Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

BBA - Molecular Cell Research 1864 (2017) 1359–1369

Contents lists available at ScienceDirect

BBA - Molecular Cell Research


journal homepage: www.elsevier.com/locate/bbamcr

Review

Transdifferentiation and reprogramming: Overview of the processes, their MARK


similarities and differences
Artur Cieślar-Pobudaa,b,1, Viktoria Knoflachc,1, Mikael V. Ringhd, Joachim Starke, Wirginia Likusf,
Krzysztof Siemianowiczg, Saeid Ghavamih,i, Andrzej Hudeckij, Jason L. Greenk, Marek J. Łosl,⁎
a
Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
b
Stem Cell Group, Nordic EMBL Partnership, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
c
Unit of Molecular Neurobiology, Department of Medical Biochemistry & Biophysics, Karolinska Institute, Stockholm, Sweden
d
Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
e
Department of Surgery, Länssjukhuset Ryhov, Jönköping, Sweden
f
Department of Anatomy, School of Health Science in Katowice Medical University of Silesia, Katowice, Poland
g
Department of Biochemistry, School of Medicine in Katowice, Medical University of Silesia, 18 Medyków Street, 40-752 Katowice, Poland
h
Department of Human Anatomy and Cell Science, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
i
Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
j
Institute of Nonferrous Metals, Gliwice, Poland
k
Duke University, School of Medicine, Durham, NC, USA
l
Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str., 30-387 Krakow, Poland

A R T I C L E I N F O A B S T R A C T

Keywords: Reprogramming, or generation of induced pluripotent stem (iPS) cells (functionally similar to embryonic stem
Reprogramming cells or ES cells) by the use of transcription factors (typically: Oct3/4, Sox2, c-Myc, Klf4) called “Yamanaka
Transdifferentiation factors” (OSKM), has revolutionized regenerative medicine. However, factors used to induce stemness are also
iPS overexpressed in cancer. Both, ES cells and iPS cells cause teratoma formation when injected to tissues. This
Teratomagenesis
raises a safety concern for therapies based on iPS derivates. Transdifferentiation (lineage reprogramming, or
Yamanaka factors
-conversion), is a process in which one mature, specialized cell type changes into another without entering a
pluripotent state. This process involves an ectopic expression of transcription factors and/or other stimuli.
Unlike in the case of reprogramming, tissues obtained by this method do not carry the risk of subsequent
teratomagenesis.

List of abbreviations while regions of chromosomal gain included oncogenes.


ES cells embryonic stem cells; pluripotent stem cells obtained
from the inner mass of a blastocyst, they are capable of
ANT altered nuclear transfer; an experimental reprogramming forming all tissues within an organism, but not extra-fetal
technique developed to satisfy research-ethical concerns. tissues like i.e. the placenta.
It employs nuclear transfer-based reprogramming, but FACS fluorescence-activated cell sorting; versatile, flow
only after the somatic cell nucleus and/or the oocyte are cytometry-based techniques commonly used for
altered in order to inhibit totipotency and eliminate the quantification of expression of cellular proteins within
full embryonal developmental potential, while still intact cells, and also for the measurement of various
allowing the formed pseudo-embryo to generate biologic processes. It is for example used for the
pluripotent stem cells. detection and quantification of stemness markers or
CNV copy number variations; interpersonal variations in the hematologic markers in the clinic.
numbers of copies of certain genes. Recently, Laurent and HIFs Hypoxia inducible factors; transcription factors that play
colleagues have described that deleted regions within the a role, among others, in the regulation of several genes
iPS-genome often contained tumor-suppressor genes, related to hypoxia, vascularization, and pluripotency in


Corresponding author at: Małopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A str., 30-387 Kraków, Poland.
E-mail address: bioappl@gmail.com (M.J. Łos).
1
These authors share first authorship.

http://dx.doi.org/10.1016/j.bbamcr.2017.04.017
Received 8 March 2017; Received in revised form 24 April 2017; Accepted 26 April 2017
Available online 28 April 2017
0167-4889/ © 2017 Elsevier B.V. All rights reserved.
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Induced-pluripotent stem cell


human embryonic stem cells cultured under hypoxic (iPS)
conditions by upregulating the expression of Oct4, Sox2
and NANOG.
iPS induced pluripotent stem (cells); functionally-
comparable/equal to ES cells pluripotent cells, obtained
by expression of ‘Yamanaka factors’ (OSKM) in somatic
cells; please consult Boxes 1 and 2 for details. Unlike ES-
cells that are usually obtained by destruction of embryos,
generation of iPS is not ethically controversial.
lincRNAs long intergenic non-coding RNAs; long non-coding RNAs
that are transcribed from non-coding DNA sequences TRANSDIFFERENTIATION
between protein-coding genes (hence, intergenic); may
harbor transposable elements and may attach to
messenger RNA to block protein production. Differentiated cell Another (partly) differentiated cell

miRNA microRNA; an ‘umbrella-term’ for several classes of Fig. 1. Basic difference between reprogramming and transdifferentiation.
small, regulatory RNAs that may affect gene Reprogramming (shown in a green arrow) requires a reversal change of a differentiated
transcription, messenger RNA (mRNA) maturation, the cell into a pluripotent stem cell (i.e. iPS), which next may undergo a differentiation
stability of mRNAs, and their translation. process (shown in a blue arrow) into another differentiated cell. Transdifferentiation
mtDNA mitochondrial DNA; mitochondrial genome, it codes for (shown in a red arrow) does not require a full reversal into iPS cells in order to transform
into another cell type. In some cases transdifferentiation is a one-step process, while in
some of the mitochondrial proteins.
others the cells pass through an intermediate state(s). Note: the drawing is schematic and
MACS magnetic-activated cell sorting; a cell-separation the differences in size and shape of pictured cells do not exactly reflect the actual
technique where cells are marked with antibodies differences.
specific to selected proteins; the antibodies carry nano-
magnet particles attached and this allows for separation molecular compounds in addition to transcription factors as well as
of the cell population marked with such antibodies from different delivery systems used. All of those methods have different
the rest. reprogramming efficiencies ranging from 0.00001 to > 1% [4–6].
OSKM Oct3/4, Sox2, Klf4, c-Myc; ‘classical’ set of transcription Another route in generating cells of a different cell type is by
factors required for reprogramming, discovered by S. utilizing transdifferentiation (Fig. 1). This concept of lineage conver-
Yamanaka, hence they are also called “Yamanaka sion is not new; already in 1987 Lassar and colleagues showed that the
factors” (please consult Boxes 1 & 2 for more details). introduction of just one cDNA converts fibroblasts into myoblasts [7].
PEG polyethyleneglicol; a chemical reagent frequently used, Instead of using transcription factors important in maintaining plur-
among others, to facilitate fusions between cells. ipotency, lineage-specific transcription factors are introduced into the
RISCs RNA-induced silencing complexes; large multi-protein cell. Recently, a predictive system was published, called Mogrify which
complex, containing mainly ribonucleoproteins, with the aims to predict the composition of transcription factors needed for
protein ARGONAUT responsible for mRNA-cleavage; optimal transdifferentiation into a given cell type [8]. It combines gene
they are a key part of the cellular mRNA-silencing expression data with regulatory network information to predict the
machinery regulated by miRNAs; RISC is also the key transdifferentiation factors necessary to induce cell conversion.
component of an innate antiviral response that degrades Whereas iPS cell reprogramming is a rather time-consuming pro-
foreign (m)RNAs. cess, transdifferentiation is often fast and highly efficient. In 2008 Zhou
ROCK Rho-associated kinases; a family of a versatile signal-
transduction proteins involved, among others, in the
regulation of cell migration (cytoskeleton dynamics) cell OSKM
cycle, and apoptosis. ROCK inhibitors enhance the A SOMATIC CELL
efficiency of iPS generation (reprogramming), at least vehicle
partially by inhibiting single-cell dissociation-induced
apoptosis.
SCNT somatic cell nuclear transfer, an early reprogramming
(cloning) technique, used i.e. in 90s for experimental
(proof of principle) cloning of sheep (please consult Box
C
3 for details).
OSKM
OSKM

1. Reprogramming and transdifferentiation — key facts and major


B
differences
OSKM
During ontogenesis, the cell fate is determined by a complex set of iPS
transcription factors and epigenetic networks that governs the differ-
entiation program, which until recently was considered unidirectional iPS
and irreversible. In the past decade, Yamanaka, and others, showed that iPS

a cocktail of just four transcription factors (Oct4, Sox2, Klf4 and c-myc,
OSKM) is sufficient to reprogram murine- [1] and human [2,3] Fig. 2. Cell reprogramming. A — Four genes (OSKM) are delivered into a nucleus of a
fibroblasts into induced pluripotent stem (iPS) cells. The group of differentiated cell. Various methods discussed in the text use various types of carriers
(“vehicle”) to introduce them into the cells' nucleus. B — After the introduction of OSKM
Yamanaka was using the same transcription factors for the reprogram-
the cell proliferates. C — Among new cells there are iPS cells (marked in blue), but also
ming of mouse fibroblasts, whereas Yu et al. exchanged c-myc and Klf4 cells that failed to (fully) reprogram, which will eventually die. Note: the drawing is
for Nanog and Lin28. Since then several protocols were established, schematic and the differences in size and shape of pictured cells do not exactly reflect the
some only using two or three transcription factors, others utilizing small actual differences.

1360
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Table 1 injection into blastocysts [1]. Subsequently, different genes have been
Examples of in vitro cellular transdifferentiation. substituted for a subset of the OSKM genes, and other gene transfer
methods have been applied, all with their respective advantages and
Source cells Transdifferentiation inducing Target cells Ref.
factors disadvantages.
A major issue with the first iPS cells was that mice derived from
Fibroblasts MyoD Myocytes [7] them were prone to tumor development. All of the original OSKM
B-cells C/EBPα, C/EBPβ Macrophages [12]
factors to some extent possess oncogenic potential, and the tumorigen-
T-cells Bcl11b deletion NK cells [111]
Pancreatic Duct Pdx1 Beta cells [112] esis in iPS cell-derived mice has been especially attributed to c-Myc.
cells Combinations of reprogramming factors excluding c-Myc evaded tu-
Pancreatic Ngn3, Mafa, Pdx1 [9] morigenesis during the study period while maintaining efficiencies
exocrine cells similar to OSKM reprogramming [14,15]. However, a non-zero long-
Hepatocytes Exendin-4, Pdx1 [113]
term risk cannot be excluded for any combination of reprogramming
Fibroblasts Hnf4α, Foxa1 or Foxa2 or Hepatocytes [114]
Foxa3 factors.
Fibroblasts Ascl1 (also know as Mash1), Neurons [115] Retroviruses, the type of vectors initially used for reprogramming,
Brn2, Myt1l integrate into the host genome and thereby drive the expression of the
miR-124, Brn2, Myt1l [116]
reprogramming factors. There is a substantial risk of insertion-muta-
Astrocytes Pax6 neurogenin 2, Ascl1 [117]
Fibroblasts Ascl1, Nurr1, Lmx1a Dopaminergic [118] genesis as a consequence of retroviral integration, severely constraining
Ascl1, Brn2, Myt1l, Lmx1a, neurons [119] use in clinical applications. Retroviral transgenes are effectively
FoxA2 inactivated in iPS cells, while the expression of the endogenous
Fibroblasts Oct4, Sox2, Klf4 and c-Myc Cardiomyocytes [120] pluripotency genes maintains pluripotency [16]. Still, the possibility
Tbx5, Mef2c, Gata-4, Mesp1 [121]
of transgene reactivation and residual expression poses a risk, especially
with regard to the tumorigenesis related to c-Myc expression.
et al. succeeded in differentiating exocrine cells into β-cells by Effective retroviral delivery can only be achieved in dividing cells
introducing three transcription factors important in the embryonic [17]. Retrovirally reprogrammed murine autologous iPS cells were
development of the pancreas. First transdifferentiated cells appeared more immunogenic compared to iPS cells reprogrammed by a non-
already on day 3 and as much as 20% of the cells were successfully integrating episomal method and autologous embryonic stem cells [18].
transdifferentiated [9]. In 2013 the first successful transdifferentiation It is unclear whether this immunogenicity extends to all iPS cells, only
of human fibroblasts towards a cardiac fate was published [10]. We to those iPS that have been obtained by employing integrating viruses
have recently transdifferentiated human fibroblasts into corneal limbal as vectors, or retrovirally reprogrammed iPS cells only. The immuno-
cells [11]. In some cases transdifferentiation is a one-step process, while genicity study underscores the fact that iPS cells may cause immuno-
in others the cells pass through an intermediate state, as shown in a rejection in syngenic transplantation and that there may exist differ-
study to reprogram B cells into macrophages where the cells express ences in immunogenicity between iPS cells produced by different
low levels of both B cell-specific genes and macrophage-specific genes reprogramming methods. Lentiviral delivery of reprogramming factors
before they completely transdifferentiate into macrophages [12]. The is another successful approach, applied e.g. in one of the two first
most important examples of in vitro transdifferentiation and factors instances of human iPS generation [3] (Box 2).
needed for cellular conversions to specific cell types are presented in
Table 1. 2.1.2. Non-integrating viral approaches
The dedifferentiated state sometimes present during transdifferen- A major advantage of non-integrating methods is the evasion of
tiation is unnatural and therefore rather unstable, whereas iPS cells potentially oncogenic insertion-mutagenesis, transgene reactivation
could be cultured indefinitely without losing their pluripotent potential. and residual expression, but the trade-off of adenoviral reprogramming
One hallmark, and also a negative aspect of pluripotency is the is considerably lower efficiency, the requirement for high viral titers
formation of teratomas when transplanted into immune deficient mice. and sometimes the need for repeated transductions [19,20]. One
Therefore it must be ensured that the differentiation of iPS cells is possible contributing variable is the dilution of reprogramming factors
uniform and the maturating cells are not contaminated with a ‘leftover’ as cells multiply.
iPS progeny. Another issue is the reactivation of the transgenes which One such non-integrating approach is adenoviral reprogramming
could lead to tumorigenic tendencies as shown by Okita el al [13]. [20,21]. Although adenoviral vectors are generally considered non-
Although some transdifferentiation protocols include a dedifferentia- integrating and adenovirally reprogrammed iPS cells have been pro-
tion step, those cells do not go back to pluripotency, therefore lowering duced with the apparent absence of viral integration, including
or eliminating the risk of tumor formation. Despite that transdiffer- negative Southern blot analysis [19,20], adenoviral vectors have been
entiation was historically described much earlier, nowadays much more reported to integrate to a small extent [22]. Sendai-virus based delivery
is known about reprogramming. Below, we discuss various aspects of of reprogramming factors (currently the most popular reprogramming
both methods. Although at first glimpse they may seem very similar, method) has been summarized in Box 2.
both have some important differences, which impact their plausible use
in regenerative therapy. The molecular principles of reprogramming 2.1.3. Non-viral delivery of reprogramming factors
have been briefly outlined in Box 1. Non-viral vectors are safer, because they are free from dangers like:
transgene activation, residual expression, insertion-mutagenesis of
integrating viruses, as well as concerns regarding the presence of
2. Reprogramming techniques viruses per se. In addition, non-viral methods allow for reprogramming
without the need for virus production and reprogramming of cell lines
2.1. Introduction of reprogramming transcription factors not limited by viral tropism.
Conventional plasmids, self-replicating episomal plasmids and
2.1.1. Integrating viral approaches minicircle plasmids are some of the non-viral methods of introducing
Upon introduction of OSKM genes into murine fibroblast via retro- reprogramming factors to generate iPS cells. Conventional plasmids do
viral infection, the cells undergo reprogramming into a pluripotent not replicate in mammalian cells, thus requiring multiple rounds of
state. They could then be differentiated into all three germ layers, and transfection for successful reprogramming, and the reprogramming
as a proof of concept, led to the production of chimeric mice after efficiency is considerably lower than in viral methods [19,23,24].

1361
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Box 1
Reprogramming — basic mechanism, and ‘Yamanaka factors’.

All cells of the body have the same genes. Pluripotent stem cells can differentiate into all cell types within the organism. This process is
accompanied by a limitation of genes that could be expressed in a differentiated cell. DNA is packed in the cell's nucleus and various
proteins associated with DNA regulate the degree of its “compression” and which genes could be expressed. In 2006, Yamanaka and co-
workers managed to generate iPS cells from fibroblasts by using a set of defined reprogramming factors [1] (Box 2, Fig. 2). The chromatin
structure in pluripotent ES cells has higher plasticity and is more open compared to differentiated cells [122] and heterochromatin
rearrangement is the first observable change in somatic cells undergoing reprogramming into iPS cells [123]. Telomeres in iPS cells have
the same epigenetic marks as ES cells, and are further elongated after reprogramming compared to their progenitors [124]. iPS generated
from the fibroblast of old donors, similarly elongate telomeres as iPS cells derived from young donor fibroblasts. Several long intergenic
non-coding RNAs (lincRNAs) have also been identified, which show increased expression in iPS cells compared to ES cells and may serve as
targets of pluripotency transcription factors [125].
Yamanaka and co-workers evaluated 24 candidate factors by co-expression in murine fibroblasts and after successive exclusion of
individual transcription factors four genes were identified as the minimal requirement for iPS cell generation. These defined factors
included Oct-3/4 (Pou5f1), Sox2, KLF4, and c-Myc (OSKM). They were then introduced into mouse fibroblasts by using retroviral vectors,
and in turn generated iPS cells [1]. These cells were morphologically similar to ES cells, including the ability to generate cells from all 3
germinal layers, and also showed comparable proliferation characteristics and expressed the same sets of surface markers. Several methods
exist to introduce Yamanaka factors to somatic cells, and more common examples are provided in Box 2.

Epstein-Barr virus-based episomal vectors, capable of self-replica- efficiency along with longer and stronger expression in both dividing
tion, enable single transfection reprogramming but suffer from low and non-dividing cells, attributed to decreased silencing, when com-
efficiency, and in several cases require more reprogramming factors in pared to conventional plasmids [6,30,31]. Yet, compared to viral
addition to the OSKM factors [25–27]. It has been suggested that the methods, minicircle delivery of reprogramming factors have yielded
limiting variables are properties intrinsic to the vector itself, including considerably lower reprogramming efficiencies even when multiple
fast methylation silencing the vector sequence, infrequently established consecutive transfections were employed [32,33]. Another non-viral
stability despite replicative potential and common loss of “established” approach to introduce reprogramming factors is the use of transposon
plasmids with cell division upon removal of selective pressure systems (Box 2).
[25,28,29]. Interestingly, one study reported less immunogenicity of
episomally reprogrammed iPS cells compared to retrovirally repro-
grammed iPS cells [18]. 2.1.4. Non-transgene factor delivery
Minicircle vectors are circular, supercoiled DNA elements, produced In order to circumvent the risks of insertion-mutagenesis and
by e.g. in vivo PhiC3-integrase based intramolecular site-specific re- oncogenic transformation, several non-transgene reprogramming tech-
combination of conventional plasmids containing sequences of interest niques such as i.e. delivering reprogramming factors in the form of
[6,30]. Non-viral and devoid of a bacterial backbone sequence, mini- mRNA or protein have been developed. Sendai viral reprogramming is
circle vectors offer an interesting safety profile and higher transfection also non-transgene, but the perks of delivering reprogramming factors
in the form of mRNA or protein is the avoidance of possible viral

Box 2
Examples of delivery methods of reprogramming factors.

Lentiviruses are integrating viruses of the Retroviridae family, and lentiviral vectors share many advantages and disadvantages with the
other retroviruses used in iPS cell production. Amongs others, the disadvantages of a lentiviral delivery method are insertional mutagenesis,
transgene activation, residual expression and immunogenicity. Lentiviruses differ from retroviruses by offering alternative tropism and are,
unlike retroviruses, capable of transducing non-dividing cells. Slower silencing of reprogramming factors makes lentiviral reprogramming
more efficient than retroviral reprogramming [16,25,36,126,127]. Differences in storage stability, the smaller maximum insert size of
lentiviruses and the safety concerns regarding lentiviruses being derived from immunodeficiency viruses are some disadvantages of
lentiviruses compared to retroviruses [128,129].
Sendai virus-mediated delivery becomes increasingly popular, as it evades the risks of insertion-mutagenesis, transgene activation and
residual expression, while still maintaining even higher reprogramming efficiencies than lentiviral methods [23,130]. As Sendai virus
constitutively replicates, concerns regarding remaining viral particles have been raised [131]. Sendai viruses are not pathogenic in humans,
but may infect epithelial cells, thus caution is advised, especially if oncogenes such as c-Myc are introduced [25,132]. Use of replication-
deficient-, and temperature sensitive Sendai viruses have shown some promise in mitigating this aspect [130,133].
Transposons are genetic mobile elements, which can be moved within a genome by a transposase, an enzyme catalyzing excision and
insertion of the transposon. The piggyBac transposon system has been used for reprogramming of both murine and human cells, and in
murine cell lines it was possible to tracelessly excise the exogenous transgene reprogramming factors when they were no longer needed for
reprogramming [134]. Another excision approach using the Cre-Lox system, where the reprogramming factors are flanked by lox sites and
excised by subsequent Cre recombinase transfection when no longer needed, may reprogram without permanent genetic alterations [90].
The excision of transgenes can prevent the risks of transgene activation and residual expression associated with many reprogramming
methods, though the risk of inefficient and imperfect excision exists. There is also the practical matter of an extra excision step. PiggyBac
reprogramming is further complicated by the fact that tranposases not only excise, but also insert transposons and screening is needed to
assess whether all transgenes have been effectively removed. Another concern is that the human genome contains piggyBac system-like
components, which potentially could interact with the piggyBac system [25,135].

1362
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

infections in patients and the lack of a need to handle viral particles. reported in in vivo induced pluripotent stem cells [41], SCNT is
Modified synthetic mRNA encoding reprogramming factors repro- currently the only known technique able to confer true totipotency
grams faster and with high efficiency as compared to other methods on somatic nuclei.
[34,35]. Unlike the DNA-based delivery of reprogramming factors, RNA An approach intended to attenuate the ethical issues regarding
is not epigenetically silenced [36]. However, foreign mRNA elicits therapeutic SCNT is altered nuclear transfer (ANT). This technique
strong innate antiviral response and is targeted for degradation by RNA- employs nuclear transfer, but only after the somatic cell nucleus and/or
induced silencing complexes (RISCs), and the in vivo half-life of mRNA the oocyte are altered in order to inhibit totipotency and eliminate the
is very short — it is degraded over the course of a few days. Therefore, full embryonal developmental potential, while still allowing the formed
repeated transfections, and large quantities of modified, high quality, pseudo-embryo to generate pluripotent stem cells [42].
long sequence mRNA, poses technical challenges (also high costs). There are other methods enabling the production of human
Another non-transgene-based method is the delivery of the repro- pluripotent stem cells without the use of neither oocytes nor embryonic
gramming transcription factors into the cell in the form of proteins. stem cells as starting materials, and which do not include formation or
Successful protein reprogramming of both murine and human cells has destruction of pre-implantation state embryos. However, nuclear
been reported, although the process was relatively slow and very transfer is still the only known method of reprogramming cells into
inefficient, especially in human cells [37,38]. The reprogramming of totipotency — a feature for which specific therapeutic applications
murine cells was made with several rounds of transfection of recombi- could theoretically be found in the future. It is also possible that the
nant proteins produced in E. coli, and the human cells were repro- resultant reprogrammed cells from nuclear transfer could differ from
grammed using cell extracts of HEK293 cells expressing recombinant cells reprogrammed using other methods. Furthermore, as non-human
proteins. Bacterially produced proteins may be inferior in reprogram- cloning is already used to produce transgenic animals for research
ming applications because of the lack of eukaryotic posttranslational purposes, along with foods and some medical products approved for
modifications and possible misfolding during purification/renaturation, human use, nuclear transfer methods have found important applica-
and cell extracts containing recombinant proteins produced in mam- tions.
malian cells might have to be enriched for higher efficiency [6,25].
Recombinant protein delivery offers reprogramming without involve-
ment of any exogenous nucleic acids and no alterations of the genome 2.3. Cell fusion
(aside from secondary epigenetic changes due to the reprogramming
process), and so it has an appealing safety profile. However, large During cell-fusion-reprogramming (Box 3), somatic cells are fused
efforts for optimizing and improving efficiency are much needed. with embryonic stem cells, thereby exposing them to the reprogram-
ming milieu of the stem cell. The method has been employed using both
murine [43,44] and human [45,46] cells. Alternative methods of
2.2. Somatic cell nuclear transfer (SCNT) inducing cell fusion include electrofusion and Sendai virus-induced
fusion. Both these methods have shown promise, though a comparative
As SCNT (Box 3) replaces the oocyte nucleus with a somatic nucleus, study found PEG to be the superior fusion inducer [47].
the mitochondrial DNA (mtDNA) of the oocyte remains in the cell. This Since it can be achieved using human embryonic stem cells, without
requires interaction between the oocyte mtDNA and a foreign nucleus, the specific need for oocytes, it makes cell fusion reprogramming a
raising concerns of incompatibility. Successful SCNT proves that this practically and ethically interesting alternative for regenerative medi-
matter can be overcome to the extent of producing viable cells, embryos cine applications. However, following fusion, the resultant hybrid cells
and even full-grown animals. The lack of somatic cell donor mtDNA contain nucleic acid material from both the somatic cell and the
transfer facilitates the therapeutic use of SCNT in diseases caused by embryonic stem cell used. Because nuclear factors are likely involved
mtDNA mutations [39,40]. Although totipotency features have been in the process of reprogramming by cell fusion [48], removal of stem

Box 3
Cellular-factors based reprogramming methods.

Independently of the reprogramming method, involving factors identified by Yamanaka, which upon ectopic expression could drive
reprogramming, others have successfully reprogrammed cells using factors readily available in stem cells or oocytes. Below we show a few
examples:
Somatic cell nuclear transfer (SCNT), often referred to as “cloning”, is achieved by transfer of a nucleus from a somatic cell into an
enucleated unfertilized oocyte, after which the oocyte cytoplasmic environment confers reprogramming on the transferred nucleus and
subsequently activating embryo development [136,137].
Cell fusion: Most modern protocols use a 1:1 ratio of somatic cells:stem cells, and fusion is induced by addition of PEG, yielding
approximately 1% fusion efficiency [45]. The resulting hybrid cells expressed reactivated pluripotent markers Oct4, Nanog and Sox2, and
both in vitro and in vivo differentiation yielded cell types from all three germ layers [45,48]. Results from one study suggests that it is
nuclear factors rather than cytoplasmic factors that are responsible for cell fusion reprogramming as fusion with karyoplasts, but not with
cytoplasts, induced Oct4 expression in the somatic genome [48].
Cell extracts: Exposure to extracts of human or murine pluripotent cells has been shown to generate pluripotent stem cell-like phenotypic
and genotypic characteristics in human somatic cells [138,139]. Upregulated expression of pluripotency-related genes Oct4, Sox2, c-Myc
and Klf, as well as downregulated expression of LMNA, a marker of differentiation [140], were indicative of pluripotency in these studies.
The human cell extract reprogrammed cells were able to differentiate into mesoderm and ectoderm lineages, but they were not
demonstrated to differentiate into all three germ layers in vivo. This could suggest that the cells may have been only partially/incompletely
reprogrammed. Cell extract method offers possible reprogramming without genetic alteration of target cells, and avoids some of the
practical and ethical pitfalls of nuclear transfer and cell fusion. However, long-term expression profiles supporting pluripotency and in vivo
differentiation into all germ layers have not yet been demonstrated. However, with the establishment of reprogramming methods capable
of producing human pluripotent cells using only a few well-defined transcription factors, miRNAs or small molecule compounds, the
importance of such methods are profoundly diminished.

1363
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Table 2
Notable advantages and disadvantages of different reprogramming methods.

Virus Method Advantages Disadvantages

Yes Retrovirus High efficiency Risk of insertional mutagenesis, transgene reactivation and residual
expression
Risk of presence of viral particles
Can only be used in dividing cells
Yes Lentivirus High efficiency Risk of insertional mutagenesis, transgene reactivation and residual
Can be used in both dividing and non-dividing cells expression
Risk of presence of viral particles
Yes Adenovirus No integration/small risk of integration Moderate efficiency
Need for repeated transductions
Risk of presence of viral particles
Yes Sendai virus High efficiency Risk of presence of viral particles
Non-transgene, no integration
No Conventional and episomal plasmids No integration Very low efficiency
No viral particles Need for repeated transfections
No Minicircle Efficiency higher than conventional plasmids Low efficiency (lower than viral methods)
No integration (Need for repeated transfections)
No viral particles
No bacterial backbone
No piggyBac transposon No viral particles Low efficiency
Theoretically possible to excise transgenes Extra excision step
Imperfect excision and transposition
Risk of insertional mutagenesis, transgene reactivation and residual
expression
Possible interactions between piggyBac system and endogenous
transposon systems
No mRNA Very high efficiency Need for repeated transfections
Non-transgene, no integration High cost
No viral particles
No Protein Non-transgene, no integration, no exogenous nucleic Very low efficiency
acids
No viral particles
No Small molecule compounds Non-transgene, no integration, no exogenous nucleic Not yet shown in human cells
acids Moderate efficiency
No viral particles
Moderate efficiency
No Direct transfection of mature miRNA Non-transgene, no integration Moderate efficiency
No viral particles
Moderate efficiency
Yes Lentiviral delivery of miRNA coding Very high efficiency Risk of insertional mutagenesis, transgene reactivation and residual
DNA expression

cell nucleic acids prior to fusion may not be a viable alternative. Other result be confirmed by others and extended to human cells, the small
approaches have aimed at selectively removing stem cell nuclei after molecule-only technique offers an interesting alternative as many of the
allowing time for reprogramming or selectively removing stem cell safety concerns regarding other techniques involving viruses, trans-
chromosomes from hybrids. Such approaches have shown promise in genes and/or non-transgene exogenous nucleic acids are evaded. Still,
murine cells, but their feasibility in human cells is yet to be confirmed since the small molecule compounds affects epigenetic cell-cycle
[49,50]. Furthermore, hybrid cells also contain non-nuclear stem cell- regulation and related functions, caution must be taken not to unleash
derived mtDNA, which could potentially pose a problem of incompat- uncontrolled proliferation or genome instability [55–57].
ibility. Thus, the presence of stem cell nucleic acids in the hybrid cells MicroRNAs (miRNAs) are short, non-coding RNAs that regulate
and the possible immunological rejection in recipients are issues that gene expression by enhancing degradation of mRNA and repressing
must be resolved before cell fusion reprogramming could be used in mRNA translation [58]. While down-regulation is the most widely
clinical applications. Another issue is the low efficiency of cell fusion recognized function of miRNA, up-regulation of target mRNA transla-
reprogramming. Several optional methods using no embryonic cells, tion by miRNA has also been reported [59]. Knockdown of key players
but only addition of a few well-defined transcription factors, miRNA or in the miRNA biogenesis results in loss of miRNA in cells and decreased
small molecule compounds exist. Cell extracts constitute another reprogramming efficiency [60,61]. Several miRNAs are able to enhance
method (Box 3). factor-induced reprogramming, other miRNAs decrease reprogramming
efficiency, and OKSM reprogramming factors and reprogramming
3. Reprogramming without OSKM-factors delivery relevant miRNAs are reciprocally regulated [61–64]. As is presumably
the case for most cellular processes, miRNAs clearly play an important
Numerous studies have identified several molecules that can role in reprogramming and maintaining pluripotency. Recently, se-
enhance reprogramming and even replace individual OSKM factors by lected miRNA clusters were used to carry reprogramming both murine
epigenetic regulation. Various combinations of a subset of reprogram- and human cells with high efficiency without the introduction of any
ming factors together with i.e. Rcor2, 5-mc-hydroxylase Tet1, valproic exogenous transcription factors or transcription factor coding nucleic
acid, kenpaullone or various miRNAs may induce pluripotency acids [52,65]. miRNA-based reprogramming offers an appealing safety
[51–53]. Finding a small molecule substitute for Oct4 long seemed profile, especially direct transfection of mature miRNA which evades
near impossible, but recently one study reported reprogramming of the concerns of insertional mutagenesis, transgene activation, residual
murine somatic cells using only seven small molecule compounds [54]. expression and presence of viral particles associated with most other
Even though the reprogramming efficiency was modest, should this reprogramming methods. Remarkably high reprogramming efficiency

1364
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

was reported when viral vectors were used to introduce miRNA coding tions reduces the chance for insertional mutagenesis, transgene reacti-
DNA [65]. The reprogramming efficiency of direct transfection of vation and residual expression. From a practical point of view, the
mature miRNAs was substantially lower, though still comparable to production and handling of only one polycistronic construct could be
the reprogramming efficiency of viral delivery of reprogramming convenient. Also, the use of polycistronic vectors ensures that all
factors [52]. One suggested rationale for the high efficiency is that successfully transduced cells express all factors. A trade-off is negatively
one single miRNA can regulate hundreds of targets, and when clusters influenced packaging efficiency of viral particles as the genome size
of miRNA are introduced, a huge number of targets can be regulated by increases [25,36,83].
several miRNAs working in great synergy [60,66,67]. On the downside,
the stability of transfected miRNA is not very high and multiple
transfections were needed for complete reprogramming. Also, the 5. Reprogramming, transdifferentiation, and carcinogenesis
detailed mechanisms of miRNA reprogramming are far from entirely
outlined, and, as is the case with all reprogramming methods, more The major challenge of translating reprogramming and transdiffer-
research is needed before miRNA reprogramming can be considered entiation research into clinical applications is the risk of the develop-
safe for clinical applications. The fact that one single miRNA can ment of cancer. OKSM reprogramming factors themselves are proto-
regulate hundreds of targets adds to this concern, as many non- oncogenes, and their expression commonly increases in metastatic
reprogramming cellular processes could be simultaneously affected. cancers. We have already discussed the dangers of insertional mutagen-
Table 2 summarizes the most important advantages and disadvantages esis posed mainly by retro- and lentiviral vectors used in some
of the described methods. reprogramming protocols. iPS cells proliferate in an uncontrolled
manner similar to cancer cells. The transplantation of iPS-derived cells
4. Methods that enhance reprogramming containing any residual iPS may result in the formation of somatic
tumors.
A plethora of methods of enhancement of reprogramming efficiency Transdifferentiation is viewed as a safer alternative to reprogram-
has been proposed. p53 pathways are upregulated in OSKM factors ming. By directly converting differentiated cells into other types of
reprogramming, and knockdown of p53 enhances reprogramming cells, one avoids the state of pluripotency. Hence, obtained by
efficiency [56,68]. But, as p53 is utterly important in maintaining the transdifferentiation autologous cells do not inherently acquire the
genomic integrity, it is unclear if p53 knockdown could safely be ability to self-renew and proliferate uncontrollably as in reprogram-
applied in the production of iPS cells for clinical use, especially when ming. While this reduces the risk of cancer development, carcinogenesis
reprogramming itself could potentially compromise the genomic integ- is still possible due to the genetic and epigenetic changes that could
rity. Targeting some selected mediators in the p53 pathway could occur during transdifferentiation.
provide a safer option [69].
Rho-associated kinases (ROCKs) are Rho-GTPase activated kinases
that serve many diverse roles, including regulation of cytoskeletal 5.1. Risk of carcinogenesis
dynamics, cell cycle control, and modulation of apoptosis [70].
Application of ROCK inhibitors could enhance the efficiency of iPS cell The association between reprogramming and carcinogenesis is
production, at least partially by inhibiting single-cell dissociation- mainly attributable to insertional mutagenesis, and genetic and epige-
induced apoptosis [71,72]. netic alterations that could occur during reprogramming in addition to
A novel way of enhancing reprogramming efficiency is mechan- the inherent tumorigenic capacity of pluripotent cells. The ability of iPS
omodulation of cells' epigenetic profile by introducing microgrooves in cells to generate teratomas in immunocompromised mice is actually
the cell-adhesive substrates [73]. The detailed mechanism is thought to used to verify their pluripotency. In vivo-teratoma formation occurs
rely on decreased histone deacetylase activity and increased H3 quicker and more efficiently for iPS cells in comparison to embryonic
methyltransferase activity, resulting in increased histone H3 acetylation stem (ES) cells, indicating their increased propensity to form tumors
and methylation. Though ROCK is involved in cytoskeletal processes, [84]. This is in part due to the factors used to induce reprogramming.
which are susceptible to mechanical stimuli, no role of ROCK was The primary method used for reprogramming is ectopic expression
proposed in this model. Application of tensile strain reduces expression of four transcription factors: Oct4, Sox2, c-Myc, and Klf4 [1]. Each of
of pluripotency-related genes via ROCK activation, and upon ROCK these factors is associated with multiple forms of cancer. The method of
inhibition the gene expressions are recovered [74]. exogenous gene delivery into cells can also induce tumorigenesis. iPS
Enhancement of iPS cell generation by culturing under hypoxic cells were initially generated by transduction of reprogramming factors
conditions has been described in both murine and human cells [75,76]. using integrating viral vectors. Integration of these genes near or within
Hypoxia inducible factors (HIFs) play a role in regulation of pluripo- gene encoding regions can inadvertently deactivate tumor suppressor
tency in human embryonic stem cells cultured under hypoxic condi- genes or activate oncogenes. Additionally, in vivo-reactivation of
tions by upregulating the expression of Oct4, Sox2 and NANOG which integrated transgenes in iPS-derived cells can result in somatic tumor
are some of the most commonly used reprogramming factors [77]. development.
Several additional compounds have been used to promote and Given the potential tumorigenic properties of the reprogramming
enhance reprogramming, in some cases increasing reprogramming factors (Box 4), ways to reduce their usage in reprogramming have been
efficiency > 100-fold when low-efficient reprogramming methods are sought. iPS cells have been generated without c-Myc and Klf4 through
used. Various histone deacetylase inhibitors and histone methyltrans- replacement with alternative pluripotency factors such as Nanog and
ferase inhibitors, SV40 T-large antigen and telomerase-reverse tran- Lin28 [3] but these proteins are also linked to different forms of cancer.
scriptase are some worth mentioning [78,79]. Knockdown of Mbd3, an Reprogramming has also been completed using only Oct4 and Klf4 by
integral part of the Mbd3/NuRD repressor complex, allowed for very addition of glycogen synthase kinase-3 (GSK3) inhibitor CHIR 99021
fast, extraordinarily efficient and synchronized OSKM factor repro- [85]. However, this was done using murine embryonic fibroblasts only,
gramming fitting a deterministic model [80]. Standard OSKM factor and so far not replicated in differentiated human cells. Even if
reprogramming is widely considered a stochastic process [81,82]. reprogramming of human cells occurs using Oct4 and Klf4 alone, two
When multiple reprogramming transgenes are used, polycistronic genes associated with cancer development are still being overexpressed
vectors allow reprogramming with less integrations than using one in cells. In addition, consistent reprogramming with reduced transgenic
vector for each factor [13,83]. At the very least one integration is still overexpression may only be feasible in cells with high endogenous
necessary for reprogramming, but minimizing the number of integra- levels of omitted reprogramming factors.

1365
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Box 4
Carcinogenesis from reprogramming factors.

Oct4 is a transcription factor that is fundamental to reverting differentiated cells back to a pluripotent state during reprogramming.
However, its promotion of pluripotency is also involved in cancer development. For example, in breast cancer, Oct4 is expressed
significantly higher in cancerous tissue and has been proposed as a biomarker for the cancers initiation, and growth [141]. It is also
correlated with reduced differentiation and increased tissue invasion in gastric cancer, resulting in a more negative prognosis [142]. These
examples demonstrate that Oct4’s function in reprogramming overlaps with its role in cancer.
c-Myc is a well-established oncogene that is expressed at increased levels in a variety of cancers. Research has shown that it has at least a
partial role in higher frequency of tumorigenesis in iPS-derived tissues. A study analyzing tumor formation from iPS-derived cells found
that 20% of tumors were attributable to reactivation of the c-Myc transgene [13]. Reprogramming has been performed without ectopic
expression of c-Myc in human adult dermal fibroblasts, demonstrating that it is not necessary for reprogramming [15]. However, the
omission of c-Myc significantly reduces the efficiency of reprogramming.
Sox2 is a transcription factor that heterodimerizes with Oct4 to activate genes involved in the maintenance of pluripotency [143]. This
heterodimer is also overexpressed in multiple forms of cancer including neuroblastoma [144] and liver cancer [145]. Sox2 is critical factor
in the self-renewal and tumorigenicity of melanoma cells [146]. Reprogramming has occurred in adult cells with only expression of Oct4
and Klf4 but this was in dermal papilla cells which have high endogenous levels of c-Myc and Sox2 [147].
Klf4 is important for maintaining self-renewal in stem cells. It influences reprogramming through direct interaction with Oct4 and Sox2
[148]. In regards to carcinogenesis, it has a dual function as both a tumor suppressor and an oncogene depending on the type of cancer. As
an oncogene, it is highly expressed in over 70% of breast cancers and is required for the maintenance of breast cancer stem cells [149]. It is
also overexpressed in colon cancer stem cell populations and its reduced expression decreases the ability of these cells to generate tumors
[150]. The function of Klf4 in cancer stem cells likely overlaps with its role in iPS cells.

5.2. Incomplete differentiation these CNV, deleted regions often contained tumor-suppressor genes,
while regions of chromosomal gain included oncogenes [92]. The rate
Clinical use of iPS-derived cells requires complete differentiation. of mutation in iPS cells has also been estimated to be ten times more
Any residual expression of genes related to pluripotency may increase frequent than in fibroblasts [93].
their tumorigenicity. Contamination of transplanted cells with iPS or
unsuccessfully reprogrammed cells cause cancer occurrence when 5.5. Epigenetic modifications
transplanted into patients. Achieving pure populations prior to intro-
duction into patients will require complete differentiation or complete Reprogramming is an epigenetic process that does not directly alter
elimination of undifferentiated cells from the pool meant for transplan- DNA sequence. During reprogramming, removal of epigenetic identities
tation. Attempts to generate homogenous differentiated cell popula- of the source cells is combined with induction of epigenetic modifica-
tions include elimination of pluripotent cells using cytotoxic antibodies tions similar to those of ES cells. Genetic alteration occurs only if
[86] and removal of undifferentiated cells based on cell surface mutations occur during reprogramming or if transgenes integrate into
molecules using magnetic-activated cell sorting (MACS) and fluores- genomes. Epigenetic changes induced by reprogramming may contri-
cence-activated cell sorting (FACS) [87]. bute to genomic instability. This could subsequently increase the rate of
genomic alterations, possibly in cancer-related genes. Accumulation of
5.3. Transgenic reactivation epigenetic alterations, especially aberrant DNA methylation, increases
the risk of carcinogenesis, particularly those associated with chronic
Reactivation of the reprogramming factors may induce carcinogen- inflammation [94,95]. iPS cells experience aberrant hyper-methylation
esis. As an example, ectopic expression of Oct3/4 [88] and Klf4 [89] in early passages which is continuously detected throughout repro-
could trigger dysplasia in vivo. Transgene reactivation is mainly a risk gramming [96].
when genes are integrated into the cell genome. To reduce genomic
integration, reprogramming factors have been introduced using non- 6. Autophagy, reprogramming and stemness maintenance
integrating Sendai viral vectors [23], episomal vectors [19], and
piggyBac transposons [90]. Reprogramming has also been induced Autophagy is a degradation process crucial for remodeling of
without transgenes, by alternatively transducing purified recombinant cellular homeostasis during cell and tissue development. Its deregula-
reprogramming proteins [38] or modified synthetic mRNAs [34]. tion has been observed in some diseases, and several types of treatments
However, these methods considerably reduce reprogramming effi- either trigger autophagy or use autophagy as a therapeutic target
ciency. [97–99]. A growing body of evidence shows that autophagy plays also
an indispensable role during reprogramming, differentiation and main-
5.4. Mutations acquired in vitro tenance of cellular stemness [100,101]. In the light of recent studies it
appears that autophagy is particularly important for early phases of cell
Therapeutic production of iPS cells requires a number of cell reprogramming during the generation of iPS cells [100,102–104]. It has
divisions as cells are cultured in vitro for weeks at a time. During this been shown that autophagy induced during reprogramming is regulated
period genetic mutations may accumulate. In addition, reprogramming by the mechanistic target of rapamycin complex 1 (mTORC1) [102] and
likely elicits stress response pathways within cells, increasing their that well-characterized mTOR inhibitors and autophagy activators
susceptibility to genetic mutations. The majority of mutations should be (PP242, rapamycin or resveratrol) notably improve the speed and
selected against but if mutations result in proliferative or antiapoptotic efficiency of iPS cells generation [103].
abilities they can be selected for during reprogramming, resulting in iPS It is also well documented that autophagy is responsible for
cells with genetic abnormalities. Whole genome sequencing has re- maintenance of stemness by preventing cellular senescence
vealed that iPS cells have a higher number of copy number variations [105,106]. Studies performed by Garcia-Prat et al. shows that satellite
(CNV) than do ES cells [91]. A separate study demonstrated that of cells with impaired autophagy machinery enter a senescence state,

1366
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

which is accompanied with mitochondrial dysfunctions and oxidative Transparency document


stress. Re-establishment of autophagy reverses senescence and restores
regenerative properties of satellite cells [106]. It is also suggested that The http://dx.doi.org/10.1016/j.bbamcr.2017.04.017 associated
autophagy may play a relevant role in protecting cellular stemness and with this article can be found, in online version.
self-renewal ability by reducing oxidative stress [107,108]. Induction of
autophagy by starvation or rapamycin, reduces ROS-induced DNA Acknowledgments
damage and helps in maintaining the stemness [107]. Studies on
human mesenchymal stem cells (hMSCs) showed that elevated ROS ACP thankfully acknowledges the support from GeCONiI
levels trigger FOXO3 induced autophagy and the knockdown of (POIG.02.03.01-24- 099/13). MJŁ and AH kindly acknowledge the
autophagy-related 7 (ATG7) results in FOXO3 downregulation and support from NCN grant #: 2016/21/B/NZ1/02812.
reduced capacity of hMSCs to handle elevated ROS levels. As a
consequence of defective autophagy hMSCs reduce their ability to References
differentiate into osteoblasts [109]. The importance of autophagy
during differentiation and tissue regeneration was also confirmed in [1] K. Takahashi, S. Yamanaka, Induction of pluripotent stem cells from mouse
the work of Saera-Vila et al. who showed that autophagy is activated in embryonic and adult fibroblast cultures by defined factors, Cell 126 (2006)
663–676.
response to zebrafish muscle injury, and blocking the autophagy using [2] K. Takahashi, et al., Induction of pluripotent stem cells from adult human
chloroquine or Atg5 knockdown reduces the rate of regeneration [110]. fibroblasts by defined factors, Cell 131 (2007) 861–872.
Although, no one so far described the role of autophagy in [3] J. Yu, et al., Induced pluripotent stem cell lines derived from human somatic cells,
Science 318 (2007) 1917–1920.
transdifferentiation, it can be presumed that its function is somehow [4] E. Farahani, et al., Cell adhesion molecules and their relation to (cancer) cell
similar to that observed during generation of iPS cells through stemness, Carcinogenesis 35 (2014) 747–759.
reprogramming. Therefore, selective blocking or inducing autophagy [5] F. González, et al., Methods for making induced pluripotent stem cells: repro-
gramming à la carte, Nat. Rev. Genet. 12 (2011) 231–242.
can be considered as a potential factor responsible of enhancing
[6] A.M. Wasik, et al., Reprogramming and carcinogenesis—parallels and distinctions,
processes of cellular conversion from one type of cell to another. Int. Rev. Cell Mol. Biol. 308 (2014) 167–203.
[7] R.L. Davis, et al., Expression of a single transfected cDNA converts fibroblasts to
myoblasts, Cell 51 (1987) 987–1000.
7. Transdifferentiation and carcinogenesis
[8] O.J. Rackham, et al., A predictive computational framework for direct repro-
gramming between human cell types, Nat. Genet. 48 (2016) 331–335.
Transdifferentiation does not carry as high a risk of carcinogenesis [9] Q. Zhou, et al., In vivo reprogramming of adult pancreatic exocrine cells to beta-
as reprogramming. This is because direct cell-to-cell conversion does cells, Nature 455 (2008) 627–632.
[10] Y.J. Nam, et al., Reprogramming of human fibroblasts toward a cardiac fate, Proc.
not require an intermediate pluripotent state, which is the source of Natl. Acad. Sci. U. S. A. 110 (2013) 5588–5593.
much of tumorigenic propensity in iPS cells. Transdifferentiated cells [11] A. Cieslar-Pobuda, et al., Human induced pluripotent stem cell differentiation and
are generally cultured for a shorter time than iPS cells and therefore are direct transdifferentiation into corneal epithelial-like cells, Oncotarget (2016).
[12] H. Xie, et al., Stepwise reprogramming of B cells into macrophages, Cell 117
not as susceptible to the accumulation of genetic mutations during in (2004) 663–676.
vitro culturing. Similar to reprogramming, transdifferentiation is pri- [13] K. Okita, et al., Generation of germline-competent induced pluripotent stem cells,
marily induced by transgene introduction. These genes are usually Nature 448 (2007) 313–317.
[14] M. Maekawa, et al., Direct reprogramming of somatic cells is promoted by
transcription factors determining the fate of the target cell type. As in maternal transcription factor Glis1, Nature 474 (2011) 225–229.
reprogramming, the characteristics of the vector used to overexpress [15] M. Nakagawa, et al., Generation of induced pluripotent stem cells without Myc
these transgenes could result in cancer by insertional mutagenesis or in from mouse and human fibroblasts, Nat. Biotechnol. 26 (2008) 101–106.
[16] A. Hotta, J. Ellis, Retroviral vector silencing during iPS cell induction: an
vivo-transgene reactivation. Similar gene delivery alternatives such as epigenetic beacon that signals distinct pluripotent states, J. Cell. Biochem. 105
non-integrating viruses, excisable vectors, and episomal vectors could (2008) 940–948.
be used to reduce this risk. Transdifferentiation also involves epigenetic [17] D.G. Miller, et al., Gene transfer by retrovirus vectors occurs only in cells that are
actively replicating at the time of infection, Mol. Cell. Biol. 10 (1990) 4239–4242.
processes and therefore, it may be subjected to the same epigenetic
[18] T. Zhao, et al., Immunogenicity of induced pluripotent stem cells, Nature 474
abnormalities that may predispose iPS cells to form tumors. It would be (2011) 212–215.
interesting to compare the cancer incidence between various implanted [19] K. Okita, et al., Generation of mouse induced pluripotent stem cells without viral
tissues obtained by reprogramming with subsequent differentiation, vectors, Science 322 (2008) 949–953.
[20] W. Zhou, C.R. Freed, Adenoviral gene delivery can reprogram human fibroblasts to
and by direct transdifferentiation. induced pluripotent stem cells, Stem Cells 27 (2009) 2667–2674.
[21] M. Stadtfeld, et al., Induced pluripotent stem cells generated without viral
8. Closing remarks integration, Science 322 (2008) 945–949.
[22] A. Harui, et al., Frequency and stability of chromosomal integration of adenovirus
vectors, J. Virol. 73 (1999) 6141–6146.
Nowadays regenerative medicine enters a new era. Reconstructive [23] N. Fusaki, et al., Efficient induction of transgene-free human pluripotent stem cells
plastic surgery, organ transplantation, various prosthesis and implants using a vector based on Sendai virus, an RNA virus that does not integrate into the
host genome, Proc. Jpn. Acad. Ser. B Phys. Biol. Sci. 85 (2009) 348–362.
are well known procedures. Generation of iPS cells offers to use patient- [24] K. Si-Tayeb, et al., Generation of human induced pluripotent stem cells by simple
specific somatic cells for tissue and organ regeneration. This new very transient transfection of plasmid DNA encoding reprogramming factors, BMC Dev.
promising perspective gives hope to all patients who at present have Biol. 10 (2010) 81.
[25] K. Hu, All roads lead to induced pluripotent stem cells: the technologies of iPSC
incurable diseases caused by the loss of a certain type of cell, i.e. type-1 generation, Stem Cells Dev. 23 (2014) 1285–1300.
diabetes and Parkinson's disease. It also offers hope for all people [26] K. Hu, I. Slukvin, Generation of transgene-free iPSC lines from human normal and
suffering from paralysis due to spinal cord injury. The use of tissues neoplastic blood cells using episomal vectors, Methods Mol. Biol. 997 (2013)
163–176.
derived from autologous iPS cells is free from all disadvantages
[27] J. Yu, et al., Human induced pluripotent stem cells free of vector and transgene
associated with organ transplantation like immunosuppression, and sequences, Science 324 (2009) 797–801.
graft versus host disease. [28] T. Kameda, et al., A severe de novo methylation of episomal vectors by human ES
Transdifferentiation seems to be a promising alternative for repro- cells, Biochem. Biophys. Res. Commun. 349 (2006) 1269–1277.
[29] E.R. Leight, B. Sugden, Establishment of an oriP replicon is dependent upon an
gramming as it could minimize the greatest risk associated with infrequent, epigenetic event, Mol. Cell. Biol. 21 (2001) 4149–4161.
reprogramming, a risk of malignant transformation. More basic re- [30] P. Mayrhofer, et al., Use of minicircle plasmids for gene therapy, Methods Mol.
search is still needed to better understand all mechanisms involved in Biol. 542 (2009) 87–104.
[31] Z.Y. Chen, et al., Minicircle DNA vectors devoid of bacterial DNA result in
these processes, factors limiting their efficiency and how to minimize persistent and high-level transgene expression in vivo, Mol. Ther. 8 (2003)
all risks associated with them. Several safety-related questions urgently 495–500.
await answers through systematic studies. [32] F. Jia, et al., A nonviral minicircle vector for deriving human iPS cells, Nat.

1367
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

Methods 7 (2010) 197–199. Reprogram. 12 (2010) 641–653.


[33] K.H. Narsinh, et al., Generation of adult human induced pluripotent stem cells [73] T.L. Downing, et al., Biophysical regulation of epigenetic state and cell repro-
using nonviral minicircle DNA vectors, Nat. Protoc. 6 (2011) 78–88. gramming, Nat. Mater. 12 (2013) 1154–1162.
[34] L. Warren, et al., Highly efficient reprogramming to pluripotency and directed [74] T. Teramura, et al., Mechanical stimulation of cyclic tensile strain induces
differentiation of human cells with synthetic modified mRNA, Cell Stem Cell 7 reduction of pluripotent related gene expressions via activation of rho/ROCK and
(2010) 618–630. subsequent decreasing of AKT phosphorylation in human induced pluripotent
[35] L. Warren, et al., Feeder-free derivation of human induced pluripotent stem cells stem cells, Biochem. Biophys. Res. Commun. 417 (2012) 836–841.
with messenger RNA, Sci. Rep. 2 (2012) 657. [75] A. Cieslar-Pobuda, et al., The expression pattern of PFKFB3 enzyme distinguishes
[36] K. Hu, Vectorology and factor delivery in induced pluripotent stem cell repro- between induced-pluripotent stem cells and cancer stem cells, Oncotarget (2015).
gramming, Stem Cells Dev. 23 (2014) 1301–1315. [76] Y. Yoshida, et al., Hypoxia enhances the generation of induced pluripotent stem
[37] D. Kim, et al., Generation of human induced pluripotent stem cells by direct cells, Cell Stem Cell 5 (2009) 237–241.
delivery of reprogramming proteins, Cell Stem Cell 4 (2009) 472–476. [77] C.E. Forristal, et al., Hypoxia inducible factors regulate pluripotency and
[38] H. Zhou, et al., Generation of induced pluripotent stem cells using recombinant proliferation in human embryonic stem cells cultured at reduced oxygen tensions,
proteins, Cell Stem Cell 4 (2009) 381–384. Reproduction 139 (2010) 85–97.
[39] G.D. Greggains, et al., Therapeutic potential of somatic cell nuclear transfer for [78] B. Feng, et al., Molecules that promote or enhance reprogramming of somatic cells
degenerative disease caused by mitochondrial DNA mutations, Sci. Rep. 4 (2014) to induced pluripotent stem cells, Cell Stem Cell 4 (2009) 301–312.
3844. [79] D. Huangfu, et al., Induction of pluripotent stem cells by defined factors is greatly
[40] S. Hiendleder, Mitochondrial DNA inheritance after SCNT, Adv. Exp. Med. Biol. improved by small-molecule compounds, Nat. Biotechnol. 26 (2008) 795–797.
591 (2007) 103–116. [80] Y. Rais, et al., Deterministic direct reprogramming of somatic cells to pluripotency,
[41] M. Abad, et al., Reprogramming in vivo produces teratomas and iPS cells with Nature 502 (2013) 65–70.
totipotency features, Nature 502 (2013) 340–345. [81] J. Hanna, et al., Direct cell reprogramming is a stochastic process amenable to
[42] W.B. Hurlbut, Ethics and embryonic stem cell research: altered nuclear transfer as acceleration, Nature 462 (2009) 595–601.
a way forward, BioDrugs 21 (2007) 79–83. [82] R. Ho, et al., Mechanistic insights into reprogramming to induced pluripotency, J.
[43] M. Tada, et al., Embryonic germ cells induce epigenetic reprogramming of somatic Cell. Physiol. 226 (2011) 868–878.
nucleus in hybrid cells, EMBO J. 16 (1997) 6510–6520. [83] L. Shao, et al., Generation of iPS cells using defined factors linked via the self-
[44] M. Tada, et al., Nuclear reprogramming of somatic cells by in vitro hybridization cleaving 2A sequences in a single open reading frame, Cell Res. 19 (2009)
with ES cells, Curr. Biol. 11 (2001) 1553–1558. 296–306.
[45] C.A. Cowan, et al., Nuclear reprogramming of somatic cells after fusion with [84] I. Gutierrez-Aranda, et al., Human induced pluripotent stem cells develop
human embryonic stem cells, Science 309 (2005) 1369–1373. teratoma more efficiently and faster than human embryonic stem cells regardless
[46] J. Yu, et al., Human embryonic stem cells reprogram myeloid precursors following the site of injection, Stem Cells 28 (2010) 1568–1570.
cell-cell fusion, Stem Cells 24 (2006) 168–176. [85] Y. Li, et al., Generation of iPSCs from mouse fibroblasts with a single gene, Oct4,
[47] J. Markova, et al., Prognostic impact of DNMT3A mutations in patients with and small molecules, Cell Res. 21 (2011) 196–204.
intermediate cytogenetic risk profile acute myeloid leukemia, Eur. J. Haematol. 88 [86] A.B. Choo, et al., Selection against undifferentiated human embryonic stem cells
(2012) 128–135. by a cytotoxic antibody recognizing podocalyxin-like protein-1, Stem Cells 26
[48] J.T. Do, H.R. Scholer, Nuclei of embryonic stem cells reprogram somatic cells, (2008) 1454–1463.
Stem Cells 22 (2004) 941–949. [87] C.Y. Fong, et al., Separation of SSEA-4 and TRA-1-60 labelled undifferentiated
[49] H. Matsumura, et al., Targeted chromosome elimination from ES-somatic hybrid human embryonic stem cells from a heterogeneous cell population using
cells, Nat. Methods 4 (2007) 23–25. magnetic-activated cell sorting (MACS) and fluorescence-activated cell sorting
[50] D. Pralong, et al., A novel method for somatic cell nuclear transfer to mouse (FACS), Stem Cell Rev. 5 (2009) 72–80.
embryonic stem cells, Cloning Stem Cells 7 (2005) 265–271. [88] K. Hochedlinger, et al., Ectopic expression of Oct-4 blocks progenitor-cell
[51] J.K. Ichida, et al., A small-molecule inhibitor of tgf-Beta signaling replaces sox2 in differentiation and causes dysplasia in epithelial tissues, Cell 121 (2005) 465–477.
reprogramming by inducing nanog, Cell Stem Cell 5 (2009) 491–503. [89] K.W. Foster, et al., Induction of KLF4 in basal keratinocytes blocks the prolifera-
[52] N. Miyoshi, et al., Reprogramming of mouse and human cells to pluripotency using tion-differentiation switch and initiates squamous epithelial dysplasia, Oncogene
mature microRNAs, Cell Stem Cell 8 (2011) 633–638. 24 (2005) 1491–1500.
[53] P. Yang, et al., RCOR2 is a subunit of the LSD1 complex that regulates ESC [90] K. Kaji, et al., Virus-free induction of pluripotency and subsequent excision of
property and substitutes for SOX2 in reprogramming somatic cells to pluripotency, reprogramming factors, Nature 458 (2009) 771–775.
Stem Cells 29 (2011) 791–801. [91] S.M. Hussein, et al., Copy number variation and selection during reprogramming
[54] P. Hou, et al., Pluripotent stem cells induced from mouse somatic cells by small- to pluripotency, Nature 471 (2011) 58–62.
molecule compounds, Science 341 (2013) 651–654. [92] L.C. Laurent, et al., Dynamic changes in the copy number of pluripotency and cell
[55] A. Cieslar-Pobuda, M.J. Los, Prospects and limitations of “click-chemistry”-based proliferation genes in human ESCs and iPSCs during reprogramming and time in
DNA labeling technique employing 5-ethynyl-2'deoxyuridine (EdU), Cytometry A culture, Cell Stem Cell 8 (2011) 106–118.
83 (2013) 977–978. [93] A. Gore, et al., Somatic coding mutations in human induced pluripotent stem cells,
[56] R.M. Marion, et al., A p53-mediated DNA damage response limits reprogramming Nature 471 (2011) 63–67.
to ensure iPS cell genomic integrity, Nature 460 (2009) 1149–1153. [94] T. Nakajima, et al., Higher methylation levels in gastric mucosae significantly
[57] E. Wiechec, Implications of genomic instability in the diagnosis and treatment of correlate with higher risk of gastric cancers, Cancer Epidemiol. Biomark. Prev. 15
breast cancer, Expert. Rev. Mol. Diagn. 11 (2011) 445–453. (2006) 2317–2321.
[58] D.P. Bartel, MicroRNAs: genomics, biogenesis, mechanism, and function, Cell 116 [95] T. Ushijima, N. Hattori, Molecular pathways: involvement of Helicobacter pylori-
(2004) 281–297. triggered inflammation in the formation of an epigenetic field defect, and its
[59] S. Vasudevan, et al., Switching from repression to activation: microRNAs can up- usefulness as cancer risk and exposure markers, Clin. Cancer Res. 18 (2012)
regulate translation, Science 318 (2007) 1931–1934. 923–929.
[60] W.T. Guo, et al., Micro-management of pluripotent stem cells, Protein Cell 5 [96] K. Nishino, et al., DNA methylation dynamics in human induced pluripotent stem
(2014) 36–47. cells over time, PLoS Genet. 7 (2011) e1002085.
[61] Z. Li, et al., Small RNA-mediated regulation of iPS cell generation, EMBO J. 30 [97] S. Ghavami, et al., Airway mesenchymal cell death by mevalonate cascade
(2011) 823–834. inhibition: integration of autophagy, unfolded protein response and apoptosis
[62] D.A. Card, et al., Oct4/Sox2-regulated miR-302 targets cyclin D1 in human focusing on Bcl2 family proteins, Biochim. Biophys. Acta 1843 (2014) 1259–1271.
embryonic stem cells, Mol. Cell. Biol. 28 (2008) 6426–6438. [98] S. Ghavami, et al., Autophagy and apoptosis dysfunction in neurodegenerative
[63] Y.J. Choi, et al., miR-34 miRNAs provide a barrier for somatic cell reprogramming, disorders, Prog. Neurobiol. 112 (2014) 24–49.
Nat. Cell Biol. 13 (2011) 1353–1360. [99] J.R. Jangamreddy, et al., Salinomycin induces activation of autophagy, mitophagy
[64] T.R. Leonardo, et al., The functions of microRNAs in pluripotency and repro- and affects mitochondrial polarity: differences between primary and cancer cells,
gramming, Nat. Cell Biol. 14 (2012) 1114–1121. Biochim. Biophys. Acta 1833 (2013) 2057–2069.
[65] F. Anokye-Danso, et al., Highly efficient miRNA-mediated reprogramming of [100] S. Wang, et al., Autophagy and cell reprogramming, Cell. Mol. Life Sci. 72 (2015)
mouse and human somatic cells to pluripotency, Cell Stem Cell 8 (2011) 376–388. 1699–1713.
[66] A. Helwak, et al., Mapping the human miRNA interactome by CLASH reveals [101] F.V. Sbrana, et al., The role of autophagy in the maintenance of stemness and
frequent noncanonical binding, Cell 153 (2013) 654–665. differentiation of mesenchymal stem cells, Stem Cell Rev. 12 (2016) 621–633.
[67] B.P. Lewis, et al., Conserved seed pairing, often flanked by adenosines, indicates [102] Y. Wu, et al., Autophagy and mTORC1 regulate the stochastic phase of somatic cell
that thousands of human genes are microRNA targets, Cell 120 (2005) 15–20. reprogramming, Nat. Cell Biol. 17 (2015) 715–725.
[68] H. Hong, et al., Suppression of induced pluripotent stem cell generation by the [103] J.A. Menendez, et al., mTOR-regulated senescence and autophagy during repro-
p53-p21 pathway, Nature 460 (2009) 1132–1135. gramming of somatic cells to pluripotency: a roadmap from energy metabolism to
[69] Y. Li, et al., The p53-PUMA axis suppresses iPSC generation, Nat. Commun. 4 stem cell renewal and aging, Cell Cycle 10 (2011) 3658–3677.
(2013) 2174. [104] T. Ma, et al., Atg5-independent autophagy regulates mitochondrial clearance and
[70] C.A. Street, B.A. Bryan, Rho kinase proteins—pleiotropic modulators of cell is essential for iPSC reprogramming, Nat. Cell Biol. 17 (2015) 1379–1387.
survival and apoptosis, Anticancer Res. 31 (2011) 3645–3657. [105] L. Garcia-Prat, et al., Autophagy: a decisive process for stemness, Oncotarget 7
[71] H. Kurosawa, Application of rho-associated protein kinase (ROCK) inhibitor to (2016) 12286–12288.
human pluripotent stem cells, J. Biosci. Bioeng. 114 (2012) 577–581. [106] L. Garcia-Prat, et al., Autophagy maintains stemness by preventing senescence,
[72] W.H. Lai, et al., ROCK inhibition facilitates the generation of human-induced Nature 529 (2016) 37–42.
pluripotent stem cells in a defined, feeder-, and serum-free system, Cell. [107] J. Hou, et al., Autophagy prevents irradiation injury and maintains stemness

1368
A. Cieślar-Pobuda et al. BBA - Molecular Cell Research 1864 (2017) 1359–1369

through decreasing ROS generation in mesenchymal stem cells, Cell Death Dis. 4 [130] K. Nishimura, et al., Development of defective and persistent Sendai virus vector: a
(2013) e844. unique gene delivery/expression system ideal for cell reprogramming, J. Biol.
[108] H. Chen, et al., Autophagy prevents oxidative stress-induced loss of self-renewal Chem. 286 (2011) 4760–4771.
capacity and stemness in human tendon stem cells by reducing ROS accumulation, [131] K. Okita, S. Yamanaka, Induced pluripotent stem cells: opportunities and
Cell. Physiol. Biochem. 39 (2016) 2227–2238. challenges, Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 366 (2011) 2198–2207.
[109] M.C. Gomez-Puerto, et al., Activation of autophagy by FOXO3 regulates redox [132] Y. Yonemitsu, et al., Efficient gene transfer to airway epithelium using recombi-
homeostasis during osteogenic differentiation, Autophagy 12 (2016) 1804–1816. nant Sendai virus, Nat. Biotechnol. 18 (2000) 970–973.
[110] A. Saera-Vila, et al., Autophagy regulates cytoplasmic remodeling during cell [133] H. Ban, et al., Efficient generation of transgene-free human induced pluripotent
reprogramming in a zebrafish model of muscle regeneration, Autophagy 12 (2016) stem cells (iPSCs) by temperature-sensitive Sendai virus vectors, Proc. Natl. Acad.
1864–1875. Sci. U. S. A. 108 (2011) 14234–14239.
[111] P. Li, et al., Reprogramming of T cells to natural killer-like cells upon Bcl11b [134] K. Woltjen, et al., piggyBac transposition reprograms fibroblasts to induced
deletion, Science 329 (2010) 85–89. pluripotent stem cells, Nature 458 (2009) 766–770.
[112] H. Noguchi, et al., PDX-1 protein containing its own antennapedia-like protein [135] A. Sarkar, et al., Molecular evolutionary analysis of the widespread piggyBac
transduction domain can transduce pancreatic duct and islet cells, Diabetes 52 transposon family and related “domesticated” sequences, Mol. Gen. Genet. MGG
(2003) 1732–1737. 270 (2003) 173–180.
[113] V. Aviv, et al., Exendin-4 promotes liver cell proliferation and enhances the PDX-1- [136] S. Miyazaki, et al., Emerging methods for preparing iPS cells, Jpn. J. Clin. Oncol.
induced liver to pancreas transdifferentiation process, J. Biol. Chem. 284 (2009) 42 (2012) 773–779.
33509–33520. [137] H. Niemann, A. Lucas-Hahn, Somatic cell nuclear transfer cloning: practical
[114] S. Sekiya, A. Suzuki, Direct conversion of mouse fibroblasts to hepatocyte-like cells applications and current legislation, Reprod. Domest. Anim. 47 (Suppl. 5) (2012)
by defined factors, Nature 475 (2011) (390-U148). 2–10.
[115] T. Vierbuchen, et al., Direct conversion of fibroblasts to functional neurons by [138] T. Bru, et al., Rapid induction of pluripotency genes after exposure of human
defined factors, Nature 463 (2010) 1035–U1050. somatic cells to mouse ES cell extracts, Exp. Cell Res. 314 (2008) 2634–2642.
[116] R. Ambasudhan, et al., Direct reprogramming of adult human fibroblasts to [139] C.K. Taranger, et al., Induction of dedifferentiation, genomewide transcriptional
functional neurons under defined conditions, Cell Stem Cell 9 (2011) 113–118. programming, and epigenetic reprogramming by extracts of carcinoma and
[117] B. Berninger, et al., Functional properties of neurons derived from in vitro embryonic stem cells, Mol. Biol. Cell 16 (2005) 5719–5735.
reprogrammed postnatal astroglia, J. Neurosci. 27 (2007) 8654–8664. [140] C.J. Hutchison, H.J. Worman, A-type lamins: guardians of the soma? Nat. Cell Biol.
[118] M. Caiazzo, et al., Direct generation of functional dopaminergic neurons from 6 (2004) 1062–1067.
mouse and human fibroblasts, Nature 476 (2011) (224-U151). [141] C.G. Liu, et al., Clinical implications of stem cell gene Oct-4 expression in breast
[119] U. Pfisterer, et al., Direct conversion of human fibroblasts to dopaminergic cancer, Ann. Surg. 253 (2011) 1165–1171.
neurons, Proc. Natl. Acad. Sci. U. S. A. 108 (2011) 10343–10348. [142] M.H. Asadi, et al., OCT4B1, a novel spliced variant of OCT4, is highly expressed in
[120] J.A. Efe, et al., Conversion of mouse fibroblasts into cardiomyocytes using a direct gastric cancer and acts as an antiapoptotic factor, Int. J. Cancer 128 (2011)
reprogramming strategy, Nat. Cell Biol. 13 (2011) 215–U261. 2645–2652.
[121] M. Ieda, et al., Direct reprogramming of fibroblasts into functional cardiomyocytes [143] V. Lefebvre, et al., Control of cell fate and differentiation by Sry-related high-
by defined factors, Cell 142 (2010) 375–386. mobility-group box (sox) transcription factors, Int. J. Biochem. Cell Biol. 39 (2007)
[122] E. Meshorer, et al., Hyperdynamic plasticity of chromatin proteins in pluripotent 2195–2214.
embryonic stem cells, Dev. Cell 10 (2006) 105–116. [144] S. Yang, et al., Oct4 and Sox2 are overexpressed in human neuroblastoma and
[123] A. Mattout, et al., Global epigenetic changes during somatic cell reprogramming to inhibited by chemotherapy, Oncol. Rep. 28 (2012) 186–192.
iPS cells, J. Mol. Cell Biol. 3 (2011) 341–350. [145] P. Huang, et al., Role of Sox2 and Oct4 in predicting survival of hepatocellular
[124] R.M. Marion, et al., Telomeres acquire embryonic stem cell characteristics in carcinoma patients after hepatectomy, Clin. Biochem. 44 (2011) 582–589.
induced pluripotent stem cells, Cell Stem Cell 4 (2009) 141–154. [146] R. Santini, et al., SOX2 regulates self-renewal and tumorigenicity of human
[125] S. Loewer, et al., Large intergenic non-coding RNA-RoR modulates reprogramming melanoma-initiating cells, Oncogene 33 (2014) 4697–4708.
of human induced pluripotent stem cells, Nat. Genet. 42 (2010) 1113–1117. [147] S.Y. Tsai, et al., Oct4 and klf4 reprogram dermal papilla cells into induced
[126] B.K. Chou, et al., Efficient human iPS cell derivation by a non-integrating plasmid pluripotent stem cells, Stem Cells 28 (2010) 221–228.
from blood cells with unique epigenetic and gene expression signatures, Cell Res. [148] Z. Wei, et al., Klf4 interacts directly with Oct4 and Sox2 to promote reprogram-
21 (2011) 518–529. ming, Stem Cells 27 (2009) 2969–2978.
[127] M. Stadtfeld, et al., Defining molecular cornerstones during fibroblast to iPS cell [149] F. Yu, et al., Kruppel-like factor 4 (KLF4) is required for maintenance of breast
reprogramming in mouse, Cell Stem Cell 2 (2008) 230–240. cancer stem cells and for cell migration and invasion, Oncogene 30 (2011)
[128] M. Patel, S. Yang, Advances in reprogramming somatic cells to induced pluripotent 2161–2172.
stem cells, Stem Cell Rev. 6 (2010) 367–380. [150] Z. Leng, et al., Kruppel-like factor 4 acts as an oncogene in colon cancer stem cell-
[129] A. Schambach, et al., Biosafety features of lentiviral vectors, Hum. Gene Ther. 24 enriched spheroid cells, PLoS One 8 (2013) e56082.
(2013) 132–142.

1369

You might also like