Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically...

9
Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with sTRAIL gene expression driven by AFP promoter Cihui Yan a, b, c, d,1 , Ming Yang a, 1 , Zhenzhen Li a , Shuangjing Li a , Xiao Hu a , Dongmei Fan a , Yanjun Zhang a, ** , Jianxiang Wang a, ** , Dongsheng Xiong a, * a State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Number 188, Nanjing Road, Heping District, Tianjin, China b Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, China c National Clinical Research Center of Cancer, China d Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China article info Article history: Received 16 November 2013 Accepted 13 December 2013 Available online 6 January 2014 Keywords: Mesenchymal stem cell TRAIL Alpha-fetoprotein Targeted therapy Hepatocarcinoma abstract Mesenchymal stem cells (MSCs) are promising vehicles for delivering therapeutic agents in tumor therapy. Human umbilical cord-derived mesenchymal stem cells (HUMSCs) resemble bone marrow- derived MSCs with respect to hepatic differentiation potential in injured livers in animals, while their hepatic differentiation under the hepatocarcinoma microenvironment is unclear. In this study, HUMSCs were isolated and transduced by lentiviral vectors coding the soluble human tumor necrosis factor- related apoptosis-inducing ligand (sTRAIL) gene driven by alpha-fetoprotein (AFP) promoter to investi- gate the therapeutic effects of these HUMSC against orthotopically implanted hepatocarcinoma in mice. We showed that HUMSCs can be transduced by lentivirus efciently. HUMSCs developed cuboidal morphology, and expressed AFP and albumin in a two-step protocol. HUMSCs were capable of migrating to hepatocarcinoma in vitro as well as in vivo. In the orthotopical hepatocarcinoma microenvironment, the AFP promoter was activated during the early hepatic differentiation of HUMSCs. After intravenous injected, MSC.AFPILZ-sTRAIL expressed sTRAIL exclusively at the tumor site, and exhibited signicant antitumor activity. This effect was stronger when in combination with 5-FU. The treatment was tolerated well in mice. Collectively, our results provide a potential strategy for targeted tumor therapy relying on the use of the tumor tropism and specic differentiation of HUMSCs as vehicles. Ó 2013 Elsevier Ltd. All rights reserved. 1. Introduction Mesenchymal stem cells (MSCs), rst described by Friedenstein et al. [1], are characterized as adherence to plastic, expression of specic surface antigens CD105, CD90 and CD73 but lack of CD34 and CD45, and multi-differentiation potential into osteoblasts, ad- ipocytes and chondroblasts [2]. MSCs hold a great promise as ve- hicles for targeted delivery and local production of agents in tumor therapy because they can be easily isolated and expanded to a large number of cells required for clinical use, have tumor tropism, and can be genetically engineered with viral vectors [3,4]. Human umbilical cord tissue represents a convenient, abundant and economic source of adult MSCs. Human umbilical cord-derived MSCs (HUMSCs) share characters of conventional bone marrow- derived MSCs, which provides an idea chose for intensive studies and application of MSCs. Gene therapy using therapeutic agents driven by the tumor- or tissue-specic promoter is a promising approach for the treatment of cancer. The alpha-fetoprotein (AFP) promoter, which is reac- tivated in hepatocellular carcinoma, has been widely applied to regulate the cytotoxic gene expression to kill the tumor cells selectively [5e7]. And the expression vectors and adenovirus were usually used as vehicles for this targeted therapy. While some problems must be concerned, such as low infective efciency of targeted cells, non-specic infection to normal tissues and poten- tial immunogenicity resulted from viral proteins. Thus, potential therapeutic strategies are urgently needed to overcome these limitations. Accumulated evidences indicate that human bone marrow, umbilical blood and adipose tissue-derived MSCs are * Corresponding author. Tel./fax: þ86 02223909404. ** Corresponding authors. E-mail address: [email protected] (D. Xiong). 1 These authors contributed equally to this work. Contents lists available at ScienceDirect Biomaterials journal homepage: www.elsevier.com/locate/biomaterials 0142-9612/$ e see front matter Ó 2013 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.biomaterials.2013.12.037 Biomaterials 35 (2014) 3035e3043

Transcript of Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically...

Page 1: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

lable at ScienceDirect

Biomaterials 35 (2014) 3035e3043

Contents lists avai

Biomaterials

journal homepage: www.elsevier .com/locate/biomater ia ls

Suppression of orthotopically implanted hepatocarcinoma in mice byumbilical cord-derived mesenchymal stem cells with sTRAIL geneexpression driven by AFP promoter

Cihui Yan a,b,c,d,1, Ming Yang a,1, Zhenzhen Li a, Shuangjing Li a, Xiao Hu a, Dongmei Fan a,Yanjun Zhang a,**, Jianxiang Wang a,**, Dongsheng Xiong a,*

a State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases,Chinese Academy of Medical Sciences & Peking Union Medical College, Number 188, Nanjing Road, Heping District, Tianjin, ChinabDepartment of Immunology, Tianjin Medical University Cancer Institute and Hospital, ChinacNational Clinical Research Center of Cancer, ChinadKey Laboratory of Cancer Immunology and Biotherapy, Tianjin, China

a r t i c l e i n f o

Article history:Received 16 November 2013Accepted 13 December 2013Available online 6 January 2014

Keywords:Mesenchymal stem cellTRAILAlpha-fetoproteinTargeted therapyHepatocarcinoma

* Corresponding author. Tel./fax: þ86 02223909404** Corresponding authors.

E-mail address: [email protected] (D. Xiong).1 These authors contributed equally to this work.

0142-9612/$ e see front matter � 2013 Elsevier Ltd.http://dx.doi.org/10.1016/j.biomaterials.2013.12.037

a b s t r a c t

Mesenchymal stem cells (MSCs) are promising vehicles for delivering therapeutic agents in tumortherapy. Human umbilical cord-derived mesenchymal stem cells (HUMSCs) resemble bone marrow-derived MSCs with respect to hepatic differentiation potential in injured livers in animals, while theirhepatic differentiation under the hepatocarcinoma microenvironment is unclear. In this study, HUMSCswere isolated and transduced by lentiviral vectors coding the soluble human tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) gene driven by alpha-fetoprotein (AFP) promoter to investi-gate the therapeutic effects of these HUMSC against orthotopically implanted hepatocarcinoma in mice.We showed that HUMSCs can be transduced by lentivirus efficiently. HUMSCs developed cuboidalmorphology, and expressed AFP and albumin in a two-step protocol. HUMSCs were capable of migratingto hepatocarcinoma in vitro as well as in vivo. In the orthotopical hepatocarcinoma microenvironment,the AFP promoter was activated during the early hepatic differentiation of HUMSCs. After intravenousinjected, MSC.AFPILZ-sTRAIL expressed sTRAIL exclusively at the tumor site, and exhibited significantantitumor activity. This effect was stronger when in combination with 5-FU. The treatment was toleratedwell in mice. Collectively, our results provide a potential strategy for targeted tumor therapy relying onthe use of the tumor tropism and specific differentiation of HUMSCs as vehicles.

� 2013 Elsevier Ltd. All rights reserved.

1. Introduction

Mesenchymal stem cells (MSCs), first described by Friedensteinet al. [1], are characterized as adherence to plastic, expression ofspecific surface antigens CD105, CD90 and CD73 but lack of CD34and CD45, and multi-differentiation potential into osteoblasts, ad-ipocytes and chondroblasts [2]. MSCs hold a great promise as ve-hicles for targeted delivery and local production of agents in tumortherapy because they can be easily isolated and expanded to a largenumber of cells required for clinical use, have tumor tropism, andcan be genetically engineered with viral vectors [3,4]. Humanumbilical cord tissue represents a convenient, abundant and

.

All rights reserved.

economic source of adult MSCs. Human umbilical cord-derivedMSCs (HUMSCs) share characters of conventional bone marrow-derived MSCs, which provides an idea chose for intensive studiesand application of MSCs.

Gene therapy using therapeutic agents driven by the tumor- ortissue-specific promoter is a promising approach for the treatmentof cancer. The alpha-fetoprotein (AFP) promoter, which is reac-tivated in hepatocellular carcinoma, has been widely applied toregulate the cytotoxic gene expression to kill the tumor cellsselectively [5e7]. And the expression vectors and adenovirus wereusually used as vehicles for this targeted therapy. While someproblems must be concerned, such as low infective efficiency oftargeted cells, non-specific infection to normal tissues and poten-tial immunogenicity resulted from viral proteins. Thus, potentialtherapeutic strategies are urgently needed to overcome theselimitations. Accumulated evidences indicate that human bonemarrow, umbilical blood and adipose tissue-derived MSCs are

Page 2: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

C. Yan et al. / Biomaterials 35 (2014) 3035e30433036

capable of differentiating into hepatocytes in vitro as well as in vivo,expressing the specific markers for hepatocytes such as AFP, albu-min (ALB), CK18 and CK19, and exhibiting hepatic function such asuptake of low density lipoprotein, synthesis of glycogen and theactivity of liver drug enzyme cytochrome P450 [8e11]. During thehepatic differentiation of MSCs, AFP expression increases at theearly stage and disappears at the later stage. As a result, the AFPpromoter, which is reactivated specifically at the early hepaticdifferentiation of MSCs, could be used to selectively control theexpression of a therapeutic agent in MSC-base gene therapyalthough there is no report so far.

TNF related apoptosis-inducing ligand (TRAIL), a member oftumor necrosis factor (TNF) superfamily, is a promising candidatefor cancer therapy as it induces apoptosis in a wide variety of hu-man cancer cell lines, while largely sparing normal cells [12,13]. Asa type 2 transmembrane protein, TRAIL can be cleaved by specificproteases and the extracellular region forms a soluble molecule.Soluble TRAIL (sTRAIL) (amino acids 114e281) forms a homotrimerwhich is a pivotal structure for receptor recognition and apoptoticfunction [12,14]. The isoleucine zipper (ILZ), a derivation of leucinezipper, is known to be a strong trimerization domain and requiredfor the apoptotic TRAIL protein secreted into the culture superna-tant [14]. Nevertheless, recombinant human sTRAIL displays a shothalf-life (30e60min) in vivo, which would limit the use of sTRAIL inclinical.

In the present study, we designed a specifically targeted thera-peutic system, in which HUMSCs were engineered to specificallysecret cytotoxic ILZ-sTRAIL protein which was regulated by AFPpromoter, and investigated its antitumor effect in combinationwith5-fluorouracil (5-FU) on the orthotopically implanted hep-atocarcinoma in mice.

2. Materials and methods

2.1. HUMSCs preparation and cell culture

HUMSCs were isolated from the gelatinous Wharton’s jelly (WJ) of the humanumbilical cord bymethods previously described [15]. HUMSCswere subcultured at adensity of 4000 cells/cm2 in DF-12 medium (Invitrogen, USA) supplemented with2 mmol/L L-glutamine and 10% FSC (Gibco, USA). Passages 3e5 were used for thefollowing experiments. The human hepatocellular carcinoma cell line HepG2, breastcancer cell line MCF-7, mouse embryonic fibroblast cell line 3T3 (Institute of He-matology & Blood Diseases Hospital Chinese Academy of Medical Sciences & PekingUnion Medical College, PUMC, Tianjin, China), and human embryonic kidney cell-derived 293 T cell line (kindly provided by Professor Cheng Tao, PUMC) were main-tained in DMEM (Invitrogen, USA) supplemented with 2 mmol/L L-glutamine,100 units/mL penicillin (Hyclone, USA), 100 ug/mL streptomycin (Hyclone, USA) and10% FCS. Cellswere incubated at 37 �C in a humidified atmosphere containing 5% CO2.

2.2. Luciferase assay for the specificity of AFP promoter

The sequence of alpha-fetoprotein (AFP) specific promoter was amplified frompUp plasmid (Cyagen Bioscience, USA) and inserted into pGL3 basic vector (Kpn Iand Nhe I) to construct pGL3 AFP Firefly luciferase reporter vector (pGL3 AFP vector).HepG2, MCF-7 and 3T3 cells incubated in the six-well culture plates were trans-fectedwith Firefly luciferase-expressing vectors (pGL3 AFP, pGL3 basic, pGL3 controland pGL3 CMV) respectively, and Renilla luciferase-expressing vector (pRL-TK) wasco-transfected into these three cell lines as an internal control. These cells werecultured for additional 2 days. Lysates were prepared and analyzed using the dualluciferase reporter assay system (Promega, USA). The relative luciferase activity wascalculated as the radio of Firefly luciferase activity to Renilla luciferase activity. Eachdata point was averaged from two replicates of three separate experiments.

2.3. Hepatogenic differentiation in vitro

Hepatogenic differentiationwas conducted in two-steps as described previously[8]. The hepatogenesis was assessed by detecting AFP and ALB expression, thespecific maker genes for liver cells, by RT-quantitative PCR and Western blot.

2.4. Transient transfection of 293 T cells

293 T cells were transfected with pLentiR.ILZ-sTRAIL, or pLentiR.CopGFP (con-trol) using lipofectamin�2000 (Invitrogen, USA). After 48 h of transfection, cellsupernatant was collected by centrifugation at 500 �g for 10 min at 4 �C to clear293 T cells and applied in vitro studies.

2.5. Production of lentiviral vectors

The lentiviral particles produced by 293 T cells were conducted according to theSystem Biosciences (SBI, Canada) protocol. The lentiviral-containing supernatantwas collected at 48 h post-transfection and spun at 500 �g for 5 min, filteredthrough a 0.44 mm pore size filter (Millipore, USA) and used to transduce HUMSCsimmediately or stored at �80 �C.

2.6. Transduction of MSCs

HUMSCs were plated at a density of 2 � 105 per well in T-25 cm plastic cultureflasks and incubated overnight at 37 �C. On the next day, mediumwas removed and3 ml of appropriate fresh medium containing lentiviral supernatants at MOI 8 and8mg/ml of polybrene (Sigma, USA)was added. Themediumwas removedafter 8hand10% FCS DF-12 medium was added. HUMSCs were incubated for the indicated timeandDsRed fluorescencewas observed underfluorescencemicroscope (Nikon, Japan).

2.7. In vitro migration study

The migratory ability of HUMSCs was determined using Transwell plates in vitroas described previously [3]. The number of cells that had migrated to the lower sideof the filter was counted under a light microscope with five high-power fields(�400). Experiments were done in triplicate.

2.8. In vivo migration of HUMSCs

The human hepatocarcinoma HepG2 cell line was used to build orthotopic livertumor model in Balb/c athymic nude mice as described previously [16]. All animals(male, age 6 weeks, Peking Union Medical College, PUMC, China) received humancare during the study and had free access to water and laboratory chow. Animalstudies were approved by the IACUC of the Institute of Hematology & Hospital ofBlood Diseases, PUMC. When the orthotopic tumor model has been successfullydeveloped after 7 days of orthotopic implantation, 5�105MSC.CMVLu cells were i.v.injected into mice for migration detection. Bioluminescence imaging (BLI) wasperformed using IVIS-Xenogen 100 system (Caliper Lifesciences, USA) at the indi-cated time. In brief, mice were anesthetized by intraperitoneal administration of100 ml of 20mg/ml pentobarbital sodium. Eachmouse received 15mg/mL D-luciferin(Promega, USA) at a dosage of 150 mg/kg, i.p. 10 min prior to imaging. All imagesrepresent a 5 min exposure time.

2.9. In vivo differentiation of HUMSCs

Firstly, MSC.AFPLu cells were orthotopically injected into the tumor-burned liverfor differentiation detection. And BLI was detected as described above. Then,MSC.AFPILZ-sTRAIL cells, in which ILZ-sTRAIL was fused with CopGFP at the N-ter-minus, were i.v. injected into mice. After 1-week of injection, mice were sacrificedand the livers were removed for the specific expression of the targeted gene byfluorescence microscope and Western blot.

2.10. Treatment of orthotopic hepatic carcinoma model

After 7 days of orthotopic tumor inoculation, mice were randomized into eightgroups (5 mice for each group) as following: (1) PBS control; (2) 5-FU; (3) HUMSCs;(4) HUMSCs and 5-FU; (5) MSC.AFPCopGFP; (6) MSC.AFPCopGFP and 5-FU; (7)MSC.AFPILZ-sTRAIL; (8) MSC.AFPILZ-sTRAIL and 5-FU. The responsive HUMSCsengineered or notwere i.v. injected at a dose of 5�105 cells in eachmouse. 5-FUwasi.p. injected at a dosage of 10 mg/kg for successive 5 days from the next day ofHUMSCs injection. At day 60 after treatment started, mice were sacrificed. The tu-mor was dissected from each mouse, measured and weighed. The volume isexpressed in mm3 using the formula: V ¼ 0.5 a � b2 where a and b are the long andshort diameters of the tumor, respectively. The serum level of liver enzymes alanineaminotransferase (ALT) and aspartate transaminase (AST) was assessed by spec-trophotometer (Nan Jing JianCheng Bioengineering Institute, China).

2.11. Statistical analysis

Data are represented as mean� SD. Differences between groups were examinedfor significant differences by ANOVA LSD or Dunnett post hoc procedure. Values ofP < 0.05 were considered to be statistically significant and that of P < 0.01 wereconsidered to be highly statistically significant.

3. Results

3.1. Hepatic differentiation of HUMSCs in vitro

In the absence of serum, cell proliferation arrested. In thepresence of HGF and bFGF, the fibroblastic morphology of HUMSCswas lost and cells developed a broadenedmorphology by the end ofthe induction step. In the presence of oncostatinM, dexamethasoneand ITSþ, elongated ends disappeared and a cuboidal morphology

Page 3: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

C. Yan et al. / Biomaterials 35 (2014) 3035e3043 3037

of hepatocytes developed with increasing time of differentiation.After prolonged culture, abundant granules appeared in the cyto-plasma of differentiated cells (Fig. 1A).

The mRNA expression of AFP, an early maker gene of hepato-cytes, increased and rank the top at day 12, then decreased sharply.The mRNA expression of ALB increased along the time of differ-entiation (Fig. 1B). The protein expression of AFP rose dramaticallyat day 6 and decreased afterward. The protein expression of ALBincreased andwas detected at all time points post-induction. Whileundifferentiated cells did not express AFP or ALB (Fig. 1C).

3.2. Construction of lentiviral expression vectors

We successfully cloned AFP specific promoter (2810 bp) frompUp plasmid. The specific transcriptional activity of AFP promoter,which was defined as the radio of Firefly luciferase (fLuc) activity toRenilla luciferase activity, was tested firstly before vector con-struction followed. The mean pGL3 AFP ratios were 49.05 � 5.47,1.69 � 0.24 and 6.88 � 0. 61 in HepG2, MCF-7 and 3T3 cells,respectively (Fig. 2A), indicating the specific transcriptional activity

Fig. 1. Hepatic differentiation of HUMSCs in vitro. (A) Morphological changes at the indicatemarker genes by quantitative real-time PCR. (C) Differentiated HUMSCs expressed hepatoc

of AFP promoter in AFP-positive cells. Then, lentiviral expressionvectors which included the targeted genes driven by CMV or AFPpromoter were successfully constructed (Fig. 2B). HUMSCs could betransduced efficiently by lentivirus containing the ILZ-sTRAIL genecontrolled by AFP promoter without affecting their growth. TheDsRed fluorescence could be observed even after 26 days oftransduction (Fig. 2C) without the CopGFP fluorescence (data notshown).

3.3. Inhibitory effect of ILZ-sTRAIL on the growth of human HepG2cells

To obtain ILZ-sTRAIL, 293 T cells were transfected with plasmidpLentiR.ILZ-sTRAIL, in which ILZ-sTRAIL expression was controlledby CMV promoter, and the supernatant containing ILZ-sTRAIL(9.475 � 0.786 ng/ml) was collected from 293 T cell culture(Fig. 3A) and used in the following studies in vitro. ILZ-sTRAILinhibited the growth of HepG2 cells in a concentration-dependent manner. And this effect was significantly enhancedwhen combined with 5-FU (Fig. 3B). The therapeutic index (CI) of

d time during differentiation. (B) Differentiated HUMSCs expressed hepatocyte-specificyte-specific proteins by Western blot.

Page 4: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

Fig. 2. Plasmid construction. (A) The specific activity of AFP promoter in different cell lines. Cells were co-transfected with firefly luciferase reporter plasmid (containing differentpromoters needed tested) and Renilla luciferase reporter plasmid (used as an internal control) at a ratio of 50:1. The activity of luciferase was quantified 48 h later. (B) Schematicrepresentation of lentiviral expression vectors constructed. , CMV promoter; , AFP promoter; , EF1 promoter; , luciferase; , signal peptide; , CopGFP; , ILZ-sTRAIL; , DsRed. (C) High transduction efficiency in HUMSCs by lentivirus.

C. Yan et al. / Biomaterials 35 (2014) 3035e30433038

combination treatment showed that ILZ-sTRAIL plus 5-FU exhibitedan addictive inhibitory effect on the proliferation of HepG2 cells(Table 1). The level of Bcl-2 protein expression decreased, and Baxexpression increased, caspases involved in apoptosis such ascaspase-8, 9, 3 and PARP were cleaved and activated. And all thesechanges were much more significant when ILZ-sTRAIL and 5-FUwere combined in the treatment (Fig. 3C).

Fig. 3. Inhibitory effect of ILZ-sTRAIL on the growth of hepatocarcinoma HepG2 cells. (A) ELIStransient transfection. (B) Cells were cultured (1 � 104 cells/well) overnight and exposed totested by CCK8 assay. **P < 0.01 compared with corresponding concentration of ILZ-sTRAI

3.4. Migration capacity of HUMSCs to hepatocarcinoma in vitro andvivo

The migration potential in HUMSCs was tested firstly usingTranswell plates in vitro. Only a few cells migrated toward serum-free medium, while the migration of HUMSCs was stimulatedsignificantly by the conditionedmedium fromHepG2 cells (Fig. 4A).

A detection measuring ILZ-sTRAIL released in the supernatant of 293 T cell culture afterdifferent concentration of ILZ-sTRAIL in the presence or absence of 5-FU for 72 h and

L treatment alone. (C) Western blot showed apoptosis induced by 4 ng/ml ILZ-sTRAIL.

Page 5: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

Table 1The therapeutic index (CI) of ILZ-sTRAIL and 5-FU combination treatment. Cells werecultured (1�104 cells/well) overnight and exposed to different concentration of ILZ-sTRAIL in the presence or absence of 5-FU for 72 h and tested by CCK8 assay. CI < 1,synergistic; CI ¼ 1, additive; CI > 1, antagonistic.

5-FU (mg/ml) ILZ-sTRAIL (ng/ml)

1 2 4

0.5 1.1 0.95 0.862 0.93 0.84 0.86

C. Yan et al. / Biomaterials 35 (2014) 3035e3043 3039

Moreover, the migratory activity of HUMSCs appeared in aconcentration-dependent manner (Fig. 4B). The migration ofmodified HUMSCs was in a similar pattern as that of unmodifiedHUMSCs. Next, to monitor tumor tropism of HUMSCs in vivo,orthotopical hepatic carcinoma model was successfully built inBalb/c mice (Fig. 4C), and HUMSCs constitutively expressing fLucreporter gene (MSC.CMVLu) were injected intravenously. Repre-sentative BLI analysis in live animals revealed that after 1 day ofinjection, intensive fLuc imaging signals were only detected in lung,suggesting that a mount of HUMSCs were entrapped by capillariesin lung. After 2 days of injection, the signals in lung decreasedsubstantially; in contrast, the signal intensity in the live slightlyincreased and strengthened on day 5 (Fig. 4D). No detectable sig-nals were observed in the other organs.

3.5. Hepatic differentiation of HUMSCs in vivo

To verify hepatic differentiation under the microenvironment oforthotopically implanted hepatocarcinoma, we inoculated HUMSCslabeled with fLuc driven specifically by AFP promoter (MSC.AFPLuc)in situ in tumor-bearing livers and found that the fLuc signal wasweak after 1 day of injection, increased and reached the peak onday 7 after injection. Then the activity decreased rapidly and wasnot detectable after 9 days of injection (Fig. 5A). We also used shamoperation as negative control and did not detect any fLuc signal in

Fig. 4. Tumor tropism of HUMSCs in vitro and vivo. (A) The migratory capacity of HUMSCsplates. SFM (serum-free medium) used as a negative control. (B) The number of cells migratpower fields (�400). **P < 0.01 compared with SFM. Experiments were done in triplicate. (Cremoved and examined histopathologically at day 7 after transplantation. (D) HUMSCs mexpressed firefly luciferase were i.v. injected into tumor-bearing mice and monitored by bi

all organs. Next, we injected MSC.AFPILZ-sTRAIL intravenously. 7days later, we found that HUMSCs migrated to the tumor site andproduced the targeted gene identified both by CopGFP fluorescence(Fig. 5B) and by ILZ-sTRAIL protein expression (Fig. 5C) owing to theactivation of AFP promoter.

3.6. Antitumor potential of MSC.AFPIZL-sTRAIL againstorthotopically implanted hepatic carcinoma

In the in vivo study, mice were sacrificed after 60 days of thestart treatment, and the tumors in livers were dissected andweighed. As shown in Fig. 6A, obvious tumor regression wasobserved in the MSC.AFPILZ-sTRAIL group (p < 0.05) and theMSC.AFPILZ-sTRAIL plus 5-FU group (p < 0.01). And the combina-tion treatment exhibited stronger antitumor effect when comparedwith MSC.AFPILZ-sTRAIL treatment alone. The pathological resultshowed that tumor cells in control groups grew vigorously and hadbigger trachychromatic nucleuses. However, tumor cells in thegroups treated with MSC.AFPILZ-sTRAIL, either with 5-FU or not,appeared pyknotic and necrotic, and had extensive lymphocyticinfiltration. This phenomenonwas even obvious in the combinationtreatment group (Fig. 6B). The serum level of ALTand AST decreasedsignificantly in these two groups (ALT, P < 0.01; AST, P < 0.05)(Fig. 6C). During the whole treatment period, there were no sig-nificant differences in the body weight in all groups (P > 0.05)(Fig. 6D).

4. Discussion

In this study, we efficiently engineered HUMSCs to specificallysecret ILZ-sTRAIL driven by AFP promoter via lentiviral trans-duction. Our results showed that these cells can migrate towardhepatocarcinoma and undergo hepatic differentiation in vitro aswell as in vivo. MSC.AFPILZ-sTRAIL exhibited significant antitumoreffect on the orthotopically implanted hepatocarcinoma, whichwas mediated by ILZ-sTRAIL induced apoptosis and strengthened

in response to conditioned medium of HepG2 cells was determined using Transwelled to the lower side of the filter was counted under a light microscope with five high-) Orthotopically implanted hepatic carcinoma model was well established. Livers wereigrated to orthotopical hepatocarcinoma in vivo. HUMSCs labeled with constitutivelyoluminescence imaging using Xenogen imaging system at the indicated time.

Page 6: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

Fig. 5. Hepatic differentiation of HUMSCs in vivo. HUMSCs expressing firefly luciferase (A) or ILZ-sTRAIL fused with CopGFP (B, C) controlled by AFP promoter were in situ implanted(A) or i.v. injected (B, C) into tumor-burned mice respectively. (A) The bioluminescence imaging for luciferase activity was detected by Xenogen imaging system at the indicatedtime. The targeted gene expression was detected by confocal microscopy (B) and Western blot using anti-TRAIL or anti-CopGFP antibody (C) after 7 days of injection. M1, M2, M3(mice 1e3).

C. Yan et al. / Biomaterials 35 (2014) 3035e30433040

when combined with 5-FU. This is an original report that thehoming capacity and differentiation potential are combinedtogether in the strategy of human derived MSCs as vehicles deliv-ering targeted agents for tumor therapy.

The effect of MSCs on tumor growth is always in debate, whichmight be due to the factors of the different source of MSCs, the ratioof each cell population performed in animal models, the location ofthe lesion, and alternative administration route et al. It is reportedthat hepatocellular carcinoma (HCC) derived MSCs promote HCCcell proliferation and invasion [17]. And tumor cells mixed withbone marrow-derived MSCs transplanted subcutaneously exhibi-ted elevated capability of proliferation, rich angiogenesis in tumortissues and highly metastatic ability [18,19]. However, HUMSCs,used in our study, did not promote the growth of orthotopicallyhepatic carcinoma. Several recent studies demonstrate thatHUMSCs express high level of pro-apoptotic and tumor suppressorgenes [20] having the properties of non-tumorigenicity [21] or anti-tumorigenicity [22], and unable transform into tumor-associatedfibroblasts (TAFs) [23]. As a result, HUMSCs represent an idealsource of MSCs in targeted therapy.

We used an orthotopical human hepatocarcinoma xenograftmodel to imitate the microenvironment of liver cancer in vivo andevaluate the migratory ability, differentiation potential and thera-peutic efficacy of HUMSCs. The tumor tropism of HUMSCs wasobserved obviously in our study. However, in healthy or shammice,HUMSCs did not home to liver and disappeared quickly in vivo (datanot shown). Tumors can be regarded as wounds that do not heal;the shared tropism of MSCs in site of an injury tissue and tumor isthought to result from the similarities of the microenvironment[24]. The exact process underlining the migration of MSCs to tumor

site is not clear. Two possible mechanisms have been proposed, oneis that the released chemokines/cytokines increases the migrationof MSCs [25]; another is that the interaction of cytokines or che-mokines with their corresponding receptors would induce themigration of MSCs towards tumor microenvironment [26]. Astumor-bearing mice had a tumor microenvironment resembling ofan unresolved wound, HUMSCs could migrate specifically andlocalize at the tumor site. Our result showed that the accumulationof HUMSCs in liver presented in delayed phase. We presume thatalthough some amount of HUMSCs were trapped by pulmonarycapillaries and a part of HUMSCs that had arrived in liver died fromthe unsuitable environment, the HUMSCs remaining in liver couldsurvive and grow after a short adaptive phase.

It has been reported that in the hepatic injured and cirrhotic rat,human MSCs from bone marrow as well as umbilical blood arecapable of differentiating into hepatocytes and improve hepaticfunction [9,27]. On the contrary, the differentiation of bonemarrowcells into mature hepatocytes is much low efficiency under physi-ologic conditions as a selection strategy is required for the differ-entiation [28]. However, the hepatic differentiation of HUMSCs islittle reported. In our experiment, hepatic differentiation ofHUMSCswas detected in the livers bearing orthotopically implanted hep-atocarcinoma, but not in the site of orthotopically implanted breastcancer (data not shown), indicating that the specific microenvi-ronment of hepatocarcinoma would act as a selective pressure tostimulate hepatic differentiation of HUMSCs. This result is alsoconfirmed by a recent study which demonstrates that HUMSCscould be a promising stem cell source to generate hepatocyte-likecells as transcription factors involved in liver development andliver progenitor markers are highly expressed in HUMSCs [29]. The

Page 7: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

Fig. 6. Antitumor potential of MSC.AFPILZ-sTRAIL against orthotopically implanted hepatic carcinoma. (A) The size of tumor removed from livers after 60 days of the starttreatment. (B) The histopathologic analysis of tumors. (C) Serum alanine transarninase (ALT) and glutamic-oxaloacetic transaminase (AST) levels in mice. (D) No difference in theweights of mice during treatment period. *P < 0.05, **P < 0.01 compared with the model group.

C. Yan et al. / Biomaterials 35 (2014) 3035e3043 3041

relative shorter expression phase of AFP here could result from thedifferent sources of MSCs, test methods and animal models.

As we found that AFP gene expressed specifically at the earlystage of hepatic differentiation of HUMSCs under the specific envi-ronmentof hepatocarcinoma,wegeneticallyengineeredHUMSCs toexpress ILZ-sTRAIL controlled byAFPpromoter to inhibit the growthof orthotopical hepatocarcinoma. This therapeutic strategy takesadvantage of the tumor tropism and hepatic differentiation ofHUMSCs together. Firstly, HUMSCs migrated to tumor site whichprovided the prerequisite of hepatic differentiation. Then, hepaticdifferentiated HUMSCs supported re-activation of AFP promoter.Consequently, ILZ-sTRAIL was expressed, secreted, localized andconcentrated in the tumor tissue exclusively but not in the peritu-moral tissue or other organs. As a result, significant antitumor effectwas achieved and no obvious side effects were detected. Theimprovement of liver function may be associated with the reducedtumor burned and repair and generationbyMSC.AFPILZ-sTRAIL. Thetumor tropism of HUMSCs both decreases the required quantity ofHUMSCs injected and escapes the probability of unrelated action

resulting from the cytotoxic agents secreted. Even though a numberof HUMSCs migrated to other organs and tissues, for example, localinflammation tissues in non-tumor-burned organs or lung capil-laries inwhichHUMSCswere trapped, the cytotoxic agent cannot beexpressed as the specific microenvironment devoting to hepaticdifferentiationwasnot provided and theAFPpromoterwould not beactive. Furthermore, we found that hepatic differentiated HUMSCstended to localize in the area of adjacent tumor or even the inner ofsolid tumor, and the expression of ILZ-sTRAIL exclusively in tumortissue but not in the peritumoral liver tissue. It seems that cytokinesinvolved in migration and differentiation also distribute gradientlyin liver just as in the overall level. As a result, HUMSCs accumulatingin tumor site are influenced more deeply by the tumor microenvi-ronment, more susceptible to hepatic differentiation and expresstargeted agents specifically. The precise mechanism should beinvestigated further in future.

It is reported recently that bone marrow-derived MSCs frommice differentiate into the stromal compartment of hep-atocarcinoma xenografts and express stromal protein Tie2 and

Page 8: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

C. Yan et al. / Biomaterials 35 (2014) 3035e30433042

CCL5. These engineering modified MSCs can deliver tissue-specificsuicide gene which expression is controlled by Tie2 or CCL5 pro-moter and suppress the growth of hepatocarcinoma in vivo [30]. AsHUMSCs are more inclined to develop hepatic differentiation [29],in our studies, we use the AFP promoter which is active in the earlyphase of hepatic differentiation to regulate specifically theexpression of ILZ-sTRAIL. Only HUMSCs developing hepatic differ-entiation in the specific microenvironment of hepatocarcinoma butnot those developing stromal differentiation could secret thetherapeutic agent, which greatly enhance the selectivity andspecification of targeted tumor therapy.

As an isoleucine zipper was added to sTRAIL to facilitate trimerformation which was the active form of sTRAIL, and the secretedILZ-sTRAIL collected from the culture supernatant of 293 T cells didnot undergo purification process but was used directly, the con-centration of ILZ-sTRAIL used in our experiments was much lowerthan that used in the previous reports. In agree with the previousstudies [31,32], we found that 5-FU sensitized tumor cells to TRAIL-induced apoptosis by regulating the expression of some membersof Bcl-2 family and activation of caspase signal pathway. Though itis reported that 5-FU can increase the expression of death receptor4 and 5 (DR4 and DR5) of TRAIL [33], we did not find significantchanges in our study (data not shown). Interesting, ILZ-sTRAILdelivered and selectively expressed by HUMSCs in vivo exhibitedmuch greater capacity compared with that in vitro. This may bepartially resulted from activation of systemic antitumor immunityvia TRAIL-induced apoptosis with the evidence that muchlymphocyte infiltration was found in the groups treated withMSC.AFPILZ-sTRAIL.

5. Conclusion

We reported here a promising therapeutic strategy of MSC-based gene therapy against hepatocarcinoma. The expression oftherapeutic agents was regulated by the activation of AFP promoterduring the early stage of hepatic differentiation of HUMSCs. Thistherapeutic strategy takes good advantages of tumor tropism andhepatic differentiation potential of HUMSCs, which provides a po-tential way for targeted therapy. However, the interaction betweenMSCs and microenvironment is complicated and the exact mech-anism should be further explored before this therapeutic strategy istranslated into clinical.

Acknowledgments

This work is supported by grants from the Chinese NationalNatural Sciences Foundation (Grant Nos. 30873091, 30971291),National High-tech R&D Program of China (863 program Grant2011AA020118) and the Natural Sciences Foundation of Tianjin,People’s Republic of China (No. 05YFGZGX02800). We thank Dr.Linlin Jiang for the animal study.

Appendix A. Supplementary data

Supplementary data related to this article can be found at http://dx.doi.org/10.1016/j.biomaterials.2013.12.037.

References

[1] Friedenstein AJ, Deriglasova UF, Kulagina NN, Panasuk AF, Rudakowa SF,Luria EA, et al. Precursors for fibroblasts in different populations of hemato-poietic cells as detected by the in vitro colony assay method. Exp Hematol1974;2:83e92.

[2] Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D,et al. Minimal criteria for defining multipotent mesenchymal stromal cells.

The International Society for Cellular Therapy position statement. Cytotherapy2006;8:315e7.

[3] Kim SM, Lim JY, Park SI, Jeong CH, Oh JH, Jeong M, et al. Gene therapy usingTRAIL-secreting human umbilical cord blood-derived mesenchymal stem cellsagainst intracranial glioma. Cancer Res 2008;68:9614e23.

[4] Mohr A, Albarenque SM, Deedigan L, Yu R, Reidy M, Fulda S, et al. Targeting ofXIAP combined with systemic mesenchymal stem cell-mediated delivery ofsTRAIL ligand inhibits metastatic growth of pancreatic carcinoma cells. StemCells 2010;28:2109e20.

[5] Willhauck MJ, Sharif Samani BR, Klutz K, Cengic N, Wolf I, Mohr L, et al. a-Fetoprotein promoter-targeted sodium iodide symporter gene therapy ofhepatocellular carcinoma sodium iodide symporter gene therapy in livercancer. Gene Ther 2008;15:214e23.

[6] Li W, Li DM, Chen K, Chen Z, Zong Y, Yin H, et al. Development of a genetherapy strategy to target hepatocellular carcinoma based inhibition of pro-tein phosphatase 2A using the alpha-fetoprotein promoter enhancer and pgkpromoter: an in vitro and in vivo study. BMC Cancer 2012;12:547e56.

[7] Kwon OJ, Kim PH, Huyn S, Wu L, Kim M, Yun CO. A hypoxia- and alpha-fetoprotein-dependent oncolytic adenovirus exhibits specific killing of hepa-tocellular carcinomas. Clin Cancer Res 2010;16:6071e82.

[8] Lee KD, Kuo TKC, Whang-Peng J, Chung YF, Lin CT, Chou SH, et al. In vitrohepatic differentiation of human mesenchymal stem cells. Hepatology2004;40:1275e84.

[9] Sato Y, Araki H, Kato J, Nakamura K, Kawano Y, Kobune M, et al. Humanmesenchymal stem cells xenografted directly to rat liver are differentiatedinto human hepatocytes without fusion. Blood 2005;106:756e76.

[10] Aurich I, Mueller LP, Aurich H, Luetzkendorf J, Tisljar K, Dollinger MM, et al.Functional integration of hepatocytes derived from human mesenchymalstem cells into mouse livers. Gut 2007;56:405e15.

[11] Yu J, Cao H, Yang J, Pan Q, Ma J, Li J, et al. In vivo hepatic differentiation ofmesenchymal stem cells from human umbilical cord blood after trans-plantation into mice with liver injury. Biochem Biophys Res Commun2012;422:539e45.

[12] Ashkenazi A, Pai RC, Fong S, Leung S, Lawrence DA, Marsters SA, et al. Safetyand antitumor activity of recombinant soluble Apo2 ligand. J Clin Invest1999;104:155e62.

[13] Walczak H, Miller RE, Ariail K, Gliniak B, Griffith TS, Kubin M, et al. Tumor-icidal activity of tumor necrosis factor-related apoptosis-inducing ligandin vivo. Nat Med 1999;5:157e63.

[14] Kim MH, Billiar TR, Seol DW. The secretable form of trimeric TRAIL, a potentinducer of apoptosis. Biochem Biophys Res Commun 2004;321:930e5.

[15] Ma L, Feng XY, Cui BL, Frieda L, Jiang XW, Yang LY, et al. Human umbilical cordWharton’s jelly-derived mesenchymal stem cells differentiation into nerve-like cells. Chin Med J (Engl) 2005;118:1987e93.

[16] Jia H, Li Y, Zhao T, Li X, Hu J, Yin D, et al. Antitumor effects of Stat3-siRNA andendostatin combined therapies, delivered by attenuated Salmonella, onorthotopically implanted hepatocarcinoma. Cancer Immunol Immunother2012;61:1977e87.

[17] Yan XL, Jia YL, Chen L, Zeng Q, Zhou JN, Fu CJ, et al. Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression:role of the S100A4-miR155-SOCS1-MMP9 axis. Hepatology 2013;57:2274e86.

[18] Zhang T, Lee YW, Rui YF, Cheng TY, Jiang XH, Li G. Bone marrow-derivedmesenchymal stem cells promote growth and angiogenesis of breast andprostate tumors. Stem Cell Res Ther 2013;4:70e84.

[19] Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesen-chymal stem cells within tumour stroma promote breast cancer metastasis.Nature 2007;449:557e63.

[20] Fong CY, Chak LL, Biswas A, Tan JH, Gauthaman K, Chan WK, et al. HumanWharton’s jelly stem cells have unique transcriptome profiles compared tohuman embryonic stem cells and other mesenchymal stem cells. Stem CellRev 2011;7:1e16.

[21] Gauthaman K, Fong CY, Suganya CA, Subramanian A, Biswas A, Choolani M,et al. Extra-embryonic human Wharton’s jelly stem cells do not inducetumorigenesis, unlike human embryonic stem cells. Reprod Biomed Online2012;24:235e46.

[22] Gauthaman K, Yee FC, Cheyyatraivendran S, Biswas A, Choolani M, Bongso A.Human umbilical cord Wharton’s jelly stem cell (hWJSC) extracts inhibitcancer cell growth in vitro. J Cell Biochem 2012;113:2027e39.

[23] Subramanian A, Shu-Uin G, Kae-Siang N, Gauthaman K, Biswas A, Choolani M,et al. Human umbilical cord Wharton’s jelly mesenchymal stem cells do nottransform to tumor-associated fibroblasts in the presence of breast andovarian cancer cells unlike bone marrow mesenchymal stem cells. J CellBiochem 2012;113:1886e95.

[24] Riss J, Khanna C, Seongjoon K, Chandramouli GRV, Yang HH, Hu Y. Cancers aswounds that do not heal: differences and similarities between renal regen-eration/repair and renal cell carcinoma. Cancer Res 2006;66:7216e24.

[25] Kosztowski T, Zaidi HA, Quinones-Hinojosa A. Applications of neural andmesenchymal stem cells in the treatment of gliomas. Expert Rev AnticancerTher 2009;9:597e612.

[26] Honczarenko M, Le Y, Swierkowski M, Ghiran I, Glodek AM, Silberstein LE.Human bone marrow stromal cells express a distinct set of biologicallyfunctional chemokine receptors. Stem Cells 2006;24:1030e41.

[27] Jung KH, Shin HP, Lee S, Lim YJ, Hwang SH, Han H, et al. Effect of humanumbilical cord blood-derived mesenchymal stem cells in a cirrhotic rat model.Liver Int 2009;29:898e909.

Page 9: Suppression of orthotopically implanted hepatocarcinoma … · Suppression of orthotopically implanted hepatocarcinoma in mice by umbilical cord-derived mesenchymal stem cells with

C. Yan et al. / Biomaterials 35 (2014) 3035e3043 3043

[28] Mallet VO, Mitchell C, Mezey E, Fabre M, Guidotti JE, Renia L, et al. Bonemarrow transplantation in mice leads to a minor population of hepatocytesthat can be selectively amplified in vivo. Hepatology 2002;35:799e804.

[29] Buyl K, De Kock J, Najar M, Lagneaux L, Branson S, Rogiers V, et al. Charac-terization of hepatic markers in human Wharton’s Jelly-derived mesenchymalstem cells. Toxicol In Vitro 2014;28:113e9.

[30] Niess H, Bao Q, Conrad C, Zischek C, Notohamiprodjo M, Schwab F, et al. Se-lective targeting of genetically engineered mesenchymal stem cells to tumorstroma microenvironments using tissue-specific suicide gene expressionsuppresses growth of hepatocellular carcinoma. Ann Surg 2011;254:767e75.

[31] Mizutani Y, Nakanishi H, Yoshida O, Fukushima M, Bonavida B, Miki T.Potentiation of the sensitivity of renal cell carcinoma cells to TRAIL-mediated

apoptosis by subtoxic concentrations of 5-fluorouracil. Eur J Cancer 2002;38:167e76.

[32] Ganten TM, Haas TL, Sykora J, Stahl H, Sprick MR, Fas SC, et al. Enhancedcaspase-8 recruitment to and activation at the DISC is critical for sensitisationof human hepatocellular carcinoma cells to TRAIL-induced apoptosis bychemotherapeutic drugs. Cell Death Differ 2004;11(Suppl. 1):S86e96.

[33] Yamamoto T, Nagano H, Sakon M, Wada H, Eguchi H, Kondo M, et al. Partialcontribution of tumor necrosis factor-related apoptosis-inducing ligand(TRAIL)/TRAIL receptor pathway to antitumor effects of interferon-a/5-fluorouracil against hepatocellular carcinoma. Clin Cancer Res 2004;10:7884e95.