human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

8
Human Mesenchymal Stem Cells Play a Dual Role on Tumor Cell Growth In Vitro and In Vivo LIN LI HUI TIAN, * WEIMING YUE, FENG ZHU, SHUHAI LI, AND WENJUN LI Department of Thoracic Surgery, Qilu Hospital, Shandong University, Jinan, P.R. China Nowadays, some evidences demonstrate that human mesenchymal stem cells (hMSCs) favor tumor growth; however, others show that hMSCs can suppress tumorigenesis and tumor growth. With the indeterminateness of the effect of hMSCs on tumors, we investigated the effect of hMSCs on lung cancer cell line A549 and esophageal cancer cell line Eca-109 in vitro and in vivo. Our results revealed that hMSCs inhibited the proliferation and invasion of A549 and Eca-109 cells, arrested tumor cells in the G1 phase of the cell cycle and induced the apoptosis of tumor cells in vitro by using a co-culture system and the hMSCs-conditioned medium. However, animal study showed that hMSCs enhanced tumor formation and growth in vivo. Western blotting and immunoprecipitation data showed that the expressions of proliferating cell nuclear antigen (PCNA), Cyclin E, phospho-retinoblastoma protein (pRb), B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-xL, and matrix metalloproteinase 2 (MMP-2) were downregulated and the formation of Cyclin E-cyclin-dependent kinase 2 (CDK2) complexes was inhibited in the tumor cells treated with the hMSCs-conditioned medium. According to the observation of tumor mass and the result of microvessel density (MVD), we found that the promoting role of hMSCs on tumor growth was related with the increase of tumor vessel formation. Our present study suggests that hMSCs have a contradictory effect on tumor cell growth between in vitro and in vivo, and therefore, the exploitation of hMSCs in new therapeutic strategies should be cautious under the malignant conditions. J. Cell. Physiol. 226: 1860–1867, 2011. ß 2010 Wiley-Liss, Inc. Mesenchymal stem cells (MSCs), the stromal progenitor stem cells found in the bone marrow (BM), which can be defined according to its ability to self-renew and differentiate into tissues of mesodermal and non-mesodermal cell lineages (osteocytes, adipocytes, chondrocytes, myocytes, cardiomyocytes, fibroblasts, myofibroblasts, epithelial cells, and neurons), play a crucial role in supporting hematopoiesis (Pittenger et al., 1999; Zhang et al., 2003). Due to their differentiation plasticity, MSCs have been widely used in cell therapy and tissue regeneration fields, such as bone and cardiovascular repair (Mathiasen et al., 2009; Undale et al., 2009). Human mesenchymal stem cells (hMSCs) used in the experiment are mostly acquired from adult BM. There is evidence showing that hMSCs have a potent immunosuppressive ability by inhibiting T-lymphocyte proliferation and thus suppressing graft-versus-host disease (Di Nicola et al., 2002; Le et al., 2004). In addition, hMSCs have received intensive attention in the field of tumors. On the one hand, along with their property of migrating to tumor sites, hMSCs have been regarded as the most promising delivery vehicle for a cell-based targeted cancer gene therapy in the future (Roorda et al., 2009); on the other hand, many studies indicate that hMSCs have several tumor-growth promoting functions in the tumor microenvironment, including expressing growth factors, enhancing tumor vessel formation, and creating tumor stem cell niches (Roorda et al., 2009). However, there are many contradictory results observed in the other studies. For example, in the Kaposi’s sarcoma and hepatoma animal models, hMSCs exerted an anti-tumorigenic effect by homing to sites of tumorigenesis via the intravenous injection or developing an animal transplantation model via the subcutaneous co-injection with tumor cell (Khakoo et al., 2006; Qiao et al., 2008). Thereafter, so far, there has not been a congruous conclusion on the effect of hMSCs on tumor progress. In order to better understand how hMSCs affect tumors, here, we investigated the influence of hMSCs on lung cancer cell line A549 and esophageal cancer cell line Eca-109 growth in vitro and in vivo. Our findings showed that hMSCs inhibited tumor cells proliferation and invasion in vitro; but, in vivo, hMSCs favored tumor formation and growth. Materials and Methods Cell culture and preparation of hMSCs-conditioned medium hMSCs (Cyagen, Chicago) were derived from BM of healthy adults and cultured in hMSCs growth medium (Cyagen) supplemented with 10% fetal bovine serum (FBS; Cyagen). Human esophageal cancer cell line Eca-109 and human lung cancer cell line A549 (SIBCB, Shanghai, China) were cultured in Rosewell’s Park Memorial Institute (RPMI) 1640 supplemented with 10% FBS (GIBCO, Gaithersburg) and 100 U/ml penicillin and 100 mg/ml streptomycin. All cells were incubated at 378C in 5% CO 2 humidified cell culture incubator. When hMSCs were 80–90% confluent in the culture flasks, the medium was collected, filtered through a 0.22-mm filter and stored at 808C until used as the hMSCs-conditioned medium. Cells co-culture and treatment with conditioned medium Co-culture systems were established by using a transwell (0.4 mm pore, polycarbonate membrane; Costar, Cambridge) of 6-well plates. A549 or Eca-109 cells suspensions (1 ml, 5 10 cells) were loaded in the upper inserts, and hMSCs cell suspensions (2 ml, 5 10 5 cells) were put into the lower compartment of the culture well. The lower compartment of control groups had only hMSCs growth medium (2 ml) not containing FBS. Every group had three Contract grant sponsor: National Natural Science Foundation of China; Contract grant number: 30571844. Contract grant sponsor: Shandong Province Natural Science Foundation in China; Contract grant number: 2009ZRB14005. *Correspondence to: Hui Tian, Department of Thoracic Surgery, Qilu Hospital, Shandong University, 107#, Wenhua Xi Road, Jinan 250012, P.R. China. E-mail: [email protected] Received 6 July 2010; Accepted 21 October 2010 Published online in Wiley Online Library (wileyonlinelibrary.com), 10 November 2010. DOI: 10.1002/jcp.22511 ORIGINAL RESEARCH ARTICLE 1860 Journal of Journal of Cellular Physiology Cellular Physiology ß 2010 WILEY-LISS, INC.

Upload: lin-li

Post on 11-Jun-2016

216 views

Category:

Documents


1 download

TRANSCRIPT

Page 1: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

Human Mesenchymal Stem CellsPlay a Dual Role on Tumor CellGrowth In Vitro and In VivoLIN LI HUI TIAN,* WEIMING YUE, FENG ZHU, SHUHAI LI, AND WENJUN LI

Department of Thoracic Surgery, Qilu Hospital, Shandong University, Jinan, P.R. China

Nowadays, some evidences demonstrate that human mesenchymal stem cells (hMSCs) favor tumor growth; however, others show thathMSCs can suppress tumorigenesis and tumor growth.With the indeterminateness of the effect of hMSCs on tumors, we investigated theeffect of hMSCs on lung cancer cell line A549 and esophageal cancer cell line Eca-109 in vitro and in vivo. Our results revealed that hMSCsinhibited the proliferation and invasion of A549 and Eca-109 cells, arrested tumor cells in the G1 phase of the cell cycle and induced theapoptosis of tumor cells in vitro by using a co-culture system and the hMSCs-conditioned medium. However, animal study showed thathMSCs enhanced tumor formation and growth in vivo. Western blotting and immunoprecipitation data showed that the expressions ofproliferating cell nuclear antigen (PCNA), Cyclin E, phospho-retinoblastoma protein (pRb), B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-xL,and matrix metalloproteinase 2 (MMP-2) were downregulated and the formation of Cyclin E-cyclin-dependent kinase 2 (CDK2)complexeswas inhibited in the tumor cells treatedwith the hMSCs-conditionedmedium. According to the observation of tumormass andthe result of microvessel density (MVD), we found that the promoting role of hMSCs on tumor growth was related with the increase oftumor vessel formation. Our present study suggests that hMSCs have a contradictory effect on tumor cell growth between in vitro and invivo, and therefore, the exploitation of hMSCs in new therapeutic strategies should be cautious under the malignant conditions.J. Cell. Physiol. 226: 1860–1867, 2011. � 2010 Wiley-Liss, Inc.

Mesenchymal stem cells (MSCs), the stromal progenitor stemcells found in the bone marrow (BM), which can be definedaccording to its ability to self-renew and differentiate intotissues of mesodermal and non-mesodermal cell lineages(osteocytes, adipocytes, chondrocytes, myocytes,cardiomyocytes, fibroblasts,myofibroblasts, epithelial cells, andneurons), play a crucial role in supporting hematopoiesis(Pittenger et al., 1999; Zhang et al., 2003). Due to theirdifferentiation plasticity, MSCs have been widely used in celltherapy and tissue regeneration fields, such as bone andcardiovascular repair (Mathiasen et al., 2009; Undale et al.,2009). Human mesenchymal stem cells (hMSCs) used in theexperiment are mostly acquired from adult BM. There isevidence showing that hMSCs have a potentimmunosuppressive ability by inhibiting T-lymphocyteproliferation and thus suppressing graft-versus-host disease (DiNicola et al., 2002; Le et al., 2004). In addition, hMSCs havereceived intensive attention in the field of tumors. On the onehand, along with their property of migrating to tumor sites,hMSCs have been regarded as the most promising deliveryvehicle for a cell-based targeted cancer gene therapy in thefuture (Roorda et al., 2009); on the other hand, many studiesindicate that hMSCs have several tumor-growth promotingfunctions in the tumor microenvironment, including expressinggrowth factors, enhancing tumor vessel formation, and creatingtumor stem cell niches (Roorda et al., 2009).

However, there are many contradictory results observed inthe other studies. For example, in the Kaposi’s sarcoma andhepatoma animal models, hMSCs exerted an anti-tumorigeniceffect by homing to sites of tumorigenesis via the intravenousinjection or developing an animal transplantation model via thesubcutaneous co-injectionwith tumor cell (Khakoo et al., 2006;Qiao et al., 2008). Thereafter, so far, there has not been acongruous conclusion on the effect of hMSCs on tumorprogress. In order to better understand how hMSCs affecttumors, here, we investigated the influence of hMSCs on lungcancer cell line A549 and esophageal cancer cell line Eca-109growth in vitro and in vivo. Our findings showed that hMSCsinhibited tumor cells proliferation and invasion in vitro; but, invivo, hMSCs favored tumor formation and growth.

Materials and MethodsCell culture and preparation of hMSCs-conditioned medium

hMSCs (Cyagen, Chicago) were derived from BM of healthy adultsand cultured in hMSCs growth medium (Cyagen) supplementedwith 10% fetal bovine serum (FBS; Cyagen). Human esophagealcancer cell line Eca-109 and human lung cancer cell line A549(SIBCB, Shanghai, China) were cultured in Rosewell’s ParkMemorial Institute (RPMI) 1640 supplemented with 10% FBS(GIBCO, Gaithersburg) and 100U/ml penicillin and 100mg/mlstreptomycin. All cells were incubated at 378C in 5% CO2

humidified cell culture incubator. When hMSCs were 80–90%confluent in the culture flasks, the medium was collected, filteredthrough a 0.22-mm filter and stored at �808C until used as thehMSCs-conditioned medium.

Cells co-culture and treatment with conditioned medium

Co-culture systems were established by using a transwell (0.4mmpore, polycarbonate membrane; Costar, Cambridge) of 6-wellplates. A549 or Eca-109 cells suspensions (1ml, 5� 10 cells) wereloaded in the upper inserts, and hMSCs cell suspensions (2ml,5� 105 cells) were put into the lower compartment of the culturewell. The lower compartment of control groups had only hMSCsgrowth medium (2ml) not containing FBS. Every group had three

Contract grant sponsor: National Natural Science Foundation ofChina;Contract grant number: 30571844.Contract grant sponsor: Shandong Province Natural ScienceFoundation in China;Contract grant number: 2009ZRB14005.

*Correspondence to: Hui Tian, Department of Thoracic Surgery,Qilu Hospital, Shandong University, 107#, Wenhua Xi Road, Jinan250012, P.R. China. E-mail: [email protected]

Received 6 July 2010; Accepted 21 October 2010

Published online in Wiley Online Library(wileyonlinelibrary.com), 10 November 2010.DOI: 10.1002/jcp.22511

ORIGINAL RESEARCH ARTICLE 1860J o u r n a l o fJ o u r n a l o f

CellularPhysiologyCellularPhysiology

� 2 0 1 0 W I L E Y - L I S S , I N C .

Page 2: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

wells. The number of tumor cells in the inserts of the co-culturesystems was counted bymicroscope after incubating for 48 h. Cellsin every insert were counted for three times, and the results werethe mean value.

For the observation of the effect of hMSCs-conditionedmediumon tumor cell proliferation, A549 and Eca-109 cells were,respectively, inoculated at a density of 5� 105 cells per wellof 6-well plates in the mixture of tumor cell culture medium(supplemented with 10% FBS) and hMSCs-conditioned medium(1:1); as controls, tumor cells grew in the mixture of tumor cellculture medium (supplemented with 10% FBS) and hMSCs growthmedium not containing FBS (1:1). Every group had three wells.After cultured for 48 h, tumor cells per well were, respectively,collected and counted, and the results were the mean value.

Flow cytometric analysis

For cell cycle and apoptosis analysis, A549 or Eca-109 cells werecultured in the mixture of RPMI 1640 medium supplemented with2% FBS which favors apoptosis and hMSCs-conditioned medium(1:1), and as controls, the cells were cultured with in the mixmedium of RPMI 1640 medium supplemented 2% FBS and hMSCsgrowth medium not containing FBS (1:1). After 24 h, a portion oftumor cells were collected and fixed with 70% ice-cold ethanol forcell-cycle analysis using Cell Cycle Detection Kit (KeyGEN,Nanjing, China) including RNase A and propidium iodide (PI). Therest of tumor cells were cultured for additional of 24 h (total 48 h)and collected by digesting with 0.25% trypsin without EDTA forapoptosis analysis using Annexin V-FITCCell Apoptosis DetectionKit (KeyGEN) including Binding Buffer, Annexin V-FITC, and PI.The cell cycle and apoptosis were analyzed by flow cytometry(FACSCalibur, Franklin Lakes). Datawere analyzed by the softwareModFit and WinMDI2.9.

Assay of caspase-3 activity

Cellular caspase-3 activity was determined by use of colorimetricassay kits (Beyotime Institute of Biotechnology, China). At first,A549 or Eca-109 cells were cultured in the mixture of RPMI 1640medium supplemented with 2% FBS and hMSCs-conditionedmedium (1:1); as the control, cells were cultured in themixmediumof RPMI 1640 medium supplemented with 2% FBS and hMSCsgrowth medium not containing FBS (1:1). After cultured for 48 h,cells were treated according to the manufacturer’s instructions.Cell lysates were prepared and assays were performed on 96-wellmicrotiter plates by incubating 10ml protein of cell lysate persample in 80ml reaction buffer (1% NP-40, 20mM Tris–HCl,137mM Nad, and 10% glycerol) containing 10ml caspase-3substrate (Ac-DEVD-pNA). Lysates were incubated at 378C for4 h. Samples were measured with the Microplate Reader (Rayto,Shenzhen, China) at an absorbance of 405 nm. Caspase-3 activitieswere expressed as the percentage of enzyme activity compared tocontrol. All experiments were carried out in triplicate.

Cell invasion experiment

Transwell chambers (6.5mm diameter inserts, 8.0mm pore,polycarbonate membrane; Costar) of 24-well plates were used incell invasion experiments. The membrane at the bottom of thetranswell chamber was evenly coated with 50ml Matrigel (BD,Franklin Lakes) before adding cells suspensions. Tumor cells werecultured for 24 h in the mixture of tumor cell culture medium andhMSCs-conditioned medium (1:1), and in control groups tumorcells were cultured for 24 h in the tumor cell culture mediumsupplemented with 50% hMSCs growth medium not containingFBS, and then tumor cells were collected and resuspended in theFBS-free RPMI 1640 medium at a concentration of 5� 105 cellper ml by cell counting for three times. Then, the cell suspensionswere put into the upper compartment of the transwell chambers(200ml/well), and RPMI 1640 medium containing 10% FBS was putinto the lower compartment (600ml/well). After cultured at 378C

for 24 h, the cells that did not penetrate the polycarbonatemembrane at the bottom of the chamber were wiped off withcotton stickers. The membrane was removed and fixed withmethanol and stained with Wright’s Stain or eosin solution. Fivevision fields were randomly selected under a ZX81 microscope(Olympus, Tokyo, Japan) and the number of cells that penetratedthe membrane was counted. Each group consisted of fourduplicates. The invasion inhibition rate was calculated using thefollowing equation: invasion inhibition rate¼ (number of countedcells in the control group� number of counted cells in the grouptreated with hMSCs-conditioned medium)/number of countedcells in the control group� 100%.

Western blotting and immunoprecipitation

Eca-109 or A549 cells were cultured in the mixture of tumor cellculture medium and hMSCs conditioned medium (1:1) for 24 h; incontrol groups, tumor cells were cultured in the tumor cell culturemedium supplemented with 50% hMSCs growth medium notcontaining FBS. For Western blotting assay, derived separatelyfromEca-109 orA549 cells by the treatment of lysis buffer, 25mg ofproteins were subjected to SDS–PAGE analysis, transferred tonitrocellulose membranes and incubated with primary antibodiesagainst proliferating cell nuclear antigen (PCNA), Cyclin D1, CyclinE, pRb, phospho-retinoblastoma protein (phospho-pRb), P27, B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-xL, Bax (Labvision,Fremont), matrix metalloproteinase 2 (MMP-2) andb-actin (Sigma,St. Louis). Protein bands were detected using an enhancedchemiluminescence reagent (Sigma) following incubation withhorseradish peroxidase-conjugated secondary antibodies (Sigma).Optical band density was quantified (Imager of Alpha Corporation,San Leandro). In order to accomplish the immunoprecipitationassay, 500mg of protein from cell lysates of A549 and Eca-109 waspre-cleared with protein A-Sepharose (Amersham Pharmacia,Uppsala, Sweden), followed by immunoprecipitation usingmonoclonal anti-cyclin-dependent kinase 2 (CDK2; Santa Cruz,Santa Cruz) antibody. Immune complexes were harvested withprotein A, and then immunoprecipitated proteinswere analyzed bySDS–PAGE, as above. Immunodetection was carried out by usingmonoclonal anti-CDK2 and -Cyclin E antibodies.

Animal experiments

Six-week-old female BALB/c nude mice were obtained from theInstitute of Zoology, Chinese Academy of Sciences. All procedureswere carried out in accordance with the advice and permission ofthe Institutional Ethical Committee. Tumor cells and hMSCs wereprepared either as single-cell type suspensions (5� 106 A549 orEca-109 cells in 0.1ml PBS) or amix of cells (5� 106 tumor cells and1� 106 hMSCs in 0.1ml PBS). Cells were injected subcutaneouslyat one site of the back of nude mice. Mice were examined one timeat 5 days and tumor growth was evaluated by measuring the lengthandwidth of tumormass. Palpable tumors at the injection siteswitha size of more than 3mm in diameters were monitored as a tumor.At the end of the experiment, animals were killed and tumormasses were removed and fixed in 10% neutral buffered formalinsolution for histologic preparations.

Immunohistochemistry

All tumor masses fixed in 10% neutral formalin were embedded inparaffin at 558C, and cut into parallel consecutive 4-mm thicksections for the subsequent immunohistochemical study. Briefly,the endogenous peroxidase activitywas blockedwith 0.1%H2O2 inmethanol for 20min. The sections were permeabilized withethylene diamine tetraacetic acid (EDTA) buffer solution (pH 9.0)for 15min with microwave. Then, the sheep serumwas applied for30min to prevent unspecific adherence of serumproteins followedby treatment with primary antibody (mouse anti-CD34 diluted for1:100; Serotec, Oxford, UK) at 378C for 2 h. After washed withPBS, sections were then incubated with biotin labeled secondary

JOURNAL OF CELLULAR PHYSIOLOGY

A D U A L R O L E O F h M S C s O N T U M O R C E L L G R O W T H 1861

Page 3: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

antibody (goat anti-mouse HRP IgG; Serotec) diluted for 1:100 at378C for 30min and washed with PBS. Peroxidase conjugatedstreptavidin was added for 20min and then washed with PBS.Finally, sections were developed with 3,3-diaminobenzidine andhydrogen peroxide and counterstained with hematoxylin. For thenegative control, PBS was used instead of primary antibody. Thesections were analyzed by light microscopy.

Detection of microvessel density (MVD)

MVD was assessed by immunohistochemical analysis withantibodies to the endothelial marker CD34 according to themethod of Weidner et al. (1991). Briefly, three most vascularizedareas detected by CD34 were initially selected (so-called hotspots) under 40� field. Then,microvesselswere counted in each ofthese areas under a 200� field. Any yellow brown stained singleendothelial cell or clusters of endothelial cells, with or without a

Fig. 1. Proliferation of tumor cells is inhibited by hMSCs in vitro.A: Eca-109 andA549 cells were, respectively, co-culturedwith hMSCsby using the transwell co-culture systems or treated with the hMSCs-conditioned medium for 48 h, and then, cells were harvested forcounting.Theresultsareshownaspercentageofcellnumber(mean)incomparison with that of controls not co-cultured with hMSCs or nottreatedwith hMSCs-conditionedmedium (MP<0.01). B:After treatedwiththehMSCs-conditionedmediumfor24 h, tumorcellsaccumulateinG1phase by flow cytometric analysis for cell cycle. The results werethe means (WSD) of three independent experiments.

Fig. 2. hMSCs induce tumor cells apoptosis in vitro. A: A549and Eca-109 cells cultivated in the presence or absence of thehMSCs-conditionedmedium for 48 hwere tested for cell apoptosis byflow cytometric analysis. The figures are representative of threeexperiments. B: The apoptotic rates of A549 and Eca-109 cellsuntreated or treated with hMSCs-conditioned medium. C: Therelative caspase-3 activity in A549 and Eca-109 cells treated with thehMSCs-conditioned medium for 48 h as compared to untreatedcontrol. The results were the means (WSD) of three experiments(MP<0.01).

JOURNAL OF CELLULAR PHYSIOLOGY

1862 L I E T A L .

Page 4: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

lumen, were considered to be individual vessels. Themean value ofthree 200� field counts was recorded as the MVD of the section.All counts were performed by three investigators in a blindedmanner. Microvessel counts were compared between theobservers and discrepant results were reassessed. The consensuswas used as the final score for analysis.

Statistical analysis

All statistical analyses were performed using SPSS 15.0 softwarepackage. Statistical significance was assessed by comparing meanvalues (means� SD). The significance level was set at P< 0.05.

ResultsProliferation of tumor cells is inhibited by hMSCs in vitro

There has been the evidence showing that the hMSCs-inducedinhibition of tumor cells proliferation was mediated by thesoluble factors in the hMSCs-conditioned medium releasedfrom hMSCs cultures (Qiao et al., 2008). Consistently, weobtained supportive results. In the co-culture systems, thenumbers of Eca-109 and A549 cells co-cultured with hMSCs for48 h were, respectively, 8.61� 0.87� 105 and9.46� 0.93� 105. However, the numbers of Eca-109 and A549cells of control groups were, respectively, 13.46� 0.83� 105

and 14.33� 1.03� 105. As compared to control groups, thehMSCs-induced inhibitory effect on tumor cells proliferationwas conspicuous (P< 0.01). Similar to the co-culture, thenumbers of Eca-109 and A549 cells treated with hMSCs-conditioned medium (8.97� 0.18� 105 and10.05� 0.19� 105) also dramatically decreased as comparedto controls (12.92� 0.29� 105 and 14.03� 0.35� 105)

(P< 0.01), and moreover, the inhibitory effect of treatmentwith conditioned medium on tumor cells proliferationapproximated to that of co-culture (Fig. 1A). Besides, cell cycleassay by flow cytometry showed that the proportion of G1phase of Eca-109 and A549 cells treated with the hMSCs-conditioned medium was far higher than that of the controlgroups (P< 0.01, Fig. 1B). This demonstrated that hMSCs couldprevent the normal running of tumor cell cycle and inhibit theproliferation of tumor cells.

hMSCs promote the apoptosis of tumor cells in vitro

In order to investigate the effect of the hMSCs-conditionedmedium on the apoptosis of tumor cells, we tested theapoptotic rate of A549 and Eca-109 cells treated with thehMSCs-conditioned medium by flow cytometry. Our findingsshowed that the apoptotic rate significantly increased whentumor cells were cultured in themixture of RPMI 1640mediumand hMSCs-conditioned medium (P< 0.01, Fig. 2A,B). Inaddition, since the activation of caspase-3 proteases is crucial inapoptotic cell death (Kumar and Lavin, 1996), we alsodetermined the caspase-3 activity in tumor cells treated withthe hMSCs-conditioned medium. The results showed thatthere was a significant and nearly twofold increase in caspase-3activity in the treated tumor cells as compared to controls(P< 0.01, Fig. 2C).

Invasion of tumor cells is inhibited by the hMSCs-conditioned medium

A549 and Eca-109 cells that penetrated the membranedisplayed, respectively, red and purple by stain with eosin

Fig. 3. Invasion of tumor cell is inhibited by the hMSCs-conditioned medium. Red A549 cells and purple Eca-109 cells that penetrated thepolycarbonate membrane by stain with eosin solution andWright’s Stain solution were counted (magnification, 200T). A549 and Eca-109 cellswere cultivated in thepresence (B,D)or absence (A,C)of hMSCs-conditionedmedium for 24 hbefore cell suspensionswereput into the transwellchambers. Figures were the representatives of invasion experiments.

JOURNAL OF CELLULAR PHYSIOLOGY

A D U A L R O L E O F h M S C s O N T U M O R C E L L G R O W T H 1863

Page 5: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

solution and Wright’s Stain solution (Fig. 3). The numbers oftumor cells treated with the hMSCs-conditioned mediumthat penetrated the membrane were significantly reduced ascompared to controls (P< 0.01). The invasion inhibition ratesof A549 and Eca-109 cells were 32% and 34%, respectively(Table 1).

Molecular changes in the tumor cells induced by thehMSCs-conditioned medium

PCNA, Bcl-2 family members (Bcl-2, Bcl-xL, and Bax) andMMP-2 are, respectively, associated with the proliferation, apoptosis,and invasion of tumor cells. In order to find out the molecularmechanism underlying the inhibitory effect of hMSCs on tumorcells in vitro, we investigated the expressions of these proteinsin tumor cells by Western blotting. The results showed thattreatment of A549 and Eca-109 cells with hMSCs-conditionedmedium resulted in the significant downregulation ofexpression levels of PCNA, Bcl-2, Bcl-xL, and MMP-2 ascompared to controls (Fig. 4A,B). In addition, Cyclin D1,

TABLE 1. Suppressive effect of hMSCs on invasion of A549 and Eca-109

cells

Groupa

Invasion

Cell numberb Inhibition ratec (%) P-value

A549 con 91.66� 6.51A549 treated 62.67� 9.07 32 P< 0.01Eca-109 con 71.33� 8.02Eca-109 treated 47.34� 7.09 34 P< 0.01

aA549 or Eca-109 con: control groups untreated with hMSCs-conditionedmedium; A549 orEca-109 treated: the groups treated with hMSCs-conditioned medium.bThe number of A549 and Eca-109 cells that penetrated the polycarbonate membrane(means� SD, n¼ 4).cInhibition rate¼ (number of counted cells in the control group� number of counted cells inthe group treated with hMSCs-conditioned medium)/number of counted cells in the controlgroup� 100%.

Fig. 4. Molecular changes in tumor cells induced by the hMSCs-conditionedmedium in vitro. A: The expressions of PCNA, Cyclin D1, Cyclin E,pRb, phospho-pRb, P27, Bcl-2, Bcl-xL, Bax, MMP-2 in tumor cells were detected byWestern blotting. After treated with hMSCs-conditionedmedium(Eca-109RorA549RandhMSCsR) for24 h,Eca-109orA549cellswere lysedand25mgofproteinsweresubjectedtoSDS–PAGEanalysis,and then transferred to nitrocellulosemembranes and incubatedwith primary antibodies against PCNA,CyclinD1, Cyclin E, pRb, phospho-pRb,P27,Bcl-2,Bcl-xL,Bax,MMP-2,andb-actin.Eca-109R(orA549R)andhMSCs-onbehalfofthecontroluntreatedwithhMSCs-conditionedmedium.B:Bargraphrepresentsof thebanddensityvalueof theseproteinsexpressions inEca-109andA549cells treatedwithhMSCs-conditionedmediumrelative to the controls. C: Analysis of Cyclin E–CDK2 complexes in tumor cells following the treatment with hMSCs-conditioned medium:cell extracts prepared from Eca-109 or A549 cells were immunoprecipitated (IP) with anti-CDk2 antibody. The precipitated complexes wereexamined by immunoblotting (IB) usingCyclin E andCDK2antibodies.D:The relative valueof banddensity ofCyclin E–CDK2complexes level intumor cells. The results were the means (WSD) of three independent experiments (MP<0.01, MMP<0.001).

JOURNAL OF CELLULAR PHYSIOLOGY

1864 L I E T A L .

Page 6: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

Cyclin E, pRb, phospho-pRb, P27 (a CDK inhibitor), and CyclinE–CDK2 are the important cell-cycle regulatory proteinsinvolved in G1 progression and G1/S transition of cell cycle(Weinberg, 1995; Yamanouchi et al., 2001). To investigate howhMSCs can inhibit cell-cycle progression (Fig. 1B), we detectedthe expressions of Cyclin D1, Cyclin E, pRb, phospho-pRb, andP27 by Western Blotting and the formation of Cyclin E–CDK2complexes by immunoprecipitation. The results showedthe presence of hMSCs-conditioned medium resulted inthe obvious downregulation of Cyclin E expression, thedecrease of pRb phosphorylation (Fig. 4A,B) and the inhibitionof Cyclin E–CDK2 formation (Fig. 4C,D) as compared tocontrols.

hMSCs favor tumor formation and growth in vivo

To investigate the effect of hMSCs on the growth of Eca-109cells and A549 cells in vivo, we assessed the kinetic growth oftumor cells in BALB/c nudemice. After 10 days, 90% and 80% ofthe mice (n¼ 10) co-injected subcutaneously with hMSCsand respective two tumor cells had developed visible tumors,whereas only 70% and 60% of the mice (n¼ 10) injectedsubcutaneously with A549 cells or Eca-109 cells alone hadformed tumors (Fig. 5A). The mean volume of tumors of themice co-injected with hMSCs and tumor cells was dramaticallybigger than that of control groups (P< 0.05, Fig. 5B,C).

Tumor vessel formation increases at the presence ofhMSCs

In order to explore the reason of hMSCs favoring tumorgrowth in vivo, we observed the formation of tumor vessels.First of all, there was much richer vessel distribution found onthe surface of tumor mass of the mice co-injected with hMSCsand tumor cells (Fig. 6A) than that of controls injected withtumor cells alone (Fig. 6B). In addition, MVDwas determined bycounting the number of the microvessels per high-power field(hpf) in the section with an antibody reactive to CD34(Fig. 6C,D). Compared with the control group injected withA549 or Eca-109 cells alone, MVD value in the tumors of themice co-injected with tumor cells and hMSCs increasedobviously (P< 0.01, Fig. 6E). This result suggests that hMSCsseem to promote tumor vessel formation to favor tumorgrowth in the tumor microenvironment.

Discussion

Solid tumors are composed of tumor cells and supportive non-tumor components known as tumor stroma, which can bedivided into four main elements, including extracellular matrix(ECM), cells of the immune system, tumor vasculature, andfibroblastic stromal cells (Hall et al., 2007). In mouse models aswell as in humans, there is evidence that tumor stromal cellscould be derived from BM-derived progenitor cells believedto originate from hematopoietic stem cells as well as non-hematopoietic stem cells, such as mesenchymal stem cells(MSCs), which can be mobilized into the circulation, migratetowards tumors, incorporate into the tumormicroenvironment, and contribute to the growth of varioustumors (De et al., 2005; Tondreau et al., 2005; Roorda et al.,2009). In fact, it has already been widely demonstrated thathMSCs favor tumorigenesis and tumor growth in vivo by severalmeans, including differentiating into tumor stromal fibroblasts(Studeny et al., 2004; Spaeth et al., 2009), promoting tumorvessel formation (Galmiche et al., 1993; Al-Khaldi et al., 2003;Hung et al., 2005; Zhu et al., 2006; Liu et al., 2007; Sun et al.,2008), constructing cancer stem cell niches and inducingimmunosuppressive effect (Djouad et al., 2003;Matsunaga et al.,2003; Ramasamy et al., 2007). However, there are also the

Fig. 5. In vivo, hMSCs favor tumor formation and growth. 1T 106

hMSCs and 5T106 Eca-109 or A549 cells (alone or as a mix) wereinjected subcutaneously into BALB/c nude mice and tumordevelopment was monitored over time. A: Tumor incidence in thegroups of mice co-injected with tumor cells and hMSCs was higherthan controls injected with tumor cells alone. B,C: The mean tumorvolume of mice that developed tumor was calculated since 15th dayafter implantation. Tumor volumeU 1/2T lengthTwidth2. Ascompared to control groups, tumor volume in the groups of miceco-injected was much bigger obviously (MP<0.05).

JOURNAL OF CELLULAR PHYSIOLOGY

A D U A L R O L E O F h M S C s O N T U M O R C E L L G R O W T H 1865

Page 7: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

evidences showing an opposite result. For example, Khakooet al. (2006) described that hMSCs inhibited tumor growth inKaposi’s sarcoma models. Qiao et al. (2008) found that hMSCscould inhibit tumorigenesis in a hepatoma model.

Our findings showed that hMSCs inhibited the proliferationand invasion of tumor cells A549 and Eca-109, arrested tumorcells in the G1 phase of the cell cycle and contributed to theapoptosis of tumor cells in vitro by using the co-culture system

Fig. 6. In vivo, tumor vessel formation is strengthened at the presence of hMSCs. On the 30th day after implantation, themice were killed, andtumormasseswereremovedandfixedin10%neutralbufferedformalin solution forhistologicpreparations.Thefiguresweretherepresentativeofthemiceco-injectedwithhMSCsandtumorcells(A)orinjectedwithtumorcellsalone(B)andtherepresentativeof immunohistochemicalstainingof CD34 (magnification, 200T) forMVD in tumors of themice co-injectedwith hMSCs and tumor cells (C) or injectedwith tumor cells alone (D).Any yellow brown stained single endothelial cells or clusters of endothelial cells, with orwithout a lumen,were considered to be individual vessel.E:Themiceco-injectedwithtumorcellsandhMSCshaveamuchhigherMVDvalue(meansWSD)ascomparedtocontrols injectedwithtumorcellsalone (MP<0.01).

JOURNAL OF CELLULAR PHYSIOLOGY

1866 L I E T A L .

Page 8: Human mesenchymal stem cells play a dual role on tumor cell growth in vitro and in vivo

and the hMSCs-conditioned medium. The in vitro moleculardata showed that the decrease in the proliferation and invasionand the increase in the apoptosis of tumor cells were,respectively, associated with the downregulation of PCNA,MMP-2, Bcl-2, and Bcl-xL expressions. In addition, thedownregulation of Cyclin E expression, the decrease of pRbphosphorylation and the inhibition of Cyclin E–CDK2 activitywere the important molecular mechanisms underlying thehMSCs-mediated arrest of tumor cells in the G1 phase of cellcycle. It has been reported thatWnt signaling can regulate genesthat are involved in cell metabolism, proliferation, cell-cycleregulation, apoptosis and invasion including PCNA, Cyclin,MMP, and Bcl-2 (Baek et al., 2003; Neth et al., 2006). So, wethought that hMSCs-mediated tumor cell inhibition in vitro wasassociated with the Wnt signaling pathway. In vitro results,similar to ours, have recently been described by Qiao et al.(2008); they observed that the anti-proliferation effect of thehMSCs-conditioned medium containing the soluble factorssecreted by hMSCs on tumor cell was similar to the effectof direct contact of two kinds of cells. Tomchuck et al.(2008) showed that hMSCs secreted some cytokines andchemokines, such as CXCL10 interferon-gamma-inducibleprotein (IP10), interleukin (IL) 6, IL8, interferon (IFN) 1b, andtumor necrosis factor a (TNF-a), which possibly played animportant role in the inhibitory effect of hMSCs on tumor cellsin vitro.

However, in vivo, we observed that hMSCs favored tumorformation and growth in the animal models of lung cancer andesophageal cancer by the subcutaneous co-injection of tumorcells and hMSCs in BALB/c nude mice. According to theobservationof tumormass and the result ofmicrovessel density(MVD), we found that the promoting role of hMSCs on tumorgrowth was related with the more formation of tumor vessel inthe animal models with the co-injection of tumor cells andhMSCs. In fact, there have been evidences showing that hMSCscan promote tumor vessel formation to favor tumor growth bythe production of pro-angiogenic growth factors and thedifferentiation of endothelial-like cells in the tumormicroenvironment (Roorda et al., 2009). In addition, it has beenknown that hMSCs can differentiate into tumor stromalfibroblasts to favor tumor growth in the tumormicroenvironment (Roorda et al., 2009; Spaeth et al., 2009).

According to our studies, we come to a conclusion thathMSCs have dual characters: on the one hand, hMSCs have theability to inhibit the proliferation and invasion of tumor cells invitro; on the other hand, in vivo, hMSCs can favor tumorformation and growth in the tumor microenvironment.Therefore, we suggest that the exploitation of hMSCs in newtherapeutic strategies should be cautious under the malignantconditions.

Acknowledgments

We are indebted to Dr. Shanrun Liu (Department ofBiochemistry and Molecular Genetics University of Alabama atBirmingham, AL, USA) for critical reading of the manuscript.We also thank the members of Institute of Basic MedicalSciences of Qilu Hospital for their technical assistance. Thisproject was supported by the National Natural ScienceFoundation of China (no. 30571844 to Hui Tian) and a grantfrom Shandong Province Natural Science Foundation in China(no. 2009ZRB14005 to Hui Tian).

Literature Cited

Al-Khaldi A, Eliopoulos N, Martineau D, Lejeune L, Lachapelle K, Galipeau J. 2003. Postnatalbonemarrowstromal cells elicit a potent VEGFdependent neoangiogenic response in vivo.Gene Ther 10:621–629.

Baek SH, Kioussi C, Briata P, Wang D, Nguyen HD, Ohgi KA, Glass CK, Wynshaw-Boris A,RoseDW, RosenfeldMG. 2003. Regulated subset of G1 growth-control genes in responseto derepression by the Wnt pathway. Proc Natl Acad Sci USA 100:3245–3250.

De Palma PM, Venneri MA, Galli R, Sergi Sergi L, Politi LS, Sampaolesi M,Naldini L. 2005. Tie2identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vesselformation and a mesenchymal population of pericyte progenitors. Cancer Cell 8:211–226.

Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, Grisanti S, GianniAM. 2002. Human bone marrow stromal cells suppress T-lymphocyte proliferationinduced by cellular or nonspecific mitogenic stimuli. Blood 99:3838–3843.

Djouad F, Plence P, Bony C, Tropel P, Apparailly F, Sany J, Noel D, Jorgensen C. 2003.Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneicanimals. Blood 102:3837–3844.

GalmicheMC,Koteliansky VE, Briere J, Herve P, Charbord P. 1993. Stromal cells fromhumanlong-term marrow cultures are mesenchymal cells that differentiate following a vascularsmooth muscle differentiation pathway. Blood 82:66–76.

Hall B, Andreeff M, Marini F. 2007. The participation of mesenchymal stem cells in tumorstroma formation and their application as targeted-gene delivery vehicles. Handb ExpPharmacol 180:263–283.

Hung SC, Deng WP, Yang WK, Liu RS, Lee CC, Su TC, Lin RJ, Yang DM, Chang CW, ChenWH,Wei HJ, Gelovani JG. 2005. Mesenchymal stem cell targeting of microscopic tumorsand tumor stroma development monitored by noninvasive in vivo positron emissiontomography imaging. Clin Cancer Res 11:7749–7756.

Khakoo AY, Pati S, Anderson SA, ReidW, Elshal MF, Rovira II, Nguyen AT, Malide D, CombsCA,HallG,Zhang J, RaffeldM,RogersTB, Stetler-StevensonW, Frank JA, ReitzM, Finkel T.2006. Human mesenchymal stem cells exert potent antitumorigenic effects in a model ofKaposi’s sarcoma. J Exp Med 203:1235–1247.

Kumar S, Lavin MF. 1996. The ICE family of cysteine proteases as effectors of cell death. CellDeath Differ 3:255–267.

Le BK, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, Ringden O. 2004.Treatment of severe acute graft-versus-host disease with third party haploidenticalmesenchymal stem cells. Lancet 363:1439–1441.

Liu JW,Dunoyer-Geindre S, Serre-Beinier V,Mai G, Lambert JF, Fish RJ, PernodG, Buehler L,Bounameaux H, Kruithof EK. 2007. Characterization of endothelial-like cells derived fromhuman mesenchymal stem cells. J Thromb Haemost 5:826–834.

Mathiasen AB, Haack-Sørensen M, Kastrup J. 2009. Mesenchymal stromal cells forcardiovascular repair: Current status and future challenges. Future Cardiol 5:605–617.

Matsunaga T, Takemoto N, Sato T, Takimoto R, Tanaka I, Fujimi A, Akiyama T, Kuroda H,Kawano Y, Kobune M, Kato J, Hirayama Y, Sakamaki S, Kohda K, Miyake K, Niitsu Y. 2003.Interaction between leukemic-cell VLA-4 and stromal fibronectin is a decisive factor forminimal residual disease of acute myelogenous leukemia. Nat Med 9:1158–1165.

Neth P, Ciccarella M, Egea V, Hoelters J, JochumM, Ries C. 2006.Wnt signalling regulates theinvasion capacity of human mesenchymal stem cells. Stem Cells 24:1892–1903.

Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA,Simonetti DW, Craig S, Marshak DR. 1999. Multilineage potential of adult humanmesenchymal stem cells. Science 284:143–147.

Qiao L, Xu Z, Zhao T, Zhao Z, Shi M, Zhao RC, Ye L, Zhang X. 2008. Suppression oftumorigenesis by human mesenchymal stem cells in a hepatoma model. Cell Res 18:500–507.

RamasamyR, Lam EW-F, Soeiro I, Tisato V, BonnetD, Dazzi F. 2007.Mesenchymal stem cellsinhibit proliferation and apoptosis of tumor cells: Impact on in vivo tumor growth.Leukemia 21:304–310.

Roorda BD, ter Elst A, KampsWA, de Bont ES., 2009. Bonemarrow-derived cells and tumorgrowth: Contribution of bone marrow-derived cells to tumor micro-environments withspecial focus on mesenchymal stem cells. Crit Rev Oncol Hematol 69:187–198.

Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, Andreeff M, Marini F. 2009.Mesenchymal stem cell transition to tumor-associated fibroblasts contributes tofibrovascular network expansion and tumor progression. PLoS ONE 4:e4992.

StudenyM, Marini FC, Dembinski JL, Zompetta C, Cabreira-HansenM, Bekele BN, ChamplinRE, Andreeff M. 2004. Mesenchymal stem cells: Potential precursors for tumor stroma andtargeted-delivery vehicles for anticancer agents. J Natl Cancer Inst 96:1593–1603.

Sun T, Sun BC, Ni CS, ZhaoXL,WangXH,Qie S, ZhangDF, GuQ,Qi H, ZhaoN. 2008. Pilotstudy on the interaction between B16 melanoma cell-line and bone-marrow derivedmesenchymal stem cells. Cancer Lett 263:35–43.

Tomchuck SL, ZwezdarykKJ, Coffelt SB,WatermanRS, Danka ES, ScandurroAB. 2008. Toll-like receptors on human mesenchymal stem cells drive their migration andimmunomodulating responses. Stem Cells 26:99–107.

Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, Mortier C, Bron D,Lagneaux L. 2005. Mesenchymal stem cells derived fromCD133-positive cells in mobilizedperipheral blood and cord blood: Proliferation, Oct4 expression, and plasticity. StemCells23:1105–1112.

Undale AH, Westendorf JJ, Yaszemski MJ, Khosla S. 2009. Mesenchymal stem cells for bonerepair and metabolic bone diseases. Mayo Clin Proc 84:893–902.

Weinberg RA. 1995. The retinoblastoma protein and cell cycle control. Cell 81:323–330.Weidner N, Semple JP, Welch WR, Folkman J. 1991. Tumor angiogenesis and metastasis—Correlation in invasive breast carcinoma. N Engl J Med 324:1–8.

Yamanouchi H, Furihata M, Fujita J, Murakami H, Yoshinouchi T, Takahara J, Ohtsuki Y. 2001.Expression of cyclin E and cyclin D1 in non-small cell lung cancers. Lung Cancer 31:3–8.

Zhang JC, Ye NL, Huang H, He X, Tong WG, Ross J, Haug J, Johnson T, Feng JQ, Harris S,Wiedemann LM, Mishina Y, Li L. 2003. Identification of the haematopoietic stem cell nicheand control of the niche size. Nature 425:836–841.

Zhu W, XuW, Jiang R, Qian H, Chen M, Hu J, Cao W, Han C, Chen Y. 2006. Mesenchymalstem cells derived from bone marrow favor tumor cell growth in vivo. Exp Mol Pathol80:267–274.

JOURNAL OF CELLULAR PHYSIOLOGY

A D U A L R O L E O F h M S C s O N T U M O R C E L L G R O W T H 1867