Elucidating the Secretion Proteome of Human Embryonic Stem Cell-derived Mesenchymal Stem Cells

10
Elucidating the Secretion Proteome of Human Embryonic Stem Cell-derived Mesenchymal Stem Cells* S Siu Kwan Sze‡, Dominique P. V. de Kleijn§¶, Ruenn Chai Lai, Eileen Khia Way Tan§, Hui Zhao, Keng Suan Yeo§, Teck Yew Low**, Qizhou Lian§, Chuen Neng Lee‡‡, Wayne Mitchell§§, Reida Menshawe El Oakley‡‡, and Sai-Kiang Lim§¶¶ Transplantation of mesenchymal stem cells (MSCs) has been used to treat a wide range of diseases, and the mechanism of action is postulated to be mediated by either differentiation into functional reparative cells that replace injured tissues or secretion of paracrine factors that promote tissue repair. To complement earlier studies that identified some of the paracrine factors, we profiled the paracrine proteome to better assess the relevance of MSC paracrine factors to the wide spectrum of MSC- mediated therapeutic effects. To evaluate the therapeutic potential of the MSC paracrine proteome, a chemically defined serum-free culture medium was conditioned by MSCs derived from human embryonic stem cells using a clinically compliant protocol. The conditioned medium was analyzed by multidimensional protein identification technology and cytokine antibody array analysis and re- vealed the presence of 201 unique gene products. 86 – 88% of these gene products had detectable transcript levels by microarray or quantitative RT-PCR assays. Com- putational analysis predicted that these gene products will significantly drive three major groups of biological processes: metabolism, defense response, and tissue differentiation including vascularization, hematopoiesis, and skeletal development. It also predicted that the 201 gene products activate important signaling pathways in cardiovascular biology, bone development, and hema- topoiesis such as Jak-STAT, MAPK, Toll-like receptor, transforming growth factor-, and mTOR (mammalian target of rapamycin) signaling pathways. This study identified a large number of MSC secretory products that have the potential to act as paracrine modulators of tissue repair and replacement in diseases of the cardio- vascular, hematopoietic, and skeletal tissues. Moreover our results suggest that human embryonic stem cell-de- rived MSC-conditioned medium has the potency to treat a variety of diseases in humans without cell transplantation. Molecular & Cellular Proteomics 6:1680 –1689, 2007. Mesenchymal stem cells (MSCs) 1 are multipotent stem cells that have been used in clinical and preclinical applica- tions to treat a wide range of diseases (1, 2) including mus- culoskeletal tissue bioengineering (3, 4) and heart disease (5, 6). They are routinely isolated from adult tissues such as bone marrow and adipose tissues and expanded ex vivo. Ex vivo expanded MSCs have lineage-restricted differentiation poten- tial and can be induced to differentiate into mesenchymal lineages such as osteoblasts, chondrocytes, adipocytes, myocytes, tendon-ligament fibroblasts, and cardiomyocytes. Transplantation of these MSCs has been shown to enhance repair of musculoskeletal injuries, reduce tissue damage and improve cardiac function in ischemic heart disease, and ame- liorate severity of graft versus host disease (1). Unlike embry- onic stem cells, these lineage-restricted stem cells have neg- ligible risk of teratoma formation (7). The therapeutic capacity of MSCs to treat a wide spectrum of diseases has been attributed to their potential to differen- tiate into many different reparative cell types. However, the efficiency of transplanted MSCs to differentiate into functional reparative cells in the injured tissues or organs and in thera- peutically relevant numbers has never been adequately doc- umented or demonstrated. Recent reports have suggested that some of these reparative effects are not mediated by From the ‡School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore, §Stem Cell and Developmental Biology, **Information and Mathematical Sciences, and §§Proteomics, Genome Institute of Singapore, 60 Biopolis Street, Singapore 138672, Singapore, ¶Experimental Car- diology, Utrecht Medical Center, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands, Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological Uni- versity, 50 Nanyang Avenue, Singapore 639798, Singapore, De- partments of ‡‡Surgery and ¶¶Biochemistry, National University of Singapore, Lower Kent Ridge Road, Singapore 117597, Singapore Received, October 12, 2006, and in revised form, April 16, 2007 Published, MCP Papers in Press, June 11, 2007, DOI 10.1074/ mcp.M600393-MCP200 1 The abbreviations used are: MSC, mesenchymal stem cell; hESC, human embryonic stem cell; hESC-MSC, hESC-derived MSC; ITS, insulin, transferrin, and selenoprotein; CM, conditioned medium; NCM, non-conditioned medium; SCX, strong cation exchange; qRT, quantitative RT; mTOR, mammalian target of rapamycin; MAPK, mi- togen-activated protein kinase; STAT, signal transducers and activa- tors of transcription; Jak, Janus kinase; TGF, transforming growth factor; DMEM, Dulbecco’s modified Eagle’s medium; PDGF, platelet- derived growth factor; FA, formic acid; GO, gene ontology; ECM, extracellular matrix; LOD, limit of detection. Research © 2007 by The American Society for Biochemistry and Molecular Biology, Inc. 1680 Molecular & Cellular Proteomics 6.10 This paper is available on line at http://www.mcponline.org

Transcript of Elucidating the Secretion Proteome of Human Embryonic Stem Cell-derived Mesenchymal Stem Cells

Elucidating the Secretion Proteome of HumanEmbryonic Stem Cell-derived MesenchymalStem Cells*□S

Siu Kwan Sze‡, Dominique P. V. de Kleijn§¶, Ruenn Chai Lai�, Eileen Khia Way Tan§,Hui Zhao�, Keng Suan Yeo§, Teck Yew Low**, Qizhou Lian§, Chuen Neng Lee‡‡,Wayne Mitchell§§, Reida Menshawe El Oakley‡‡, and Sai-Kiang Lim§¶¶��

Transplantation of mesenchymal stem cells (MSCs) hasbeen used to treat a wide range of diseases, and themechanism of action is postulated to be mediated byeither differentiation into functional reparative cells thatreplace injured tissues or secretion of paracrine factorsthat promote tissue repair. To complement earlier studiesthat identified some of the paracrine factors, we profiledthe paracrine proteome to better assess the relevance ofMSC paracrine factors to the wide spectrum of MSC-mediated therapeutic effects. To evaluate the therapeuticpotential of the MSC paracrine proteome, a chemicallydefined serum-free culture medium was conditioned byMSCs derived from human embryonic stem cells using aclinically compliant protocol. The conditioned mediumwas analyzed by multidimensional protein identificationtechnology and cytokine antibody array analysis and re-vealed the presence of 201 unique gene products. 86–88% of these gene products had detectable transcriptlevels by microarray or quantitative RT-PCR assays. Com-putational analysis predicted that these gene productswill significantly drive three major groups of biologicalprocesses: metabolism, defense response, and tissuedifferentiation including vascularization, hematopoiesis,and skeletal development. It also predicted that the 201gene products activate important signaling pathways incardiovascular biology, bone development, and hema-topoiesis such as Jak-STAT, MAPK, Toll-like receptor,transforming growth factor-�, and mTOR (mammaliantarget of rapamycin) signaling pathways. This studyidentified a large number of MSC secretory productsthat have the potential to act as paracrine modulators of

tissue repair and replacement in diseases of the cardio-vascular, hematopoietic, and skeletal tissues. Moreoverour results suggest that human embryonic stem cell-de-rived MSC-conditioned medium has the potency to treat avariety of diseases in humans without cell transplantation.Molecular & Cellular Proteomics 6:1680–1689, 2007.

Mesenchymal stem cells (MSCs)1 are multipotent stemcells that have been used in clinical and preclinical applica-tions to treat a wide range of diseases (1, 2) including mus-culoskeletal tissue bioengineering (3, 4) and heart disease (5,6). They are routinely isolated from adult tissues such as bonemarrow and adipose tissues and expanded ex vivo. Ex vivoexpanded MSCs have lineage-restricted differentiation poten-tial and can be induced to differentiate into mesenchymallineages such as osteoblasts, chondrocytes, adipocytes,myocytes, tendon-ligament fibroblasts, and cardiomyocytes.Transplantation of these MSCs has been shown to enhancerepair of musculoskeletal injuries, reduce tissue damage andimprove cardiac function in ischemic heart disease, and ame-liorate severity of graft versus host disease (1). Unlike embry-onic stem cells, these lineage-restricted stem cells have neg-ligible risk of teratoma formation (7).

The therapeutic capacity of MSCs to treat a wide spectrumof diseases has been attributed to their potential to differen-tiate into many different reparative cell types. However, theefficiency of transplanted MSCs to differentiate into functionalreparative cells in the injured tissues or organs and in thera-peutically relevant numbers has never been adequately doc-umented or demonstrated. Recent reports have suggestedthat some of these reparative effects are not mediated by

From the ‡School of Biological Sciences, Nanyang TechnologicalUniversity, 60 Nanyang Drive, Singapore 637551, Singapore, §StemCell and Developmental Biology, **Information and MathematicalSciences, and §§Proteomics, Genome Institute of Singapore, 60Biopolis Street, Singapore 138672, Singapore, ¶Experimental Car-diology, Utrecht Medical Center, Heidelberglaan 100, 3584 CX,Utrecht, the Netherlands, �Division of Bioengineering, School ofChemical and Biomedical Engineering, Nanyang Technological Uni-versity, 50 Nanyang Avenue, Singapore 639798, Singapore, De-partments of ‡‡Surgery and ¶¶Biochemistry, National University ofSingapore, Lower Kent Ridge Road, Singapore 117597, Singapore

Received, October 12, 2006, and in revised form, April 16, 2007Published, MCP Papers in Press, June 11, 2007, DOI 10.1074/

mcp.M600393-MCP200

1 The abbreviations used are: MSC, mesenchymal stem cell; hESC,human embryonic stem cell; hESC-MSC, hESC-derived MSC; ITS,insulin, transferrin, and selenoprotein; CM, conditioned medium;NCM, non-conditioned medium; SCX, strong cation exchange; qRT,quantitative RT; mTOR, mammalian target of rapamycin; MAPK, mi-togen-activated protein kinase; STAT, signal transducers and activa-tors of transcription; Jak, Janus kinase; TGF, transforming growthfactor; DMEM, Dulbecco’s modified Eagle’s medium; PDGF, platelet-derived growth factor; FA, formic acid; GO, gene ontology; ECM,extracellular matrix; LOD, limit of detection.

Research

© 2007 by The American Society for Biochemistry and Molecular Biology, Inc.1680 Molecular & Cellular Proteomics 6.10This paper is available on line at http://www.mcponline.org

differentiation of MSCs but rather by paracrine factors se-creted by MSCs (8). These factors are postulated to promotearteriogenesis through paracrine mechanisms (7); support thestem cell crypt in the intestine (9); protect against ischemicrenal (10, 11), myocardial (12–15), and limb tissue injury (16);support and maintain hematopoiesis (17); and support theformation of megakaryocytes and proplatelets (18).

This paracrine hypothesis will introduce a radically differentdimension to the use of MSCs in regenerative medicine. In-stead of using cells, repair of injured tissues will be mediatedby enhancing endogenous tissue repair using biologics se-creted by MSCs. This will bypass the present confoundingissues associated with cell-based therapy, i.e. immunecompatibility, tumorigenicity, xenozootic infections, costs,and waiting time for ex vivo expansion of autologous cellpreparations. Such an approach will have a greater potentialfor the development of “off-the-shelf” MSC-based thera-peutics at affordable costs and with better quality controland consistency.

Numerous reports have invoked the secretion of paracrinefactors as a mechanism for the reparative effects of MSCs oninjured tissues (8, 19). However, there has been no systematicor comprehensive profiling of the paracrine proteome that willenable an adequate assessment of the general validity of theparacrine hypothesis and the relevance of this paracrine hy-pothesis to the wide spectrum of MSC-mediated therapeuticeffects.

In the present study we assessed the secretion proteome ofMSCs by performing multidimensional protein identificationtechnology (20) and cytokine antibody array analysis on achemically defined culture medium conditioned by theHuES9.E1 MSC line, one of three lines that we have previouslyderived from a human embryonic stem cell (hESC) line usinga clinically compliant protocol (21). These hESC-derived MSC(hESC-MSC) lines fulfilled the minimal criteria of a multipotentMSC and share the basic distinctive characteristics of adulttissue-derived MSCs, i.e. these cell lines are plastic-adherentwhen maintained in standard culture conditions; expressCD105, CD73, and CD90 and lack expression of CD45 andCD34; and can differentiate into osteoblasts, adipocytes, andchondroblasts (21, 22). Although hESC-derived MSCs resem-ble adult bone marrow-derived MSCs, there are differences.For example, genes that were preferentially expressed in theHuES9.E1 MSC line are associated with embryonic processessuch as proliferation and early developmental processes ofembryogenesis and segmentation, whereas those in bonemarrow-derived MSCs were over-represented in biologicalprocesses associated with more mature cell types such asmetabolic processes, cell structure, and late developmentalprocesses of skeletal development and muscle development(21). This phenomenon is consistent with the observation thatthe biology of MSCs isolated from fetal and adult tissues isdifferent and is characteristic of the developmental stage oftheir tissue of origin (24–26). In general, MSCs from younger

donors or developmentally less mature tissues are more pro-liferative and have a more robust differentiation potential andgreater therapeutic efficacy (24, 25). Therefore by extrapola-tion, MSCs derived from embryonic hESC lines will be moreproliferative and have a more robust differentiation potentialand greater therapeutic efficacy than those derived from adulttissues.

The use of these hESC-MSCs to characterize the secretoryproteome of MSCs offers several advantages over the use ofMSCs derived from adult tissues. 1) hESC-MSCs can bestably propagated in culture for at least 80 population dou-bling time as monitored by genome-wide gene expressionprofile and karyotyping using spectral karyotyping analysis(21), thereby ensuring a stable and renewable source of cellsfor repeated verifications during proteomics profiling. Thisadvantage is further bolstered by the reproducible generationof highly similar MSC cultures from either the same hESC lineor different hESC lines (21). 2) The use of hESCs in contrast tothe use of adult tissues, e.g. bone marrow as the tissuesource of MSCs, virtually ensures an infinitely renewable andconsistent tissue source and enhances the reproducible andconsistent batch to batch preparation of MSCs and thereforesecretory products. 3) We have also demonstrated that thehESC-MSC lines derived using our protocol are highly similarto single cell-derived hESC-MSC cultures and are thereforehomogenous (21).

Analysis of the secretory proteome of hESC-MSCs revealeda total of 201 unique gene products. 29 of these gene prod-ucts have been previously reported to be secreted by adulttissue-derived MSCs. Four other proteins that were reportedto be secreted by adult tissue-derived MSCs, namely IGFBP3,MIP3�, Oncostatin M, and TGF�3, were not present in our listof 201 gene products. The spectrum of secreted gene prod-ucts is consistent with the reported paracrine effects of MSCson different diverse cellular systems and diseases (9–18, 27)and provides a molecular basis for the use of hESC-MSC-conditioned medium in local or systemic treatment of dis-eases including repair of the heart after myocardial infarction.

EXPERIMENTAL PROCEDURES

Preparation of Conditioned Medium—HuES9.E1 cells were cul-tured as described previously (21). 80% confluent HuES9.E1 cellcultures were washed three times with PBS and cultured overnight ina chemically defined medium consisting of DMEM without phenol red(catalog number 31053, Invitrogen) and supplemented with insulin,transferrin, and selenoprotein (ITS) (Invitrogen), 5 ng/ml FGF2 (Invitro-gen), 5 ng/ml PDGF AB (Peprotech, Rocky Hill, NJ), glutamine-peni-cillin-streptomycin, and �-mercaptoethanol. The cultures were thenrinsed three times with PBS, and then fresh defined medium wasadded. After 3 days, the medium was collected and centrifuged at500 � g, and the supernatant was filtered using a 0.2-�m filter. ForLC-MS/MS analysis, the conditioned medium was placed in dialysiscassettes with molecular weight cutoff of 3500 (Pierce), dialyzedagainst three changes of 10 volumes of 0.9% NaCl, then concen-trated 20 times using Slide-A-Lyzer concentrating solution, and thendialyzed against 10 changes of 100 volumes of 0.9% NaCl beforefiltering with a 0.2-�m filter. The same volume of non-conditioned

Secretion Proteome of MSC

Molecular & Cellular Proteomics 6.10 1681

medium was dialyzed and concentrated in parallel with the condi-tioned medium.

Cytokine Antibody Blot Assays—1 ml of conditioned or non-con-ditioned medium was assayed for the presence of cytokines andother proteins using RayBio� Cytokine Antibody Arrays according tomanufacturer’s instructions (RayBio, Norcross, GA).

LC-MS/MS Analysis—Proteins in 2 ml of dialyzed conditioned me-dium (CM) or non-conditioned medium (NCM) were reduced, alky-lated, and digested with trypsin as described previously (20). Thesamples were then desalted by passing the digested mixture througha conditioned Sep-Pak C18 solid phase extraction cartridge (Waters,Milford, MA), washed twice with a 3% ACN (J. T. Baker Inc.) and 0.1%formic acid (FA) buffer, and eluted with a 70% ACN and 0.1% FAbuffer. The eluted samples were then dried to about 10% of theirinitial volumes by removing organic solvent in a SpeedVac. The sam-ples were kept at 4 °C prior to LC-MS/MS analysis. The desaltedpeptide mixture was analyzed by multidimensional protein identifica-tion technology with an LC-MS/MS system (LTQ, ThermoFinnigan,San Jose, CA). The sample was loaded into a strong cation exchange(SCX) column (Biobasic SCX, 5 �m, Thermo Electron, San Jose, CA)and fractionated by six salt steps with 50 �l of buffers (0, 2, 5, 10, 100,and 1000 mM ammonium chloride in 5% ACN and 0.1% FA) in the firstdimension. The peptides eluted from the SCX column were concen-trated and desalted in a Zorbax peptide trap (Agilent Technologies,Palo Alto, CA). The second dimensional chromatographic separationwas carried out with a home-packed nanobored C18 column (75-�minner diameter x 10 cm, 5-�m particles) directly into a PicoFrit nano-spray tip (New Objective, Woburn, MA) operating at a flow rate of 200nl/min with a 120-min gradient. The LTQ was operated in a data-de-pendent mode by performing MS/MS scans for the three most intensepeaks from each MS scan. The mass spectrometer was operated ata nanospray voltage of 1.8 kV, without sheath gas and auxiliary gasflow, at an ion transfer tube temperature of 180 °C, at a collision gaspressure of 0.85 millitorr, and with normalized collision energy at 35%for MS2. The ion selection threshold was set to 500 counts forinitiating MS2; activation quadrupole electric field was set to 0.25, andactivation time was set to 30 ms. The MS scan range was 400–1800m/z. Dynamic exclusion was activated with an exclusion duration of 1min. For each experiment, MS/MS (dta) spectra of the six salt stepswere extracted from the raw data files by the extract_msn program inBioworks 3.2 (ThermoFinnigan). The extracted dta files were com-bined into a single file with Mascot generic file format by a home-written program. Except the conversion of precursor mass from MH�

in dta to m/z in the Mascot generic file, the fragment ion m/z andintensity values were used as is without any manipulation. Proteinidentification was achieved by searching the combined data againstthe Internation Protein Index (IPI) human protein database (version3.15, 58,009 entries) via an in-house Mascot server (version 2.01). Thesearch parameters were as follows: a maximum of three missedcleavages using trypsin; fixed modification, carbamidomethylation;and variable modification, oxidation of methionine. The mass toler-ances were set to 2.0 and 0.8 Da for peptide precursor and fragmentions, respectively. The averaged Mascot identity score with signifi-cance threshold p � 0.05 is 41. A protein was accepted as a truepositive if two or more different peptides from the same protein werefound with ion scores greater than their Mascot identity score.

Bioinformatics—The validated proteins were collated by removingthe background proteins identified in the non-conditioned medium.The IPI identifier of each protein was then converted to the genesymbol by using the protein cross-reference table. Gene productswere classified into the different biological processes or pathways ofthe gene ontology (GO) classification system using GeneSpring GX7.3Expression Analysis software (Agilent Technologies), the frequency ofgenes in each process or pathway was compared with that in the

GenBankTM human genome database, and those processes of path-ways with significantly higher gene frequency (p � 0.05) were as-sumed to be significantly modulated by the secretion of MSC.

qRT-PCR—Total RNA was extracted from HuES9.E1 cells withTRIzol reagent (Invitrogen) and purified over a spin column (Nucle-ospin RNA II System, Macherey-Nagel GmbH and Co., Duren, Ger-many) according to the manufacturer’s protocol. 1 �g of total RNAwas converted to cDNA with random primers in a 50-�l reactionvolume using a High Capacity cDNA Archive kit (Applied Biosystems,Foster City, CA). The cDNA was diluted with distilled water to avolume of 100 �l. 1 �l was used for each primer set in pathway-specific RT2 Profiler PCR Arrays (SuperArray, Frederick, MD) accord-ing to the manufacturer’s protocol. The plates used for the analysiswere Chemokines and Receptors PCR Array (catalog number APH-022), NF�B Signaling Pathway PCR Array (catalog number APH-025),Inflammatory Cytokines and Receptors PCR Array (catalog numberAPH-011), Common Cytokine PCR Array (catalog number APH-021),and JAK/STAT Signaling Pathway PCR Array (catalog numberAPH-039).

RESULTS

Preparation of CM and NCM—To ensure that there wasminimal contamination of conditioned medium by mediumsupplements such as serum replacement medium, HuES9.E1MSCs were grown to about 80% confluency, washed threetimes with PBS, and incubated overnight in a chemicallydefined medium consisting of DMEM supplemented with ITS,5 ng/ml FGF2, 5 ng/ml PDGF AB, glutamine-penicillin-strep-tomycin, and �-mercaptoethanol. HuES9.E1 MSCs can bepropagated in this minimal medium for at least a week. Thenext day, the cell culture was again washed three times withPBS and incubated with the fresh defined medium. The me-dium was collected after 3 days of conditioning. The CM wasalways analyzed or processed in parallel with an equivalentvolume of NCM. For LC-MS/MS analysis, the medium wasconcentrated �10� before dialyzing extensively against0.9% saline as described under “Experimental Procedures.”The average protein concentrations of concentrated CM andNCM were 98.0 � 17.9 and 41.6 � 1.2 �g/ml (n � 3), respec-tively. The conditioning of medium by MSCs was monitoredby running aliquots of the medium on protein gels. Proteincomposition of the medium increased in complexity with time(Fig. 1). The CM had a more complex protein compositionthan NCM.

Analysis of MSC-conditioned Medium by LC-MS/MS andAntibody Array—LC-MS/MS analysis identified 247 proteinsthat were present in two independently prepared batches ofCM but not in a similarly processed NCM (Supplemental Table1). Together these 247 proteins are encoded by 132 uniqueknown genes (Supplemental Table 2a). There were 29 un-known proteins (Supplemental Table 2b).

MSCs have been shown to secrete a broad spectrum ofcytokines and growth factors that affect cells in their vicinity(8). Many of these factors are small molecules that are noteasily detectable during shotgun LC-MS/MS analysis. There-fore, the CM and NCM were also analyzed by hybridization tofive different antibody arrays that together carried antibodies

Secretion Proteome of MSC

1682 Molecular & Cellular Proteomics 6.10

against 101 cytokines/growth factors (Fig. 2 and Supplemen-tal Table 3a). 72 of the cytokines/growth factors were found tobe reproducibly secreted by HuES9.E1 MSCs in at least threeof four independently prepared batches of CM but not in NCM(Supplemental Table 3, b and c). However, only three geneproducts, namely IGFBP2, TIMP1, and TIMP2, were also de-tected by LC-MS/MS analysis possibly because many of cy-tokines and growth factors are small molecules and are notdetectable during conventional LC-MS/MS analysis.

Because the lists of 132 and 72 gene products identified byLC-MS/MS and antibody array analysis, respectively, havethree common genes, namely IGFBP2, TIMP1, and TIMP2,

the final tally was 201 unique gene products (Table I). 29 ofthese gene products, namely ENA78 (CXCL5), FGF4, FGF7,FGF9, GCP2 (CXCL6), granulocyte colony-stimulating factor,granulocyte/macrophage colony-stimulating factor, GRO-a,NCC4 (CCL16), hepatocyte growth factor, IGFBP1, IGFBP2,IGFBP4, IL1�, IL6, IL8, IP10 (CXCL10), leukocyte migrationinhibitory factor, MCP1 (CCL2), macrophage colony-stimulat-ing factor, macrophage migration inhibitory factor, Osteopro-tegerin, pulmonary and activation-regulated chemokine, pla-centa growth factor, stem cell factor, TGF�2, TIMP1, TIMP2,and vascular endothelial growth factor, have been previouslyreported to be secreted by adult tissue-derived MSCs (7, 13,16, 19, 28). Four other proteins that were reported to besecreted by adult tissue-derived MSCs, namely IGFBP3,MIP3�, Oncostatin M, and TGF�3 (Supplemental Table 3b),were not present in our list of 201 gene products

Verification by Genome-wide Gene Expression Analysis—Comparison of the 201 gene products to a genome-widegene expression profile of the hESC-MSCs generated byhybridizing total RNA to an Illumina BeadArray revealed that134 or 67% of the gene products had gene transcript levelsthat were present at above the limit of detection (LOD) with a99% confidence (Table I). Although 115 or 88% of the 132gene products identified by LC-MS/MS had detectable tran-script levels (Table I), only 27 or 38% of the 72 gene productsidentified by antibody array had detectable transcript levels,and 45 or 62% had no detectable transcript level (Table I).Probes for two of the gene products, ENO1B and SVEP1,were not present on the Illumina BeadArray. It is possible thattranscript levels for most of the 72 gene may be too low inabundance for detection by Illumina BeadArray because

FIG. 1. Protein analysis of medium conditioned by HuES9.E1MSC culture. 80% confluent HuES9.E1 cell cultures were washedthree times with PBS and cultured overnight in a chemically definedmedium consisting of DMEM without phenol red and supplementedwith ITS, 5 ng/ml FGF2, 5 ng/ml PDGF AB, glutamine-penicillin-streptomycin, and �-mercaptoethanol. The cultures were then rinsedthree times with PBS, and then fresh defined medium was added. Analiquot of medium was removed at 0, 24, 48, and 72 h and centrifugedat 500 � g, and the supernatant was filtered using a 0.2-�m filter. 10�l of the medium were separated by 4–12% SDS-PAGE, and proteinswere stained with silver.

FIG. 2. Antibody array. 1 ml of CM or NCM was incubated with a RayBio Cytokine Antibody Arrays according to the manufacturer’sinstructions. The antibody map for each array is listed in Supplemental Table 3a. Binding of ligands to specific antibody was visualized usinga horseradish peroxidase-based chemiluminescence assay. Different exposures of each membrane were analyzed. An antigen was scored aspresent if a signal was present on the membrane hybridized with CM but absent on that hybridized with NCM. Data from analysis of fourindependent batches of CM and NCM are summarized in Supplemental Table 1b.

Secretion Proteome of MSC

Molecular & Cellular Proteomics 6.10 1683

mRNAs encoding for cytokines/chemokines are known tocontain AU-rich elements that caused rapid degradation ofthe mRNA during translation (29, 30). More sensitive qRT-PCR assays were therefore performed. 42 of the 72 geneproducts were randomly selected and tested. 36 or 86% ofthe 42 gene products had detectable transcript levels definedas having a normalized Ct value of �35 (Table II). This fre-quency was similar to the 88% frequency observed for geneproducts identified by LC-MS/MS and whose gene transcriptswere detectable by Illumina BeadArray. In addition, all 15 ofthe 42 genes tested that had detectable transcript levels byIllumina BeadArray also had detectable transcript levels byqRT-PCR (Table II). 21 of 27 (78%) gene products that did nothave detectable transcript levels by Illumina BeadArray hadtranscript levels detectable by qRT-PCR (Table II).

Biological Processes That Are Modulated by the SecretedProteins—To investigate whether the secreted products havethe potential to repair the injured tissues or organs, geneproducts were first classified according to their biologicalprocesses and pathways according to GO. The frequency ofunique genes in the secreted MSC proteome associated witheach process or pathway was then compared with the genefrequency for the respective pathway or process in a data-base collated from Unigene, Entrez, and GenBank usingGeneSpring GX7.3 Expression Analysis software. Significantly

higher frequencies of genes (p � 0.05) were associated with58 biological processes and 30 pathways (Supplemental Ta-ble 4). The 58 biological processes could be approximatedinto the three major groups of metabolism, defense response,and tissue differentiation, whereas the 30 pathways could bebroadly categorized into receptor binding, signal transduc-tion, cell-cell interaction, cell migration, immune response,and metabolism (Figs. 3 and 4). The postulated biologicalprocesses and pathways both suggest that the secreted pro-teins have a major impact on the cellular metabolism that will

TABLE IAlphabetical list of 201 unique gene products identified by LC-MS/MS

and antibody array

The proteins identified by LC-MS/MS and antibody array as listed inSupplemental Tables 2a and 3b were combined and are representedby their gene symbol. The transcript level for each gene was assessedusing a high throughput Illumina BeadArray.

TABLE IIQuantitative RT-PCR assay for the presence of transcripts

42 of the 72 gene products identified by antibody array wererandomly selected for qRT-PCR analysis. 12 gene products havetranscript levels that were above the LOD at 99% confidence on theIllumina BeadArray, a high throughput genome-wide gene expressionassay. The Ct value for each gene was normalized against �-actin.

Symbol Illumina BeadArray Normalized Ct

1. BDNF �LOD 13.382. CCL2 �LOD 11.283. CCL7 �LOD 17.354. CCL8 �LOD 30.285. CXCL1 �LOD 14.096. CXCL12 �LOD 16.837. CXCL5 �LOD 19.938. IL1A �LOD 16.69. IL1B �LOD 11.4410. IL6 �LOD 18.9211. IL8 �LOD 8.5312. MIF �LOD 16.2313. MMP3 �LOD 15.9614. TGFB2 �LOD 15.1615. TNFRSF11B �LOD 12.2816. CCL1 �LOD 34.6717. CCL11 �LOD 26.1318. CCL23 �LOD 37.3319. CCL24 �LOD 10.8220. CCL26 �LOD 30.3821. CCL5 �LOD 27.9722. CSF1 �LOD 14.3523. CSF2 �LOD 23.9224. CSF3 �LOD 32.4725. CX3CL1 �LOD 25.0426. CXCL11 �LOD 22.3827. CXCR3 �LOD 28.4128. IL10 �LOD 27.429. IL12B �LOD 22.1730. IL13 �LOD 14.9631. IL16 �LOD 32.9832. IL2 �LOD 31.8733. IL3 �LOD 32.2434. IL7 �LOD 22.335. TGFB1 �LOD 10.6336. TNF �LOD 31.7237. CCL15 �LOD �3538. CCL16 �LOD �3539. CXCL13 �LOD �3540. IFNG �LOD �3541. VEGF �LOD �3542. XCL1 �LOD �35

Secretion Proteome of MSC

1684 Molecular & Cellular Proteomics 6.10

modulate energy production and the breakdown, biosynthe-sis, and secretion of macromolecules, processes essential forthe removal of damaged tissues and regeneration of newtissues (Figs. 3 and 4). Consistent with the predominant pres-ence of cytokines and chemokines in the MSC-conditionedmedium, the analysis also predicted that the secreted factorscould elicit many cellular responses that are dependent onexternal stimuli, .e.g. chemotaxis, taxis, and many immuneresponses (Fig. 3). Notably the conditioned medium couldalso induce biological processes that are important in tissuedifferentiation, particularly processes that promote vascular-ization, hematopoiesis, and bone development (Fig. 3). Inthose pathways predicted to be modulated by the secretedproteome, receptor-mediated binding of cytokine and ECMpathways were consistent with the predominance of cyto-kines and ECM components in the secreted proteome (Fig. 4).The main signal transduction pathways that could be acti-

vated by the secreted proteome include Jak-STAT signalingpathway, MAPK signaling pathway, Toll-like receptor signal-ing pathway, TGF� signaling pathway, mTOR signaling path-way, Fc�RI signaling pathway, and epithelial cell signaling inHelicobacter pylori infection. The computational analysis ofthe secreted proteome also suggested that MSC secretioncould enhance cell-cell interaction, migration, and immuneresponses.

DISCUSSION

MSCs have been used in preclinical and clinical trials totreat a myriad of diseases (3–5, 31–34). However, the under-lying mechanism remains unclear. Although MSCs have thepotential to differentiate into numerous cell type, e.g. endo-thelial cells, cardiomyocytes, and chondrocytes, that can po-tentially repair or regenerate damaged tissues, differentiationof MSCs into reparative cell types is generally too inefficient to

FIG. 3. Distribution of 201 gene products into biological processes. The 201 genes were classified into different biological processes inthe GO classification system. 58 biological processes that were over-represented by the frequency of genes in the secretory proteome relativeto the frequency of the genes in a database collated from Unigene, Entrez, and GenBank with a p value of �0.05 were grouped into three majorgroups: metabolism, defense response, and tissue differentiation (Supplemental Tables 4 and 5).

Secretion Proteome of MSC

Molecular & Cellular Proteomics 6.10 1685

Secretion Proteome of MSC

1686 Molecular & Cellular Proteomics 6.10

effectively replace damaged tissue or restore tissue function.There is increasing evidence that some of the therapeuticeffects of MSCs may be mediated by paracrine factors se-creted by MSCs (8). Therefore, elucidating the molecularcomposition of the secretion from MSCs will enhance ourunderstanding of the paracrine effects of MSCs. The signifi-cant similarity between hESC-derived MSCs and adult tissue-derived MSCs (21) suggests that conditioned medium of ei-ther MSC culture is likely to have similar biological activities.hESC-derived MSCs, however, have several advantages overadult tissue-derived MSCs. It has been observed that the ageof the donor and the developmental stage of the tissue fromwhich MSCs are derived have an impact on the proliferation,differentiation potential, and therapeutic efficacy of the MSCs.In general, younger donors and developmentally less maturetissues generate MSCs that are more proliferative and have amore robust differentiation potential and greater therapeuticefficacy (24–26). The use of hESC cell lines as a tissue sourceof MSC constitutes an infinitely renewable and expansibletissue source and enhances the reproducible and consistentbatch to batch preparation of MSCs. We have shown previ-ously that our protocol for generating MSCs from hESCs isnot only clinically compliant but also generates MSC culturesthat are homogenous and highly similar (21). The use ofhESC-MSC lines also enhances the scalability of preparingCM and the potential of developing low cost off-the-shelftherapeutics. In addition, the development of a serum-freechemically defined medium for the preparation of hESC-derived MSCs (CM) reduces confounding and variable con-taminants associated with complex medium supplementssuch as serum or serum replacement medium.

Here we describe the composition of the secreted pro-teome of hESC-MSCs through a combination of two tech-niques, LC-MS/MS and antibody arrays. Although shotgunproteomics analysis by LC-MS/MS is a sensitive techniqueand has high throughput capability, it is difficult to detectsmall proteins/peptides that include most of the cytokines,chemokines, and growth factors. This was partially mitigatedby the use of antibody arrays. The qualitative proteomic pro-file of the MSC secretion using the two techniques was highlyreproducible. Proteins identified by LC-MS/MS were presentin two independently prepared batches of CM, whereas thoseidentified by antibody array were present in at least three offour independently prepared batches of CM. The resultingproteomic profile of secretion by hESC-MSCs included al-most all the factors that were previously reportedly secretedby adult tissue-derived MSCs (7, 13, 16, 19, 28) includingtwice as many factors that have not been described. Therobustness of the proteomics profiling was further substanti-

ated by the detection of transcripts for 86–88% of geneproducts in the proteomic profile using a high throughputgenome-wide gene expression assay and quantitative RT-PCR assays. Although this proteomic profile was developedusing a population of cells rather than single cells, it is nev-ertheless a robust representation of hESC-derived MSC se-cretion for several reasons. First, the cell population used inthe elucidation of the secreted proteome can be reproduciblygenerated from either the same parental hESC line or a dif-ferent hESC line with a high degree of similarity betweensorted single cell-derived lines and unsorted population-derived lines (21). Second, the cell population maintained astable gene expression profile and karyotype for at least 60population doublings (21). Third, �99% of cells in this cellpopulation differentiate to form adipocytes when exposed toan adipocyte differentiation medium, and �90% of the cellsproduce proteoglycans when exposed to chondrocyte differ-entiation medium, suggesting that a large majority of cells areleast bipotential (21). Therefore, the elucidated proteome islargely reflective of an hESC-derived MSC population. How-ever, it remains to be determined whether this secreted pro-teome is unique to hESC-derived MSCs and not to otherhESC-derived cell populations or differentiated MSCs. A ma-jor hindrance in resolving this issue is that, unlike hESC-derived MSCs, we have not been able to maintain any hESC-derived cell population or differentiate MSCs using achemically defined medium without serum or serum replace-ment medium. The high protein content in serum and serumreplacement medium precludes any meaningful proteomicsanalysis of secretion by cells into the medium.

To evaluate and assess the potential functions of the MSCsecretion on a global scale, we utilized the more readily avail-able computational tools for gene expression analysis. Con-sistent with the predominance of cytokines and chemokinesin the secretion, computational analysis not unexpectedlypredicted many processes and pathways that are generallyassociated with the functions of cytokines and chemokinessuch as chemotaxis, taxis, cellular response to external stim-uli, breakdown, biosynthesis, and secretion of macromole-cules, cytokine-cytokine receptor interactions, cell-cell com-munication, and basal metabolism, e.g. glucose and aminoacid metabolism. Although these processes and pathwaysare not specific to the process of injury, repair, and regener-ation in any particular cell or tissue type, their facilitation ofimmune cell migration to the site of injury, ECM remodeling,and an increase in the cellular metabolism will have reparativeeffects on most injured or diseased tissues.

Aside from these generic pathways associated with cyto-kines and chemokines, computational analysis also predicted

FIG. 4. Distribution of 201 gene products into pathways. The 201 genes were classified into different pathways in the GO classificationsystem. 30 biological processes that were over-represented by the frequency of genes in the secretory proteome relative to the frequency ofthe genes in a database collated from Unigene, Entrez, and GenBank with a p value of �0.05 were categorized into several major categories:receptor binding, signal transduction, cell-cell interaction, cell migration, immune response, and metabolism (Supplemental Tables 4 and 5).

Secretion Proteome of MSC

Molecular & Cellular Proteomics 6.10 1687

that the secreted proteins regulate many processes involvedin vascularization, hematopoiesis, and skeletal development.Coincidentally most reported MSC-mediated tissue repairand regeneration are associated with cardiovascular, hema-topoietic, and musculoskeletal tissues (3–5, 31–34). Pathwayanalysis further uncovered candidate pathways that may beinvolved in mediating some of the paracrine effects of MSCs.In fact, many of these candidate pathways have already beenimplicated in many aspects of cardiovascular, hematopoietic,and musculoskeletal biology. For example, Jak-STAT signal-ing is associated with cardioprotection (35), hematopoiesis(36, 37), and skeletal repair and remodeling (38, 39); MAPKsignaling plays a crucial role in many aspects of cardiovas-cular responses (40, 41), skeletal repair and remodeling (39,42), and hematopoiesis (43); Toll-like receptor signaling hasbeen implicated in the initiation and progression of cardiovas-cular pathologies (44) and modulation of innate and adaptiveimmunity (45); TGF� signaling is critical in correct heart de-velopment cardiac remodeling, progression to heart failure,and vascularization (46–48), hematopoiesis (49), formationand remodeling of bone and cartilage (34, 50), and generalwound healing (51); and mTOR as an important regulator ofcell growth and proliferation plays a nonspecific yet criticalrole in both normal physiology and diseases (23, 52, 53).

In conclusion, our analysis of the secreted proteome inhESC-derived MSCs not only identified many of the cytokinesthat were previously reportedly to be secreted by adult tissue-derived MSCs but also more than 150 proteins that were notknown to be secreted by MSCs. Collectively the secretedproteome could potentially exert modulating effects on tissuerepair and regeneration particularly in the cardiovascular, he-matopoietic, and musculoskeletal tissues and therefore pro-vide molecular support for an MSC-mediated paracrine effecton tissue repair and regeneration in MSC transplantationstudies. Although the composition of secretion by MSCs invitro is consistent with the hypothesis that paracrine factorscould potentially mediate the tissue repair and regenerationobserved in MSC transplantation, validation of the hypothesiswill require demonstration that transplanted MSCs in vivo alsosecrete a similar spectrum of proteins or a spectrum of pro-teins that could support tissue repair and regeneration. Ourpresent elucidation of the in vitro MSC secretion proteome willnot only facilitate verification of this hypothesis by providingcandidates for in vivo verification, it also uncovered manyhighly testable hypotheses for the molecular mechanisms inMSC-mediated tissue repair and also potential “druggable”targets to modulate tissue repair and regeneration. Therefore,our elucidation of the CM is very relevant to the translation ofMSC-based biologics to clinical applications.

* This work was funded by a Biomedical Research Council (BMRC)grant (BMRC Project 01/1/21/17/045 to S. K. L.). The costs of publica-tion of this article were defrayed in part by the payment of page charges.This article must therefore be hereby marked “advertisement” in ac-cordance with 18 U.S.C. Section 1734 solely to indicate this fact.

□S The on-line version of this article (available at http://www.mcponline.org) contains supplemental material.

�� To whom correspondence and requests for materials should beaddressed: Inst. of Medical Biology, 11 Biopolis St., Helios 02-02,Singapore 138667, Singapore. E-mail: [email protected].

REFERENCES

1. Le Blanc, K., and Pittenger, M. (2005) Mesenchymal stem cells: progresstoward promise. Cytotherapy 7, 36–45

2. Reiser, J., Zhang, X. Y., Hemenway, C. S., Mondal, D., Pradhan, L., and LaRussa, V. F. (2005) Potential of mesenchymal stem cells in gene therapyapproaches for inherited and acquired diseases. Expert Opin. Biol. Ther.5, 1571–1584

3. Hui, J. H., Ouyang, H. W., Hutmacher, D. W., Goh, J. C., and Lee, E. H.(2005) Mesenchymal stem cells in musculoskeletal tissue engineering: areview of recent advances in National University of Singapore. Ann.Acad. Med. Singapore 34, 206–212

4. Caplan, A. I. (2005) Review: mesenchymal stem cells: cell-based recon-structive therapy in orthopedics. Tissue Eng. 11, 1198–1211

5. Menasche, P. (2005) The potential of embryonic stem cells to treat heartdisease. Curr. Opin. Mol. Ther. 7, 293–299

6. Laflamme, M. A., and Murry, C. E. (2005) Regenerating the heart. Nat.Biotechnol. 23, 845–856

7. Kinnaird, T., Stabile, E., Burnett, M. S., and Epstein, S. E. (2004) Bone-marrow-derived cells for enhancing collateral development: mecha-nisms, animal data, and initial clinical experiences. Circ. Res. 95,354–363

8. Caplan, A. I., and Dennis, J. E. (2006) Mesenchymal stem cells as trophicmediators. J. Cell. Biochem. 98, 1076–1084

9. Leedham, S. J., Brittan, M., McDonald, S. A., and Wright, N. A. (2005)Intestinal stem cells. J. Cell. Mol. Med. 9, 11–24

10. Togel, F., Hu, Z., Weiss, K., Isaac, J., Lange, C., and Westenfelder, C.(2005) Administered mesenchymal stem cells protect against ischemicacute renal failure through differentiation-independent mechanisms.Am. J. Physiol. 289, F31–F42

11. Patschan, D., Plotkin, M., and Goligorsky, M. S. (2006) Therapeutic use ofstem and endothelial progenitor cells in acute renal injury: ca ira. Curr.Opin. Pharmacol. 6, 176–183

12. Miyahara, Y., Nagaya, N., Kataoka, M., Yanagawa, B., Tanaka, K., Hao, H.,Ishino, K., Ishida, H., Shimizu, T., Kangawa, K., Sano, S., Okano, T.,Kitamura, S., and Mori, H. (2006) Monolayered mesenchymal stem cellsrepair scarred myocardium after myocardial infarction. Nat. Med. 12,459–465

13. Gnecchi, M., He, H., Noiseux, N., Liang, O. D., Zhang, L., Morello, F., Mu,H., Melo, L. G., Pratt, R. E., Ingwall, J. S., and Dzau, V. J. (2006) Evidencesupporting paracrine hypothesis for Akt-modified mesenchymal stemcell-mediated cardiac protection and functional improvement. FASEB J.20, 661–669

14. Gnecchi, M., He, H., Liang, O. D., Melo, L. G., Morello, F., Mu, H., Noiseux,N., Zhang, L., Pratt, R. E., Ingwall, J. S., and Dzau, V. J. (2005) Paracrineaction accounts for marked protection of ischemic heart by Akt-modifiedmesenchymal stem cells. Nat. Med. 11, 367–368

15. Mayer, H., Bertram, H., Lindenmaier, W., Korff, T., Weber, H., and Weich, H.(2005) Vascular endothelial growth factor (VEGF-A) expression in humanmesenchymal stem cells: autocrine and paracrine role on osteoblasticand endothelial differentiation. J. Cell. Biochem. 95, 827–839

16. Nakagami, H., Maeda, K., Morishita, R., Iguchi, S., Nishikawa, T., Takami,Y., Kikuchi, Y., Saito, Y., Tamai, K., Ogihara, T., and Kaneda, Y. (2005)Novel autologous cell therapy in ischemic limb disease through growthfactor secretion by cultured adipose tissue-derived stromal cells. Arte-rioscler. Thromb. Vasc. Biol. 25, 2542–2547

17. Van Overstraeten-Schlogel, N., Beguin, Y., and Gothot, A. (2006) Role ofstromal-derived factor-1 in the hematopoietic-supporting activity of hu-man mesenchymal stem cells. Eur. J. Haematol. 76, 488–493

18. Cheng, L., Qasba, P., Vanguri, P., and Thiede, M. A. (2000) Human mes-enchymal stem cells support megakaryocyte and pro-platelet formationfrom CD34� hematopoietic progenitor cells. J. Cell. Physiol. 184, 58–69

19. Liu, C. H., and Hwang, S. M. (2005) Cytokine interactions in mesenchymalstem cells from cord blood. Cytokine 32, 270–279

20. Washburn, M. P., Wolters, D., and Yates, J. R., III (2001) Large-scaleanalysis of the yeast proteome by multidimensional protein identification

Secretion Proteome of MSC

1688 Molecular & Cellular Proteomics 6.10

technology. Nat. Biotechnol. 19, 242–24721. Lian, Q., Lye, E., Suan Yeo, K., Khia Way Tan, E., Salto-Tellez, M., Liu, T. M.,

Palanisamy, N., El Oakley, R. M., Lee, E. H., Lim, B., and Lim, S. K. (2007)Derivation of clinically compliant MSCs from CD105�, CD24� differen-tiated human ESCs. Stem Cells 25, 425–436

22. Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F.,Krause, D., Deans, R., Keating, A., Prockop, D., and Horwitz, E. (2006)Minimal criteria for defining multipotent mesenchymal stromal cells. TheInternational Society for Cellular Therapy position statement. Cyto-therapy 8, 315–317

23. Tee, A. R., and Blenis, J. (2005) mTOR, translational control and humandisease. Semin. Cell Dev. Biol. 16, 29–37

24. Gotherstrom, C., West, A., Liden, J., Uzunel, M., Lahesmaa, R., and LeBlanc, K. (2005) Difference in gene expression between human fetal liverand adult bone marrow mesenchymal stem cells. Haematologica 90,1017–1026

25. Le Blanc, K. (2003) Immunomodulatory effects of fetal and adult mesen-chymal stem cells. Cytotherapy 5, 485–489

26. Zhang, H., Fazel, S., Tian, H., Mickle, D. A., Weisel, R. D., Fujii, T., and Li,R. K. (2005) Increasing donor age adversely impacts beneficial effects ofbone marrow but not smooth muscle myocardial cell therapy. Am. J.Physiol. 289, H2089–H2096

27. Kinnaird, T., Stabile, E., Burnett, M. S., Shou, M., Lee, C. W., Barr, S.,Fuchs, S., and Epstein, S. E. (2004) Local delivery of marrow-derivedstromal cells augments collateral perfusion through paracrine mecha-nisms. Circulation 109, 1543–1549

28. Haynesworth, S. E., Baber, M. A., and Caplan, A. I. (1996) Cytokine ex-pression by human marrow-derived mesenchymal progenitor cells invitro: effects of dexamethasone and IL-1�. J. Cell. Physiol. 166, 585–592

29. Barreau, C., Paillard, L., and Osborne, H. B. (2005) AU-rich elements andassociated factors: are there unifying principles? Nucleic Acids Res. 33,7138–7150

30. Espel, E. (2005) The role of the AU-rich elements of mRNAs in controllingtranslation. Semin. Cell Dev. Biol. 16, 59–67

31. Bhatia, R., and Hare, J. M. (2005) Mesenchymal stem cells: future sourcefor reparative medicine. Congest. Heart Fail. 11, 87–93

32. Mauney, J. R., Volloch, V., and Kaplan, D. L. (2005) Role of adult mesen-chymal stem cells in bone tissue engineering applications: current statusand future prospects. Tissue Eng. 11, 787–802

33. Zimmet, J. M., and Hare, J. M. (2005) Emerging role for bone marrowderived mesenchymal stem cells in myocardial regenerative therapy.Basic Res. Cardiol. 100, 471–481

34. Lin, Z., Willers, C., Xu, J., and Zheng, M. H. (2006) The chondrocyte: biologyand clinical application. Tissue Eng. 12, 1971–1984

35. Negoro, S., Kunisada, K., Tone, E., Funamoto, M., Oh, H., Kishimoto, T.,

and Yamauchi-Takihara, K. (2000) Activation of JAK/STAT pathwaytransduces cytoprotective signal in rat acute myocardial infarction. Car-diovasc Res. 47, 797–805

36. Ward, A. C., Touw, I., and Yoshimura, A. (2000) The Jak-Stat pathway innormal and perturbed hematopoiesis. Blood 95, 19–29

37. Ungureanu, D., and Silvennoinen, O. (2005) SLIM trims STATs: ubiquitin E3ligases provide insights for specificity in the regulation of cytokine sig-naling. Sci. STKE 2005, pe49

38. Blair, H. C., Robinson, L. J., and Zaidi, M. (2005) Osteoclast signallingpathways. Biochem. Biophys. Res. Commun. 328, 728–738

39. Malemud, C. J. (2004) Protein kinases in chondrocyte signaling and osteo-arthritis. Clin. Orthop. Relat. Res. Suppl. 427, S145–S151

40. Nigro, J., Osman, N., Dart, A. M., and Little, P. J. (2006) Insulin resistanceand atherosclerosis. Endocr. Rev. 27, 242–259

41. Pandya, N., Santani, D., and Jain, S. (2005) Role of mitogen-activatedprotein (MAP) kinases in cardiovascular diseases. Cardiovasc. Drug Rev.23, 247–254

42. Berenbaum, F. (2004) Signaling transduction: target in osteoarthritis. Curr.Opin. Rheumatol. 16, 616–622

43. Platanias, L. C. (2003) Map kinase signaling pathways and hematologicmalignancies. Blood 101, 4667–4679

44. de Kleijn, D., and Pasterkamp, G. (2003) Toll-like receptors in cardiovas-cular diseases. Cardiovasc. Res. 60, 58–67

45. Ozato, K., Tsujimura, H., and Tamura, T. (2002) Toll-like receptor signalingand regulation of cytokine gene expression in the immune system.BioTechniques Suppl: 66–68, 70, 72 passim

46. Euler-Taimor, G., and Heger, J. (2006) The complex pattern of SMADsignaling in the cardiovascular system. Cardiovasc. Res. 69, 15–25

47. Bertolino, P., Deckers, M., Lebrin, F., and ten Dijke, P. (2005) Transforminggrowth factor-� signal transduction in angiogenesis and vascular disor-ders. Chest 128, 585S–590S

48. Bobik, A. (2006) Transforming growth factor-�s and vascular disorders.Arterioscler. Thromb. Vasc. Biol. 26, 1712–1720

49. Ruscetti, F. W., Akel, S., and Bartelmez, S. H. (2005) Autocrine transforminggrowth factor-� regulation of hematopoiesis: many outcomes that de-pend on the context. Oncogene 24, 5751–5763

50. Janssens, K., ten Dijke, P., Janssens, S., and Van Hul, W. (2005) Trans-forming growth factor-�1 to the bone. Endocr. Rev. 26, 743–774

51. Faler, B. J., Macsata, R. A., Plummer, D., Mishra, L., and Sidawy, A. N.(2006) Transforming growth factor-� and wound healing. Perspect. Vasc.Surg. Endovasc. Ther. 18, 55–62

52. Wullschleger, S., Loewith, R., and Hall, M. N. (2006) TOR signaling ingrowth and metabolism. Cell 124, 471–484

53. Proud, C. G. (2004) Ras, PI3-kinase and mTOR signaling in cardiac hyper-trophy. Cardiovasc. Res. 63, 403–413 Vol. , No.

Secretion Proteome of MSC

Molecular & Cellular Proteomics 6.10 1689