Immortalization and malignant transformation of eucaryotic cells
This review is aimed at understanding the mechanisms of cell immortalization by different «immortalizing agents», oncogene-induced cell transformation of immortalized cells and moderate response of the advanced tumors to anticancer therapy in the light of tumor «oncogene and chromosome addiction», i...
Gespeichert in:
Datum: | 2012 |
---|---|
Hauptverfasser: | , |
Format: | Artikel |
Sprache: | English |
Veröffentlicht: |
Інститут клітинної біології та генетичної інженерії НАН України
2012
|
Schriftenreihe: | Цитология и генетика |
Schlagworte: | |
Online Zugang: | http://dspace.nbuv.gov.ua/handle/123456789/126463 |
Tags: |
Tag hinzufügen
Keine Tags, Fügen Sie den ersten Tag hinzu!
|
Назва журналу: | Digital Library of Periodicals of National Academy of Sciences of Ukraine |
Zitieren: | Immortalization and malignant transformation of eucaryotic cells / O.A. Stepanenko, V.M. Kavsan // Цитология и генетика. — 2012. — Т. 46, № 2. — С. 36-75. — Бібліогр.: 413 назв. — англ. |
Institution
Digital Library of Periodicals of National Academy of Sciences of Ukraineid |
irk-123456789-126463 |
---|---|
record_format |
dspace |
spelling |
irk-123456789-1264632017-11-25T03:02:58Z Immortalization and malignant transformation of eucaryotic cells Stepanenko, O.A. Kavsan, V.M. Обзорные статьи This review is aimed at understanding the mechanisms of cell immortalization by different «immortalizing agents», oncogene-induced cell transformation of immortalized cells and moderate response of the advanced tumors to anticancer therapy in the light of tumor «oncogene and chromosome addiction», intra-/intertumor heterogeneity, and chromosome instability. Целью настоящего обзора является понять механизмы клеточной иммортализации различными «иммортализующими агентами» ,онкоген-индуцируемой клеточной трансформации иммортализированных клеток и умеренный ответ на терапию из-за «склонности» опухоли к приобретению многочисленных генных и хромосомных изменений, внутри- и межопухолевой гетерогенности. Мета даного огляду зрозуміти механізми клітинної іморталізації різними «іморталізуючими агентами» ,онкогеніндукованої клітинної трансформації іморталізованих клітин і помірну відповідь на терапію через «схильність» пухлини до придбання численних генних та хромосомних змін та гетерогенністю усередині і між пухлинами. 2012 Article Immortalization and malignant transformation of eucaryotic cells / O.A. Stepanenko, V.M. Kavsan // Цитология и генетика. — 2012. — Т. 46, № 2. — С. 36-75. — Бібліогр.: 413 назв. — англ. 0564-3783 http://dspace.nbuv.gov.ua/handle/123456789/126463 576.385.5 en Цитология и генетика Інститут клітинної біології та генетичної інженерії НАН України |
institution |
Digital Library of Periodicals of National Academy of Sciences of Ukraine |
collection |
DSpace DC |
language |
English |
topic |
Обзорные статьи Обзорные статьи |
spellingShingle |
Обзорные статьи Обзорные статьи Stepanenko, O.A. Kavsan, V.M. Immortalization and malignant transformation of eucaryotic cells Цитология и генетика |
description |
This review is aimed at understanding the mechanisms of cell immortalization by different «immortalizing agents», oncogene-induced cell transformation of immortalized cells and moderate response of the advanced tumors to anticancer therapy in the light of tumor «oncogene and chromosome addiction», intra-/intertumor heterogeneity, and chromosome instability. |
format |
Article |
author |
Stepanenko, O.A. Kavsan, V.M. |
author_facet |
Stepanenko, O.A. Kavsan, V.M. |
author_sort |
Stepanenko, O.A. |
title |
Immortalization and malignant transformation of eucaryotic cells |
title_short |
Immortalization and malignant transformation of eucaryotic cells |
title_full |
Immortalization and malignant transformation of eucaryotic cells |
title_fullStr |
Immortalization and malignant transformation of eucaryotic cells |
title_full_unstemmed |
Immortalization and malignant transformation of eucaryotic cells |
title_sort |
immortalization and malignant transformation of eucaryotic cells |
publisher |
Інститут клітинної біології та генетичної інженерії НАН України |
publishDate |
2012 |
topic_facet |
Обзорные статьи |
url |
http://dspace.nbuv.gov.ua/handle/123456789/126463 |
citation_txt |
Immortalization and malignant transformation of eucaryotic cells / O.A. Stepanenko, V.M. Kavsan // Цитология и генетика. — 2012. — Т. 46, № 2. — С. 36-75. — Бібліогр.: 413 назв. — англ. |
series |
Цитология и генетика |
work_keys_str_mv |
AT stepanenkooa immortalizationandmalignanttransformationofeucaryoticcells AT kavsanvm immortalizationandmalignanttransformationofeucaryoticcells |
first_indexed |
2025-07-09T05:04:25Z |
last_indexed |
2025-07-09T05:04:25Z |
_version_ |
1837144440821514240 |
fulltext |
36 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
© O.A. STEPANENKO, V.M. KAVSAN, 2012
UDK 576.385.5
A.A. STEPANENKO, V.M. KAVSAN
Institute of Molecular Biology and Genetics, NAS of Ukraine, Kyiv
E-mail: kavsan@imbg.org.ua
IMMORTALIZATION
AND MALIGNANT TRANSFORMATION
OF EUKARYOTIC CELLS
The process of cellular transformation has been amply
studied in vitro using immortalized cell lines. Immortal-
ized cells never have the normal diploid karyotype, nev-
ertheless, they cannot grow over one another in cell cul-
ture (contact inhibition), do not form colonies in soft agar
(anchorage-dependent growth) and do not form tumors
when injected into immunodeficient rodents. All these
characteristics can be obtained with additional chromo-
some changes. Multiple genetic rearrangements, includ-
ing whole chromosome and gene copy number gains and
losses, chromosome translocations, gene mutations are
necessary for establishing the malignant cell phenotype.
Most of the experiments detecting transforming ability of
genes overexpressed and/or mutated in tumors (onco-
genes) were performed using mouse embryonic fibroblasts
(MEFs), NIH3T3 mouse fibroblast cell line, human
embryonic kidney 293 cell line (HEK293), and human
mammary epithelial cell lines (mainly HMECs and MC-
F10A). These cell lines have abnormal karyotypes and
are prone to progress to malignantly transformed cells.
This review is aimed at understanding the mechanisms of
cell immortalization by different «immortalizing agents»,
oncogene-induced cell transformation of immortalized
cells and moderate response of the advanced tumors to
anticancer therapy in the light of tumor «oncogene and
chromosome addiction», intra-/intertumor heterogeneity,
and chromosome instability.
Introduction. Malignant transformation is the
process by which cells acquire the properties of
cancer. The first successful malignant transfor-
mation in vitro was achieved with the polyoma
virus on Syrian hamster embryo cells, followed
by transformation with chemical carcinogens in
the mid-1960th (reviewed in [1]). Reports of hu-
man cell transformation using viruses and viral
oncogenes appeared only in the late 1970th [1]. In
early 80th it was shown that immortalized NIH3T3
mouse fibroblast cells introduced with total ge-
nomic DNA from human tumors were converted
into cancer cells; later it was found that H-RAS
gene harboring a point mutation induced trans-
formation of NIH3T3 cells in culture and con-
ferred on them the ability to induce tumors in
nude mice (reviewed in [2]). These discoveries
marked an advent of the intense searching for the
abnormal genes influencing the development of
human cancer that continues today [3].
Early works stated that in vitro transformation
of human cells by a single carcinogenic agent in
contrast to rodent cells was an extraordinarily
rare event [1]. Moreover, a spontaneous im-
mortalization following senescence was also an
extremely rare event in human fibroblasts and
epithelial cells, although it occurred commonly
in rodent cells with varying frequencies depend-
ing on species from which the cells were derived
[1, 4]. One of the explanations of intrinsic anti-
neoplastic mechanisms of human cells was dif-
ferences in telomere biology between human and
murine cells. Mouse cells begin their replication
ex vivo with extremely long telomeres: 3–10-fold
longer than in identical human cells and the ten-
dency for progressive telomere erosion might ef-
fectively be countered by the basal telomerase ac-
tivity that is constitutively present in mouse cells
[1, 4–8]. Additionally, the basal metabolic rate is
about 7-fold higher in mice than in humans and
this affects the levels of endogenous oxidants and
other mutagens that are produced as by-products
of normal oxidative metabolism resulting in 18-
fold more breakdown products of DNA in mice
[4]. Moreover, the rates of metabolic conversion
of procarcinogens to carcinogens and the detoxi-
fication of many other potential mutagens can
occur with greatly differing kinetics [4]. Further-
more, humans have more efficient DNA repair
system, and the rate of 5-methylcytosine decline
during cellular senescence is much slower in hu-
man cells than in mouse cells [1].
Îáçîðíûå ñòàòüè
37ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
Nevertheless, telomerase-deficient primary
mouse embryonic fibroblasts (MEFs) could be
immortalized/transformed in culture, and gener-
ated tumors in nude mice following transforma-
tion [9]. It was concluded that telomerase is not
required for establishment of immortalized cell
lines, oncogenic transformation, or tumor forma-
tion in mice. Another research group transformed
human primary fibroblasts and human primary
mesodermal cells introducing simultaneously three
oncogenes E1A, MDM2, and H-RASV12. These
cells formed colonies in soft agar and tumors in
mice, but they and the majority of the tumors
derived from them lacked telomerase activity,
and telomere erosion was observed [10]. Authors
have deduced that telomere maintenance is not
obligatory for tumorigenic conversion. To the
point, human primary melanomas show telomere
maintenance as a late event in tumor progression
(metastatic melanoma), thus, telomere mainte-
nance/immortalization is associated with pro-
gression rather than initiation of melanoma [11].
Furthermore, like primary human cells, pri-
mary MEFs require combination of two «hits»
to acquire the capacity to form tumors [9, 12-
19]. There are also cases of a conversion of nor-
mal primary rodent [20–23] and human [24–27]
cells to fully transformed cells with a single on-
cogene under specific experimental (significant
overexpression of oncogene) and culture con-
ditions. Culture conditions significantly affect
proliferative (before senescence) [5] and trans-
formation potential of cells [8]. For example,
wild type MEFs grown in serum-free medium
supplemented with defined growth components
(EGF, PDGF, insulin, high density lipoprotein,
fibronectin, and transferrin) were refractory to
transformation by oncogenic RAS + E1A [7].
Moreover, RAS + E1A-induced chromosome in-
stability, colony formation and tumorigenesis of
the p53 –/– serum free-MEFs also could be at-
tenuated by treating the cells with the free-radical
scavenger N-acetylcysteine [7].
Finally, humans live, on average, 30–50 times
longer than mice and undergo about 105 more
cell divisions in a lifetime (1016 versus 1011 mi-
toses) [4]. Nevertheless, epidemiological studies
have revealed that the life-time risk of develop-
ing cancer is comparable in both species. About
30 % of laboratory rodents have cancer by the
end of their 2–3 year life-span and about 30 %
of people have cancer by the end of their 70–80
year life-span [4].
Thus, it seems that in vitro (and likely in vivo)
transformation process may be fundamentally
similar in rodent and human cells and be sig-
nificantly affected by non-physiological culture
conditions in vitro.
Senescence. In contrast to germ cells and cer-
tain stem cells somatic cells have a limited lifes-
pan, gradually slow in growth, and stop dividing, a
process known as replicative senescence [28]. The
finite replicative life span of normal cells in culture
was first described approximately 50 years ago by
Leonard Hayflick [29], and is often termed as the
«Hayflick limit» [30]. The precise number of rep-
licative doublings exhibited by cultured cells before
they reach senescence depends on the species from
which the cells are derived, the tissue of origin,
and the age of the donor organism [31]. Cultured
human primary fibroblastic cells generally display
50 to 80 population doublings (PD) [7, 32, 33],
whereas explanted MEFs can divide just for 15–30
PD before undergoing senescence [5, 7]. Primary
normal human astrocytes perform only about 20
PD before reaching senescence [34]. Human ke-
ratinocytes have an in vitro life span of 15–20 PD
in serum-free chemically defined media, whereas
keratinocytes grown on feeder fibroblasts proliferate
for up to 50 PD [7, 32] and in F medium on feeder
fibroblasts for up to 80 PD before senescencing
[35]. Most published reports on cultured human
epithelial cells have shown active growth for only
10 to 30 PD [32]. Significantly, simple changes in
the culture conditions (defined growth factors in-
stead of serum) could permit active growth of hu-
man mammary epithelial cells for up to 60 PD,
whereas addition of oxytocin (endogenous antioxi-
dant) gave about 20 PD of increased proliferation
[32]. MEFs proliferate for more than 60 PD with
no signs of replicative senescence under physiologi-
cal oxygen levels (3 % versus 21 %) [7, 8]. Thus,
primary cells undergo stress-associated senescence
due to in vitro non-physiological standard cultur-
ing conditions, including disruption of cell-cell
contacts, lack of heterotypic interactions between
different cell types, the medium-to-cell ratio, per-
sistent signaling pathways activation by mitogens,
absence of appropriate survival factors, hyperoxia,
and plating on plastic [5].
38 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
The process of senescence occurs both in
vitro and in vivo. Cellular senescence in vivo is
now recognized to play an active role as a tu-
mor suppressor pathway [36, 37], in the loss of
regenerative potential in aging tissues and in the
pathogenesis of cardiovascular diseases [38]. Se-
nescence in vitro is marked by the appearance of
large, flattened vacuolated cells and character-
ized by the inability of cells to proliferate despite
the presence of a steady supply of abundant nu-
trients, mitogens [39], ample room for expansion
[33], and by the maintenance of cell viability/
resistance to apoptosis and metabolic activity for
months [37, 38, 40]. Once senescence is triggered,
cells are not capable of re-entering the cell cycle
or developing into tumors [36]. Moreover, senes-
cent cells secrete a plethora of factors primarily
involved in insulin-like growth factor and trans-
forming growth factor signaling, extracellular
matrix remodeling, and inflammation. Altogether
these secreted factors were referred to as the «Se-
nescence-Messaging Secretome» or the «Senes-
cence-Associated Secretory Phenotype» [33].
Senescent cells can be distinguished from pre-
senescent, immortal, quiescent or terminally dif-
ferentiated cells by histochemical detection of the
biomarker senescence-associated -galactosidase
[41]. Senescence accompanies changes in nuclear
morphology and formation of a distinct chro-
matin structure, called senescence-associated
heterochromatic foci (SAHF). These foci are
characterized by the accumulation of histone H3
trimethylated at lysine 9 and recruit heterochro-
matin proteins to the genes that are to be stably
repressed during senescence [41]. Importantly,
formation of SAHF and silencing of genes re-
quire an intact pRB pathway, since inhibition of
p16INK4A prevents SAHF formation and leads to
DNA replication [33, 41].
The onset of senescence is partly attributable
to the shortening of telomeres by approximately
50–200 base pairs with each cell division to a
threshold where it is recognized as DNA dam-
age and thus initiates replicative senescence [31,
33, 38]. Critical telomere shortening and even-
tual dysfunction triggers a classical DNA dam-
age response involving a number of cellular pro-
teins, including ataxia telangiectasia mutated
protein (ATM), check point kinase 1/2 (CHK1
and CHK2), p16INK4A, p53, 53 binding protein 1
(53BP1), p21CIP1, nijmegan breakage syndrome
1 protein (NBS1), plasminogen activator protein
1 (PAI1), and phosphorylated histone -H2A.X
[41]. These cellular factors cooperate to initiate
senescence, thereby preventing cellular prolifera-
tion in the presence of damaged chromosomes
and hence limiting the acquisition of potential
pathogenic mutations [33, 41].
However, telomere attrition is not the only
stimulus for replicative senescence. Oxidative
stress can induce or accelerate the onset, a phe-
nomenon referred to as stress-induced replicative
senescence. It occurs in several ways (reviewed
in [38]): oxidative stress can activate critical cell
cycle tumor suppressor proteins p53 and pRB by
oxidative-stress-induced DNA damage such as
double strand breaks. Oxidative stress can result
in oxidative modifications of triple guanine re-
peats (TTAGGG) in sequences of telomeric ends
making them more susceptible to breaks and en-
hancing the rate of telomere attrition. Oxidative-
stress-induced premature senescence might be
a function of a direct suppression of telomerase
activity. hTERT gene (encodes catalytic subunit
of human telomerase) expression is regulated by
many transcription factors, including AP1, SP1
and NF- B, all of which are redox regulated
[42]. In any case, oxidative stress results in the
loss of chromosomal integrity as manifested by
chromosomal fusions, recombination and deg-
radation, and contributes to DNA damage re-
sponses that eventually lead to the irreversible
cell-cycle arrest/senescence or cell death through
the activation of p53- and pRB-dependent func-
tions [24, 43].
Immortalization. Senescence (telomere erosion-
induced, oncogene-induced [33], or stress-in-
duced [44]) forms a barrier against tumorigen-
esis. Overcoming of senescence and acquisition
of immortality is an essential step in the pro-
cess of malignant transformation [33, 45]. Cel-
lular immortalization allows a cell to indefi-
nitely proliferate while accumulating genetic
abnormalities [27, 46]. Immortalized cells can-
not grow over one another (contact inhibition)
[47, 48], their proliferation ability is growth fac-
tor dependent [49] and their growth is anchor-
age-dependent (cells do not form colonies in
soft agar) [50–52]. There are several mutually
complementary mechanisms that contribute to
39ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
a cell being able to escape senescence and be-
come immortal, including telomere length sta-
bilization, epigenetic gene silencing by selective
promoter methylation, oxidative DNA damage,
inactivation of cell cycle regulatory genes such as
p16INK4A, p53, pRB, p21CIP1, overexpression of a
cellular oncogenic proteins such as c-MYC, BMI1
or through expression of viral oncogenes [41].
In vitro immortalization of various cell types
was successfully performed by the introduction of
viral genomes/oncogenes (Table 1), telomerase
catalytic subunit (hTERT) (Table 2) as well as by
enforced expression of transcription factors (e.g.
c-MYC, BMI1, ZNF217, or -catenin).
Genomes of viruses encode a number of regu-
latory and structural proteins but «immortalizating
effect» can be attributed only to several of them.
For SV40 viral oncoproteins responsible for im-
mortalization correspond to the portion of the
viral chromosome expressed early after infection,
which encodes two proteins, the large T-antigen
and the small t-antigen [92, 93]. For the adenovi-
ruses viral oncoproteins are encoded by a subset of
the early genes and termed the E1A proteins and
the E1B proteins [93]. Experiments with the hu-
man papilloma viruses uncovered a similar set of
early proteins called the E6 and E7 proteins [93,
94]. In the cell these oncoproteins bind to pRB
and p53 causing their ubiquitin-dependent pro-
teasomal degradation. It allows going through the
cell cycle checkpoints in an uncontrolled manner.
Role of pRB and p53 signaling pathways in cell
cycle regulation is presented in Figure. Cells ex-
pressing these viral oncogenes continue proliferat-
ing beyond the population doubling level, at which
their untreated counterparts become senescent,
but they eventually cease proliferating in a state
referred to as crisis [95–97]. A small number of
cells within the population may acquire the ability
to escape from crisis and form an immortalized
cell line. In all such cell lines examined, escape
from crisis has been shown to be associated with
activation of a telomere maintenance mechanism.
Viral oncoproteins can bind to multiple other
cellular proteins [92, 98], including several tran-
scription factor complexes involved in hTERT
transcription regulation [94]. For example, hu-
man papilloma virus E6 protein, via direct bind-
ing, increases c-MYC efficiency in activating
the hTERT promoter and, on the other hand,
E6 is able, through its association with E6AP, to
promote the degradation of the hTERT promoter
transcriptional repressor NFX1-91 [94]. Usage of
both viral oncogene and hTERT to induce immor-
talization has also been reported. For instance,
pre-adipocytes, bone marrow stromal cells and
ovarian surface epithelial cells were immortal-
ized by introduction of HPV E7 and hTERT [96].
Mechanistically, thus, process of immortalization
induced by viruses corresponds to a process of cell
cycle checkpoint proteins inactivation (pRB and/
or p53) and restoration of telomerase activity re-
sulting in telomere ends stabilization.
Ectopic expression of hTERT alone in pre-
senescent or still dividing cells can effectively
Schematic representation of pRB and p53 signaling
pathways. p16INK4A inhibits CDK4 and CDK6
preventing interaction with D-cyclins. CDK4 and
CDK6 phosphorylate pRB leading to a partial loss of its
ability to repress the E2F. When pRB-E2F suppressive
interaction is relaxed, E2F transactivates genes involved
in G1/S transition and in the initiation of DNA
replication in S phase. CDK2-cyclin E complexes can
further phosphorylate pRB resulting to complete its
release from interacting with E2F and, thus, promoting
S phase progression. P14ARF/p19ARF is an antagonist
for MDM2 which, in turn, regulates p53 stability
through its ubiquitin ligase activity. ARF sequesters
MDM2 resulting in p53 activation and stabilization.
p53 induces p21CIP1 expression. p21CIP1 associates with
cyclin D-CDK4/6, E/A-CDK2, and cyclin B-CDK1
complexes, and has a universal inhibitory activity
towards these CDKs thereby regulating G1/S transition,
S and M phases of cell cycle
40 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
immortalize them (Table 2). The native hTERT
locus is embedded in a large nuclease-resistant
chromatin domain in most normal human cells
[97]. As a result, the hTERT promoter is strin-
gently repressed in somatic primary cells. But in
immortalized and tumor cell lines hTERT is of-
ten up-regulated, and these cells are capable to
maintain stable telomere lengths by activation of
a telomere maintenance mechanism; also, there
is non-telomerase alternative mechanism of telo-
mere maintenance [95–100]. Except cases with
ectopic hTERT expression, in otherwise immor-
talized (and tumor) cells hTERT gene activa-
tion can occur in several ways probably mutu-
ally complementary to each other: through gene
amplification; nonreciprocal translocation by
chromosomal breakage at the hTERT locus and
subsequent ligation to heterologous sequences by
non-homologous end joining (NHEJ) mecha-
nisms resulting in the chromosomal rearrange-
ments upstream of its promoter; the activation
of c-MYC and inhibition of histone deacetylases
(HDACs) (reviewed in [97]). Hyperoxia or ad-
dition of exogenous H2O2 was shown to induce
senescence of fibroblasts despite hTERT overex-
pression and exogenous H2O2 prevented hTERT-
dependent immortalization of normal endothe-
lial cells, whereas N-acetylcystein (antioxidant)
permitted hTERT-dependent immortalization of
endothelial cells [38]. Indeed, oxidative stress re-
gulates hTERT at many levels, such as its gene ex-
pression, activity, and sub-cellular localization [42].
Immortalization of human and rodent cells was
also achieved by different transcription factors.
The c-MYC protein is a basic helix-loop-helix
leucine zipper transcription factor that modulates
Table 1
Immortalization of human cells by viral genomes/oncogenes
Indications. SV40 – simian polyomavirus; HPV16/18 – human papilloma virus types 16 and 18; Ad5 and 12 –
adenovirus type 5 and 12.
Cell type Immortalizing agent Ref.
Astrocytes
HS74BM diploid fetal bone marrow fibroblasts
IMR-90 diploid lung fibroblasts
Ciliary epithelial cells
Fetal liver epithelial cell
Mammary epithelial cells
Prostate epithelial cells
Tracheal epithelial cells
Uroepithelia cells
Cervical epithelial cells
Epidermal keratinocytes
Epidermal keratinocytes
Esophageal epithelial cells
Foreskin keratinocytes
Gingival keratinocytes
HFE keratinocytes
Mammary epithelial cells
Mammary epithelial cells
Uroepithelial cells (from ureteral uroepithelium)
Urothelial cells (from ureteric or bladder tissue)
WHE-7 fetal fibroblast
Embrionic kidney cells HEK293
Bronchial epithelial cells
Epidermal keratinocytes
SV40 T antigen
SV40
SV40
SV40
SV40 T antigen
SV40
SV40
SV40
SV40
HPV16/18
HPV16
HPV16 E6+E7
HPV16 E6/E7
HPV16/18/ 31/ 33
HPV16 E6
HPV16 E7
HPV16
HPV16 E6+E7
HPV16 E7
HPV16 E6+E7
HPV16 E6
Ad5
Ad12-SV40 fusion genome
Ad12-SV40 fusion genome
[53]
[54]
[55, 56]
[57]
[58]
[59]
[60]
[61]
[62]
[63, 64]
[65, 66]
[67, 68]
[69]
[70, 71]
[72]
[73]
[74]
[75]
[76]
[77]
[72]
[78]
[79]
[80]
41ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
Table 2
Molecular characteristics of immortalized human cell lines
Indications. RPE-340 – retinal pigment epithelial-340 cells; MDAH 087 – skin fibroblasts derived from a
patient with Li-Fraumeni syndrome; Y-27632 – Rho kinase (ROCK) inhibitor; 4-NQO 4-nitroquinoline 1-ox-
ide; ALT – alternative telomere maintenance mechanism; « » – overexpressed; « » – downregulated; «+» –
unchanged level; * for hTERT – overexpressed; «–» – undetected; N/A – not analyzed.
Cells Immort. agent p16INK4A pRB p53 p21CIPI hTERT* Ref.
Adenoid epithelial
cells and foreskin
keratinocytes
Ameloblastoma
cells
BJ fibroblasts,
RPE-340 cells
BJ fibroblasts
Cen3 fibroblasts
Esophageal epithe-
lial cells
Foreskin fibroblasts
Gingival and perio-
dontal ligament
fibroblasts
Dermal keratino-
cytes
Mammary epithe-
lial cells
Epi gingival kera-
tinocytes
Esophageal epithe-
lial cells
Mammary epithe-
lial cells
WHE-7 fetal fibro-
blasts
Foreskin fibro-
blasts
Prostate epithe-
lial cells
Mammary epithe-
lial cells
Oral keratinocytes
Foreskin keratino-
cytes
KMST-6 fibroblasts
MDAH 087 skin
fibroblasts
OUMS-24F fibro-
blasts
hTERT
hTERT
hTERT
hTERT
hTERT
hTERT
hTERT
hTERT
hTERT
hTERT
HPV E6
HPV16 E6/E7
HPV16 E6 + E7
HPV16 E6
c-MYC
c-MYC
ZNF217
Cyclin D1 + domi-
nant-negative p53
Y-27632
60Co
Aflatoxin B1/X-rays
4-NQO
–
–
+/–
N/A
/ –
deletion
–
–
–
–
N/A
–
+
–
N/A
N/A
+
–
–
+
+ hyper
phosphorylated
N/A
+
+ unaffected
N/A
+ unaffected
+ hyper
phosphorylated
+ hyper
phosphorylated
N/A
E2F1 elevated
+ hyper
phosphorylated
+ hyper
phosphorylated
N/A
N/A
+ unaffected
N/A
N/A
+ hyper
phosphorylated
+ hyper
phosphorylated
+ hyper
phosphorylated
+
+
+
+
+ mutated
+
+/ mutated
N/A
+
+
N/A
N/A
+
+
+
+
mutated
+
N/A
+
mutated
N/A
+
+
N/A
N/A
+/–
N/A
N/A
+
+
+
N/A
N/A
N/A
+
+
+
+
+
+
+
+
+
+
+
+
N/A
N/A
N/A
+
+
ALT
+
ALT
ALT
ALT
[81]
[82]
[83]
[84]
[24]
[69]
[85]
[72]
[86]
[87]
[72]
[69]
[75]
[72]
[88]
[89]
[90]
[91]
[28]
[72]
[72]
[72]
42 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
expression of a cohort of genes, including those
that function to promote cell growth and cell cy-
cle entry [88, 101]. c-MYC up-regulates certain
cyclin-dependent kinases (CDK4) and cyclins
(A, B1, D1, D2, and E), and represses cycline-
dependent kinase inhibitors (p15INK4B, p21CIP1,
and p27KIP1) [48, 101, 102]. Mechanisms can be
direct or indirect: for example, c-MYC directly
binds to the cyclin B1 promoter, but optimal in-
duction of expression occurs only when p53 is
concurrently inactivated [102], whereas cyclin D1
expression is positively regulated through MYC/
miR-378/TOB2/cyclin D1 functional module in
human mammary epithelial cells [103]. p21CIP1
expression is regulated both negatively and posi-
tively by c-MYC [104]. Induction of p21CIP1 by
c-MYC overexpression was p53-dependent in nor-
mal human and mouse fibroblasts and was associ-
ated with G2 arrest, whereas, inversely, c-MYC
repressed p21CIP1 transcription in p53-null mouse
cells and in a human adenocarcinoma cell line
[105]. Moreover, the hTERT promoter contains
the MYC binding site (E-box) and is a direct tran-
scriptional target of c-MYC [106]. c-MYC expres-
sion was reported to result in successful immor-
talization of rat kidney cells [22], mouse neural
precursor cells [107], human neural stem cells (by
v-MYC and c-MYC T58A mutant) [49], prostate
epithelial cells [89], and foreskin fibroblast cells
[88]. Interestingly, foreskin fibroblast cells had in-
creased levels of p16INK4A and p53 and functional
both p16INK4A–pRB (pRB phosphorylation was
reduced) and p53–p21CIP1 parthways. Prostate
epithelial cells preserved functional p53-p21CIP1
pathway and had elevated p16INK4A but, never-
theless, pRB phosphorylation was maintained.
Moreover, c-MYC allowed to tolerate ectopically
overexpressed p16INK4A in prostate epithelial cells,
whereas p16INK4A overexpression in foreskin fibro-
blast resulted in senescence. Foreskin fibroblast
cells also showed epigenetically silenced p14ARF
(unfortunately, p14ARF status was not analysed in
prostate epithelial cells). Rodent cells immortal-
ized by c-MYC characteristically inactivate the
ARF–p53–p21CIP1 pathway by loss of either func-
tional p53 or 19ARF [88]. p14ARF/p19ARF is unique
among c-MYC regulators. It selectively inactivates
the hyperproliferative and transforming func-
tions of c-MYC without affecting normal cell
cycle progression or preventing c-MYC-mediated
apoptosis [108]. Thus, p53 or p14ARF/p19ARF in-
activation is likely beneficial in cells immortalized
by c-MYC.
Other transcription factor BMI1, a member
of the Polycomb group of transcriptional repres-
sors [109], was initially identified as an oncogene
that cooperates with c-MYC in lymphomagenesis
[110]. Moreover, BMI1 is positively regulated by
c-MYC [37]. It was reported that overexpression
of BMI1 down-regulated p16INK4A and p19ARF
expression in mouse embryonic fibroblasts and
resulted in their immortalization [96], immortal-
ized primary human mammary epithelial cells
(HMEC) [111] and nasopharyngeal cells [112].
In both latter cases immortalization was accom-
panied by telomerase activation. BMI1 caused
the bypass of replicative senescence in normal
human oral keratinocytes but did not immortal-
ize them (no hTERT activation) [113]. BMI1
introduction along with human papilloma virus
E6 gene but not with E7 immortalized oral ke-
ratinocytes and it was associated with telomerase
activation [113]. Introduction of BMI1 as well
as p16INK4A-specific short hairpin RNA into hu-
man epithelial cells derived from skin, mammary
gland and lung suppressed p16INK4A expression
and extended cells life span; subsequent intro-
duction of hTERT in these cells resulted in their
efficient immortalization with following mainte-
nance of near normal diploidy [96]. The reason
why some cell types become immortalized after
BMI1 introduction alone, whereas other cells do
not is unclear. It was speculated that BMI1-in-
duced immortalization mechanism may be tissue-
dependent or because the cultured cells already
underwent critical steps towards immortalization
[113]. The mechanism whereby BMI1 promotes
evasion of senescence involves, among other tar-
gets, transcriptional silencing of CDKN2A (cy-
clin-dependent kinase inhibitor 2A, which en-
codes both p16INK4A and p14ARF) and CDKN2B,
which encodes p15INK4B [42, 110, 114].
Transcription factor ZNF217 was able to im-
mortalize human primary HMECs disturbing
ARF-p53-p21CIP1 pathway [90]. -Catenin, a
member of the Armadillo (ARM) repeat protein
superfamily, activates transcription of target genes
primarily by associating with the T cell factor/lym-
phoid enhancer-binding factor (TCF/LEF) family
[115, 116]. Expression of -catenin immortalized
43ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
primary mouse melanocytes directly repressing the
expression of p16INK4A by binding to its promoter
[117]. Other well known targets of -catenin are
c-MYC and cyclin D1 [115].
In fact, all mentioned targets of discussed tran-
scription factors are only the «top of an iceberg».
It was established that c-MYC regulates a total
of 1469 target genes in HeLa cells and human
primary fibroblasts [118], -catenin in HCT116
colorectal carcinoma cells directly bound in vivo
to more than 400 target genes [115]. ZNF217 was
shown to target 103 genes in breast cancer cell line
MCF7, 44 genes in colon cancer cell line SW480,
and 101 genes in teratocarcinoma cell line Ntera2
[119]. Moreover, NF- B, STAT3, ER , JUN, ELK4,
CEBP, and ETS1 were found among transcrip-
tional regulator genes up-regulated by c-MYC
in human B cell line P493 [120], and EPAS1,
ERF, FHL2, JUN, MNT, MYT1, RPO1-2, SOX4,
TEAD4, TIEG1, and ZFP28 in pancreatic -cells
[121], which, in turn, regulate additional gene
cohorts resulting eventually in global change in
gene expression. C-MYC can regulate overall up
to 10–15 % of all genes [120, 122], among which
there are those regulating replication and repara-
tion in S phase and chromosome separation dur-
ing M phase [121–124].
Spontaneously immortalized cells emerge at
an extremely low frequency (about 10–7) during
crisis in vitro [85, 97], but show the same gen-
eral changes in cell cycle checkpoint pathways
as all otherwise immortalized cells [125–127].
Thus, overwhelming majority of immortalized
cells irrespective of «immortalizing agent» do not
express p16INK4A cell cycle suppressor and this
correlates with pRB hyperphosphorylation (inac-
tivation) (Table 2). Indeed, it has been estimated
that more than 70% of human immortalized and
cancer cell lines lack functional p16INK4A due to
promoter methylation, mutation, or homozygous
deletion. In many instances the deletions affect
both p16INK4A and p14ARF/p19ARF, but a substan-
tial proportion of the missense mutations exclu-
sively affect p16INK4A, suggesting that p16INK4A
itself plays significant and non-redundant role in
tumor suppression [128]. Spontaneous reduction
in p16INK4A expression due to promoter methyla-
tion (most often) or otherwise mechanisms during
in vitro propagation of normal primary cells has
been documented, for example, in HMEC [96,
129], fibroblasts derived from lung [96], human
keratinocytes [96, 130], and human astrocytes
[34]. Molecular mechanism of p16INK4A gene in-
activation by epigenetic deregulating methylation
during progression from primary cells to immor-
talized and pre-malignant cells is complex; it is
under intensive investigation and may be differ-
ent in mouse and human cells [128, 131–137].
Nevertheless, in contrast to frequent loss of
p16INK4A expression in vitro, another well doc-
umented fact is that p16INK4A is overexpressed
in certain samples of different cancer types. In
tumors increased p16INK4A expression correlates
statistically with RB loss of heterozygosity [138].
In spite of being tumor suppressor, overexpres-
sion of p16INK4A, nevertheless, correlates with a
poor prognosis and seems to be an unfavorable
prognostic indicator [138].
ARF–p53–p21CIP1 pathway is likely less critical
for immortalization of human cells than
p16INK4A–pRB, because approximately in a half
of analyzed works it was apparently functional
(Table 2). Moreover, Odell et al. [139] exam-
ined more than a hundred spontaneously im-
mortalized MEF cell lines and found that at
least half of them had neither a p53 mutation
nor loss of p19ARF. Nevertheless, it is neces-
sary to take into account that analysis of ARF-
p53-p21CIP1 pathway in most works (where it
was shown functional) was performed once in
certain population doubling (PD), but it could
be inactivated later. For instance, hTERT im-
mortalized cen3tel fibroblasts up to 108 PD had
wild-type p53 sequence, whereas at late PDs (165
and 366 PDs) had a mutation in codon 161 [24].
Thus, it is possible to conclude that for suc-
cessful immortalization cells must overcome se-
nescence by inactivating p16INK4A–pRB and /or
ARF-p53-p21CIP1 and crisis by maintaining their
telomeres by activation of hTERT expression
or by an alternative mechanism for lengthening
telomeres (ALT) [99, 100]. However, it needs to
keep in mind that every oncogene/«immortalizing
agent» introduced into a cell has a great number
of targets and multidirectional effects rather than
being one-way agent. Thus, the process of im-
mortalization is not simply a number of well de-
fined events like inactivation of cell cycle nega-
tive regulators and activation of telomerase but,
instead, is associated with karyotype/genome ab-
44 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
normalities (aneuploidy/gain or loss of additional
chromosomes, translocations, deletions and am-
plifications) and, as a consequence, with global
changes in gene expression [165]. All immortal-
ized cells have abnormal karyotypes irrespectively
of «immortalizing agent» (Table 3).
Also, significant changes in global gene ex-
pression can be reached through aberrant meth-
ylation of promoters. In HMECs during im-
mortalization global aberrant DNA methylation
changes occured in a stepwise fashion [129]. The
first aberrant DNA methylation step coincided
with overcoming stasis, and resulted in few to
hundreds of changes, depending on how stasis
was overcome (stress-inducing serum-free me-
dium, benzo(a)pyrene or p16INK4A shRNA). A
second step coincided with crisis/immortaliza-
tion and resulted in hundreds of additional DNA
methylation changes regardless of the immortal-
ization pathway [129].
All together, it explains why across cell types
and model systems genes in the cell cycle path-
way, cytoskeletal genes, IFN pathway, IGF path-
way, MAP kinase pathway, and oxidative stress
pathway were identified as regulators of senes-
cence/immortalization [41, 166]. It is worth no-
ticing that virus oncoproteins induce more pro-
found karyotype changes because of simultaneous
ablation of pRB and p53 pathways, inactivation
of which is directly linked with aneuploidy/poly-
ploidy. In contrast, hTERT alone immortalized
cells are suggested to be apparently genetically
stable frequently showing near diploid karyotypes
with lower abnormalities than otherwise immor-
talized cells [47, 142, 143, 147, 167–171]. It is
clear that telomerase introduction into a cell can
Table 3
Karyotype abnormalities in immortalized cells
Immortalizing agent Ref. Immortalizing agent Ref.
Cells immortalized by hTERT
Human adenoid epithelial cells and
foreskin keratinocytes
Human fibroblasts from two
centenarian individuals
Human normal fibroblasts
Sheep fibroblasts
Human bone marrow endothelial cells
Human small airway epithelial cells
Human mammary epithelial cells
Human myometrial and uterine
leiomyoma cells
Swine umbilical vein endothelial cells
Human mesenchymal stem cells
Human fetal hepatocytes
Human meibomian gland epithelial
cells
Cells immortalized by SV40 T
large antigen
Rabbit kidney epithelial cells
Human corneal epithelial cells
Human gingival keratinocytes
Human nasopharyngeal epithelial cells
Human bronchial epithelial cells
Human mammary epithelial cells
[81]
[140]
[83, 84,
141, 142]
[143]
[144]
[145]
[46]
[146]
[47]
[147]
[148]
[149]
[150]
[151]
[152]
[153]
[154]
[155]
Cells immortalized by HPV16/18 E6/E7
Human epidermal keratinocytes
Human smooth muscle cells
Human nasopharyngeal epithelial cells
Human extravillous cytotrophoblasts
Human bronchial epithelial cells
Spontaneously immortalized cells
MEFs
Syrian hamster embryo cells
Human epidermal cells
Human keratinocytes
Murine neural crest-derived corneal
progenitor cells
Immortalized by otherwise ways
Mouse embryos cells by v-SIS or K51
oncogenes
Human mesenchymal stem cells by E6/E7
plus hTERT or BMI1, E6 plus hTERT
Human meibomian gland epithelial cells by
SV40 LT antigen plus hTERT
Human bronchial epithelial cells by hTERT
plus CDK4 or HPV16 E6/E7
Human bronchial epithelial cells by BMI1
plus hTERT
Human prostate epithelial cells by c-MYC
Oral keratinocytes by cyclin D1 plus mutant p53
[156]
[157]
[153]
[158]
[159]
[160]
[161]
[125]
[162]
[126]
[163]
[147]
[149]
[159]
[164]
[89]
[91]
45ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
stabilize telomeres preventing numerous chromo-
somal aberrations occuring via telomere dysfunc-
tion and the breakage-fusion-bridge mechanism
[165]. Nevertheless, although BMI1 and hTERT
immortalized human embryonic stem cells at
passage 40 [172], as well as hTERT immortalized
human neural progenitor cells isolated from the
ventral telencephalons of first trimester embryos
after more than 40 passages (80–120 PD) [170]
had normal karyotypes, in contrast, hTERT im-
mortalized bone marrow endothelial cell clones
showed numerous abnormalities after 75 PD
(more than 160 days) [144]. Interestingly, mass
culture of these cells at 65 PD (120 days) had 47
chromosomes without any structural abnormali-
ties and served this karyotype at 135 PD [144].
Also, prolonged cultivation of telomerase-immor-
talized human fibroblasts led to a premalignant
phenotype, although hTERT-immortalized cells
behaved similarly to primary cells during the first
150 PDs [141]. The possible pitfall of «normal»
or near diploid karyotypes of hTERT immortal-
ized cells can result from exploiting conventional
cytogenetic techniques for karyotyping, which do
not allow detecting the subchromosomal aberra-
tions. In contrast, for example, SNP and CGH
arrays revealed multiple genomic abnormalities
in tumors with near diploid katyotypes.
How do immortalized and tumor cells become
aneuploid? p53, a well known «genome safeguard»,
plays multiple roles in maintaining genomic stability
in somatic cells. Loss of p53 functions promotes on-
cogenesis by inducing chromosomal instability and
aneuploidy [173-176] and enabling efficient accu-
mulation of genetic mutations [177]. Loss or muta-
tional inactivation of p53 results in a high frequency
of centrosome amplification in part via allowing the
activation of CDK2-cyclin E (as well as CDK2-
cyclin A), which is a critical factor for the initiation
of centrosome duplication [178]. It also allows im-
mature escaping from cell cycle G2 checkpoint ar-
rest through inability of p21CIP1 to inactivate CDK2,
and this leads to reinforcement of CDK2-dependent
NF-Y phosphorylation and NF-Y dependent tran-
scription of the cell cycle G2-regulatory genes, in-
cluding CDK1, CDC25, cyclin A and B [179].
There is also a link between pRB inactiva-
tion, cell aneuploidy and chromosome instabil-
ity (CIN). Aneuploidy and CIN results from
persistent defects in mitotic fidelity, and several
mechanisms have been described that cause cells
to missegregate whole chromosomes [176, 180,
181]. More than 50 proteins are able to trigger
polyploidy/aneuploidy when are appropriately
misregulated (mutation, depletion, knockdown
or overexpression) [182]. If the dosage of any
one of many proteins involved in ensuring chro-
mosome segregation fidelity is disrupted by the
missegregation of the chromosome carrying that
gene, the resulting imbalance can further com-
promise chromosome segregation accuracy [176].
Importantly, pRB-E2Fs pathway directly regu-
lates genes involved in bipolar spindle formation,
chromosome-spindle association, chromosome
cohesion, and the spindle assembly checkpoint
(SAC) [183]. Acute pRB suppression in IMR90
cells [184], HCT116 cells [185], primary hu-
man fibroblasts [186], mouse embryonic fibro-
blasts [180], and mouse adult fibroblasts [187]
caused misregulation of chechpoint genes [180,
185, 186] and, as a consequence, gave rise to
centrosome amplification, multipolar spindles,
anaphase bridges, lagging chromosomes, and mi-
cronuclei harbouring whole chromosomes result-
ing in polyploidy/aneuploidy and cancerogenesis
[180, 184–188]. Moreover, pRB influences mi-
totic chromosome condensation in E2F-inde-
pendent manner, and loss of pRB function can
influence chromosome loss irrespectively of pro-
liferation [189]. pRB can interact with the con-
densin II subunit CAP-D3, and this interaction is
necessary for chromosome compaction in mitosis
[189]. pRB depletion compromises centromeric
localization of CAP-D3/condensin II and chro-
mosome cohesion, leading to an increase in in-
tercentromeric distance and deformation of cen-
tromeric structure [181]. These defects promotes
merotelic attachment (occurs when one kineto-
chore is attached to both mitotic spindle poles),
resulting in failure of chromosome congression
and an increased propensity for lagging chromo-
somes following mitotic delay [181].
In contrast to pRB, p107 and p130 (also
members of retinoblastoma family) are rarely
found inactivated in human tumors [190], and
this fact determined predominant research on
pRB, but it hardly proves the unique importance
of pRB in cell cycle regulation. Equally weighty
roles of p107 and p130 may well be masked by
a functional redundancy that they have with one
46 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
another. Such redundancy would drastically re-
duce the likelihood of their elimination from tu-
mor cell genomes during tumor progression [191].
Indeed, these proteins are part of a «tumor-sur-
veillance» mechanism and can suppress tumori-
genesis [190, 192–194].
Another consequence of pRB and p53 path-
ways deregulation is compromised DNA damage
surveillance and repair. More than 70 genes have
been identified that have roles in DNA damage
surveillance and repair [195], and many of them
are regulated by pRB and p53 [183, 195].
pRB and/or p53 pathways deregulation and
hTERT expression (or ALT) are markers and indis-
pensable conditions of immortalization. Deregu-
lated pRB and/or p53 pathways inevitably leads
to numerical and structural chromosome aber-
rations. A cell attains immortality by a global
change in gene expression, which accompanies
karyotype changes. Although a few common chro-
mosome aberrations might have been observed in
different immortalized cell, karyotype changes,
in general, have stochastic nature. Immortalized
cells with aberrant karyotypes are prone to malig-
nant transformation.
Transformation. Cancer cells display several
hallmarks that can be distinguished from those of
normal counterparts. These include immortaliza-
tion (bypass of senescence), evasion of apoptosis,
immune destruction and anti-growth signals,
growth factor independence, reprogramming of ener-
gy metabolism (enhanced glycolysis), anchorage-
independence, resistance to contact inhibition, mi-
gration, invasion/degradation of matrix components,
angiogenesis, metastasis, inflammation, and geno-
me instability, which generates the genetic diver-
sity accelerating acquisition of all listed hallmarks
[31, 196]. In addition to cancer cells, tumors ex-
hibit another dimension of complexity: they contain
a repertoire of recruited, ostensibly normal cells
that contribute to the acquisition of hallmark traits
by creating the «tumor microenvironment» [196].
Cancer genes are often classified according to
whether they function in a dominant or recessive
manner at the level of the cancer cell. Dominant
cancer genes (also known as oncogenes) require
only one of the two parental alleles present in
a normal cell to be mutated, and the encoded
protein is usually constitutively activated by the
mutations. Recessive cancer genes (also known
as tumor suppressor genes) require mutation of
both parental alleles, and these usually result in
inactivation of the encoded protein. More than
80 % of the currently known cancer genes are
dominantly acting [3]. Census of cancer genes
lists 467 genes (data on December 2011, www.
sanger.ac.uk/genetics/CGP/Census), which are
supposed to be causally implicated in cancer de-
velopment when appropriately changed (point
mutations, deletions, translocations or ampli-
fications) [197]. However, studies in mice have
suggested that more than 3000 genes, when ap-
propriately altered, may have the potential to
contribute to cancer development (see reference
in [198]).
The process of cellular transformation has been
intensively studied in vitro using cell-culture tech-
niques. Most research works on this issue satisfy four
criteria: the cells are immortalized, i.e., can grow
indefinitely in culture; the cells can efficiently form
colonies in soft agar; the cells can develop tumor in
immunodeficient mice; the xenograft or orthotopic
tumor in the mouse shows malignant histology to
exclude a pseudo-tumor or a benign tumor [267].
Actually, there are a few interesting outliers. For
example, cells obtained by stable overexpression of
cyclooxygenase 1 in spontaneously immortalized
human umbilical vein endothelial cells underwent
contact inhibition, failed to grow under anchor-
age-independent conditions but grew aggressively
as tumors in mice [202]. Human primary foreskin
fibroblasts in which E1A+H-RASV12+MDM2 were
introduced, although able to form colonies in soft
agar and tumors in nude mice, were not immor-
tal and, if maintained in culture for an extended
period of time (40–50 generations), underwent a
crisis phase characterized by dramatically reduced
proliferation and adoption of a senescent pheno-
type [10]. Cells were telomerase-negative, only few
of them eventually survived this phase and these
cells became telomerase-positive [10].
MAPK and PI3K-AKT signaling pathways in
transformation: a double-edged sword. One prom-
inent hallmark of transformed in vitro cells irre-
spectively of transforming agent is upregulation
of RAS-dependent extracellular signal-regulated
kinases 1 and 2 (ERK1/2) mitogen-activated
protein kinase (MAPK) pathway, phosphoinosit-
ide-3-kinase (PI3K)-mammalian target of ra-
pamycin (mTOR)-AKT pathway and overex-
47ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
pression of CDKs/cyclins. Actually, besides
MAPK and PI3K/AKT signaling, other path-
ways can also be activated. Nevertheless, RAS-
dependent extracellular signal-regulated kinase
1/2 (ERK1/2) mitogen-activated protein (MAP)
kinase [268–270] and phosphoinositide-3-ki-
nase (PI3K)-mammalian target of rapamycin
(mTOR)-AKT cascades [271] are the key signal
transduction pathways responsible for integrat-
ing the different environmental signals and re-
laying the information to the cell cycle control
system. Both these pathways are hyperactivated
frequently in transformed cells in vitro and tu-
mors in vivo, and are involved in regulation of all
aspects of normal and tumor cell biology (e.g.,
cell growth, proliferation, apoptosis, migration,
invasion etc).
As it is reviewed in [268-270], ERK1/2 are
required for cyclin D1 expression via regulation
of FOS family members and c-MYC transcrip-
tion factors, as well as inhibition of TOB1 and
JUND, cyclin D1 expression negative regula-
tors. The ERK pathway may assist in both the
assembly and stabilization of cyclin D1-CDK4/6
complexes via HSC70. There are also a few re-
ports implicating MAPK pathway in the regu-
lation of cyclin D2 and cyclin D3 expression.
ERK activity is required for proper nuclear
translocation of CDK2, and in the nucleus ERK
regulates phosphorylation of a CDK2 activat-
ing site. ERK can phosphorylate two of four
phosphorylation sites of the cytoplasmic reten-
tion sequence of cyclin B1, which are neces-
sary for nuclear localization of cyclin B1. ERK
futher contributes to CDK1-cyclin B activation
via RSK/MYT1/CDK1-cyclin B pathway. The
RAS-ERK signaling pathway is involved in the
mitogen-induced downregulation of p27KIP1. The
degradation of p27KIP1 at the G1/S transition
depends on the accumulation of cyclin E and
concomitant activation of CDK2, events that
are conditional on earlier activation of cyclin D-
CDK4/6 complexes by the ERK pathway.
Activation of ERK markedly enhances c-
MYC protein stability, which can transcription-
ally upregulate expression of certain cycline-
dependent kinases (CDK4) and cyclins (A, B1,
D1, D2 and E), and represses cyclin-dependent
kinase inhibitors (p15INK4B, p21CIP1 and p27KIP1)
[48, 101,102, 272]. ERK1/2 dislodge pRB from
its interaction with lamin A, thereby facilitating
its rapid phosphorylation and consequently pro-
moting E2F activation and cell cycle entry [273].
Pyrimidine nucleotides serve as essential precur-
sors for the synthesis of RNA and DNA, phos-
pholipids, UDP-sugars and glycogen [268, 270].
The rate-limiting step in the pyrimidine pathway
is catalysed by the carbamoyl-phosphate synthe-
tase enzyme, which is part of the large multifunc-
tional protein CAD [268, 270]. ERK2 directly
phosphorylates CAD activating it. ERK may im-
pact on global protein synthesis through direct
regulation of ribosomal gene transcription [268].
Over a hundred putative AKT substrates have
been reported. Targets among cell cycle regulating
proteins are reviewed in [43, 274, 275]. GSK3-
mediated phosphorylation of cyclin D and cy-
clin E and the transcripton factors c-JUN and
c-MYC, which all play a central role in the G1-
to-S phase cell-cycle transition, targets them for
proteasomal degradation. Phosphorylation and
inhibition of GSK3 by AKT enhances the sta-
bility of these proteins. AKT reduces p21CIP1
protein level through downregulation of p53-
mediated transcription and activation of MDM2,
and inhibits p27KIP1 expression via inactivation
of FOXO family of transcription factors. AKT
phosphorylates both p21CIP1 and the p27KIP1 cy-
clin-dependent kinase inhibitors leading to their
cytosolic sequestration and phosphorylates and
deactivates pRB leading to the activation of E2F.
AKT induces p27KIP1 degradation via GSK3 /c-
MYC/p27KIP1.
Aberrant activation of mTORC1 is a common
molecular event in a variety of cancers [276, 277].
Activation of the AKT and ERK pathways acts
in a synergistic manner to promote mTORC1
signaling through phosphorylation of a tuberous
sclerosis complex 2 (TSC2), GTPase activator
protein (GAP), leading to the disruption of the
TSC1-TSC2 complex as an inhibitor of RHEB,
which in turn regulates mTORC1. AKT phos-
phorylates residues of TSC2 distinct from those
phosphorylated by ERK [278]. Furthermore, the
kinase RSK, a direct downstream substrate of ERK,
can also phosphorylate TSC2 to inhibit the func-
tion of TSC1/TSC2 complex [270]. The S6K1
and 4E-BP1/eIF4E pathways represent critical
mediators of mTORC1-dependent cell cycle con-
trol [279, 280] by promoting the cap-dependent
48 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
translation of many target mRNAs, including
those encoding cyclins and c-MYC [274, 281].
mTORC2 also contributes to cell size and cell
cycle regulation via AKT activation and, thus,
contributing to TSC2 inactivation [275].
Nevetheless, it should be noted that the induc-
tion of cell cycle arrest by hyperactivation of the
ERK1/2 pathway does occur in some cell lines
and is frequently observed in non-immortalized
primary cells. Expression of activated forms of
RAS, RAF or MEK1 was shown to elicit cell
cycle arrest in primary fibroblasts, Schwann cells,
hepatocytes, T lymphocytes, keratinocytes, astro-
cytes, and epithelial intestinal cells (reviewed in
[268]). Notably, the proliferation arrest observed
in primary fibroblasts, astrocytes and epithelial
intestinal cells is permanent and phenotypi-
cally related to cellular senescence [268]. This
phenomenon is not restricted to MAPK path-
way. AKT overexpression induced senescence of
primary and immortalized esophageal epithelial
cells [282], primary MEFs [283], primary human
aortic endothelial cells, human dermal microvas-
cular endothelial cells, and human umbilical vein
endothelial cells [284, 285]. Moreover, senes-
cence can be triggered in human cells by overex-
pression of E2F1/3, CDC6, MOS or deletion of
PTEN and NF1 (reviewed in [36]).
A robust and prolonged activation of ERK1/2
causes G1 arrest due to long-term p21CIP1 in-
duction [286, 287] and CDK2 inhibition and
also induces the expression of p53 and the CDK
inhibitors p16INK4A and p15INK4B in certain cell
lines [36, 268]. Indeed, ERK pathway can induce
p21CIP1 transcription [287], translation [288],
mRNA stabilization [289] and block proteasome-
mediated p21CIP1 degradation [287]. Constitutive
activation of AKT promotes senescence-like ar-
rest of cell growth via a p53/p21CIP1-dependent
pathway and this action is at least partly medi-
ated by the forkhead transcription factor [284].
On the other hand, hyperactivated MAPK
and PI3K-AKT pathways were documented in
most, if not all, tumors and elevated p21CIP1 ex-
pression was highlighted, for example, in rectal
stromal tumors [290], lung adenocarcinomas
[291], bladder tumors [292], colorectal carci-
nomas [293], ependymomas and astrocytomas
[294], hepatocellular carcinomas [295], choroidal
melanoma tumors [296], and rhabdomyosarcoma
cells [288]. Importantly, in these cancers p21CIP1
expression was associated with tumor malignancy
and poor prognosis but not with long-term sur-
vival as it was expected.
Thus, oncogene-induced senescence occurs in
primary cells, some immortalized and in benign
but not in advanced tumors. It suggests that tu-
mor cells gain resistance to p21CIP1-mediated
scenesence and inhibition of CDK/cyclin com-
plexes. Oncogene-induced senescence can be
bypassed by inactivating pRB and p53. Accord-
ingly, if pRB and/or p53 are inactivated in a cell
before an oncogenic event, senescence should be
averted what is supported by numerous in vivo
mouse modeling studies and by genetic analysis
of human tumors [36]. Moreover, RB deficiency
sharply increases the ability of RAS to bind gua-
nine nucleotides, resulting in its activation [297].
Importantly, p21CIP1 (and p27KIP1 to a lesser
degree) functions as an assembly- and activity-
promoting factor for cyclin D-CDK4, cyclin
E/A-CDK2, and cyclin B-CDK1 complexes
when p21CIP1 level is below a certain threshold,
after which the presence of excess p21CIP1 be-
comes inhibitory. Stoichiometry of p21CIP1 is
critical to allow or inhibit kinase activity [104,
286]. When one p21CIP1 molecule is binding to
cyclin-CDK, the complex is catalytically active,
while binding of several p21CIP1 subunits inhibits
the complex. Thus, simultaneous overexpression
of CDKs/cyclins with p21CIP1 would create more
active complexes fostering cell cycle progression
and resistance to antimitotic stimuli.
Interestingly, in contrast, loss or decrease of
cyclin dependent kinase inhibitor p27KIP1 is com-
monly seen in many human cancers as lung, breast
and prostate adenocarcinomas, gastrointestinal
malignancies, brain tumors, and lymphoprolifera-
tive neoplasms [298]. Level of p27KIP1 in epithe-
lial cancers correlates with the pathologic tumor
grade: high-grade, poorly differentiated tumors
showing significantly lower p27KIP1 protein than
their well-differentiated counterparts [298]. Thus,
selection of tumor cells against p27KIP1 is likely
beneficial, and p27KIP1 has less profound role in
CDK-cyclin complex assembly than p21CIP1.
Karyotype evolution, selection and tumorige-
necity. Accoding to the Duesberg's evolutionary
chromosomal cancer theory [299–302], «activated
oncogenes induce neoplastic transformation by
49ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
inducing random aneuploidy. Aneuploidy destabi-
lizes the karyotype by unbalancing teams of proteins
that segregate, synthesize and repair chromosomes in
proportion to the degree of aneuploidy. Aneuploidy
initiates and maintains karyotypic evolutions
automatically because of the inherent instability
of aneuploidy. Occasionally, rare cancer-causing
karyotypes evolve stochastically. These cancer-
causing karyotypes are then stabilized against
the inherent instability of aneuploidy by selection
for transforming function within narrow clonal limits
of variation. Flexibility and heterogeneity of cancer
karyotypes is the basis for the further, spontaneous
evolutions that are known as tumor progression, such
as metastasis and drug resistance». Most oncogenes
deregulate DNA replication, centrosome amplification
and chromosome segregation and lead to formation
of DNA double strand breaks and chromosome
instability. Indeed, oncogene and carcinogene induced
chromosome instability is a driving force of cell
immortalization and tumor evolution (Stepanenko
and Kavsan, in preparation). For example, activated
RAS induces DNA double strand breaks in NIH3T3
fibroblasts within a single cell cycle; other oncogenes,
including MYC, cyclin E, MOS, CDC25A, E2F1
and sustained delivery of growth factors have similar
effects in various cell types and in animal models (re-
viewed in [303]). Importantly, most of experiments
detecting transforming ability of genes overexpressed
and/or mutated in tumors (oncogenes) were per-
formed using mouse and human cell lines (Table 4
and 5), which represent already immortalized cells
with abnormal karyotypes (poly-/aneuploids with se-
vere chromosome rearrangements) and are prone to
progress to completely transformed cells under culture
conditions.
Human embryonic kidney 293 cells (also of-
ten referred to as 293 cells, HEK 293, or less
precisely HEK cells) is widely used human cell
line both for basic molecular studies and as a ve-
hicle for the production of recombinant proteines
and viruses [304]. Originally named simply «293
cells» («293» designates a number of experiment),
they were obtained by exposing human embry-
onic kidney cell culture to mechanically sheared
fragments of adenovirus type 5 DNA (Ad5) [78].
After transformation the cells subcultured more
than 100 times could be considered as an estab-
lished/immortalized line and contained 4 to 5
fragments of Ad5 genome [78]. The transform-
ing region of the human adenovirus is within the
left 11 % of the viral genome encoding E1A and
E1B proteins which are necessary and sufficient
for mammalian cell transformation by Ads [305].
The integration site of the adenoviral DNA was
mapped to chromosome region 19q13.2 [305].
Adenovirus-induced chromosome aberra-
tions in human cells are well documented fact
[306, 307]. Bylund et al. [304] performed cyto-
genetic studies on the 293 cells obtained from
different sources. Karyotype analysis (G-banding
and spectral karyotyping) showed that 293 cells
(from ECACC, Salisbury, UK) cultured for less
than ten days prior to harvesting was near trip-
loid with 62–70 chromosomes/cell and had lots
of chromosomal abnormalities. No additional
chromosomal changes were found between 293
cells and 293aged cells (in culture 6 months, more
100 PD). Thus, 293 cells exhibit the cytogenetic
stability during culturing. Another work on 293
cell karyotype (cells were obtained from ATCC,
Manassas, VA, USA) also showed triploidy of
these cells [308] but with only partial overlap in
chromosome gains/losses comparing with cells
analysed by Bylund et al. [304]. Interestingly,
original 293 cells obtained by Graham et al. [78]
and tested at passage 8 were near-tetraploid and
retained this ploidy at passage 38. On the other
hand, it was revealed that 293 cell tumorigenic
potential correlated with number of passages,
that is, low-passage cells (less 52 passages) could
not form tumors in mice in 8 weeks, whereas
tumorigenicity reached 100% when the passage
had exceeded 65 (2 107 cells per injection; 10 of
10 mice had about 0.5 cm3 in size tumors within
2 weeks, after 4 weeks tumor was as large as 2.0
1.5 1.3 cm3) [309]. Nevertheless, there is no
correlation between long-time culturing-induced
tumorigenesis of parental 293 cells and karyotype
instability [304, 309].
The possible reason why prolonged cultiva-
tion drives tumorigenic potential of parental
cells might be deduced from investigations with
NIH3T3 murine fibroblasts, which were used for
transformation assays much more frequently than
any other cells (Table 5). NIH3T3 cells were ob-
tained in 1962 as spontaneously immortalized
cells during long culturing using «3T3 proto-
col» [160]. Rubin documented [310–315] that
spontaneous transformation of NIH3T3 (also of
50 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
Table 4
Transformation of human cells
Indications. HMEC – human mammary epithelial cells; SV7tert cells – derived from angiomyolipoma; FoxM1B –
member of the Forkhead box transcription factor; RPMS1 – ORF of Epstein-Barr virus; PTTG1 – pituitary
tumor-transforming 1; MutCCK2R – cholecystokinin-2 (CCK2)/gastrin receptor intron 4 retained; ESM1 –
endocan, dermatan sulfate proteoglycan; CD74 – major histocompatibility complex, class II invariant chain;
HCCR1 – human cervical cancer oncogene 1; PDX1 – pancreatic and duodenal homeobox-factor 1; ROBO1 –
a member of round about family of transmembrane receptors.
Cell type Immortalizing agent Transforming agent Ref.
Astrocytes
Barrett's epithelial cells
BJ fibroblasts
cen3tel fibroblasts
Colorectal crypt cells
HUVEC
Embryonic esophageal
epithelial cells
FHC fetal colon cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
293 cells
HMEC
HMEC
HMEC
HMEC
MCF-10A
MCF-12A
MCF-10A, 12A
MCF-10A
MCF-10A
MCF-10A
MCF-10A
MCF-10A
Oral epithelial cells
Ovarian epithelial cells
Prostatic epithelial cells
SV7tert cells
HPV E6 + E7 + hTERT
hTERT
Primary normal cells
hTERT
hTERT + SV40 large T antigen
Spontaneous
HPV18 E6 + E7
Primary normal cells
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
Ad5
hTERT
Primary normal cells
hTERT
Primary normal cells
Spontaneous
Spontaneous
Spontaneous
Spontaneous
Spontaneous
Spontaneous
Spontaneous
Spontaneous
E6/E7
SV40 LT + hTERT
SV40
SV40 LT antigen + hTERT
FoxM1B
H-RASG12V + p53 knockdown
E1A + H-RASG12V + MDM2
Culture propagation/spontaneous
MCLR (cyclic hepatotoxin peptide)
COX1 (cyclooxygenase1)
Culture propagation/spontaneous
MET wt or MET mutated
FAP (fibroblast activation protein)
PTTG1
RPMS1
HER2 ( exon 16)
CnB (calcineurin B subunit)
VEGF111, 121, and 165
CCK2R mutated
ESM1
CD74
HCCR1
hBD3 ( -defensin 3)
PDX1
ROBO1
SV40 LT and st, p110 , RASG12V
WNT1
c-MYC transcription factor
MYCT58A
EphA2
ESE1 transcription factor
B-crystallin
CD8-IGF-IR chimera
hGH (human growth hormone)
EGFR + c-SRC
HOXA1 transcription factor
HER2V664E
ErbB2
H-RAS mutated or ErbB2
FGF7 (fibroblast growth factor)
PDGF (platelet derived growth factor)
[199]
[200]
[10]
[24]
[201]
[202]
[203]
[25]
[204]
[205]
[206]
[207]
[208]
[209]
[210]
[211]
[212]
[213]
[214]
[215]
[216]
[217]
[26]
[48]
[27]
[218]
[219]
[220]
[221]
[222]
[109]
[223]
[137]
[224]
[225]
[226]
[227]
51ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
Balb/c 3T3 mouse fibroblasts) cell line in mono-
layer culture is common event especially if cells
were allowed to reach high density in routine
passages (transformation of a diploid line of rat
liver cells is also accelerated by the constraint of
confluence). It also occurs in low density pas-
sages supplemented with low concentrations of
serum. If however the cells are kept continuously
and rapidly multiplying at low density in high se-
rum concentration, not only do they remain non-
transformed but they gradually lose the capacity
for transformation under standard conditions
[310–315]. Grown at different growth conditions
(confluence, concentrations of serum) karyotypes
of NIH3T3 cells analyzed at different passages
(24, 253 and 385 passages) showed that although
the chromosome complement of each of the pas-
sages was near triploid/hypotetraploid (76 ± 2.65,
74 ± 3.2 and 72 ± 2.3, respectively, instead of
the normal 40 chromosomes in mice), there
were more marker (i.e., abnormal) chromosomes
in passage 385 cells than in the earlier passages
[312]. Moreover, every one of the karyotyped
cells of each passage was unique in the precise
distribution of chromosomes. These results sug-
gest that passaging and culture conditions can in-
fluence on aneuploid karyotype of NIH3T3 cells.
It worth recalling that 293 cells also retained near
triploid karyotype through more 100 doublings (6
months in culture) but modal number of chro-
mosomes ranged from 62 to 70, that is, there are
cell populations inside cell line that differ from
each other [304]. In fact, genotypic and pheno-
typic variants constantly appear in the cell line
populations. In addition to the passage number
and the media, the selection of variants is also
modulated by the temperature, humidity, and
CO2 concentration. Some cells can occur to be
Table 5
NIH3T3 cells transformation
Indications. mAChR – muscarinic acetylcholine receptor; CDC42HsF28L – full GTPase activity but sponta-
neous GTP-GDP exchange; EEF1A2 – protein elongation factor 1A2; EGFR – epidermal growth factor receptor;
F-LANa – a member of Derlin family; G o Q205L and G q Q209L – lack of guanosine triphosphatase (GTPase)
activity; G6PD – glucose-6-phosphate dehydrogenase; HCCR1 – human cervical cancer oncogene 1; HCCRBP1 –
human cervical cancer oncogene binding protein 1; IMUP1 and 2 – immortalization-up-regulated protein 1 and 2;
LIN28 and LIN28B – the RNA-binding proteins that block let-7 precursors from being processed to mature
miRNAs and consequently derepress let-7 target genes; Matrigel – extract of basement-membrane proteins;
Midkine – a heparin-binding growth factor; Mina53 – Myc-induced nuclear antigen; PAR - prostate androgen
regulated; PDGF – platelet-derived growth factor; RET – receptor tyrosine kinase for members of the glial cell
line-derived neurotrophic factor family; STAT3-C – constitutively dimerizable STAT3.
Transforming agent Ref. Transforming agent Ref.
mAChR
AKT1myr
AzI (antizyme inhibitor)
BCR-ABL + IL3R
BI1 (Bax inhibitor 1)
CDC42Hs (F28L)
Cyclin T1
EEF1A2
EGFR mutant
FGF (fibroblast growth factor)
F-LANa
G q Q209L, wtG q, G o Q205L
G6PD
HCCR1
HCCRBP1
HPV E7 truncated
IMUP1 and 2
[228]
[229]
[230]
[231]
[232]
[233]
[234]
[235]
[236]
[237–239]
[240]
[241–243]
[244]
[213]
[245]
[246]
[247]
KIT (stem cell factor receptor)
Lin28, Lin28B
Matrigel
Midkine
Mina53
MUC4 (mucin)
Nanog transcription factor
Ornithine decarboxylase
PAR
PDGF
Pleiotrophin
Polyamines
RET mutants
c-SRC + nuclear oncogenes
v-SRC
v-SRC, STAT3-C
14-3-3
[248]
[249]
[250]
[251]
[252]
[253]
[254]
[255, 256]
[257]
[258, 259]
[260]
[261]
[262]
[263]
[264]
[265]
[266]
52 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
more physiologically advantaged to grow during
culture monolayer constrains (e.g. confluence).
Subsequent selection and clonal propagation of
such cells can progressively replace the rest more
growth restrained cells leading eventually to full
replacement with the cell population with superior
growth properties [316]. Selection explains why
culturing 52 passages retained 293 cells growth
restrained (cells did not form tumors in mice),
whereas additional only 13 passages were enough
to make cells fully transformed. To the point, au-
thors stated that 293 cells were propagated as the
cells grown to a 90 % monolayer [309]. Trans-
formation can arise by the continuous fluctuation
of growth states within cells, accompanied by the
progressive selection of those states best suited to
function under the selecting constraint [310–315].
The selection may foster cells carrying alterations
that confer the capability to proliferate and survive
more effectively than their neighbors [198]. Cell-
cell contact interactions can conditionally deter-
mine suppression or selection of the neoplastic
phenotype [315]. It is selection that plays a major
role in the spontaneous neoplastic transformation
of cells in culture [310–316].
Selection and evolution of cells in vitro and in
vivo is universe phenomenon. Nielsen and Briand
[317] demonstrated chromosome abnormalities
and karyotypic evolution in a nontumorigenic
(tested in nude mice) and noninvasive (tested
in vitro), spontaneously immortalized cell line
HMT-3522, derived from a fibrocystic breast le-
sion. During 205 passages, gain and loss of mark-
ers, loss of normal chromosomes, and duplica-
tion of the chromosome complement could be
demonstrated. The variability increased during
in vitro growth. This variability led to cells with
different growth capacities from which sidelines
might be selected and become stem lines. Selec-
tion in both directions (non-tumorigenic cells to
tumorigenic and vice versa) was also described
[154]. This work is of special interest and all ob-
servations documented by authors are presented
here. The karyotypic changes were associated
with the spontaneous acquisition of tumorigenic-
ity in an immortalized human bronchial epithe-
lial cell line NL20, which had been established
by transfection of human bronchial epithelial
cells with the SV40 T-antigen. When cells from
passage 184 were inoculated into nude mice, a
transplantable tumor was obtained. Subsequent
passages of the NL20 cells in vitro did not yield
further tumors by passage 205. Furthermore, the
original tumorigenic NL20T cells lost the neo-
plastic phenotype after 25 passages in vitro and
reverted to the nontumorigenic karyotype ob-
served at passage 189. In contrast to the loss
of the tumorigenic phenotype and karyotype,
which occurred with in vitro passaging of the
original tumor, when the NL20T cells were pas-
saged in other nude mice, they continued to give
rise to tumors; cells from the secondary tumors
(NL20T-A cells) maintained a stable karyotype
and remained tumorigenic even after 64 passages
in vitro. A mixture of 10 % tumorigenic NL20T-
A and 90 % nontumorigenic NL20 cells formed
tumors in nude mice when cultured in vitro on
fibronectin, but not on plastic; cytogenetic analy-
sis demonstrated that the tumors and cell cultures
were composed of tumorigenic NL20T-A cells,
whereas cells cultured on plastic were identical to
the nontumorigenic NL20 cells. Thus, neoplastic
transformation in original cell line arose from in
vivo selection of a small mutant clone, which had
arisen in culture and was subsequently selected
in vivo but was lost in in vitro culture [154]. The
degree of karyotype heterogeneity determines
selection rate and correlates with tumor latent
period [318]. The karyotypes of tumors formed
by spontaneously transformed Chinese hamster
cells of high tumorigenic potential after a short
latent period were similar to each other and to
the injected cells. The karyotypes of tumors from
cells of low tumorigenic potential and long latent
periods were diverse, however. No chromosome
aberration was common to every tumor. These
results suggested that preneoplastic cells whose
phenotypes were not directly capable of tumor
formation could progress in vivo and that karyo-
type instability played an important role in pro-
viding cell variants for tumor progression [318].
NIH3T3 cells were fully transformed (showed
both anchorage independent growth in soft agar
and tumor formation in mice) by a number of
transforming agents depicted in Table 5. Actu-
ally, as Rubin emphasized «the effectiveness of
the NIH3T3 cell line as a target for demonstrat-
ing the transforming capacity of oncogenes de-
pends on its partially transformed state, which
needs only a nudge from an added oncogene to
53ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
progress to more advanced transformation» [315,
319]. Unfortunately, only several works traced
karyotype changes accompanied by oncogene
transformation. For example, karyotypic analysis
of parent NIH3T3 cells and NIH3T3 contain-
ing an activated N-RAS oncogene showed that,
although the modal chromosome number was
comparable for both cell types, number of un-
stable chromosomes and forms of abnormalities
were different [320]. Another work demonstrated
that parental NIH3T3 cells contained 71 chro-
mosomes (hypotetraploid), whereas EJ-NIH3T3
(NIH3T3 cell line carrying the transfected human
activated H-RAS sequence of EJ human blad-
der carcinoma cells) contained 60 chromosomes
(triploid) [321]. Moreover, when the latter cells
were treated with mutagenes (ethyl methane-
sulfonate and 8-azaguanine) and mutant clones
were selected, they were resistant to retransfor-
mation by Kirsten sarcoma virus, DNA from EJ-
NIH 3T3 cells, H-RAS, v-SRC, v-MOS, simian
virus 40 large T-antigen, or polyomavirus middle
T antigen [321]. Karyotype analysis showed that
resistant clones had hyperpentaploid karyotypes
(103 ± 9.7 chromosomes) [321]. Transfection
of vector containing the mitochondrial D-loop
gene from colorectal cancer cell line SW480
into NIH3T3 cells resulted in that NIH3T3 cells
had significantly greater percentage of multi-
ploid and aberrant chromosomes than control
NIH3T3, and this correlated with ability to form
colonies in soft agar [322]. Also, other group of
investigations suggests that for stable transforma-
tion profound changes in genome must occur
[232, 238, 246, 323].
MCF10A cell line is spontaneously immortal-
ized human mammary epithelial cells with near-
diploid karyotype harboring a number of chro-
mosome abnormalities. Besides being frequently
used in in vitro transformation assays (Table 4),
MCF10A cell line was used for comprehensive
analysis of the MCF10A series of cell lines rep-
resenting progression towards obvious malignancy
[165, 324]. The MCF10A progression model con-
sists of three directly derived cell lines: the spon-
taneously immortalized MCF10A cells (do not
show any characteristics of invasiveness or tumor
formation), MCF10AT1 cells (MCF10A cells
transformed by H-RAS), and MCF10CA1a cells
(obtained from tumor in immunodeficient mice
after xenograft transplantation of MCF10AT1
cells) [165, 324]. 47 chromosomes were found
in MCF10A (gained additional chromosome 8)
and the MCF10AT1 cell lines (additional chro-
mosome 8 was deleted, but chromosome 9 was
gained), whereas the malignant MCF10CA1a cell
line had 50 chromosomes [165]. Four marker
chromosomes were identified in MCF10A and
MCF10AT1 and nine in the malignant MCF-
10CA1a cell line [165]. Spectral karyotyping anal-
ysis showed that the premalignant MCF10AT1
gained additional translocations to the MCF10A,
whereas the malignant MCF10CA1a had more
translocations extra to both MCF10A and MC-
F10AT1 [165]. Array comparative genome hy-
bridization (aCGH) showed that MCF10A had
a number of gains and losses of different chro-
mosome regions and progression towards full ma-
lignancy was accompanied by much more severe
genomic aberrations. Importantly, regions of ge-
nomic loss/gain overlapped only partially among
these three cell lines [165]. Another investigation
exploiting the same model (MCF10A series of cell
lines) confirmed the stepwise genome changes ac-
companying progression to full malignancy [324].
Moreover, combining SNP array with Gene Array
authors showed correlation between DNA copy
number gains and increased expression levels for
genes located in these regions [324].
Analysis of tumorigenic potential of established
seven VERO cell line strains (African green mon-
key kidney cells, the normal chromosome number
is 60), of which 1 strain was hypotetraploid and the
rest strains were hypodiploid, and 3 strains of HeLa
cell line (all strains were hyperdiploid) showed that
the cell strains were comparatively stable in terms
of their heritable characters [325]. There were only
little significant changes between passages but the
tumorigenicity of strains was different among dif-
ferent karyotypic cells (from 7 VERO strains 2 ap-
peared tumorigenic: 73 ± 3, and 68 ± 3 or 65 ± 4,
hyperdiploids and 5 were nontumorigenic: 54 ± 2,
55 ± 2, 54 ± 2, 54 ± 2, 54 ± 2, all hypodiploids),
all HeLa strains were hyperdiploid and tumori-
genic [325]. The chromosome number variation
of strains had positive relationships with their car-
cinogenesis and the chromosome number varia-
tion of cell line could be significantly changed
when it developed to tumor in nude mice [325].
These observations were confirmed in experiments
54 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
with other cell cultures. Meningioma cells with
multiple chromosomal abnormalities grew rapidly
in vitro and induced tumors in 49 of 50 animals,
whereas with simple karyotypes (less or 1 chromo-
somal abnormality) grew slowly in vitro and gave
small, nongrowing tumors in mice [326]. Also, the
average number of chromosomes in 293N cells (a
subline of just obtained 293 cells in 1977 year de-
rived from a tumor developed in a nude mouse)
was significantly lower than for the parental line [78].
Thus, progression of immortalized or pre-neo-
plastic cells towards obvious malignancy is always
accompanied by karyotype changes. The degree and
diversity of karyotype changes determine tumori-
genic potential of cell culture and latency period
of tumor formation necessary for creation and/or
selection of the most competitive malignant cells.
Gene copy number, mRNA and protein lev-
els relationships in tumor cells. Actually, primary
tumor cells and cancer cell lines are always polip-
loid/aneuploid, and have karyotypes ranging from
40 to 60 but occasionally exceeding 70 or more
chromosomes [327]. Moreover, numerical large-
scale and focal chromosome aberrations (losses/
gains/deletions/ translocations) were found in all
samples of each type and subtype of tumors ana-
lyzed up to now (Table 6). Roschke et al. [328]
using spectral karyotyping provided a description
of the chromosomal complement of the NCI-60
cell line panel developed by the National Cancer
Institute (NCI) for in vitro anticancer drug screen-
ing and reflecting diverse cell lineages (lung, renal,
colorectal, ovarian, breast, prostate, central nervous
system, melanoma, and hematological malignan-
cies). 23 cell lines were identified as near-diploid (a
chromosome modal number between 35 and 57),
22 as near-triploid (the chromosome modal num-
ber between 58 and 80), 13 as near-tetraploid (a
chromosome modal number between 81 and 103),
and 1 as near-pentaploid (chromosome modal
number between 104 and 126) on the basis of the
International System for Chromosome Nomen-
clature. The range of numerical changes (clonal
chromosome gains and losses) ranged from 1 to 28.
Number of structurally rearranged chromosomes
(contained translocations, deletions, duplications,
insertions, inversions, or homogeneously staining
regions) ranged from 1 to 45 (38 cell lines had 10
and more structurally rearranged chromosomes). In
addition, in 24 of the 59 cell lines ploidy heteroge-
neity was found (i.e., if the majority of cells had a
near-triploid karyotype there might be an additional
small populations of cells with a near-pentaploid
or near-hexaploid count, or, in few cases, with a
near-diploid count). Chromosome numerical and
structural heterogeneity between cells in the same
cell line was also documented [328].
Later, this NCI-60 cell line panel was used
to elucidate correlation between gene copy num-
ber and mRNA levels for the same gene. The
data showed a generally positive correlation be-
tween a given gene’s copy number and its ex-
pression at the mRNA level supporting the gen-
eralization that DNA copy number is one factor
(among others) that can influence gene expres-
sion [329]. In another work authors performed a
global analysis of both mRNA and protein levels
based on sequence-based transcriptome analysis
(RNA-seq) and SILAC-based mass spectrom-
etry analysis [330]. The study was performed
in three functionally different human cell lines
(the glioblastoma cell line U251MG, the epi-
dermoid carcinoma cell line A431 and the osteo-
sarcoma cell line U2OS). The changes of mRNA
and protein levels in the cell lines using SILAC
and RNA ratios showed high correlations, even
though the genome-wide dynamic range was sub-
stantially higher for the proteins as compared with
the transcripts [330]. Also, there was a moderate
but significant correlation between global mRNA
(RNA-seq) and protein levels (SILAC) of 1710
genes affected by amplification or deletion (SNP
and CGH arrays) in seven human metastatic mela-
noma cell lines [331]. Whether does it mean that
the end point of gene expression, the level of pro-
tein, is affected proportionally to copy number of
gene in tumor cells? Measurement of expression
levels of 6735 proteins was directly compared to
the gene copy number in MCF7 breast cancer cell
line [332]. Authors found that in the majority of
cases, there was no direct correspondence between
the gene copy number change and the correspond-
ing protein change. Nevertheless, proteins encoded
by amplified oncogenes were often overexpressed,
while adjacent amplified genes, which presumably
did not promote growth and survival, were attenua-
ted [332]. Furthermore, authors revealed that the
proteins of such complexes as the proteasome, ribo-
some, spliceosome, and NADH dehydrogenase al-
ways maintained equal protein ratios, despite varia-
55ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
tion in the gene copy number of their subunits. This
was strictly true for the core complexes components,
but to a lesser degree for peripheral proteins, which
could also be involved in other processes [332]. In-
terestingly, levels of protein expression in aneuploid
yeast strains largely scale with chromosome copy
numbers, following the same trend as that observed
for the transcriptome [333]. Thus, eventually to be
definitely concluded, relationships between gene
copy number, mRNA level and protein level of indi-
vidual genes across the whole cancer genome should
be analyzed. It would give the comprehensive under-
standing to which degree regulation of gene expres-
sion on different levels operates in tumor cells and
which groups of genes are predominantly imposed
on such regulation.
Inter- and intratumor heterogeneity. It is sup-
posed that common (clonal) chromosome changes
are the «drivers» of neoplastic transformation
whereas rare chromosome changes (non-clonal)
are likely the «passengers» in this process, which
may be either nonfunctional or functional but con-
stitute secondary events [3, 374, 375]. Nevertheless,
non-clonal aberrations reflect the significant tumor
feature: genome/chromosome instability and, as a
consequence, inter- and intratumor genome het-
erogeneity [376]. Importantly, the main determi-
nant of the ability of a population to evolve is the
extent of heritable variation within the population
[377]. Numerous studies have proved that intra-tu-
mor genetic heterogeneity/clonal diversity is a key
force driving transformation and tumor evolution
(Stepanenko and Kavsan, in preparation).
Nobusawa et al. [374] have analysed by
aCGH separate tumor areas of 14 primary glio-
blastomas (total, 41 tumor areas). They revealed
Table 6
Tumors and cell lines with multiple chromosomal abnormalities
Indications. aCGH – array comparative genome hybridization; SNP array – single nucleotide polymorphism
array; Sequencing – massively parallel paired-end sequencing; MLDPA – multiple ligation-dependent probe
amplification.
Cancer type Method
Number
of samples Ref.
Acute myeloid leukemia
Bladder cancer
Bladder carcinomas
Breast cancer
Cervical carcinomas
Colorectal carcinomas and adenomas
Endometrial carcinomas and carcinosarcomas
Ewing’s cancer
Gastric cancer
Germ cell cancer
Glioma
Head and neck squamous cell carcinomas
Lung cancer
Myelodysplastic syndromes and related
myeloid malignancies
Oral carcinomas
Oropharynx and hypopharynx squamous cell
carcinomas
Ovarian epithelial tumors
Pancreatic carcinomas
Prostate tumours
Thyroid carcinomas and adenomas
29 different tissues
26 different tissues
aCGH
aCGH
aCGH
aCGH, SNP array, sequencing,
aCGH, SNP array
aCGH, SNP array
aCGH and karyotyping
aCGH, spectral karyotyping
aCGH
aCGH
aCGH, SNP array, karyotyping
aCGH
Sequencing, SNP array, karyotyping
SNP array, karyotyping
aCGH
aCGH
aCGH
aCGH, sequencing
Sequencing
aCGH
aCGH
SNP array
17
22
109
1143
40
129
82
7
31
24
248
43
80
430
60
20
47
37
7
28
598
3131
[334 ]
[335]
[336, 337]
[338–342]
[343, 344]
[345–348]
[349]
[350]
[351]
[352]
[353–359]
[360, 361]
[362, 363]
[364]
[365]
[366]
[367]
[368, 369]
[370]
[371]
[372]
[373]
56 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
that chromosomal imbalances significantly dif-
fered among glioblastomas. In addition, there
were numerous tumor area-specific genomic im-
balances. Analysis of disseminated single cells in
minimal residual disease has shown that there is a
high level of genomic heterogeneity within indi-
vidual lesions as well as between primary tumors
and metastatic cells [376]. Giving comments on
reports of breast cancer genomes analyses with
high-throughput genomics thechnics [339, 342],
Swanton et al. [378] concluded that results from
these studies had revealed «perplexing breast can-
cer genome complexity with very few aberrations
occurring in common between breast cancers. In
addition, such complexity is compounded by evi-
dence of profound genomic heterogeneity within
individual breast tumors (intratumoral hetero-
geneity), where multiple tumor subpopulations
have been identified, each with distinct genomic
profiles heterogeneity occurring within individual
breast cancers». Moreover, recurrent tumors al-
ways show appearance of new chromosome im-
balances and gene mutations distinct from those,
which were observed in most cells of a primary
tumor but could be harbored by a small group of
cells within a primary tumor or acquired de novo
[371, 377, 380–389].
Intratumor genomic heterogeneity is created
and fostered by chromosome instability (CIN. Al-
though defects in chromosome cohesion, kinet-
ochore-microtubule misattachments, assembly
of multipolar mitotic spindles [182, 390–396],
translocations containing breakpoints within fra-
gile sites [397], satellite repeats in heterochro-
matin [398], cell-in-cell formation by entosis (as
a result, cytokinesis frequently fails, generating
binucleate cells that produce aneuploid cell lin-
eages) [399], random fragmentation of the entire
chromosome (chromothripsis) in which chromo-
somes are broken into many pieces and then
randomly stitched back together [400, 401] can
contribute to CIN, in cancer cells mechanism
of centrosome amplification and clustering is
proposed to be the major contributor to CIN.
Importantly, there is compelling evidence that
diverse oncogenes or carcinogenes induce cen-
trosome deregulation and CIN (Stepanenko and
Kavsan, in preparation).
Inter- and intratumor heterogeneity of gene
mutations was also revealed by sequencing of all
protein coding genes in several solid tumors, in-
cluding glioblastomas, colorectal, pancreatic and
breast cancers. It was found that individual solid
organ tumors harbor approximately 40–80 clonal
mutations per tumor in the coding regions of dif-
ferent genes, and although a few of these genes are
mutated in a high proportion of tumors, the preva-
lence at which the majority are mutated among dif-
ferent tumors of the same cancer type is low [376,
377, 402]. 2576 somatic mutations were identified
across 1800 megabases of DNA representing 1507
coding genes from 441 tumors comprising breast,
lung, ovarian and prostate cancer types and sub-
types [403]. Authors found that mutation rates and
the sets of mutated genes varied substantially across
tumor types and subtypes. Results of sequencing
COLO-829 cancer cell line derived, before treat-
ment, from a metastasis of a malignant melanoma
demonstrated a total of 292 somatic base substitu-
tions in protein-coding sequences [404]. As Fox et
al. stated «each tumor displays a unique and diverse
profile of mutated genes, but no new prevalently
mutated genes are identified... Within an individual
neoplasm, a few mutations are present throughout
the population, a greater number are present in mi-
nority subclones, and the majority is found in only
one or a few cells» [376]. Actually, if to calculate all
abnormalities of noncoding genome regions, there
are usually between 1000 and 10000 somatic sub-
stitutions in the genomes of most adult tumors, in-
cluding breast, ovary, colorectal, pancreas cancers,
and glioma [3]. All these data imply that tumors are
really «oncogene addicted», but it has not revealed
a possible «Achilles’ heel» within the cancer cell
that can be exploited therapeutically [3, 405], be-
cause «instead of long-anticipated common muta-
tions, a large number of stochastic gene mutations
were detected for each individual with the same
cancer type» [406].
Chromosome instability and drug resistance.
Cancer cells rapidly acquire resistance against
numerous cytotoxic drugs or are even intrinsically
resistant [379]. The chromosomes of cancer cells
are extremely unstable compared to those of normal
cells: 1 in 100 highly aneuploid human cancer cells
loses or gains or rearranges a chromosome per
cell generation [407]. Genetic variation within a
cancer cell population reflects dynamics of clonal
evolution and, importantly, serves as a reservoir
of genetic diversity from which therapy-resistant
57ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
clones may arise [376]. In vitro studies have con-
firmed that CIN cells acquire multidrug resistance
at an accelerated rate compared with diploid cells
resulting from the selection pressure influenced by
drug exposure. Moreover, mouse CIN cells be-
came multidrug resistant even after deletion of all
known multidrug resistance genes [319, 379]. To
identify distinct therapeutic approaches to specifi-
cally limit the growth of CIN tumors, Lee et al.
[408] focused on a panel of colorectal cancer cell
lines, previously classified as either chromosom-
ally unstable, CIN (+), or diploid/near-diploid,
CIN (–), and treated them individually with a
library of kinase inhibitors targeting components
of signal transduction, cell cycle, and transmem-
brane receptor signaling pathways. CIN (+) cell
lines displayed significant intrinsic multidrug re-
sistance compared with CIN (–) cancer cell lines,
and this seemed to be independent of somatic mu-
tation status and proliferation rate [408].
According to Duesberg et al., karyotype plays
the central role in drug resistance [379]. «When
cancer cells acquire resistance against drugs, they
acquire new karyotypic alterations and/or they
lose old ones». Indeed, gene expression profiles
of drug resistant cells differ from those of parental
drug sensitive cells in the over- or underexpression
of hundreds to thousands genes [379]. Comparison
of the structures of the puromycin resistance-
specific chromosomal alterations in four different
human colon cancer lines indicates, that most
but not all of resistance-specific chromosomal
alterations were unique for each cancer cell [379].
Drug resistance correlates with chromosomal
alterations [319, 407]. It is generated de novo in
cancer cells by chromosome re-assortments [379].
The resulting level of resistance is proportional to
the numbers of resistance-specific chromosomal
alterations or «tumor heterogeneity» [379]. In the
presence of cytotoxic drugs resistance-specific
alterations are selected from the resultining vari-
ants by classical Darwinian mechanisms [379].
Ñhromosome instability, intertumoral and intra-
tumoral heterogeneity present a challenge to per-
sonalized therapeutic approaches [378].
Conclusion. The intense searching for the ab-
normal genes influencing the development of hu-
man cancer revealed about 200000 somatic mu-
tations in cancer genomes (COSMIC database)
since the first somatic mutation that was found in
H-RAS the quarter of a century ago [198]. Hun-
dreds of genes are being considered and dozens
genes/proteins have been used already as poten-
tial drug targets in clinical trials. However, at
present benefits from oncogene directed therapy
are still moderate. Large-scale tumor genome se-
quencing have failed to reveal «universal» can-
cer genes and, instead, «large numbers of diverse
mutations have been identified dominating the
cancer genome landscape» [406]. «A future of
multiple targeted therapies and patient stratifica-
tion, based on a mutational signature of defined
key genes for each cancer type, seems less hope-
ful than initially anticipated» [376].
Now it is clear that «cancer progression is a
stochastic process both at the genome and gene
level, and is not a stepwise process defined by
sequential genetic aberrations. Stochastic pro-
cess frequently occurs prior to the key stages of
immortalization, transformation and metasta-
sis and results in inability to detect type- and
stage-specific recurrent aberrations in solid tu-
mors» [406]. Numerous somatic rearrangements,
including whole chromosome and copy number
gains and losses, chromosome translocations,
and gene mutations participate in establishing
the malignant cell phenotype. «Multiple rounds
of proliferation, often counter-balanced by cell
death, are required to produce macroscopic tu-
mors, and genomic instability, observed in most
cancers, is expected to constantly produce new
mutations, which serve as raw material on which
tumor evolution can work» [409]. The selection
may weed out cells that have acquired deleteri-
ous mutations or it may foster cells carrying al-
terations that confer the capability to proliferate
and survive more effectively than their neighbors
[198]. A single cell can acquire a set of suffi-
ciently advantageous mutations that allows it to
proliferate autonomously, invade tissues and me-
tastasize [198].
CIN is the most common genetic abnormality
of cancer cells and tumorigenic cell lines. The fre-
quent losses and gains of whole chromosomes dur-
ing cell divisions in CIN cancer cells trigger rapid
alterations in gene dosage [379]. In vitro studies
have confirmed that CIN cells acquire multi-drug
resistance at an accelerated rate compared with
diploid cells [379]. Furthermore, although most
cancers are of monoclonal origin, the expansion of
58 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
the population size, which occurs after malignant
transformation, coupled with the constant acquisi-
tion of mutations promotes the diversion into sub-
clones and a dramatic increase in genetic tumor
heterogeneity [377]. High genetic heterogeneity
of tumors means high probability of pre-existent
clones that are resistant to therapeutic intervention
and can be selected by therapy resulting in therapy
failure [409]. Severe genome rearrangements and
intratumoral heterogeneity challenge oncogene di-
rected therapy, while chromosome instability and
karyotype evolution make each tumor «a moving
rather than frozen» target.
Thus, a main driver of evolutionary adaptation
during drug treatment is the genetic heterogeneity,
which is fostered by CIN. New tools are necessary
to study heterogeneity and to analyze changes in
heterogeneity and clonal composition during drug
treatment [410–412]. This would allow new in-
sights into these processes and provide the basis
to improve therapeutic outcomes based on tumor
evolution and the specific targeting of distinct ge-
nomic instability mechanisms [377, 413].
This work was supported in part by grant SFFR
F46/457-2011 «State key laboratory of molecular
and cellular biology» and by budget topic of the
Institute of Molecular Biology and Genetics, NAS
of Ukraine, «Functional characterization of genes
associated with initiation and progression of glial
and connective tissue tumors of human brain».
À.À. Ñòåïàíåíêî, Â.Ì. Êàâñàí
ÈÌÌÎÐÒÀËÈÇÀÖÈß
È ÇËÎÊÀ×ÅÑÒÂÅÍÍÀß ÒÐÀÍÑÔÎÐÌÀÖÈß
ÝÓÊÀÐÈÎÒÈ×ÅÑÊÈÕ ÊËÅÒÎÊ
×òîáû ñòàòü ïîëíîñòüþ òðàíñôîðìèðîâàííîé îïó-
õîëåâîé êëåòêîé, íîðìàëüíàÿ êëåòêà äîëæíà ïðå-
îäîëåòü ðÿä âíóòðåííèõ êëåòî÷íûõ áàðüåðîâ è ïðè-
îáðåñòè áîëüøîå ÷èñëî õðîìîñîìíûõ èçìåíåíèé.
Ïåðâûì è íåîáõîäèìûì øàãîì â çëîêà÷åñòâåííîé
òðàíñôîðìàöèè ÿâëÿåòñÿ ïðåîäîëåíèå ñòàðåíèÿ, èëè
èììîðòàëèçàöèÿ êëåòêè. Èììîðòàëèçèðîâàííûå
êëåòêè ìîãóò áåñêîíå÷íî äîëãî ïðîëèôåðèðîâàòü â
ïðèñóòñòâèè ðîñòîâûõ ôàêòîðîâ è ïèòàòåëüíûõ âå-
ùåñòâ. Èììîðòàëèçèðîâàííûå êëåòêè íèêîãäà íå
èìåþò íîðìàëüíîãî äèïëîèäíîãî êàðèîòèïà, xoòÿ
âî âðåìÿ ðîñòà ïîäâåðãàþòñÿ êîíòàêòíîìó èíãèáè-
ðîâàíèþ, íå ôîðìèðóþò êîëîíèé â ìÿãêîì àãàðå (ò.å.
çàâèñèìûé îò ïîäëîæêè ðîñò) è íå ôîðìèðóþò îïóõîëåé
ïðè ââåäåíèè èììóíîäåôèöèòíûì ìûøàì. Âñå ýòè
ñâîéñòâà ìîãóò áûòü ïðèîáðåòåíû ñ äîïîëíèòåëüíû-
ìè õðîìîñîìíûìè èçìåíåíèÿìè. Ìíîæåñòâåííûå ãå-
íåòè÷åñêèå èçìåíåíèÿ, âêëþ÷àÿ ïðèîáðåòåíèå/ïîòå-
ðþ öåëûõ õðîìîñîì èëè îòäåëüíûõ ó÷àñòêîâ/ëîêó-
ñîâ, òðàíñëîêàöèþ õðîìîñîì è ãåííûå ìóòàöèè,
íåîáõîäèìû äëÿ óñòàíîâëåíèÿ òðàíñôîðìèðîâàííî-
ãî ôåíîòèïà. Ïðîöåññ êëåòî÷íîé òðàíñôîðìàöèè äî-
ñòàòî÷íî õîðîøî èçó÷åí íàêëåòî÷íûõ êóëüòóðàõ
in vitro. Áîëüøèíñòâî ýêñïåðèìåíòîâ, âûÿâèâøèõ
òðàíñôîðìèðóþùóþ ñïîñîáíîñòü ãåíîâ (îíêîãåíîâ),
íàäýêñïðåññèðîàííûõ è/èëè ìóòèðîâàííûõ â îïóõî-
ëÿõ, áûëî âûïîëíåíî ñ èñïîëüçîâàíèåì òàêèõ êëåòî÷-
íûõ êóëüòóð, êàê ìûøèíûå ýìáðèîíàëüíûå ôèáðîáëà-
ñòû (MEFs), ìûøèíàÿ êëåòî÷íàÿ ëèíèÿ ôèáðîáëàñòîâ
NIH3T3, êëåòî÷íàÿ ëèíèÿ ÷åëîâå÷åñêîé ýìáðèî-
íàëüíîé ïî÷êè 293 (293 êëåòêè) è ýïèòåëèàëüíûå
êëåòî÷íûå ëèíèè ìîëî÷íîé æåëåçû ÷åëîâåêà (ãëàâ-
íûì îáðàçîì, HMECs è MCF10A), êîòîðûå ïðåäñòàâ-
ëÿþò ñîáîé èììîðòàëèçèðîâàííûå êëåòêè (êðî-
ìå ïåðâè÷íûõ ìûøèíûõ ôèáðîáëàñòîâ) ñ èçìåíåí-
íûìè ãåíîìàìè (ïîëè-/àíåóïëîèäû ñî çíà÷èòåëü-
íûìè õðîìîñîìíûìè ïåðåñòðîéêàìè) è ñêëîííûå ê
ïîëíîé çëîêà÷åñòâåííîé òðàíñôîðìàöèè ïðè êóëü-
òèâèðîâàíèÿ. Íåäàâíî îáíîâëåííûé ñïèñîê îíêî-
ãåíîâ âêëþ÷àåò áîëåå 467 ãåíîâ, êîòîðûå, êàê ïî-
ëàãàþò, âîâëå÷åíû â ðàçâèòèå îïóõîëè, êîãäà ñî-
îòâåòñòâåííûì îáðàçîì èçìåíåíû (òî÷êîâûå ìóòà-
öèè, äåëåöèè, òðàíñëîêàöèè èëè àìïëèôèêàöèè).
Îäíàêî èññëåäîâàíèÿ íà ìûøàõ ñâèäåòåëüñòâóþò,
÷òî áîëåå 3000 ãåíîâ ìîãóò âíîñèòü âêëàä â
ðàçâèòèå îïóõîëè. Öåëüþ íàñòîÿùåãî îáçîðà ÿâ-
ëÿåòñÿ ïîíÿòü ìåõàíèçìû êëåòî÷íîé èììîðòàëè-
çàöèè ðàçëè÷íûìè «èììîðòàëèçóþùèìè àãåíòàìè»,
îíêîãåí-èíäóöèðóåìîé êëåòî÷íîé òðàíñôîðìàöèè
èììîðòàëèçèðîâàííûõ êëåòîê è óìåðåííûé îòâåò íà
òåðàïèþ èç-çà «ñêëîííîñòè» îïóõîëè ê ïðèîáðåòå-
íèþ ìíîãî÷èñëåííûõ ãåííûõ è õðîìîñîìíûõ èçìå-
íåíèé, âíóòðè- è ìåæîïóõîëåâîé ãåòåðîãåííîñòè.
Î.À. Ñòåïàíåíêî, Â.Ì. Êàâñàí
²ÌÎÐÒÀ˲ÇÀÖ²ß
ÒÀ ÇËÎßʲÑÍÀ ÒÐÀÍÑÔÎÐÌÀÖ²ß
ÅÓÊÀвÎÒÈ×ÍÈÕ Ê˲ÒÈÍ
Ùîá ñòàòè ïîâí³ñòþ òðàíñôîðìîâàíîþ ïóõ-
ëèííîþ êë³òèíîþ, íîðìàëüíà êë³òèíà ïîâèííà
ïîäîëàòè íèçêó âíóòð³øí³õ êë³òèííèõ áàð’ºð³â ³
ïðèäáàòè âåëèêó ê³ëüê³ñòü õðîìîñîìíèõ çì³í. Ïåð-
øèì íåîáõ³äíèì êðîêîì ó çëîÿê³ñí³é òðàíñôîðìàö³¿
º ïîäîëàííÿ ñòàð³ííÿ, àáî ³ìîðòàë³çàö³ÿ êë³òèíè.
²ìîðòàë³çîâàí³ êë³òèíè ìîæóòü íåñê³í÷åííî äîâãî
ïðîë³ôåðóâàòè â ïðèñóòíîñò³ ðîñòîâèõ ôàêòîð³â ³
ïîæèâíèõ ðå÷îâèí. ²ìîðòàë³çîâàí³ êë³òèíè ìàéæå
í³êîëè íå ìàþòü íîðìàëüíîãî äèïëî¿äíîãî êàð³î-
òèïó, òèì íå ìåíø âîíè ï³ä ÷àñ ðîñòó ï³ääàþòüñÿ
êîíòàêòíîìó ³íã³áóâàííþ, íå ôîðìóþòü êîëîí³é â
ì’ÿêîìó àãàð³ (òîáòî çàëåæíå â³ä ï³äêëàäêè çðîñ-
59ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
òàííÿ) ³ íå ôîðìóþòü ïóõëèí ïðè ââåäåíí³ ³ìóíî-
äåô³öèòíèì ìèøàì. Âñ³ ö³ âëàñòèâîñò³ ñòàá³ëüíî
ìîæóòü áóòè ïðèäáàí³ ç äîäàòêîâèìè õðîìîñîì-
íèìè çì³íàìè. Ìíîæèíí³ ãåíåòè÷í³ çì³íè, âêëþ÷à-
þ÷è íàáóòòÿ àáî âòðàòó ö³ëèõ õðîìîñîì àáî îêðåìèõ
ä³ëÿíîê/ëîêóñ³â, òðàíñëîêàö³ÿ õðîìîñîì ³ ãåíí³ ìó-
òàö³¿, º íåîáõ³äíèìè äëÿ âñòàíîâëåííÿ òðàíñôîðìî-
âàíîãî ôåíîòèïó. Ïðîöåñ êë³òèííî¿ òðàíñôîðìàö³¿
äîñèòü äîáðå âèâ÷åíèé íà êë³òèííèõ êóëüòóðàõ
in vitro. Á³ëüø³ñòü åêñïåðèìåíò³â ç âèÿâëåííÿ
òðàíñôîðìóþ÷î¿ çäàòíîñò³ ãåí³â (îíêîãåí³â), íàä-
åêñïðåñîâàíèõ òà/àáî ìóòîâàíèõ â ïóõëèíàõ, áóëî
âèêîíàíî ç âèêîðèñòàííÿì òàêèõ êë³òèííèõ êóëü-
òóð, ÿê ìèøà÷³ åìáð³îíàëüí³ ô³áðîáëàñòè (MEFs),
êë³òèííà ë³í³ÿ ìèøà÷èx ô³áðîáëàñò³â NIH3T3,
êë³òèííà ë³í³ÿ ëþäñüêî¿ åìáð³îíàëüíî¿ íèðêè 293
(293 êë³òèíè) ³ åï³òåë³àëüí³ êë³òèíí³ ë³í³¿ ìî-
ëî÷íî¿ çàëîçè ëþäèíè (ãîëîâíèì ÷èíîì, HMECs
³ MCF10A), ÿê³ ïðåäñòàâëÿþòü ñîáîþ ³ìîðòàë³-
çîâàí³ êë³òèíè (êð³ì ïåðâèííèõ ìèøà÷èõ ô³áðî-
áëàñò³â) ç³ çì³ííèìè êàð³îòèïàìè (ïîë³-/àíåó-
ïëî¿äè ç³ çíà÷íèìè õðîìîñîìíèìè ïåðåáóäîâàìè)
³ ñõèëüí³ äî ïîâíî¿ çëîÿê³ñíî¿ òðàíñôîðìàö³¿ ïðè
êóëüòèâóâàíí³. Íåùîäàâíî îíîâëåíèé ñïèñîê îíêî-
ãåí³â âêëþ÷ຠïîíàä 467 ãåí³â, ùî çàëó÷åí³, ÿê
ââàæàþòü, äî ðîçâèòêó ïóõëèíè, êîëè â³äïîâ³äíèì
÷èíîì çì³íåí³ (òî÷êîâ³ ìóòàö³¿, äåëåö³¿, òðàíñëîêàö³¿
àáî àìïë³ô³êàö³¿). Îäíàê äîñë³äæåííÿ íà ìèøàõ
ñâ³ä÷àòü ïðîòå, ùî ïîíàä 3000 ãåí³â ìîæóòü ðî-
áèòè âíåñîê ó ðîçâèòîê ïóõëèíè. Ìåòà äàíîãî
îãëÿäó çðîçóì³òè ìåõàí³çìè êë³òèííî¿ ³ìîðòàë³çàö³¿
ð³çíèìè «³ìîðòàë³çóþ÷èìè àãåíòàìè», îíêîãåí-
³íäóêîâàíî¿ êë³òèííî¿ òðàíñôîðìàö³¿ ³ìîðòàë³çîâà-
íèõ êë³òèí ³ ïîì³ðíó â³äïîâ³äü íà òåðàï³þ ÷åðåç
«ñõèëüí³ñòü» ïóõëèíè äî ïðèäáàííÿ ÷èñëåííèõ
ãåííèõ òà õðîìîñîìíèõ çì³í òà ãåòåðîãåíí³ñòþ
óñåðåäèí³ ³ ì³æ ïóõëèíàìè.
REFERENCES
1. Kuroki T., Huh N.H. Why are human cells resistant
to malignant cell transformation in vitro? // Jpn. J.
Cancer Res. – 1993. – 84, ¹ 11. – P. 1091–1100.
2. Malumbres M., Barbacid M. RAS oncogenes: the
first 30 years // Nat. Rev. Cancer. – 2003. – 3,
¹ 6. – P. 459–465.
3. Stratton M.R. Exploring the genomes of cancer
cells: progress and promise // Science. – 2011. –
331, ¹ 6024. – P. 1553–1558.
4. Rangarajan A., Weinberg R.A. Comparative biology
of mouse versus human cells: modelling human
cancer in mice // Nat. Rev. Cancer. – 2003. – 3,
¹ 12. – P. 952–959.
5. Sherr C.J., DePinho R.A. Cellular senescence:
mitotic clock or culture shock? // Cell. – 2000. –
102, ¹ 4. – P. 407–410.
6. Hahn W.C., Weinberg R.A. Modelling the mole-
cular circuitry of cancer // Nat. Rev. Cancer. –
2002. – 2, ¹ 5. – P. 331–341.
7. Woo R.A., Poon R.Y. Activated oncogenes promote
and cooperate with chromosomal instability for
neoplastic transformation // Genes Dev. – 2004. –
18, ¹ 11. – P. 1317–1330.
8. Woo R.A., Poon R.Y. Gene mutations and aneu-
ploidy: the instability that causes cancer // Cell
Cycle. – 2004. – 3, ¹ 9. – P. 1101–1103.
9. Blasco M.A., Lee H.W., Hande M.P. et al. Telo-
mere shortening and tumor formation by mouse
cells lacking telomerase RNA // Cell. – 1997. –
91, ¹ 1. – P. 25–34.
10. Seger Y.R., García-Cao M., Piccinin S. et al. Trans-
formation of normal human cells in the absence of
telomerase activation // Cancer Cell. – 2002. – 2,
¹ 5. – P. 401–413.
11. Soo J.K., Mackenzie Ross A.D., Kallenberg D.M.
et al. Malignancy without immortality? Cellular
immortalization as a possible late event in mela-
noma progression // Pigment Cell Melanoma
Res. – 2011. – 24, ¹ 3. – P. 490–503.
12. Kamijo T., Zindy F., Roussel M.F. et al. Tumor sup-
pression at the mouse INK4a locus mediated by
the alternative reading frame product p19ARF //
Cell. – 1997. – 91, ¹ 5. – P. 649–659.
13. Peeper D.S., Shvarts A., Brummelkamp T. et al. A
functional screen identifies hDRIL1 as an
oncogene that rescues RAS-induced senescence //
Nat. Cell Biol. – 2002. – 4, ¹ 2. – P. 148–153.
14. Petrenko O., Zaika A., Moll U.M. deltaNp73 facilitates
cell immortalization and cooperates with oncogenic
Ras in cellular transformation in vivo // Mol. Cell.
Biol. – 2003. – 23, ¹ 16. – P. 5540–5555.
15. Guo F., Zheng Y. Involvement of Rho family GTPases
in p19Arf- and p53-mediated proliferation of pri-
mary mouse embryonic fibroblasts // Mol. Cell.
Biol. – 2004. – 24, ¹ 3. – P. 1426–1438.
16. Maeda T., Hobbs R.M., Merghoub T. et al. Role of
the proto-oncogene Pokemon in cellular trans-
formation and ARF repression // Nature. – 2005. –
433, ¹ 7023. – P. 278–285.
17. Kobayashi K., Era T., Takebe A. et al. ARID3B in-
duces malignant transformation of mouse embryo-
nic fibroblasts and is strongly associated with
malignant neuroblastoma // Cancer Res. – 2006. –
66, ¹ 17. – P. 8331–8336.
18. Sun H., Taneja R. Analysis of transformation and tu-
morigenicity using mouse embryonic fibroblast cells //
Meth. Mol. Biol. – 2007. – 383. – P. 303–310.
19. Ivetac I., Gurung R., Hakim S., et al. Regulation of
PI(3)K/Akt signalling and cellular transformation
by inositol polyphosphate-4-phosphatase-1 //
EMBO Rep. – 2009. – 10, ¹ 5. – P. 487–493.
60 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
20. Spandidos D.A., Wilkie N.M. Malignant transfor-
mation of early passage rodent cells by a single
mutated human oncogene // Nature. – 1984. –
310, ¹ 5977. – P. 469–475.
21. Land H., Chen A.C., Morgenstern J.P. et al. Behavior
of myc and ras oncogenes in transformation of rat
embryo fibroblasts // Mol. Cell Biol. – 1986. – 6,
¹ 6. – P. 1917–1925.
22. Kelekar A., Cole M.D. Immortalization by c-myc,
H-ras, and Ela oncogenes induces differential cel-
lular gene expression and growth factor responses //
Mol. Cell. Biol. – 1987. – 7, ¹ 11. – P. 3899–
3907.
23. Sugiyama K., Otori K., Esumi H. Neoplastic trans-
formation of rat colon epithelial cells by expression
of activated human K-ras // Japan. J. Cancer Res. –
1998. – 89, ¹ 6. – P. 615–625.
24. Zongaro S., de Stanchina E., Colombo T. et al.
Stepwise neoplastic transformation of a telo-
merase immortalized fibroblast cell line // Can-
cer Res. – 2005. – 65, ¹ 24. – P. 11411–11418.
25. Boon E.M., Kovarikova M., Derksen P.W., van der
Neut R. MET signalling in primary colon epithelial
cells leads to increased transformation irrespective
of aberrant Wnt signalling // Brit. J. Cancer. –
2005. – 92, ¹ 6. – P. 1078–1083.
26. Ayyanan A., Civenni G., Ciarloni L. et al. Increased
Wnt signaling triggers oncogenic conversion
of human breast epithelial cells by a Notch-
dependent mechanism // Proc. Nat. Acad. Sci.
USA. – 2006. – 103, ¹ 10. – P. 3799–3804.
27. Thibodeaux C.A., Liu X., Disbrow G.L. et al.
Immortalization and transformation of human
mammary epithelial cells by a tumor-derived Myc
mutant // Breast Cancer Res. Treat. – 2009. –
116, ¹ 2. – P. 281–294.
28. Chapman S., Liu X., Meyers C. et al. Human
keratinocytes are efficiently immortalized by a
Rho kinase inhibitor // J. Clin. Invest. – 2010. –
120, ¹ 7. – P. 2619–2626.
29. Hayflick L. The limited in vitro lifetime of human
diploid cell strains // Exp. Cell Res. – 1965. – 37. –
P. 614–636.
30. Ohtani N., Mann D.J., Hara E. Cellular senescence:
its role in tumor suppression and aging // Cancer
Sci. – 2009. – 100, ¹ 5. – P. 792–797.
31. Lleonart M.E., Artero-Castro A., Kondoh H. Senes-
cence induction: a possible cancer therapy // Mol.
Cancer. – 2009. – 8, ¹ 1. – P. 3.
32. Garbe J.C., Bhattacharya S., Merchant B. et al.
Molecular distinctions between stasis and telomere
attrition senescence barriers shown by long-term
culture of normal human mammary epithelial cells //
Cancer Res. – 2009. – 69, ¹ 19. – P. 7557–7568.
33. McDuff F.K., Turner S.D. Jailbreak: oncogene-indu-
ced senescence and its evasion // Cell Signal. –
2011. – 23, ¹ 1. – P. 6–13.
34. Evans R.J., Wyllie F.S., Wynford-Thomas D. et al.
A P53-dependent, telomere-independent proli-
ferative life span barrier in human astrocytes
consistent with the molecular genetics of glioma
development // Cancer Res. – 2003. – 63, ¹ 16. –
P. 4854–4861.
35. Fu B., Quintero J., Baker C.C. Keratinocyte growth
conditions modulate telomerase expression, senescence,
and immortalization by human papillomavirus type 16
E6 and E7 oncogenes // Cancer Res. – 2003. – 63,
¹ 22. – P. 7815–7824.
36. Courtois-Cox S., Jones S.L., Cichowski K. Many
roads lead to oncogene-induced senescence //
Oncogene. – 2008. – 27, ¹ 20. – P. 2801–2809.
37. Hydbring P., Larsson L.G. Cdk2: a key regulator of
the senescence control function of Myc // Aging
(Albany NY). – 2010. – 2, ¹ 4. – P. 244–250.
38. Voghel G., Thorin-Trescases N., Mamarbachi A.M.
et al. Endogenous oxidative stress prevents telo-
merase-dependent immortalization of human endo-
thelial cells // Mech. Ageing Dev. – 2010. – 131,
¹ 5. – P. 354–363.
39. Yaswen P., Stampfer M.R. Molecular changes accom-
panying senescence and immortalization of cul-
tured human mammary epithelial cells // Int. J.
Biochem. Cell Biol. – 2002. – 34, ¹ 11. – P. 1382–
1394.
40. Evan G.I., d’Adda di Fagagna F. Cellular senescence:
hot or what? // Curr. Opin. Genet. Dev. – 2009. –
19, ¹ 1. – P. 25–31.
41. Fridman A.L., Tainsky M.A. Critical pathways in
cellular senescence and immortalization revealed
by gene expression profiling // Oncogene. – 2008. –
27, ¹ 46. – P. 5975–5987.
42. Indran I.R., Hande M.P., Pervaiz S. Tumor cell
redox state and mitochondria at the center of
the non-canonical activity of telomerase reverse
transcriptase // Mol. Aspects Med. – 2010. – 31,
¹ 1. – P. 21–28.
43. Kim D., Chung J. Akt: versatile mediator of cell
survival and beyond // J. Biochem. Mol. Biol. –
2002. – 35, ¹ 1. – P. 106–115.
44. Bitto A., Sell C., Crowe E., et al. Stress-induced
senescence in human and rodent astrocytes // Exp.
Cell Res. – 2010. – 316, ¹ 17. – P. 2961–2968.
45. Heeg S., Doebele M., von Werder A., Opitz O.G. In
vitro transformation models: modeling human
cancer // Cell Cycle. – 2006. – 5, ¹ 6. – P. 630–
634.
46. Rao K., Alper O., Opheim K.E. et al. Cytogenetic
characterization and H-ras associated transformation
of immortalized human mammary epithelial cells //
Cancer Cell Int. – 2006. – 6, ¹ 15.
61ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
47. Hong H.X., Zhang Y.M., Xu H. et al. Immortalization
of swine umbilical vein endothelial cells with
human telomerase reverse transcriptase // Mol.
Cells. – 2007. – 24, ¹ 3. – P. 358–363.
48. Cowling V.H., Cole M.D. E-cadherin repression
contributes to c-Myc-induced epithelial cell trans-
formation // Oncogene. – 2007. – 26, ¹ 24. –
P. 3582–3586.
49. De Filippis L., Ferrari D., Rota Nodari L. et al.
Immortalization of Human Neural Stem Cells
with the c-Myc Mutant T58A // PLoS One. –
2008. – 3, ¹ 10. – P. e3310.
50. Abken H., Bützler C., Willecke K. Immortalization
of human lymphocytes by transfection with DNA
from mouse L929 cytoplasts // Proc. Nat. Acad.
Sci. USA. – 1988. – 85, ¹ 2. – P. 468–472.
51. Shay J.W., Wright W.E., Werbin H. Defining the
molecular mechanisms of human immortalization //
Biochim. Biophys. Acta. – 1991. – 1072, ¹ 1. –
P. 1–7.
52. Yuan H., Veldman T., Rundell K., Schlegel R. Simian
virus 40 small tumor antigen activates AKT and
telomerase and induces anchorage-independent
growth of human epithelial cells // J. Virol. –
2002. – 76, ¹ 21. – P. 10685–10691.
53. Price T.N., Burke J.F., Mayne L.V. A novel human
astrocyte cell line (A735) with astrocyte- specific
neurotransmitter function // In Vitro Cell Dev.
Biol. Anim. – 1999. – 35, ¹ 5. – P. 279–288.
54. Radna R.L., Caton Y., Jha K.K. et al. Growth of
immortal simian virus 40 tsA-transformed human
fibroblasts is temperature dependent // Mol. Cell.
Biol. – 1989. – 9, ¹ 7. – P. – 3093–3096.
55. Shay J.W., Wright W.E. Quantitation of the
frequency of immortalization of normal human
diploid fibroblasts by SV40 large T-antigen // Exp.
Cell Res. – 1989. – 184, ¹ 1. – P. 109–118.
56. Wright W.E., Pereira-Smith O.M., Shay J.W. Reversible
cellular senescence: implications for immortalization of
normal human diploid fibroblasts // Mol. Cell. Biol. –
1989. – 9, ¹ 7. – P. 3088–3092.
57. Coca-Prados M., Wax M.B. Transformation of
human ciliary epithelial cells by simian virus 40:
induction of cell proliferation and retention of
beta 2-adrenergic receptors // Proc. Nat. Acad.
Sci. USA. – 1986. – 83, ¹ 22. – P. 8754–8758.
58. Ishida T., Ando H., Nomura S. et al. Establishment
and characterization of human fetal liver epithelial
cell line transfected with SV40 T antigen // Proc.
Soc. Exp. Biol. Med. – 1995. – 209, ¹ 3. – P. 251–
256.
59. Chang S.E., Keen J., Lane E.B., Taylor-Papadimit-
riou J. Establishment and characterization of SV40-
transformed human breast epithelial cell lines //
Cancer Res. – 1982. – 42, ¹ 5. – P. 2040–2053.
60. Kaighn M.E., Reddel R.R., Lechner J.F. et al.
Transformation of human neonatal prostate epithelial
cells by strontium phosphate transfection with a
plasmid containing SV40 early region genes // Cancer
Res. – 1989. – 49, ¹ 11. – P. 3050–3056.
61. Gruenert D.C., Basbaum C.B., Welsh M.J. et al.
Characterization of human tracheal epithelial cells
transformed by an origin-defective simian virus 40 //
Proc. Nat. Acad. Sci. USA. – 1988. – 85, ¹ 16. –
P. 5951–5955.
62. Christian B.J., Loretz L.J., Oberley T.D., Reznikoff C.A.
Characterization of human uroepithelial cells
immortalized in vitro by simian virus 40 // Cancer
Res. – 1987. – 47, ¹ 22. – P. 6066–6073.
63. Woodworth C.D., Bowden P.E., Doniger J. et al. Cha-
racterization of normal human exocervical epithe-
lial cells immortalized in vitro by papillomavirus
types 16 and 18 DNA // Cancer Res. – 1988. –
48, ¹ 16. – P. 4620–4628.
64. Pecoraro G., Morgan D., Defendi V. Differential ef-
fects of human papillomavirus type 6, 16, and 18
DNAs on immortalization and transformation of
human cervical epithelial cells // Proc. Nat. Acad.
Sci. USA. – 1989. – 86, ¹ 2. – P. 563–567.
65. Dürst M., Dzarlieva-Petrusevska R.T., Boukamp P.
et al. Molecular and cytogenetic analysis of im-
mortalized human primary keratinocytes obtained
after transfection with human papillomavirus type
16 DNA // Oncogene. – 1987. – 1, ¹ 3. – P. 251–
256.
66. Pirisi L., Yasumoto S., Feller M. et al. Transformation
of human fibroblasts and keratinocytes with human
papillomavirus type 16 DNA // J. Virol. – 1987. –
61, ¹ 4. – P. 1061–1066.
67. Münger K., Werness B.A., Dyson N. et al. Complex
formation of human papillomavirus E7 proteins with
the retinoblastoma tumor suppressor gene product //
EMBO J. – 1989. – 8, ¹ 13. – P. 4099–4105.
68. Hudson J.B., Bedell M.A., McCance D.J., Laimi-
nis L.A. Immortalization and altered differentiation
of human keratinocytes in vitro by the E6 and E7
open reading frames of human papillomavirus type
18 // J. Virol. – 1990. – 64, ¹ 2. – P. 519–526.
69. Zhang H., Jin Y., Chen X. et al. Papillomavirus
type 16 E6/E7 and human telomerase reverse
transcriptase in esophageal cell immortalization and
early transformation // Cancer Lett. – 2007. – 245,
¹ 1–2. – P. 184–194.
70. Woodworth C.D., Doniger J., DiPaolo J.A. Immor-
talization of human foreskin keratinocytes by
various human papillomavirus dnas corresponds
to their association with cervical carcinoma // J.
Virol. – 1989. – 63, ¹ 1. – P. 159–164.
71. Blanton R.A., Perez-Reyes N., Merrick D.T.,
McDougall J.K. Epithelial cells immortalized by
62 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
human papillomaviruses have premalignant charac-
teristics in organotypic culture // Amer. J. Pathol. –
1991. – 138, ¹ 3. – P. 673–685.
72. Tsutsui T., Kumakura S., Yamamoto A. et al. Associ-
ation of p16 (INK4a) and pRb inactivation with
immortalization of human cells // Carcinogenesis. –
2002. – 23, ¹ 12. – P. 2111–2117.
73. Halbert C.L., Demers G.W., Galloway D.A. The E7
gene of human papillomavirus type 16 is sufficient
for immortalization of human epithelial cells // J.
Virol. – 1991. – 65, ¹ 1. – P. 473–478.
74. Band V., Zajchowski D., Kulesa V., Sager R. Human
papilloma virus DNAs immortalize normal human
mammary epithelial cells and reduce their growth
factor requirements // Proc. Nat. Acad. Sci. USA. –
1990. – 87, ¹ 1. – P. 463–467.
75. Wazer D.E., Liu X.L., Chu Q., et al. Immortalization
of distinct human mammary epithelial cell types by
human papilloma virus 16 E6 or E7 // Proc. Nat.
Acad. Sci. USA. – 1995. – 92, ¹ 9. – P. 3687–
3691.
76. Reznikoff C.A., Belair C., Savelieva E., et al. Long-
term genome stability and minimal genotypic and
phenotypic alterations in HPV16 E7-, but not E6-
immortalized human uroepithelial cells // Genes
Dev. – 1994. – 8, ¹ 18. – P. 2227–2240.
77. Carmean N., Kosman J.W., Leaf E.M. et al. Immor-
talization of human urothelial cells by human
papillomavirus type 16 E6 and E7 genes in a de-
fined serum-free system // Cell Prolif. – 2007. –
40, ¹ 2. – P. 166–184.
78. Graham F.L., Smiley J., Russell W.C., Nairn R.
Characteristics of a human cell line transformed
by DNA from human adenovirus type 5 // J. Gen.
Virol. – 1977. – 36, ¹ 1. – P. 59–74.
79. Reddel R.R., Ke Y., Gerwin B.I. et al. Transformation
of human bronchial epithelial cells by infection
with SV40 or adenovirus-12 SV40 hybrid virus,
or transfection via strontium phosphate copreci-
pitation with a plasmid containing SV40 early
region genes // Cancer Res. – 1988. – 48, ¹ 7. –
P. 1904–1909.
80. Rhim J.S., Jay G., Arnstein P. et al. Neoplastic
transformation of human epidermal keratinocytes
by AD12-SV40 and Kirsten sarcoma viruses //
Science. – 1985. – 227, ¹ 4691. – P. 1250–
1252.
81. Farwell D.G., Shera K.A., Koop J.I. et al. Genetic
and epigenetic changes in human epithelial cells
immortalized by telomerase // Amer. J. Pathol. –
2000. – 156, ¹ 5. – P. 1537–1547.
82. Tao Q., Lv B., Qiao B. et al. Immortalization of
ameloblastoma cells via reactivation of telomerase
function: Phenotypic and molecular characteristics //
Oral Oncol. – 2009. – 45, ¹ 12. – P. 239–244.
83. Jiang X.R., Jimenez G., Chang E. et al. Telomerase
expression in human somatic cells does not induce
changes associated with a transformed phenotype //
Nat. Genet. – 1999. – 21, ¹ 1. – P. 111–114.
84. Morales C.P., Holt S.E., Ouellette M. et al. Absence
of cancer-associated changes in human fibroblasts
immortalized with telomerase // Nat. Genet. –
1999. – 21, ¹ 1. – P. 115–118.
85. Noble J.R., Zhong Z.H., Neumann A.A. et al.
Alterations in the p16(INK4a) and p53 tumor
suppressor genes of hTERT-immortalized human
fibroblasts // Oncogene. – 2004. – 23, ¹ 17. –
P. 3116–3121.
86. Dickson M.A., Hahn W.C., Ino Y. et al. Human
keratinocytes that express hTERT and also bypass
a p16(INK4a)-enforced mechanism that limits life
span become immortal yet retain normal growth
and differentiation characteristics // Mol. Cell.
Biol. – 2000. – 20, ¹ 4. – P. 1436–1447.
87. Kim H., Farris J., Christman S.A. et al. Events
in the immortalizing process of primary human
mammary epithelial cells by the catalytic subunit
of human telomerase // Biochem. J. – 2002. –
365, ¹ 3. – P. 765–772.
88. Benanti J.A., Wang M.L., Myers H.E. et al.
Epigenetic down-regulation of ARF expression
is a selection step in immortalization of human
fibroblasts by c-Myc // Mol. Cancer Res. – 2007. –
5, ¹ 11. – P. 1181–1189.
89. Gil J., Kerai P., Lleonart M. et al. Immortalization
of primary human prostate epithelial cells by
c-Myc // Cancer Res. – 2005. – 65, ¹ 6. –
P. 2179–2185.
90. Nonet G.H., Stampfer M.R., Chin K. et al. The
ZNF217 gene amplified in breast cancers promotes
immortalization of human mammary epithelial
cells // Cancer Res. – 2001. – 61, ¹ 4. – P. 1250–
1254.
91. Opitz O.G., Suliman Y., Hahn W.C. et al. Cyclin
D1 overexpression and p53 inactivation immortalize
primary oral keratinocytes by a telomerase-independent
mechanism // J. Clin. Invest. – 2001. – 108, ¹ 5. –
P. 725–732.
92. Pipas J.M. SV40: Cell transformation and tumori-
genesis // Virology. – 2009. – 384, ¹ 2. – P. 294–
303.
93. Levine A.J. The common mechanisms of
transformation by the small DNA tumor viruses:
The inactivation of tumor suppressor gene
products: p53 // Virology. – 2009. – 384, ¹ 2. –
P. 285–293.
94. Ghittoni R., Accardi R., Hasan U. et al. The bio-
logical properties of E6 and E7 oncoproteins from
human papillomaviruses // Virus Genes. – 2010. –
40, ¹ 1. – P. 1–13.
63ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
95. Huschtscha L.I., Reddel R.R. p16(INK4a) and
the control of cellular proliferative life span //
Carcinogenesis. – 1999. – 20, ¹ 6. – P. 921–
926.
96. Haga K., Ohno S., Yugawa T. et al. Efficient
immortalization of primary human cells by
p16INK4a-specific short hairpin RNA or Bmi-1,
combined with introduction of hTERT // Cancer
Sci. – 2007. – 98, ¹ 2. – P. 147–154.
97. Zhao Y., Wang S., Popova E.Y. et al. Rearrangement
of upstream sequences of the hTERT gene during
cellular immortalization // Genes Chromosomes
Cancer. – 2009. – 48, ¹ 11. – P. 963–974.
98. Frisch S.M., Mymryk J.S. Adenovirus-5 E1A:
paradox and paradigm // Nat. Rev. Mol. Cell.
Biol. – 2002. – 3, ¹ 6. – P. 441–452.
99. Cesare A.J., Reddel R.R. Telomere uncapping and
alternative lengthening of telomeres // Mech.
Ageing Dev. – 2008. – 129, ¹ 1/2. – P. 99–
108.
100. Henson J.D., Reddel R.R. Assaying and investigating
alternative lengthening of telomeres activity in
human cells and cancers // FEBS Lett. – 2010. –
584, ¹ 17. – P. 3800–3811.
101. Adhikary S., Eilers M. Transcriptional regulation
and transformation by Myc proteins // Nat. Rev.
Mol. Cell. Biol. – 2005. – 6, ¹ 8. – P. 635–645.
102. Yin X.Y., Grove L., Datta N.S. et al. Inverse regulation
of cyclin B1 by c-Myc and p53 and induction of
tetraploidy by cyclin B1 overexpression // Cancer
Res. – 2001. – 61, ¹ 17. – P. 6487–6493.
103. Feng M., Li Z., Aau M. et al. Myc/miR-378/TOB2/
cyclin D1 functional module regulates oncogenic
transformation // Oncogene. – 2011. – 30, ¹ 19. –
P. 2242–2251.
104. Jung Y.S., Qian Y., Chen X. Examination of the
expanding pathways for the regulation of p21
expression and activity // Cell Signal. – 2010. –
22, ¹ 7. – P. 1003–1012.
105. Gartel A.L., Ye X., Goufman E. et al. Myc represses
the p21(WAF1/CIP1) promoter and interacts with
Sp1/Sp3 // Proc. Nat. Acad. Sci. USA. – 2001. –
98, ¹ 8. – P. 4510–4515.
106. Lin C.P., Liu C.R., Lee C.N. et al. Targeting
c-Myc as a novel approach for hepatocellular
carcinoma // World J Hepatol. – 2010. – 2, ¹
1. – P. 16–20.
107. Bartlett P.F., Reid H.H., Bailey K.A., Bernard O.
Immortalization of mouse neural precursor cells
by the c-myc oncogene // Proc. Nat. Acad. Sci.
USA. – 1988. – 85, ¹ 9. – P. 3255–3259.
108. Gregory M.A., Qi Y., Hann S.R. The ARF tumor
suppressor: keeping Myc on a leash // Cell Cycle. –
2005. – 4, ¹ 2. – P. 249–252.
109. Dimri M., Naramura M., Duan L. et al. Modeling
breast cancer-associated c-Src and EGFR over-
expression in human MECs: c-Src and EGFR
cooperatively promote aberrant three- dimensional
acinar structure and invasive behavior // Cancer
Res. – 2007. – 67, ¹ 9. – P. 4164–4172.
110. Mills A.A. Throwing the cancer switch: reciprocal
roles of polycomb and trithorax proteins // Nat.
Rev. Cancer. – 2010. – 10, ¹ 10. – P. 669–682.
111. Dimri G.P., Martinez J.L., Jacobs J.J. et al. The
Bmi-1 oncogene induces telomerase activity and
immortalizes human mammary epithelial cells //
Cancer Res. – 2002. – 62, ¹ 16. – P. 4736–4745.
112. Song L.B., Zeng M.S., Liao W.T. et al. Bmi-1
is a novel molecular marker of nasopharyngeal
carcinoma progression and immortalizes primary
human nasopharyngeal epithelial cells // Cancer
Res. – 2006. – 66, ¹ 12. – P. 6225–6232.
113. Kim R.H., Kang M.K., Shin K.H. et al. Bmi-1
cooperates with human papillomavirus type 16 E6
to immortalize normal human oral keratinocytes //
Exp. Cell Res. – 2007. – 313, ¹ 3. – P. 462–72.
114. Meng S., Luo M., Sun H. et al. Identification and
characterization of Bmi-1-responding element
within the human p16 promoter // J. Biol. Chem. –
2010. – 285, ¹ 43. – P. 33219–33229.
115. Yochum G.S., McWeeney S., Rajaraman V. et al.
Serial analysis of chromatin occupancy identifies
beta-catenin target genes in colorectal carcinoma
cells // Proc. Nat. Acad. Sci. USA. – 2007. –
104, ¹ 9. – P. 3324–3329.
116. Mosimann C., Hausmann G., Basler K. Beta-
catenin hits chromatin: regulation of Wnt target
gene activation. // Nat. Rev. Mol. Cell Biol. –
2009. – 10, ¹ 4. – P. 276–286.
117. Delmas V., Beermann F., Martinozzi S. et al. -Ca-
tenin induces immortalization of melanocytes by
suppressing p16INK4a expression and cooperates
with N-Ras in melanoma development // Genes
Dev. – 2007. – 21, ¹ 22. – P. 2923–2935.
118. Kim J., Lee J.H., Iyer V.R. Global Identification
of Myc Target Genes Reveals Its Direct Role in
Mitochondrial Biogenesis and Its E-Box Usage In
Vivo // PLoS ONE. – 2008. – 3, ¹ 3. – P. e1798.
119. Krig S.R., Jin V.X., Bieda M.C. et al. Identification
of genes directly regulated by the oncogene
ZNF217 using chromatin immunoprecipitation
(ChIP)-chip assays // J. Biol. Chem. – 2007. –
282, ¹ 13. – P. 9703–9712.
120. Zeller K.I., Zhao X., Lee C.W. et al. Global mapping
of c-Myc binding sites and target gene networks in
human B cells // Proc. Nat. Acad. Sci. USA. –
2006. – 103, ¹ 47. – P. 17834–17839.
121. Lawlor E.R., Soucek L., Brown-Swigart L. et al.
Reversible kinetic analysis of Myc targets in vivo
provides novel insights into Myc-mediated tumo-
64 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
rigenesis // Cancer Res. – 2006. – 66, ¹ 9. – P. 4591–
4601.
122. Wu C.H., Sahoo D., Arvanitis C. et al. Combined
analysis of murine and human microarrays and
ChIP analysis reveals genes associated with the
ability of MYC to maintain tumorigenesis // PLoS
Genet. – 2008. – 4, ¹ 6. – P. e1000090.
123. Menssen A., Hermeking H. Characterization of the
c-MYC-regulated transcriptome by SAGE: identi-
fication and analysis of c-MYC target genes // Proc.
Nat. Acad. Sci. USA. – 2002. – 99, ¹ 9. – P. 6274–
6279.
124. O’Connell B.C., Cheung A.F., Simkevich C.P. et al. A
large scale genetic analysis of c- Myc-regulated gene
expression patterns // J. Biol. Chem. – 2003. –
278, ¹ 14. – P. 12563–12573.
125. Rea M.A., Zhou L., Qin Q. et al. Spontaneous
immortalization of human epidermal cells with
naturally elevated telomerase // J. Invest. Dermatol . –
2006. – 126, ¹ 11. – P. 2507–2515.
126. Brandl C., Kaesbauer J., Weber B.H., Morsczeck C.
Spontaneous immortalization of neural crest-
derived corneal progenitor cells after chromosomal
aberration // Cell Prolif. – 2010. – 43, ¹ 4. – P. 372–
377.
127. Zhao C., Meng L., Hu H. et al. Spontaneously
immortalised bovine mammary epithelial cells
exhibit a distinct gene expression pattern from the
breast cancer cells // BMC Cell Biol. – 2010. –
11, ¹ 82.
128. Ohtani N., Yamakoshi K., Takahashi A., Hara E.
Real-time in vivo imaging of p16 gene expression:
a new approach to study senescence stress signaling
in living animals // Cell Div. – 2010. – 5, ¹ 1.
129. Novak P., Jensen T.J., Garbe J.C. et al. Stepwise
DNA methylation changes are linked to escape
from defined proliferation barriers and mammary
epithelial cell immortalization // Cancer Res. –
2009. – 69, ¹ 12. – P. 5251–5258.
130. Darbro B.W., Lee K.M., Nguyen N.K. et al. Methy-
lation of the p16(INK4a) promoter region in
telomerase immortalized human keratinocytes co-
cultured with feeder cells // Oncogene. – 2006. –
25, ¹ 56. – P. 7421–7433.
131. Hinshelwood R.A., Melki J.R., Huschtscha L.I. et al.
Aberrant de novo methylation of the p16INK4A
CpG island is initiated post gene silencing in
association with chromatin remodelling and
mimics nucleosome positioning // Hum. Mol.
Genet. – 2009. – 18, ¹ 16. – P. 3098–3109.
132. Agherbi H., Gaussmann-Wenger A., Verthuy C. et al.
Polycomb mediated epigenetic silencing and
replication timing at the INK4a/ARF locus during
senescence // PLoS One. – 2009. – 4, ¹ 5. – P.
e5622.
133. Maertens G.N., El Messaoudi-Aubert S., Racek T. et
al. Several distinct polycomb complexes regulate
and co-localize on the INK4a tumor suppressor
locus // PLoS One. – 2009. – 4, ¹ 7. – P. e6380.
134. Agger K., Cloos P.A., Rudkjaer L. et al. The H3K27me3
demethylase JMJD3 contributes to the activation of
the INK4A-ARF locus in response to oncogene- and
stress-induced senescence // Genes Dev. – 2009. –
23, ¹ 10. – P. 1171–1176.
135. Maertens G.N., El Messaoudi-Aubert S., Elderkin S.
et al. Ubiquitin-specific proteases 7 and 11 mo-
dulate Polycomb regulation of the INK4a tumour
suppressor // EMBO J. – 2010. – 29, ¹ 15. –
P. 2553–2565.
136. Negishi M., Saraya A., Mochizuki S. et al. A novel
zinc finger protein Zfp277 mediates transcriptional
repression of the Ink4a/arf locus through polycomb
repressive complex 1 // PLoS One. – 2010. – 5,
¹ 8. – P. e12373.
137. Meng L., Gabai V.L., Sherman M.Y. Heat-shock
transcription factor HSF1 has a critical role in
human epidermal growth factor receptor-2-in-
duced cellular transformation and tumorigenesis //
Oncogene. – 2010. – 29, ¹ 37. – P. 5204–5213.
138. Romagosa C., Simonetti S., López-Vicente L. et al.
p16(Ink4a) overexpression in cancer: a tumor sup-
pressor gene associated with senescence and high-
grade tumors // Oncogene. – 2011. – 30, ¹ 18. –
P. 2087–2097.
139. Odell A., Askham J., Whibley C., Hollstein M. How
to become immortal: let MEFs count the ways //
Aging (Albany NY). – 2010. – 2, ¹ 3. – P. 160–
165.
140. Mondello C., Chiesa M., Rebuzzini P. et al. Karyo-
type instability and anchorage-independent growth
in telomerase-immortalized fibroblasts from two
centenarian individuals // Biochem. Biophys. Res.
Communs. – 2003. – 308, ¹ 4. – P. 914–921.
141. Milyavsky M., Shats I., Erez N. et al. Prolonged
culture of telomerase-immortalized human fibroblasts
leads to a premalignant phenotype // Cancer Res. –
2003. – 63, ¹ 21. – P. 7147–7157.
142. Pirzio L.M., Freulet-Marrière M.A., Bai Y. et al. Human
fibroblasts expressing hTERT show remarkable karyo-
type stability even after exposure to ionizing radiation //
Cytogenet. Genome Res. – 2004. – 104, ¹ 1–4. –
P. 87–94.
143. Cui W., Aslam S., Fletcher J. et al. Stabilization of
telomere length and karyotypic stability are di-
rectly correlated with the level of hTERT gene
expression in primary fibroblasts // J. Biol. Chem. –
2002. – 277, ¹ 41. – P. 38531–38539.
144. Wen V.W., Wu K., Baksh S. et al. Telomere-driven
karyotypic complexity concurs with p16INK4a
inactivation in TP53-competent immortal endo-
65ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
thelial cells // Cancer Res. – 2006. – 66, ¹ 22. –
P. 10691–10700.
145. Piao C.Q., Liu L., Zhao Y.L. et al. Immortalization
of human small airway epithelial cells by ectopic
expression of telomerase // Carcinogenesis. –
2005. – 26, ¹ 4. – P. 725–731.
146. Varella-Garcia M., Chen L., Zheng X. et al. Karyotypic
characteristics of human uterine leiomyoma and myo-
metrial cell lines following telomerase induction //
Cancer Genet. Cytogenet. – 2006. – 170, ¹ 1. – P. 71–
75.
147. Takeuchi M., Takeuchi K., Kohara A. et al. Chro-
mosomal instability in human mesenchymal stem
cells immortalized with human papilloma virus E6,
E7, and hTERT genes // In Vitro Cell. Dev. Biol.
Anim. – 2007. – 43, ¹ 3/4. – P. 129–138.
148. Haker B., Fuchs S., Dierlamm J. et al. Absence of
oncogenic transformation despite acquisition of
cytogenetic aberrations in long-term cultured telo-
merase-immortalized human fetal hepatocytes //
Cancer Lett. – 2007. – 256, ¹ 1. – P. 120–127.
149. Liu S., Hatton M.P., Khandelwal P., Sullivan D.A.
Culture, immortalization, and characterization of
human meibomian gland epithelial cells // Invest.
Ophthalmol. Vis. Sci. – 2010. – 51, ¹ 8. –
P. 3993–4005.
150. MacDonald C., Watts P., Stuart B. et al. Studies
on the phenotype and karyotype of immortalized
rabbit kidney epithelial cell lines // Exp. Cell. Res. –
1991. – 195, ¹ 2. – P. 458–461.
151. Yamasaki K., Kawasaki S., Young R.D. et al. Ge-
nomic aberrations and cellular heterogeneity in
SV40-immortalized human corneal epithelial cells //
Invest. Ophthalmol. Vis. Sci. – 2009. – 50, ¹ 2. –
P. 604–613.
152. Kubo C., Tsutsui T.W., Tamura Y. et al. Immorta-
lization of normal human gingival keratinocytes
and cytological and cytogenetic characterization
of the cells // Odontology. – 2009. – 97, ¹ 1. –
P. 18–31.
153. Tsao S.W., Wang X., Liu Y. et al. Establishment
of two immortalized nasopharyngeal epithelial cell
lines using SV40 large T and HPV16 E6/E7 viral
oncogenes // Biochim. Biophys. acta . – 2002. –
1590, ¹ 1–3. – P. 150–158.
154. Schiller J.H., Bittner G., Wu S.Q., Meisner L.
Karyotypic changes associated with spontaneous
acquisition and loss of tumorigenicity in a human
transformed bronchial epithelial cell line: evidence
for in vivo selection of transformed clones // In
Vitro Cell Dev. Biol. Anim. – 1998. – 34, ¹ 4. –
P. 283–289.
155. Toouli C.D., Huschtscha L.I., Neumann A.A. et al.
Comparison of human mammary epithelial cells
immortalized by simian virus 40 T-Antigen or by
the telomerase catalytic subunit // Oncogene. –
2002. – 21, ¹ 1. – P. 128–139.
156. Hashida T., Yasumoto S. Induction of chromosome
abnormalities in mouse and human epidermal
keratinocytes by the human papillomavirus type
16 E7 oncogene // J. Gen. Virol. – 1991. – 72,
¹ 7. – P. 1569–1577.
157. Bonin L.R., Madden K., Shera K. et al. Generation
and characterization of human smooth muscle
cell lines derived from atherosclerotic plaque //
Arterioscler., Thromb. Vasc. Biol. – 1999. – 19,
¹ 3. – P. 575–587.
158. Jin Y., Feng H.C., Deng W. et al. Immortalization
of human extravillous cytotrophoblasts by human
papilloma virus gene E6/E7: sequential cytogenetic
and molecular genetic characterization // Cancer
Genet. Cytogenet. – 2005. – 163, ¹ 1. – P. 30–37.
159. Ramirez R.D., Sheridan S., Girard L. et al. Immor-
talization of human bronchial epithelial cells in
the absence of viral oncoproteins // Cancer Res. –
2004. – 64, ¹ 24. – P. 9027–9034.
160. Todaro G.J., Green H. Quantitative studies of the
growth of mouse embryo cells in culture and their
development into established lines // J. Cell. Biol. –
1963. – ¹ 17. – P. 299–313.
161. Endo S., Metzler M., Hieber L. Nonrandom karyo-
typic changes in a spontaneously immortalized and
tumourigenic Syrian hamster embryo cell line //
Carcinogenesis. – 1994. – 15, ¹ 10. – P. 2387–
2390.
162. Allen-Hoffmann B.L., Schlosser S.J., Ivarie C.A. et al.
Normal growth and differentiation in a sponta-
neously immortalized near-diploid human kerati-
nocyte cell line, NIKS // J. Invest. Dermatol. –
2000. – 114, ¹ 3. – P. 444–455.
163. Grinchuk T.M., Pugovkina N.A., Tarunina M.V. et
al. Immortalized cell lines from murine embryos
are characterized by progressive destabilization of
their karyotype structure // Tsitologiia. – 2004. –
46, ¹ 1. – P. 62–68.
164. Fulcher M.L., Gabriel S.E., Olsen J.C. et al. Novel
human bronchial epithelial cell lines for cystic
fibrosis research // Amer. J. Physiol. Lung Cell
Mol. Physiol. – 2009. – 296, ¹ 1. – P. 82–91.
165. Marella N.V., Malyavantham K.S., Wang J. et al.
Cytogenetic and cDNA microarray expression
analysis of MCF10 human breast cancer progression
cell lines // Cancer Res. – 2009. – 69, ¹ 14. – P. 5946–
5953.
166. Fridman A.L., Tang L., Kulaeva O.I. et al. Expres-
sion profiling identifies three pathways altered in
cellular immortalization: interferon, cell cycle,
and cytoskeleton // J. Gerontol. A Biol. Sci. Med.
Sci. – 2006. – 61, ¹ 9. – P. 879–889.
167. Nakamura H., Fukami H., Hayashi Y. et al. Esta-
66 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
blishment of immortal normal and ataxia telangi-
ectasia fibroblast cell lines by introduction of the
hTERT gene // J. Radiat. Res. (Tokyo). – 2002. – 43,
¹ 2. – P. 167–174.
168. Cui W., Wylie D., Aslam S. et al. Telomerase-immor-
talized sheep fibroblasts can be reprogrammed by
nuclear transfer to undergo early development //
Biol. Reprod. – 2003. – 69, ¹ 1. – P. 15–21.
169. Krikun G., Mor G., Huang J. et al. Metalloproteinase
expression by control and telomerase immortalized
human endometrial endothelial cells // Histol.
Histopathol. – 2005. – 20, ¹ 3. – P. 719–724.
170. Zhang H., Wang Y., Zhao Y. et al. Immortalized
human neural progenitor cells from the ventral
telencephalon with the potential to differentiate
into GABAergic neurons // J. Neurosci. Res. –
2008. – 86, ¹ 6. – P. 1217–1226.
171. Steele S.L., Wu Y., Kolb R.J. et al. Telomerase
immortalization of principal cells from mouse
collecting duct // Amer. J. Physiol. Renal. Physiol. –
2010. – 299, ¹ 6. – P. 1507–1514.
172. Unger C., Gao S., Cohen M. et al. Immortalized hu-
man skin fibroblast feeder cells support growth
and maintenance of both human embryonic and
induced pluripotent stem cells // Hum. Reprod. –
2009. – 24, ¹ 10. – P. – 2567–2581.
173. Donehower L.A., Lozano G. 20 years studying p53
functions in genetically engineered mice // Nat.
Rev. Cancer. – 2009. – 9, ¹ 11. – P. 831–841.
174. Li M., Fang X., Baker D.J. et al. The ATM-p53 path-
way suppresses aneuploidy-induced tumorigenesis //
Proc. Nat. Acad. Sci. USA. – 2010. – 107, ¹ 32. –
P. 14188–14193.
175. Weiss M.B., Vitolo M.I., Mohseni M. et al. Deletion
of p53 in human mammary epithelial cells causes
chromosomal instability and altered therapeutic
response // Oncogene. – 2010. – 29, ¹ 33. –
P. 4715–4724.
176. Thompson S.L., Compton D.A. Proliferation of
aneuploid human cells is limited by a p53-
dependent mechanism // J. Cell. Biol. – 2010. –
188, ¹ 3. – P. 369–381.
177. Zhao T., Xu Y. p53 and stem cells: new developments
and new concerns // Trends Cell. Biol. – 2010. –
20, ¹ 3. – P. 170–175.
178. Fukasawa K. P53, cyclin-dependent kinase and
abnormal amplification of centrosomes // Biochim.
Biophys. acta. – 2008. – 1786, ¹ 1. – P. 15–23.
179. Yun U.J., Park H.D., Shin D.Y. p53 prevents
immature escaping from cell cycle G2 checkpoint
arrest through inhibiting cdk2-dependent NF-Y
phosphorylation // Cancer Res. Treat. – 2006. –
38, ¹ 4. – P. 224–228.
180. Iovino F., Lentini L., Amato A., Di Leonardo A. RB
acute loss induces centrosome amplification and
aneuploidy in murine primary fibroblasts // Mol.
Cancer. – 2006. – 5, ¹ 38.
181. Manning A.L., Longworth M.S., Dyson N.J. Loss of
pRB causes centromere dysfunction and chromoso-
mal instability // Genes Dev. – 2010. – 24, ¹ 13. –
P. 1364–1376.
182. Thompson S.L., Bakhoum S.F., Compton D.A.
Mechanisms of chromosomal instability // Curr.
Biol. – 2010. – 20, ¹ 6. – P. 285–295.
183. Knudsen E.S., Knudsen K.E. Tailoring to RB: tu-
mour suppressor status and therapeutic response //
Nat. Rev. Cancer. – 2008. – 8, ¹ 9. – P. 714–
724.
184. Hernando E., Nahlé Z., Juan G. et al. Rb inactivation
promotes genomic instability by uncoupling cell
cycle progression from mitotic control // Nature. –
2004. – 430, ¹ 7001. – P. 797–802.
185. Amato A., Lentini L., Schillaci T. et al. RNAi media-
ted acute depletion of retinoblastoma protein (pRb)
promotes aneuploidy in human primary cells via
micronuclei formation // BMC Cell. Biol. – 2009. –
10, ¹ 79.
186. Amato A., Schillaci T., Lentini L., Di Leonardo A.
CENPA overexpression promotes genome instabi-
lity in pRb-depleted human cells // Mol. Cancer. –
2009. – 8, ¹ 119.
187. Srinivasan S.V., Mayhew C.N., Schwemberger S. et al.
RB loss promotes aberrant ploidy by deregulating
levels and activity of DNA replication factors // J.
Biol. Chem. – 2007. – 282, ¹ 33. – P. 23867–
23877.
188. Bourgo R.J., Ehmer U., Sage J., Knudsen E.S.
RB deletion disrupts coordination between DNA
replication licensing and mitotic entry in vivo //
Mol. Biol. Cell. – 2011. – 22, ¹ 7. – P. 931–
939.
189. Coschi C.H., Martens A.L., Ritchie K. et al. Mito-
tic chromosome condensation mediated by the
retinoblastoma protein is tumor-suppressive //
Genes Dev. – 2010. – 24, ¹ 13. – P. 1351–1363.
190. Wirt S.E., Sage J. p107 in the public eye: an Rb
understudy and more // Cell Div. – 2010. – 5, ¹ 9.
191. Weinberg R.A. Cell the retinoblastoma protein and
cell cycle control // Cell. – 1995. – 81, ¹ 3. –
P. 323–330.
192. Dannenberg J.H., Schuijff L., Dekker M. et al. Tissue-
specific tumor suppressor activity of retinoblastoma
gene homologs p107 and p130 // Genes Dev. –
2004. – 18, ¹ 23. – P. 2952–2962.
193. Dannenberg J.H., te Riele H.P. The retinoblastoma
gene family in cell cycle regulation and suppression
of tumorigenesis // Results Probl. Cell Differ. –
2006. – 42, ¹. – P. 183–225.
194. Schaffer B.E., Park K.S., Yiu G. et al. Loss of p130
accelerates tumor development in a mouse model
67ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
for human small-cell lung carcinoma // Cancer
Res. – 2010. – 70, ¹ 10. – P. 3877– 3883.
195. Albertson D.G., Collins C., McCormick F., Gray J.W.
Chromosome aberrations in solid tumors // Nat.
Genet. – 2003. – 34, ¹ 4. – P. 369–376.
196. Hanahan D., Weinberg R.A. Hallmarks of cancer:
the next generation // Cell. – 2011. – 144, ¹ 5. –
P. 646–674.
197. Santarius T., Shipley J., Brewer D. et al. A census
of amplified and overexpressed human cancer
genes // Nat. Rev. Cancer. – 2010. – 10, ¹ 1. –
P. 59–64.
198. Stratton M.R., Campbell P.J., Futreal P.A. The can-
cer genome // Nature. – 2009. – 458, ¹ 7239. –
P. 719–724.
199. Dai B., Pieper R.O., Li D. et al. FoxM1B regulates
NEDD4-1 expression, leading to cellular trans-
formation and full malignant phenotype in immor-
talized human astrocytes // Cancer Res. – 2010. –
70, ¹ 7. – P. 2951–2961.
200. Zhang X., Yu C., Wilson K. et al. Malignant trans-
formation of non-neoplastic Barrett’s epithelial
cells through well-defined genetic manipulations //
PLoS One. – 2010. – 5, ¹ 9. – P. e13093.
201. Zhu Y., Zhong X., Zheng S. et al. Transformation of
immortalized colorectal crypt cells by microcystin
involving constitutive activation of Akt and MAPK
cascade // Carcinogenesis. – 2005. – 26, ¹ 7. –
P. 1207–1214.
202. Narko K., Ristimäki A., MacPhee M. et al. Tumo-
rigenic transformation of immortalized ECV
endothelial cells by cyclooxygenase-1 overexpres-
sion // J. Biol. Chem. – 1997. – 272, ¹ 34. –
P. 21455–21460.
203. Shen Z.Y., Xu L.Y., Chen M.H. et al. Progressive trans-
formation of immortalized esophageal epithelial
cells // World J. Gastroenterol. – 2002. – 8, ¹ 6. –
P. 976–981.
204. Cheng J.D., Dunbrack R.L. Jr., Valianou M. et al.
Promotion of tumor growth by murine fibroblast
activation protein, a serine protease, in an animal
model // Cancer Res. – 2002. – 62, ¹ 16. –
P. 4767–4772.
205. Hamid T., Malik M.T., Kakar S.S. Ectopic expression
of PTTG1/securin promotes tumorigenesis in
human embryonic kidney cells // Mol. Cancer. –
2005. – 4 (1), ¹ 3.
206. Li A., Zhang X.S., Jiang J.H. et al. Transcriptional
expression of RPMS1 in nasopharyngeal carcinoma
and its oncogenic potential // Cell Cycle. – 2005. –
4, ¹ 2. – P. 304–309.
207. Castiglioni F., Tagliabue E., Campiglio M. et al.
Role of exon-16-deleted HER2 in breast carcino-
mas // Endocr. Relat. Cancer. – 2006. – 13, ¹
1. – P. 221–232.
208. Wang Y.L., Wang Y., Tong L., Wei Q. Over-
expression of calcineurin B subunit (CnB) enhan-
ces the oncogenic potential of HEK293 cells //
Cancer Sci. – 2008. – 99, ¹ 6. – P. 1100–
1108.
209. Mineur P., Colige A.C., Deroanne C.F. et al. Newly
identified biologically active and proteolysis-resistant
VEGF-A isoform VEGF111 is induced by genotoxic
agents // J. Cell. Biol. – 2007. – 179, ¹ 6. –
P. 1261–1273.
210. Chao C., Goluszko E., Lee Y.T. et al. Constitutively
active CCK2 receptor splice variant increases Src-
dependent HIF-1 alpha expression and tumor
growth // Oncogene. – 2007. – 26, ¹ 7. – P. 1013–
1019.
211. Depontieu F., Grigoriu B.D., Scherpereel A. et al.
Loss of Endocan tumorigenic properties after
alternative splicing of exon 2 // BMC Cancer. –
2008. – 8, ¹ 14.
212. Liu Y.H., Lin C.Y., Lin W.C. et al. Up-regulation of
vascular endothelial growth factor-d expression in
clear cell renal cell carcinoma by CD74: A critical
role in cancer cell tumorigenesis // J. Immunol. –
2008. – 181, ¹ 9. – P. 6584–6594.
213. Ha S.A., Kim H.K., Yoo J. et al. Transdifferentiation-
inducing HCCR-1 oncogene // BMC Cell. Biol. –
2010. – 11, ¹ 49.
214. Jin G., Kawsar H.I., Hirsch S.A. et al. An anti-
microbial peptide regulates tumor-associated macro-
phage trafficking via the chemokine receptor CCR2,
a model for tumorigenesis // PLoS One. – 2010. –
5, ¹ 6. – P. e10993.
215. Liu S.H., Patel S., Gingras M.C. et al. PDX-1:
demonstration of oncogenic properties in pancreatic
cancer // Cancer. – 2011. – 117, ¹ 4. – P. 723–
733.
216. Zhou W.J., Geng Z.H., Chi S. et al. Slit-Robo signaling
induces malignant transformation through Hakai-
mediated E-cadherin degradation during colorectal
epithelial cell carcinogenesis // Cell Res. – 2011. –
21, ¹ 4. – P. 609–626.
217. Zhao J.J., Gjoerup O.V., Subramanian R.R. et al.
Human mammary epithelial cell transformation
through the activation of phosphatidylinositol
3-kinase // Cancer Cell. – 2003. – 3, ¹ 5. – P. 483–
495.
218. Zelinski D.P., Zantek N.D., Stewart J.C. et al. EphA2
overexpression causes tumorigenesis of mammary
epithelial cells // Cancer Res. – 2001. – 61, ¹ 5. –
P. 2301–2306.
219. Prescott J.D., Koto K.S., Singh M., Gutierrez-Hart-
mann A. The ETS transcription factor ESE-1
transforms MCF-12A human mammary epithelial
cells via a novel cytoplasmic mechanism // Mol.
Cell. Biol . – 2004. – 24, ¹ 12. – P. 5548–5564.
68 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
220. Moyano J.V., Evans J.R., Chen F. et al. AlphaB-
crystallin is a novel oncoprotein that predicts
poor clinical outcome in breast cancer // J. Clin.
Invest. – 2006. – 116, ¹ 1. – P. 261–270.
221. Kim H.J., Litzenburger B.C., Cui X. et al. Con-
stitutively active type I insulin-like growth factor
receptor causes transformation and xenograft
growth of immortalized mammary epithelial cells
and is accompanied by an epithelial-to-mesen-
chymal transition mediated by NF-kappaB and
snail // Mol. Cell. Biol. – 2007. – 27, ¹ 8. –
P. 3165–3175.
222. Zhu T., Starling-Emerald B., Zhang X. et al.
Oncogenic transformation of human mammary
epithelial cells by autocrine human growth hor-
mone // Cancer Res. – 2005. – 65, ¹ 1. –
P. 317–24.
223. Mohankumar K.M., Perry J.K., Kannan N. et al.
Transcriptional activation of signal transducer and
activator of transcription (STAT) 3 and STAT5B
partially mediate homeobox A1- stimulated onco-
genic transformation of the immortalized human
mammary epithelial cell // Endocrinology . –
2008. – 149, ¹ 5. – P. 2219–2229.
224. Al Moustafa A.E., Foulkes W.D., Benlimame N. et al.
E6/E7 proteins of HPV type 16 and ErbB-2
cooperate to induce neoplastic transformation of
primary normal oral epithelial cells // Oncogene. –
2004. – 23, ¹ 2. – P. 350–358.
225. Kusakari T., Kariya M., Mandai M. et al. C-erbB-2
or mutant Ha-ras induced malignant transformation of
immortalized human ovarian surface epithelial cells
in vitro // Brit. J. Cancer. – 2003. – 89, ¹ 12. –
P. 2293–2298.
226. Ropiquet F., Huguenin S., Villette J.M. et al. FGF7/
KGF triggers cell transformation and invasion on im-
mortalised human prostatic epithelial PNT1A cells //
Int. J. Cancer. – 1999. – 82, ¹ 2. – P. 237–243.
227. Govindarajan B., Shah A., Cohen C. et al. Malig-
nant transformation of human cells by constitutive
expression of platelet-derived growth factor-BB //
J. Biol. Chem. – 2005. – 280, ¹ 14. – P. 13936–
13943.
228. Gutkind J.S., Novotny E.A., Brann M.R., Robbins K.C.
Muscarinic acetylcholine receptor subtypes as ago-
nist-dependent oncogenes // Proc. Nat. Acad. Sci.
USA. – 1991. – 88, ¹ 11. – P. 4703–4707.
229. Sun M., Wang G., Paciga J.E. et al. AKT1/PKBalpha
kinase is frequently elevated in human cancers and
its constitutive activation is required for oncogenic
transformation in NIH3T3 cells // Amer. J. Pathol. –
2001. – 159, ¹ 2. – P. 431–437.
230. Keren-Paz A., Bercovich Z., Porat Z. et al.
Overexpression of antizyme-inhibitor in NIH3T3
fibroblasts provides growth advantage through
neutralization of antizyme functions // Oncogene. –
2006. – 25, ¹ 37. – P. 5163–5172.
231. Tao W.J., Lin H., Sun T. et al. BCR-ABL oncogenic
transformation of NIH 3T3 fibroblasts requires the
IL-3 receptor BCR-ABL transformation of 3T3
cells requires IL-3R // Oncogene. – 2008. – 27,
¹ 22. – P. 3194–3200.
232. Xiang-yong L., Yang-chao C., Ke-yuan Z. et al.
Overexpression of Bax inhibitor-1 (BI-1) induces
cell transformation in NIH3T3 cells // Cell. Biol.
Int. – 2010. – 34, ¹ 11. – P. 1099–1104.
233. Lin R., Bagrodia S., Cerione R., Manor D. A Novel
Cdc42Hs mutant induces cellular transformation //
Curr. Biol. – 1997. – 7, ¹ 10. – P. 794–797.
234. Moiola C., De Luca P., Gardner K. et al. Cyclin T1
overexpression induces malignant transformation
and tumor growth // Cell Cycle. – 2010. – 9, ¹ 15. –
P. 3119–3126.
235. Anand N., Murthy S., Amann G. et al. Protein
elongation factor EEF1A2 is a putative oncogene in
ovarian cancer // Nat. Genet. – 2002. – 31, ¹ 3. –
P. 301–305.
236. Greulich H., Chen T.H., Feng W. et al. Oncogenic
transformation by inhibitor-sensitive and resistant
EGFR mutants // PLoS Med. – 2005. – 2, ¹ 11. –
P. e313.
237. Moscatelli D., Quarto N. Transformation of NIH
3T3 cells with basic fibroblast growth factor or
the hst/K-fgf oncogene causes downregulation of
the fibroblast growth factor receptor: reversal of
morphological transformation and restoration of
receptor number by suramin // J. Cell. Biol. –
1989. – 109, ¹ 5. – P. 2519–2527.
238. Fuller-Pace F., Peters G., Dickson C. Cell trans-
formation by kFGF requires secretion but not
glycosylation // J. Cell. Biol. – 1991. – 115, ¹ 2. –
P. 547–555.
239. MacArthur C.A., Lawshé A., Shankar D.B. et al.
FGF-8 isoforms differ in NIH3T3 cell transforming
potential // Cell Growth Differ. – 1995. – 6, ¹ 7. –
P. 817–825.
240. Hu Y., Ying H., Xu Y. hF-LANa, a human homologue
of Derlin family, regulating the expression of can-
cer-related genes promotes NIH3T3 cell trans-
formation // Cancer Lett. – 2007. – 258, ¹ 2. –
P. 171–180.
241. De Vivo M., Chen J., Codina J., Iyengar R. Enhan-
ced phospholipase C stimulation and transfor-
mation in NIH-3T3 cells expressing Q209LGq-
alpha-subunits // J. Biol. Chem. – 1992. – 267,
¹ 26. – P. 18263–18266.
242. Kroll S.D., Chen J., De Vivo M. et al. The
Q205LGo-alpha subunit expressed in NIH-3T3
cells induces transformation // J. Biol. Chem. –
1992. – 267, ¹ 32. – P. 23183–23188.
69ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
243. Ram P.T., Horvath C.M., Iyengar R. Stat3-mediated
transformation of NIH-3T3 cells by the constitutively
active Q205L Galphao protein // Science. – 2000. –
287, ¹ 5450. – P. 142–144.
244. Kuo W., Lin J., Tang T.K. Human glucose-6-
phosphate dehydrogenase (G6PD) gene transforms
NIH 3T3 cells and induces tumors in nude mice //
Int. J. Cancer. – 2000. – 85, ¹ 6. – P. 857–864.
245. Ha S.A., Shin S.M., Lee Y.J. et al. HCCRBP-1
directly interacting with HCCR-1 induces
tumorigenesis through P53 stabilization // Int. J.
Cancer. – 2008. – 122, ¹ 3. – P. 501–508.
246. Gao G., Peng M., Zhu L. et al. Human papillomavirus
16 variant E7 gene induces transformation of NIH 3T3
cells via up-regulation of cdc25A and cyclin À // Int. J.
Gynecol. Cancer. – 2009. – 19, ¹ 4. –P. 494–499.
247. Ryoo Z.Y., Jung B.K., Lee S.R. et al. Neoplastic
transformation and tumorigenesis associated with
overexpression of IMUP-1 and IMUP-2 genes in
cultured NIH/3T3 mouse fibroblasts // Biochem.
Biophys. Res. Communs. – 2006. – 349, ¹ 3. –
P. 995–1002.
248. Caruana G., Cambareri A.C., Gonda T.J., Ashman L.K.
Transformation of NIH3T3 fibroblasts by the
c-Kit receptor tyrosine kinase: effect of receptor
density and ligand-requirement // Oncogene. –
1998. – 16, ¹ 2. – P. 179–190.
249. Viswanathan S.R., Powers J.T., Einhorn W. et al.
Lin28 promotes transformation and is associated
with advanced human malignancies // Nat.
Genet. – 2009. – 41, ¹ 7. – P. 843–848.
250. Fridman R., Sweeney T.M., Zain M. et al. Malig-
nant transformation of NIH-3T3 cells after
subcutaneous co-injection with a reconstituted
basement membrane (matrigel) // Int. J. Cancer . –
1992. – 51, ¹ 5. – P. 740–754.
251. Kadomatsu K., Hagihara M., Akhter S. et al. Midkine
induces the transformation of NIH3T3 cells // Brit.
J. Cancer. – 1997. – 75, ¹ 3. – P. 354–359.
252. Komiya K., Sueoka-Aragane N., Sato A. et al.
Mina53, a novel c-Myc target gene, is frequently
expressed in lung cancers and exerts oncogenic
property in NIH/3T3 cells // J. Cancer Res. Clin.
Oncol. – 2010. – 136, ¹ 3. – P. 465–473.
253. Bafna S., Singh A.P., Moniaux N. et al. MUC4,
a multifunctional transmembrane glycoprotein,
induces oncogenic transformation of NIH3T3
mouse fibroblast cells // Cancer Res. – 2008. –
68, ¹ 22. – P. 9231–9238.
254. Piestun D., Kochupurakkal B.S., Jacob-Hirsch J. et al.
Nanog transforms NIH3T3 cells and targets cell-
type restricted genes // Biochem. Biophys. Res.
Communs. – 2006. – 343, ¹ 1. – P. 279–285.
255. Moshier J.A., Dosescu J., Skunca M., Luk G.D. Trans-
formation of NIH/3T3 cells by ornithine decar-
boxylase overexpression // Cancer Res. – 1993. –
53, ¹ 11. – P. 2618–2622.
256. Moshier J.A., Malecka-Panas E., Geng H. et al. Orni-
thine decarboxylase transformation of NIH/3T3
cells is mediated by altered epidermal growth
factor receptor activity // Cancer Res. – 1995. –
55, ¹ 22. – P. 5358–5365.
257. Platica M., Ivan E., Ionescu A. et al. Transformation
of NIH 3T3 cells by enhanced PAR expression //
Biochem. Biophys. Res. Communs. – 2004. – 314,
¹ 3. – P. 891–896.
258. MacArthur L.H., Clarke M.F., Westin E.H. Malig-
nant transformation of NIH 3T3 fibroblasts by
human c-sis is dependent upon the level of
oncogene expression // Mol. Carcinog. – 1992. –
5, ¹ 4. – P. 311–319.
259. Kim H.R., Upadhyay S., Korsmeyer S., Deuel T.F.
Platelet-derived growth factor (PDGF) B and A
homodimers transform murine fibroblasts depen-
ding on the genetic background of the cell // J.
Biol. Chem. – 1994. – 269, ¹ 48. – P. 30604–
30608.
260. Chauhan A.K., Li Y.S., Deuel T.F. Pleiotrophin
transforms NIH 3T3 cells and induces tumors in
nude mice // Proc. Nat. Acad. Sci. USA. – 1993. –
90, ¹ 2. – P. 679–682.
261. Tabib A., Bachrach U. Role of polyamines in
mediating malignant transformation and oncogene
expression // Int. J. Biochem. Cell Biol. – 1999. –
31, ¹ 11. – P. 1289–1295.
262. Cranston A., Carniti C., Martin S. et al. A Novel acti-
vating mutation in the RET tyrosine kinase do-
main mediates neoplastic transformation // Mol.
Endocrinol. – 2006. – 20, ¹ 7. – P. 1633– 1643.
263. Shalloway D., Johnson P.J., Freed E.O. et al.
Transformation of NIH 3T3 Cells by Cotransfection
with c-src and Nuclear Oncogenes // Mol. Cell
Biol. – 1987. – 7, ¹ 10. – P. 3582–3590.
264. Shalloway D., Coussens P.M., Yaciuk P. Overexpres-
sion of the c-src protein does not induce
transformation of NIH 3T3 cells // Proc. Nat.
Acad. Sci. USA. – 1984. – 81, ¹ 22. – P. 7071–
7075.
265. Bromberg J.F., Wrzeszczynska M.H., Devgan G. et al.
Stat3 as an oncogene // Cell. – 1999. – 98, ¹ 3. –
P. 295–303.
266. Radhakrishnan V.M., Martinez J.D. 14-3-3gamma
induces oncogenic transformation by stimulating
MAP kinase and PI3K signaling // PLoS One. –
2010. – 5, ¹ 7. – P. e11433.
267. Wang C., Lisanti M.P., Liao D.J. Reviewing once
more the c-myc and Ras collaboration: converging
at the cyclin D1-CDK4 complex and challenging
basic concepts of cancer biology // Cell Cycle. –
2011. – 10, ¹ 1. – P. 57–67.
70 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
268. Meloche S., Pouysségur J. The ERK1/2 mitogen-
activated protein kinase pathway as a master
regulator of the G1- to S-phase transition //
Oncogene. – 2007. – 26, ¹ 22. – P. 3227–3239.
269. McCubrey J.A., Steelman L.S., Chappell W.H. et al.
Roles of the Raf/MEK/ERK pathway in cell
growth, malignant transformation and drug re-
sistance // Biochim. Biophys. acta. – 2007. – 1773,
¹ 8. – P. 1263–1284.
270. Chambard J.C., Lefloch R., Pouysségur J., Lenor-
mand P. ERK implication in cell cycle regulation //
Biochim. Biophys. acta. – 2007. – 1773, ¹ 8. –
P. 1299–1310.
271. Jiang B.H., Liu L.Z. PI3K/PTEN signaling in
angiogenesis and tumorigenesis // Adv. Cancer
Res. – 2009. – 102. – P. 19–65.
272. Dang C.V., O’Donnell K.A., Zeller K.I. et al. The
c-Myc target gene network // Semin. Cancer
Biol. – 2006. – 16, ¹ 4. – P. 253–264.
273. Rodríguez J., Calvo F., González J.M. et al. ERK1/2
MAP kinases promote cell cycle entry by rapid,
kinase-independent disruption of retinoblastoma-
lamin A complexes // J. Cell. Biol. – 2010. – 191,
¹ 5. – P. 967–979.
274. Manning B.D., Cantley L.C. AKT/PKB signaling:
navigating downstream // Cell. – 2007. – 129, ¹
7. – P. 1261–1274.
275. Rosner M., Fuchs C., Siegel N. et al. Functional
interaction of mammalian target of rapamycin
complexes in regulating mammalian cell size and cell
cycle // Hum. Mol. Genet. – 2009. – 18, ¹ 17. –
P. 3298–3310.
276. Menon S., Manning B.D. Common corruption of
the mTOR signaling network in human tumors //
Oncogene. – 2008. – Suppl 2:S43–51.
277. Zoncu R., Efeyan A., Sabatini D.M. mTOR: from
growth signal integration to cancer, diabetes and
ageing // Nat. Rev. Mol. Cell Biol. – 2011. – 12,
¹ 1. – P. 21–35.
278. Winter J.N., Jefferson L.S., Kimball S.R. The ERK
and akt signaling pathways function through parallel
mechanisms to promote mTORC1 // Amer. J.
Physiol. Cell Physiol. – 2011. – 300, ¹ 5. –
P. 1172–1180.
279. Fingar D.C., Richardson C.J., Tee A.R. et al.
mTOR controls cell cycle progression through its
cell growth effectors S6K1 and 4E-BP1/eukaryotic
translation initiation factor 4E // Mol. Cell. Biol. –
2004. – 24, ¹ 1. – P. 200–216.
280. Dowling R.J., Topisirovic I., Alain T. et al. mTORC1-
mediated cell proliferation, but not cell growth,
controlled by the 4E-BPs // Science. – 2010. –
328, ¹ 5982. – P. 1172–1176.
281. Tarn W.Y, Lai M.C. Translational control of cyclins //
Cell Div. – 2011. – 6, ¹ 1.
282. Oyama K., Okawa T., Nakagawa H. et al. AKT
induces senescence in primary esophageal epithelial
cells but is permissive for differentiation as revealed
in organotypic culture // Oncogene. – 2007. – 26,
¹ 16. – P. 2353–2364.
283. Nogueira V., Park Y., Chen C.C. et al. Akt determines
replicative senescence and oxidative or oncogenic
premature senescence and sensitizes cells to oxidative
apoptosis // Cancer Cell. – 2008. – 14, ¹ 6. –
P. 458–470.
284. Miyauchi H., Minamino T., Tateno K., et al. Akt
negatively regulates the in vitro lifespan of human
endothelial cells via a p53/p21-dependent pathway //
EMBO J. – 2004. – 23, ¹ 1. – P. 212–220.
285. Minamino T., Miyauchi H., Tateno K. et al. Akt-
induced cellular senescence: implication for human
disease // Cell Cycle. – 2004. – 3, ¹ 4. – P. 449–
451.
286. Besson A., Yong V.W. Involvement of p21(Waf1/
Cip1) in protein kinase C alpha-induced cell cycle
progression // Mol. Cell Biol. – 2000. – 20, ¹
13. – P. 4580–4590.
287. Coleman M.L., Marshall C.J., Olson M.F. Ras promo-
tes p21(Waf1/Cip1) protein stability via a cyclin D1-
imposed block in proteasome-mediated degradation //
EMBO J. – 2003. – 22, ¹ 9. – P. 2036–2046.
288. Ciccarelli C., Marampon F., Scoglio A. et al. p21WAF1
expression induced by MEK/ERK pathway activation
or inhibition correlates with growth arrest, myogenic
differentiation and onco– phenotype reversal in
rhabdomyosarcoma cells // Mol. Cancer. – 2005. –
4, ¹ 41.
289. Yang X., Wang W., Fan J. et al. Prostaglandin
A2-mediated stabilization of p21 mRNA through
an ERK-dependent pathway requiring the RNA–
binding protein HuR // J. Biol. Chem. – 2004. –
279, ¹ 47. – P. 49298–49306.
290. Hu T.H., Tai M.H., Chuah S.K. et al. Elevated p21
expression is associated with poor prognosis of
rectal stromal tumors after resection // J. Surg.
Oncol. – 2008. – 98, ¹ 2. – P. 117–123.
291. Takeshima Y., Yamasaki M., Nishisaka T. et al.
p21WAF1/CIP1 expression in primary lung adeno-
carcinomas: heterogeneous expression in tumor
tissues and correlation with p53 expression and
proliferative activities // Carcinogenesis. – 1998. –
19, ¹ 10. – P. 1755–1761.
292. Korkolopoulou P., Konstantinidou A.E., Thomas-
Tsagli E. et al. WAF1/p21 protein expression is
an independent prognostic indicator in superficial
and invasive bladder cancer // Appl. Immun. Mol.
Morphol. – 2000. – 8, ¹ 4. – P. 285–292.
293. Abdulamir A.S., Hafidh R.R., Mahdi L.K. et al. The
Interplay between p53 and p21 Tumor suppressor
proteins in the transformation of colorectal
71ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
adenoma to carcinoma // Amer. J. Immunol. –
2008. – 4, ¹ 2. – P. 14–22.
294. Kamiya M., Nakazato Y. The expression of
p73, p21 and MDM2 proteins in gliomas // J.
Neurooncol. – 2002. – 59, ¹ 2. – P. 143–149.
295. Zhang M.F., Zhang Z.Y., Fu J. et al. Correlation bet-
ween expression of p53, p21/WAF1, and MDM2
proteins and their prognostic significance in prima-
ry hepatocellular carcinoma // J. Transl. Med. –
2009. – 7, ¹ 110.
296. Mouriaux F., Maurage C.A., Labalette P. et al. Cyclin-
dependent kinase inhibitory protein expression in
human choroidal melanoma tumors // Invest. Op-
hthalmol. Vis. Sci. – 2000. – 41, ¹ 10. – P. 2837–
2843.
297. Peeper D.S. Ras and pRb: the relationship gets yet
more intimate // Cancer Cell. – 2009. – 15, ¹ 4. –
P. 243–245.
298. Slingerland J., Pagano M. Regulation of the cdk
inhibitor p27 and its deregulation in cancer // J.
Cell Physiol. – 2000. – 183, ¹ 1. – P. 10–17.
299. Nicholson J.M., Duesberg P. On the karyotypic origin
and evolution of cancer cells // Cancer Genet.
Cytogenet. – 2009. – 194, ¹ 2. – P. 96–110.
300. Klein A., Li N., Nicholson J.M., McCormack A.A.
et al. Transgenic oncogenes induce oncogene-
independent cancers with individual karyotypes and
phenotypes // Cancer Genet. Cytogenet. – 2010. –
200, ¹ 2. – P. 79–99.
301. Li L., McCormack A.A., Nicholson J.M. et al. Cancer-
causing karyotypes: chromosomal equilibria between
destabilizing aneuploidy and stabilizing selection for
oncogenic function // Cancer Genet. Cytogenet. –
2009. – 188, ¹ 1. – P. 1–25.
302. Duesberg P., Mandrioli D., McCormack A.,
Nicholson J.M. Is carcinogenesis a form of
speciation? // Cell Cycle. – 2011. – 10, ¹ 13.
303. Halazonetis T.D., Gorgoulis V.G., Bartek J. An onco-
gene-induced DNA damage model for cancer
development // Science. – 2008. – 319, ¹ 5868. –
P. 1352–1355.
304. Bylund L., Kytölä S., Lui W.O. et al. Analysis of
the cytogenetic stability of the human embryonal
kidney cell line 293 by cytogenetic and STR pro-
filing approaches // Cytogenet. Genome Res. –
2004. – 106, ¹ 1. – P. 28–32.
305. Louis N., Evelegh C., Graham F.L. Cloning and se-
quencing of the cellular-viral junctions from the
human adenovirus type 5 transformed 293 cell line //
Virology. – 1997. – 233, ¹ 2. – P. 423–429.
306. Zur Hausen H. Induction of specific chromosomal
aberrations by adenovirus type 12 in human embryonic
kidney cells // J. Virol. – 1967. – 1, ¹ 6. – P. 1174–
1185.
307. McDougall J.K. Adenovirus-induced chromosome
aberrations in human cells // J. Gen. Virol . –
1971. – 12, ¹ 1. – P. 43–51.
308. Kim J.H., Choi E.Y., Jung E.-S. et al. Characterization
of clones of human cell line infected with porcine
endogenous retrovirus (PERV) from porcine cell
line, PK-15 // Infection and Chemotherapy. –
2009. – 41, ¹ 1. – P. 1–8.
309. Shen C., Gu M., Song C. et al. The tumorigenicity
diversification in human embryonic kidney 293
cell line cultured in vitro // Biologicals. – 2008. –
36, ¹ 4. – P. 263–268.
310. Rubin A.L., Arnstein P., Rubin H. Physiological in-
duction and reversal of focus formation and tumo-
rigenicity in NIH 3T3 cells. // Proc. Nat. Acad.
Sci. USA. – 1990. – 87, ¹ 24. – P. 10005–10009.
311. Rubin A.L., Ellison B.J. Induction of transformation
in NIH3T3 cells by moderate growth constraint:
evidence that neoplasia is driven by adaptational
change // Carcinogenesis. – 1991. – 12, ¹ 10. –
P. 1801–1806.
312. Rubin H. Cellular epigenetics: effects of passage
history on competence of cells for «spontaneous»
transformation // Proc. Nat. Acad. Sci. USA. –
1993. – 90, ¹ 22. – P. 10715–10719.
313. Rubin H. The role of selection in progressive neo-
plastic transformation // Adv. Cancer Res. – 2001. –
83. – P. 159–207.
314. Rubin H. Multistage carcinogenesis in cell culture //
Dev. Biol (Basel). – 2001. – 106. – P. 61–66.
315. Rubin H. Cell-cell contact interactions conditionally
determine suppression and selection of the neoplastic
phenotype // Proc. Nat. Acad. Sci. USA. – 2008. –
105, ¹ 17. – P. 6215–6221.
316. Millau J.F., Mai S., Bastien N., Drouin R. p53
functions and cell lines: have we learned the
lessons from the past?// Bioessays. – 2010. – 32,
¹ 5. – P. 392–400.
317. Nielsen K.V., Briand P. Cytogenetic analysis of in
vitro karyotype evolution in a cell line established
from nonmalignant human mammary epithelium //
Cancer Genet. Cytogenet. – 1989. – 39, ¹ 1. –
P. 103–118.
318. Bartholdi M.F., Ray F.A., Cram L.S., Kraemer P.M.
Karyotype instability of Chinese hamster cells du-
ring in vivo tumor progression // Somat. Cell.
Mol. Genet. – 1987. – 13, ¹ 1. – P. 1–10.
319. Duesberg P., Rasnick D. Aneuploidy, the somatic
mutation that makes cancer a species of its own // Cell
Motil. Cytoskeleton. – 2000. – 47, ¹ 2. – P. 81–107.
320. Thorgeirsson U.P., Turpeenniemi-Hujanen T., Wil-
liams J.E. et al. NIH/3T3 cells transfected with
human tumor DNA containing activated ras
oncogenes express the metastatic phenotype in
nude mice // Mol. Cell. Biol. – 1985. – 5, ¹ 1. –
P. 259–262.
72 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
321. Kuzumaki N., Ogiso Y., Oda A. et al. Resistance
to oncogenic transformation in revertant R1 of
human ras-transformed NIH 3T3 cells // Mol.
Cell. Biol. – 1989. – 9, ¹ 5. – P. 2258–2263.
322. Li Y., Weibing S., Liu H. et al. Mitochondrial DNA
from colorectal cancer cells promotes the malignant
phenotype of NIH3T3 cells. // Cell Biol. Int. –
2008. – 32, ¹ 8. – P. 979–983.
323. Zhang K., Sun J., Liu N. et al. Transformation
of NIH 3T3 Cells by HER3 or HER4 Receptors
Requires the Presence of HER1 or HER2 // J.
Biol. Chem. – 1996. – 271, ¹ 7. – P. 3884–3890.
324. Kadota M., Yang H.H., Gomez B. et al. Delineating
genetic alterations for tumor progression in the
MCF10A series of breast cancer cell lines // PLoS
One. – 2010. – 5, ¹ 2. – P. e9201.
325. Zhang D.L., Ji L., Li L.J., Huang G.S. Systematically
experimental investigation on carcinogenesis or
tumorigenicity of VERO cell lines of different
karyotypes in nude mice in vivo used for viral
vaccine manufacture // Acta Genet. Sin. – 2004. –
31, ¹ 7. – P. 647–660.
326. Ragel B.T., Couldwell W.T., Gillespie D.L. et al. A
comparison of the cell lines used in meningioma
research // Surg. Neurol. – 2008. – 70, ¹ 3. – P. 295–
307.
327. Thompson S.L., Compton D.A. Examining the link
between chromosomal instability and aneuploidy
in human cells // J. Cell Biol. – 2008. – 180,
¹ 4. – P. 665–672.
328. Roschke A.V., Tonon G., Gehlhaus K.S. et al. Karyo-
typic ñomplexity of the NCI-60 drug- screening
panel // Cancer Res. – 2003. – 63, ¹ 24. – P. 8634–
8647.
329. Bussey K.J., Chin K., Lababidi S. et al. Integrating
data on DNA copy number with gene expression
levels and drug sensitivities in the NCI-60 cell line
panel // Mol. Cancer Ther. – 2006. – 5, ¹ 4. –
P. 853–867.
330. Lundberg E., Fagerberg L., Klevebring D. et al.
Defining the transcriptome and proteome in three
functionally different human cell lines // Mol.
Syst. Biol. – 2010. – 6, ¹ 450.
331. Valsesia A., Rimoldi D., Martinet D. et al. Network-
guided analysis of genes with altered somatic copy
number and gene expression reveals pathways
commonly perturbed in metastatic melanoma //
PLoS One. – 2011. – 6, ¹ 4. – P. e18369.
332. Geiger T., Cox J., Mann M. Proteomic changes resulting
from gene copy number variations in cancer cells //
PLoS Genet. – 2010. – 6, ¹ 9. – P. e1001090.
333. Pavelka N., Rancati G., Zhu J. et al. Aneuploidy
confers quantitative proteome changes and pheno-
typic variation in budding yeast // Nature. – 2010. –
468, ¹ 7321. – P. 321–325.
334. Bajaj R., Xu F., Xiang B. et al. Evidence-based
genomic diagnosis characterized chromosomal and
cryptic imbalances in 30 elderly patients with
myelodysplastic syndrome and acute myeloid leu-
kemia // Mol. Cytogenet. – 2011. – 4, ¹ 3.
335. Hurst C.D., Fiegler H., Carr P. et al. High-resolution
analysis of genomic copy number alterations in
bladder cancer by microarray-based comparative
genomic hybridization // Oncogene. – 2004. – 23,
¹ 12. – P. 2250–2263.
336. Qin S.L., Chen X.J., Xu X. et al. Detection of
chromosomal alterations in bladder transitional cell
carcinomas from northern China by comparative
genomic hybridization // Cancer Lett. – 2006. –
238, ¹ 2. – P. 230–239.
337. Prat E., del Rey J., Ponsa I. et al. Comparative geno-
mic hybridization analysis reveals new different sub-
groups in early-stage bladder tumors // Urology. –
2010. – 75, ¹ 2. – P. 347–355.
338. Nikolsky Y., Sviridov E., Yao J. et al. Genome-
wide functional synergy between amplified and
mutated genes in human breast cancer // Cancer
Res. – 2008. – 68, ¹ 22. – P. 9532–9540.
339. Stephens P.J., McBride D.J., Lin M.L. et al.
Complex landscapes of somatic rearrangement in
human breast cancer genomes // Nature. – 2009. –
462, ¹ 7276. – P. 1005–1010.
340. Russnes H.G., Vollan H.K., Lingjaerde O.C. et al.
Genomic architecture characterizes tumor progres-
sion paths and fate in breast cancer patients // Sci.
Transl. Med. – 2010. – 2, ¹ 38. – P. 38–47.
341. Smid M., Hoes M., Sieuwerts A.M. et al. Patterns
and incidence of chromosomal instability and their
prognostic relevance in breast cancer subtypes. //
Breast Cancer Res. Treat. – 2011. – 128, ¹ 1. –
P. 23–30.
342. Navin N., Krasnitz A., Rodgers L. et al. Inferring
tumor progression from genomic heterogeneity //
Genome Res. – 2010. – 20, ¹ 1. – P. 68–80.
343. Heselmeyer K., Macville M., Schröck E. et al.
Advanced-stage cervical carcinomas are defined by
a recurrent pattern of chromosomal aberrations
revealing high genetic instability and a consistent
gain of chromosome arm 3q // Genes Chrom.
Cancer. – 1997. – 19, ¹ 4. – P. 233–240.
344. Kloth J.N., Oosting J., van Wezel T. et al. Combined
array-comparative genomic hybridization and
single-nucleotide polymorphism-loss of hetero-
zygosity analysis reveals complex genetic altera-
tions in cervical cancer // BMC Genom. – 2007. –
8, ¹ 53.
345. He Q.J., Zeng W.F., Sham J.S. et al. Recurrent ge-
netic alterations in 26 colorectal carcinomas and 21
adenomas from Chinese patients // Cancer Genet.
Cytogenet. – 2003. – 144, ¹ 2. – P. 112–118.
73ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
346. Gaasenbeek M., Howarth K., Rowan A.J. et al.
Combined array-comparative genomic hybridization
and single-nucleotide polymorphism-loss of hetero-
zygosity analysis reveals complex changes and multiple
forms of chromosomal instability in colorectal
cancers // Cancer Res. – 2006. – 66, ¹ 7. –
P. 3471–3479.
347. Xiao X.Y., Zhou X.Y., Yan G. et al. Chromosomal
alteration in Chinese sporadic colorectal carci-
nomas detected by comparative genomic hybridi-
zation // Diagn. Mol. Pathol. – 2007. – 16, ¹ 2. –
P. 96–103.
348. Castorina S., Barresi V., Luca T. et al. Recent
advances in molecular diagnostics of colorectal cancer
by genomic arrays: proposal for a procedural shift in
biological sampling and pathological report // Ital. J.
Anat. Embryol. – 2010. – 115, ¹ 1/2. – P. 39–45.
349. Micci F., Teixeira M.R., Haugom L. et al. Genomic
aberrations in carcinomas of the uterine corpus //
Genes Chrom. Cancer. – 2004. – 40, ¹ 3. – P. 229–
246.
350. Shing D.C., Morley-Jacob C.A., Roberts I. et al. Ewing's
tumour: novel recurrent chromosomal abnormalities
demonstrated by molecular cytogenetic analysis of
seven cell lines and one primary culture // Cytogenet.
Genome Res. – 2002. – 97, ¹ 1/2. – P. 20–27.
351. Takada H., Imoto I., Tsuda H. et al. Screening of DNA
copy-number aberrations in gastric cancer cell lines
by array-based comparative genomic hybridization //
Cancer Sci. – 2005. – 96, ¹ 2. – P. 100–110.
352. Veltman I., Veltman J., Janssen I. et al. Identification
of recurrent chromosomal aberrations in germ cell
tumors of neonates and infants using genomewide
array-based comparative genomic hybridization //
Genes Chrom. Cancer. – 2005. – 43, ¹ 4. –
P. 367–376.
353. Kim D.H., Mohapatra G., Bollen A. et al.
Chromosomal abnormalities in glioblastoma
multiforme tumors and glioma cell lines detected
by comparative genomic hybridization // Int. J.
Cancer. – 1995. – 60, ¹ 6. – P. 812–819.
354. Inda M.M., Fan X., Muñoz J. et al. Chromosomal
abnormalities in human glioblastomas: gain in
chromosome 7p correlating with loss in chromo-
some 10q // Mol. Carcinog. – 2003. – 36, ¹ 1. –
P. 6–14.
355. Roversi G., Pfundt R., Moroni R.F. et al. Identification
of novel genomic markers related to progression
to glioblastoma through genomic profiling of 25
primary glioma cell lines // Oncogene. – 2006. –
25, ¹ 10. – P. 1571–1583.
356. Vranová V., Necesalová E., Kuglík P. et al. Screening of
genomic imbalances in glioblastoma multiforme using
high-resolution comparative genomic hybridization //
Oncol. Rep. – 2007. – 17, ¹ 2. – P. 457–464.
357. Gardina P.J., Lo K.C., Lee W. et al. Ploidy status and
copy number aberrations in primary glioblastomas
defined by integrated analysis of allelic ratios,
signal ratios and loss of heterozygosity using 500K
SNP Mapping Arrays // BMC Genom. – 2008. –
9, ¹ 489.
358. Lo K.C., Bailey D., Burkhardt T. et al. Comprehensive
analysis of loss of heterozygosity events in glioblastoma
using the 100K SNP mapping arrays and comparison
with copy number abnormalities defined by BAC array
comparative genomic hybridization // Genes Chrom.
Cancer. – 2008. – 47, ¹ 3. – P. 221–227.
359. Dahlback H.S., Brandal P., Meling T.R. et al. Geno-
mic aberrations in 80 cases of primary glioblastoma
multiforme: Pathogenetic heterogeneity and putative
cytogenetic pathways // Genes Chrom. Cancer. –
2009. – 48, ¹ 10. – P. 908–924.
360. Speicher M.R., Howe C., Crotty P. et al. Compa-
rative genomic hybridization detects novel dele-
tions and amplifications in head and neck
squamous cell carcinomas // Cancer Res. – 1995. –
55, ¹ 5. – P. 1010–1013.
361. Bockmühl U., Schwendel A., Dietel M., Petersen I.
Distinct patterns of chromosomal alterations in
high- and low-grade head and neck squamous cell
carcinomas // Cancer Res. – 1996. – 56, ¹ 23. –
P. 5325–5329.
362. Campbell P.J., Stephens P.J., Pleasance E.D. et al.
Identification of somatically acquired rearrange-
ments in cancer using genome-wide massively
parallel paired-end sequencing // Nat. Genet. –
2008. – 40, ¹ 6. – P. 722–729.
363. Belloni E., Veronesi G., Micucci C. et al. Genomic
characterization of asymptomatic CT- detected
lung cancers // Oncogene. – 2011. – 30, ¹ 9. –
P. 1117–1126.
364. Tiu R.V., Gondek L.P., O’Keefe C.L. et al. Prognostic
impact of SNP array karyotyping in myelodysplastic
syndromes and related myeloid malignancies //
Blood. – 2011. – 117, ¹ 17. – P. 4552–4560.
365. Ambatipudi S., Gerstung M., Gowda R. et al.
Genomic profiling of advanced-stage oral cancers
reveals chromosome 11q alterations as markers of
poor clinical outcome // PLoS One. – 2011. – 6,
¹ 2. – P. e17250.
366. Welkoborsky H.J., Bernauer H.S., Riazimand H.S.,
et al. Patterns of chromosomal aberrations in
metastasizing and nonmetastasizing squamous cell
carcinomas of the oropharynx and hypopharynx //
Ann. Otol. Rhinol. Laryngol. – 2000. – 109, ¹ 4. –
P. 401–410.
367. Hauptmann S., Denkert C., Koch I. et al. Genetic
alterations in epithelial ovarian tumors analyzed
by comparative genomic hybridization // Hum.
Pathol. – 2002. – 33, ¹ 6. – P. 632–641.
74 ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
A.A. Stepanenko, V.M. Kavsan
368. Mahlamäki E.H., Höglund M., Gorunova L. et al.
Comparative genomic hybridization reveals frequent
gains of 20q, 8q, 11q, 12p, and 17q, and losses of
18q, 9p, and 15q in pancreatic cancer // Genes
Chrom. Cancer. – 1997. – 20, ¹ 4. – P. 383–391.
369. Campbell P.J., Yachida S., Mudie L.J. et al. The
patterns and dynamics of genomic instability in
metastatic pancreatic cancer // Nature. – 2010. –
467, ¹ 7319. – P. 1109–1113.
370. Berger M.F., Lawrence M.S., Demichelis F. et al.
The genomic complexity of primary human pros-
tate cancer // Nature. – 2011. – 470, ¹ 7333. –
P. 214–220.
371. Wada N., Duh Q.Y., Miura D. et al. Chromosomal
aberrations by comparative genomic hybridization
in hürthle cell thyroid carcinomas are associated
with tumor recurrence // J. Clin. Endocrinol.
Metab. – 2002. – 87, ¹ 10. – P. 4595–4601.
372. Mattison J., Kool J., Uren A.G. et al. Novel candidate
cancer genes identified by a large- scale cross-species
comparative oncogenomics approach // Cancer
Res. – 2010. – 70, ¹ 3. – P. 883–895.
373. Beroukhim R., Mermel C.H., Porter D. et al. The
landscape of somatic copy-number alteration across
human cancers // Nature. – 2010. – 463, ¹ 7283. –
P. 899–905.
374. Nobusawa S., Lachuer J., Wierinckx A. et al.
Intratumoral patterns of genomic imbalance in
glioblastomas // Brain Pathol. – 2010. – 20, ¹ 5. –
P. 936–944.
375. Tyson J., Majerus T.M., Walker S., Armour J.A.
Screening for common copy-number variants in
cancer genes // Cancer Genet. Cytogenet. – 2010. –
203, ¹ 2. – P. 316–323.
376. Fox E.J., Salk J.J., Loeb L.A. Cancer genome
sequencing – an interim analysis // Cancer Res. –
2009. – 69, ¹ 12. – P. 4948–4950.
377. Gerlinger M., Swanton C. How Darwinian models
inform therapeutic failure initiated by clonal
heterogeneity in cancer medicine // Brit. J.
Cancer. – 2010. – 103, ¹ 8. – P. 1139–1143.
378. Swanton C., Burrell R.A., Futreal P.A. Breast cancer
genome heterogeneity: a challenge to personalised
medicine? // Breast Cancer Res. – 2011. – 13,
¹ 1. – P. 104.
379. Duesberg P., Li R., Sachs R. et al. Cancer drug
resistance: the central role of the karyotype // Drug
Resist. Updat. – 2007. – 10, ¹ 1– 2. – P. 51–
58.
380. Monni O., Joensuu H., Franssila K., Knuutila S.
DNA copy number changes in diffuse large B-cell
lymphoma – comparative genomic hybridization
study // Blood. – 1996. – 87, ¹ 12. – P. 5269–
5278.
381. Frank C.J., McClatchey K.D., Devaney K.O., Ca-
rey T.E. Evidence that loss of chromosome 18q is
associated with tumor progression // Cancer Res. –
1997. – 57, ¹ 5. – P. 824–827.
382. Rickert C.H., Dockhorn-Dworniczak B., Busch G. et
al. Increased chromosomal imbalances in recurrent
pituitary adenomas // Acta Neuropathol. – 2001. –
102, ¹ 6. – P. 615–620.
383. Nishio J., Iwasaki H., Ishiguro M. et al. Synovial
sarcoma with a secondary chromosome change
der(22)t(17;22)(q12;q12) // Cancer Genet. Cyto-
genet. – 2002. – 137, ¹ 1. – P. 23–28.
384. Gömöri E., Fülöp Z., Mészáros I. et al. Microsatellite
analysis of primary and recurrent glial tumors
suggests different modalities of clonal evolution
of tumor cells // J. Neuropathol. Exp. Neurol. –
2002. – 61, ¹ 5. – P. 396–402.
385. Waldman F.M., DeVries S., Chew K.L. et al.
Chromosomal alterations in ductal carcinomas in
situ and their in situ recurrences // J. Nat. Cancer
Inst. – 2000. – 92, ¹ 4. – P. 313–320.
386. Ness G.O., Lybaek H., Arnes J., Rødahl E.
Chromosomal imbalances in a recurrent solitary
fibrous tumor of the orbit // Cancer Genet.
Cytogenet. – 2005. – 162, ¹ 1. – P. 38–44.
387. Heinrich U.R., Brieger J., Gosepath J. et al.
Frequent chromosomal gains in recurrent juvenile
nasopharyngeal angiofibroma // Cancer Genet.
Cytogenet. – 2007. – 175, ¹ 2. – P. 138–143.
388. Korshunov A., Benner A., Remke M. et al. Accumulation
of genomic aberrations during clinical progression of
medulloblastoma // Acta Neuropathol. – 2008. –
116, ¹ 4. – P. 383–390.
389. Zieger K., Wiuf C., Jensen K.M. et al. Chromosomal
imbalance in the progression of high-risk non-
muscle invasive bladder cancer // BMC Cancer. –
2009. – 9, ¹ 149.
390. Cimini D., Degrassi F. Aneuploidy: a matter of bad
connections // Trends Cell. Biol. – 2005. – 15,
¹ 8. – P. 442–451.
391. Li J.J., Li S.A. Mitotic kinases: the key to dup-
lication, segregation, and cytokinesis errors, chro-
mosomal instability, and oncogenesis // Pharma-
col. Ther. – 2006. – 111, ¹ 3. – P. 974–984.
392. Cimini D. Merotelic kinetochore orientation, aneu-
ploidy, and cancer // Biochim. Biophys. acta. –
2008. – 1786, ¹ 1. – P. 32–40.
393. Holland A.J., Cleveland D.W. Boveri revisited:
chromosomal instability, aneuploidy and tumo-
rigenesis // Nat. Rev. Mol. Cell. Biol. – 2009. –
10, ¹ 7. – P. 478–487.
394. McClelland S.E., Burrell R.A., Swanton C. Chromo-
somal instability: a composite phenotype that
influences sensitivity to chemotherapy // Cell
Cycle. – 2009. – 8, ¹ 20. – P. 3262–3266.
395. Chandhok N.S., Pellman D. A little CIN may cost
75ISSN 0564–3783. Öèòîëîãèÿ è ãåíåòèêà. 2012. ¹ 2
Immortalization and malignant transformation of eukaryotic cells
a lot: revisiting aneuploidy and cancer // Curr.
Opin. Genet. Dev. – 2009. – 19, ¹ 1. – P. 74–
81.
396. Rao C.V., Yamada H.Y., Yao Y., Dai W. Enhanced
genomic instabilities caused by deregulated micro-
tubule dynamics and chromosome segregation: a
perspective from genetic studies in mice // Carcino-
genesis. – 2009. – 30, ¹ 9. – P. 1469–1474.
397. Dillon L.W., Burrow A.A., Wang Y.H. DNA
instability at chromosomal fragile sites in cancer //
Curr. Genom. – 2010. – 11, ¹ 5. – P. 326–337.
398. Ting D.T., Lipson D., Paul S. et al. Aberrant
overexpression of satellite repeats in pancreatic
and other epithelial cancers // Science. – 2011. –
331, ¹ 6017. – P. 593–596.
399. Krajcovic M., Johnson N.B., Sun Q. et al. A Non-
genetic route to aneuploidy in human cancers //
Nat. Cell. Biol. – 2011. – 13, ¹ 3. – P. 324–330.
400. Kitada K., Taima A., Ogasawara K. et al. Chromosome-
specific segmentation revealed by structural analysis of
individually isolated chromosomes // Genes Chrom.
Cancer. – 2011. – 50, ¹ 4. – P. 217–227.
401. Tubio J.M., Estivill X. Cancer : When catastrophe
strikes a cell // Nature. – 2011. – 470, ¹ 7335. –
P. 476–477.
402. Parsons D.W., Li M., Zhang X. et al. The genetic
landscape of the childhood cancer medulloblastoma //
Science. – 2011. – 331, ¹ 6016. – P. 435–439.
403. Kan Z., Jaiswal B.S., Stinson J. et al. Diverse somatic
mutation patterns and pathway alterations in human
cancers // Nature. – 2010. – 466, ¹ 7308. – P. 869–
873.
404. Pleasance E.D., Cheetham R.K., Stephens P.J. et al.
A comprehensive catalogue of somatic mutations
from a human cancer genome // Nature. – 2010. –
463, ¹ 7278. – P. 191–196.
403. Kan Z., Jaiswal B.S., Stinson J. et al. Diverse somatic
mutation patterns and pathway alterations in human
cancers // Nature. – 2010. – 466, ¹ 7308. – P. 869–
873.
405. Sharma S.V., Settleman J. Oncogene addiction:
setting the stage for molecularly targeted cancer
therapy // Genes Dev. – 2007. – 21, ¹ 24. – P. 3214–
3231.
406. Heng H.H. Cancer genome sequencing: the challen-
ges ahead // Bioessays. – 2007. – 29, ¹ 8. – P. 783–
794.
407. Duesberg P., Li R., Fabarius A., Hehlmann R. The
chromosomal basis of cancer // Cell Oncol. –
2005. – 27, ¹ 5–6. – P. 293–318.
408. Lee A.J., Endesfelder D., Rowan A.J. et al.
Chromosomal instability confers intrinsic
multidrug resistance // Cancer Res. – 2011. – 71,
¹ 5. – P. 1858–1870.
409. Marusyk A., Kornelia P. Tumor heterogeneity: causes
and consequences // Biochimica et Biophys. acta. –
2010. – 1805, ¹ 1. – P. 105–117.
410. Navin N., Kendall J., Troge J. et al. Tumour
evolution inferred by single – cell sequencing //
Nature. – 2011. – 472, ¹ 7341. – P. 90–94.
411. Navin N., Hicks J. Future medical applications
of single – cell sequencing in cancer // Genome
Med. – 2011. – 3, ¹ 51.
412. Heng H.H., Liu G., Stevens J.B. et al. Decoding
the genome beyond sequencing: The new phase
of genomic research // Genomics. – 2011.
doi:10.1016/j.ygeno.2011.05.008
413. Heng H.H., Stevens J.B., Bremer S.W. et al. The
evolutionary mechanism of cancer // J. Cell
Biochem. – 2010. – 109, ¹ 6. – P. 1072–1084.
Received 06.07.11
<<
/ASCII85EncodePages false
/AllowTransparency false
/AutoPositionEPSFiles true
/AutoRotatePages /None
/Binding /Left
/CalGrayProfile (Dot Gain 20%)
/CalRGBProfile (sRGB IEC61966-2.1)
/CalCMYKProfile (U.S. Web Coated \050SWOP\051 v2)
/sRGBProfile (sRGB IEC61966-2.1)
/CannotEmbedFontPolicy /Error
/CompatibilityLevel 1.4
/CompressObjects /Tags
/CompressPages true
/ConvertImagesToIndexed true
/PassThroughJPEGImages true
/CreateJobTicket false
/DefaultRenderingIntent /Default
/DetectBlends true
/DetectCurves 0.0000
/ColorConversionStrategy /CMYK
/DoThumbnails false
/EmbedAllFonts true
/EmbedOpenType false
/ParseICCProfilesInComments true
/EmbedJobOptions true
/DSCReportingLevel 0
/EmitDSCWarnings false
/EndPage -1
/ImageMemory 1048576
/LockDistillerParams false
/MaxSubsetPct 100
/Optimize true
/OPM 1
/ParseDSCComments true
/ParseDSCCommentsForDocInfo true
/PreserveCopyPage true
/PreserveDICMYKValues true
/PreserveEPSInfo true
/PreserveFlatness true
/PreserveHalftoneInfo false
/PreserveOPIComments false
/PreserveOverprintSettings true
/StartPage 1
/SubsetFonts true
/TransferFunctionInfo /Apply
/UCRandBGInfo /Preserve
/UsePrologue false
/ColorSettingsFile ()
/AlwaysEmbed [ true
]
/NeverEmbed [ true
]
/AntiAliasColorImages false
/CropColorImages true
/ColorImageMinResolution 300
/ColorImageMinResolutionPolicy /OK
/DownsampleColorImages true
/ColorImageDownsampleType /Bicubic
/ColorImageResolution 1200
/ColorImageDepth -1
/ColorImageMinDownsampleDepth 1
/ColorImageDownsampleThreshold 1.50000
/EncodeColorImages false
/ColorImageFilter /DCTEncode
/AutoFilterColorImages true
/ColorImageAutoFilterStrategy /JPEG
/ColorACSImageDict <<
/QFactor 0.15
/HSamples [1 1 1 1] /VSamples [1 1 1 1]
>>
/ColorImageDict <<
/QFactor 0.15
/HSamples [1 1 1 1] /VSamples [1 1 1 1]
>>
/JPEG2000ColorACSImageDict <<
/TileWidth 256
/TileHeight 256
/Quality 30
>>
/JPEG2000ColorImageDict <<
/TileWidth 256
/TileHeight 256
/Quality 30
>>
/AntiAliasGrayImages false
/CropGrayImages true
/GrayImageMinResolution 300
/GrayImageMinResolutionPolicy /OK
/DownsampleGrayImages true
/GrayImageDownsampleType /Bicubic
/GrayImageResolution 1200
/GrayImageDepth -1
/GrayImageMinDownsampleDepth 2
/GrayImageDownsampleThreshold 1.50000
/EncodeGrayImages false
/GrayImageFilter /DCTEncode
/AutoFilterGrayImages true
/GrayImageAutoFilterStrategy /JPEG
/GrayACSImageDict <<
/QFactor 0.15
/HSamples [1 1 1 1] /VSamples [1 1 1 1]
>>
/GrayImageDict <<
/QFactor 0.15
/HSamples [1 1 1 1] /VSamples [1 1 1 1]
>>
/JPEG2000GrayACSImageDict <<
/TileWidth 256
/TileHeight 256
/Quality 30
>>
/JPEG2000GrayImageDict <<
/TileWidth 256
/TileHeight 256
/Quality 30
>>
/AntiAliasMonoImages false
/CropMonoImages true
/MonoImageMinResolution 1200
/MonoImageMinResolutionPolicy /OK
/DownsampleMonoImages true
/MonoImageDownsampleType /Bicubic
/MonoImageResolution 1200
/MonoImageDepth -1
/MonoImageDownsampleThreshold 1.50000
/EncodeMonoImages false
/MonoImageFilter /CCITTFaxEncode
/MonoImageDict <<
/K -1
>>
/AllowPSXObjects false
/CheckCompliance [
/None
]
/PDFX1aCheck false
/PDFX3Check false
/PDFXCompliantPDFOnly false
/PDFXNoTrimBoxError true
/PDFXTrimBoxToMediaBoxOffset [
0.00000
0.00000
0.00000
0.00000
]
/PDFXSetBleedBoxToMediaBox true
/PDFXBleedBoxToTrimBoxOffset [
0.00000
0.00000
0.00000
0.00000
]
/PDFXOutputIntentProfile (None)
/PDFXOutputConditionIdentifier ()
/PDFXOutputCondition ()
/PDFXRegistryName ()
/PDFXTrapped /False
/CreateJDFFile false
/Description <<
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
/BGR <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>
/CHS <FEFF4f7f75288fd94e9b8bbe5b9a521b5efa7684002000410064006f006200650020005000440046002065876863900275284e8e9ad88d2891cf76845370524d53705237300260a853ef4ee54f7f75280020004100630072006f0062006100740020548c002000410064006f00620065002000520065006100640065007200200035002e003000204ee553ca66f49ad87248672c676562535f00521b5efa768400200050004400460020658768633002>
/CHT <FEFF4f7f752890194e9b8a2d7f6e5efa7acb7684002000410064006f006200650020005000440046002065874ef69069752865bc9ad854c18cea76845370524d5370523786557406300260a853ef4ee54f7f75280020004100630072006f0062006100740020548c002000410064006f00620065002000520065006100640065007200200035002e003000204ee553ca66f49ad87248672c4f86958b555f5df25efa7acb76840020005000440046002065874ef63002>
/CZE <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>
/DAN <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>
/DEU <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>
/ESP <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>
/ETI <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>
/FRA <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>
/GRE <FEFF03a703c103b703c303b903bc03bf03c003bf03b903ae03c303c403b5002003b103c503c403ad03c2002003c403b903c2002003c103c503b803bc03af03c303b503b903c2002003b303b903b1002003bd03b1002003b403b703bc03b903bf03c503c103b303ae03c303b503c403b5002003ad03b303b303c103b103c603b1002000410064006f006200650020005000440046002003c003bf03c5002003b503af03bd03b103b9002003ba03b103c42019002003b503be03bf03c703ae03bd002003ba03b103c403ac03bb03bb03b703bb03b1002003b303b903b1002003c003c103bf002d03b503ba03c403c503c003c903c403b903ba03ad03c2002003b503c103b303b103c303af03b503c2002003c503c803b703bb03ae03c2002003c003bf03b903cc03c403b703c403b103c2002e0020002003a403b10020005000440046002003ad03b303b303c103b103c603b1002003c003bf03c5002003ad03c703b503c403b5002003b403b703bc03b903bf03c503c103b303ae03c303b503b9002003bc03c003bf03c103bf03cd03bd002003bd03b1002003b103bd03bf03b903c703c403bf03cd03bd002003bc03b5002003c403bf0020004100630072006f006200610074002c002003c403bf002000410064006f00620065002000520065006100640065007200200035002e0030002003ba03b103b9002003bc03b503c403b103b303b503bd03ad03c303c403b503c103b503c2002003b503ba03b403cc03c303b503b903c2002e>
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
/HRV (Za stvaranje Adobe PDF dokumenata najpogodnijih za visokokvalitetni ispis prije tiskanja koristite ove postavke. Stvoreni PDF dokumenti mogu se otvoriti Acrobat i Adobe Reader 5.0 i kasnijim verzijama.)
/HUN <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>
/ITA <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>
/JPN <FEFF9ad854c18cea306a30d730ea30d730ec30b951fa529b7528002000410064006f0062006500200050004400460020658766f8306e4f5c6210306b4f7f75283057307e305930023053306e8a2d5b9a30674f5c62103055308c305f0020005000440046002030d530a130a430eb306f3001004100630072006f0062006100740020304a30883073002000410064006f00620065002000520065006100640065007200200035002e003000204ee5964d3067958b304f30533068304c3067304d307e305930023053306e8a2d5b9a306b306f30d530a930f330c8306e57cb30818fbc307f304c5fc59808306730593002>
/KOR <FEFFc7740020c124c815c7440020c0acc6a9d558c5ec0020ace0d488c9c80020c2dcd5d80020c778c1c4c5d00020ac00c7a50020c801d569d55c002000410064006f0062006500200050004400460020bb38c11cb97c0020c791c131d569b2c8b2e4002e0020c774b807ac8c0020c791c131b41c00200050004400460020bb38c11cb2940020004100630072006f0062006100740020bc0f002000410064006f00620065002000520065006100640065007200200035002e00300020c774c0c1c5d0c11c0020c5f40020c2180020c788c2b5b2c8b2e4002e>
/LTH <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>
/LVI <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>
/NLD (Gebruik deze instellingen om Adobe PDF-documenten te maken die zijn geoptimaliseerd voor prepress-afdrukken van hoge kwaliteit. De gemaakte PDF-documenten kunnen worden geopend met Acrobat en Adobe Reader 5.0 en hoger.)
/NOR <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>
/POL <FEFF0055007300740061007700690065006e0069006100200064006f002000740077006f0072007a0065006e0069006100200064006f006b0075006d0065006e007400f300770020005000440046002000700072007a0065007a006e00610063007a006f006e00790063006800200064006f002000770079006400720075006b00f30077002000770020007700790073006f006b00690065006a0020006a0061006b006f015b00630069002e002000200044006f006b0075006d0065006e0074007900200050004400460020006d006f017c006e00610020006f007400770069006500720061010700200077002000700072006f006700720061006d006900650020004100630072006f00620061007400200069002000410064006f00620065002000520065006100640065007200200035002e0030002000690020006e006f00770073007a0079006d002e>
/PTB <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>
/RUM <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>
/RUS <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>
/SKY <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>
/SLV <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>
/SUO <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>
/SVE <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>
/TUR <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>
/UKR <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>
/ENU (Use these settings to create Adobe PDF documents best suited for high-quality prepress printing. Created PDF documents can be opened with Acrobat and Adobe Reader 5.0 and later.)
>>
/Namespace [
(Adobe)
(Common)
(1.0)
]
/OtherNamespaces [
<<
/AsReaderSpreads false
/CropImagesToFrames true
/ErrorControl /WarnAndContinue
/FlattenerIgnoreSpreadOverrides false
/IncludeGuidesGrids false
/IncludeNonPrinting false
/IncludeSlug false
/Namespace [
(Adobe)
(InDesign)
(4.0)
]
/OmitPlacedBitmaps false
/OmitPlacedEPS false
/OmitPlacedPDF false
/SimulateOverprint /Legacy
>>
<<
/AddBleedMarks false
/AddColorBars false
/AddCropMarks false
/AddPageInfo false
/AddRegMarks false
/ConvertColors /ConvertToCMYK
/DestinationProfileName ()
/DestinationProfileSelector /DocumentCMYK
/Downsample16BitImages true
/FlattenerPreset <<
/PresetSelector /MediumResolution
>>
/FormElements false
/GenerateStructure false
/IncludeBookmarks false
/IncludeHyperlinks false
/IncludeInteractive false
/IncludeLayers false
/IncludeProfiles false
/MultimediaHandling /UseObjectSettings
/Namespace [
(Adobe)
(CreativeSuite)
(2.0)
]
/PDFXOutputIntentProfileSelector /DocumentCMYK
/PreserveEditing true
/UntaggedCMYKHandling /LeaveUntagged
/UntaggedRGBHandling /UseDocumentProfile
/UseDocumentBleed false
>>
]
>> setdistillerparams
<<
/HWResolution [2400 2400]
/PageSize [612.000 792.000]
>> setpagedevice
|