Teratoma Formation in Immunocompetent Mice After (1)

7

Click here to load reader

Transcript of Teratoma Formation in Immunocompetent Mice After (1)

Page 1: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 1/7

 Asian Pacic Journal of Cancer Prevention, Vol 14, 2013 5705

 DOI:http://dx.doi.org/10.7314/APJCP.2013.14.10.5705

Teratoma Formation in Mice after Syngeneic and Allogeneic Implantation of Mouse Embryonic Stem Cells

 Asian Pac J Cancer Prev, 14 (10), 5705-5711

Introduction

  The use of embryonic stem cells (ESCs) in the

treatment of human diseases has long been muted, because

such a treatment must not only be efcacious but must

also be shown to be completely safe (Baker et al., 2007;

Lukovic et al., 2013). Indeed, in many previous studies,

the use of injected ESCs was associated with the risk of

teratoma development (Wakitani et al., 2003; Baharvand

and Matthaei, 2004; Przyborski, 2005; Nussbaum et al.,

2007; Dressel et al., 2008; Prokhorova et al., 2009; Zhang

et al., 2012; Li et al., 2013). In a clinical setting, this is

a critical limitation of such a treatment. Therefore, much

1Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University and King Khalid University Hospital, Riyadh,

Kingdom of Saudi Arabia, 4 Biology Department, College of Science, King Abdulaziz University, Saudi Arabia, 2KMEB, Department of

 Endocrinology, University of Southern Denmark, Odense, Denmark , 3 Histology Department, Faculty of Medicine, Cairo University,

 Egypt, 5Stem Cell and Gene Targeting Laboratory, The John Curtin School of Medical Research, The Australian National University,

Canberra ACT Australia *For correspondence: [email protected], [email protected]

Abstract

  Background: Embryonic stem cells (ESCs) have the potential to form teratomas when implanted into

immunodecient mice, but data in immunocompetent mice are limited. We therefore investigated teratoma

formation after implantation of three different mouse ESC (mESC) lines into immunocompetent mice. Materials

and Methods: BALB/c mice were injected with three highly germline competent mESCs (129Sv, BALB/c and

C57BL/6) subcutaneously or under the kidney capsule. After 4 weeks, mice were euthanized and examined

histologically for teratoma development. The incidence, size and composition of teratomas were compared using

Pearson Chi-square, t-test for dependent variables, one-way analysis of variance and the nonparametric Kruskal-

Wallis analysis of variance and median test. Results: Teratomas developed from all three cell lines. The incidence

of formation was signicantly higher under the kidney capsule compared to subcutaneous site and occurred in

both allogeneic and syngeneic mice. Overall, the size of teratoma was largest with the 129Sv cell line and under

the kidney capsule. Diverse embryonic stem cell-derived tissues, belonging to the three embryonic germ layers,

were encountered, reecting the pluripotency of embryonic stem cells. Most commonly represented tissues

were nervous tissue, keratinizing stratied squamous epithelium (ectoderm), smooth muscle, striated muscle,cartilage, bone (mesoderm), and glandular tissue in the form of gut- and respiratory-like epithelia (endoderm).

Conclusions: ESCs can form teratomas in immunocompetent mice and, therefore, removal of undifferentiated

ESC is a pre-requisite for a safe use of ESC in cell-based therapies. In addition the genetic relationship of the

origin of the cell lines to the ability to transplant plays a major role. 

Keywords: Mouse embryonic stem cells - teratoma - immunocompetent - syngeneic - allogeneic 

RESEARCH ARTICLE

Teratoma Formation in Immunocompetent Mice After

Syngeneic and Allogeneic Implantation of Germline Capable

Mouse Embryonic Stem Cells

Abdullah Aldahmash1,2*, Muhammad Atteya1,3, Mona Elsafadi1, May Al-

Nbaheen1, Husain Adel Al-Mubarak1, Radhakrishnan Vishnubalaji1, Ali Al-

Roalle1, Suzan Al-Harbi4, Muthurangan Manikandan1, Klaus Ingo Matthaei1,5,

Amer Mahmood1*

interest among the scientic community has been on the

understanding of these processes. Most studies have used

immunodecient recipient mice into which these cellswere implanted (Wakitani et al., 2003; Baharvand and

Matthaei, 2004; Przyborski, 2005; Nussbaum et al., 2007;

Dressel et al., 2008; Prokhorova et al., 2009; Li et al.,

2013). The resultant teratomas consisted of all three germ

layers (ectoderm, mesoderm, endoderm) representing all

tissue types in a whole individual (Prokhorova et al., 2009).

The use of immunodecient mice appears logical since it

could be expected that immunocompetent animals would

reject these cells. However, one study (Dressel et al., 2008)

has surprisingly reported that the syngeneic implantation

Page 2: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 2/7

 Abdullah Aldahmash et al

 Asian Pacic Journal of Cancer Prevention, Vol 14, 20135706

of 129Sv ESCs into 129Sv immunocompetent mice also

gave rise to teratomas consisting of all three germ layers,

and at a high frequency. We wondered, therefore, if the

state of undifferentiation, or pluripotency, of the injected

cells may play a critical role in this process. The present

study was, therefore, designed specically to investigate

this possibility by using three mouse ESC lines of

early passage that have been proven to have germlinetransmission capacity at similar passage stages. Moreover,

we were interested in determining whether these cells

could form teratomas not only in syngeneic but also in

allogeneic immunocompetent hosts. Here we report that

ESCs can form teratomas in immunocompetent mice after

allogeneic as well as syngeneic implantation. 

Materials and Methods

 Ethics statement 

  All animal experimental procedures of this study

were approved by the Institutional Review Board (Ethical

Approval ID 11/3215/IRB), College of Medicine, King

Saud University, Riyadh, Kingdom of Saudi Arabia.

Cell culture

  Three mouse ESC (mESC) lines were used in this

study: 129Sv W9.5 passage (p) 19, BALB/c p11, and

C57BL/6 p13. The cell lines were provided by the Stem

Cell and Gene Targeting Laboratory, John Curtin School

of Medical Research, the Australian National University,

Canberra ACT 0200, Australia, and were transferred to

King Saud University in liquid nitrogen. All three cell

lines have been published previously and were shown to

be germline competent at comparable passages; 129Sv(Szabó and Mann, 1994; Yang et al., 2006), BALB/c

(Royan C1) (Baharvand and Matthaei, 2004), and

C57BL/6 (Royan B1) (Baharvand and Matthaei, 2004;

Sadraei et al., 2009).

  The mESCs were grown on mitomycin C-treated

embryonic feeder cells in complete ESC medium

(Dulbecco’s modied Eagle’s medium (DMEM) (Gibco

ES cell qualied 1829, Invitrogen) supplemented with

15% ES qualied fetal bovine serum (Gibco 16141-079,

Invitrogen), 2mM Glutamine (Gibco, 25030, Invitrogen),

0.1 mM MEM nonessential amino acids (Gibco, 11140,

Invitrogen), and 0.1mM 2-mercaptoethanol (Sigma,

M7522), 50 U penicillin, and 50 U streptomycin (GIBCO

15140-148, Invitrogen), and 1,000 U/mL leukemia

inhibitory factor (LIF) (ESGRO ESG1106, Millipore) as

previously described (Matthaei, 2009) but in 5%CO2 in

air.

  Mouse ESCs were grown to near conuence. Medium

was aspirated from the ask. The adhered cells were

washed twice with Ca2+ Mg2+-free phosphate-buffered

saline (PBS) (Gibco, 20012), and incubated at room

temperature for 2 minutes with PBS containing 0.5mM

ethylene glycol tetra-acetic acid (EGTA) (Sigma-Aldrich

E3889, St. Louis, USA), then for 1-2 minutes with

0.05% trypsin-ethylenediaminetetraacetic acid (EDTA)(Gibco 25300, Invitrogen) (Matthaei, 2009). To obtain

a single cell suspension, the cells were triturated rapidly

in the trypsin solution several times through a small-bore

pipette. The trypsin was inactivated by the addition of

complete ESC medium. The cell suspension was plated

into a 10 cm tissue culture dish and incubated at 37°C for

15-30 minutes to allow mouse embryonic feeder (MEF)

cells to attach. The ESCs were then carefully aspirated,

the cell suspension counted using a hemocytometer and

centrifuged at 1000 rpm for 5 minutes to obtain a cell

pellet for appropriate dilution.

 Immunocytochemical staining

  For immunostaining, cells were grown to 80-90%

conuence on 4 Permanox chamber slides. The slides were

xed in 4% formaldehyde for 10 minutes and then washed

three times in PBS. Nonspecific immunoglobulin G

binding sites were blocked for 20 minutes with 5% bovine

serum albumin (BSA) in PBS. The cells were labeled

with primary antibodies [anti-mouse Oct3/4 (1-5 µg/ml,

Abcam), anti-mouse Sox2 (3 µg/ml, Abcam), and anti-

mouse SSEA1(1/100, Abcam)], incubated for 1-2 hours,

and then stained with anti-rat or anti-rabbit FITC (Abcam)

secondary antibodies. All cells were counterstained with

DAPI for nuclei.

Teratoma formation

  Forty-six BALB/c mice (a kind gift by the Stem Cell

and Gene Targeting Laboratory, John Curtin School of

Medical Research, the Australian National University,

Canberra ACT 0200, Australia) 6-10 weeks old were

used as host animals for the implantation of three mESC

lines (129Sv, BALB/c and C57BL/6). Injections were

performed under the kidney capsule in 21 mice and into

the subcutaneous tissue in 25 mice. For renal subcapsular

injection, 15106 mESCs/injection were resuspendedthoroughly in 20µl cold complete ESC medium with

20mM HEPES (Gibco, 15630) in 1.5ml sterile Eppendorf

tubes. For subcutaneous injection, 15106 mESCs/injection

were resuspended in 150µl cold complete ESC medium

in 1.5ml sterile Eppendorf tubes and kept on ice prior to

injection. Subcutaneous injection was performed at 2 sites

(ank and neck) in each animal.

 Injection under the kidney capsule

  The mice were anesthetized with Sevourane and the

right kidney was exposed via a right paralumbar incision.

A transverse tear was made in the ventral aspect of the

kidney capsule using microforceps. Air was then injected

using a 22G intravenous cannula through the tear to create

a pocket. The same size cannula was used to inject the

cells into the pocket. Then the tear in the renal capsule

was closed with cautery. The abdominal wall musculature

and overlying skin were closed with 6-0 vicryl sutures.

Subcutaneous injection

  The mice were anesthetized with Sevourane. The

cells were resuspended gently using a 25G syringe needle

and then drawn into a 1ml syringe immediately before

injection. The skin at the site of injection was lifted and

the cells were injected into the subcutaneous space.

 Histological analysis

  Four weeks after the injection, the animals were

Page 3: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 3/7

 Asian Pacic Journal of Cancer Prevention, Vol 14, 2013 5707

 DOI:http://dx.doi.org/10.7314/APJCP.2013.14.10.5705

Teratoma Formation in Mice after Syngeneic and Allogeneic Implantation of Mouse Embryonic Stem Cells

euthanized and the sites of implantation were dissected

for histological examination. Harvested tissues were xed

for 48hrs in 4% paraformaldehyde, parafn sectioned

at 5µm and stained with hematoxylin and eosin. High-

resolution whole-slide digital scans of all glass slides

were then created with a ScanScope scanner (Aperio

Technologies, Inc.). The digital slide images were then

viewed and analyzed using the viewing and image analysistools of Aperio’s ImageScope software. The size of each

teratoma in terms of its cut-sectional area was measured

in mm2. The composition of each teratoma was estimated

semi-quantitatively by visual inspection of digital slide

images. Differentiated elements in the teratomas were

classied into ectodermal (skin, neural), mesodermal

(cartilage, bone, muscle), and endodermal (glandular

structures) components as a percentage of entire tissue.

The percentages of undifferentiated tissue, cysts, necrosis

and hemorrhage were also recorded. The remaining

spindle-shaped stroma was included under mesoderm.

Statistical analysis

  The incidence of teratoma was compared using

Pearson Chi-square. For composition of teratoma, t-test

for dependent variables was used for within group

comparisons, while one-way analysis of variance was

used for between group comparisons. Because the size

varied widely among teratomas, it was compared between

groups using the nonparametric Kruskal-Wallis analysis

of variance and median test. Results were considered

signicant when p<0.05.

Results

  The three mESC lines were cultured as undifferentiated

ESCs in the presence of MEFs and LIF. Colonies from

all cell lines exhibited positive staining for Oct3/4,

Sox2, and SSEA1 (Figure 1). We then analyzed the

tumor formation ability of ESCs (129Sv, BALB/c and

C57BL/6) by syngeneic and allogeneic implantation

into immunocompetent BALB/c mice. We found that

the incidence of teratoma formation after syngeneic

implantation of BALB/c ESCs into immunocompetent

BALB/c mice was 16.67% (Table 1). Unexpectedly,

however, we observed that when we implanted 129Sv

and C57BL/6 ESCs into immunocompetent BALB/c mice

(allogeneic implantation) teratomas were also formed in

33.3% and 30.43%, respectively (Table 1).

An encapsulated solid tumor was found at the

implantation site in 30% and 40% of 129Sv-implanted

mice, in 0% and 37.5% of BALB/c-implanted mice,

and in 25% and 66.7% of C57BL/6-implanted mice, for

subcutaneous and renal implantation, respectively (Table

1, Figure 2A). The overall teratoma development success

rate was 20 out of 71 injections (28.17%). The overall

incidence was highest with 129Sv (33.3%) followed byC57BL/6 (30.43%) and lowest with BALB/c (16.67%).

For all cell lines combined, the incidence of teratoma was

signicantly higher under the kidney capsule (42.86%)

than in the subcutaneous tissue (22%; p=0.0395). No

teratoma development was observed in mice that had

received the BALB/c cell line subcutaneously.

  To investigate whether the site of injection plays a role

in the size of teratoma formation we analyzed the size

of teratomas based on the site of injection. The overall

tumor size appeared largest when derived from 129Sv

(66.07±99.43 mm2) followed by C57BL/6 (35.02±35.37

mm2) and smallest with BALB/c (5.30±4.77 mm2) ESCs.

However, these differences did not reach statistical

signicance. Similarly the overall size appeared largest

under the kidney capsule (53.80±109.84 mm2) followed

by the subcutaneous tissue (39.78±29.79 mm2). The

cut-sectional area of teratomas developed by 129Sv

Figure 1. Characterization of the Three Different

mESC Lines. Upper panel shows live cell image and colony

formation of the three mESC lines grown under standard ESC

conditions. Lower 3 panels show immunouorescence cellstaining of pluripotent colony cells with positivity for Oct4, Sox2

and SSEA1. Nuclear stain DAPI was used as a counter staining

Table 1. Incidence, Mean Size and Percentages of Components of Teratomas Formed After Subcutaneous and

Renal Subcapsular Injection of Three mESC Lines

Cell Line Site Incidence Size (mm2) Ectoderm Mesoderm Endoderm Undiff. H/N Cysts

129Sv SC 30.00% (6/20) 35.40±23.99 40.33%±17.91 36.33%±21.69 9.33%±5.61 4.83%±2.93 2.17%±3.92 5.33%±10.03

  Renal 40.00% (4/10) 112.09±154.89 53.5%±34.58 7.75%±4.50 8.00%±4.00 14.5%±20.60 3.75%±1.50 12.5%±9.57

  Total 33.30% (10/30) 66.07±99.43 45.6%±24.96 24.9%±22.05 8.8%±4.83 8.7%±13.08 2.8%±3.16 8.2%±10.01

BALB/c SC 0.00% (0/10) – – – – – – –

  Renal 37.50% (3/8) 5.30±4.77 83.33%±15.28 5.0%±5.00 3.33%±2.89 5.67%±5.13 1.67%±1.53 1.0%±1.73

  Total 16.67% (3/18) 5.30±4.77 83.33%±15.28 5.0%±5.00 3.33%±2.89 5.67%±5.13 1.67%±1.53 1.0%±1.73

C57BL/6 SC 25.00% (5/20) 45.04±37.88 73.60%±6.12 14.40%±7.54 6.20%±2.77 3.00%±1.00 1.60%±1.53 1.2%±2.17

  Renal 66.70% (2/3) 9.97±2.60 80.00%±1.41 4.5%±2.12 3.5%±0.71 10.00%±0.00 1.00%±0.00 1.00%±1.41

  Total 30.43% (7/23) 35.02±35.37 75.43%±5.91 11.57%±7.87 5.43%±2.64 5.00%±3.51 1.43%±1.72 1.14%±1.86

Overall 28.17% (20/71) 46.09±74.82 61.7%±24.71 17.25%±17.86 6.8%±4.34 6.95%±9.54 2.15%±2.52 4.65%±7.88

*SC, subcutaneous; undiff., undifferentiated tissue; H/N, hemorrhage and necrosis

Page 4: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 4/7

 Abdullah Aldahmash et al

 Asian Pacic Journal of Cancer Prevention, Vol 14, 20135708

cell line measured an average of 35.40±23.99 mm2 and

112.09±154.89 mm2 in the subcutaneous tissue and under

kidney capsule, respectively. No teratoma development

was observed in the subcutaneous tissue of mice that

received the BALB/c cell line, while those that developed

under the kidney capsule were only small and measured

an average of 5.3±4.77 mm2 (Figure 2B). The teratomas

that developed from the C57BL/6 cell line measured anaverage of 45.04±37.88 mm2 in the subcutaneous tissue

and 9.97±2.60 mm2 under the kidney capsule (Table 1,

Figure 2B). The largest sizes were observed with the

129Sv cell line injected under the kidney capsule and with

the C57BL/6 cell line in the subcutaneous tissue (Figure

2B). Notably, the larger the teratoma had grown, the more

tissue types could be identied histologically. We also

found that the incidence and size of teratoma formation

are site-dependent; injection under the kidney capsule

having the highest success rate and producing the largest

teratoma sizes (Figure 3A).

  Diverse ESC-derived tissues, belonging to the three

embryonic germ layers, were encountered, reecting the

pluripotency of the ESCs. Most commonly present tissues

were nervous tissue, keratinizing stratied squamous

epithelium (ectoderm), smooth muscle, striated muscle,

cartilage, bone (mesoderm), and glandular tissue in the

form of gut- and respiratory-like epithelia (endoderm), in

that order (Figure 3B). The overall average composition of

teratomas (n=20) was toward signicantly more ectoderm

(61.70%±24.71) than mesoderm (17.25%±17.85,

p=0.0001) or endoderm (6.80%±4.34, p=0.0000) and

signicantly more mesoderm than endoderm (p=0.0085)

(Table 1, Figure 4D). Teratomas that developed from

the 129Sv cell line in the subcutaneous tissue showed

significantly more mesoderm than teratomas that

developed under the kidney capsule (p=0.017) (Figure4A). However, C57BL/6 and BALB/c did not show any

signicant difference in germ layer composition between

injection sites (Figure 4B-C). Teratomas that developed

Figure 2. Development of Teratomas by Three Different

mESC Lines in BALB/c Mice. A) Average teratoma

development at the different sites as a percentage; and B) Average

teratoma size at the different sites (mm2). SC, subcutaneous;

*Signicant difference (p<0.05)

Figure 4. Germ Layer Composition of Teratomas

Derived from the Different Cell Lines. A) to C) comparison

between sites for the 129Sv, C57BL/6 and BALB/c, respectively;

and D) Overall comparison of germ layer composition of all

teratomas formed, **Signicant difference from meso- and

endoderm; *Signicant difference from endoderm (p<0.05)

Figure 3. Histological Analysis of Teratomas. A) Gross

anatomy and overview of teratoma cut-section; B) Histological

analysis of teratomas identifying the presence of derivatives of

all three germ layers: neural tissue and keratinizing epithelium

(ectoderm), cartilage, striated muscle and trabecular bone

(mesoderm) and respiratory-like, gut-like and mucinous

glandular epithelium (endoderm); and C) Other tissue elements:

undifferentiated tissue with malignant nuclear criteria includingpleomorphism, hyperchromatism, frequent mitotic figures

(arrowheads) and tumor giant cells with bizarre nuclei (white

arrows), necrotic tissue and cysts lined by cuboidal epithelium

(black arrow). Scale bars represent 100 μm

Figure 5. Germ Layer Composition of Teratomas

Derived from the Different Cell Lines. Comparison of

cell lines in subcutaneous A) and renal subcapsular B) teratomas

Page 5: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 5/7

 Asian Pacic Journal of Cancer Prevention, Vol 14, 2013 5709

 DOI:http://dx.doi.org/10.7314/APJCP.2013.14.10.5705

Teratoma Formation in Mice after Syngeneic and Allogeneic Implantation of Mouse Embryonic Stem Cells

from the C57BL/6 cell line in the subcutaneous site

contained signicantly more ectoderm than teratomas

that developed from the 129Sv cell line at the same

site (p=0.0017) (Table 1, Figure 5A). Teratomas that

developed under the kidney capsule did not show any

signicant difference in germ layer composition between

the three cell lines; however they all showed a high

tendency towards ectoderm (Figure 5B).  Clusters of undifferentiated cells with malignant nuclear

criteria (including pleomorphism, hyperchromatism

and frequent mitotic figures) were common in most

of the tumors (Figure 3C). The overall percentage of

undifferentiated cells was higher with 129Sv (8.7%±13.08)

than BALB/c (5.67%±5.13) or C57BL/6 (5%±3.51) with

no statistically signicant differences (Table 1). Site

comparison showed that, overall, renal subcapsular

implantation was associated with a signicantly higher

percentage of undifferentiated cells than subcutaneous

implantation.

Discussion

The use of stem cell therapy for the treatment of

human diseases would dramatically improve our ability

to alleviate debilitating conditions. The potential of using

ESCs for this purpose has been muted since these cells

have the ability to differentiate into every cell type in

an entire individual. This would suggest that all organs

could be treated once methods had been developed to

direct the ESCs to differentiate into specic target tissues

thereby eliminating the problem of limited donor tissue for

treatments. However, concerns have been raised over the

safety of this promising therapeutic approach, includingwhether the transplanted ESCs have the potential to form

tumors after engraftment because of their unlimited self-

renewal and high differentiation potential (Arnhold et al.,

2004; Amariglio et al., 2009; Lukovic et al., 2013).

A number of studies (Thomson et al., 1998; Wakitani

et al., 2003; Stojkovic et al., 2005; Nussbaum et al.,

2007; Dressel et al., 2008; Prokhorova et al., 2009;

Zhang et al., 2012; Li et al., 2013; Lukovic et al., 2013)

have shown the potential of ESCs to form teratomas by

injecting undifferentiated ESCs into mice. Most studies

used immunodecient hosts such as severe combined

immunodecient (SCID) mice (Thomson et al., 1998;

Wakitani et al., 2003; Stojkovic et al., 2005; Zhang et

al., 2012). Dressel et al. (2008) surprisingly showed that

teratoma development could also occur after injection of

ESCs into syngeneic immunocompetent mice. This made

us consider whether the state of differentiation of the ESCs

could inuence the outcome. The question, therefore, is

whether the more undifferentiated ESCs are, the more

they are prone to form teratomas.

We have for many years been using mouse ESCs from

3 different mouse strains to generate genetically modied

mice. This process is completely reliant on the ability of

the ESCs to remain totally pluripotent and integrate into

the germline of chimera after injection into blastocysts;thereby producing ESC-derived offspring after mating

with wild-type mice. Our cell lines had all been proven

to be fully undifferentiated and germline competent using

this technique (Campbell et al., 2002; Baharvand and

Matthaei, 2004; Yang et al., 2006; Meyer et al., 2010). We

therefore decided to test the ability of all 3 mouse ESC

lines to form teratomas in immunocompetent mice, both

in syngeneic as well as allogeneic combinations.

The present study shows that our ESCs had the

capability to form teratomas after injection into syngeneic

as well as allogeneic immunocompetent mice (Table 1).The incidence of teratomas was higher after implantation

of ESCs from the 129Sv strain when compared to ESCs

from the C57BL/6 and BALB/c strains. This might be

expected since the 129Sv strain is most commonly used

due to its high germline competence and stability during

continued cell culture and expansion. Interestingly, in this

study, syngeneic implantation of the BALB/c cells into

BALB/c mice failed to produce teratomas in subcutaneous

tissue while those that developed under the kidney capsule

were only small. Previous studies reported teratomas

developing after syngeneic implantation (129Sv ESCs

into 129Sv mice (Arnhold et al., 2004; Swijnenburg et

al., 2005; Dressel et al., 2008) or C57BL/6 ESCs into

C57BL/6 mice (Nussbaum et al., 2007), but syngeneic

implantation of BALB/c ESCs into BALB/c mice has not

been described previously. In previous studies (Nussbaum

et al., 2007; Xie et al., 2007) allogeneic ESCs also caused

teratomas. However, in these previous studies, allogeneic

teratomas initially detected were gradually rejected by an

increasing inammatory immune response. These ndings

suggested that immunogenicity of ESCs increases on their

differentiation, i.e. once ESCs reach a more differentiated

state, they can be more effectively recognized and rejected

by the recipient immune system. In the present study, the

reason why we did not encounter such immune rejectionafter allogeneic implantation is, at this stage, not known.

Histologic analysis of the teratomas showed that

the most commonly present tissues were nervous

tissue, keratinizing stratified squamous epithelium

(ectoderm), smooth muscle, striated muscle, cartilage,

bone (mesoderm), and glandular tissue in the form of

gut- and respiratory-like epithelia (endoderm), in that

order. Hence diverse ESC-derived tissues, belonging

to the three embryonic germ layers, were encountered,

thereby reecting the pluripotency of the ESCs (Thomson

et al., 1998; Oosterhuis and Looijenga, 2005; Mahmood

et al., 2010; Bustamante-Marín et al., 2013; Pal et al.,

2013). Importantly, this suggests that the differentiation

potential of the 3 mouse ESC lines used is independent

of microenvironmental factors that may be encountered

at the injection site. These results are in agreement

with previous studies that implanted a heterogeneous

differentiated ESC population into brain (Riess et al.,

2007), heart (Xie et al., 2007) and retina (Cui et al.,

2013) and found that any remaining undifferentiated cells

formed teratomas. Hence, our data also support that care

should be taken that ESCs are fully differentiated before

their use for cell-based therapy (Prokhorova et al., 2009),

since any remaining undifferentiated cells, if irresponsive

to microenvironmental differentiation signals, can formteratomas. Moreover, even after thorough selection of a

highly puried population of ESC-derived neural (Arnhold

et al., 2004; Lukovic et al., 2013) and islet (Fujikawa et al.,

Page 6: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 6/7

 Abdullah Aldahmash et al

 Asian Pacic Journal of Cancer Prevention, Vol 14, 20135710

2005) precursor cells, there remains a high risk of teratoma

formation after engraftment. However, transplantation

of FACS purified ESC-derived neural precursors

expressing the neural precursor marker Sox1 into mouse

brains (Fukuda et al., 2006) and targeting hESC-specic

antiapoptotic factor(s) (i.e., survivin or Bcl10) represent

an efcient strategy to selectively eliminate pluripotent

cells with teratoma potential (Lee et al., 2013).We found, in accordance with Cooke et al. (2006),

that the incidence and size of teratoma formation are

site-dependent; injection under the kidney capsule

having the highest success rate and producing the largest

teratoma sizes. Because of its rich vascular bed, the renal

subcapsular space of mice is a suitable environment

for growing and studying solid tumors (Press et al.,

2008). This makes renal subcapsular implantation better

to subcutaneous implantation for teratoma induction

experiments, in terms of success rate and size of teratomas

produced.

In this study, clusters of undifferentiated cells

exhibiting malignant nuclear criteria (including

pleomorphism, hyperchromatism and frequent mitotic

gures) were common in most of the teratomas. These

can be considered as malignant teratomas based on the

presence of tissues of the three germ cell layers and the

presence of both anaplastic foci and immature tissues

(López and Múrcia, 2008). The overall percentage

of undifferentiated cells (malignant-looking clusters)

was higher with 129Sv than BALB/c or C57BL/6

ESCs. Renal subcapsular implantation was associated

with a higher percentage of undifferentiated cells than

subcutaneous implantation. This agrees with Cooke

et al. (2006) who reported that subcutaneous implantswere signicantly slower growing and formed tumors

composed of differentiated tissues, in contrast to cells

grafted into the liver which rapidly produced large tumors

containing predominantly immature cells. It is still largely

unknown why ESCs, which lack overt chromosomal

abnormalities, are tumorigenic. One hypothesis is based

on the identication of the canonical WNT signaling

as a critical determinant for the tumorigenicity and

therapeutic function of ESC-derived retinal progenitors.

The function of WNT signaling is primarily mediated by

TCF7, which directly induces expression of Sox2 and

Nestin (Cui et al., 2013). Also, similarity between ESCs

and cancer cells has been proposed. The embryonic stem

cell factor Sox2 is reported to be expressed in a variety

of early stage postmenopausal breast carcinomas and

metastatic lymph nodes (Lengerke et al., 2011). The study

of Wong et al. (2008) demonstrated that embryonic stem

cells and multiple types of human cancer cells share a

genetic expression pattern that is repressed in normal

differentiated cells. Recent discoveries also indicated that

testicular carcinoma in situ (CIS) cells have a striking

phenotypic and genotypic similarity to ESCs (Almstrup

et al., 2006). The similarities between ESCs and cancer

cells should be further investigated.

In the present study, implantation of mESCscaused teratomas in both syngeneic and allogeneic

immunocompetent mice. It could be argued that our data

are not consistent with a previously published report

(Dressel et al., 2008) in which allogeneic transplants of

mESC did not result in teratomas in immunocompetent

hosts. However, the state of pluripotency of the ESCs

used in that study (Dressel et al., 2008) was not clearly

described, in contrast to our ESCs with proven germline

competence. In addition, because of their early stage of

development, ESCs are considered “immune privileged,”

i.e, unrecognizable by the recipient immune defenses(Magliocca et al., 2006; Ichiryu and Fairchild, 2013).

Moreover, it has been reported that mESCs are resistant

to antigen-specic cytotoxic T lymphocyte (CTL) due to

the expression of serpin-6, an endogenous inhibitor of

granzyme B (Abdullah et al., 2007). Dressel et al. (2008)

stated that the MPI-II cells they used were susceptible to

granule exocytosis-mediated killing by rat NK cells and

also by peptide-specic mouse CTL. So their cells may

not have been protected against cellular cytotoxicity and

hence their allogeneic transplants into immunocompetent

mice were rejected soon after injection. In contrast, our

cells were not rejected after allogeneic implantation

in immunocompetent mice since they may have been

immune privileged and thereby escaped cytotoxic immune

destruction and formed teratomas. Nevertheless, such

argument warrants that the susceptibility of different ES

cell lines to the cytotoxic immune response be further

investigated.

In conclusion, we have conrmed that ESCs can form

teratomas in immunocompetent mice. Furthermore, we

showed that germline competent ESC can form teratomas

not only in syngeneic but also in allogeneic hosts. It is

clear, therefore, that in a clinical setting, it must be ensured

that no undifferentiated cells remain which could form

teratomas. Without this assurance, the use of ESCs shouldnot be considered as a therapeutic option for patients with

degenerative disease.

Acknowledgements

This work was supported by a grant (No. 10-BIO1304-

02) and (10-BIO1308-02) from the National Plan for

Sciences and Technology Program, King Saud University

and a grant from the International Scientic Twinning

Program, King Saud University. Our thanks must also go

to Professor Moustapha Kassem.

References

Abdullah Z, Saric T, Kashkar H, et al (2007). Serpin-6 expression

protects embryonic stem cells from lysis by antigen-specic

CTL. J Immunol, 178, 3390-9.

Almstrup K, Sonne SB, Hoei-Hansen CE, et al (2006). From

embryonic stem cells to testicular germ cell cancer-- should

we be concerned? Int J Androl, 29, 211-8.

Amariglio N, Hirshberg A, Scheithauer BW, et al (2009).

Donor-derived brain tumor following neural stem cell

transplantation in an ataxia telangiectasia patient. PLoS

 Med , 6, 1000029.

Arnhold S, Klein H, Semkova I, Addicks K, Schraermeyer U

(2004). Neurally selected embryonic stem cells induce tumor

formation after long-term survival following engraftment

into the subretinal space.  Invest Ophthalmol Vis Sci, 45,

4251-5.

Page 7: Teratoma Formation in Immunocompetent Mice After (1)

8/10/2019 Teratoma Formation in Immunocompetent Mice After (1)

http://slidepdf.com/reader/full/teratoma-formation-in-immunocompetent-mice-after-1 7/7

 Asian Pacic Journal of Cancer Prevention, Vol 14, 2013 5711

 DOI:http://dx.doi.org/10.7314/APJCP.2013.14.10.5705

Teratoma Formation in Mice after Syngeneic and Allogeneic Implantation of Mouse Embryonic Stem Cells

Baharvand H, Matthaei KI (2004). Culture condition difference

for establishment of new embryonic stem cell lines from the

C57BL/6 and BALB/c mouse strains. In vitro Cell Dev Biol

 Anim, 40, 76-81.

Baker DEC, Harrison NJ, Maltby E, et al (2007). Adaptation to

culture of human embryonic stem cells and oncogenesis in

vivo. Nat Biotechnol, 25, 207-15.

Bustamante-Marín X, Garness JA, Capel B (2013). Testicular

teratomas: an intersection of pluripotency, differentiationand cancer biology. Int J Dev Biol, 57, 201-10.

Campbell HD, Fountain S, McLennan IS, et al (2002). Fliih,

a gelsolin-related cytoskeletal regulator essential for early

mammalian embryonic development.  Mol Cell Biol, 22,

3518-26.

Cooke MJ, Stojkovic M, Przyborski SA (2006). Growth of

teratomas derived from human pluripotent stem cells is

inuenced by the graft site. Stem Cells Dev, 15, 254-9.

Cui L, Guan Y, Qu Z, et al (2013). WNT signaling determines

tumorigenicity and function of ESC-derived retinal

progenitors. J Clin Invest , 123, 1647-61.

Dressel R, Schindehütte J, Kuhlmann T, et al (2008). The

tumorigenicity of mouse embryonic stem cells and in vitro

differentiated neuronal cells is controlled by the recipients’immune response. PLoS ONE , 3, 2622.

Fujikawa T, Oh S-H, Pi L, et al (2005). Teratoma formation leads

to failure of treatment for type I diabetes using embryonic

stem cell-derived insulin-producing cells.  Am J Pathol,

166, 1781-91.

Fukuda H, Takahashi J, Watanabe K, et al (2006). Fluorescence-

activated cell sorting-based purication of embryonic stem

cell-derived neural precursors averts tumor formation after

transplantation. Stem Cells, 24, 763-71.

Ichiryu N, Fairchild PJ (2013). Immune privilege of stem cells.

 Methods Mol Biol, 1029, 1-16.

Lee M-O, Moon SH, Jeong H-C, et al (2013). Inhibition of

pluripotent stem cell-derived teratoma formation by small

molecules. Proc Natl Acad Sci USA, 110, 3281-90.

Lengerke C, Fehm T, Kurth R, et al (2011). Expression of the

embryonic stem cell marker SOX2 in early-stage breast

carcinoma. BMC Cancer, 11, 42.

Li P, Chen Y, Xiaoming M, et al (2013). Suppression of

malignancy by Smad3 in mouse embryonic stem cell formed

teratoma. Stem Cell Rev, [Epub ahead of print].

López RM, Múrcia DB (2008). First description of malignant

retrobulbar and intracranial teratoma in a lesser kestrel (Falco

naumanni). Avian Pathol, 37, 413-4.

Lukovic D, Stojkovic M, Moreno-Manzano V, Bhattacharya

SS, Erceg S (2013). Perspectives and future directions of

human pluripotent stem cell-based therapies. Lessons from

Geron’s clinical trial for spinal cord injury.Stem Cells Dev

,[Epub ahead of print].

Magliocca JF, Held IKA, Odorico JS (2006). Undifferentiated

murine embryonic stem cells cannot induce portal tolerance

but may possess immune privilege secondary to reduced

major histocompatibility complex antigen expression. Stem

Cells Dev, 15, 707-17.

Mahmood A, Harkness L, Schrøder HD, Abdallah BM, Kassem

M (2010). Enhanced differentiation of human embryonic

stem cells to mesenchymal progenitors by inhibition of

TGF-beta/activin/nodal signaling using SB-431542. J Bone

 Miner Res, 25, 1216-33.

Matthaei KI (2009). Current developments in genetically

manipulated mice. In trends in stem cell biology and

technology, H. baharvand, ed. Humana Press, New York,pp 123-36.

Meyer S, Nolte J, Opitz L, Salinas-Riester G, Engel W (2010).

Pluripotent embryonic stem cells and multipotent adult

germline stem cells reveal similar transcriptomes including

pluripotency-related genes. Mol Hum Reprod , 16, 846-55.

Nussbaum J, Minami E, Laflamme MA, et al (2007).

Transplantation of undifferentiated murine embryonic stem

cells in the heart: teratoma formation and immune response.

FASEB J , 21, 1345-57.

Oosterhuis JW, Looijenga LHJ (2005). Testicular germ-cell

tumours in a broader perspective. Nat Rev Cancer, 5, 210-22.

Pal R, Mamidi MK, Das AK, Rao M, Bhonde R (2013).Development of a multiplex PCR assay for characterization

of embryonic stem cells. Methods Mol Biol, 1006, 147-66.

Press JZ, Kenyon JA, Xue H, et al (2008). Xenografts of primary

human gynecological tumors grown under the renal capsule

of NOD/SCID mice show genetic stability during serial

transplantation and respond to cytotoxic chemotherapy.

Gynecol Oncol, 110, 256-64.

Prokhorova TA, Harkness LM, Frandsen U, et al (2009).

Teratoma formation by human embryonic stem cells is site

dependent and enhanced by the presence of Matrigel. Stem

Cells Dev, 18, 47-54.

Przyborski SA (2005). Differentiation of human embryonic stem

cells after transplantation in immune-decient mice. Stem

Cells, 23, 1242-50.Riess P, Molcanyi M, Bentz K, et al (2007). Embryonic stem

cell transplantation after experimental traumatic brain injury

dramatically improves neurological outcome, but may cause

tumors. J Neurotrauma, 24, 216-25.

Sadraei H, Abtahi SR, Nematollahi M, et al (2009). Assessment

of potassium current in Royan B(1) stem cell derived

cardiomyocytes by patch-clamp technique. Res Pharm Sci,

4, 85-97.

Stojkovic P, Lako M, Stewart R, et al (2005). An autogeneic

feeder cell system that efficiently supports growth of

undifferentiated human embryonic stem cells. Stem Cells,

23, 306-14.

Swijnenburg R-J, Tanaka M, Vogel H, et al (2005). Embryonic

stem cell immunogenicity increases upon differentiation

after transplantation into ischemic myocardium. Circulation,

112, 166-72.

Szabó P, Mann JR (1994). Expression and methylation of

imprinted genes during in vitro differentiation of mouse

parthenogenetic and androgenetic embryonic stem cell lines.

 Development , 120, 1651-60.

Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al (1998).

Embryonic stem cell lines derived from human blastocysts.

Science, 282, 1145-7.

Wakitani S, Takaoka K, Hattori T, et al (2003). Embryonic stem

cells injected into the mouse knee joint form teratomas and

subsequently destroy the joint.  Rheumatology (Oxford),

42, 162-5.Wong DJ, Liu H, Ridky TW, et al (2008). Module map of stem

cell genes guides creation of epithelial cancer stem cells.

Cell Stem Cell, 2, 333-44.

Xie X, Cao F, Sheikh AY, et al (2007). Genetic modication of

embryonic stem cells with VEGF enhances cell survival and

improves cardiac function. Cloning Stem Cells, 9, 549-63.

Yang T, Riehl J, Esteve E, et al (2006). Pharmacologic and

functional characterization of malignant hyperthermia in

the R163C RyR1 knock-in mouse.  Anesthesiology, 105,

1164-75.

Zhang WY, de Almeida PE, Wu JC (2012). Teratoma formation:

A tool for monitoring pluripotency in stem cell research.

In ‘StemBook’, (Cambridge (MA): Harvard Stem Cell

Institute).