You are on page 1of 10

Tumor Biol.

DOI 10.1007/s13277-016-5214-8

ORIGINAL ARTICLE

Primary cultures of human colon cancer as a model to study


cancer stem cells
Sergey Koshkin 1 & Anna Danilova 2 & Grigory Raskin 3 & Nikolai Petrov 1 &
Olga Bajenova 4,5 & Stephen J. O’Brien 4 & Alexey Tomilin 1 & Elena Tolkunova 1

Received: 22 December 2015 / Accepted: 14 July 2016


# International Society of Oncology and BioMarkers (ISOBM) 2016

Abstract The principal cause of death in cancer in- Keywords Colon cancer . Cancer stem cells . Oct4 . CD133 .
volves tumor progression and metastasis. Since only a Drug resistance . 5-Fu
small proportion of the primary tumor cells, cancer stem
cells (CSCs), which are the most aggressive, have the
capacity to metastasize and display properties of stem Colorectal cancer is one of the leading causes of morbidity
cells, it is imperative to characterize the gene expression and mortality worldwide [1]. The 5-year survival rate for pa-
of diagnostic markers and to evaluate the drug sensitiv- tients with local colorectal cancer is 80–90 %, whereas that
ity in the CSCs themselves. Here, we have examined with distant metastasis is only 10–20 % [2, 3]. Such unfavor-
the key genes that are involved in the progression of able outcome for this pathology is caused by frequent late
colorectal cancer and are expressed in cancer stem cells. diagnosis, treatment failure at advanced stages, and
Primary cultures of colorectal cancer cells from a pa- metastasis.
tient’s tumors were studied using the flow cytometry The current standard treatment for metastatic colorectal
and cytological methods. We have evaluated the clinical cancer includes FOLFOX (oxaliplatin plus infusional 5-
and stem cell marker expression in these cells, their fluorouracil (5-FU) and leucovorin) or FOLFIRI (5-FU,
resistance to 5-fluorouracil and irinotecan, and the abil- leucovorin, and irinotecan). Irinotecan is a camptothecin de-
ity of cells to form tumors in mice. The data shows the rivative that exerts cytotoxic effects on cancer cells by
role of stem cell marker Oct4 in the resistance of pri- inhibiting topoisomerase I, and FOLFIRI has been proven to
mary colorectal cancer tumor cells to 5-fluorouracil. be effective as a second-line treatment of patients with meta-
static colorectal cancer [4, 5].
Based on the concept of cancer stem cells (CSCs) [6], a
tumor represents a mixed-cell population that consists of
healthy and cancerous cells with varying degrees of differen-
* Sergey Koshkin tiation. The most differentiated cancer cells in the primary
Koshkin31@mail.ru; http://www.cytspb.rssi.ru tumor have the features of epithelial tissue from which the
tumor originated, enabling its identification. The least differ-
1
Institute of Cytology, Russian Academy of Sciences, Tikhoretsky entiated cells have the characteristics of stem cells that confer
ave. 4, St. Petersburg, Russia 194064 to the tumor the ability to de- and re-differentiate, metastasize,
2
N.N. Petrov Oncology Research Institute, St. Petersburg 197758, and acquire drug resistance. CSCs, according to publications,
Russia comprise about 0.1–3 % of the whole cell number in a tumor
3
Russian Research Centre for Radiology and Surgical Technologies, and differ from the remaining mass of differentiated cancer
St. Petersburg 197758, Russia cells of primary tumors in their abilities of assymetric division,
4
Theodosius Dobzhansky Center for Genome Bioinformatics, St. self-renewal and differentiation, elevated resistance to radio-
Petersburg State University, St. Petersburg 199034, Russia therapy and chemotherapy, and expression of some stem cell
5
Department of Genetics and Biotechnology, St. Petersburg State markers and telomere length [7–10]. CSCs have the ability to
University, St. Petersburg 199034, Russia reproduce the parental tumor in vivo. In fact, as with any other
Tumor Biol.

kind of tissue, a malignant tumor has a structure composed of with CK20, 90 % of the cells with CDX2, and membrane
genetically heterogenous cells in different state: pre-cancer cytoplasmic localization of beta-catenin in 90 % of cells; the
cells, primary cancer cells, differentiated cells, migrating cells, index of Ki-67 was 60 %. From the first tissue sample later on,
and CSC, which play a role in the development of the disease the BSC-1 cell line was selected. The tissue sample from
[11]. which we obtained the BSC-6 cell line was also characterized
It is well established that cancer stem-like cells are charac- by a high index of Ki-67 of 60 % and a high level of expres-
terized by expression of several stem cell markers [12, 13]. sion of CDX2. It was defined as a solid tumor in the wall of
The most common membrane markers used for CSC cell the colon.
sorting and analysis are CD133, CD44, Lgr5, and aldehyde The cell isolation from the primary tissue was carried out
dehydrogenase (ALDH1). CD133+ colorectal cancer cells on the basis of recommendations of Yu et al. [17]. Each sam-
were shown to form tumors in the immune-deficient mice. ple was cut into pieces of approximately 1 mm−3 in size and
However, the correlation between CD133 expression and the incubated in 10-fold (by volume) excess of trypsin (Gibco,
clinical-pathological factors in colorectal cancer remains USA) overnight in a refrigerator at 4 °C, followed by an hour
unclear. at 37 °C. The action of trypsin was inhibited by RPMI medi-
Lgr5 was first identified in human colon cancer as a Wnt um with 10 % FBS. This suspension of disintegrated cells was
target gene [14, 15]. It was shown to be overexpressed in other filtrated through a Falcon Cell strainer (100 μm) and centri-
human malignancies such as ovarian, hepatocellular, esopha- fuged to collect the cells (150 g for 5 min).
geal, and basal cell carcinomas [16]. Lgr5-expressing cells are The remaining Bundigested tissue^ was treated with a so-
responsible for the complete restoration of small and large lution of collagenase in RPMI medium with serum containing
intestine epithelium in vivo [17]. The available evidence sug- 500 units/ml of enzyme, incubated in a Petri dish at 37 °C for
gests that Lgr5 might represent a useful marker to identify and 1 h. The released cells had been collected after filtration by
target CSCs in colon cancer. centrifugation (150 g for 5 min) combined with cells previ-
Most methods rely on specific surface antigen recognition ously obtained after trypsin treatment and were placed in
and thus are restricted by the availability of highly specific DMEM medium (Gibco, USA) with 10 % FBS (Sigma,
antibodies. In addition, labeling of cell-surface markers by USA) for culturing in the incubator. The cells were passaged
antibodies could trigger signaling pathways and induce cell using trypsin solution (Gibco, USA), once for 3 days at a ratio
modification and differentiation. Therefore, the development of 1:3.
of methods that do not rely on marker labeling is vital.
In the recent years, many studies have demonstrated that
Cell culturing in order to obtain spheroids
aberrant expression of stem cell-associated nuclear transcrip-
tion factors, such as Oct4, Sox2, Nanog, and Klf4, can con-
To obtain cell spheroids, we cultured cells in plastic dishes
tribute to tumorigenesis in various cancers [18, 19].
with a low-adhesive surface and in the serum-free culture
The aim of this study is to investigate the significance of
medium of the following composition: DMEM medium with
Oct4 and CD133 in the development of colon cancer and
6 mg/ml glucose, 1 mg/ml sodium bicarbonate, 5 mM
delineate their CSC properties. We make an attempt to extract
HEPES, 2 mM L-glutamine, 4 mg/ml heparin, 4 mg/ml
a population of cancer stem cells from primary cell culture of
BSA, 10 ng/ml bFGF, 20 ng/ml EGF, 100 μg/ml
colorectal cancer. We have also assessed the role of Oct4 tran-
apotransferrin, 25 μg/ml insulin, 9.6 μg/ml putrescine,
scription factor in drug resistance properties of colorectal can-
30 nM anhydrous sodium selenite, and 20 nM final concen-
cer cells.
tration of progesterone in a concentration of 300,000 cells per
milliliter. Such culture conditions were chosen to preferential-
ly maintain immature (poorly differentiated) tumor cells,
Material and methods
which proliferate and produce cell aggregates called spher-
oids. The above medium composition was described by
Processing of primary samples
Todaro et al. [18].
The material for this study was obtained on the basis of pa-
tients’ informed consent from the LG Sokolov Clinical Packing of viral particles and infected cells in vitro
Hospital No. 122. The resulting fragments were placed in a with a virus
saline solution for 2 h and brought to the laboratory. This
study included six tumor samples. From six endoscopic tissue To produce the viral particles, we used human embryonic
samples, two cell lines were obtained: BSC-1 and BSC-6. kidney cell line HEK293T, which was cultured in DMEM,
Based on the immuno-histochemical studies, the first tissue containing 10 % fetal bovine serum (Sigma, USA). Cells were
sample was characterized by positive staining of 20 % of cells split every 3 days at a ratio of 1:4.
Tumor Biol.

To assemble the viral particles, 293T cells were transfected medium, put into PBS containing 0.5 mg/ml MTT, and incu-
with the lentivirus using the calcium phosphate method. This bated for 2 h at 37 °C under 5 % CO2. The resulting precip-
method was based on the integration of the lentiviral vector itates (crystals) of formazan were dissolved in DMSO (Sigma,
and helper plasmid for the assembly of viral particles USA). The absorbance was measured at 570 nm with the
(http://tcf.epfl.ch/page-6766-en.html). The medium with Multiskan EX spectrophotometer (Thermo Electron, USA).
transfected cells was collected every 48 h, and the virus
particles were concentrated by ultracentrifugation using a Analysis of drug resistance
standard technique (http://tcf.epfl.ch/page-6764.html).
To determine the sensitivity of CRC cells to the cytostatic
Lentivirus infection of cancer cells and selection drugs (5-FU and irinotecan), the cells were seeded in 96-
of resistant clones well plates at 50,000 per well in a volume of 100 μl. Each
experimental point was measured six times, and the standard
Cells were plated in 24-well plates 1 day before the infection error of the mean was calculated. The experiment was repeat-
at a density of 105 cells per well. The infection was performed ed three times with obtaining similar results. The final con-
by adding 10 μl of concentrated virus to 100 μl DMEM/10 % centration of 5-FU was 100 ng/ml, for IT 20 ng/ml. For each
FBS medium per well. After 2 days, cells were trypsinized and studied cell lines, we had six wells of control, without the
plated on a 10-cm culture dish (Falcon, USA). The selection cytostatic. After 48 h, 10 μl of MTT solution (5 mg/ml in
was carried out by changing the media next day per medium PBS buffer) was added to each well and the cells were incu-
with antibiotic puromycin (Sigma, USA) at a concentration of bated for 2 h. Then, PBS buffer was removed and formazan
30 μg/ml. We change the medium every 2 days during crystals were dissolved in 100 μl DMSO. Further measure-
14 days. Grown colonies were picked individually, ments were carried out with a Multiskan EX spectrophotom-
trypsinized, and plated on 3-cm plates. After a week in culture, eter (Thermo Electron, USA). Results were expressed as per-
the cells were frozen in the liquid nitrogen. centage of cells surviving after the drug treatment vs the con-
trol group (untreated cells).
Test on the formation of tumors in mice Histological and enzyme-linked immunosorbent staining
was performed by G.A. Raskin at The Department of
Cell cultures were trypsinized to obtain single cell suspension, Pathology of the Russian Scientific Center of Radiology and
and cells were washed three times with Hank’s solution. After Surgical Technologies, headed by Dr. K.M. Pozharissky. The
counting, the cell concentration was adjusted to 106 cells/ml. selection of CD133+ by specific antibodies was performed
The resulting suspension was injected into immuno-deficient according to the manufacturer’s CD133 MicroBead human
nude mice subcutaneously into the thigh region, 106 cells per Kit protocol (MACS Miltenyi Biotec, Germany).
injection. To prevent any leakage of fluid from the syringe, the
needle was introduced through the top of the thigh muscles
into the gaps between the skin and muscles of the lower side. Results
Each mouse has received two injections—to the right and left
thighs. Group of animals injected with original cells served as Primary tumors cell culture
a control standard to the experimental group, injected with
Oct4+ cells. Grown tumors were measured every 10 days Our work was carried out with the biopsies of six patients that
starting from the 20th day after the injection. Measurements were processed as described in the BMaterials and methods^
were made with a caliper by the maximum diameter up to section. From six biopsy specimens taken from different pa-
1 mm. We measured the greatest longitudional diameter, x, tients, we were able to select two cell lines. The resulting cell
and the greatest transverse diameter, y. The tumor volume lines, called BSC-1 and BSC-6, were further characterized by
was calculated from these two transcutaneous diameters (1/2 their ability to grow in the form of a monolayer and spheroids.
L × W2) [20]. To obtain the monolayer cultures, CRC cells, obtained
from the endoscopic material, were cultured in DMEM medi-
MTT viability test um with 10 % FBS as a monolayer. Through the passages, the
structure of the cell population changed (Fig. 1). The initial
Assessment of cell viability was performed by colorimetric cell suspension contained a large amount of red blood cells,
MTT assay (Sigma, USA). This method is based on the fact sometimes visible as Brouleaux.^ After 5–7 days in a culture,
that in the live cells mitochondrial oxidoreductase reduces small colonies of fibroblast-like cells appeared. As illustrated
yellow MTT to purple formazan. The amount of the devel- in Fig. 1, the cells initially had fibroblast-like morphology
oped formazan is correlated with the number of viable cells in (Fig. 1a). After 1 month in culture, the primary cells devel-
the population. The cells were spun down from the RPMI oped dense colonies of polygonal cells, similar in morphology
Tumor Biol.

Fig. 1 Primary colon cancer cell


cultures were grown as a
monolayer or spheroids on the
low-adhesive substrate. a Small
colonies appearing on the fifth
day in culture. b–d Colonies
developed after 1 month in culture
in a monolayer. e Spheroids are
formed when cells are plated on
the low-adhesive plastic in the
serum-free medium. f Spheroid
formed on agarose on the third
day in culture

to the embryonic stem cells surrounded by a small number of figure shows a positive staining by anti-CK-PAN antibodies
fibroblast-like cells (Fig. 1b–d). in 80 % of cells and by the anti-CDX2 antibody in 100 % of
These cells were able to grow in size in a serum-free me- the cells, which is similar to the staining of original cancer
dium supplemented with growth factors and to form spheroids biopsy.
on a low-adhesive plastic (Fig. 1e). When agarose was used as
a low-adhesive substrate, the primary cells formed spheroids Enrichment of endogenous Oct4 expression by infection
(Fig. 1f). Aggregates of five to eight cells could be seen, fol- with the lentiviral construct
lowing the seeding on low-adhesive plastic, suggesting that
these spheres are not the cell clones but cell aggregates. We We have recently developed a bicistronic DNA construct
noticed that tumor cells have high adhesion to the plastic. If 2A2B-TKiresPur which features the thymidine kinase (TK)
the serum-free medium was not changed every day, the and puromycin resistance (Puror) genes under the control of
spheres will have started attaching to the plastic and spread- the 2A2B enhancer from the pluripotency-associated gene
ing, looking similar to the cells in the monolayer culture. On a Oct4 [21]. When stably integrated into the pluripotent cell
highly adhesive plastic in a serum-free medium, the colonies genome, the construct allows highly selective enrichment
could detach, and they often started growing as spheres. (via Puror gene) or elimination (via TK gene) of Oct4+ undif-
Next, we compared the expression of clinical CRC markers ferentiated pluripotent cells from the mixed populations, fol-
in the BSC-6 cell line and in the original primary tumor tissue lowing the exposure to puromycin or ganciclovir (GCV), re-
using specific staining with the antibodies to PAN-cytokeratin spectively [21]. In the current study, we used the 2A2B-
and receptor CDX2 (Fig. 2). The data show that after ten TKiresPuro construct embedded into the lentiviral vector
passages in culture, the BSC-6 cell line retains the expression (Fig. 2b). It was suggested that the same strategy would allow
of PAN-cytokeratin and CDX2 (Fig. 2b, d, f), which corre- to positively select CSC cells that, according to our working
sponds to the staining of initial material—a moderately differ- hypothesis [22], express Oct4. Transfected colorectal cancer
entiated adenocarcinoma of the colon (Fig. 2a, c, e). The cells were selected in DMEM medium with 10 % FBS,
Tumor Biol.

Fig. 2 Original colon cancer


tumor (a, c, e) and derived thereof
cell line BSC-6 (b, d, f) were
stained with hematoxylin-eosin
(a, b) or with the antibodies to the
clinical markers CK-20 (c, d) or
CDX2 (e, f), followed by
peroxidase-conjugated secondary
antibodies. The figure shows a
positive staining by anti-CK-PAN
antibodies in 80 % of cells and by
anti-CDX2 antibody in 100 % of
the cells, which is similar to the
staining of original cancer biopsy

containing 2 μg/ml puromycin. Resistant cell lines were called (Oct4+) and BSC-6R (Oct4+) and of parental cell lines are
BSC-1R (Oct4+) and BSC-6R (Oct4+). To keep the cells pu- similar.
romycin resistant, we cultured them in media, containing pu- To study the nature of the tumors, they were restrained in
romycin. After five passages, cells continue to express Oct4, polyformaldehyde and stained with anti-CK-20, beta-catenin,
which was checked by RT-PCR with Oct4-specific primers CDX2, and ALDH1 antibodies (Fig. 4). The data shows that
(data not shown). As a matter of controlling lentiviral trans- after prolonged passaging, and regardless of culturing
duction efficiency, we used lentivirus-expressing green fluo- methods, the established CRC cell lines preserve the ability
rescent protein (EGFP). The data shows the high efficiency of
infection—60–70 % of cells (Fig. 3a, b).

In vivo mouse experiments

In our next step, we evaluated the ability of CRC cells, cul-


tured in monolayer or as spheroids, to give rise to tumors in
immune-deficient nude mice (Table 1). In this step, after 10
passages, culture BSC-1 cells were injected subcutaneously
into mice; the presence and the size of the tumors were esti-
mated after 30 days. Tumor measurements were made with a
caliper by the maximum diameter. BSC-1 cells cultured in a Fig. 3 Primary colon cancer cells infected with the lentiviral construct.
Upper panel—a scheme of the suicidal lentiviral construct carrying the
monolayer and the same cells cultured as spheroids gave rise 2A2B-TKtkPur cassette. Lower panel—cell lines BSC-1 (a) and BSC-6
to tumors of a similar size (data not shown). Our data show (b) that are transfected with the virus expressing green fluorescent pro-
that tumorigenic potentials of puromycin-selected BSC-1R tein. The figure shows high efficiency of virus transduction
Tumor Biol.

Table 1 Size of tumors,


developed in vivo after injection Cell line Average volume, Percentage of animals that developed Numbers of injected
of colorectal cancer cells in mice mm3 tumors mice

BSC-1 388 ± 124 75 % 4


BSC-1R 407 ± 343 50 % 4
BSC-6 372 ± 308 57 % 7
BSC-6 150 ± 46 60 % 8
R

BSC-1 (four mice) and BSC-6 (seven mice) original cell lines served as a control standard for experimental
groups, injected with Puror (Oct4+) cells (four mice for BSC-1R and eight mice for BSC-6R)

to form tumors in nude mice (Fig. 4b, d, f, h) and are suitable and Lgr5 markers of 16 BSC-6R (Oct4+) clones confirmed
to study CSCs. The data revealed that the expression patterns that the levels of Oct4+ cells (98–100 %) and Sox2+ cells (98–
of these clinical markers are very similar to those in primary 100 %) were similar in all of them. For the Oct4+ cell clones,
human tumor tissues (compare Fig. 4a, c, e, g). which have also shown a high number of cells with enhanced
To find out if there is a genetic heterogeneity in puromycin- Sox2 expression, we observed a different expression of colo-
resistant cell cultures, we have characterized individual clones rectal cancer stem cell marker Lgr5. There were substantial
of the BSC-6R (Oct4+) cell line. The quantification by flow differences in the proportions of cells expressing the Lgr5
cytometry of the percentage of cells expressing Oct4, Sox2, marker in different clones (21 to 92 % of cells were Lgr5+).

Fig. 4 Immuno-histochemical
staining of initial colorectal
cancer tumor tissue (a, c, e, g),
and tumors developed in mice (b,
d, f, h) by BSC-1 cells. Samples
were stained with antibodies to
CK-20 (a, b); beta-catenin (c, d);
CDX2 (e, f); ALDH1 (g, h). The
data show the same pattern of
expression of colon cancer
markers in original tumor, and
developed in mice
Tumor Biol.

Not all cells, expressing Oct4, express also Lgr5. When cells
were regrown in culture, they were able to reconstitute the cell
heterogeneity of the starter culture.

Drug resistance of colorectal cancer cells to irinotecan


and 5-FU

5-FU is the Buniversal^ anti-cancer drug. It is included in the


therapeutic regimens received by more than half of all cancer
patients, although the response to the treatment with 5-FU
alone in patients is no more than 10–15 % [23]. Irinotecan
targets DNA topoisomerase I, causing the inhibition of DNA
Fig. 5 The differences in the resistance of colorectal cancer cell lines
replication and subsequent cell death—a standard treatment BSC-1 and BSC-6 to irinotecan (20 mkM) and 5-FU (100 mkM). Y-axis
regimen for metastatic colorectal cancer, especially in combi- shows the percentage of cells that survived treatment vs the control cells
nation with oxaliplatin [24]. It improves the response rate of (no drug treatment) measured by optical density in MTT assay. The data
patients with colorectal cancer in up to 40–50 %. The effects show that both lines are resistant to irinotecan treatment, but have differ-
ent sensitivity to 5-FU. BSC-1 is resistant; BSC-6 is sensitive
of these two anti-cancer drugs were tested on BSC-1 and
BSC-6 cell lines. The results show (Fig. 5) that BSC cells
Discussion
derived from different patients responded differently to 5-
FU. BSC-6 cells were sensitive to 5-FU treatment as more
Gastrointestinal cancers account for approximately 30 % of
than 50 % of cells could not survive, whereas BSC-1 cells
the total cancer patient population worldwide [26, 27]. 5-FU
were resistant to 5-FU treatment (Fig. 5).
with oxaliplatin for colorectal cancer has shown limited clin-
We choose the BSC-6 cell line to further assess how these
ical utility [28, 29], especially due to the high rate of tumor
cells, enriched for CSC marker Oct4 and CD133, will respond
recurrence. Emerging evidence suggests that the poor re-
to the indicated drugs. The stem cell marker enrichment has
sponse to the current treatment modalities for epithelial can-
been done in two ways. First, the cells were transfected with
cers is due to the aberrations in multiple signaling pathways
the lentivirus carrying the suicidal vector that allows to select
together with the presence of a small subpopulation of drug-
cells expressing endogenous Oct4, and second, by sorting
resistant CSCs that have the propensity to promote tumor
cells with the antibodies to the surface antigen CD133.
recurrence, invasion, and metastasis [30]. Although chemo-
The following enriched populations were analyzed (Fig. 6):
therapies target the majority of tumor cells, the CSCs in the
the parental cells BSC-6 cells, (BSC-6), the same cells after
tumor mass are nonresponsive, resulting in tumor recurrence
MACS for CD133 (CD133+) and the flow-through (CD133
[31].
−), BSC-6R cells selected on puromycin (Oct4+), and the
same cells after MACS for CD133 (Oct4+CD133+) and the
flow-through (Oct4+CD133−). The data shows that these cell
populations express similar response to irinotecan treatment
(Fig. 7a). In contrast, sensitivity of selected cell populations to
5-FU is different. As is shown in Fig. 7b, the cell population
enriched in endogenous expression of Oct4 (Oct+) has 33 %
higher resistance to 5-FU than the original cell line. CD133+
cells, otherwise, did not obtain a high resistance compared to
CD133− cells. CD133− cells are more resistant to 5-FU than
CD133+ cells. According to our results, CD133+ cells are
twice more sensitive to 5-FU than CD133− cells. That is cor-
rect for both original cell lines and for Oct4+ enriched cells.
The fact that the cell population enriched by the endoge-
Fig. 6 Scheme of BSC6 cell enrichment by CSC markers Oct4 and
nous expression of nuclear factor Oct4 has significantly great- CD133. Cells were infected with reporter construct and selected on
er resistance to cytostatic drugs is consistent with the obser- puromycin. Starting cell line (BSC-6) and Puror (Oct4+) cells were sorted
vations that CSCs have higher resistance to modern by MACS Technology (Miltenyi Biotec). CD133+ and CD133− cells
chemotherapeutic agents [25]. The lack of resistance to were separated. As a result of the two-step enrichment technique, we
received six cell lines: starting cell line (BSC-6), Oct4+-enriched line
fluorouracil of CD133+ cells makes the possibility of using and CD133+ and CD133− lines from starting line (BSC-6 CD133+,
CD133 as a marker for CSC population in colorectal cancer BSC-6 CD133−) and from Oct4+-enriched line (BSC-6 Oct4+CD133+,
questionable. BSC-6 Oct4+CD133−)
Tumor Biol.

networks supporting their resistant behavior are poorly under-


stood. Identification of a specific marker of CSCs could have a
significant impact on the design of effective therapies against
drug-resistant cancers [35]. For the first time, the new method
of enrichment of the endogenous Oct4 gene expression by
infection with lentiviral construct was developed. This suicid-
al vector allows not only to enhance the proportion of cells
with endogenous expression of Oct4 protein in the CSC pop-
ulation on puromycin but also to select the population of Oct4-
expressing cells using another antibiotic, ganciclovir.
Cancer stem cells possess a stronger tumorigenetic activity
and resistance to radiotherapy and chemotherapy [36]. CSCs
may be responsible for the treatment failure in multiple tu-
mors, because they are more resistant to chemotherapy and
radiotherapy than other tumor cells [37–39]. However, any
correlation of chemotherapeutic drug resistance and expres-
sion of Oct4 in colorectal cancer remains unclear. The fact that
in our experiments the cell population enriched in endogenous
Oct4 expression had significantly greater resistance to 5-FU is
consistent with the fact that CSCs can survive chemotherapy
[40].
As recently shown, transcription factor Oct4 is involved in
cancer development and causes epithelial dysplasia of somatic
cells [25]. The presence of Sox2- and Oct4-positive cells in
Fig. 7 The resistance of BSC-6 cells, enriched by Oct4 and CD133
markers to irinotecan (a) and 5-FU (b). Y-axis shows the percentage of endometrial adenocarcinomas was recently confirmed by
cells that survived the treatment vs control cells (no drug treatment) Pitynski et al. [41]. High levels of nuclear expression of both
measured by optical density in MTT assay. (*P < 0.05, Mann-Whitney Sox2 and Oct4 have been observed in cervical cancer, small
U test). The data show that all cell lines are resistant to irinotecan treat- cell lung cancer, and squamous cell esophageal cancer
ment. Oct4+ has higher resistance to 5-FU than the original cell line.
CD133+ cells are twice more sensitive to 5-FU than CD133− cells. [42–44]. In patients with squamous cell esophageal cancer,
That is correct for both original cell lines and for Oct4+-enriched cells both Sox2 and Oct4 overexpression were related to signifi-
cantly shorter overall survival rate compared to patients with
negative stained tumors. There is a higher risk of distant re-
A number of studies have highlighted the detection of low- currence in rectal cancer if Sox2 and Oct4 are overexpressed
level expression of Oct4 in tumor cells and its enrichment in a in the primary tumor [45]. Also, Oct4 expression has been
subpopulation of CSCs. Based on such information, Oct4 in implicated in the malignancy and prognosis of glioblastoma
CSCs act via mechanisms different from those in pluripotent [46]. Osteosarcoma also contained stem-like cells involved in
stem cells [32]. To investigate the significance of Oct4 expres- tumor development that express Oct4 and Nanog stem cell
sion in acquiring cancer stem-like properties, we established markers.
the cell lines from primary tumors in monolayer forms that As was shown by Kashihara et al., CD133 expression is
have maintained the stem cell markers of the primary tumors. correlated with poor prognosis in colorectal cancer. [47].
Low level of Oct4 protein in somatic tumors makes it difficult Vincent et al. [48] claim that CD133 is the only reliable mark-
to detect it by Western hybridization and to enrich the CSC er for CSC characterization in the Colo205 CRC cell line. It
subpulation by sorting [33, 34]. We have found that the level also has been shown that Wnt pathway activity could be re-
of Oct4 expression in primary cultures of CRC is sufficient to sponsible for the chemoresistance of CD133+ cells in colorec-
enrich this subpopulation by integration of the 2A2B- tal cancer [49]. Deng et al. [50] demonstrated that 5-FU can
containing construct. Our study has indicated that primary upregulate Wnt activity in CD133+ colon cancer stem-like
colorectal cancer cells retain their tumorigenic potential after cells. Moreover, if Dickkopf-1 is overexpressed in CRC cells,
being cultured, regardless of the culturing method (monolayer then the expression of CD133 and Lgr5 markers will be also
or spheroids). The results of this research indicate that obtain- decreased in CRC cells. It also reduced the proliferation, mi-
ed cell lines can be used as in vitro model to study colorectal gration, and invasion of CRC cells [51].
cancer stem-like cells. In our experiments, to identify colorectal cancer stem cells,
Populations of immature stem-like cells were successfully we are using early passages of newly established colorectal
isolated from primary cancer tissues, but the molecular cancer cell lines from clinical specimens. Based on the recent
Tumor Biol.

data, the sorting by surface markers (CD133, CD44) did not 3. Ishida H, Fujita K, Akiyama Y, Sunakawa Y, Yamashita K, Mizuno
K, et al. Regimen selection for first-line FOLFIRI and FOLFOX
reliably enrich for stemness when the CRC cell lines
based on UGT1A1 genotype and physical background is feasible in
established a long time ago were used [52]. The authors sug- Japanese patients with advanced colorectal cancer. Jpn J Clin
gest to use the newly established cell lines and cells from Oncol. 2011;41:617–23.
clinical specimens to identify the stem cell-enriched popula- 4. Sanli UA, Karabulut B, Uslu R, Korkut M, Goker E. Single-agent
tion. We expect, via CD133 sorting, to obtain chemoresistant irinotecan for recurrent/metastatic colorectal cancer: a retrospective
analysis. Med Princ Pract. 2006;15:288–92.
tumor stem cell enrichment. Contrary to expectations, there 5. Valent P, Bonnet D, De Maria R, Lapidot T, Copland M, Melo JV,
was no increase in the resistance to 5-FU in the CD133+ et al. Cancer stem cell definitions and terminology: the devil is in
subpopulation of cancer cells, which makes the possibility of the details. Nat Rev Cancer. 2012;12:767–75.
using CD133 as a marker for cancer stem-like cell population 6. Lapidot T, Sirard C, Vormoor J, Murdoch B, Hoang T, Caceres-
Cortes J, et al. A cell initiating human acute myeloid leukaemia
from clinical colon cancer specimens questionable. after transplantation into SCID mice. Nature. 1994;367:645–8.
Concerning the use of Oct4 as such a marker, our data show 7. Ricci-Vitiani L, Fabrizi E, Palio E, De Maria R. Colon cancer stem
that the expression of the Oct4 transcription factor can be used cells. J Mol Med (Berl). 2009;87:1097–104.
as a marker of colorectal cancer and a potential target for 8. Fabian A, Barok M, Vereb G, Szollosi J. Die hard: are cancer stem
cancer therapy. cells the Bruce Willises of tumor biology? Cytometry A. 2009;75:
67–74.
In this study, we demonstrated differences in the levels of 9. Vaiopoulos AG, Kostakis ID, Koutsilieris M, Papavassiliou AG.
Lgr5 expression in cell clones with Oct4 expression. Since Colorectal cancer stem cells. Stem Cells. 2012;30:363–71.
Lgr5 plays an important role not only in the early but also in 10. Mathonnet M, Perraud A, Christou N, Akil H, Melin C, Denizot Y,
the late event of tumorigenesis, we consider that the reason for et al. Hallmarks in colorectal cancer: angiogenesis and cancer stem-
like cells. World J Gastroenterol. 2014;20:4189–96.
this different proportion of positive cells in the final Oct4-
11. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer
positive clones may reflect the original heterogeneity of tumor cell capable of initiating tumour growth in immunodeficient mice.
cell culture, which was used for CSC enrichment. Lgr5 is a Nature. 2007;445:106–10.
marker of normal intestinal stem cells and also could have an 12. Van de Wetering M, Sancho E, Verweij C, de Lau W, Oving I,
important role in cancer cell stemness [53, 54]. We think that Hurlstone A, van der Horn K, Clevers H. The beta-catenin/TCF-4
complex imposes a crypt progenitor phenotype on colorectal cancer
in vivo experiments will help us to validate our speculation, cells. Cell. 2002;111:241–50.
which states that the level of Lgr5 can be important for the 13. XS W, Xi HQ, Chen L. Lgr5 is a potential marker of colorectal
tumor development in nude mice and may be used as addi- carcinoma stem cells that correlates with patient survival. World J
tional marker of CSCs. Surg Oncol. 2012;10:244.14.
14. Ritsma L, Ellenbroek SI, Zomer A, Snippert HJ, de Sauvage FJ, van
Altogether, this approach can be used in developing new Rheenen J, et al. Intestinal crypt homeostasis revealed at single-
therapeutic strategies aimed at eradicating the tumorigenic stem-cell level by in vivo live imaging. Nature. 2014;507:362–5.
subpopulation of CSCs within colorectal cancer tumors. Our 15. Gargett CE, Schwab KE, Zillwood RM, Nguyen HP, Wu D.
results provide the rationale that new therapeutic methods of Isolation and culture of epithelial progenitors and mesenchymal
stem cells from human endometrium. Biol Reprod. 2009;80:
targeting of Oct4-positive cancer cells should be developed to
1136–45.
gain the maximal clinical benefits. 16. Ebben JD, Treisman DM, Zorniak M, Kutty RG, Clark PA, Kuo JS.
The cancer stem cell paradigm: a new understanding of tumor de-
velopment and treatment. Expert Opin Ther Targets. 2010;14:621–
Compliance with ethical standards 32.
17. CS Y, Huang AC, Lai KC, Huang YP, Lin MW, Chung JG, et al.
Conflicts of interest None Diallyltrisulfide induces apoptosis in human primary colorectal
cancer cells. Oncol Rep. 2012;28:94954.
Grant support This work was financially supported by the Russian 18. Todaro M, Orlando V, Cicero G, Caccamo N, Meraviglia S, Dieli F,
Science Foundation (project 14-50-00068) to the Molecular Stem Cell et al. Chemotherapy sensitizes colon cancer initiating cells to
Biology Laboratory, Institute of Cytology RAS, and by the Government Vy9V52 T cell-mediated cytotoxicity. PLoS One. 2013;8.
of the Russian Federation mega grant 11G34.31.0068 to Dr. S. J. O’Brien. 19. Khan IN, Al-Karim S, Bora RS, Chaudhary AG, Saini KS. Cancer
stem cells: a challenging paradigm for designing targeted drug ther-
apies. Drug Discov Today. 2015;20(10):1205–16.
20. Tomayko MM, Reynolds CP, Euhus DM, Hudd C, LaRegina MC,
Johnson FE. Determination of subcutaneous tumor size in athymic
(nude) mice. Cancer Chemother Pharmacol. 1989;24(3):148–54.
21. Liskovykh MA, Chuikin IA, Ranjan A, Safina DA, Tolkunova EN,
References Tomilin AN, et al. Generation of rat induced pluripotent stem cells:
the analysis of reprogramming and culturing media. Tsitologiia.
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer 2011;53:939–45.
J Clin. 66(1):7–30. 22. Davydov-Sinitsyn AP, Bazhenova OV, Liskovykh MA,
2. Chang GJ, CY H, Eng C, Skibber JM, Rodriguez-Bigas MA. Ponomartsev CV, Tomilin AN, Tolkunova EN, et al. In vitro der-
Practical application of a calculator for conditional survival in colon ivation and characterization of a colorectal cancer stem cell sub-
cancer. J Clin Oncol. 2009;27:5938–43. population. Tsitologiia. 2013;55:318–23.
Tumor Biol.

23. Wolmark N, Rockette H, Fisher B, Wickerham DL, Redmond C, 40. Pointer KB, Clark PA, Zorniak M, Alrfaei BM, Kuo JS.
Petrelli NJ, et al. The benefit of leucovorin-modulated fluorouracil Glioblastoma cancer stem cells: biomarker and therapeutic ad-
as postoperative adjuvant therapy for primary colon cancer: results vances. Neurochem Int. 2014;71:1–7.
from National Surgical Adjuvant Breast and Bowel Project protocol 41. Pitynski K, Banas T, Pietrus M, Milian-Ciesielska K, Ludwin A,
C-03. J ClinOncol. 1993;11:187987. Okon K. SOX-2, but not Oct4, is highly expressed in early-stage
24. Cunningham D, Pyrhonen S, James RD, Punt CJ, Hickish TF, endometrial adenocarcinoma and is related to tumour grading. Int J
Herait P, et al. Randomised trial of irinotecan plus supportive care Clin Exp Pathol. 2015;8:8189–98.
versus supportive care alone after fluorouracil failure for patients 42. Ji J, Wei X, Wang Y. Embryonic stem cell markers Sox-2 and OCT4
with metastatic colorectal cancer. Lancet. 1998;352:1413–8. expression and their correlation with WNT signal pathway in cer-
25. Hochedlinger K, Yamada Y, Beard C, Jaenisch R. Ectopic expres- vical squamous cell carcinoma. Int J Clin Exp Pathol. 2014;7:2470–
sion of Oct-4 blocks progenitor-cell differentiation and causes dys- 86.
plasia in epithelial tissues. Cell. 2005;121:465–77. 43. Yang F, Gao Y, Geng J, Qu D, Han Q, Chen G, et al. Elevated
26. DeSantis CE, Lin CC, Mariotto AB, Siegel RL, Stein KD, Jemal A. expression of SOX2 and FGFR1 in correlation with poor prognosis
Cancer treatment and survivorship statistics, 2014. CA Cancer J in patients with small cell lung cancer. Int J Clin Exp Pathol.
Clin. 2014;64:252–71. 2013;6:2846–54.
27. Siegel R, DeSantis C, Jemal A. Colorectal cancer statistics, 2014. 44. Wang Q, He W, Lu C, Wang Z, Wang J, Suo Z, et al. Oct3/4 and
CA Cancer J Clin. 2014;64:104–17. Sox2 are significantly associated with an unfavorable clinical out-
28. Gungor C, Hofmann BT, Wolters-Eisfeld G, Bockhorn M. come in human esophageal squamous cell carcinoma. Anticancer
Pancreatic cancer. Br J Pharmacol. 2014;171:849–58. Res. 2009;29:1233–41.
29. Kaddis N, Saif MW. Second-line treatment for pancreatic cancer. 45. Saigusa S, Tanaka K, Toiyama Y, Yokoe T, Okugawa, Kusunoki M,
JOP. 2014;15:344347. et al. Correlation of CD133, OCT4, and SOX2 in rectal cancer and
30. Tanase CP, Neagu AI, Necula LG, Mambet C, Enciu AM, their association with distant recurrence after chemoradiotherapy.
Albulescu R, et al. Cancer stem cells: involvement in pancreatic Ann Surg Oncol. 2009;16:3488–98.
cancer pathogenesis and perspectives on cancer therapeutics. World 46. Hosokawa Y, Takahashi H, Inoue A, Kawabe Y, Funahashi Y,
J Gastroenterol. 2014;20:10790–801. Tanaka J, et al. Oct-3/4 modulates the drug- resistant phenotype
31. Muqbil I, Bao GW, El-Kharraj R, Shah M, Mohammad RM, Azmi of glioblastoma cells through expression of ATP binding cassette
AS et al. Systems and network pharmacology approaches to cancer transporter G2. Biochim Biophys Acta. 2015;1850:1197–205.
stem cells research and therapy. J Stem Cell Res Ther. 2012;suppl 7.
47. Kashihara H, Shimada M, Kurita N, Iwata T, Sato H, Matsumoto N,
32. Wang YJ, Herlyn M. The emerging roles of Oct4 in tumor-initiating
et al. CD133 expression is correlated with poor prognosis in colo-
cells. Am J Phys Cell Phys. 2015;309(11):C709–18.
rectal cancer. Hepato-Gastroenterology. 2014;61:1563–7.
33. Poursani EM, Mohammad Soltani B, Mowla SJ. Differential ex-
48. Vicente-Duenas C, Gutierrez de Diego J, Rodriguez FD, Jimenez R,
pression of OCT4 pseudogenes in pluripotent and tumor cell lines.
Cell J. 2016;18(1):28–36. Cobaleda C. The role of cellular plasticity in cancer development.
Curr Med Chem. 2009;16:3676–85.
34. Kobayashi I, Takahashi F, Nurwidya F, Nara T, Hashimoto M,
Takahashi K. Oct4 plays a crucial role in the maintenance of 49. Ren F, Sheng WQ, Du X. CD133: a cancer stem cells marker, is
gefitinib-resistant lung cancer stem cells. Biochem Biophys Res used in colorectal cancers. World J Gastroenterol. 2013;19:2603–
Commun. 2016;473(1):125–32. 11.
35. Fazlul H. Sarkar. Novel holistic approaches for overcoming therapy 50. Deng YH, XX P, Huang MJ, Xiao J, Zhou JM, Lin EH, et al. 5-
resistance in pancreatic and colon cancers. Med Princ Pract 2015. Fluorouracil upregulates the activity of Wnt signaling pathway in
36. Meacham CE, Morrison SJ. Tumour heterogeneity and cancer cell CD133-positive colon cancer stemlike cells. Chin J Cancer.
plasticity. Nature. 2013;501:328–37. 2010;29:810–5.
37. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeu-len L, 51. Qi L, Sun B, Liu Z, Li H, Gao J, Leng X. Dickkopf-1 inhibits
Stassi G. Colon cancer stem cells dictate tumor growth and resist epithelial-mesenchymal transition of colon cancer cells and contrib-
cell death by production of interleukin-4. Cell Stem Cell. 2007;1: utes to colon cancer suppression. Cancer Sci. 2012;103:828–35.
389–402. 52. Fan F, Bellister S, Lu J, Ye X, Boulbes DR, Ellis LM, et al. The
38. Dallas NA, Xia L, Fan F, Gray MJ, Gaur P, Ellis LM, et al. requirement for freshly isolated human colorectal cancer (CRC)
Chemoresistant colorectal cancer cells, the cancer stem cell pheno- cells in isolating CRC stem cells. Br J Cancer. 2015;112:539–46.
type, and increased sensitivity to insulin-like growth factor-I recep- 53. Barker N, Clevers H. Leucine-rich repeat-containing G-protein-
tor inhibition. Cancer Res. 2009;69:1951–7. coupled receptors as markers of adult stem cells.
39. Dieter SM, Ball CR, Hoffmann CM, Nowrouzi A, Herbst F, Glimm Gastroenterology. 2010;138:1681–96.
H, et al. Distinct types of tumor-initiating cells form human colon 54. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal
cancer tumors and metastases. Cell Stem Cell. 2011;9:357–65. stem cells. Nat Genet. 2008;40:915–20.

You might also like