Nothing Special   »   [go: up one dir, main page]

Academia.eduAcademia.edu
938 REVIEW ARTICLE Journal of Regeneration of Articular Cartilage by Adipose Tissue Derived Mesenchymal Stem Cells: Perspectives From Stem Cell Biology and Molecular Medicine LING WU,1,2 XIAOXIAO CAI,1 SHU ZHANG,1 MARCEL KARPERIEN,2 AND Cellular Physiology YUNFENG LIN1* 1 State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, P.R. China 2 Department of Developmental BioEngineering, MIRA-Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, the Netherlands Adipose-derived stem cells (ASCs) have been discovered for more than a decade. Due to the large numbers of cells that can be harvested with relatively little donor morbidity, they are considered to be an attractive alternative to bone marrow derived mesenchymal stem cells. Consequently, isolation and differentiation of ASCs draw great attention in the research of tissue engineering and regenerative medicine. Cartilage defects cause big therapeutic problems because of their low self-repair capacity. Application of ASCs in cartilage regeneration gives hope to treat cartilage defects with autologous stem cells. In recent years, a lot of studies have been performed to test the possibility of using ASCs to re-construct damaged cartilage tissue. In this article, we have reviewed the most up-to-date articles utilizing ASCs for cartilage regeneration in basic and translational research. Our topic covers differentiation of adipose tissue derived mesenchymal stem cells into chondrocytes, increased cartilage formation by co-culture of ASCs with chondrocytes and enhancing chondrogenic differentiation of ASCs by gene manipulation. J. Cell. Physiol. 228: 938–944, 2013. ß 2012 Wiley Periodicals, Inc. Cartilage defects due to trauma, tumor ablation, or age-related abrasion, lead to constant pain and functional limitations of joints and cause serious medical and social problems. It is believed that even small lesions can severely affect the structure and function of articular cartilage and may predispose to the development of osteoarthritis (Alford and Cole, 2005a). The reason for this is quite obvious: no vascularization is present in articular cartilage tissues. Therefore, normal events in tissue repair like inflammation and fibrin clot formation do not happen in cartilage defects. Only chondrocyte and synoviocytes which reside in the local environment can fill up the defects by slow proliferation and matrix deposition (Mankin, 1982; Vincent et al., 2002). In cartilage defects deep into the subchondral bone, bone marrow cells as well as blood cells can migrate to the articular surface by bleeding to fill the gaps with rapid proliferation and matrix synthesis (Furukawa et al., 1980). However, the newly synthesized matrix is usually fibrous. And fibrous cartilage is inferior to hyaline cartilage in mechanical properties (Nehrer et al., 1999). Troubled by the poor selfregeneration of cartilage tissue, clinicians and basic scientists have been working for years on new techniques to find the perfect treatment for cartilage defects. The most popular treatments for cartilage defects nowadays, are micro-drilling and autologous chondrocytes implantation (ACI). In the micro-drilling technique also known as microfracturing, tiny fractures are induced into the subchondral bone plate by drilling small holes which allow blood and bone marrow to seep out in the defect. This creates a blood clot with incorporated pluripotent mesenchymal stem cells (MSCs). These MSCs eventually heal the defect with scar tissue consisting of a mixture of fibrous tissue, fibrocartilage, and hyaline-like cartilage (Gilbert, 1998). Regarding the clinical outcome, improvements in joint function and pain relief have been reported in 75% of young patients, with even higher success rates in young athletes (Sledge, 2001). However, the quality of the newly formed cartilage is generally out of control, ß 2 0 1 2 W I L E Y P E R I O D I C A L S , I N C . since it may depend on various factors including the gender and age of the patients, the size and location of the defects, the surgical protocols used, and the post-surgery rehabilitation (Alford and Cole, 2005b). In addition, the mechanical properties of scar tissue are inferior compared to native cartilage which may predispose the defected joint to early onset osteoarthritis in the medium to long run. Another treatment called ACI was first introduced by Brittberg et al. (1994). The rational behind ACI is to fill the cartilage defects with autologous chondrocytes which are expanded in vitro. The classical procedure includes arthroscopic excision of biopsies from low-weight bearing areas of healthy cartilage, isolation and expansion of chondrocytes in the laboratory, and implantation of chondrocyte suspension into the defects which is then covered by a periosteal flap sutured to the surrounding healthy tissues. Nowadays, new technique called matrix-induced autologous chondrocyte implantation (MACI) is becoming more popular. Instead of injection into defects as cell suspension, chondrocytes were seeded on a bilayer of porcine-derived type I/type III collagen, after in vitro expansion. The MACI membrane is then secured directly to the defect by fibrin glue without a cover (Bartlett et al., 2005). Clinical studies with a The authors have declared that no competing interests exist. *Correspondence to: Yunfeng Lin, State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, P.R. China. E-mail: yunfenglin@scu.edu.cn Manuscript Received: 26 May 2012 Manuscript Accepted: 27 September 2012 Accepted manuscript online in Wiley Online Library (wileyonlinelibrary.com): 5 October 2012. DOI: 10.1002/jcp.24255 REGENERATION OF ARTICULAR CARTILAGE follow-up period of 2–10 years indicated that 90% of treated patients developed well-integrated tissue in the defect sites (Peterson et al., 2003). Despite the success of ACI in clinical practice, there are some drawbacks of this therapeutic method that limit its broader application. One major issue is that the success rate of the procedure severely drops with age limiting the application of ACI to patients under the age of 50 years. Other drawbacks include expensive surgical procedures, donor site morbidity, and dedifferentiation of chondrocytes during in vitro expansion. In vitro expansion is required since relatively large quantities of healthy chondrocytes from the patient are required to fill up the defect site. Replacement of chondrocytes with other cell sources like stem cells gives hope to tackle this problem. Differentiation of Adipose Tissue Derived Mesenchymal Stem Cells Into Chondrocytes Adipose tissue, like bone marrow, is derived from the embryonic mesenchyme and contains a stroma that can be easily isolated. It was first reported in 2001, that a group of multipotent cells can be isolated from the stromal vascular fraction (SVF) of collagenase digested human adipose tissue (Zuk et al., 2001). These cells called adipose tissue-derived stromal cells or adipose stem cells (ASCs) can differentiate into adipocytes, osteoblasts, chondrocytes, and myocytes under specific culture conditions in vitro (Zuk et al., 2002). From that point on, many documents have emerged to describe the chondrogenic potential of ASCs isolated from diverse animal models including mouse (Lin et al., 2005b), rat (Lopez and Spencer, 2011), rabbit (Han et al., 2009), dog (Reich et al., 2012), and pig (Wang et al., 2008). Chondrogenic potential of ASCs When cultured in medium containing proper growth factors (TGFb-1, TGFb-2, TGFb-3, BMP-2, BMP-6, or BMP-7), ASCs differentiate into chondrocytes in vitro (Lin et al., 2005b; Knippenberg et al., 2006; Mehlhorn et al., 2007). With a few days pre-conditioning in chondrogenic medium, ASCs could form cartilage tissue in vivo (Lin et al., 2005a). Unlike bone marrow stromal cells (BMSCs), ASCs can be isolated in large quantities with minimal morbidity and discomfort clinically (Parker and Katz, 2006). In view of these practical advantages, ASCs are an alternative for chondrocytes or BMSCs in cell based cartilage regeneration strategies. Regarding the application of ASCs in cartilage repair, infrapatellar fat pad (IFP) could be a more attractive clinical source of ASCs. IFP can give rise to cells that fulfill all the criteria of MSCs, including most importantly significant chondrogenic potential (Dragoo et al., 2003; Khan et al., 2008; Buckley et al., 2010). It was even reported that ASCs derived from osteoarthritic (OA) IFP showed higher chondrogenic capacity than that of bone marrow MSCs and subcutaneous fat-derived ASCs (Sakaguchi et al., 2005; Mochizuki et al., 2006). Moreover, it was reported that chondrogenic potential of IFP derived ASCs was better preserved during in vitro expansion process compared to OA-cartilage derived chondrocytes which rapidly lose their phenotype (English et al., 2007). Micro-environment needed for cartilage matrix deposition of ASCs The differentiation medium required to induce chondrogenic differentiation of ASCs usually contains a cocktail of growth factors. Transforming growth factor-b (TGF-b) is considered as the most important component. There are three TGF-b isoforms: TGF-b1, -b2, and -b3. Their distinct roles in embryonic development have been studied intensively in mouse JOURNAL OF CELLULAR PHYSIOLOGY and human (Gatherer et al., 1990; Millan et al., 1991; Schmid et al., 1991). However, their differential functions on extracellular matrix (ECM) formation were just discovered recently. Studies showed that TGF-b3 and TGF-b2 led to significantly higher collagen type II expression and glycosaminoglycans deposition of BMSC than TGF-b1 (Barry et al., 2001). Cals et al. (2012) reported that no significant differences in total collagen and glycosaminoglycans (GAGs) formation could be observed among BMSCs cultured in medium containing the three TGF-b isoforms respectively. However cells induced by TGF-b3 had significantly higher mineralization level than cells cultured in TGF-b1 containing medium. Although we did not find any study in which the differences of TGF-b isoforms on chondrogenic differentiation of ASCs were tested, these data suggest that differences between isoforms of TGF-b may affect ASCs differentiation and ECM deposition as well. BMP-6 is another important growth factor commonly used in the differentiation medium. It was reported that BMP-6 when combined with TGF-b significantly increased chondrogenesis of ASCs by up-regulating the expression of aggrecan and collagen II with minimal side-effects such as increased collagen type X expression or other characteristics of a hypertrophic phenotype (Estes et al., 2006). The mechanism of the synergistic effects of BMP-6 and TGF-b is that BMP-6 could induce the expression of TGF-b receptor 1 which is usually not expressed by ASCs (Hennig et al., 2007). BMP-2 was used as a stimulator for osteogenic differentiation of ASCs (Lin et al., 2008b). However, BMP-2 was also applied to promote the chondrogenic differentiation of MSCs (Kurth et al., 2007; Noth et al., 2007). The cross talk between TGF and BMP signaling suggests an important role of BMP-2 in cartilage matrix deposition (Luyten et al., 1992; Keller et al., 2011). Notably, BMP-2 induced chondrogenic differentiation of MSCs would eventually lead to hypertrophy and endochondralossification (Carlberg et al., 2001; Steinert et al., 2009). BMP-4 is traditionally considered as a trigger of adipogenic differentiation of embryonic stem cells (Taha et al., 2006). A recent article presented BMP-4 as a promising growth factor for ASCs’ in vitro expansion since a low dose of BMP-4 increased their viability and maintained their multipotency (Vicente et al., 2011). Addition of BMP-4 in the differentiation medium significantly enhanced the chondrogenic phenotype of ASCs compared to TGF-b1 alone (Kim et al., 2010). The role of BMP-7 in ASCs differentiation is not as clearly defined as other BMPs. On one hand, BMP-7 has been shown to be an important regulator of brown fat adipogenesis and energy expenditure (Tseng et al., 2008); on the other hand, it is also commonly used in bone tissue engineering to promote healing of critical size bone defects (Yang et al., 2005; Koh et al., 2008; Zhu et al., 2010). To make it even more complex, there are reports claiming that BMP-7 could initiate a more chondrogenic phenotype in ASCs than BMP-2 (Knippenberg et al., 2006). It looks like BMP-7 is involved in all the three mesenchymal lineages and might play multiple roles in the differentiation of ASCs. In many studies, serum free medium was used for chondrogenic differentiation. It was reported that serum free medium maintained the expression of Sox 9 in chondrocytes during in vitro expansion and sustained their phenotype, while serum caused the de-differentiation of chondrocytes (Malpeli et al., 2004). Another report claimed that fetal bovine serum (FBS) in the differentiation medium inhibited the production of glycosaminoglycans (GAGs) and type II collagens in synovial cells (Bilgen et al., 2007). However, the negative effects of serum on chondrogenic differentiation of ASCs appears to be weak, since differentiation of ASCs towards chondrocytes was observe with the presence of serum (Ogawa et al., 2004; Lin et al., 2005b). 939 940 WU ET AL. Conventionally, chondrocytes or MSCs must be placed in a three dimensional culture environment such as a micro-mass or a pellet culture before they start depositing cartilage matrix (Koch and Gorti, 2002). One misconception is that 3D (3 dimensional) culture is required for chondrogenic differentiation of ASCs. Actually, chondrogenic differentiation of ASCs involves two biological events: commitment into chondrogenic lineage and deposition of cartilage matrix. There is ample evidence showing that 3D culture environment is not essential for chondrogenic commitment of ASCs. In vitro induction of ASCs in 2D culture was sufficient to make these cells express chondrogenic genes and form cartilage tissue in nude mice (Lin et al., 2005a; Merceron et al., 2011). Molecular cascades in ASCs during chondrogenic differentiation We previously identified a group of osteo-adipo progenitors (OAPs) in SVF from adipose tissue (Lin et al., 2008c). This group of cells possess bidirectional differentiation potential which are derived from the Scal-1 negative cell population. They simultaneously express adipogenic and osteogenic genes (RUNX2 and PPAR-g). Interestingly, PPAR-g moved from cytoplasm to the nucleus when OAPs differentiated into adipocytes, while RUNX2 stayed in the cytoplasm. In contrast, RUNX2 moved from cytoplasm to the nucleus when OAPs differentiated into osteoblast, while PPAR-g remained in the cytoplasm (Lin et al., 2008c). This article together with other studies (Enomoto et al., 2004; Heim et al., 2004; Backesjo et al., 2006) demonstrated an interesting reciprocal relationship between osteogenesis and adipogenesis: osteogenic induction enhanced expression of osteogenic genes and inhibited expression of adipogenic genes, while adipogenic induction enhanced expression of adipogenic genes and inhibited expression of osteogenic genes. When ASCs lose their potential to differentiate into the adipogenic lineage, they seem to be able to differentiate into both chondrocytes and osteoblasts. From a developmental point of view, osteoblasts and chondrocytes share the same progenitor (Zou et al., 2006). During endochondral ossification, mesenchymal progenitors first differentiate into an intermediate bipotential progenitor cell that can give rise to both the chondrocytes which give rise to primary growth plate and the osteoblasts in the bone collar. After a period of proliferation, growth plate chondrocytes become hypertrophic, die and are replaced by osteoblasts depositing bone on the cartilaginous matrix (Mackie et al., 2008). Osteochondral progenitors are not only observed during development, but are also found in vitro. A number of bipotential cell lines have been described to differentiate into both the osteogenic and chondrogenic lineages simultaneously (Grigoriadis et al., 1990; Tominaga et al., 2009). Reciprocal relationship between osteogenesis and chondrogenesis was also found in osteochondral progenitors. Hypertrophic differentiation of chondrocytes, is tightly controlled by the balance of Sox9 and Runx2: Sox9 preserves the chondrogenic phenotype, while Runx2 accelerates hypertrophic differentiation. RunX2 also acts as the master transcription regulator of osteoblastic differentiation (Ding et al., 2011; Dy et al., 2012). Once ASCs are committed to the chondrogenic lineage, molecular events become clear and simple. Cells stably express Sox9, and then Sox9 triggers the expressions of cartilage matrix proteins, including collagen type II (COL II), collagen type IX (COLIX), aggrecan (ACAN), and cartilage oligomeric matrix protein (COMP) (Lin et al., 2005a). Then a group of cytokines is secreted by mature chondroyctes to maintain the expression of Sox9 and other chondrogenic marker genes such as COL II and ACAN (Polacek et al., 2011). The molecular events regulating JOURNAL OF CELLULAR PHYSIOLOGY the step-wise differentiation from tri-potential ASCs into bi-potential osteochondral progenitors and then into committed chondrocytes are summarized in Figure 1. Increased Cartilage Formation by Co-Culture of ASCs With Chondrocytes Cartilage is a unique tissue in which only one cell population resides. Cellular interactions between chondrocytes and other cell types are rare occasions that can only occur at the superficial zone of cartilage and at the interphase between cartilage and the subchondral bone. When co-culture was first introduced into the cartilage field as a research tool (Goldring et al., 1984), it was mainly used to study the pathophysiology of rheumatoid-arthritis and osteoarthritis by investigating the cross-talk between chondrocytes on one hand and synoviocytes on the other (Lubke et al., 2005), or between chondrocytes and osteoblasts (Sanchez et al., 2005). Only recently, it has become clear that co-culture has great potential in cartilage regeneration (Hendriks et al., 2007). Synergistic effects in co-culture of ASCs and chondrocytes To reduce the cell number need for ACI, chondrocytes may be partially replaced by other more easily obtained cell types. Tsuchiya et al. (2004) first reported that co-culture of BMSCs and articular chondrocytes enhanced matrix production. The synergistic effects of co-culture were confirmed by other researchers in similar co-culture models (Mo et al., 2009; Hendriks et al., 2010). Meanwhile, increased cartilage matrix formation was also reported in co-culture of chondrocytes with ASCs (Hildner et al., 2009). To explain the mechanism of increased cartilage formation in co-cultures, two hypotheses have been proposed: (1) increased cartilage formation is due to chondrogenic differentiation of MSCs triggered by signals from chondrocytes; (2) increased cartilage matrix is a result of enhanced activity of chondrocytes stimulated by MSCs. Two hypotheses are illustrated in Figure 2. Fig. 1. Schematic representation of molecular events during stepwise differentiation of ASCs from tri-potential stem cells into bipotential osteochondral progenitors and eventually into committed chondrocytes. LPL, lipoprotein lipase; AP2, adipocyte fatty acidbinding protein 2; OCN, osteocalcin; OPN, osteopotin; COL I, collagen type I, ACAN, aggrecan; COL II, collagen type II; COMP, cartilage oligomeric matrix protein. REGENERATION OF ARTICULAR CARTILAGE Fig. 2. Two hypotheses explaining the mechanism of increased cartilage formation in co-culture of MSCs and chondrocytes. In hypothesis I (left), signals from chondrocytes induce the chondrogenic differentiation of MSCs, while in hypothesis II (right), MSCs secreted soluble factors to increase the proliferation and matrix deposition of chondrocytes. Chondrocytes promote differentiation of ASCs It was suggested that beneficial effects of co-culturing chondrocytes with MSCs are largely due to the differentiation of MSCs into chondrocytes. Soluble factors released from chondrocytes have been shown to support chondrogenesis in an indirect co-culture model of human embryonic stem cells (hESCs) and primary chondrocytes by significantly enhancing the expression of proteoglycans, collagen I and II (Vats et al., 2006). Conditioned medium of chondrocytes could induce osteo-chondrogenic differentiation of BMSCs (Hwang et al., 2007). It was also reported that co-culture of BMSCs and chondrocytes in a 3D environment induced chondrogenic gene expression in BMSCs (Vadala et al., 2008). In a trans-well coculture system, chondrogenic differentiation of BMSCs is increased by chondrocytes (Chen et al., 2009). More specifically, several studies revealed that ASC could respond to soluble factors released by nuclear pulposus cells by upregulating cartilage-specific gene expression such as of COL II and aggrecan (Li et al., 2005a; Lu et al., 2007, 2008). A conflicting study reported that direct cell–cell contact was required for the differentiation of BMScs when co-cultured with nucleus pulposus cells (Yamamoto et al., 2004; Richardson et al., 2006b). Nevertheless, many studies so far indicate secreted soluble factors may be responsible for the differentiation of BMSCs in co-culture with chondrocytes. Trophic effects of MSCs In a recently published work, we tracked the two cell populations by using a xenogenic co-culture model of human MSCs and bovine chondrocytes (Wu et al., 2011). Their contributions to cartilage matrix formation were therefore separately studied. Our data showed a significant decrease of MSCs in co-culture pellets, resulting in an almost homogeneous cartilage tissue. Thus the beneficial effect of co-culture is largely due to increased chondrocyte proliferation and matrix formation. Chondrogenic differentiation MSCs was shown to be a minor contribution to cartilage formation. Furthermore, these observations are not specific to certain species (combination) or donors. It is the first time a trophic role of MSCs has been demonstrated in stimulating chondrocyte proliferation and matrix production. JOURNAL OF CELLULAR PHYSIOLOGY Arnold Caplan first proposed MSCs as a trophic mediator for tissue repair (Caplan and Dennis, 2006). Term TROPHIC traditionally refers to the non-neurotransmitters bioactive molecules produced by nerve terminals in neurology (Singer, 1974). When first being introduced in the field of MSCs, the term ‘‘trophic effect’’ referred to the effects that MSCs secrete factors that stimulate releasing of functional bioactive factors from surrounding cells (Caplan and Dennis, 2006). Its definition then expanded to the MSC produced factors that promote cell viability, proliferation, and matrix production in the surrounding environment. The picture has been changed about the roles MSCs played in tissue repair since the introduction of trophic effects into MSCs research. Based on the first pioneer studies, people tend to believe that MSCs repair damaged tissues by differentiating into specific cell types and replacing lost cells (Bruder et al., 1994). But now, more and more researchers considered the trophic roles of the MSC as more important feature of MSCs in tissue repair (Kassis et al., 2011). Evidences supporting the trophic role of MSCs in tissue repair include MSCs improved gain of coordinated functions into brain stroked rats without differentiating into any neuronal related cell type (Li et al., 2005b) and MSCs stimulated cardiomyocyte proliferation (Sassoli et al., 2011) and vascular regeneration (Tang et al., 2005). As illustrated by recent co-culture studies (Wu et al., 2011; Acharya et al., 2012), the trophic effects of MSCs in cartilage regeneration can be dissected into several layers: (1) MSCs promoted ECM formation of chondrocytes; (2) MSCs increase proliferation of chondrocytes; (3) MSCs died overtime in the co-culture with chondrocytes. Furthermore, our follow-up study demonstrated that the trophic effects MSCs in co-culture pellets stimulating cartilage formation are independent of the culture conditions or MSCs origins (Wu et al., 2012). Co-culture pellets grow in medium stimulating chondrogenic differentiation gave similar results as pellets cultured in proliferation medium. The origins of the MSCs are also proved to be unimportant for their trophic effects since co-culturing chondrocytes with MSCs isolated from bone marrow, adipose tissue and synovial membrane all showed similar results. This implies that it is a very general observation that the MSCs play as trophic mediators in co-cultures with chondrocytes. Enhanced Chondrogenic Differentiation of ASCs by Gene Manipulation Besides co-culture ASCs with chondrocytes, over-expression of regulatory genes in ASCs is another strategy to enhance chondrogenic differentiation (Gafni et al., 2004). Our previous studies have shown that ASCs are good cell source for genetic modification (Wu et al., 2007, 2008; Lin et al., 2008b). Genes related to muscle-skeleton development have been introduced into ASCs to improve the differentiation of ASCs (Gimble et al., 2011; Peng et al., 2011). On the list of genes involved in cartilage development, there are generally two groups of genes which are potentially useful for genetic manipulation to boost cartilage regeneration (Trippel et al., 2004). These are genes encoding anabolic growth factors, such as TGF-b, BMPs and insulin-like growth factor (IGF), and transcription factors like Sox-5, -6, and -9 that control chondrogenesis. Growth factors: TGF-b TGF-b1 has been regarded as the most powerful chondrogenic growth factor, which induces significant chondrogenic phenotype of ASCs both in vitro and in vivo (Lin et al., 2005a; Lin et al., 2005b). Guo et al. (2006a) reported that a plasmid DNA encoding TGF-b1 could be entrapped into a chitosan-gelatin based biomaterial to enhance ECM deposition of chondrocytes which were incorporated in the same materials. In a similar 941 942 WU ET AL. study, Guo et al. (2006b) used a slightly different strategy in which a plasmid encoding TGF-b1 was transfected into BMSCs, then transfected cells were applied to repair full-thickness articular cartilage defects in a rabbit model. There are no reports on expressing TGF-b1 or TGF-b3 in ASCs. In contrast, TGF-b2 transduced ASCs have been used. In these studies PLGA (poly-lactic-co-glycolic acid)/alginate compound materials have been used to potentiate the differentiation of the genetically manipulated ASCs (Jin et al., 2007, 2008). It has also been demonstrated that TGF-b2 transfected ASCs could repair articular cartilage defects in rabbits (Yang and Tian, 2008). Growth factors: BMPs and others Exogenic expression of BMPs in ASCs normally leads to osteogenic differentiation. For example, BMP-2 transfected ASCs developed an osteoblastic phenotype and after loading in an alginate gel were used to repair critical size cranial defects in rat models (Lin et al., 2008b). BMP-7 was also transduced into ASCs to promote bone formation both in vitro and in vivo (Kang et al., 2007). However, there are some BMPs found to induce cartilage matrix formation when over-expressed in pluoripotent stem cells or de-differentiated chondrocytes. These BMPs might be useful to boost cartilage formation when overexpressed in ASCs. Kuroda et al. (2006) reported that BMP-4 transduced muscle derived stem cells (MDSCs) acquired chondrocyte-like characteristics in vitro and formed better cartilage in knee repair models in rats. The repairing results could even be better if BMP-4 was co-tranduced with sFit-1 (Matsumoto et al., 2009). Lin et al. (2008a) demonstrated that BMP-4 could induce re-differentiation of chondrocytes which lost their typical phenotype. The only BMP that has been ectopically expressed in ASCs is BMP-6, due to the special effects of BMP-6 that induces the expression of TGF-b receptor 1 on ASCs (Hennig et al., 2007). Diekman et al. (2010) reported a model of alginate beads to culture ASCs transfected with a pcDNA3-BMP-6 construct and confirmed the induction of chondrogenic differentiation of ASCs. Other growth factors that were considered for overexpression in ASCs for cartilage tissue engineering purposes are IFG-1, fibroblast growth factors (FGF), and epidermal growth factors (EGF). Results from a previous study suggest that dynamic compression combined with IGF-1 overexpression could benefit cartilage tissue formation of ASCs seeded in chitosan/gelatin scaffolds (Li et al., 2012). Although FGF and EGF are believed to benefit the proliferation of ASCs while keeping their chondrogenic potential (Kilroy et al., 2007; Lee et al., 2009), no transgenic studies have ever been conducted in ASCs with these two groups of factors so far. Transcription factors: Sox 9 and its family members Sox 9 is considered as the ‘‘master regulator’’ of chondrogenic differentiation (de Crombrugghe et al., 2000), since it directly controls the synthesis of collagen type II and other ECM matrix in cartilage tissue (Lefebvre et al., 1997; Zhao et al., 1997). A few researchers used adenovirus to deliver exogenic Sox 9 gene in chondrocytes and disc cells to increase the deposition of cartilage specific ECM (Paul et al., 2003). With respect to tissue engineering, Sox 9 was over-expressed in BMSCs by adenoviral transduction (Tsuchiya et al., 2003; Richardson et al., 2006a). Infected BMSCs express higher level of Collagen II than cells without transduction. Recently researchers started expressing exogenous Sox 9 in ASCs in an attempt to boost cartilage matrix formation. Yang et al. (2011b) infected ASCs with a retrovirus expressing Sox 9. In this study, they found that collagen II and proteoglycan production was increased in Sox 9 engineered ASCs. Furthermore, co-culture of Sox-9 JOURNAL OF CELLULAR PHYSIOLOGY transduced ASCs and nuclear pulposus cells in alginate beads resulted in an increase of collagen II and GAGs production. A new trend in these studies is to co-transfect ASCs with SOX Trio (Sox 5, 6, and 9 genes), since Sox 5 and 6 are believed to cooperate with Sox 9 in cartilage development (Han and Lefebvre, 2008; Dy et al., 2010). Studies showed that transfection of SOX Trio initiated the differentiation of ASCs into chondrocyte-like cells both in vitro and in vivo (Yang et al., 2011a). It was even reported that SOX Trio retroviraltransduced ASCs seeded in fibrin gel promoted the healing of osteochondral defects and prevented the progression of experimental osteoarthritis in a rat model (Lee and Im, 2012). Besides plasmid transfection and viral transduction, the delivery method could also be seeding ASCs on PLGA hydrogel incorporated with the pcDNA vector expressing SOX Trio. This method has been successfully used to treat osteochondral defects on the patellar groove of a rabbit model (Im et al., 2011). Conclusion Many efforts have been made to improve cartilage regeneration during the last few decades. Advances have been achieved to efficiently differentiate ASCs into chondrocyte-like cells. These findings can be potentially translated into stem cell-based therapies for treating large size cartilage defects. Achievements in this field have shown a wide range of prospects and promise to support cartilage regeneration in the future. Acknowledgments This work was funded by National Natural Science Foundation of China (31170929, 81071273, 81201211), Foundation for the Author of National Excellent Doctoral Dissertation of China (FANEDD 200977), Funding for Distinguished Young Scientists in Sichuan (2010JQ0066). Literature Cited Acharya C, Adesida A, Zajac P, Mumme M, Riesle J, Martin I, Barbero A. 2012. Enhanced chondrocyte proliferation and mesenchymal stromal cells chondrogenesis in coculture pellets mediate improved cartilage formation. J Cell Physiol 227:88–97. Alford JW, Cole BJ. 2005a. Cartilage restoration, part 1: Basic science, historical perspective, patient evaluation, and treatment options. Am J Sports Med 33:295–306. Alford JW, Cole BJ. 2005b. Cartilage restoration, part 2: Techniques, outcomes, and future directions. Am J Sports Med 33:443–460. Backesjo CM, Li Y, Lindgren U, Haldosen LA. 2006. Activation of Sirt1 decreases adipocyte formation during osteoblast differentiation of mesenchymal stem cells. J Bone Miner Res 21:993–1002. Barry F, Boynton RE, Liu B, Murphy JM. 2001. Chondrogenic differentiation of mesenchymal stem cells from bone marrow: Differentiation-dependent gene expression of matrix components. Exp Cell Res 268:189–200. Bartlett W, Skinner JA, Gooding CR, Carrington RW, Flanagan AM, Briggs TW, Bentley G. 2005. Autologous chondrocyte implantation versus matrix-induced autologous chondrocyte implantation for osteochondral defects of the knee: A prospective, randomised study. J Bone Joint Surg Br 87:640–645. Bilgen B, Orsini E, Aaron RK, Ciombor DM. 2007. FBS suppresses TGF-beta1-induced chondrogenesis in synoviocyte pellet cultures while dexamethasone and dynamic stimuli are beneficial. J Tissue Eng Regen Med 1:436–442. Brittberg M, Lindahl A, Nilsson A, Ohlsson C, Isaksson O, Peterson L. 1994. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N Engl J Med 331:889–895. Bruder SP, Fink DJ, Caplan AI. 1994. Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy. J Cell Biochem 56:283–294. Buckley CT, Vinardell T, Thorpe SD, Haugh MG, Jones E, McGonagle D, Kelly DJ. 2010. Functional properties of cartilaginous tissues engineered from infrapatellar fat pad-derived mesenchymal stem cells. J Biomech 43:920–926. Cals FL, Hellingman CA, Koevoet W, Baatenburg de Jong RJ, van Osch GJ. 2012. Effects of transforming growth factor-beta subtypes on in vitro cartilage production and mineralization of human bone marrow stromal-derived mesenchymal stem cells. J Tissue Eng Regen Med 6:68–76. Caplan AI, Dennis JE. 2006. Mesenchymal stem cells as trophic mediators. J Cell Biochem 98:1076–1084. Carlberg AL, Pucci B, Rallapalli R, Tuan RS, Hall DJ. 2001. Efficient chondrogenic differentiation of mesenchymal cells in micromass culture by retroviral gene transfer of BMP-2. Differentiation 67:128–138. Chen WH, Lai MT, Wu AT, Wu CC, Gelovani JG, Lin CT, Hung SC, Chiu WT, Deng WP. 2009. In vitro stage-specific chondrogenesis of mesenchymal stem cells committed to chondrocytes. Arthritis Rheum 60:450–459. de Crombrugghe B, Lefebvre V, Behringer RR, Bi W, Murakami S, Huang W. 2000. Transcriptional mechanisms of chondrocyte differentiation. Matrix Biol 19:389–394. REGENERATION OF ARTICULAR CARTILAGE Diekman BO, Estes BT, Guilak F. 2010. The effects of BMP6 overexpression on adipose stem cell chondrogenesis: Interactions with dexamethasone and exogenous growth factors. J Biomed Mater Res A 93:994–1003. Ding M, Lu Y, Abbassi S, Li F, Li X, Song Y, Geoffroy V, Im HJ, Zheng Q. 2011. Targeting Runx2 expression in hypertrophic chondrocytes impairs endochondral ossification during early skeletal development. J Cell Physiol 227:3446–3456. Dragoo JL, Samimi B, Zhu M, Hame SL, Thomas BJ, Lieberman JR, Hedrick MH, Benhaim P. 2003. Tissue-engineered cartilage and bone using stem cells from human infrapatellar fat pads. J Bone Joint Surg Br 85:740–747. Dy P, Smits P, Silvester A, Penzo-Mendez A, Dumitriu B, Han Y, de la Motte CA, Kingsley DM, Lefebvre V. 2010. Synovial joint morphogenesis requires the chondrogenic action of Sox5 and Sox6 in growth plate and articular cartilage. Dev Biol 341:346–359. Dy P, Wang W, Bhattaram P, Wang Q, Wang L, Ballock RT, Lefebvre V. 2012. Sox9 directs hypertrophic maturation and blocks osteoblast differentiation of growth plate chondrocytes. Dev Cell 22:597–609. English A, Jones EA, Corscadden D, Henshaw K, Chapman T, Emery P, McGonagle D. 2007. A comparative assessment of cartilage and joint fat pad as a potential source of cells for autologous therapy development in knee osteoarthritis. Rheumatology (Oxford) 46:1676–1683. Enomoto H, Furuichi T, Zanma A, Yamana K, Yoshida C, Sumitani S, Yamamoto H, EnomotoIwamoto M, Iwamoto M, Komori T. 2004. Runx2 deficiency in chondrocytes causes adipogenic changes in vitro. J Cell Sci 117:417–425. Estes BT, Wu AW, Guilak F. 2006. Potent induction of chondrocytic differentiation of human adipose-derived adult stem cells by bone morphogenetic protein 6. Arthritis Rheum 54:1222–1232. Furukawa T, Eyre DR, Koide S, Glimcher MJ. 1980. Biochemical studies on repair cartilage resurfacing experimental defects in the rabbit knee. J Bone Joint Surg Am 62:79–89. Gafni Y, Turgeman G, Liebergal M, Pelled G, Gazit Z, Gazit D. 2004. Stem cells as vehicles for orthopedic gene therapy. Gene Ther 11:417–426. Gatherer D, Ten Dijke P, Baird DT, Akhurst RJ. 1990. Expression of TGF-beta isoforms during first trimester human embryogenesis. Development 110:445–460. Gilbert JE. 1998. Current treatment options for the restoration of articular cartilage. Am J Knee Surg 11:42–46. Gimble JM, Grayson W, Guilak F, Lopez MJ, Vunjak-Novakovic G. 2011. Adipose tissue as a stem cell source for musculoskeletal regeneration. Front Biosci (Schol Ed) 3:69–81. Goldring SR, Dayer JM, Krane SM. 1984. Rheumatoid synovial cell hormone responses modulated by cell-cell interactions. Inflammation 8:107–121. Grigoriadis AE, Heersche JN, Aubin JE. 1990. Continuously growing bipotential and monopotential myogenic, adipogenic, and chondrogenic subclones isolated from the multipotential RCJ 3.1 clonal cell line. Dev Biol 142:313–318. Guo T, Zhao J, Chang J, Ding Z, Hong H, Chen J, Zhang J. 2006a. Porous chitosan-gelatin scaffold containing plasmid DNA encoding transforming growth factor-beta1 for chondrocytes proliferation. Biomaterials 27:1095–1103. Guo X, Zheng Q, Yang S, Shao Z, Yuan Q, Pan Z, Tang S, Liu K, Quan D. 2006b. Repair of fullthickness articular cartilage defects by cultured mesenchymal stem cells transfected with the transforming growth factor beta1 gene. Biomed Mater 1:206–215. Han Y, Lefebvre V. 2008. L-Sox5 and Sox6 drive expression of the aggrecan gene in cartilage by securing binding of Sox9 to a far-upstream enhancer. Mol Cell Biol 28:4999–5013. Han Y, Wei Y, Wang S, Song Y. 2009. Enhanced chondrogenesis of adipose-derived stem cells by the controlled release of transforming growth factor-beta1 from hybrid microspheres. Gerontology 55:592–599. Heim M, Frank O, Kampmann G, Sochocky N, Pennimpede T, Fuchs P, Hunziker W, Weber P, Martin I, Bendik I. 2004. The phytoestrogen genistein enhances osteogenesis and represses adipogenic differentiation of human primary bone marrow stromal cells. Endocrinology 145:848–859. Hendriks J, Riesle J, van Blitterswijk CA. 2007. Co-culture in cartilage tissue engineering. J Tissue Eng Regen Med 1:170–178. Hendriks JAA, Miclea RL, Schotel R, de Bruijn E, Moroni L, Karperien M, Riesle J, van Blitterswijk CA. 2010. Primary chondrocytes enhance cartilage tissue formation upon coculture with a range of cell types. Soft Matter 6:5080–5088. Hennig T, Lorenz H, Thiel A, Goetzke K, Dickhut A, Geiger F, Richter W. 2007. Reduced chondrogenic potential of adipose tissue derived stromal cells correlates with an altered TGFbeta receptor and BMP profile and is overcome by BMP-6. J Cell Physiol 211:682–691. Hildner F, Concaro S, Peterbauer A, Wolbank S, Danzer M, Lindahl A, Gatenholm P, Redl H, van Griensven M. 2009. Human adipose-derived stem cells contribute to chondrogenesis in coculture with human articular chondrocytes. Tissue Eng Part A 15:3961–3969. Hwang NS, Varghese S, Puleo C, Zhang Z, Elisseeff J. 2007. Morphogenetic signals from chondrocytes promote chondrogenic and osteogenic differentiation of mesenchymal stem cells. J Cell Physiol 212:281–284. Im GI, Kim HJ, Lee JH. 2011. Chondrogenesis of adipose stem cells in a porous PLGA scaffold impregnated with plasmid DNA containing SOX trio (SOX-5,-6 and -9) genes. Biomaterials 32:4385–4392. Jin X, Sun Y, Zhang K, Wang J, Shi T, Ju X, Lou S. 2007. Ectopic neocartilage formation from predifferentiated human adipose derived stem cells induced by adenoviral-mediated transfer of hTGF beta2. Biomaterials 28:2994–3003. Jin XB, Sun YS, Zhang K, Wang J, Shi TP, Ju XD, Lou SQ. 2008. Tissue engineered cartilage from hTGF beta2 transduced human adipose derived stem cells seeded in PLGA/alginate compound in vitro and in vivo. J Biomed Mater Res A 86:1077–1087. Kang Y, Liao WM, Yuan ZH, Sheng PY, Zhang LJ, Yuan XW, Lei L. 2007. In vitro and in vivo induction of bone formation based on adeno-associated virus-mediated BMP-7 gene therapy using human adipose-derived mesenchymal stem cells. Acta Pharmacol Sin 28:839–849. Kassis I, Vaknin-Dembinsky A, Karussis D. 2011. Bone marrow mesenchymal stem cells: Agents of immunomodulation and neuroprotection. Curr Stem Cell Res Ther 6:63–68. Keller B, Yang T, Chen Y, Munivez E, Bertin T, Zabel B, Lee B. 2011. Interaction of TGFbeta and BMP signaling pathways during chondrogenesis. PLoS ONE 6:e16421. Khan WS, Tew SR, Adesida AB, Hardingham TE. 2008. Human infrapatellar fat pad-derived stem cells express the pericyte marker 3G5 and show enhanced chondrogenesis after expansion in fibroblast growth factor-2. Arthritis Res Ther 10:R74. Kilroy GE, Foster SJ, Wu X, Ruiz J, Sherwood S, Heifetz A, Ludlow JW, Stricker DM, Potiny S, Green P, Halvorsen YD, Cheatham B, Storms RW, Gimble JM. 2007. Cytokine profile of human adipose-derived stem cells: Expression of angiogenic, hematopoietic, and proinflammatory factors. J Cell Physiol 212:702–709. Kim BS, Kang KS, Kang SK. 2010. Soluble factors from ASCs effectively direct control of chondrogenic fate. Cell Prolif 43:249–261. Knippenberg M, Helder MN, Zandieh Doulabi B, Wuisman PI, Klein-Nulend J. 2006. Osteogenesis versus chondrogenesis by BMP-2 and BMP-7 in adipose stem cells. Biochem Biophys Res Commun 342:902–908. JOURNAL OF CELLULAR PHYSIOLOGY Koch RJ, Gorti GK. 2002. Tissue engineering with chondrocytes. Facial Plast Surg 18:59–68. Koh JT, Zhao Z, Wang Z, Lewis IS, Krebsbach PH, Franceschi RT. 2008. Combinatorial gene therapy with BMP2/7 enhances cranial bone regeneration. J Dent Res 87:845–849. Kuroda R, Usas A, Kubo S, Corsi K, Peng H, Rose T, Cummins J, Fu FH, Huard J. 2006. Cartilage repair using bone morphogenetic protein 4 and muscle-derived stem cells. Arthritis Rheum 54:433–442. Kurth T, Hedbom E, Shintani N, Sugimoto M, Chen FH, Haspl M, Martinovic S, Hunziker EB. 2007. Chondrogenic potential of human synovial mesenchymal stem cells in alginate. Osteoarthritis Cartilage 15:1178–1189. Lee JM, Im GI. 2012. SOX trio-co-transduced adipose stem cells in fibrin gel to enhance cartilage repair and delay the progression of osteoarthritis in the rat. Biomaterials 33:2016–2024. Lee SY, Lim J, Khang G, Son Y, Choung PH, Kang SS, Chun SY, Shin HI, Kim SY, Park EK. 2009. Enhanced ex vivo expansion of human adipose tissue-derived mesenchymal stromal cells by fibroblast growth factor-2 and dexamethasone. Tissue Eng Part A 15:2491–2499. Lefebvre V, Huang W, Harley VR, Goodfellow PN, de Crombrugghe B. 1997. SOX9 is a potent activator of the chondrocyte-specific enhancer of the pro alpha1(II) collagen gene. Mol Cell Biol 17:2336–2346. Li X, Lee JP, Balian G, Greg Anderson D. 2005a. Modulation of chondrocytic properties of fatderived mesenchymal cells in co-cultures with nucleus pulposus. Connect Tissue Res 46:75–82. Li Y, Chen J, Zhang CL, Wang L, Lu D, Katakowski M, Gao Q, Shen LH, Zhang J, Lu M, Chopp M. 2005b. Gliosis and brain remodeling after treatment of stroke in rats with marrow stromal cells. Glia 49:407–417. Li J, Zhao Q, Wang E, Zhang C, Wang G, Yuan Q. 2012. Dynamic compression of rabbit adipose-derived stem cells transfected with insulin-like growth factor 1 in chitosan/gelatin scaffolds induces chondrogenesis and matrix biosynthesis. J Cell Physiol 227:2003–2012. Lin Y, Luo E, Chen X, Liu L, Qiao J, Yan Z, Li Z, Tang W, Zheng X, Tian W. 2005a. Molecular and cellular characterization during chondrogenic differentiation of adipose tissue-derived stromal cells in vitro and cartilage formation in vivo. J Cell Mol Med 9:929–939. Lin Y, Tian W, Chen X, Yan Z, Li Z, Qiao J, Liu L, Tang W, Zheng X. 2005b. Expression of exogenous or endogenous green fluorescent protein in adipose tissue-derived stromal cells during chondrogenic differentiation. Mol Cell Biochem 277:181–190. Lin L, Zhou C, Wei X, Hou Y, Zhao L, Fu X, Zhang J, Yu C. 2008a. Articular cartilage repair using dedifferentiated articular chondrocytes and bone morphogenetic protein 4 in a rabbit model of articular cartilage defects. Arthritis Rheum 58:1067–1075. Lin Y, Tang W, Wu L, Jing W, Li X, Wu Y, Liu L, Long J, Tian W. 2008b. Bone regeneration by BMP-2 enhanced adipose stem cells loading on alginate gel. Histochem Cell Biol 129:203– 210. Lin YF, Jing W, Wu L, Li XY, Wu Y, Liu L, Tang W, Long J, Tian WD, Mo XM. 2008c. Identification of osteo-adipo progenitor cells in fat tissue. Cell Prolif 41:803–812. Lopez MJ, Spencer ND. 2011. In vitro adult rat adipose tissue-derived stromal cell isolation and differentiation. Methods Mol Biol 702:37–46. Lu ZF, Zandieh Doulabi B, Wuisman PI, Bank RA, Helder MN. 2007. Differentiation of adipose stem cells by nucleus pulposus cells: Configuration effect. Biochem Biophys Res Commun 359:991–996. Lu ZF, Doulabi BZ, Wuisman PI, Bank RA, Helder MN. 2008. Influence of collagen type II and nucleus pulposus cells on aggregation and differentiation of adipose tissue-derived stem cells. J Cell Mol Med 12:2812–2822. Lubke C, Ringe J, Krenn V, Fernahl G, Pelz S, Kreusch-Brinker R, Sittinger M, Paulitschke M. 2005. Growth characterization of neo porcine cartilage pellets and their use in an interactive culture model. Osteoarthritis Cartilage 13:478–487. Luyten FP, Yu YM, Yanagishita M, Vukicevic S, Hammonds RG, Reddi AH. 1992. Natural bovine osteogenin and recombinant human bone morphogenetic protein-2B are equipotent in the maintenance of proteoglycans in bovine articular cartilage explant cultures. J Biol Chem 267:3691–3695. Mackie EJ, Ahmed YA, Tatarczuch L, Chen KS, Mirams M. 2008. Endochondral ossification: How cartilage is converted into bone in the developing skeleton. Int J Biochem Cell Biol 40:46–62. Malpeli M, Randazzo N, Cancedda R, Dozin B. 2004. Serum-free growth medium sustains commitment of human articular chondrocyte through maintenance of Sox9 expression. Tissue Eng 10:145–155. Mankin HJ. 1982. The response of articular cartilage to mechanical injury. J Bone Joint Surg Am 64:460–466. Matsumoto T, Cooper GM, Gharaibeh B, Meszaros LB, Li G, Usas A, Fu FH, Huard J. 2009. Cartilage repair in a rat model of osteoarthritis through intraarticular transplantation of muscle-derived stem cells expressing bone morphogenetic protein 4 and soluble Flt-1. Arthritis Rheum 60:1390–1405. Mehlhorn AT, Niemeyer P, Kaschte K, Muller L, Finkenzeller G, Hartl D, Sudkamp NP, Schmal H. 2007. Differential effects of BMP-2 and TGF-beta1 on chondrogenic differentiation of adipose derived stem cells. Cell Prolif 40:809–823. Merceron C, Portron S, Masson M, Lesoeur J, Fellah BH, Gauthier O, Geffroy O, Weiss P, Guicheux J, Vinatier C. 2011. The effect of two and three dimensional cell culture on the chondrogenic potential of human adipose-derived mesenchymal stem cells after subcutaneous transplantation with an injectable hydrogel. Cell Transplant 20:1575–1588. Millan FA, Denhez F, Kondaiah P, Akhurst RJ. 1991. Embryonic gene expression patterns of TGF beta 1, beta 2 and beta 3 suggest different developmental functions in vivo. Development 111:131–143. Mo XT, Guo SC, Xie HQ, Deng L, Zhi W, Xiang Z, Li XQ, Yang ZM. 2009. Variations in the ratios of co-cultured mesenchymal stem cells and chondrocytes regulate the expression of cartilaginous and osseous phenotype in alginate constructs. Bone 45:42–51. Mochizuki T, Muneta T, Sakaguchi Y, Nimura A, Yokoyama A, Koga H, Sekiya I. 2006. Higher chondrogenic potential of fibrous synovium- and adipose synovium-derived cells compared with subcutaneous fat-derived cells: Distinguishing properties of mesenchymal stem cells in humans. Arthritis Rheum 54:843–853. Nehrer S, Spector M, Minas T. 1999. Histologic analysis of tissue after failed cartilage repair procedures. Clin Orthop Relat Res 365:149–162. Noth U, Rackwitz L, Heymer A, Weber M, Baumann B, Steinert A, Schutze N, Jakob F, Eulert J. 2007. Chondrogenic differentiation of human mesenchymal stem cells in collagen type I hydrogels. J Biomed Mater Res A 83:626–635. Ogawa R, Mizuno H, Hyakusoku H, Watanabe A, Migita M, Shimada T. 2004. Chondrogenic and osteogenic differentiation of adipose-derived stem cells isolated from GFP transgenic mice. J Nihon Med Sch 71:240–241. Parker AM, Katz AJ. 2006. Adipose-derived stem cells for the regeneration of damaged tissues. Expert Opin Biol Ther 6:567–578. Paul R, Haydon RC, Cheng H, Ishikawa A, Nenadovich N, Jiang W, Zhou L, Breyer B, Feng T, Gupta P, He TC, Phillips FM. 2003. Potential use of Sox9 gene therapy for intervertebral degenerative disc disease. Spine (Phila Pa 1976) 28:755–763. 943 944 WU ET AL. Peng LH, Fung KP, Leung PC, Gao JQ. 2011. Genetically manipulated adult stem cells for wound healing. Drug Discov Today 16:957–966. Peterson L, Minas T, Brittberg M, Lindahl A. 2003. Treatment of osteochondritis dissecans of the knee with autologous chondrocyte transplantation: Results at two to ten years. J Bone Joint Surg Am 85-A:17–24. Polacek M, Bruun JA, Elvenes J, Figenschau Y, Martinez I. 2011. The secretory profiles of cultured human articular chondrocytes and mesenchymal stem cells: Implications for autologous cell transplantation strategies. Cell Transplant 20:1381–1393. Reich CM, Raabe O, Wenisch S, Bridger PS, Kramer M, Arnhold S. 2012. Isolation, culture and chondrogenic differentiation of canine adipose tissue- and bone marrow-derived mesenchymal stem cells-a comparative study. Vet Res Commun 36:139–148. Richardson SM, Curran JM, Chen R, Vaughan-Thomas A, Hunt JA, Freemont AJ, Hoyland JA. 2006a. The differentiation of bone marrow mesenchymal stem cells into chondrocyte-like cells on poly-L-lactic acid (PLLA) scaffolds. Biomaterials 27:4069–4078. Richardson SM, Walker RV, Parker S, Rhodes NP, Hunt JA, Freemont AJ, Hoyland JA. 2006b. Intervertebral disc cell-mediated mesenchymal stem cell differentiation. Stem Cells 24:707–716. Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. 2005. Comparison of human stem cells derived from various mesenchymal tissues: Superiority of synovium as a cell source. Arthritis Rheum 52:2521–2529. Sanchez C, Deberg MA, Piccardi N, Msika P, Reginster JY, Henrotin YE. 2005. Subchondral bone osteoblasts induce phenotypic changes in human osteoarthritic chondrocytes. Osteoarthritis Cartilage 13:988–997. Sassoli C, Pini A, Mazzanti B, Quercioli F, Nistri S, Saccardi R, Orlandini SZ, Bani D, Formigli L. 2011. Mesenchymal stromal cells affect cardiomyocyte growth through juxtacrine Notch1/Jagged1 signaling and paracrine mechanisms: Clues for cardiac regeneration. J Mol Cell Cardiol 51:399–408. Schmid P, Cox D, Bilbe G, Maier R, McMaster GK. 1991. Differential expression of TGF beta 1, beta 2 and beta 3 genes during mouse embryogenesis. Development 111:117–130. Singer M. 1974. Trophic functions of the neuron. VI. Other trophic systems. Neurotrophic control of limb regeneration in the newt. Ann N Y Acad Sci 228:308–322. Sledge SL. 2001. Microfracture techniques in the treatment of osteochondral injuries. Clin Sports Med 20:365–377. Steinert AF, Proffen B, Kunz M, Hendrich C, Ghivizzani SC, Noth U, Rethwilm A, Eulert J, Evans CH. 2009. Hypertrophy is induced during the in vitro chondrogenic differentiation of human mesenchymal stem cells by bone morphogenetic protein-2 and bone morphogenetic protein-4 gene transfer. Arthritis Res Ther 11:R148. Taha MF, Valojerdi MR, Mowla SJ. 2006. Effect of bone morphogenetic protein-4 (BMP-4) on adipocyte differentiation from mouse embryonic stem cells. Anat Histol Embryol 35:271–278. Tang YL, Zhao Q, Qin X, Shen L, Cheng L, Ge J, Phillips MI. 2005. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann Thorac Surg 80:229–236. Tominaga H, Maeda S, Miyoshi H, Miyazono K, Komiya S, Imamura T. 2009. Expression of osterix inhibits bone morphogenetic protein-induced chondrogenic differentiation of mesenchymal progenitor cells. J Bone Miner Metab 27:36–45. Trippel SB, Ghivizzani SC, Nixon AJ. 2004. Gene-based approaches for the repair of articular cartilage. Gene Ther 11:351–359. Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, Tran TT, Suzuki R, Espinoza DO, Yamamoto Y, Ahrens MJ, Dudley AT, Norris AW, Kulkarni RN, Kahn CR. 2008. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454:1000–1004. Tsuchiya H, Kitoh H, Sugiura F, Ishiguro N. 2003. Chondrogenesis enhanced by overexpression of sox9 gene in mouse bone marrow-derived mesenchymal stem cells. Biochem Biophys Res Commun 301:338–343. Tsuchiya K, Chen G, Ushida T, Matsuno T, Tateishi T. 2004. The effect of coculture of chondrocytes with mesenchymal stem cells on their cartilaginous phenotype in vitro. Mater Sci Eng C 24:6. JOURNAL OF CELLULAR PHYSIOLOGY Vadala G, Studer RK, Sowa G, Spiezia F, Iucu C, Denaro V, Gilbertson LG, Kang JD. 2008. Coculture of bone marrow mesenchymal stem cells and nucleus pulposus cells modulate gene expression profile without cell fusion. Spine (Phila Pa 1976) 33:870–876. Vats A, Bielby RC, Tolley N, Dickinson SC, Boccaccini AR, Hollander AP, Bishop AE, Polak JM. 2006. Chondrogenic differentiation of human embryonic stem cells: The effect of the micro-environment. Tissue Eng 12:1687–1697. Vicente Lopez, Vazquez MA, Garcia MN, Entrena A, Olmedillas Lopez S, Garcia-Arranz M, Garcia-Olmo D, Zapata A. 2011. Low doses of bone morphogenetic protein 4 increase the survival of human adipose-derived stem cells maintaining their stemness and multipotency. Stem Cells Dev 20:1011–1019. Vincent T, Hermansson M, Bolton M, Wait R, Saklatvala J. 2002. Basic FGF mediates an immediate response of articular cartilage to mechanical injury. Proc Natl Acad Sci USA 99:8259–8264. Wang KH, Kao AP, Wangchen H, Wang FY, Chang CH, Chang CC, Lin SD. 2008. Optimizing proliferation and characterization of multipotent stem cells from porcine adipose tissue. Biotechnol Appl Biochem 51:159–166. Wu L, Wu Y, Lin Y, Jing W, Nie X, Qiao J, Liu L, Tang W, Tian W. 2007. Osteogenic differentiation of adipose derived stem cells promoted by overexpression of osterix. Mol Cell Biochem 301:83–92. Wu L, Zhu F, Wu Y, Lin Y, Nie X, Jing W, Qiao J, Liu L, Tang W, Zheng X, Tian W. 2008. Dentin sialophosphoprotein-promoted mineralization and expression of odontogenic genes in adipose-derived stromal cells. Cells Tissues Organs 187:103–112. Wu L, Leijten JC, Georgi N, Post JN, van Blitterswijk CA, Karperien M. 2011. Trophic effects of mesenchymal stem cells increase chondrocyte proliferation and matrix formation. Tissue Eng Part A 17:1425–1436. Wu L, Prins HJ, Helder M, van Blitterswijk C, Karperien M. 2012. Trophic effects of mesenchymal stem cells in chondrocyte co-cultures are independent of culture conditions and cell sources. Tissue Eng Part A 18:1542–1551. Yamamoto Y, Mochida J, Sakai D, Nakai T, Nishimura K, Kawada H, Hotta T. 2004. Upregulation of the viability of nucleus pulposus cells by bone marrow-derived stromal cells: Significance of direct cell-to-cell contact in coculture system. Spine (Phila Pa 1976) 29:1508–1514. Yang YR, Tian H. 2008. [Repair of articular cartilage defects by autologous adipose derived mesenchymal stem cell: Experiment with rabbits]. Zhonghua Yi Xue Za Zhi 88: 2214–2218. Yang M, Ma QJ, Dang GT, Ma K, Chen P, Zhou CY. 2005. In vitro and in vivo induction of bone formation based on ex vivo gene therapy using rat adipose-derived adult stem cells expressing BMP-7. Cytotherapy 7:273–281. Yang HN, Park JS, Woo DG, Jeon SY, Do HJ, Lim HY, Kim SW, Kim JH, Park KH. 2011a. Chondrogenesis of mesenchymal stem cells and dedifferentiated chondrocytes by transfection with SOX Trio genes. Biomaterials 32:7695–7704. Yang Z, Huang CY, Candiotti KA, Zeng X, Yuan T, Li J, Yu H, Abdi S. 2011b. Sox-9 facilitates differentiation of adipose tissue-derived stem cells into a chondrocyte-like phenotype in vitro. J Orthop Res 29:1291–1297. Zhao Q, Eberspaecher H, Lefebvre V, De Crombrugghe B. 1997. Parallel expression of Sox9 and Col2a1 in cells undergoing chondrogenesis. Dev Dyn 209:377–386. Zhu L, Chuanchang D, Wei L, Yilin C, Jiasheng D. 2010. Enhanced healing of goat femur-defect using BMP7 gene-modified BMSCs and load-bearing tissue-engineered bone. J Orthop Res 28:412–418. Zou L, Zou X, Li H, Mygind T, Zeng Y, Lu N, Bunger C. 2006. Molecular mechanism of osteochondroprogenitor fate determination during bone formation. Adv Exp Med Biol 585:431–441. Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, Benhaim P, Lorenz HP, Hedrick MH. 2001. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng 7:211–228. Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. 2002. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell 13:4279–4295.