ISSN 0006-2979, Biochemistry (Moscow), 2024, Vol. 89, No. 5, pp. 799-816 © Pleiades Publishing, Ltd., 2024.
799
REVIEW
Dysregulation of Immune Tolerance to Autologous iPSCs
and Their Differentiated Derivatives
Margarita E. Bogomiakova
1,2,a
*, Alexandra N. Bogomazova
1,2
,
and Maria A. Lagarkova
1,3
1
Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency,
119435 Moscow, Russia
2
Center for Precision Genome Editing and Genetic Technologies for Biomedicine,
Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency,
119435 Moscow, Russia
3
Lomonosov Moscow State University, 119991 Moscow, Russia
a
e-mail: margbog@rcpcm.ru
Received October 12, 2023
Revised December 21, 2023
Accepted February 13, 2024
AbstractInduced pluripotent stem cells (iPSCs), capable of differentiating into any cell type, are a promising tool
for solving the problem of donor organ shortage. In addition, reprogramming technology makes it possible to ob-
tain a personalized, i.e., patient-specific, cell product transplantation of which should not cause problems related
to histocompatibility of the transplanted tissues and organs. At the same time, inconsistent information about the
main advantage of autologous iPSC-derivatives– lack of immunogenicity– still casts doubt on the possibility of
using such cells beyond immunosuppressive therapy protocols. This review is devoted to immunogenic properties
of the syngeneic and autologous iPSCs and their derivatives, as well as to the reasons for dysregulation of their
immune tolerance.
DOI: 10.1134/S0006297924050031
Keywords: induced pluripotent stem cells, immune response, immunogenicity, immunotolerance, T-cells, NK-cells,
differentiation
Abbreviations: B2M,beta-2-microglobulin; CD,clusters of differentiation; ESCs, embryonic stem cells; HLA, human leuko-
cyte antigen; iPSCs,induced pluripotent stem cells; KIR,killer-cell immunoglobulin-like receptor; NK,natural killer cells;
PSCs,pluripotent stem cells; RPE,retinal pigment epithelium; SMCs,smooth muscle cells.
* To whom correspondence should be addressed.
INTRODUCTION
Human pluripotent stem cells (PSCs), which in-
clude embryonic stem cells (ESCs) and induced plu-
ripotent stem cells (iPSCs), can unlimitedly proliferate,
and differentiate into almost any type of somatic cells
[1, 2]. These unique properties make them an attrac-
tive and promising tool for modeling various diseases
and drug development [3,4]. Hopes are pinned on dif-
ferentiated derivatives of PSCs as a source of materi-
al for cell therapy, which should solve the problem of
shortage of donor organs and tissues [5].
It is still too early to discuss widespread introduc-
tion of the PSC technology into clinical practice. Only
26  years have passed since the discovery of human
ESCs in 1998 [1], and active work is currently ongoing
to improve protocols for differentiating PSCs into spe-
cialized cells and obtaining three-dimensional struc-
tured tissues in vitro [4]. Another stumbling block to
integrating PSCs into the clinic is high cost of the tech-
nology. According to the recent estimate, generating
a clinical grade iPSCs line under good manufacturing
practice (GMP) costs approximately U$800,000 [4]. An-
other limiting factor is long time required to obtain a
new iPSC line and its subsequent differentiation into
the desired cell type [6]. Additionally, there are cur-
rently no developed standardization parameters that
would be applied to both iPSCs [7] and their differ-
BOGOMIAKOVA et al.800
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
entiated derivatives [8]. Thus, at least in the coming
years, patient-specific iPSC-based therapy is unlikely
to become widespread. Interestingly, almost half of the
clinical trials of PSC-derivatives used cell products de-
rived from only five ESC lines; however, the number
of studies of cells derived from iPSCs has increased
significantly over the past few years [9]. According to
the Federal Law No. 180, using ESCs to develop, pro-
duce, and apply biomedical cell products is prohibited
in Russia. Therefore, only iPSC-derivatives can be used
in clinical practice.
Despite the obvious economic advantages asso-
ciated with production and “scaling-up” of allogeneic
PSC-derivatives, the issue of immune rejection remains
unresolved due to the high polymorphism of the genes
of the major histocompatibility complex (HLA). To pre-
vent immune rejection during allogeneic tissue and
organ transplantation, patients must undergo lifelong
immunosuppressive treatment and its associated side
effects [10]. Initially, it was believed that personalized
therapy based on autologous iPSC-derivatives could cir-
cumvent the problem of histocompatibility [11]. How-
ever, some researchers report that immune response
against the syngeneic and autologous iPSCs is still
possible, which casts doubt on the main advantage
of autologous iPSCs– lack of immunogenicity [12-15].
Thereasons for this phenomenon have yet to be thor-
oughly studied. In this review, we tried to shed light
on the mechanisms of impaired immune tolerance
to autologous iPSCs. It is worth emphasizing that un-
derstanding effects of the significant immune effector
cells– T and NK-cells– on various types of cells and,
in the long-term perspective, tissues derived from
iPSCs will help to find approaches to their suppres-
sion and will be of great importance for successful de-
velopment of translational medicine.
IMMUNOGENICITY OF SYNGENEIC
AND AUTOLOGOUS iPSCs
AND THEIR DERIVATIVES TOWARD T-CELLS
The possibility that cells differentiated from au-
tologous iPSCs can provoke an immune response was
widely considered only after the publication of Zhao
etal. [12]. In this study, the authors showed that sub-
cutaneous administration of iPSCs to syngeneic, i.e.,
linear, mice led to formation of teratomas, where T-cell
infiltration zones were found. Moreover, the process
was accompanied by subsequent necrosis and regres-
sion of the resulting teratomas. At the same time, ter-
atomas formed after administration of the syngeneic
ESCs with the same genetic background caused an
immune response much less often. The authors not-
ed that the teratomas from iPSCs were rarely rejected
when episomal reprogramming was used. However,
in this case, the formed teratomas were rejected with
the frequency of 10-20%, and most of them were also
infiltrated by T-cells. Nevertheless, these results were
met with some skepticism by the scientific community,
mainly because undifferentiated iPSCs are not consid-
ered as a source of cells for clinical use [11].
More recent studies have been somewhat contra-
dictory, although most still indicated lack of immuno-
genicity of the syngeneic iPSCs derivatives. The oppo-
site findings were reported only in a few studies. Thus,
additional evidence of immunogenicity of the cells
derived from syngeneic iPSCs was presented in 2013
by Araki etal. [16]. The authors reported similar fre-
quency of teratoma rejection formed by both syngene-
ic iPSCs and ESCs. It was suggested that the immune
response to teratomas is potentially related to the ex-
pression of genes regulating pluripotency. In particu-
lar, the authors relied on the previously obtained data
that the transcription factor OCT4 may have immuno-
genic properties [17]. In addition, Araki et al., for the
first time, found signs of immune response to the ter-
minally differentiated derivatives of syngeneic iPSCs.
Transplantation of cardiomyocytes differentiated from
the iPSCs led to the significant T-cell infiltration of the
graft in the syngeneic mice [16].
Another study reported complete survival of ter-
atomas formed by the syngeneic ESCs, although some
teratomas still showed areas of T-cell infiltration [18].
Furthermore, the authors compared immunogenicity
of endothelial cells, hepatocytes, and neuron precur-
sors differentiated from the syngeneic ESCs and iPSCs.
Differentiated iPSCs derivatives did not induce signs
of specific T-cell response either in the in  vitro model
or after transplantation into the syngeneic mice. Thus,
Guha etal. [18] showed that the degree of immunoge-
nicity of iPSCs can decrease in the process of differen-
tiation. The authors of another work [19] came to the
same conclusion. Analysis of the functional state of im-
mune cells found in the transplantation area showed
that teratomas were infiltrated predominantly by the
cytotoxic T-cells, while endothelial cells differentiat-
ed from iPSCs – by the regulatory T-cells and macro-
phages [19]. Another study reported complete survival
of teratomas formed by the syngeneic ESCs, although
some teratomas still showed areas of T-cell infiltration
[18]. Moreover, the authors compared immunogenicity
of endothelial cells, hepatocytes, and neuron precur-
sors differentiated from the syngeneic ESCs and iPSCs.
Differentiated iPSCs derivatives did not induce signs of
specific T-cell response either in the invitro model or
after transplantation into syngeneic mice. Thus, Guha
et al. [18] showed that the degree of immunogenicity
of iPSCs can decrease in the differentiation process.
The authors of another work [19] came to the same
conclusion. Analysis of the functional state of immune
cells found in the transplantation area showed that
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 801
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
teratomas were infiltrated predominantly by the cyto-
toxic T-cells, but endothelial cells differentiated from
iPSCs– by the regulatory T-cells and macrophages [19].
Another study with the non-human primate mod-
el was published in 2013 [20]. The authors compared
immune response to autologous and allogeneic trans-
plantation of the iPSC-differentiated midbrain dopa-
minergic neurons into Macaca fascicularis brains [20].
It was found that the significant amounts of microglia
and T-cells infiltrated the allografts, while autologous
neurons elicited minimal immune cell response. Simi-
lar work was done with the neural precursors of iPSCs,
where no significant infiltration was observed in the
autologous cell transplantation areas [21,22]. Interest-
ingly, Morizane etal. detected a limited T-cell response
to the autologous dopaminergic neurons in some ex-
perimental groups, if they were differentiated from
the iPSCs derived by retroviral transfection [20]. How-
ever, when the iPSCs were obtained by the episomal
reprogramming system, their derivatives did not lead
to the immune response of autologous T-cells. These
data indicate that viral integration of pluripotency fac-
tors during the iPSCs production may affect immuno-
genicity of the cell products.
Some limitations of the work of Morizane et al.
[20] are worth noting. First, the authors were able to
analyze immune response only after euthanasia of the
animals, approximately 3-4 months after transplan-
tation. Attempts were made to track dynamics of the
immune response using positron emission tomography
and measuring cytokine content in blood and cerebro-
spinal fluid. However, the results were highly variable
and correlated poorly with the postmortem histologic
data. Additional time points could more accurately de-
termine likelihood of the immune response to trans-
plantation of autologous iPSC-derivatives. Moreover,
although the authors demonstrated that the degree
of immune response was higher when the allogeneic
dopaminergic neurons were transplanted, rejection
was not observed even without immunosuppressive
therapy. This phenomenon could be explained by the
brain being an immune-privileged organ. Moreover,
these data are consistent with the clinical observations
of long-term survival of the allogeneic dopaminergic
neurons derived from the fetal material in the patients
with Parkinson’s disease who received only short-term
or no immunosuppression [23,24].
All previous studies have described immunogenic-
ity of the animal iPSCs– mice and primates. However,
studying immunogenicity of the autologous human
iPSC-derivatives is essential for clinical application.
In 2015, Zhao etal. studied this issue in a humanized
mouse model with the reconstructed human immune
system [13]. They found T-cell infiltration and tissue
necrosis areas in the most teratomas formed from
iPSCs. However, the degree of immune response to
the autologous iPSCs was weaker than to the allogene-
ic ESCs. Therefore, the authors hypothesized that only
certain derivatives of iPSCs could induce rejection.
In addition, deep sequencing of the T-cell receptor
(TCR) repertoire of the infiltrating lymphocytes re-
vealed their oligoclonal character, pointing to the an-
tigen-specific response of T-lymphocytes to the autol-
ogous iPSCs. For allogeneic ESCs, the polyclonal TCR
repertoire was revealed.
Furthermore, histological sections of the terato-
mas infiltrated with T-cells were analyzed to identify
potentially immunogenic tissues. The authors found
that the desmin-positive smooth muscle cells (SMCs)
were significantly more frequently surrounded by
the infiltrating T-cells. In contrast, the retinal pigment
epithelial cells (RPE) were almost never infiltrated by
T-cells. Next, the authors compared immunogenicity of
the two cell types, SMCs and RPE. It turned out that the
autologous SMCs were more immunogenic due to dys-
regulated expression of the tumor-associated genes,
particularly HORMAD1 and ZG16. Ectopic expression
of ZG16 in the RPE cells resulted in the significant
T-cell response in autologous recipients.
Thus, the data on immunogenicity of syngeneic
and autologous iPSCs derivatives against T-cells are
contradictory. Nevertheless, most of them are encour-
aging, such as, for example, relatively recent works
performed with the pigs [25], monkeys [26], humanized
mice [27] models as well as invitro studies [15,28], where
immune tolerance to the cellular products derived
from iPSCs was demonstrated.
IMMUNOGENICITY OF SYNGENEIC
AND AUTOLOGOUS iPSCs
AND THEIR DERIVATIVES TOWARD NK-CELLS
While primary function of T-lymphocytes is to
recognize foreign molecules, including neoepitopes,
NK-cells have a different principle of immunological
recognition. The classical “missing self” hypothesis
assumes that NK-cells recognize and destroy all cells
lacking HLA class I molecules [29]. Based on the cur-
rent viewpoint, activation of NK-cells is a more com-
plex concept and is determined by interaction of the
signals from two types of receptors on their surface:
activating and inhibitory [30]. Predominance of the
inhibitory signals upon interaction with the target cell
does not disturb the anergy of NK-cells, while predom-
inance of the activating signals triggers their cytotoxic
program. In turn, predominance of the activating sig-
nals can be caused by increase in the amount (level) of
the ligands for activating NK-cell receptors on the tar-
get cells and decrease in the inhibitory ligands, mainly
HLA class I molecules. Such imbalance in physiologi-
cal conditions is determined by various pathological
BOGOMIAKOVA et al.802
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
processes, including oncogenesis, viral and bacterial
infections, and stress [31].
Activity of NK-cells against undifferentiated PSCs,
including syngeneic and autologous ones, was previ-
ously highlighted in several in  vitro studies [32,  33].
Generally, low expression of HLA class I molecules is
a characteristic feature of PSCs [34], so high activity of
NK-cells is primarily due to the absence of inhibitory
signals. However, some studies indirectly noted contri-
bution of the activating ligands. For example, Frenzel
et  al. found that the preliminary blocking of activat-
ing receptor NKG2D significantly reduces cytotoxicity
of the NK-cells co-cultured with the syngeneic mouse
ESCs [35]. Another study showed that the NK-cells with
knockout of the Klrk1
–/–
gene encoding the NKG2D re-
ceptor lysed a significantly lower percentage of the
ESCs than the wild-type NK-cells [36]. Interestingly,
experiments with blocking antibodies have shown a
role for another activating receptor, DNAM-1, for hu-
man PSCs [33]. High sensitivity of PSCs to NK-cells is
due to two factors simultaneously: low expression of
HLA-I molecules and increased expression of activat-
ing ligands.
As for the in  vivo immune response, NK-cells are
known to limit teratoma formation after subcutaneous
injection of both syngeneic [37] and autologous iPSCs
[38]. These findings suggest that the residual PSCs,
which could potentially remain in the graft among
the differentiated cells, would fail to form teratomas
and would be rejected by the NK-cells. Melendez etal.
reported that NK-cells can act as an internal barrier
during reprogramming in  vitro and in  vivo [39]. It was
demonstrated that NK-cells can recognize and destroy
the partially reprogrammed cells shown to express the
ligands for activating NKG2D and DNAM-1 receptors.
Further, using the transgenic mouse line [40] express-
ing four reprogramming factors from the Yamanaka
cocktail (OSKM) under doxycycline treatment, the au-
thors showed that partial reprogramming in  vivo oc-
curs more efficiently when NK-cells are depleted and,
on the contrary, is significantly reduced when they are
adoptively transferred [39].
Response of the syngeneic and autologous NK-cells
to differentiated iPSC-derivatives needs to be better
understood. For example, increased sensitivity of the
hepatocyte-like cells differentiated from the murine
iPSCs (iPS-HLC) to syngeneic NK-cells in  vitro has been
reported [41]. At the same time, syngeneic somatic
cellshepatocytes– practically did not induce the NK-
cell response. Interestingly, this work also determined
immune response of NK-cells to the hepatocyte-like
cells derived from ESCs (ES-HCs). The authors found
that the lysed ES-HCs percentage was almost twice
as high as that of the lysed iPS-HCs. ES-HCs, but not
iPS-HCs, appeared to have ligands for activating the
NKG2D receptor. In addition, knockout of the NKG2D
receptors in the NK-cells significantly reduced percent-
age of the lysed ES-HSCs but not iPS-HSCs. Thus, this
work confirmed previous findings that elimination of
the murine PSCs and their differentiated derivatives is
mainly due to interaction of the NKG2D receptor with
its ligands [35, 36, 42].
In another work, the NK-cell response to trans-
plantation of cardiomyocytes differentiated from the
syngeneic iPSCs (miPSC-CMs) was studied in a mouse
model [14]. It was shown that survival rate of the sub-
cutaneously transplanted miPSC-CMs was significant-
ly higher in the NK-cell-depleted mice. In the control
mice, in addition to infiltration of the graft by NK-cells,
there were signs of the NK-cell degranulation and re-
jection of the miPSC-CMs. Analysis of the NK-cell li-
gand expression showed that the miPSC-CMs weakly
expressed the MHC class I molecules and were stained
with antibodies to the ligands for NKG2D and DNAM-1
receptors. Blocking of the NKG2D and DNAM-1 recep-
tors or increasing the MHC-I expression by the IFNγ
(interferon gamma) pretreatment mitigated cytotoxic
properties of the NK-cells in  vitro. Also, it decreased
the NK-cell infiltration into the transplanted areas and
necrosis of miPSC-CMs in  vivo.
At the same time, immunogenicity of the human
iPSC-derivatives to NK-cells is poorly investigated.
It was studied mainly upon engineering of the im-
mune-evasive or “universal” PSCs. This approach is
believed to be an alternative to the traditional immu-
nosuppressive therapy, since derivatives of such cells
will be suitable for any recipient [43, 44]. The most
commonly used strategy to create hypoimmunogenic
cells is to completely “turn off” expression of HLA mole-
cules, both classes  I and II. To suppress the HLA class  I
expression, the beta-2-microglobulin (B2M) gene, which
encodes the light subunit required for stable heterodi-
mer formation, is usually knocked out [45-47]. To sup-
press the HLA class  II expression, the CIITA gene, tran-
scription factor required for the HLA-II expression, is
usually knocked out [48-50]. Cells devoid of HLA mol-
ecules should become completely invisible to the re-
cipient’s T-cells, both CD8
+
and CD4
+
[51]. On the other
hand, elimination of the HLA class  I molecules makes
the PSC-derivatives sensitive to cytotoxic properties
of NK-cells [46, 52, 53]. Therefore, obtaining PSC lines
with the reduced immunogenicity usually involves
two steps: first, HLA expression should be suppressed,
and then additional factors should be added to avoid
NK-cell response [53-57].
A  priori studies of low-immunogenic iPSC-de-
rivatives were performed with an allogeneic model.
Since NK-cells can not recognize foreign molecules,
immune tolerance to the iPSC-derivatives can be eval-
uated using NK-cells of allogeneic origin but with one
stipulation. It is known that the NK-cells alloreactiv-
ity can theoretically be caused by mismatch of the
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 803
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 1. Mechanism of NK-cell alloreactivity exemplified by the ligand mismatch for KIR2DL receptors. HLA-C1/C1 NK-cells are
activated by interaction with HLA-C2/C2 target cells (no inhibitory signal via KIR2DL3-receptor). HLA-C2/C2 NK-cells are acti-
vated by interaction with HLA-C1/C1 target cells (no inhibitory signaling through KIR2DL1 receptor). HLA-C1/C2 NK-cells are
activated by interaction with HLA-C1/C1 target cells (no inhibitory signaling through KIR2DL1-receptor) and by interaction with
HLA-C2/C2 target cells (no inhibitory signaling through KIR2DL3-receptor).
KIR family (killer-cell immunoglobulin-like receptor)
ligands via the mechanism of “missing-self” recogni-
tion. All HLA-C alleles are divided into two groups
HLA-C1 and HLA-C2 – depending on the sequence of
amino acids at positions 77 and 80 of the alpha chain,
which determines their ability to bind to the NK-cell
receptors KIR2DL3 and KIR2DL1, respectively [58].
According to this principle, all donors and recipients
can be categorized into the following groups: HLA-C1/
C1, HLA-C1/C2, and HLA-C2/C2. NK-cells, in the process
of licensing or “learning” during maturation, acquire
tolerance to the specific set of HLA-C alleles on their
own cells. The conditions of NK-cell response to HLA-C
allele mismatch by interaction with the KIR2DL recep-
tors are presented in Fig.  1. Thus, if derivatives dif-
ferentiated from the HLA-homozygous iPSC lines are
used as target cells, heterozygous HLA-C1/C2 NK-cells
will respond to the absence of any of the KIR-ligands.
It was confirmed in the work by Ichise et al. [59].
The authors showed [59] that the HLA-C1/C2 NK-cells
isolated from the blood of healthy donors lysed T-cells
and endothelial cells differentiated from the HLA-C1/
C1 iPSCs. In turn, ectopic expression of HLA-C2 in the
differentiated HLA-C1/C1 derivatives diminished the
NK-cell response. In addition to alloreactivity to the
mismatched ligands of the KIR2DL receptors, the NK-
cell responses to the absence of ligands for KIR3DL1
(epitope Bw4) and KIR3DL2 (HLA-A3, A11 alleles) re-
ceptors have also been reported [60]. Thus, the alloge-
neic model can be used to assess immunogenicity of
the iPSCs derivatives toward NK-cells; however, only
under condition that the donors participating in the
study are typed.
Interestingly, in some studies with hypoimmuno-
genic PSC-derivatives, no significant difference was
found in the response of NK-cells to PSC knockout
derivatives and wild-type PSC-derivatives. However,
it should be noted that in all these studies, the target
cells and the donors involved were not typed. Dif-
ferences in cytotoxicity and number of CD107a
+
, i.e.,
degranulated NK-cells cocultured with the wild-type
SMCs or SMCs not expressing HLA classI, were statisti-
cally insignificant [55]. In turn, the wild-type RPE cells
induced extremely high NK-cell cytotoxicity, compa-
rable to the knockout PSC-derivatives [50]. Moreover,
percentage of the degranulated NK-cells also did not
differ between the wild-type and knockout RPE cells,
although it varied greatly depending on the donor.
High cytotoxicity of NK-cells was also observed against
the cardiomyocytes differentiated from ESCs [57].
BOGOMIAKOVA et al.804
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Endothelial cells induced the same level of NK-cell
degranulation regardless of the HLA class I expres-
sion, although NK-cell cytotoxicity was higher against
the ESC-derivatives with the B2M gene knockout [61].
Itshould be noted that in the studies mentioned above,
the authors did not focus on the NK-cell response to
the wild-type PSC-derivatives. Instead, they described
absence of hypersensitivity of the HLA-negative cells
to the NK-cells.
In our recent work, we also found that the fibro-
blast-like iPSC-derivatives with the B2M gene knock-
out (ΔiPS-fibro) show the same degree of sensitivity
to allogeneic and autologous NK-cells as the wild-type
fibroblast-like cells – iPS-fibro [15]. Unlike other au-
thors, we used parental fibroblasts as a negative con-
trol for the NK-cell reaction. This comparison enabled
us to detect absence of complete immunologic tol-
erance to the differentiated iPSC-derivatives from
the autologous NK-cells. Transcriptome analysis re-
vealed a significant imbalance of the NK-cell ligands
in the iPS- fibro. Compared to the parental fibroblasts,
iPS- fibro simultaneously showed significant decrease
in the expression of HLA-I molecules and increase in
the expression of ligands for activating DNAM-1 and
NKG2D receptors. Further in this work, it was shown
that the NK-cell ligands in the differentiated iPSC-de-
rivatives can be balanced by pretreatment of the cell
cultures with IFNγ [15].
Another study noted sensitivity of the renal iPSC-
derivatives to the autologous NK-cells [28]. The au-
thors found that percentage of the activated NK-cells
cocultured with the proximal epithelial cell precursors
was lower than with the more “mature” iPSC-deriva-
tives. However, these differences were not discussed in
detail in the article. According to the RNA-sequencing
data, the level of HLA class  I transcripts increased in
the proximal epithelial cells during prolonged cultur-
ing. These data are in agreement with the results ob-
tained with iPS-fibro [15] as well as with some activat-
ing NK-cell ligands, particularly MICA and NECTIN2.
Itexplains predominance of the activating signals and
initiation of cytotoxic program in the NK-cells. It is also
interesting to note that, in contrast to the earlier work
[33], Rossbach etal. [28] did not observe high activity
of NK-cells against the undifferentiated human iPSCs.
It should be noted that the role of NK-cells in the
solid organ transplantation remains quite controver-
sial [60,  62]. There is evidence that some subsets of
NK-cells may play a role in regulation of allograft tol-
erance, and that NK-cells are, nevertheless, involved
in the T-cell-mediated and antibody-mediated allograft
rejection [63]. Without immunosuppressive therapy,
which affects cytotoxic activity and adjusts degranu-
lation properties, the activated NK-cells produce IFNγ
that could contribute to the development of chronic in-
flammation and enhance the predominantly T-cell-me-
diated immune response [64]. Thus, the iPSC-based cell
therapies should also consider immunogenicity of the
iPSC-derivatives toward NK-cells.
POSSIBLE REASONS OF THE IMMUNE RESPONSE
TO AUTOLOGOUS IPSC DERIVATIVES
It is still unclear what the crucial factor for some-
times-observed immunogenicity of the autologous iPSC-
derivatives is. Generally, the T-cell response can be
explained by formation of neoantigens and aberrant
gene expression (Fig.2a), and the NK-cell response can
be explained by imbalance between the activating and
inhibitory ligands in the target cells (Fig.2b).
It was initially assumed that using different repro-
gramming vectors could be the reason for iPSC immu-
nogenicity. Even in the first work, it was shown that
the teratomas formed by retroviral iPSCs were more
often rejected in the syngeneic hosts [12]. In turn, us-
ing episomes as a reprogramming vector significantly
reduced percentage of the rejected teratomas. Similar
results were obtained during transplantation of dopa-
minergic neurons into the primate brain [20]. It is well-
known that the retroviral and lentiviral constructs are
predominantly integrated into the transcriptionally ac-
tive sites that could cause mutations, genome instabili-
ty, and chromosomal aberrations. In addition, there is
evidence that the subsequent activation of transgenes
correlates with the aberrant production of the immu-
nogenic protein OCT4 [17].
It is assumed that immunogenicity of the “inte-
gration-free” iPSCs should be lower than of the iPSCs
obtained by retro- and lentiviral transfection. However,
to the best of our knowledge, there are no detailed
studies on this topic. The data comparing genomic in-
stability in the iPSC lines obtained by various repro-
gramming methods are contradictory. Thus, some re-
searchers report a similar number of point mutations
[65], as well as copy number variations (CNVs) [66].
On the contrary, others showed 2-fold lower number
of mutations in the integration-free” iPSCs [67, 68],
which means lower probability of neoepitope forma-
tion. The frequency of genetic variations was also low
in the human iPSCs obtained with episomal constructs
[69]. In the recent years, other methods of reprogram-
ming without integration have been suggested, partic-
ularly involving endogenous pluripotent genes using
the CRISPR/Cas9 system [70]. However, as far as we
know, no additional information about their genomic
instability has been provided. In any case, non-integra-
tive reprogramming methods are currently the safest
and most effective for further clinical use [4].
Commonly, differences in immunogenicity are ex-
plained by mutations and, consequently, by formation
of neoepitopes [71]. First, these may be mutations that
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 805
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 2. Immunogenicity of autologous iPSCs and their derivatives. a)Immune response of T-cells to autologous iPSCs and their
differentiated derivatives could be due to recognition of immunogenic neoepitopes formed as a result of mutations or aberrant
expression of immunogenic genes. b) Immune response of NK-cells to autologous iPSCs and their differentiated derivatives
could be due to imbalance of the NK-cell ligands in the target cells. Predominance of activating signals triggers cytotoxic pro-
gram in NK-cells.
existed in the parental somatic cells. Thus, fibroblasts
are one of the most frequently used sources of cells
for reprogramming [72]. It has been reported that mu-
tations acquired due to UV-induced mutagenesis are
present in ~50% of human iPSCs reprogrammed from
the skin fibroblasts [73]. Such mutations are charac-
terized by the C-to-T or CC-to-TT substitutions and are
often observed in melanomas [74]. Other somatic cells
can act as an alternative to fibroblasts. For example, it
has been reported that the iPSCs obtained from hema-
topoietic stem cells contain significantly fewer point
mutations, insertions, and deletions than the iPSCs
obtained from the skin fibroblasts  [75]. Peripheral
blood cells are also very often used as a source of cells
for reprogramming. Rouhani et al. revealed that the
blood-derived iPSCs contained fewer mutations than
the fibroblast-derived iPSCs  [76]. At the same time,
there is evidence that mutations in the blood cells
also accumulate with age [77]. Thus, the work using 16
iPSC lines obtained from the blood cells of donors aged
21-100 years demonstrated that the frequency of muta-
tions in the iPSC increases linearly with the donor age
[78]. In addition, frequency of the mitochondrial DNA
mutations in human iPSCs also increases with the do-
nor age, and this can lead to metabolic defects in iPSCs
[79]. Although denovo mutations in the mitochondrial
DNA are usually rare for iPSCs [80, 81], they can lead
to formation of immunogenic neoepitopes and pro-
voke immune response even during the autologous
transplantation, as was shown earlier [82].
Thus, somatic cells of the young donors may have
a comparative advantage for deriving iPSCs. This is
also confirmed by the results of recent work, where
iPSCs were obtained from the umbilical cord blood
erythroblasts and did not contain mutations in the
protein-coding regions [83]. In addition, age of the do-
nors could affect cultural properties of iPSCs. Thus,
it was shown that the iPSCs obtained from the older
mice proliferated not as good as the iPSCs obtained
from the young mice [84].
Mutations in iPSCs could also occur during re-
programming. Such mutations are usually similar to
the mutations caused by oxidative stress (C-to-A sub-
stitutions) and are predominantly found in the lami-
na-associated domains – condensed regions of heter-
ochromatin positioned at the nuclear periphery [85].
BOGOMIAKOVA et al.806
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Earlier studies showed that up to 75% of point muta-
tions in iPSCs occur during reprogramming [86, 87].
Presence of various mutations in the isogenic iPSC
clones and lower frequency of point mutations in the
isogenic ESC presumably indicates that such mutations
were not obtained from the parental somatic cells [67].
Additional information on the mutations that occur
during reprogramming was provided by Rouhani etal.
[88]. The authors identified unique mutations in the
isogenic iPSC lines derived from the monoclonal-origin
endothelial precursors.
It is believed that mutations occur at the earliest
stages of reprogramming before the first cell division
after the introduction of reprogramming factors or
immediately after the first or second division [67,  83].
The authors of the latest work also found that tempo-
rary deficit of the control components of the G1/S cell
cycle at the initial stage of the reprogramming process
leads to accumulation of mutations [83]. At the same
time, the data on mutations that occur during repro-
gramming are pretty ambiguous. Recent studies have
shown that up to 90% of the various SNPs and indels in
iPSCs originate from somatic cells [73,  89]. Moreover,
Kosanke et al. showed that only 2% of the mutations
detected in iPSCs were not detected in the parental en-
dothelial cells used for reprogramming [90].
The third reason for mutations in iPSCs is long-
term cultivation. Such mutations are formed stochasti-
cally and are much less common than the pre-existing
somatic mutations or reprogramming-induced muta-
tions. Mutations caused by prolonged cultivation are
believed to provide proliferative benefits [91, 92]. For
example, one iPSCs line carried four additional point
mutations in the late passages compared with the cells
of early passages [86]. Nevertheless, according to the
recent data, frequency and spectrum of the mutations
induced by long-term cultivation of iPSCs do not differ
from the mutations occurring at the pre-gastrulation
stage of embryogenesis [93]. Another study showed
that the rate of accumulated mutations in the long-
term cultured iPSCs is lower than in the intestinal and
liver stem cells [94]. The authors found that more than
a third of mutations were caused by the C-to-A substi-
tutions associated with oxidative stress. Cell cultiva-
tion under hypoxic conditions (3% oxygen) for three
months significantly reduced the number of single
nucleotide substitutions. Similar results were obtained
for the ESCs: the authors observed a more than two-
fold decrease in the frequency of mutations under hy-
poxia [95]. The obtained data can be used to optimize
conditions of IPSC cultivation.
For clinical use, it is crucial to understand how
mutations can affect the iPSC phenotype, including
whether they can trigger carcinogenesis. There are
studies indicating that point mutations are predom-
inantly found in the cancer-associated genes [86].
Repeated mutations in the TP53 tumor suppressor
gene were detected both by the whole exome sequenc-
ing of ESC and by analyzing the publicly available RNA
sequencing data from 120 PSC lines [92]. In addition to
mutations in the TP53 gene, repeated mutations were
found in other tumor-associated genes, such as CDK12,
EGFR, and PATZ1 [96,  97]. A recent study revealed mu-
tations in the BCOR gene, often found in hematological
diseases, in more than 25% of the analyzed iPSC lines
[76]. In contrast, no association with the tumor-associ-
ated genes has been found in other studies [65, 69, 73,
94]. Point mutations were mostly found in the areas
of inactive chromatin, so they were unlikely to cause
undesirable effects. However, unique mutations in var-
ious isogenic iPSC clones were usually found in active
promoters and could alter gene expression [73]. This,
in turn, could lead to formation of immunogenic deter-
minants or affect effectiveness of differentiation into
the desired cell type [98], which is also essential for re-
generative medicine.
In addition to mutations induced by reprogram-
ming or cultivation, epigenetic changes that regulate
expression of various proteins should also be consid-
ered. First of all, it applies to disruption in DNA meth-
ylation in the PSC lines. Some studies noted that ab-
errant methylation pattern in iPSCs may be similar to
the tumor cells [99-101]. Moreover, it was shown that
methylation deregulation can persist in differentiated
cells [102]. However, it is worth noting that DNA meth-
ylation in the PSCs can be dynamic, respond to culture
conditions, and vary depending on the cell line [95].
It is also believed that some cells are not fully
undergoing the reprogramming process, and iPSCs
could to a large extent retain transcriptional and epi-
genetic memory of their origin [103]. Nevertheless, re-
sults of the studies on this topic are quite
contradictory,
but according to the modern concepts, molecular and
functional differences in different iPSC lines are lost
during prolonged cultivation [104]. At the same time,
it was shown in the recent study that reprogramming
through the stage of naive iPSCs (TNT-reprogramming)
completely erases epigenetic memory and corrects
epigenetic aberrations that have arisen [105]. Such
TNT-iPSCs turned out to be more similar to ESCs from
the molecular and functional point of view than the
iPSCs obtained by the standard method.
Epigenetic peculiarities could explain abnormal
expression of immunogenic proteins. Thus, at least two
studies have demonstrated that the “somatic memory”
phenomenon could influence immunogenicity of the
iPSCs [106,  107]. Mouse iPSCs obtained from the Sertoli
cells, anatomically related to the immune-privileged
regions, formed teratomas more efficiently than the
iPSCs obtained from the embryonic fibroblasts [106].
Moreover, differentiated derivatives of the syngene-
ic ESCs demonstrated a reduced in  vitro activation
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 807
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
of allogeneic T-cells compared to the iPSCs obtained
from the embryonic fibroblasts. However, it is worth
noting that in the later passages, the authors did not
observe differences in immunogenicity of the iPSCs
obtained from different somatic cells. These results
confirm that “somatic memory” in the iPSCs may be
present only in early passages [106]. Another study
showed that the umbilical cord mesenchymal cells are
a less immunogenic source of cells for reprogramming
than the skin fibroblasts [107].
Impaired expression of the genes associated with
the NK-cell response is another reason for immunoge-
nicity of iPSCs and their derivatives [15]. Both increase
in the signals from activating receptors and decrease
in the signals from inhibitory receptors can trigger
cytotoxic program of NK-cells. In other words, proper
balance between the inhibitory and activating ligands
could make the target cell invisible to NK-cells [108].
In contrast, an impaired balance of the NK-cell ligands
in the iPSC-derivatives could cause excessive activa-
tion of NK-cells. Thus, intensity of the HLA class  I mol-
ecule expression and activating ligands and adhesion
molecules would influence the degree of immune re-
sponse. Previously, we showed that all these factors
were responsible for the increased NK-cell response
to iPSC-derivatives [15]. First, we observed a relatively
low gene expression of the HLA-I molecules, major in-
hibitory ligands in the fibroblast-like iPSC-derivatives
(iPS-fibro). Second, the genes coding for the main ac-
tivating NK-cell ligands were upregulated in the iPS-
fibro. Expression of the stress-induced molecule MICA
(NKG2D ligand) gene was more than 1.5 times higher
in the iPS-fibro than in their parental fibroblasts. The
DNAM-1 ligands, NECTIN2 (CD112) and PVR (CD155),
and the NKp30 ligand, NCR3LG1 (B7-H6), underwent
a more noticeable increase in the gene expression
with more than 3-fold-change in the iPS-fibro. Third,
the genes of some adhesion molecules were also over-
expressed in iPS-fibro. Interaction of the adhesion
molecules with their receptors on NK-cells facilitates
formation of tight junctions between the NK-cell and
the target cell and leads to assembly of immunologi-
cal synapses essential for the target cell killing [109].
The ICAM-1 (LFA-1 ligand) and VCAM-1 (VLA-4 or α4β1
integrin ligand) genes were upregulated in the iPSC-
derivatives. Hence, imbalance between the NK-cell
ligands in iPSC-derivatives was determined simultane-
ously by low intensity of the inhibitory signals and ele-
vated intensity of the activating signals [15].
Vulnerability to the action of NK-cells can be ex-
plained by insufficient maturity of the differentiated
iPSC-derivatives and low level of the HLA-I class mol-
ecules compared to the parental somatic cells. Thus,
increase in the HLA-I expression was shown during
prolonged cultivation or passaging, at least for the RPE
cells [50], proximal renal epithelium cells [28], and
iPS-fibro [15]. Another risk of immature phenotype is
expression of embryonic or fetal proteins, which are
also typical for some cancers (for example, alpha-fe-
toprotein) [110]. Despite the active development of
differentiation protocols, several cell types can be dif-
ferentiated in  vitro only to an immature phenotype, in
particular, cardiomyocytes [111], hepatocytes [112], or
beta-cells [113].
Increased expression of the activating NK-cell li-
gands is worth noting separately. Analysis of the pub-
licly available RNA-seq datasets [114-116] showed that
expression of the NECTIN2, PVR, CADM1, and CD70
genes was upregulated in the independently derived
fibroblast-like cells compared to the isogenic fibro-
blasts used for reprogramming [15]. Imperfect micro-
environment during in  vitro differentiation may affect
proper balance between the ligands for the NK-cell
receptors in this type of iPSC-derivatives. In addition,
high levels of the MICA and NECTIN2 gene expression
were observed in the proximal epithelial cells of the
kidney [28]. Since each cell type expresses its own set
of proteins, it will be necessary to determine expres-
sion pattern of the ligands of the NK-cell receptors for
clinical use. It is worth noting that the cells that belong
to immune-privileged tissues could have immunomod-
ulatory functions to suppress immune response. It was
shown that some types of the differentiated PSC-deriv-
atives, in particular RPE cells [117,  118], retinal gan-
glion cells [119], neuron precursors [120-122], neural
crest cells [123, 124], and chondrocytes [125] demon-
strate reduced immunogenicity even to allogeneic lym-
phocytes.
Different cultivation conditions could affect im-
munogenicity of iPSCs and their derivatives. As men-
tioned earlier, prolonged cultivation could lead to ac-
cumulation of mutations in the cells at later passages
[86,  94]. The cryo-pause method, i.e., storing iPSCs as
ready-to-use aliquots from one passage, can reduce
frequency of genomic aberrations caused by passaging
and prolonged cultivation of iPSCs [126]. Considering
that oxidative process during reprogramming and pro-
longed cultivation could lead to C-to-A substitutions
[85, 88, 94], antioxidants could reduce mutagenic load
in the iPSCs. In particular, antioxidants were report-
ed to reduce CNVs in the iPSCs [127]. A recent study
also indicated that introduction of antioxidant trans-
genes, such as superoxide dismutase  1 (SOD1) and 2
(SOD2), glutathione peroxidase  1 (GPX1), and N-acetyl-
cysteine (NAC), reduced the number of transversions
iniPSCs[83].
Selection of the reagents used for cultivation and
differentiation could affect immunogenic properties
of iPSCs and their derivatives. Using xenogeneic
materials for PSC cultivation could complicate fur-
ther clinical use of the PSC-derivatives. For example,
ESCs and embryoid bodies were shown to absorb
BOGOMIAKOVA et al.808
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
Fig. 3. Potential reasons of impaired immune tolerance to the autologous iPSCs and their differentiated derivatives.
N- glycolylneuraminic acid (Neu5Gc) from the feeder
layers of embryonic fibroblasts and from nutrient me-
dia containing animal serum [128]. This poses a sig-
nificant risk since antibodies to Neu5Gc circulate in
human blood [129]. Currently, PSCs are usually culti-
vated under feeder-free conditions. In addition, repro-
gramming and differentiation protocols have been de-
veloped without using animal components (xeno-free),
in which the amount of Neu5Gc is reduced or it is
completely absent. However, these methods are more
expensive [130,  131]. On the other hand, commer-
cial xeno-free media may contain elevated levels of
ascorbate, which could affect methylation of the CD30
promoter, a marker of malignant neoplasms [132].
However, it is worth noting that CD30 more likely is
the marker of undifferentiated cells than the marker
of transformed cells [133]. In any case, the risk is not
limited to animal products. For new formulations of
nutrient media, it is necessary to determine their bio-
logical effects on the cultured cells, including balance
of the NK-cell ligands.
Hence, several factors could potentially affect
immunogenicity of the final cellular product obtained
from PSCs (Fig.  3). Systematization of the possible rea-
sons listed above should contribute to the develop-
ment of quality criteria to ensure safety of the PSC-
based therapy in clinical practice. Since each type of
the differentiated cells expresses different genes and
proteins, we assume that screening of each cell type
for immunogenicity would be necessary for subse-
quent clinical use [134].
CONCLUSION
Accumulating data on the lack of complete im-
mune tolerance to derivatives of autologous iPSCs [13-
15] raises concerns about their transplantation without
immunosuppression. Nevertheless, the most eloquent
response to these concerns are the results of ongoing
clinical trials. According to the https://clinicaltrials.gov,
about 20 cell products based on autologous iPSCs are
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 809
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
undergoing clinical trials. Primary results of the three
of them have been published [135-137]. In the first two
cases, RPE cells were transplanted to treat age-relat-
ed macular degeneration; in the third, dopaminergic
progenitor cells were transplanted to treat Parkinson’s
disease. Immunosuppressive therapy was not used in
any transplantation, but no side effects were report-
ed. It should be noted that in all cases transplantation
of homogeneous cultures of the iPSC-derivatives was
carried out into the immune-privileged organs – eye
and brain. It remains to be studied whether the unde-
sirable immune reactions would occur during trans-
plantation of autologous iPSC-derivatives, including
complex cellular products, into the organs and tissues
deprived of immune-privileged status. More recently,
there has been a report on the absence of side effects
in the case of transfusion of the platelets differentiated
from autologous iPSCs [138].
Impaired immune tolerance and high cost and
time necessary for deriving a new iPSC line currently
do not allow us to consider personalized therapy as a
promising tool for broad medical practice. In this re-
gard, allogeneic derivatives of PSCs are currently the
preferred source for regenerative medicine. In the
recent years, it was suggested that derivation of the
“universal” PSCs could solve the problem of histocom-
patibility and prevent immune rejection since their de-
rivatives would be suitable for any recipient [43, 44,
51]. As mentioned earlier, various “immune masking”
strategies are used to obtain such iPSCs, from elimi-
nation of the HLA molecules to inhibit T-lymphocytes
[45-50] to introduction of the immunomodulatory fac-
tors to suppress NK-cells [53-57]. It was shown that the
differentiated derivatives of the modified iPSCs with
reduced immunogenicity demonstrate long-term sur-
vival in the fully immunocompetent animals: 50 days
in the mouse model [56] and 40 weeks in rhesus mon-
keys [139]. In both studies, “blinding” of the allogeneic
immune system was achieved by blocking expression
of the HLA classI andII by knocking out the B2M and
CIITA genes and introducing the CD47 transgene as
an immune checkpoint to NK-cells [140]. In the recent
study, simultaneous introduction of 8 immunomodula-
tory factors (Pdl1, Cd200, Cd47, H2-M3, Fasl, Serpinb9,
Ccl21, and Mfge8) to the mouse ESC ensured long-term
survival of teratomas in the allogeneic model [141].
Insome groups of animals, the observation period was
nine months. In February 2022, ViaCyte and CRISPR
Therapeutics announced launch of the PhaseI trials of
VCTX210, ESC-based therapy for type 1 diabetes with-
out the need for immunosuppression. The used CyT49
line has a B2M gene knockout and expresses the CD274
transgene encoding immunological checkpoint PD-L1
for additional protection against the T-cell attack [142].
Despite attractiveness of the “universal” approach,
the problems associated with safety of such therapy
are worth noting [43, 143, 144]. First, they relate to
immune evasion, which means increase in the risks
of tumor transformation of such cells. By itself, ab-
sence of the MHC/HLA molecules should not contrib-
ute to oncogenesis [145]. Therefore, the probability
of malignant degeneration is unlikely to exceed that
in the adult tissues. However, it is evident that in the
HLA-negative cells, it would be more challenging to
eliminate it using conventional immune mechanisms.
It was proposed to solve this problem by introduc-
ing “suicide cassettes” that can be activated in the
case of malignant transformation or viral infection
of the graft [43, 51]. The herpes simplex virus 1 thy-
midine kinase (HSV-TK) gene can be introduced under
the promoter of both pluripotent genes [146] and the
genes that play a key role in regulation of the cell cy-
cle, such as the CDK1 gene [147]. The latter approach
has recently been applied to the immunomodified hu-
man ESCs [141]. Another approach is a suicidal system
based on the inducible caspase-9 (iCas9) [148]. Taking
into account this issue, the current Viacyte clinical tri-
al is safer since a semipermeable membrane encap-
sulates beta-cells and cannot exit the capsule in the
case of degeneration [149]. In any case, it is evident
that for safety reasons it is necessary to thoroughly
investigate “universal” lines for potential oncogenic
mutations and activating proto-oncogenes [51]. Still,
despite the unresolved issues, development of the ge-
netically modified pluripotent stem cells can contrib-
ute to the large-scale production of the “off-the-shelf
cell products and solve the problem of tissue and
organ shortage.
Acknowledgments. M.A.L. is a member of the
Interdisciplinary Scientific and Educational School of
Moscow University “Molecular Technologies of the Liv-
ing Systems and Synthetic Biology.”
Contributions. M.E.B. conducted background re-
search, wrote the manuscript, and created the figures;
A.N.B. wrote and edited the manuscript; M.A.L. provid-
ed the concept of this review and edited the final man-
uscript.
Funding. The study was performed with financial
support of the State Assignment no.123032900030-7.
Ethics declarations. This work does not contain
any studies involving human and animal subjects.
Theauthors of this work declare that they have nocon-
flicts of interest.
REFERENCES
1. Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S. S., Wak-
nitz, M.A., Swiergiel, J.J., etal. (1998) Embryonic stem
cell lines derived from human blastocysts, Science,
282, 1145-1147, doi:10.1126/science.282.5391.1145.
BOGOMIAKOVA et al.810
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
2. Takahashi, K., and Yamanaka, S. (2006) Induction of
pluripotent stem cells from mouse embryonic and
adult fibroblast cultures by defined factors, Cell, 126,
663-676, doi:10.1016/j.cell.2006.07.024.
3. Lebedeva, O.S., and Lagarkova, M.A. (2018) Pluripo-
tent stem cells for modelling and cell therapy of Par-
kinson’s disease, Biochemistry (Moscow), 83, 1318-
1330, doi:10.1134/S0006297918090067.
4. Doss, M.X., and Sachinidis,A. (2019) Current challeng-
es of iPSC-based disease modeling and therapeutic im-
plications, Cells, 8, 403, doi:10.3390/cells8050403.
5. Bogomazova, A.N., Vassina, E.M., Kiselev, S.I., Lagar-
kova, M.A., Lebedeva, O.S., etal. (2015) Genetic cell
reprogramming: a new technology for basic research
and applied usage, Genetika, 51, 466-478, doi:10.7868/
S0016675815040025.
6. Huang, C. Y., Liu, C. L., Ting, C. Y., Chiu, Y. T., Cheng,
Y.C., et al. (2019) Human iPSC banking: barriers and
opportunities, J. Biomed. Sci., 26, 87, doi: 10.1186/
s12929-019-0578-x.
7. Rehakova,D., Souralova,T., and Koutna,I. (2020) Clin-
ical-grade human pluripotent stem cells for cell thera-
py: characterization strategy, Int.J. Mol. Sci., 21, 2435,
doi:10.3390/ijms21072435.
8. Sullivan, S., Stacey, G. N., Akazawa, C., Aoyama, N.,
Baptista, R., et al. (2018) Quality control guidelines
for clinical-grade human induced pluripotent stem
cell lines, Regen. Med., 13, 859-866, doi: 10.2217/rme-
2018-0095.
9. Kobold,S., Guhr,A., Mah,N., Bultjer,N., Seltmann,S.,
etal. (2020) A manually curated database on clinical
studies involving cell products derived from human
pluripotent stem cells, Stem Cell Rep., 15, 546-555,
doi:10.1016/j.stemcr.2020.06.014.
10. Bolton, E.M., and Bradley, J.A. (2015) Avoiding immu-
nological rejection in regenerative medicine, Regen.
Med., 10, 287-304, doi:10.2217/rme.15.11.
11. Barrilleaux, B., and Knoepfler, P. S. (2011) Inducing
iPSCs to escape the dish, Cell Stem Cell, 9, 103-111,
doi:10.1016/j.stem.2011.07.006.
12. Zhao,T., Zhang, Z.N., Rong,Z., and Xu,Y. (2011) Immu-
nogenicity of induced pluripotent stem cells, Nature,
474, 212-215, doi:10.1038/nature10135.
13. Zhao,T., Zhang, Z.N., Westenskow, P.D., Todorova,D.,
Hu,Z., etal. (2015) Humanized mice reveal differen-
tial immunogenicity of cells derived from autologous
induced pluripotent stem cells, Cell Stem Cell, 17,
353-359, doi:10.1016/j.stem.2015.07.021.
14. Nakamura,Y., Miyagawa,S., Yoshida,S., Sasawatari,S.,
Toyofuku, T., et al. (2019) Natural killer cells impede
the engraftment of cardiomyocytes derived from in-
duced pluripotent stem cells in syngeneic mouse mod-
el, Sci. Rep., 9, 10840, doi:10.1038/s41598-019-47134-3.
15. Bogomiakova, M.E., Sekretova, E.K., Anufrieva, K.S.,
Khabarova, P. O., Kazakova, A. N., et al. (2023) iPSC-
derived cells lack immune tolerance to autologous
NK-cells due to imbalance in ligands for activating
and inhibitory NK-cell receptors, Stem Cell Res. Ther.,
14, 77, doi:10.1186/s13287-023-03308-5.
16. Araki, R., Uda, M., Hoki, Y., Sunayama, M., Nakamu-
ra,M., etal. (2013) Negligible immunogenicity of ter-
minally differentiated cells derived from induced plu-
ripotent or embryonic stem cells, Nature, 494, 100-104,
doi:10.1038/nature11807.
17. Dhodapkar, K.M., Feldman,D., Matthews,P., Radfar,S.,
Pickering,R., etal. (2010) Natural immunity to plurip-
otency antigen OCT4 in humans, Proc. Natl. Acad. Sci.
USA, 107, 8718-8723, doi:10.1073/pnas.0915086107.
18. Guha, P., Morgan, J. W., Mostoslavsky, G., Rodrigues,
N.P., and Boyd, A.S. (2013) Lack of immune response
to differentiated cells derived from syngeneic induced
pluripotent stem cells, Cell Stem Cell, 12, 407-412,
doi:10.1016/j.stem.2013.01.006.
19. de Almeida, P. E., Meyer, E. H., Kooreman, N. G.,
Diecke, S., Dey, D., et al. (2014) Transplanted termi-
nally differentiated induced pluripotent stem cells
are accepted by immune mechanisms similar to
self- tolerance, Nat. Commun., 5, 3903, doi: 10.1038/
ncomms4903.
20. Morizane, A., Doi, D., Kikuchi, T., Okita, K., Hotta, A.,
et al. (2013) Direct comparison of autologous and al-
logeneic transplantation of iPSC-derived neural cells
in the brain of a non-human primate, Stem Cell Rep.,
1, 283-292, doi:10.1016/j.stemcr.2013.08.007.
21. Emborg, M. E., Liu, Y., Xi, J., Zhang, X., Yin, Y., et al.
(2013) Induced pluripotent stem cell-derived neu-
ral cells survive and mature in the nonhuman
primate brain, Cell Rep., 3, 646-650, doi: 10.1016/
j.celrep.2013.02.016.
22. Hallett, P. J., Deleidi, M., Astradsson, A., Smith, G. A.,
Cooper,O., etal. (2015) Successful function of autolo-
gous iPSC-derived dopamine neurons following trans-
plantation in a non-human primate model of Parkin-
son’s disease, Cell Stem Cell, 16, 269-274, doi:10.1016/
j.stem.2015.01.018.
23. Kordower, J.H., Chu,Y., Hauser, R.A., Freeman, T.B.,
and Olanow, C.W. (2008) Lewy body-like pathology in
long-term embryonic nigral transplants in Parkinson’s
disease, Nat. Med., 14, 504-506, doi:10.1038/nm1747.
24. Mendez, I., Viñuela, A., Astradsson, A., Mukhida, K.,
Hallett,P., etal. (2008) Dopamine neurons implanted
into people with Parkinson’s disease survive with-
out pathology for 14 years, Nat. Med., 14, 507-509,
doi:10.1038/nm1752.
25. Strnadel,J., Carromeu,C., Bardy,C., Navarro,M., Pla-
toshyn,O., et al. (2018) Survival of syngeneic and al-
logeneic iPSC-derived neural precursors after spinal
grafting in minipigs, Sci. Transl. Med., 10, eaam6651,
doi:10.1126/scitranslmed.aam6651.
26. Lu,M., Peng,L., Ming,X., Wang,X., Cui,A., etal. (2019)
Enhanced wound healing promotion by immune re-
sponse-free monkey autologous iPSCs and exosomes
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 811
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
vs. their allogeneic counterparts, EBioMedicine, 42,
443-457, doi:10.1016/j.ebiom.2019.03.011.
27. Benabdallah, B., Désaulniers-Langevin, C., Goyer,
M.L., Colas,C., Maltais,C., etal. (2021) Myogenic pro-
genitor cells derived from human induced pluripo-
tent stem cell are immune-tolerated in humanized
mice, Stem Cells Transl. Med., 10, 267-277, doi:10.1002/
sctm.19-0452.
28. Rossbach, B., Hariharan, K., Mah, N., Oh, S. J., Volk,
H. D., et al. (2022) Human iPSC-derived renal cells
change their immunogenic properties during matu-
ration: implications for regenerative therapies, Cells,
11, 1328, doi:10.3390/cells11081328.
29. Ljunggren, H. G., and Kärre, K. (1990) In search of
the ‘missing self’: MHC molecules and NK cell rec-
ognition, Immunol. Today, 11, 237-244, doi: 10.1016/
0167-569990097-s.
30. Lanier, L.L. (2005) NK cell recognition, Annu. Rev. Im-
munol., 23, 225-274, doi: 10.1146/annurev.immunol.
23.021704.115526.
31. Smyth, M.J., Cretney, E., Kelly, J.M., Westwood, J. A.,
Street, S. E., et al. (2005) Activation of NK cell cyto-
toxicity, Mol. Immunol., 42, 501-510, doi: 10.1016/
j.molimm.2004.07.034.
32. Dressel, R., Nolte, J., Elsner, L., Novota, P., Guan, K.,
et al. (2010) Pluripotent stem cells are highly sus-
ceptible targets for syngeneic, allogeneic, and xeno-
geneic natural killer cells, FASEB J., 24, 2164-2177,
doi:10.1096/fj.09-134957.
33. Kruse, V., Hamann, C., Monecke, S., Cyganek, L., El-
sner, L., et al. (2015) Human induced pluripotent
stem cells are targets for allogeneic and autologous
natural Killer (NK) cells and killing is partly mediat-
ed by the activating NK receptor DNAM-1, PLoS One,
10, e0125544, doi:10.1371/journal.pone.0125544.
34. Drukker, M., Katz, G., Urbach, A., Schuldiner, M.,
Markel,G., etal. (2002) Characterization of the expres-
sion of MHC proteins in human embryonic stem cells,
Proc. Natl. Acad. Sci. USA, 99, 9864-9869, doi:10.1073/
pnas.142298299.
35. Frenzel, L. P., Abdullah, Z., Kriegeskorte, A. K., Di-
eterich,R., Lange,N., etal. (2009) Role of natural-kill-
er group 2 member D ligands and intercellular adhe-
sion molecule 1 in natural killer cell-mediated lysis
of murine embryonic stem cells and embryonic stem
cell-derived cardiomyocytes, Stem Cells, 27, 307-316,
doi:10.1634/stemcells.2008-0528.
36. Gröschel, C., Hübscher, D., Nolte, J., Monecke, S.,
Sasse,A., etal. (2017) Efficient killing of murine plu-
ripotent stem cells by natural killer (NK) cells requires
activation by cytokines and partly depends on the ac-
tivating NK receptor NKG2D, Front. Immunol., 8, 870,
doi:10.3389/fimmu.2017.00870.
37. Dressel, R., Schindehütte, J., Kuhlmann, T., Elsner, L.,
Novota, P., et al. (2008) The tumorigenicity of mouse
embryonic stem cells and in vitro differentiated neu-
ronal cells is controlled by the recipients’ immune
response, PLoS One, 3, e2622, doi: 10.1371/journal.
pone.0002622.
38. Benabdallah, B., Désaulniers-Langevin, C., Colas, C.,
Li,Y., Rousseau,G., etal. (2019) Natural killer cells pre-
vent the formation of teratomas derived from human
induced pluripotent stem cells, Front. Immunol., 10,
2580, doi:10.3389/fimmu.2019.02580.
39. Melendez, E., Chondronasiou, D., Mosteiro, L.,
Martínez de Villarreal,J., Fernández-Alfara,M., etal.
(2022) Natural killer cells act as an extrinsic barri-
er for in vivo reprogramming, Development, 149,
dev200361, doi:10.1242/dev.200361.
40. Abad,M., Mosteiro,L., Pantoja,C., Cañamero,M., Ray-
on, T., et al. (2013) Reprogramming in vivo produc-
es teratomas and iPS cells with totipotency features,
Nature, 502, 340-345, doi:10.1038/nature12586.
41. Cisneros,T., Dillard, D.W., Qu,X., Arredondo-Guerre-
ro,J., Castro, M., et al. (2019) Differential role of nat-
ural killer group 2D in recognition and cytotoxicity
of hepatocyte-like cells derived from embryonic stem
cells and induced pluripotent stem cells, Am.J. Trans-
plant., 19, 1652-1662, doi:10.1111/ajt.15217.
42. Phillips, L. K., Gould, E. A., Babu, H., Krams, S. M.,
Palmer, T.D., and Martinez, O.M. (2013) Natural kill-
er cell-activating receptor NKG2D mediates innate
immune targeting of allogeneic neural progenitor
cell grafts, Stem Cells, 31, 1829-1839, doi: 10.1002/
stem.1422.
43. Zheng, D., Wang, X., and Xu, R. H. (2016) Concise re-
view: one stone for multiple birds: generating univer-
sally compatible human embryonic stem cells, Stem
Cells, 34, 2269-2275, doi:10.1002/stem.2407.
44. Bogomiakova, M. E., Eremeev, A. V., and Lagarkova,
M.A. (2019) At home among strangers: is it possible to
create hypoimmunogenic pluripotent stem cell lines?
Mol. Biol., 53, 725-740, doi:10.1134/S0026898419050045.
45. Riolobos, L., Hirata, R. K., Turtle, C. J., Wang, P. R.,
Gornalusse, G.G., etal. (2013) HLA engineering of hu-
man pluripotent stem cells, Mol. Ther., 21, 1232-1241,
doi:10.1038/mt.2013.59.
46. Wang, D., Quan, Y., Yan, Q., Morales, J. E., and Wet-
sel, R. A. (2015) Targeted disruption of the β2-micro-
globulin gene minimizes the immunogenicity of hu-
man embryonic stem cells, Stem Cells Transl. Med.,
4, 1234-1245, doi:10.5966/sctm.2015-0049.
47. Bogomiakova, M. E., Sekretova, E. K., Eremeev,
A. V., Shuvalova, L. D., Bobrovsky, P. A., et al. (2021)
Derivation of induced pluripotent stem cells line
(RCPCMi007-A-1) with inactivation of the beta-2-mi-
croglobulin gene by CRISPR/Cas9 genome editing, Stem
Cell Res., 55, 102451, doi:10.1016/j.scr.2021.102451.
48. Chen, H., Li, Y., Lin, X., Cui, D., Cui, C., et al. (2015)
Functional disruption of human leukocyte antigen
II in human embryonic stem cell, Biol. Res., 48, 59,
doi:10.1186/s40659-015-0051-6.
BOGOMIAKOVA et al.812
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
49. Mattapally, S., Pawlik, K. M., Fast, V. G., Zumaque-
ro, E., Lund, F. E., et al. (2018) Human leukocyte an-
tigen class I and II knockout human induced plurip-
otent stem cell-derived cells: universal donor for cell
therapy, J. Am Heart Assoc., 7, e010239, doi: 10.1161/
JAHA.118.010239.
50. Petrus-Reurer, S., Winblad, N., Kumar, P., Gorchs, L.,
Chrobok, M., et al. (2020) Generation of retinal pig-
ment epithelial cells derived from human embryon-
ic stem cells lacking human leukocyte antigen class
I and II, Stem Cell Rep., 14, 648-662, doi: 10.1016/
j.stemcr.2020.02.006.
51. Lanza, R., Russell, D. W., and Nagy, A. (2019) Engi-
neering universal cells that evade immune detection,
Nat. Rev. Immunol., 19, 723-733, doi: 10.1038/s41577-
019-0200-1.
52. Kim, A., Lee, K. G., Kwon, Y., Lee, K. I., Yang, H. M.,
etal. (2021) Off-the-shelf, immune-compatible human
embryonic stem cells generated via CRISPR-mediat-
ed genome editing, Stem Cell Rev Rep, 17, 1053-1067,
doi:10.1007/s12015-020-10113-7.
53. Wang, B., Iriguchi, S., Waseda, M., Ueda, N., Ueda, T.,
et al. (2021) Generation of hypoimmunogenic T cells
from genetically engineered allogeneic human in-
duced pluripotent stem cells, Nat. Biomed. Eng.,
5, 429-440, doi:10.1038/s41551-021-00730-z.
54. Gornalusse, G.G., Hirata, R.K., Funk, S.E., Riolobos,L.,
Lopes, V. S., et al. (2017) HLA-E-expressing pluripo-
tent stem cells escape allogeneic responses and lysis
by NK cells, Nat. Biotechnol., 35, 765-772, doi:10.1038/
nbt.3860.
55. Han, X., Wang,M., Duan, S., Franco, P.J., Kenty, J.H.,
et al. (2019) Generation of hypoimmunogenic hu-
man pluripotent stem cells, Proc. Natl. Acad. Sci. USA,
116, 10441-10446, doi:10.1073/pnas.1902566116.
56. Deuse,T., Hu,X., Gravina,A., Wang,D., Tediashvili,G.,
etal. (2019) Hypoimmunogenic derivatives of induced
pluripotent stem cells evade immune rejection in fully
immunocompetent allogeneic recipients, Nat. Biotech-
nol., 37, 252-258, doi:10.1038/s41587-019-0016-3.
57. Shi,L., Li,W., Liu,Y., Chen,Z., Hui,Y., etal. (2020) Gen-
eration of hypoimmunogenic human pluripotent stem
cells via expression of membrane-bound and secreted
β2m-HLA-G fusion proteins, Stem Cells, 38, 1423-1437,
doi:10.1002/stem.3269.
58. Moffett, A., and Colucci, F. (2015) Co-evolution of NK
receptors and HLA ligands in humans is driven by re-
production, Immunol. Rev., 267, 283-297, doi:10.1111/
imr.12323.
59. Ichise,H., Nagano,S., Maeda,T., Miyazaki,M., Miyaza-
ki,Y., etal. (2017) NK Cell Alloreactivity against KIR-li-
gand-mismatched HLA-haploidentical tissue derived
from HLA haplotype-homozygous iPSCs, Stem Cell
Rep., 9, 853-867, doi:10.1016/j.stemcr.2017.07.020.
60. Zamir, M. R., Shahi, A., Salehi, S., and Amirzargar, A.
(2022) Natural killer cells and killer cell immunoglob-
ulin-like receptors in solid organ transplantation:
Protectors or opponents? Transplant. Rev. (Orlando),
36, 100723, doi:10.1016/j.trre.2022.100723.
61. Song,C., Wang,L., Li,Q., Liao,B., Qiao,W., etal. (2022)
Generation of individualized immunocompatible en-
dothelial cells from HLA-I-matched human pluripo-
tent stem cells, Stem Cell Res. Ther., 13
, 48, doi:10.1186/
s13287-022-02720-7.
62. Villard,J. (2011) The role of natural killer cells in hu-
man solid organ and tissue transplantation, J. Innate
Immun., 3, 395-402, doi:10.1159/000324400.
63. Pontrelli,P., Rascio,F., Castellano,G., Grandaliano,G.,
Gesualdo,L., and Stallone,G. (2020) The role of natural
killer cells in the immune response in kidney trans-
plantation, Front. Immunol., 11, 1454, doi: 10.3389/
fimmu.2020.01454.
64. Adenugba, A. (2017) NK cells in transplantation,
Transplantation, 101, 2262-2264, doi: 10.1097/
TP.0000000000001914.
65. Bhutani,K., Nazor, K.L., Williams,R., Tran,H., Dai,H.,
etal. (2016) Whole-genome mutational burden analy-
sis of three pluripotency induction methods, Nat.
Commun., 7, 10536, doi:10.1038/ncomms10536.
66. Popp, B., Krumbiegel, M., Grosch, J., Sommer, A.,
Uebe, S., et al. (2018) Need for high-resolution ge-
netic analysis in iPSC: results and lessons from the
ForIPS consortium, Sci. Rep., 8, 17201, doi: 10.1038/
s41598-018-35506-0.
67. Sugiura, M., Kasama, Y., Araki, R., Hoki, Y., Sunaya-
ma,M., etal. (2014) Induced pluripotent stem cell gen-
eration-associated point mutations arise during the
initial stages of the conversion of these cells, Stem Cell
Rep., 2, 52-63, doi:10.1016/j.stemcr.2013.11.006.
68. Kang, X., Yu, Q., Huang, Y., Song, B., Chen, Y., et al.
(2015) Effects of integrating and non-integrating re-
programming methods on copy number variation and
genomic stability of human induced pluripotent stem
cells, PLoS One, 10, e0131128, doi: 10.1371/journal.
pone.0131128.
69. Cheng,L., Hansen, N.F., Zhao,L., Du,Y., Zou,C., etal.
(2012) Low incidence of DNA sequence variation in
human induced pluripotent stem cells generated by
nonintegrating plasmid expression, Cell Stem Cell,
10, 337-344, doi:10.1016/j.stem.2012.01.005.
70. Weltner, J., Balboa, D., Katayama, S., Bespalov, M.,
Krjutškov, K., et al. (2018) Human pluripotent repro-
gramming with CRISPR activators, Nat. Commun.,
9, 2643, doi:10.1038/s41467-018-05067-x.
71. Liu, X., Li, W., Fu, X., and Xu, Y. (2017) The immuno-
genicity and immune tolerance of pluripotent stem
cell derivatives, Front. Immunol., 8, 645, doi:10.3389/
fimmu.2017.00645.
72. Hanna, J. H., Saha, K., and Jaenisch,R. (2010) Plurip-
otency and cellular reprogramming: facts, hypothe-
ses, unresolved issues, Cell, 143, 508-525, doi:10.1016/
j.cell.2010.10.008.
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 813
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
73. D’Antonio, M., Benaglio,P., Jakubosky, D., Greenwald,
W. W., and Matsui, H. (2018) Insights into the muta-
tional burden of human induced pluripotent stem
cells from an integrative multi-omics approach, Cell
Rep., 24, 883-894, doi:10.1016/j.celrep.2018.06.091.
74. Martincorena, I., Roshan, A., Gerstung, M., Ellis, P.,
Van Loo, P., et al. (2015) Tumor evolution. High bur-
den and pervasive positive selection of somatic mu-
tations in normal human skin, Science, 348, 880-886,
doi:10.1126/science.aaa6806.
75. Wang,K., Guzman, A.K., Yan,Z., Zhang,S., Hu, M.Y.,
et al. (2019) Ultra-high-frequency reprogramming
of individual long-term hematopoietic stem cells
yields low somatic variant induced pluripotent
stem cells, Cell Rep., 26, 2580-2592.e7, doi: 10.1016/
j.celrep.2019.02.021.
76. Rouhani, F. J., Zou, X., Danecek, P., Badja, C., Amar-
ante, T. D., et al. (2022) Substantial somatic genomic
variation and selection for BCOR mutations in hu-
man induced pluripotent stem cells, Nat. Genet., 54,
1406-1416, doi:10.1038/s41588-022-01147-3.
77. Genovese, G., Kähler, A. K., Handsaker, R. E., Lind-
berg,J., Rose, S.A., etal. (2014) Clonal hematopoiesis
and blood-cancer risk inferred from blood DNA se-
quence, N.Engl. J.Med., 371, 2477-2487, doi: 10.1056/
NEJMoa1409405.
78. Lo Sardo,V., Ferguson,W., Erikson, G.A., Topol, E.J.,
Baldwin, K.K., and Torkamani,A. (2017) Influence of
donor age on induced pluripotent stem cells, Nat. Bio-
technol., 35, 69-74, doi:10.1038/nbt.3749.
79. Kang,E., Wang,X., Tippner-Hedges,R., Ma,H., Folmes,
C.D., etal. (2016) Age-related accumulation of somat-
ic mitochondrial DNA mutations in adult-derived hu-
man iPSCs, Cell Stem Cell, 18, 625-636, doi: 10.1016/
j.stem.2016.02.005.
80. Wei, W., Gaffney, D. J., and Chinnery, P. F. (2021) Cell
reprogramming shapes the mitochondrial DNA land-
scape, Nat. Commun., 12, 5241, doi: 10.1038/s41467-
021-25482-x.
81. Kosanke,M., Davenport,C., Szepes,M., Wiehlmann,L.,
Kohrn,T., etal. (2021) iPSC culture expansion selects
against putatively actionable mutations in the mi-
tochondrial genome, Stem Cell Rep., 16, 2488-2502,
doi:10.1016/j.stemcr.2021.08.016.
82. Deuse, T., Hu, X., Agbor-Enoh, S., Koch, M., Spitzer,
M.H., etal. (2019) De novo mutations in mitochondri-
al DNA of iPSCs produce immunogenic neoepitopes
in mice and humans, Nat Biotechnol., 37, 1137-1144,
doi:10.1038/s41587-019-0227-7.
83. Araki,R., Hoki, Y., Suga, T., Obara,C., Sunayama, M.,
et al. (2020) Genetic aberrations in iPSCs are intro-
duced by a transient G1/S cell cycle checkpoint defi-
ciency, Nat. Commun., 11, 197, doi: 10.1038/s41467-
019-13830-x.
84. Wang, B., Miyagoe-Suzuki, Y., Yada, E., Ito, N., Nishi-
yama,T., etal. (2011) Reprogramming efficiency and
quality of induced pluripotent stem cells (iPSCs) gen-
erated from muscle-derived fibroblasts of mdx mice
at different ages, PLoS Curr., 3, RRN1274, doi:10.1371/
currents.RRN1274.
85. Yoshihara, M., Araki, R., Kasama, Y., Sunayama, M.,
Abe,M., etal. (2017) Hotspots of denovo point muta-
tions in induced pluripotent stem cells, Cell Rep., 21,
308-315, doi:10.1016/j.celrep.2017.09.060.
86. Gore, A., Li, Z., Fung, H. L., Young, J. E., Agarwal, S.,
et al. (2011) Somatic coding mutations in human
induced pluripotent stem cells, Nature, 471, 63-67,
doi:10.1038/nature09805.
87. Ji,J., Ng, S.H., Sharma,V., Neculai,D., Hussein,S., etal.
(2012) Elevated coding mutation rate during the repro-
gramming of human somatic cells into induced plurip-
otent stem cells, Stem Cells, 30, 435-440, doi: 10.1002/
stem.1011.
88. Rouhani, F. J., Nik-Zainal, S., Wuster, A., Li, Y., Con-
te,N., etal. (2016) Mutational history of a human cell
lineage from somatic to induced pluripotent stem
cells, PLoS Genet., 12, e1005932, doi: 10.1371/journal.
pgen.1005932.
89. Kwon, E. M., Connelly, J. P., Hansen, N. F., Donovan,
F. X., Winkler, T., et al. (2017) iPSCs and fibroblast
subclones from the same fibroblast population con-
tain comparable levels of sequence variations, Proc.
Natl. Acad. Sci. USA, 114, 1964-1969, doi: 10.1073/
pnas.1616035114.
90. Kosanke, M., Osetek, K., Haase, A., Wiehlmann, L.,
Davenport, C., et al. (2021) Reprogramming enrich-
es for somatic cell clones with small-scale mutations
in cancer-associated genes, Mol. Ther., 29, 2535-2553,
doi:10.1016/j.ymthe.2021.04.007.
91. International Stem Cell Initiative, Amps, K., An-
drews, P.W., Anyfantis,G., Armstrong,L., etal. (2011)
Screening ethnically diverse human embryonic stem
cells identifies a chromosome 20 minimal amplicon
conferring growth advantage, Nat. Biotechnol., 29,
1132-1144, doi:10.1038/nbt.2051.
92. Merkle, F.T., Ghosh,S., Kamitaki,N., Mitchell,J., etal.
(2017) Human pluripotent stem cells recurrently ac-
quire and expand dominant negative P53 mutations,
Nature, 545, 229-233, doi:10.1038/nature22312.
93. Hasaart, K. A. L., Manders, F., Ubels, J., Verheul, M.,
van Roosmalen, M. J., et al. (2022) Human induced
pluripotent stem cells display a similar mutation bur-
den as embryonic pluripotent cells in vivo, iScience,
25, 103736, doi:10.1016/j.isci.2022.103736.
94. Kuijk, E., Jager, M., van der Roest, B., Locati, M. D.,
Van Hoeck, A., et al. (2020) The mutational impact
of culturing human pluripotent and adult stem
cells, Nat. Commun., 11, 2493, doi: 10.1038/s41467-
020-16323-4.
95. Thompson, O., von Meyenn, F., Hewitt, Z., Alexan-
der, J., Wood, A., et al. (2020) Low rates of mutation
in clinical grade human pluripotent stem cells under
BOGOMIAKOVA et al.814
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
different culture conditions, Nat. Commun., 11, 1528,
doi:10.1038/s41467-020-15271-3.
96. Avior,Y., Lezmi,E., Eggan,K., and Benvenisty,N. (2021)
Cancer-related mutations identified in primed hu-
man pluripotent stem cells, Cell Stem Cell, 28, 10-11,
doi:10.1016/j.stem.2020.11.013.
97. Merkle, F.T., Ghosh,S., Genovese,G., Handsaker, R.E.,
Kashin,S., etal. (2022) Whole-genome analysis of hu-
man embryonic stem cells enables rational line se-
lection based on genetic variation, Cell Stem Cell, 29,
472-486.e7, doi:10.1016/j.stem.2022.01.011.
98. Puigdevall,P., Jerber,J., Danecek,P., Castellano,S., and
Kilpinen,H. (2023) Somatic mutations alter the differ-
entiation outcomes of iPSC-derived neurons, Cell Gen-
om., 3, 100280, doi:10.1016/j.xgen.2023.100280.
99. Ohm, J. E., Mali, P., Van Neste, L., Berman, D. M.,
Liang, L., et al. (2010) Cancer-related epigenome
changes associated with reprogramming to induced
pluripotent stem cells, Cancer Res., 70, 7662-7673,
doi:10.1158/0008-5472.CAN-10-1361.
100. Planello, A.C., Ji,J., Sharma,V., Singhania,R., Mbabaa-
li, F., et al. (2014) Aberrant DNA methylation repro-
gramming during induced pluripotent stem cell gen-
eration is dependent on the choice of reprogramming
factors, Cell Regen., 3, 4, doi:10.1186/2045-9769-3-4.
101. Weissbein, U., Plotnik, O., Vershkov, D., and Ben-
venisty, N. (2017) Culture-induced recurrent epigen-
etic aberrations in human pluripotent stem cells,
PLoS Genet., 13, e1006979, doi: 10.1371/journal.pgen.
1006979.
102. Ruiz,S., Diep,D., Gore,A., Panopoulos, A.D., Montser-
rat,N., et al. (2012) Identification of a specific repro-
gramming-associated epigenetic signature in human
induced pluripotent stem cells, Proc. Natl. Acad. Sci.
USA, 109, 16196-16201, doi:10.1073/pnas.1202352109.
103. Chin, M.H., Mason, M.J., Xie,W., Volinia,S., Singer,M.,
et al. (2009) Induced pluripotent stem cells and em-
bryonic stem cells are distinguished by gene expres-
sion signatures, Cell Stem Cell, 5, 111-123, doi:10.1016/
j.stem.2009.06.008.
104. Poetsch, M.S., Strano,A., and Guan,K. (2022) Human
induced pluripotent stem cells: from cell origin, ge-
nomic stability, and epigenetic memory to transla-
tional medicine, Stem Cells, 40, 546-555, doi:10.1093/
stmcls/sxac020.
105. Buckberry,S., Liu,X., Poppe,D., Tan, J.P., Sun,G., etal.
(2023) Transient naive reprogramming corrects hiPS
cells functionally and epigenetically, Nature, 620,
863-872, doi:10.1038/s41586-023-06424-7.
106. Wang,X., Qin,J., Zhao, R.C., and Zenke,M. (2014) Re-
duced immunogenicity of induced pluripotent stem
cells derived from Sertoli cells, PLoS One, 9, e106110,
doi:10.1371/journal.pone.0106110.
107. Liu,P., Chen,S., Li,X., Qin,L., Huang,K., etal. (2013)
Low immunogenicity of neural progenitor cells differ-
entiated from induced pluripotent stem cells derived
from less immunogenic somatic cells, PLoS One, 8,
e69617, doi:10.1371/journal.pone.0069617.
108. Pegram, H.J., Andrews, D.M., Smyth, M.J., Darcy, P.K.,
and Kershaw, M. H. (2011) Activating and inhibitory
receptors of natural killer cells, Immunol. Cell Biol.,
89, 216-224, doi:10.1038/icb.2010.78.
109. Netter, P., Anft, M., and Watzl, C. (2017) Termination
of the activating NK Cell immunological synapse is an
active and regulated process, J. Immunol., 199, 2528-
2535, doi:10.4049/jimmunol.1700394.
110. Simpson, A. J., Caballero, O. L., Jungbluth, A., Chen,
Y. T., and Old, L. J. (2005) Cancer/testis antigens, ga-
metogenesis and cancer, Nat. Rev. Cancer, 5, 615-625,
doi:10.1038/nrc1669.
111. Hong,Y., Zhao,Y., Li,H., Yang,Y., Chen,M., etal. (2023)
Engineering the maturation of stem cell-derived car-
diomyocytes, Front. Bioeng. Biotechnol., 11, 1155052,
doi:10.3389/fbioe.2023.1155052.
112. Tricot,T., Verfaillie, C. M., and Kumar, M. (2022) Cur-
rent status and challenges of human induced pluripo-
tent stem cell-derived liver models in drug discovery,
Cells, 11, 442, doi:10.3390/cells11030442.
113. Diane, A., Mohammed, L. I., and Al-Siddiqi, H. H.
(2023) Islets in the body are never flat: transition-
ing from two-dimensional (2D) monolayer culture to
three-dimensional (3D) spheroid for better efficien-
cy in the generation of functional hPSC-derived pan-
creatic β cells in vitro, Cell Commun. Signal., 21, 151,
doi:10.1186/s12964-023-01171-8.
114. Ma, H., Folmes, C. D., Wu, J., Morey, R., Mora-Castil-
la, S., et al. (2015) Metabolic rescue in pluripotent
cells from patients with mtDNA disease, Nature, 524,
234-238, doi:10.1038/nature14546.
115. Cacchiarelli, D., Trapnell, C., Ziller, M. J., Soumil-
lon, M., Cesana, M., et al. (2015) Integrative analyses
of human reprogramming reveal dynamic nature of
induced pluripotency, Cell, 162, 412-424, doi:10.1016/
j.cell.2015.06.016.
116. Choi, J., Lee, S., Mallard, W., Clement, K., Tagliazuc-
chi, G. M., et al. (2015) A comparison of genetically
matched cell lines reveals the equivalence of hu-
man iPSCs and ESCs, Nat. Biotechnol., 33, 1173-1181,
doi:10.1038/nbt.3388.
117. Idelson, M., Alper, R., Obolensky, A., Yachimovich-
Cohen,N., Rachmilewitz,J., etal. (2018) Immunologi-
cal properties of human embryonic stem cell-derived
retinal pigment epithelial cells, Stem Cell Rep., 11, 681-
695, doi:10.1016/j.stemcr.2018.07.009.
118. Yamasaki, S., Sugita, S., Horiuchi, M., Masuda, T., Fu-
jii,S., etal. (2021) Low immunogenicity and immuno-
suppressive properties of human ESC- and iPSC-de-
rived retinas, Stem Cell Rep., 16, 851-867, doi:10.1016/
j.stemcr.2021.02.021.
119. Edo, A., Sugita, S., Futatsugi, Y., Sho, J., Onishi, A.,
et al. (2020) Capacity of retinal ganglion cells de-
rived from human induced pluripotent stem cells to
DYSREGULATION OF IMMUNE TOLERANCE TO AUTOLOGOUS iPSCs 815
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
suppress T-cells, Int.J. Mol. Sci., 21, 7831, doi:10.3390/
ijms21217831.
120. Liu, J., Götherström, C., Forsberg, M., Samuelsson,
E.B., Wu,J., etal. (2013) Human neural stem/progeni-
tor cells derived from embryonic stem cells and fetal
nervous system present differences in immunogenic-
ity and immunomodulatory potentials in vitro, Stem
Cell Res., 10, 325-337, doi:10.1016/j.scr.2013.01.001.
121. Itakura, G., Ozaki, M., Nagoshi, N., Kawabata, S.,
Nishiyama, Y., et al. (2017) Low immunogenicity of
mouse induced pluripotent stem cell-derived neural
stem/progenitor cells, Sci. Rep., 7, 12996, doi:10.1038/
s41598-017-13522-w.
122. Ozaki,M., Iwanami, A., Nagoshi, N., Kohyama,J., Ita-
kura,G., etal. (2017) Evaluation of the immunogenic-
ity of human iPS cell-derived neural stem/progenitor
cells invitro, Stem Cell Res., 19, 128-138, doi:10.1016/
j.scr.2017.01.007.
123. Fujii,S., Yoshida,S., Inagaki,E., Hatou,S., Tsubota,K.,
et al. (2019) Immunological properties of neural
crest cells derived from human induced pluripotent
stem cells, Stem Cells Dev., 28, 28-43, doi: 10.1089/
scd.2018.0058.
124. Mehler, V. J., Burns, C. J., Stauss, H., Francis, R. J.,
and Moore, M. L. (2020) Human iPSC-derived neu-
ral crest stem cells exhibit low immunogenicity, Mol.
Ther. Methods Clin. Dev., 16, 161-171, doi: 10.1016/
j.omtm.2019.12.015.
125. Kimura,T., Yamashita,A., Ozono,K., and Tsumaki,N.
(2016) Limited immunogenicity of human induced
pluripotent stem cell-derived cartilages, Tissue Eng.
PartA, 22, 1367-1375, doi:10.1089/ten.TEA.2016.0189.
126. Wong, K. G., Ryan, S. D., Ramnarine,K., Rosen, S. A.,
Mann, S. E., et al. (2017) CryoPause: a new method
to immediately initiate experiments after cryopres-
ervation of pluripotent stem cells, Stem Cell Rep., 9,
355-365, doi:10.1016/j.stemcr.2017.05.010.
127. Ji,J., Sharma, V., Qi, S., Guarch, M. E., Zhao, P., et al.
(2014) Antioxidant supplementation reduces genom-
ic aberrations in human induced pluripotent stem
cells, Stem Cell Rep., 2, 44-51, doi: 10.1016/j.stemcr.
2013.11.004.
128. Martin, M.J., Muotri,A., Gage,F., and Varki,A. (2005)
Human embryonic stem cells express an immuno-
genic nonhuman sialic acid, Nat. Med., 11, 228-232,
doi:10.1038/nm1181.
129. Tangvoranuntakul,P., Gagneux,P., Diaz,S., Bardor,M.,
Varki, N., et al. (2003) Human uptake and incorpo-
ration of an immunogenic nonhuman dietary sial-
ic acid, Proc. Natl. Acad. Sci. USA, 100, 12045-12050,
doi:10.1073/pnas.2131556100.
130. Rodríguez-Pizà, I., Richaud-Patin, Y., Vassena, R.,
González, F., Barrero, M. J., et al. (2010) Reprogram-
ming of human fibroblasts to induced pluripotent
stem cells under xeno-free conditions, Stem Cells,
28, 36-44, doi:10.1002/stem.248.
131. Swistowski,A., Peng,J., Liu,Q., Mali,P., Rao, M.S., etal.
(2010) Efficient generation of functional dopaminergic
neurons from human induced pluripotent stem cells
under defined conditions, Stem Cells, 28, 1893-1904,
doi:10.1002/stem.499.
132. Chung, T.L., Turner, J.P., Thaker, N.Y., Kolle,G., Cooper-
White, J.J., etal. (2010) Ascorbate promotes epigenetic
activation of CD30 in human embryonic stem cells,
Stem Cells, 28, 1782-1793, doi:10.1002/stem.500.
133. Lagarkova, M. A., Volchkov, P. Y., Philonenko, E. S.,
Pfannkuche, K., Prokhorovich, M. A., et al. (2008)
CD 30 is a marker of undifferentiated human em-
bryonic stem cells rather than a biomarker of trans-
formed hESCs, Cell Cycle, 7, 3610-3612, doi: 10.4161/
cc.7.22.6981.
134. Kaneko, S., and Yamanaka, S. (2013) To be immuno-
genic, or not to be: that’s the iPSC question, Cell Stem
Cell, 12, 385-386, doi:10.1016/j.stem.2013.03.008.
135. Mandai, M., Watanabe, A., Kurimoto, Y., Hirami, Y.,
Morinaga, C., et al. (2017) Autologous induced stem-
cell-derived retinal cells for macular degenera-
tion, N. Engl. J. Med., 376, 1038-1046, doi: 10.1056/
NEJMoa1608368.
136. Takagi,S., Mandai,M., Gocho,K., Hirami,Y., Yamamo-
to, M., et al. (2019) Evaluation of transplanted autol-
ogous induced pluripotent stem cell-derived retinal
pigment epithelium in exudative age-related mac-
ular degeneration, Ophthalmol. Retina, 3, 850-859,
doi:10.1016/j.oret.2019.04.021.
137. Schweitzer, J.S., Song,B., Herrington, T.M., Park, T.Y.,
Lee, N., et al. (2020) Personalized iPSC-derived dopa-
mine progenitor cells for Parkinson’s disease, N.Engl.
J.Med., 382, 1926-1932, doi:10.1056/NEJMoa1915872.
138. Sugimoto, N., Kanda, J., Nakamura, S., Kitano, T.,
Hishizawa, M., et al. (2022) iPLAT1: the first-in-hu-
man clinical trial of iPSC-derived platelets as a phase
1 autologous transfusion study, Blood, 140, 2398-2402,
doi:10.1182/blood.2022017296.
139. Hu,X., White,K., Olroyd, A.G., DeJesus,R., Dominguez,
A. A., et al. (2023) Hypoimmune induced pluripotent
stem cells survive long term in fully immunocompe-
tent, allogeneic rhesus macaques, Nat. Biotechnol.,
doi:10.1038/s41587-023-01784-x.
140. Deuse,T., Hu,X., Agbor-Enoh,S., Jang, M.K., Alawi,M.,
et al. (2021) The SIRPα-CD47 immune checkpoint in
NK cells, J. Exp. Med., 218, e20200839, doi: 10.1084/
jem.20200839.
141. Harding, J., Vintersten-Nagy, K., Yang, H., Tang, J. K.,
Shutova, M., et al. (2023) Immune-privileged tissues
formed from immunologically cloaked mouse embry-
onic stem cells survive long term in allogeneic hosts,
Nat. Biomed. Eng., doi:10.1038/s41551-023-01133-y.
142. Viacyte. CRISPR Therapeutics and ViaCyte, Inc. an-
nounce first patient dosed in phase 1 clinical trial
of novel gene-edited cell replacement therapy for
treatment of type1 diabetes (T1D). February 2, 2022,
BOGOMIAKOVA et al.816
BIOCHEMISTRY (Moscow) Vol. 89 No. 5 2024
URL: https://crisprtx.com/about-us/press-releases-and-
presentations/crispr-therapeutics-and-viacyte-inc-
announce-first-patient-dosed-in-phase-1-clinical-trial-
of-novel-gene-edited-cell-replacement-therapy-for-
treatment-of-type-1-diabetes-t1d.
143. González, B. J., Creusot, R. J., Sykes, M., and Egli, D.
(2020) How safe are universal pluripotent stem
cells? Cell Stem Cell, 26, 307-308, doi: 10.1016/j.stem.
2020.02.006.
144. Harding, J., Vintersten-Nagy, K., and Nagy, A. (2020)
Universal stem cells: making the unsafe safe, Cell Stem
Cell, 27, 198-199, doi:10.1016/j.stem.2020.07.004.
145. Zimmer, J., Andrès, E., Donato, L., Hanau, D., Hent-
ges, F., and de la Salle, H. (2005) Clinical and immu-
nological aspects of HLA class I deficiency, QJM, 98,
719-727, doi:10.1093/qjmed/hci112.
146. Hara, A., Aoki, H., Taguchi, A., Niwa, M., Yamada, Y.,
Kunisada, T., and Mori, H. (2008) Neuron-like differen-
tiation and selective ablation of undifferentiated em-
bryonic stem cells containing suicide gene with Oct-4
promoter, Stem Cells Dev., 17, 619-627, doi: 10.1089/
scd.2007.0235.
147. Liang,Q., Monetti,C., Shutova, M.V., Neely, E.J., Ha-
cibekiroglu,S., etal. (2018) Linking a cell-division gene
and a suicide gene to define and improve cell thera-
py safety, Nature, 563, 701-704, doi: 10.1038/s41586-
018-0733-7.
148. Wu, Y., Chang, T., Long, Y., Huang, H., Kandeel, F.,
and Yee, J. K. (2019) Using gene editing to establish
a safeguard system for pluripotent stem-cell-based
therapies, iScience, 22, 409-422, doi: 10.1016/j.isci.
2019.11.038.
149. Viacyte. CRISPR Therapeutics and ViaCyte present
positive in vitro data towards a potential immune-eva-
sive cell replacement therapy for diabetes at EASD
2019. September 17, 2019, URL: https://crisprtx.com/
about-us/press-releases-and-presentations/crispr-
therapeutics-and-via-cyte-present-positive-in-vitro-
data-towards-a-potential-immune-evasive-cell-
replacement-therapy-for-diabetes-at-easd-2019.
Publishers Note. Pleiades Publishing remains
neutral with regard to jurisdictional claims in pub-
lished maps and institutional affiliations.