- J4 S! U
Sherwood JK, Riley SL, Palazzolo R et al. A three-dimensional osteochondral composite scaffold for articular cartilage repair. Biomaterials 2002;23:4739–4751.; i! y( T! Q" S4 X3 n
2 G1 f5 L& I/ v" b( f
Chen G, Sato T, Ushida T et al. The use of a novel PLGA fiber/collagen composite web as a scaffold for engineering of articular cartilage tissue with adjustable thickness. J Biomed Mater Res 2003;67:1170–1180.# D/ |5 c6 }3 }5 T  U4 E

" K9 K6 m$ L2 J* ]Aigner J, Tegeler J, Hutzler P et al. Cartilage tissue engineering with novel nonwoven structured biomaterial based on hyaluronic acid benzyl ester. J Biomed Mater Res 1998; 42:172–181.
  d  b0 r) u& L$ c2 _" O, w4 d  C9 T' _
Girotto D, Urbani S, Brun P et al. Tissue-specific gene expression in chondrocytes grown on three-dimensional hyaluronic acid scaffolds. Biomaterials 2003;24:3265–3275.
- M& p  a) s* ]5 [
0 |. M9 u1 G  r9 l: u" X. lvan Lent PL, Blom AB, van der Kraan P et al. Crucial role of synovial lining macrophages in the promotion of transforming growth factor beta-mediated osteophyte formation. Arthritis Rheum 2004;50:103–111.
, r, y# W# {3 s
6 G; ^. ]6 m8 }1 U" HWong M, Tuan RS. Interactive cellular modulation of chondrogenic differentiation in vitro by subpopulations of chick embryonic calvarial cells. Dev Biol 1995;167:130–147.; T3 B3 d* r$ f9 q& G, \  }
& ~5 b' c1 B# \- l
Stewart, R., Przyborski, S. Non-neural adult stem cells: tools for brain repair? Bioessays 2002;24:708–713.
6 s! F: B( l: R: D# h* L4 l; F# x# j' D+ R4 A$ f$ K7 t8 p( c1 e
Herzenberg LA, Parks D, Sahaf B, et al. The history and future of the fluorescence activated cell sorter and flow cytometry: a view from Stanford. Clin Chem 2002;48: 1819–1827." ~  \$ |! N: r$ q" I8 @

0 a9 D* d) n4 ?Siegel DL. Selecting antibodies to cell-surface antigens using magnetic sorting techniques. Methods Mol Biol 2002;178:219–226.
, m4 X+ ^" T/ R
1 ?. U! S+ X0 r  |) w) }; DElder SH. Conditioned medium of mechanically compressed chick limb bud cells promotes chondrocyte differentiation. J Orthop Sci 2002;7:538–543.
0 d  g5 L& d( N, P5 p; Z/ t. V% p4 `% h3 x9 E
Di Nino DL, Long F, Linsenmayer TF. Regulation of endochondral cartilage growth in the developing avian limb: cooperative involvement of perichondrium and periosteum. Dev Biol 2001;240:433–442.
/ C6 w( G9 f( _0 S& I3 T  I; V% I2 q2 n% y/ y' K$ Q
Solursh M, Meier S. A conditioned medium (CM) factor produced by chondrocytes that promotes their own differentiation. Dev Biol 1973;30:279–289.# ?) X( C9 x& \3 j1 \+ `
6 v2 |; P& m. E& Q  S& J. G
Solursh M, Reiter RS. The enhancement of in vitro survival and chondrogenesis of limb bud cells by cartilage conditioned medium. Dev Biol 1975;44:278–287.
) x) H+ `3 ^  o1 O1 Q- L4 e' o2 q( Y3 H. H% I8 k0 p7 p  x2 }) B% T3 N7 y+ O
Shen V, Rifas L, Kohler G et al. Fetal rat chondrocytes sequentially elaborate separate growth- and differentiation-promoting peptides during their development in vitro. Endocrinology 1985;116:920–925.9 x2 M( ^. L/ K7 N' J& T9 g
0 p" G+ w" x4 X# }4 H1 ^
Johnson LF, deSerres S, Herzog SR et al. Antigenic cross-reactivity between media supplements for cultured keratinocyte grafts. J Burn Care Rehabil 1991;12:306–312.) @: J1 t1 d' ?% [, X0 I& R

& t$ X% n; r7 F4 [3 dHodgetts SI, Beilharz MW, Scalzo AA et al. Why do cultured transplanted myoblasts die in vivo? DNA quantification shows enhanced survival of donor male myoblasts in host mice depleted of CD4  and CD8  cells or Nk1.1  cells. Cell Transplant 2000;9:489–502.+ A  M9 }2 l' g9 ~2 Z- ~5 x* {

7 y. ~) D3 o- I( w* K9 k6 [Smythe GM, Grounds MD. Exposure to tissue culture conditions can adversely affect myoblast behavior in vivo in whole muscle grafts: implications for myoblast transfer therapy. Cell Transplant 2000;9:379–393.
- l& I) l- C. {: K! J. x# H. r' w: |/ C5 i2 O' |! I/ d
de Crombrugghe B, Lefebvre V, Behringer RR et al. Transcriptional mechanisms of chondrocyte differentiation. Matrix Biol 2000;19:389–394.
6 v7 W5 p2 D) v
8 U( i0 @& b' @% W& `9 {) S. W" tFernandez-Lloris R, Vinals F, Lopez-Rovira T et al. Induction of the Sry-related factor SOX6 contributes to bone morphogenetic protein-2-induced chondroblastic differentiation of C3H10T1/2 cells. Mol Endocrinol 2003;17:1332–1343.
; r  d! v! @# s# X4 u9 u4 {0 A7 [; d5 D( A  \9 t, n
Chimal-Monroy J, Rodriguez-Leon J, Montero JA et al. Analysis of the molecular cascade responsible for mesodermal limb chondrogenesis: Sox genes and BMP signaling. Dev Biol 2003;257:292–301.
1 h* n& r! m8 j3 m- Y9 O! K& {% k" _
Ng LJ, Wheatley S, Muscat GE et al. SOX9 binds DNA, activates transcription, and coexpresses with type II collagen during chondrogenesis in the mouse. Dev Biol 1997; 183:108–121.4 M: {/ V1 r  T: _2 `9 E& q8 \  g8 J
* V& p* \! v3 u5 V, w- t- R
Mori-Akiyama Y, Akiyama H, Rowitch DH et al. Sox9 is required for determination of the chondrogenic cell lineage in the cranial neural crest. Proc Natl Acad Sci U S A 2003;100:9360–9365.+ p! s! `9 D: N0 E

$ P2 k( R1 s! R* P- X! t/ PHuang W, Lu N, Eberspaecher H, De Crombrugghe B. A new long form of c-Maf cooperates with Sox9 to activate the type II collagen gene. J Biol Chem 2002;277:50668–50675.9 Z+ p3 F$ d3 G5 j1 i

" L& a( Q7 w. V$ H( J% C& WTsuda M, Takahashi S, Takahashi Y et al. Transcriptional co-activators CREB-binding protein and p300 regulate chondrocyte-specific gene expression via association with Sox9. J Biol Chem 2003;278:27224–27229.# O: q# H9 U# i# t+ K
# D" r* R1 P* Y
Murtaugh LC, Zeng L, Chyung JH et al. The chick transcriptional repressor Nkx3.2 acts downstream of Shh to promote BMP-dependent axial chondrogenesis. Dev Cell 2001;1: 411–422.
( Z/ l9 p. ^( J" @. M9 q! G
! {4 E- C; K# ^3 @; IZeng L, Kempf H, Murtaugh LC et al. Shh establishes an Nkx3.2/Sox9 autoregulatory loop that is maintained by BMP signals to induce somitic chondrogenesis. Genes Dev 2002;16:1990–2005.2 R4 B& w# K1 S4 q, L5 U! J! @" a

, U% C9 i6 V4 w" I: H- y" _0 TRodrigo I, Hill RE, Balling R et al. Pax1 and Pax9 activate Bapx1 to induce chondrogenic differentiation in the sclerotome. Development 2003;130:473–482.
7 G, E0 n2 Y: A' |8 z* K- T$ P6 @
+ W9 v" @" I  Z* p$ o! IShen M, Yoshida E, Yan W et al. Basic helix-loop-helix protein DEC1 promotes chondrocyte differentiation at the early and terminal stages. J Biol Chem 2002;277:50112–50120., o! w, ^9 n& B% C$ x
2 f' @+ H& E$ X0 ^
Hoffmann A, Czichos S, Kaps C et al. The T-box transcription factor Brachyury mediates cartilage development in mesenchymal stem cell line C3H10T1/2. J Cell Sci 2002; 115:769–781.+ j* T/ O2 L1 P( n

8 x3 J( H+ B1 QHatakeyama Y, Nguyen J, Wang X et al. Smad signaling in mesenchymal and chondroprogenitor cells. J Bone Joint Surg Am 2003;85(suppl 3):13–18.* l8 s# ^/ Y8 y* B

+ c3 h$ _0 {9 [" k; K( LTsuchiya H, Kitoh H, Sugiura F et al. Chondrogenesis enhanced by overexpression of sox9 gene in mouse bone marrow-derived mesenchymal stem cells. Biochem Bio-phys Res Commun 2003; 301:338–343.
- t8 I* v$ H) n# i* X* J; Q. O4 g6 u0 L5 H4 s! }/ |6 {! ]9 Y: L3 M8 @
Madry H, Zurakowski D, Trippel SB. Overexpression of human insulin-like growth factor-I promotes new tissue formation in an ex vivo model of articular chondrocyte transplantation. Gene Ther 2001;8:1443–1449.
. _! c& \' s$ \7 t' G8 |- w9 p3 H( f* O8 O3 E* d
Madry H, Padera R, Seidel J et al. Gene transfer of a human insulin-like growth factor I cDNA enhances tissue engineering of cartilage. Hum Gene Ther 2002;13:1621–1630.- V4 p* X$ G8 S" R- b

4 d( w! b% [9 R" LWang WG, Lou SQ, Ju XD et al. In vitro chondrogenesis of human bone marrow-derived mesenchymal progenitor cells in monolayer culture: activation by transfection with TGF-beta2. Tissue Cell 2003;35:69–77.+ K2 X+ u2 X" [: R2 M
3 ]; o8 b* R; [1 p5 ?
Ohta S, Muramatsu H, Senda T et al. Midkine is expressed during repari of bone fracture and promotes chongrogenesis. J Bone Miner Re 1999;14:1132–1144.
* D- z3 c9 Z; U  O4 B* c# J2 ?- f% Y  o6 w1 O8 W
Widelitz RB, Jiang TX, Murray BA et al. Adhesion molecules in skeletogenesis, II: neural cell adhesion molecules mediate precartilaginous mesenchymal condensations and enhance chondrogenesis. J Cell Physiol 1993;156:399–411.5 }- C3 I7 {  R' s9 z

; y$ _# z3 n' R  s  f+ Y/ t% kFischer L, Boland G, Tuan RS. Wnt signaling during BMP-2 stimulation of mesenchymal chondrogenesis. J Cell Biochem 2002;84:816–831.# V1 r) r% {" \! v) l; X8 L
& C+ ~1 {5 M2 ^" `; M; C- p( y4 \
Shimizu-Sato S, Huq E, Tepperman JM et al. A light-switchable gene promoter system. Nat Biotechnol 2002;20: 1041–1044.
, E' q; }5 `8 _! ]$ g2 C! o$ W5 _+ l5 q/ C1 M& \8 q  Z, l5 O) D
Conget PA, Minguell JJ. Adenoviral-mediated gene transfer into ex vivo expanded human bone marrow mesenchymal progenitor cells. Exp Hematol 2000;28:382–390.$ o% K: m% [. M1 w

1 D$ w% g' z. x2 @Pawelek JM. Effects of thyroxine and low oxygen tension on chondrogenic expression in cell culture. Dev Biol 1969;19:52–72.
6 C* x* o) Z. T& `$ l/ Y8 H' D  Y3 u  p% Y
O’Driscoll SW, Fitzsimmons JS, Commisso CN. Role of oxygen tension during cartilage formation by periosteum. J Orthop Res 1997;15:682–687.  b. c( M2 H. a

# d1 k3 p$ p4 iBrighton CT, Jensen L, Pollack SR et al. Proliferative and synthetic response of bovine growth plate chondrocytes to various capacitively coupled electrical fields. J Orthop Res 1989;7:759–765.
! Q# y. j7 T/ t* v& f0 [% K8 o" `) b& j9 p
Ciombor DM, Lester G, Aaron RK et al. Low frequency EMF regulates chondrocyte differentiation and expression of matrix proteins. J Orthop Res 2002;20:40–50.
! B. k; b) }  s/ J) I9 m$ J" L% c6 B' j+ ]" I" V) d3 X
Waldman SD, Spiteri CG, Grynpas MD et al. Effect of biomechanical conditioning on cartilaginous tissue formation in vitro. J Bone Joint Surg Am 2003;85(suppl 2):101–105.8 K" q/ l' Y: V1 q! ?

$ T# V' B; w7 h7 [: e$ }Hojo T, Fujioka M, Otsuka G et al. Effect of heat stimulation on viability and proteoglycan metabolism of cultured chondrocytes: preliminary report. J Orthop Sci 2003;8:396–399.
+ Z) y( _( v$ ]5 X  g+ w7 u  U3 F
Zhang W, Green C, Stott NS. Bone morphogenetic protein-2 modulation of chondrogenic differentiation in vitro involves gap junction-mediated intercellular communication. J Cell Physiol 2002;193:233–243.9 |: j- y9 g  k
. a" o# O& t- d. l1 T% J
Torricelli P, Giavaresi G, Fini M et al. Laser biostimulation of cartilage: in vitro evaluation. Biomed Pharmacother 2001;55:117–120.
. @9 v: r5 v3 |" W% g+ ^+ l
4 n- C/ b; \8 n5 [/ T# n" WCoelho CN, Kosher RA. Gap junctional communication during limb cartilage differentiation. Dev Biol 1991;144: 47–53.
5 N: y+ G' m3 n- m; n9 K5 U( k$ r% R
Zimmermann B, Scharlach E, Kaatz R. Cell contact and surface coat alterations of limb-bud mesenchymal cells during differentiation. J Embryol Exp Morphol 1981;72:1–18.6 N/ E3 ?3 H1 n; A9 S. k
% {" ]& ?# v; e: R0 Z' H1 z3 n
Zimmermann B. Assembly and disassembly of gap junctions during mesenchymal cell condensation and early chondrogenesis in limb buds of mouse embryos. J Anat 1984;138:351–363.* Z5 S4 L# }  L5 X
( I3 x: _. i& e2 e5 e( p
Tonon R, D’Andrea P. The functional expression of connexin 43 in articular chondrocytes is increased by interleukin 1beta: evidence for a Ca2 -dependent mechanism. Biorheology 2002;39:153–160.
5 s0 |5 Q8 q' x" C
4 F% B' u" w/ |/ Z7 L/ Q1 r" ~0 `Zhang ZJ, Huckle J, Francomano CA et al. The effects of pulsed low-intensity ultrasound on chondrocyte viability, proliferation, gene expression and matrix production. Ultrasound Med Biol 2003;29:1645–1651.
9 H3 r0 I) _5 T% {* H( @
; X$ o' A7 o1 y. G, g* YElfervig MK, Graff RD, Lee GM et al. ATP induces Ca2  signaling in human chondrons cultured in three-dimensional agarose films. Osteoarthritis Cartilage 2001;9:518–526.. A& L" h$ R/ E2 l
, v. X3 |) A7 h3 c3 g" s/ L- }
H?kelien AM, Landsverk HB, Robl JM et al. Reprogramming fibroblasts to express T-cell functions using cell extracts. Nat Biotechnol 2002;20:460–466.+ I' Y& N7 U6 ?7 E1 ^$ v. }. ^

- I$ y; ?5 P- {1 q1 q& R0 [H?kelien AM, Collas P. Novel approaches to transdifferentiation. Cloning Stem Cells 2002;4:379–387." o( k9 N; r# p  D* c

3 _  i8 z) w' M. qGoldman RD, Pollack R, Hopkins NH. Preservation of normal behavior by enucleated cells in culture. Proc Natl Acad Sci U S A 1973;70:750–754.6 r3 _4 r, d- Q8 ?

3 a$ y( o5 n- L1 ?( D$ L% p: l3 WWigler MH, Weinstein IB. A preparative method for obtaining enucleated mammalian cells. Biochem Biophys Res Commun 1975;63:669–674.
" S" v* W: Z+ N0 m) |1 M1 b, P6 k. i% N2 F! e5 h
Stocco DM Rapid, quantitative isolation of mitochondria from rat liver using Ficoll gradients in vertical rotors. Anal Biochem 1983;131:453–457.
# z3 ]) \% m  ^; s0 f
) p$ y( t8 S0 h& s7 v# {Sligh JE, Levy SE, Waymire KG et al. Maternal germ-line transmission of mutant mtDNAs from embryonic stem cell-derived chimeric mice. Proc Natl Acad Sci U S A 2000;97: 14461–14466.1 G" [4 U2 y' H2 b$ W

! `/ ?3 e. v6 X) v3 EDavidson RL, Gerald PS. Induction of mammalian somatic cell hybridization by polyethylene glycol. Methods Cell Biol 1977;15:325–238.
% [0 o5 ~; G7 N- ]5 t. F
, Y/ J* H, z. j; {2 ?Lucas JJ, Kates JR. The construction of viable nuclear-cytoplasmic hybrid cells by nuclear transplantation. Cell 1976;7:397–405.
; m' w7 g( D0 s1 t" h) p
& I& z) J  @' bIwakura Y, Nozaki M, Asano M et al. Pleiotropic phenotypic expression in cybrids derived from mouse teratocarcinoma cells fused with rat myoblast cytoplasts. Cell 1985;43:777–791.
& K2 p% z% H$ H
; t- ?, c2 J+ l; Q& g" }' c5 cTosu M, Terasaki T, Iwakura Y et al. Clonal isolation and characterization of myoblast-like reconstituted cells formed by fusion of karyoplasts from mouse teratocarcinoma cells with rat myoblast cytoplasts. Cell Struct Funct 1988;13:249–266.( ]5 O" p# s( m( k

, }- i- y7 B3 W% {' h# {Watanabe T, Nomura S, Oishi M. Induction of erythroid differentiation by cytoplast fusion in mouse erythroleukemia (Friend) cells. Exp Cell Res 1985;159:224–234.
. i, `% {' ~  R% K3 y: D7 _: D( ?9 V
Watanabe T, Nomura S, Kaneko T et al. Cytoplasmic factors involved in erythroid differentiation in mouse erythroleukemia (MEL) cells. Cell Differ Dev 1988;25(suppl): 105–109
( M' V3 G5 ]+ U4 _  Y! f8 c- F2 F" a& A1 |, S
Okazaki T, Kato Y, Tashima M et al. Evidence of intracellular and trans-acting differentiation-inducing activity in human promyelocytic leukemia HL-60 cells: its possible involvement in process of cell differentiation from a commitment step to a phenotype-expression step. J Cell Physiol 1988;134:261–268.* f, @0 k+ [' a% A5 Q$ b
, P8 ?8 x/ {9 Y
Martin I, Vunjak-Novakovic G, Yang J et al. Mammalian chondrocytes expanded in the presence of fibroblast growth factor 2 maintain the ability to differentiate and regenerate three-dimensional cartilaginous tissue. Exp Cell Res 1999; 253:681–688.(Boon Chin Heng, Tong Cao,)

Rank: 2

积分
162 
威望
162  
包包
1746  
沙发
发表于 2015-5-31 20:10 |只看该作者
偶真幸运哦...  

Rank: 2

积分
79 
威望
79  
包包
1769  
藤椅
发表于 2015-7-14 19:44 |只看该作者
看看..  

Rank: 2

积分
72 
威望
72  
包包
1859  
板凳
发表于 2015-7-30 11:43 |只看该作者
干细胞之家微信公众号
挺好啊  

Rank: 2

积分
162 
威望
162  
包包
1746  
报纸
发表于 2015-8-10 20:30 |只看该作者
彪悍的人生不需要解释。  

Rank: 2

积分
104 
威望
104  
包包
1772  
地板
发表于 2015-8-17 14:35 |只看该作者
一个有信念者所开发出的力量,大于99个只有兴趣者。  

Rank: 2

积分
56 
威望
56  
包包
1853  
7
发表于 2015-9-4 11:43 |只看该作者
不错,看看。  

Rank: 2

积分
77 
威望
77  
包包
1730  
8
发表于 2015-9-9 16:35 |只看该作者
一个有信念者所开发出的力量,大于99个只有兴趣者。  

Rank: 2

积分
75 
威望
75  
包包
2193  
9
发表于 2015-9-17 19:53 |只看该作者
活着,以死的姿态……  

Rank: 2

积分
76 
威望
76  
包包
1772  
10
发表于 2015-9-18 07:41 |只看该作者
进行溜达一下  
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-25 15:55

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.