SlideShare a Scribd company logo
1 of 13
Download to read offline
Human mesenchymal stem cells with enhanced telomerase activity
acquire resistance against oxidative stress-induced genomic damage
VARVARA TRACHANA1
, SPYROS PETRAKIS2
, ZISIS FOTIADIS3
, EVANGELIA K. SISKA2,4
,
VASILEIOS BALIS3
, EFSTATHIOS S. GONOS5
, MARTHA KALOYIANNI3
&
GEORGE KOLIAKOS2,4
1
Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece,
2
Biohellenika Biotechnology Company, 57001 Thessaloniki, Greece, 3
Department of Zoology, School of Biology,
Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece, 4
Department of Biological Chemistry, Medical School,
Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece, and 5
National Hellenic Research Foundation, 48Vas.
Konstantinou Str, 11635 Athens, Greece
Abstract
Background. Human mesenchymal stem cells (MSC) are important tools for several cell-based therapies. However, their
use in such therapies requires in vitro expansion during which MSCs quickly reach replicative senescence. Replicative se-
nescence has been linked to macromolecular damage, and especially oxidative stress-induced DNA damage. Recent studies
on the other hand, have implicated telomerase in the cellular response to oxidative damage, suggesting that telomerase has
a telomere-length independent function that promotes survival. Methods. Here, we studied the DNA damage accumula-
tion and repair during in vitro expansion as well as after acute external oxidative exposure of control MSCs and MSCs that
overexpress the catalytic subunit of telomerase (hTERT MSCs). Results. We showed that hTERT MSCs at high passages
have a significant lower percentage of DNA lesions as compared to control cells of the same passages. Additionally, less
damage was accumulated due to external oxidative insult in the nuclei of hTERT overexpressing cells as compared to the
control cells. Moreover, we demonstrated that oxidative stress leads to diverse nucleus malformations, such as multillobular
nuclei or donut-shaped nuclei, in the control cells whereas hTERT MSCs showed significant resistance to the formation
of such defects. Finally, hTERT MSCs were found to possess higher activities of the basic antioxidant enzymes, superox-
ide dismutase and catalase, than control MSCs. Discussion. On the basis of these results, we propose that hTERT enhancement
confers resistance to genomic damage due to the amelioration of the cell’s basic antioxidant machinery.
Key Words: mesenchymal stem cells, telomerase, oxidative stress, genomic damage
Abbreviations: MSCs, human Mesenchymal Stem Sells; ASCs, Adipose Stem Cells; WJ, Wharton’s Jelly; hTERT, catalytic
subunit of human telomerase; ROS, Reactive Oxygen Species; SAC, Spindle Assembly Checkpoint.
Introduction
Several studies have demonstrated the therapeutic po-
tential of human mesenchymal stem cells (MSCs).Their
self-renewal ability and multilineage differentiation
(stemness) have made them a rather attractive candi-
date for cell based therapies. In fact, clinical trials are
currently taking place using MSCs as the main ther-
apeutic agent for different human diseases [1]. Even
though MSCs can be easily isolated from bone marrow,
adipose tissue, the umbilical cord and other tissues, their
expansion in vitro is not free of concerns [2,3]. It is ac-
tually well documented that MSCs reachreplicative
senescence rather fast in culture conditions [4,5] which
impairs their regenerative potential [6,7].
Replicative senescence is a common feature to all
somatic cells and most stem cells; after a period of rig-
orous proliferation the rate of cell division drops until
cells reach a state where they are still alive but unable
to divide further.The phenomenon is accompanied by
a number of characteristic changes, such as increase in
size and flattening, nuclear changes as well as several
changes in gene and protein expression patterns [8,9].
The telomere shortening hypothesis is currently the best
explanation for the limited proliferation potential of the
cells.Telomeres are hexanucleotide repeats which extend
Correspondence: Varvara Trachana, PhD, Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa,
Greece. E-mail: vtrachana@med.uth.gr
(Received 20 February 2017; accepted 25 March 2017)
ISSN 1465-3249 Copyright © 2017 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.jcyt.2017.03.078
Cytotherapy, 2017; 19: 808–820
over several thousand base pairs (bp) in length, forming
functional “caps” at the end of the chromosomes.These
repeats due to the “end-replication-problem” are sub-
jected to erosion with each round of DNA replication.
Telomeric DNA is synthesized by a specialized reverse
transcriptase called telomerase [10].When telomerase
is not present or is expressed at very low levels, pro-
gressive shortening of the telomeres is taking place in
every cell division until the telomeres reach a crucial
length. At this point cellular senescence is triggered by
activating the DNA damage checkpoint that prevents
cells from further cycling [11].
For MSCs the proliferation limit determined by
the length of their telomeres bears another obstacle:
the age of the donor of the cells, since it has been
shown to be correlated with the initial telomeres length,
and therefore the time that MSCs can be main-
tained in culture [12]. The initial telomere length of
the donor’s MSCs could therefore be a limiting factor
of the MSCs therapeutic potential since it will affect
the survival and integration ability of the trans-
planted cells to the adult tissue [13–15]. In fact there
is increasing evidence that the age of the donor affects
several properties of MSCs [16].
On the other hand, it has been demonstrated that
the erosion of the telomeres is not only due to the way
DNA is replicated, but it can also be accelerated or even
induced by environmental factors, such as the oxida-
tive stress (OS) [17,18]. Different human cells subjected
to long-term exposure to mild OS have shown to
undergo accelerated telomere erosion that triggers pre-
mature senescence [17]. This phenomenon has been
collectively termed ‘stress induced premature senes-
cence’ (SIPS) [19]. It was also reported that hydrogen
peroxide (H2O2) can cause predominant DNA damage
at the 5΄ site of 5′-GGG-3′ in the telomere sequence.
Furthermore, H2O2 induced the formation of 8-oxo-
7,8-dihydro-2′-deoxyguanosine (8-oxo-dG), an oxidized
derivative of deoxyguanosine and major product of DNA
oxidation [20], in telomere sequences more efficient-
ly than that in non-telomere sequences [21]. Different
groups, however, have reported that SIPS could also
be independent of telomeric damage [22]. Despite the
controversies on the matter, it is widely acknowl-
edged that the progressive oxidative damage to the
macromolecules, and especially to DNA, can induce
cellular senescence [23]. Even though many different
groups have studied the involvement of OS in MSCs
senescence in culture [24–26], the exact underlying mo-
lecular mechanism remains unclear.
Interestingly, it has been recently reported that
telomerase has also non-canonical or extratelomeric
functions, that are involved in processes such resis-
tance to stress and especially to oxidative stress. In fact,
Borras et al. [27] showed that glutathione, a physio-
logical antioxidant regulates telomerase activity,
highlighting therefore the interplay between telomerase
and the cellular oxidative status. Moreover, emerg-
ing data demonstrate that telomerase could be directly
involved in the reduction of reactive oxygen species
(ROS) generation and therefore decreased oxidative
DNA damage [28–31].
In order to investigate the antioxidant properties
of telomerase, and their implication in the DNA
damage driven decline in the cellular proliferation po-
tential, in the present study we analyzed the oxidative
stress-induced DNA damage response of MSCs ge-
netically modified with the catalytic subunit of human
telomerase (hTERT). In the analysis we included
adipose derived MSCs (ASCs) from adult individu-
als as well as umbilical cord’s Wharton Jelly derived
cells (WJ-MSCs), in an attempt to determine possi-
ble differences that could be attributed to the donor’s
initial telomeres length. We believe that this ap-
proach provides insights regarding the recently
proposed antioxidant role for telomerase, which could
contribute in developing strategies that will over-
come the senescence-associated impairment of the
regenerative potential of MSCs.
Materials and methods
Construction of the hTERT transposon
An entry clone encoding the human telomerase reverse
transcriptase catalytic subunit (hTERT), (GeneID:
7015, clone ID: IOH36343) was shuttled into a pT2-
CAGGS-EYFP-GW plasmid [32] using LR clonase
(Life Technologies), according to manufacturer’s in-
structions. The reaction mixture was used for
transformation of Mach1 E. coli cells and transformants
were selected on agar plates supplemented with 100 µg/
ml Ampicillin. Plasmid isolation was performed using
NucleoSpin plasmid isolation kit (Macherey-Nagel).
The identity of pT2-CAGGS-EYFP-hTERT plasmid
was verified by BsrGI restriction digestion.
Generation of hTERT mesenchymal stem cells
MSCs were enzymatically isolated from adipose tissue
(ASCs) of healthy donors (four different individu-
als, n = 4, age range of 49–60 years) by lipoaspiration.
Informed consent was given for the collection of
samples from all donors and the collection was per-
formed in accordance with established guidelines. For
the isolation of ASCs from the adipose tissue, tissues
were treated with collagenase (2.7 mg/ml) and hyal-
uronidase (0.7 mg/ml) solution for 1h at 37°C followed
by incubation with trypsin (2.5%). Cell suspension was
diluted with equal volume of PBS, passed through a
sterile 0.2 µm filter and centrifuged at 500g for 30 min
at room temperature (RT).We also isolated and used
MSCs from theWharton Jelly of umbilical cords (WJ
Telomerase protects against genomic instability 809
MSCs) from term-gestation newborns after birth (two
different individuals, n = 2), having obtained consent
from the parents. The isolating method used in our
laboratory was previously described [33].
After isolation ASCs and WJ MSCs were diluted
in Dulbecco’s modified Eagle’s medium (DMEM,
Sigma-Aldrich, Saint Louis, MO, USA) supple-
mented with 10% fetal bovine serum (FBS, Sigma-
Aldrich), penicillin (100 IU/mL) and streptomycin
(100 µg/mL) and placed in a 6-well plates. 2x105
cells
at passage 4 were grown to 70–80% confluency and
then nucleofected with 7.5 µg DNA encoding Sleep-
ing Beauty 100X transposase [34], pT2-CAGGS-
YFP-hTERT and pT2-SV40-neoR transposon
plasmids (1:8:1 ratio) using a Nucleofector device
(Lonzabio). hTERT ASCs and hTERTWJ MSCs were
selected with 100 µg/ml G418. All following analy-
ses were performed in four individual cultures of ASCs
(four control ASCs and four hTERT ASCs) and in
two individual cultures ofWJ MSCs (two controlWJ-
MSCs and two hTERT WJ-MSCs.
Culture conditions
Cells were cultured under standard cell-culture in
DMEM (supplemented with 10% FBS, penicillin
(100 IU/mL) and streptomycin (100 µg/mL)) in a hu-
midified incubator set to 37°C and 5% CO2 and 20%
O2. Medium was changed twice a week and cells were
passed when reached confluency. Cell number was de-
termined in duplicates using an hemocytometer. For
exposure to hydrogen peroxide (H2O2), 200.000 cells
(hTERT or control MSCs) were seeded per well on a
6 well plate and at 65–70% confluence cells were
exposed to 300µΜ H2O2 for 30 min in serum-free
medium. The medium was then replaced with fresh
complete medium and cells were normally cultured for
48 h (recovery time) in order to recover from the stress.
Antibodies
Mouse monoclonal anti-γH2AX (05–636; phosphor
S139, clone JBW301), and anti-53BP1 (clone BP13,)
were from Millipore (MA, USA). Anti- 8-hydroxy-
2’- deoxyguanosine (8-oxo-dG, Clone 2E2), was
purchased from Trevigen. For the western blots anti-
hTERT (NB120-32020) was from Novus and anti-
β-actin antibody (sc-47778) from Santa Cruz.
Secondary antibodies for confocal microscopy, Alexa
Fluor 594 anti-mouse IgG (A11005) and AlexaFluor
488 anti-mouse IgG (A11001 were obtained from Mo-
lecular Probes (Invitrogen).
qRT-PCR
Total RNA was purified from hTERT or control MSCs
using the NucleoSpin RNA kit (Macherey-Nagel),
according to manufacturer’s instructions. cDNA gen-
eration and qRT-PCR reactions were performed using
the KAPA SYBR Fast 1-step Green qRT-PCR kit
(KAPA Biosystems) in a Rotor-Gene 6000 operat-
ing system. Each analysis was performed in triplicates.
The correct size of amplified qRT-PCR products was
verified by electrophoresis in a 2% agarose gel.
Western blot analysis
hTERT or control MSCs were lysed in 1% SDS in
PBS containing Protease Inhibitors Cocktail Set III
(Calbiochem) and Benzonase (Novagen). Protein con-
centrations were determined by the Bradford method
with bovine serum albumin as standard (Bio-Rad
Laboratories, CA, USA). Samples(<20 µg total protein)
were analyzed by SDS-PAGE and transferred onto ni-
trocellulose membranes. Membranes were blocked with
5% weight per volume (w/v) non-fat dry milk in PBS/
0.1%Tween20 probed with the appropriate antibodies.
Secondary antibodies conjugated with alkaline-
phosphatase were detected using nitro blue tetrazolium
chloride/5-bromo-4-chloro-3-indolyl phosphate
p-toluidine salt (NBT/BCIP) substrate (Applichem).
Equal protein loading was verified by reprobing each
membrane with the antibody against β-actin.
Quantification of Telomerase activity
Real Time-PCR based telomeric repeat amplification
protocol (TRAP) assay was performed according to the
manufacturer’s protocol (TeloTAGGGTelomerase PCR
ELIZAPLUS-Roche Applied Science, Indianapolis, IN,
USA). In brief, 2 × 105
hTERT or control MSCs were
lysed in 200 µl lysis buffer and the cell lysate was
centrifuged at 16,000 g for 20 min at 4°C. Cell extract
(0.5 µg of total protein) was used for PCR reaction.
The amplification product (3 µl) was transferred for the
hybridization, and the ELISA assay was performed in
triplicates, in which two served as controls (one con-
tained a negative control, and a second containing the
high telomerase activity positive control). Telomerase
activity of each sample was calculated as a percentage
of the relative telomerase activity of the positive control
cells.
Flow cytometry
For phenotypic characterization, hTERT and control
MSCs were stained with phycoerythrin (PE)-
conjugated antibodies against hemopoietic (CD34, 45)
or mesenchymal (CD29, 73, 90, 105 and 146) stem
cell markers. Unstained cells or cells stained with IgG
isotype antibodies were used as negative controls. Cells
were measured in a Cytomics FC500 flow cytom-
eter (Beckman Coulter) using the CXP2.2 software.
810 V.Trachana et al.
Multilineage differentiation
For osteogenic or adipogenic differentiation, hTERT
and control MSCs were grown to 90% confluency and
cultured for 28 days either in StemPro Osteogenesis
or StemPro Adipogenesis medium (Life Technolo-
gies). Differentiation of cells into osteocytes or
adipocytes was monitored by Alizarin Red or Oil Red
staining, respectively.
Immunofluorescence
Immunofluorescence experiments were performed as
previously described [35]. Briefly, MSCs were grown
on coverslips and subjected to the above described oxi-
dative treatment or left to reach the desired passage
and then fixed in 4% paraformaldehyde. Fixed samples
were incubated with primary antibodies of interest (γ-
H2AX, 53BP1 or 8-oxo-dG) and the appropriate
secondary antibodies. Coverslips were embedded in
10 µl of Vectashield mounting medium for fluores-
cence with 4,6-diamidino-2-phenylindole (DAPI,Vector
Laboratories, CA, USA) to visualize the nuclei, and
analyzed on ZEISS Axio Imager Z2 fluorescent
microscope. Images were captured on confocal
microscope ZEISS LSM780 and ZEN 2011 program.
For calculations of damaged nuclei at least 5 ran-
domly selected fields were analyzed for each culture
condition. Cells with >5 γ-H2AX, 53BP1 or 8-oxo-
dG stained foci in their nuclei were counted as positive
for DNA damage by a single observer blinded to treat-
ment regimen.
Measurement of antioxidant enzymes activity
Total superoxide dismutase (SOD) and catalase ac-
tivities were measured in MSCs cell lysates using
commercially available kits (Superoxide Dismutase
Assay Kit 706002, Catalase Assay Kit 707002 Cayman
Chemical, Ann Arbor, MI) according to the manu-
facturer’s recommendations.
Statistical analysis
For the statistical analysis of the results the GraphPad
Prism 5 software (GraphPad Software, San Diego, Cal-
ifornia USA) was used. P values less than 0.05 were
considered significant and the significance is ex-
pressed with asterisks: * = P < 0.05; ** = P < 0.01;
*** = P < 0.001. Results are reported as mean ± stan-
dard error (means ± S.Ε.) unless otherwise stated.
Results
Genetically modified MSCs have increased expression of
hTERT and enhanced telomerase activity
Ectopic expression of hTERT in hTERT ASCs was
confirmed by qRT-PCR analysis (Figure 1A).The iden-
tity of amplified hTERT and actin beta (ACTB)
housekeeping gene fragments was verified by agarose
electrophoresis (Figure 1B). Production of recombi-
nant yellow fluorescent protein (YFP)-TERT was
evaluated by Western blotting. A protein band with a
molecular weight of ~160 kDa was detected in hTERT
ASCs corresponding toYFP-TERT, whereas endog-
enous hTERT was detected in all samples at the
expected molecular weight (127 kDa) (Figure 1C).
These results verify successful overexpression of
hTERT in ASCs.
hTERT WJ-MSCs showed similarly increased
mRNA levels of the telomerase catalytic subunit and
production of the recombinant YFP-TERT protein
(Supplementary Figure S1A and B respectively).
Finally, realTime-PCR basedTRAP assay (Roche
Applied Science) was performed according to the man-
ufacturer’s protocol.The assay confirmed that ASCs
genetically modified with the hTERT (ASCs1-ASCS4)
Figure 1. Genetically modified ASCs have increased messenger RNA (mRNA) levels of hTERT and produce recombinant YFP-TERT
protein. (A) mRNA levels of control and hTERT-overexpressing ASCs normalized to housekeeping β-actin (ACTB) gene. Values showed
are the means ± SE. *P < 0.05 versus control ASCs. (B) qRT-PCR–amplified fragments analyzed in a 2% agarose electrophoresis. (C)Western
blot analysis for hTERT in control and hTERT-ASCs.The analysis was performed in all control and hTERT ASC individual cultures and
a characteristic image is demonstrated. Molecular weight markers are run in the middle lane. β-actin was used as a loading control.
Telomerase protects against genomic instability 811
have increased telomerase activity when compared to
the control cells (Figure 2).
MSCs overexpressing hTERT retain their
mesenchymal properties
hTERT MSCs derived from adipose tissue were further
characterized in order to verify that the genetic mod-
ification does not affect their mesenchymal properties.
hTERT ASCs were plastic-adherent and had a similar
fibroblastic morphology as control cells. Flow cytometry
of hTERT ASCs showed that these cells do not express
the hemopoietic CD34 marker, whereas they express
typical mesenchymal stem cell CD markers 29, 73,
90, 105 and 146 at similar levels to control ASCs
(Table I). Similarly, hTERTWJ-MSCs do not express
the CD34 marker and express the typical mesenchy-
mal CD29 and CD105 markers (Supplementary
Table SI).
The multilineage differentiation potential of hTERT
ASCs was also assessed. Cells were differentiated
towards adipocytes and osteocytes, as indicated by Oil
Red and Alizarin Red staining, respectively.At early pas-
sages (p ≤ 8) both control as well as genetically modified
ASCs were able to differentiate towards both adipocytes
and osteocytes (data not shown). As shown in Figure 3,
control ASCs at passage 10 were able to differentiate
towards adipocytes but not osteocytes, whereas hTERT
ASCs of the same passage retained their osteogenic dif-
ferentiation capacity (Figure 3A, B).
Moreover, as recently demonstrated (in a paral-
lel work done by members of our team [36],) control
WJ-MSCs maintain their differentiation ability towards
adipocytes and osteocytes even at high passage (passage
40). Here, we demonstrate that hTERT overexpression
did not affect this differentiation ability ofWJ-MSCs,
as indicated in Figure S1C.
The above data prove that the genetic modifica-
tion did not alter the mesenchymal properties of adipose
derived or WJ-derived cells. It also implies that the
overexpression of hTERT may contribute in retain-
ing their mesenchymal properties for longer (at higher
passages).
hTERT overexpression offers protection against DNA
damage accumulation during long-term culture
MSCs were kept in culture until their proliferation rate
dropped to less than 2 cumulative population doublings
per 4 weeks. At this stage control MSCs had the char-
acteristic senescent-like morphology (large, flattened
vacuolated cells), suggesting that they have reached their
proliferation limit. In accordance with previously re-
ported data [37], control ASCs reached this state at
passage 20 ± 2.3, whereas hTERT ASCs at did not seem
to enter this state even at high passages (passage > 30).
Similarly, control WJ-MSCs reached their prolifera-
tion limit at passage 22 ± 1.3, whereas hTERT
overexpressing WJ-MSCs did not show any reduction
in their duplication time until at least passage 30.
Our observations, together with those previously re-
ported [37], reveal the beneficial effect of hTERT
overexpression on the growth potential of MSCs, which,
based on our results, is not limited to adipose-derived
cells but apply to MSCs from different sources.
In an attempt to explain the amelioration in the
growth potential of hTERT overexpressing cells and
given the importance of DNA damage accumulation
in limiting cellular proliferation capacity [38], we
analysed the DNA damage in the nuclei of growing cells.
We evaluated the amount of DNA damage in control
ASCs as well hTERT ASCs at early passage (p6),
middle passage (p12) and high passage (p20).The pres-
ence of 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxo-
dG), major product of DNA oxidation as mentioned
earlier, was assessed. 8-oxo-dG lesions could lead to
reduced efficiency of the base excision repair process
leading, in turn, to persistent DNA single strand breaks
Figure 2. ASCs genetically modified with hTERT have increased
telomerase activity. RT-PCR–basedTRAP assay was used to measure
telomerase activity in hTERT ASCs at passage 7. Telomerase ac-
tivity of the hTERT ASCs is depicted as fold change relative to
the activity of the control ASCs of the same passage.The analysis
was performed in triplicate with four control ASCs and four hTERT
ASCs. Values showed are the means ± SE.
Table I. Flow cytometry analysis results for CD stem cell markers
in control ASCs and hTERT-overexpressing ASCs (hTERT ASCs)
at passage 7.
Stem cell marker
hTERT ASCs
passage 7
Control ASCs
passage 7
CD34 0% 2%
CD29 99.6% 99.8%
CD73 99.4% 99.7%
CD90 99.3% 99.3%
CD105 99.7% 97.5%
CD146 96.4% 93.8%
812 V.Trachana et al.
(SSB) intermediates; the latter could then act as double
strand breaks (DSB)-prone sites in subsequent rounds
of DNA replication [39]. DSB activate ataxia-
telangiectasia mutated/ataxia telangiectasia and Rad3-
related serine/threonine-protein kinases (ATM/ATR)
that bind to the DNA and induce phosphorylation of
many downstream proteins, such as H2A.X and 53BP1,
in order to both mark the location and initiate repair
of the damage. In order to evaluate DSB formation in
long-term cultures of MSCs, we therefore assessed
control ASCs as well as hTERT ASCs of different pas-
sages (p6, p12 and p20) for the presence of the
phosphorylated form (on Ser139) of the histone variant
H2AX (γH2AX) and the 53BP1 protein. Figure 4A
shows that growing ASCs accumulate DSB as well as
oxidative lesions, as indicated by the increased per-
centage of cells positive for γΗ2ΑΧ/ 53BP1 (i, ii) and
8-oxo-dG staining (iii), respectively. All nuclei bearing
>5 foci (8-oxo-dG, γΗ2ΑΧ or 53BP1) were counted as
positive. Importantly, as depicted in the same figure
(Figure 4A), our result demonstrate that cells geneti-
cally modified with the catalytic subunit of hTERT
accumulate significantly less damage than the control
cells in all passages tested. Figure 4B shows confocal
microscopy characteristic images of foci positive for
γΗ2ΑΧ, 53BP1 and 8-oxo-dG in the nuclei of control
as well hTERT overexpressing cells. It is obvious that
hTERT overexpressing cells accumulate a lot less DNA
damage associated foci in their nuclei at high passage
(p = 20).The latter suggest that the elevated activity of
telomerase protects cells against oxidative lesions and
DSB accumulation which could explain hTERT cells’
enhanced proliferation capacity.
Ectopic expression of hTERT results in ameliorated
response to exogenous oxidative stress-induced
DNA damage
The protection acquired by hTERT overexpressing cells
against DNA lesions accumulating during growth of
ASCs, prompt us to investigate whether similar pro-
tection occurs after exposure to acute external oxidative
stress.Therefore, control ASCs as well as hTERT ASCs
were treated with H2O2 for 30 min in order to evaluate
DNA damage accumulation. One hour after treat-
ment nuclei with >5 foci (for all markers) were counted
as positive. As Figure 5 shows, both control ASCs as
well as hTERT ASCs at passage 6 (early), passage 12
(middle) and passage 20 (high) treated with H2O2, ac-
cumulate similarly high amounts of DNA damage
associated foci (Figure 5A–C, p(6/12/20) + H2O2). Both
control ASCs and hTERT ASCs at early and middle
Figure 3. ASCs that overexpress hTERT retain their differentiation potential. (A, B) Passage 10 ASCs and hTERT ASCs cultured under
standard conditions (control medium) or under conditions to induce adipogenic/osteogenic differentiation (differentiation medium). Under
differentiation conditions, cells began to accumulate lipid droplets and acquired a foamy appearance with the characteristic staining for oil
red, typically seen in adipocytes (A), or exhibited calcium deposition, with the characteristic staining for alizarin red, which is typically
seen in osteocytes (B) (Scale bar: 10 µmol/L). (For interpretation of the references to colour in this figure legend, the reader is referred to
the web version of this article.)
Telomerase protects against genomic instability 813
passage were, however, able to, at least partially, repair
the damage when left to recover in fresh medium for
48 h (p6/12 + H2O2 recovery 48h), but not at high
passage (p20 + H2O2 recovery 48h). This ability was
found to be enhanced in hTERT ASCs, since signifi-
cantly less amount of DNA damage was measured in
these cells 48 h after the oxidative treatment at passages
6 and 12, than in control ASCs. The latter was grad-
ually diminished as cells reached higher passages (p20).
Overall these results suggest that ASCs of early and
middle passage genetically modified with the cata-
lytic subunit of hTERT have enhanced ability to repair
the externally provoked oxidative stress-induced DNA
lesions.
A
B
Figure 4. hTERT overexpression in ASCs offers protection against DNA damage accumulation in long-term culture. (A) Percentage of
control ASCs and hTERT-overexpressing ASCs (hTERT ASCs) with DNA damage measured by immunofluorescence using specific an-
tibodies against γH2AX (i), 53BP1 (ii) or 8-oxo-dG (iii) at the indicated passages. Nuclei bearing >5 foci stained with 8-oxo-dG, γΗ2ΑΧ
or 53BP1 antibody were counted as positive. Values shown are the means ± SE. *P < 0.05, **P < 0.01 and ***P < 0.001 versus control
ASCs in the passage indicated. (B) Characteristic images of control and hTERT ASCs at low (passage 6) and high passage (passage 20)
with DNA damage evaluated by confocal microscope and after staining with γH2AX, 53BP1 or 8-oxo-dG antibody (green). Nuclei are
stained with DAPI (blue). Scale bar 20 µm. (For interpretation of the references to colour in this figure legend, the reader is referred to
the web version of this article.)
814 V.Trachana et al.
Figure 5. hTERT overexpression in ASCs offers protection against external oxidative insult. Percentage of control ASCs and hTERT-
overexpressing ASCs (hTERT ASCS) with DNA damage measured by IF using γH2AX (A), 53BP1 (B) or 8-oxo-dG (C) antibodies at
low (passage 6), middle (passage 12) and high (passage 20) passages after treatment with 300 µmol/L H2O2 for 30 min (passage 6/12/
20 + H2O2) and after 48 h of recovery time (passage 6/12/20 + H2O2 recovery 48 h). Nuclei bearing >5 foci stained with 8-oxo-dG, γΗ2ΑΧ
or 53BP1 antibody were counted as positive. Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in each
passage indicated.
Telomerase protects against genomic instability 815
Enhanced TERT activity protects against oxidative stress
provoked nuclei malformations
It has been suggested that oxidative stress could lead
to aberrant nucleus morphology, such as giant nuclei,
donut-shaped nuclei, micronuclei or multilobular
nuclei, which is also considered a characteristic of se-
nescent cells [40]. Indeed, as shown in Figure 6A both
control as well as hTERT ASCs demonstrate an in-
crease in such malformations (i.e mitotic defects) in
passage 20 (senescence reaching) as compared to low
passage 6. Nevertheless, hTETR overexpressing ASCs
had significantly less nucleus defects than control cells
in both time points tested (p6, p20). Figure 6B shows
characteristic nuclei aberration of high passage 20
control ASCs, such as giant nucleus, donut-shaped
nucleus, micronucleus or multilobular nucleus
(Figure 6B, i, ii, iii, iv respectively).
Additionally, we tested whether the externally pro-
voked oxidative stress could lead to an increase of such
nuclei malformations. Both control ASCs and hTERT
ASCs at low (p6) and high (p20) passage treated with
300 µΜ Η2Ο2 for 30 min and left to recover for 48 h,
were DAPI-stained in order to evaluate their nuclear
morphology. Firstly, as indicated in Figure 6A, both
control ASCs and hTERT showed an increase in
nucleus malformations in both passages tested after
treatment with H2O2 (p6/20 + H2O2 recovery 48h) in
comparison with the same passages without treat-
ment (p6/20). However, significantly less aberrant
nuclei were present in hTERT ASCs 48 after treat-
ment with Η2Ο2 at both low and high passages in
comparison with the control cells. The latter indi-
cates that the hTERT overexpression was able protect
against oxidative stress-induced nuclei malforma-
tions, which, together with the previous observations,
suggests an overall ameliorated antioxidant response.
Genetically modified MSCs with hTERT demonstrate
increased activity of the basic antioxidant enzymes
Superoxide dismutases (SOD) are metaloenzymes that
catalyze the dismutation of the superoxide anion to mo-
lecular oxygen and H2O2 and thus form a crucial part
of the cellular antioxidant defense mechanism [41].
Catalase (CAT) is a ubiquitous antioxidant enzyme in-
volved in the detoxification of H2O2 by catalyzing its
conversion to molecular oxygen and water [42]. In an
A
B
Figure 6. hTERT-overexpressing ASCs show resistance to aberrant nuclei formation. (A) Percentage of cells with mitotic defects (*giant
nuclei, donut-shaped nuclei, micronuclei and multilobular nuclei) evaluated using DAPI staining of control ASCs and hTERT-
overexpressing ASCs (hTERT ASCs) at low (passage 6) and high passage (passage 20). Same nuclei aberrations were assessed at passages
6 and 20 after the cells were treated with 300 µmol/L Η2Ο2 for 30 min and left to recover for 48 h (passage 6/20 + H2O2 recovery 48 h).
Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in the passage indicated. (B) Characteristic images of
DAPI-stained (blue) giant nuclei (arrow in (i)), donut-shaped nuclei (ii), micronucleus (arrow in (iii)) and multilobular nuclei (iv) of control
ASCs at high passage (passage 20) (Scale bar 25 µm). (For interpretation of the references to colour in this figure legend, the reader is
referred to the web version of this article.)
816 V.Trachana et al.
attempt to explain the amelioration of the oxidative re-
sponse observed in hTERT overexpressing cells we
analysed the basic antioxidant capacity of ASCs. For
this reason, we measured the activities of both enzymes
(SOD and CAT) using commercially available kits
(Cayman, Chemical, Ann Arbor, MI). As shown in
Figure 7A ASCs genetically modified with hTERT
(hTERT ASCs) have similar activity of SOD with
control cells at low passage (p6). At high passage (p20)
however, control cells show a decrease in their SOD
activity whereas hTERT overexpressing cells main-
tain significantly higher levels, similar to the levels of
the low passage. Regarding CAT activity, we observed
that both control as well as hTERT overexpressing cells
have increased activity at higher passages (Figure 7B,
p20), which is in accordance with previously reported
data. In specific, it was reported that senescent cul-
tures of human lung fibroblasts have elevated H2O2
generation which stimulate cells to increase their cata-
lase activity [43]. Besides confirming that, we report
here, that hTERT ASCs have significantly higher CAT
activity levels than control cells, both at passage 6 as
well as in passage 20 (Figure 7B). The above results
reveal an enhancement of the basic antioxidant enzy-
matic defense of the cells due to hTERT overexpression.
Discussion
Endogenous oxidants generated by normal cellular oxi-
dative metabolism are a major obstacle to overcome
in cell based therapies that require ex vivo long-term
cell culture. The DNA damage that oxidative stress
(OS) provokes could lead to cellular senescence and
therefore limit the number of cells with the self-
renewal and multilineage differentiation ability
(stemness) that is required for successful transplan-
tation. Besides DNA damage, it has been reported that
OS -either normally occurred due to metabolism or
through exposure to external oxidants- leads to ab-
errant nuclear morphology. It was demonstrated that
a significant number of senescent or progeroid human
fibroblasts exhibit nuclei malformations, such as
multilobbular nuclei and one or multiple micronu-
clei that are attributed to mitotic slippage [40]. Mitotic
slippage is a known consequence of a dysfunctional
spindle assembly checkpoint (SAC) [44] and OS was
proposed to be a major cause of its impairment [45].
Moreover, it was recently demonstrated that, exog-
enously induced OS is able to promote aneuploidy in
MSCs [37] which could also be attributed to an im-
paired SAC. Actually, it was proposed that OS could
affect the functionality of the SAC by regulating the
expression of two key SAC mediators, BubR1 and
Mad2 [46]. All the above suggest that OS provokes
genomic instability, i.e. DNA damage, nuclear defor-
mations and aneuploidy that reduce the stemness of
MSCs in culture and limit transplantation success.
On the other hand, accumulating recent evi-
dence link the catalytic subunit of telomerase to
functions other than telomere length maintenance. As
a matter of fact, one of these extratellomeric func-
tions of telomerase that attracted a lot of attention
recently is its implication in the antioxidant defense
mechanism(s) of the cell [47]. The precise molecu-
lar mechanism of telomerase’s telomere independent
functions is becoming an important-still mostly
unanswered-question.
In order to shed light onto these issues, in the present
study we genetically modified human mesenchymal stem
cells derived from different sources (adult adipose tissue
and umbilical cord’s Wharton’s Jelly) with the cata-
lytic subunit of telomerase (hTERT) in order to
elucidate its contribution in overcoming endogenously
occurring or externally provoked OS. Firstly, we showed
Figure 7. ASCs genetically modified with hTERT have higher basic antioxidant enzymes activities. Analysis of the SOD (A) and CAT
(B) enzymatic activity of control ASCs and ASCS overexpressing hTERT (hTERT ASCs) at low (passage 6) and high (passage 20) pas-
sages. Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in the passage indicated.
Telomerase protects against genomic instability 817
that genetic modification with hTERT did not alter the
mesenchymal properties of adipose orWharton’s Jelly
derived cells, in agreement with previously reported data
on human bone marrow stromal cells [48] and mouse
adipose derived MSCs [49]. Furthermore, we report
that overexpression of hTERT in ASCs may contrib-
ute in retaining their mesenchymal properties for longer,
an observation which is in agreement with previous
results [50]. Importantly, we proved that the genetic
modification of MSCs with hTERT results in de-
creased DNA damage accumulation, that occurs either
as a result of long term culture or due to acute oxida-
tive external insult. It has been suggested that hTERT
overexpression ameliorates the DNA damage re-
sponse by either accelerating DNA repair kinetics
through increased levels of intracellular deoxynucleotides
(dNTP) and ribonucleotides (NTPs) [51], or by at-
tracting DNA repair proteins to the sites of DNA
damage [52]. Even though the hTERT participation
in DNA damage repair is a rather attractive notion and
some supportive data do exist, the exact mechanism
remains elusive. Based on our data it is, at this point,
safer to hypothesize that the protection against DNA
damage accumulation results from an hTERT driven
enhancement of the antioxidant cellular response. In
support, recent studies showed that the telomerase lo-
calizes to subcellular locations other than the nucleus,
such as the mitochondria. hTERT was shown to shuttle
dynamically from the nucleus to the mitochondria,
where it decreases mitochondrial reactive oxygen species
(ROS) generation [28–31]. Furthermore, strong evi-
dence has been provided that increased hTERT
expression not only reduces the total basal ROS levels,
but also significantly antagonizes the increase in cel-
lular ROS in response to exogenous ROS triggers
(H2O2) [53].
We also showed that MSCs genetically modified
with hTERT exhibit resistance to nuclei malforma-
tions that arise either through prolonged culture or
due to H2O2 treatment. Nuclei malformations, as men-
tioned above, could be the result of an OS-induced
SAC impairment.The resistance against OS-induced
aberrant nuclei formation demonstrated here is a novel
indication of telomerase’s ability to counteract OS and
promote cell survival.
We know that in order to resist ROS, the cells use
enzymatic antioxidants, with superoxide dismutase
(SOD) and catalase (CAT) being the major ones [54].
Our data demonstrate that hTERT overexpression
results in increased SOD and CAT activities. It was
previously reported that hTERT modified MSC dem-
onstrated elevated protein levels of the principal protein
scavenger of mitochondrial superoxide, MnSOD
(SOD2), suggesting that hTERT overexpression could
be a key regulator of the metabolic status [37]. Here
we demonstrate that hTERT overexpression could
directly lead to an enhancement of the antioxidant ma-
chinery of the cell by elevating the activity of the key
enzymes. In support, one previous study has shown
that mouse embryonic fibroblasts isolated from
telomerase-deficient mice, have decreased catalase ac-
tivity which results in redox imbalance in these mice
[55].
Finally, it has been reported that the donor’s initial
telomeres length might be a critical restriction for the
maintenance of the MSCs in prolonged cell culture
[56]. Following transplantation, the telomere length
might limit the therapeutic potential of MSCs, by de-
creasing either the duration of cell survival in the tissue
or their ability to integrate in the adult tissue [57,58].
Here, we prove that both adult adipose derived stem
cells as well as umbilical cord’sWharton’s Jelly derived
cells genetically modified with hTERT have an en-
hanced growth potential that could be attributed to
their improved antioxidant capacity. Therefore, this
genetic manipulation implemented by us, and others
[48,59], could overcome the obstacle of shorter initial
telomere length of the adult stem cells. Understand-
ing the restrains of this strategy we believe that synthetic
or natural compounds that activate the telomerase
might be a better approach.
As a matter of fact, Tichon et al. [60] demon-
strated that two novel compounds, designated AGS-
499 and AGS-500, when administrated to MSCs
increased the average telomere length, preserved
genome integrity and allowed MSCs to differentiate
into various lineage. The authors also demonstrated
that these effects were telomerase depended. Our study
confirms these previously reported results but also
demonstrates further novel effects of the hTERT
overexpression. We showed that hTERT enhance-
ment, via direct genetic manipulation, confers higher
growth potential to the cells, most probably due to
the amelioration of the cellular response to the OS that
either occurs normally during cellular senescence or
it is exogenously provoked. Moreover, we demon-
strated that this ameliorated OS response of the
hTERT overexpressing cells comes from direct en-
hancement of the cell’s basic enzymatic antioxidant
machinery, which in turn protects against OS-induced
genomic damage. In other words, our study pro-
vides novel data in support of the recently proposed
antioxidant role of telomerase. Besides that, we believe
that our analysis offers a comprehensive evaluation of
the outcomes of an augmented telomerase activity in
human MSCs that might help overcome the adult stem
cell-based therapies limitations.
Acknowledgments
This work was funded by a SYNERGASIA project
(11ΣΥΝ_1_1112). Authors wish to thank Dr. Z. Ivics
818 V.Trachana et al.
and Dr. Z. Izsvák for kindly providing the Sleeping
Beauty 100X transposon system, A. Kalkavouri and
A. Ntoga for technical assistance and Dr. E. Panteris
for his help with the microscopy techniques.
Disclosure of interests:The authors declare no com-
mercial or financial conflict of interest.
References
[1] Wei X, Yang X, Han Z, Qu F, Shao L, Shi Y. Mesenchymal
stem cells: a new trend for cell therapy. Acta Pharmacol Sin
2013;34:747–54. doi:10.1038/aps.2013.50.
[2] Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells:
revisiting history, concepts, and assays. Cell Stem Cell
2008;2:313–19. doi:10.1016/j.stem.2008.03.002.
[3] Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ,
et al. Multilineage cells from human adipose tissue:
implications for cell-based therapies. Tissue Eng 2001;7:211–
28. doi:10.1089/107632701300062859.
[4] Baxter MA, Wynn RF, Jowitt SN, Wraith JE, Fairbairn LJ,
Bellantuono I. Study of telomere length reveals rapid aging
of human marrow stromal cells following in vitro expansion.
Stem Cells 2004;22:675–82. doi:10.1634/stemcells.22-5
-675.
[5] Brandl A, Meyer M, Bechmann V, Nerlich M, Angele P.
Oxidative stress induces senescence in human mesenchymal
stem cells. Exp Cell Res 2011;317:1541–7. doi:10.1016/
j.yexcr.2011.02.015.
[6] Gu Y, Li T, Ding Y, Sun L, Tu T, Zhu W, et al. Changes in
mesenchymal stem cells following long-term culture in vitro.
Mol Med Rep 2016;13:5207–15. doi:10.3892/mmr.2016
.5169.
[7] Kasper G, Mao L, Geissler S, Draycheva A, Trippens J,
Kühnisch J, et al. Insights into mesenchymal stem cell aging:
involvement of antioxidant defense and actin cytoskeleton.
Stem Cells 2009;27:1288–97. doi:10.1002/stem.49.
[8] Campisi J. The biology of replicative senescence. Eur J Cancer
A 1997;33:703–9. doi:10.1016/S0959-8049(96)00058-5.
[9] Cristofalo VJ, Lorenzini A, Allen RG, Torres C, Tresini M.
Replicative senescence: a critical review. Mech Ageing Dev
2004;125:827–48. doi:10.1016/j.mad.2004.07.010.
[10] Collins K, Mitchell JR. Telomerase in the human organism.
Oncogene 2002;21:564–79. doi:10.1038/sj/onc/.
[11] Smogorzewska A, de Lange T. Different telomere damage
signaling pathways in human and mouse cells. EMBO J
2002;21:4338–48. doi:10.1093/emboj/cdf433.
[12] Siegel G, Kluba T, Hermanutz-Klein U, Bieback K, Northoff
H, Schafer R. Phenotype, donor age and gender affect function
of human bone marrow-derived mesenchymal stromal cells.
BMC Med 2013;11:146. doi:10.1186/1741-7015-11-146.
[13] Huang K, Zhou DH, Huang SL, Liang SH. [Age-related
biological characteristics of human bone marrow mesenchymal
stem cells from different age donors]. Zhongguo ShiYan Xue
Ye Xue Za Zhi 2005;13:1049–53.
[14] Stenderup K, Justesen J, Clausen C, Kassem M. Aging is
associated with decreased maximal life span and accelerated
senescence of bone marrow stromal cells. Bone 2003;33:919–
26.
[15] Stolzing A, Jones E, McGonagle D, Scutt A. Age-related
changes in human bone marrow-derived mesenchymal stem
cells: consequences for cell therapies. Mech Ageing Dev
2008;129:163–73. doi:10.1016/j.mad.2007.12.002.
[16] Kalaszczynska I, Ferdyn K. Wharton’s jelly derived
mesenchymal stem cells: future of regenerative medicine?
Recent findings and clinical significance. Biomed Res Int
2015;2015:430847. doi:10.1155/2015/430847.
[17] Von Zglinicki T. Oxidative stress shortens telomeres.
Trends Biochem Sci 2002;27:339–44. doi:10.1016/S0968
-0004(02)02110-2.
[18] Houben JMJ, Moonen HJJ, van Schooten FJ, Hageman GJ.
Telomere length assessment: biomarker of chronic oxidative
stress? Free Radic Biol Med 2008;44:235–46. doi:10.1016/
j.freeradbiomed.2007.10.001.
[19] Toussaint O, Medrano EE, von Zglinicki T. Cellular and
molecular mechanisms of stress-induced premature senescence
(SIPS) of human diploid fibroblasts and melanocytes. Exp
Gerontol 2000;35:927–45.
[20] Fraga CG, Shigenaga MK, Park JW, Degan P, Ames BN.
Oxidative damage to DNA during aging: 8-hydroxy-2’-
deoxyguanosine in rat organ DNA and urine. Proc Natl
Acad Sci USA 1990;87:4533–7. doi:10.1073/pnas.87.12
.4533.
[21] Oikawa S, Kawanishi S. Site-specific DNA damage at GGG
sequence by oxidative stress may accelerate telomere
shortening. FEBS Lett 1999;453:365–8. doi:10.1016/S0014
-5793(99)00748-6.
[22] Chen QM, Prowse KR, Tu VC, Purdom S, Linskens MH.
Uncoupling the senescent phenotype from telomere shortening
in hydrogen peroxide-treated fibroblasts. Exp Cell Res
2001;265:294–303. doi:10.1006/excr.2001.5182.
[23] Finkel T, Holbrook NJ. Oxidants, oxidative stress and the
biology of ageing. Nature 2000;408:239–47. doi:10.1038/
35041687.
[24] Choi KM, Seo YK, Yoon HH, Song KY, Kwon SY, Lee HS,
et al. Effect of ascorbic acid on bone marrow-derived
mesenchymal stem cell proliferation and differentiation. J Biosci
Bioeng 2008;105:586–94. doi:10.1263/jbb.105.586.
[25] Lin TM, Tsai JL, Lin SD, Lai CS, Chang CC. Accelerated
growth and prolonged lifespan of adipose tissue-derived
human mesenchymal stem cells in a medium using reduced
calcium and antioxidants. Stem Cells Dev 2005;14:92–102.
doi:10.1089/scd.2005.14.92.
[26] Tsai CC, Chen YJ, Yew TL, Chen LL, Wang JY, Chiu CH,
et al. Hypoxia inhibits senescence and maintains mesenchymal
stem cell properties through down-regulation of E2A-p21 by
HIF-TWIST. Blood 2010;117:459–69. doi:10.1182/blood
-2010-05-287508.
[27] Borrás C, Esteve JM, Viña JB, Sastre J, Viña J, Pallardó FV.
Glutathione regulates telomerase activity in 3T3
fibroblasts. J Biol Chem 2004;279:34332–5. doi:10.1074/
jbc.M402425200.
[28] Ahmed S, Passos JF, Birket MJ, Beckmann T, Brings S, Peters
H, et al. Telomerase does not counteract telomere shortening
but protects mitochondrial function under oxidative stress. J
Cell Sci 2008;121:1046–53. doi:10.1242/jcs.019372.
[29] Singhapol C, Pal D, Czapiewski R, Porika M, Nelson G,
Saretzki GC. Mitochondrial telomerase protects cancer cells
from nuclear DNA damage and apoptosis. PLoS ONE
2013;8:e52989. doi:10.1371/journal.pone.0052989.
[30] Haendeler J, Drose S, Buchner N, Jakob S, Altschmied J, Goy
C, et al. Mitochondrial telomerase reverse transcriptase binds
to and protects mitochondrial DNA and function from
damage. Arterioscler Thromb Vasc Biol 2009;29:929–35.
doi:10.1161/ATVBAHA.109.185546.
[31] Saretzki G. Telomerase, mitochondria and oxidative stress. Exp
Gerontol 2009;44:485–92. doi:10.1016/j.exger.2009.05.004.
[32] Petrakis S, Rasko T, Mates L, Ivics Z, Izsvak Z,
Kouzi-Koliakou K, et al. Gateway-compatible transposon
vector to genetically modify human embryonic kidney and
adipose-derived stromal cells. Biotechnol J 2012;7:891–7.
doi:10.1002/biot.201100471.
Telomerase protects against genomic instability 819
[33] Tsagias N, Koliakos I, Karagiannis V, Eleftheriadou M,
Koliakos GG. Isolation of mesenchymal stem cells using
the total length of umbilical cord for transplantation
purposes. Transfus Med 2011;21:253–61. doi:10.1111/
j.1365-3148.2011.01076.x.
[34] Mates L, Chuah MK, Belay E, Jerchow B, Manoj N,
Acosta-Sanchez A, et al. Molecular evolution of a novel
hyperactive Sleeping Beauty transposase enables robust stable
gene transfer in vertebrates. Nat Genet 2009;41:753–61.
doi:10.1038/ng.343.
[35] Tsolou A, Nelson G, Trachana V, Chondrogianni N, Saretzki
G, Von Zglinicki T, et al. The 19S proteasome subunit Rpn7
stabilizes DNA damage foci upon genotoxic insult. IUBMB
Life 2012;64:432–42. doi:10.1002/iub.1018.
[36] Kapetanou M, Chondrogianni N, Petrakis S, Koliakos G,
Gonos ES. Proteasome activation enhances stemness and
lifespan of human mesenchymal stem cells. Free Radic Biol
Med 2017;103:226–35. doi:10.1016/j.freeradbiomed.2016
.12.035.
[37] Estrada JC, Torres Y, Benguría A, Dopazo A, Roche E,
Carrera-Quintanar L, et al. Human mesenchymal stem
cell-replicative senescence and oxidative stress are closely linked
to aneuploidy. Cell Death Dis 2013;4:e691. doi:10.1038/
cddis.2013.211.
[38] Chen JH, Hales CN, Ozanne SE. DNA damage, cellular
senescence and organismal ageing: causal or correlative?
Nucleic Acids Res 2007;35:7417–28. doi:10.1093/nar/
gkm681.
[39] Eccles LJ, O’Neill P, Lomax ME. Delayed repair of radiation
induced clustered DNA damage: friend or foe? Mutat Res
2011;711:134–41. doi:10.1016/j.mrfmmm.2010.11.003.
[40] Ohshima S. Abnormal mitosis in hypertetraploid cells causes
aberrant nuclear morphology in association with H2O2-
induced premature senescence. Cytometry A 2008;73:808–15.
doi:10.1002/cyto.a.20604.
[41] Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase
multigene family: a comparison of the CuZn-SOD (SOD1),
Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures,
evolution, and expression. Free Radic Biol Med 2002;33:337–
49. doi:10.1016/S0891-5849(02)00905-X.
[42] Radi R, Turrens JF, Chang LY, Bush KM, Crapo JD, Freeman
BA. Detection of catalase in rat heart mitochondria. J Biol
Chem 1991;266:22028–34.
[43] Allen RC, Tresini M, Keogh BP, Doggett DL, Cristofalo VJ.
Differences in electron transport potential, antioxidant
defenses, and oxidant generation in young and senescent fetal
lung fibroblasts (WI-38). J Cell Physiol 1999;180:114–22.
doi:10.1002/(SICI)1097-4652(199907)180:1<114::AID-
JCP13>3.0.CO;2-0.
[44] Trachana V, Van Wely KHM, Guerrero AA, Fütterer A,
Martinez-A C. Dido disruption leads to centrosome
amplification and mitotic checkpoint defects compromising
chromosome stability. Proc Natl Acad Sci USA 2007;104:
2691–6. doi:10.1073/pnas.0611132104.
[45] D’Angiolella V, Santarpia C, Grieco D. Oxidative stress
overrides the spindle checkpoint. Cell Cycle 2007;6:576–9.
doi:10.4161/cc.6.5.3934.
[46] Ikawa-Yoshida A, Ando K, Oki E, Saeki H, Kumashiro R,
Taketani K, et al. Contribution of BubR1 to oxidative stress-
induced aneuploidy in p53-deficient cells. Cancer Med
2013;2:447–56. doi:10.1002/cam4.101.
[47] Saretzki G. Extra-telomeric functions of human telomerase:
cancer, mitochondria and oxidative stress. Curr Pharm Des
2014;20:6386–403. doi:10.2174/1381612820666140630
095606.
[48] Bocker W, Yin Z, Drosse I, Haasters F, Rossmann O, Wierer
M, et al. Introducing a single-cell-derived human mesenchymal
stem cell line expressing hTERT after lentiviral gene transfer.
J Cell Mol Med 2008;12:1347–59. doi:10.1111/j.1582
-4934.2008.00299.x.
[49] Madonna R, Taylor DA, Geng YJ, De Caterina R, Shelat H,
Perin EC, et al. Transplantation of mesenchymal cells
rejuvenated by the overexpression of telomerase and myocardin
promotes revascularization and tissue repair in a murine
model of hindlimb ischemia. Circ Res 2013;113:902–14.
doi:10.1161/CIRCRESAHA.113.301690.
[50] Kang SK, Putnam L, Dufour J, Ylostalo J, Jung JS, Bunnell
BA. Expression of telomerase extends the lifespan and
enhances osteogenic differentiation of adipose tissue-derived
stromal cells. Stem Cells 2004;22:1356–72. doi:10.1634/
stemcells.2004-0023.
[51] Sharma GG, Gupta A, Wang H, Scherthan H, Dhar S, Gandhi
V, et al. hTERT associates with human telomeres and
enhances genomic stability and DNA repair. Oncogene
2003;22:131–46. doi:10.1038/sj.onc.1206063.
[52] Shin KH, Kang MK, Dicterow E, Kameta A, Baluda MA,
Park NH. Introduction of human telomerase reverse
transcriptase to normal human fibroblasts enhances DNA
repair capacity. Clin Cancer Res 2004;10:2551–60.
[53] Indran IR, Hande MP, Pervaiz S. hTERT overexpression
alleviates intracellular ROS production, improves mitochondrial
function, and inhibits ROS-mediated apoptosis in cancer cells.
Cancer Res 2010;71:266–76. doi:10.1158/0008-5472.CAN-
10-1588.
[54] Caradonna F, Mauro M. Role of the antioxidant defence
system and telomerase in arsenic-induced genomic instability.
Mutagenesis 2016;31:661–7. doi:10.1093/mutage/gew034.
[55] Perez-Rivero G, Ruiz-Torres MP, Diez-Marques ML, Canela
A, Lopez-Novoa JM, Rodriguez-Puyol M, et al. Telomerase
deficiency promotes oxidative stress by reducing catalase
activity. Free Radic Biol Med 2008;45:1243–51. doi:10.1016/
j.freeradbiomed.2008.07.017.
[56] Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH,
Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell
in vitro. BMC Cell Biol 2006;7:14. doi:10.1186/1471-2121
-7-14.
[57] Ju Z, Jiang H, Jaworski M, Rathinam C, Gompf A, Klein C,
et al. Telomere dysfunction induces environmental alterations
limiting hematopoietic stem cell function and engraftment.
Nat Med 2007;13:742–7. doi:10.1038/nm1578.
[58] Mangerini R, Lanino E, Terranova P, Faraci M, Pistillo MP,
Gaetani GF, et al. Telomere length of donors influences
granulocyte recovery in children after hematopoietic stem cell
transplantation. Ann Hematol 2009;88:1029–31. doi:10.1007/
s00277-009-0712-z.
[59] Nakahara H, Misawa H, Hayashi T, Kondo E, Yuasa T,
Kubota Y, et al. Bone repair by transplantation of
hTERT-immortalized human mesenchymal stem cells in
mice. Transplantation 2009;88:346–53. doi:10.1097/TP
.0b013e3181ae5ba2.
[60] Tichon A, Eitan E, Kurkalli BG, Braiman A, Gazit A, Slavin
S, et al. Oxidative stress protection by novel telomerase
activators in mesenchymal stem cells derived from healthy and
diseased individuals. Curr Mol Med 2013;13:1010–22.
Appendix: Supplementary material
Supplementary data to this article can be found online
at doi:10.1016/j.jcyt.2017.03.078.
820 V.Trachana et al.

More Related Content

What's hot

The moringa antioxidant assortment
The moringa antioxidant assortmentThe moringa antioxidant assortment
The moringa antioxidant assortmentfb.com/malaysiamlm
 
A putative mesenchymal stem cells population isolated from adult human testes
A putative mesenchymal stem cells population isolated from adult human testesA putative mesenchymal stem cells population isolated from adult human testes
A putative mesenchymal stem cells population isolated from adult human testes◂ Justin (M) Gaines ▸
 
Thesis Poster
Thesis PosterThesis Poster
Thesis PosterTravis Tu
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordComprehensiveBiologi
 
Heraud Et Al. S C R
Heraud Et Al.  S C RHeraud Et Al.  S C R
Heraud Et Al. S C Ruvperson
 
Gari et al bmc medical genetics
Gari et al bmc medical geneticsGari et al bmc medical genetics
Gari et al bmc medical geneticsTariq Mohammed
 
Antinuclear antobdy testing
Antinuclear antobdy testingAntinuclear antobdy testing
Antinuclear antobdy testingLAB IDEA
 
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-Bhaswati Sarcar
 
A Primer to Bioinformatics: 29 September 2017
A Primer to Bioinformatics: 29 September 2017A Primer to Bioinformatics: 29 September 2017
A Primer to Bioinformatics: 29 September 2017DocSoc2017
 
Neuromics presentation november 2021
Neuromics presentation november 2021Neuromics presentation november 2021
Neuromics presentation november 2021Pete Shuster
 
Terzic and Maxon et al., 2016
Terzic and Maxon et al., 2016Terzic and Maxon et al., 2016
Terzic and Maxon et al., 2016Jake Maxon
 
Insects as Model in Aging Research
Insects as Model in Aging ResearchInsects as Model in Aging Research
Insects as Model in Aging ResearchBharati Singh
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsComprehensiveBiologi
 
Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Shashaanka Ashili
 
Cellular Senescence Paper
Cellular Senescence PaperCellular Senescence Paper
Cellular Senescence PaperStephen Liu
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)ComprehensiveBiologi
 

What's hot (20)

The moringa antioxidant assortment
The moringa antioxidant assortmentThe moringa antioxidant assortment
The moringa antioxidant assortment
 
A putative mesenchymal stem cells population isolated from adult human testes
A putative mesenchymal stem cells population isolated from adult human testesA putative mesenchymal stem cells population isolated from adult human testes
A putative mesenchymal stem cells population isolated from adult human testes
 
Thesis Poster
Thesis PosterThesis Poster
Thesis Poster
 
Peripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcordPeripheral neuropathyumbilicalcord
Peripheral neuropathyumbilicalcord
 
Heraud Et Al. S C R
Heraud Et Al.  S C RHeraud Et Al.  S C R
Heraud Et Al. S C R
 
Fphys 07-00180
Fphys 07-00180Fphys 07-00180
Fphys 07-00180
 
Gari et al bmc medical genetics
Gari et al bmc medical geneticsGari et al bmc medical genetics
Gari et al bmc medical genetics
 
nihms419319
nihms419319nihms419319
nihms419319
 
Antinuclear antobdy testing
Antinuclear antobdy testingAntinuclear antobdy testing
Antinuclear antobdy testing
 
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-
Sarcar B et al.,-Mol Cancer Ther-2011-Sarcar-
 
A Primer to Bioinformatics: 29 September 2017
A Primer to Bioinformatics: 29 September 2017A Primer to Bioinformatics: 29 September 2017
A Primer to Bioinformatics: 29 September 2017
 
Neuromics presentation november 2021
Neuromics presentation november 2021Neuromics presentation november 2021
Neuromics presentation november 2021
 
Terzic and Maxon et al., 2016
Terzic and Maxon et al., 2016Terzic and Maxon et al., 2016
Terzic and Maxon et al., 2016
 
Vascular_Poster
Vascular_PosterVascular_Poster
Vascular_Poster
 
Insects as Model in Aging Research
Insects as Model in Aging ResearchInsects as Model in Aging Research
Insects as Model in Aging Research
 
Excellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterialsExcellent extracellular matrix_inspired_biomaterials
Excellent extracellular matrix_inspired_biomaterials
 
Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...Method for physiologic phenotype characterization at the single-cell level in...
Method for physiologic phenotype characterization at the single-cell level in...
 
Whartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_mWhartons jelly ms_cs_a4_m
Whartons jelly ms_cs_a4_m
 
Cellular Senescence Paper
Cellular Senescence PaperCellular Senescence Paper
Cellular Senescence Paper
 
Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)Umbilical cord wj_viable_msc.watson2015_(1)
Umbilical cord wj_viable_msc.watson2015_(1)
 

Similar to 16 cytotherapy 2017

Aging and the telomere connection dr. Jerry w. Shay - april 2012
Aging and the telomere connection   dr. Jerry w. Shay - april 2012Aging and the telomere connection   dr. Jerry w. Shay - april 2012
Aging and the telomere connection dr. Jerry w. Shay - april 2012Life Length
 
Biotechnology and ageing. pptx.
Biotechnology and ageing.           pptx.Biotechnology and ageing.           pptx.
Biotechnology and ageing. pptx.AbhishekUppal18
 
Journal of Stem Cells Research, Reviews & Reports
Journal of Stem Cells Research, Reviews & ReportsJournal of Stem Cells Research, Reviews & Reports
Journal of Stem Cells Research, Reviews & ReportsAustin Publishing Group
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicineSpringer
 
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...Minia university, Faculty of Medicine
 
Wojciechowski stevens 2021_a_dynamic_duo
Wojciechowski stevens 2021_a_dynamic_duoWojciechowski stevens 2021_a_dynamic_duo
Wojciechowski stevens 2021_a_dynamic_duoFranProfesor1
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionAnkita-rastogi
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionAnkita-rastogi
 
Chancellor's Award - Nathan Jayne - Project Description
Chancellor's Award - Nathan Jayne - Project DescriptionChancellor's Award - Nathan Jayne - Project Description
Chancellor's Award - Nathan Jayne - Project DescriptionNathan Jayne
 
Biological Basis of Aging
Biological Basis of AgingBiological Basis of Aging
Biological Basis of AgingFarhad Zargari
 
Biological and molecular Characterization of a canine hemangiosarcoma-derived...
Biological and molecular Characterization of a canine hemangiosarcoma-derived...Biological and molecular Characterization of a canine hemangiosarcoma-derived...
Biological and molecular Characterization of a canine hemangiosarcoma-derived...Rodrigo Shamed Cedillo Flores
 

Similar to 16 cytotherapy 2017 (20)

Aging and the telomere connection dr. Jerry w. Shay - april 2012
Aging and the telomere connection   dr. Jerry w. Shay - april 2012Aging and the telomere connection   dr. Jerry w. Shay - april 2012
Aging and the telomere connection dr. Jerry w. Shay - april 2012
 
Biotechnology and ageing. pptx.
Biotechnology and ageing.           pptx.Biotechnology and ageing.           pptx.
Biotechnology and ageing. pptx.
 
Journal of Stem Cells Research, Reviews & Reports
Journal of Stem Cells Research, Reviews & ReportsJournal of Stem Cells Research, Reviews & Reports
Journal of Stem Cells Research, Reviews & Reports
 
Biology of ageing
Biology of ageingBiology of ageing
Biology of ageing
 
Regenerative medicine
Regenerative medicineRegenerative medicine
Regenerative medicine
 
BBL2
BBL2BBL2
BBL2
 
Trehalose_Shhyam Moorthy
Trehalose_Shhyam MoorthyTrehalose_Shhyam Moorthy
Trehalose_Shhyam Moorthy
 
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...
3. possible protective-effect-of-bms cs -against-the-remote-liver-injury-indu...
 
Wojciechowski stevens 2021_a_dynamic_duo
Wojciechowski stevens 2021_a_dynamic_duoWojciechowski stevens 2021_a_dynamic_duo
Wojciechowski stevens 2021_a_dynamic_duo
 
Stem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder DysfunctionStem Cell Therapy in Bladder Dysfunction
Stem Cell Therapy in Bladder Dysfunction
 
Stem cells for Bladder Dysfunction
Stem cells for Bladder DysfunctionStem cells for Bladder Dysfunction
Stem cells for Bladder Dysfunction
 
Chancellor's Award - Nathan Jayne - Project Description
Chancellor's Award - Nathan Jayne - Project DescriptionChancellor's Award - Nathan Jayne - Project Description
Chancellor's Award - Nathan Jayne - Project Description
 
Editing genome
Editing genomeEditing genome
Editing genome
 
Editing genome
Editing genome Editing genome
Editing genome
 
Editing genome
Editing genomeEditing genome
Editing genome
 
Editing genome
Editing genomeEditing genome
Editing genome
 
Editing genome
Editing genomeEditing genome
Editing genome
 
Editing genome
Editing genomeEditing genome
Editing genome
 
Biological Basis of Aging
Biological Basis of AgingBiological Basis of Aging
Biological Basis of Aging
 
Biological and molecular Characterization of a canine hemangiosarcoma-derived...
Biological and molecular Characterization of a canine hemangiosarcoma-derived...Biological and molecular Characterization of a canine hemangiosarcoma-derived...
Biological and molecular Characterization of a canine hemangiosarcoma-derived...
 

Recently uploaded

Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...CALL GIRLS
 
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipur
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls JaipurCall Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipur
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipurparulsinha
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Miss joya
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Serviceparulsinha
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...narwatsonia7
 
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...narwatsonia7
 
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...Garima Khatri
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...narwatsonia7
 
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...Miss joya
 
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girlsnehamumbai
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Miss joya
 
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls ServiceCall Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Servicenarwatsonia7
 
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...Miss joya
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliRewAs ALI
 
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Service
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls ServiceKesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Service
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Servicemakika9823
 
Call Girl Chennai Indira 9907093804 Independent Call Girls Service Chennai
Call Girl Chennai Indira 9907093804 Independent Call Girls Service ChennaiCall Girl Chennai Indira 9907093804 Independent Call Girls Service Chennai
Call Girl Chennai Indira 9907093804 Independent Call Girls Service ChennaiNehru place Escorts
 
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls ServiceCALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls ServiceMiss joya
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalorenarwatsonia7
 

Recently uploaded (20)

Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
 
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipur
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls JaipurCall Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipur
Call Girls Service Jaipur Grishma WhatsApp ❤8445551418 VIP Call Girls Jaipur
 
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
Low Rate Call Girls Pune Esha 9907093804 Short 1500 Night 6000 Best call girl...
 
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCREscort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
Escort Service Call Girls In Sarita Vihar,, 99530°56974 Delhi NCR
 
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort ServiceCall Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
Call Girls Service In Shyam Nagar Whatsapp 8445551418 Independent Escort Service
 
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
Russian Call Girl Brookfield - 7001305949 Escorts Service 50% Off with Cash O...
 
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
 
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...
VIP Mumbai Call Girls Hiranandani Gardens Just Call 9920874524 with A/C Room ...
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
 
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...
VIP Call Girls Pune Vrinda 9907093804 Short 1500 Night 6000 Best call girls S...
 
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Yelahanka Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy GirlsCall Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
Call Girls In Andheri East Call 9920874524 Book Hot And Sexy Girls
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
 
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls ServiceCall Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
 
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...
VIP Call Girls Pune Vani 9907093804 Short 1500 Night 6000 Best call girls Ser...
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas Ali
 
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Service
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls ServiceKesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Service
Kesar Bagh Call Girl Price 9548273370 , Lucknow Call Girls Service
 
Call Girl Chennai Indira 9907093804 Independent Call Girls Service Chennai
Call Girl Chennai Indira 9907093804 Independent Call Girls Service ChennaiCall Girl Chennai Indira 9907093804 Independent Call Girls Service Chennai
Call Girl Chennai Indira 9907093804 Independent Call Girls Service Chennai
 
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls ServiceCALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune)  Girls Service
CALL ON ➥9907093804 🔝 Call Girls Baramati ( Pune) Girls Service
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
 

16 cytotherapy 2017

  • 1. Human mesenchymal stem cells with enhanced telomerase activity acquire resistance against oxidative stress-induced genomic damage VARVARA TRACHANA1 , SPYROS PETRAKIS2 , ZISIS FOTIADIS3 , EVANGELIA K. SISKA2,4 , VASILEIOS BALIS3 , EFSTATHIOS S. GONOS5 , MARTHA KALOYIANNI3 & GEORGE KOLIAKOS2,4 1 Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece, 2 Biohellenika Biotechnology Company, 57001 Thessaloniki, Greece, 3 Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece, 4 Department of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece, and 5 National Hellenic Research Foundation, 48Vas. Konstantinou Str, 11635 Athens, Greece Abstract Background. Human mesenchymal stem cells (MSC) are important tools for several cell-based therapies. However, their use in such therapies requires in vitro expansion during which MSCs quickly reach replicative senescence. Replicative se- nescence has been linked to macromolecular damage, and especially oxidative stress-induced DNA damage. Recent studies on the other hand, have implicated telomerase in the cellular response to oxidative damage, suggesting that telomerase has a telomere-length independent function that promotes survival. Methods. Here, we studied the DNA damage accumula- tion and repair during in vitro expansion as well as after acute external oxidative exposure of control MSCs and MSCs that overexpress the catalytic subunit of telomerase (hTERT MSCs). Results. We showed that hTERT MSCs at high passages have a significant lower percentage of DNA lesions as compared to control cells of the same passages. Additionally, less damage was accumulated due to external oxidative insult in the nuclei of hTERT overexpressing cells as compared to the control cells. Moreover, we demonstrated that oxidative stress leads to diverse nucleus malformations, such as multillobular nuclei or donut-shaped nuclei, in the control cells whereas hTERT MSCs showed significant resistance to the formation of such defects. Finally, hTERT MSCs were found to possess higher activities of the basic antioxidant enzymes, superox- ide dismutase and catalase, than control MSCs. Discussion. On the basis of these results, we propose that hTERT enhancement confers resistance to genomic damage due to the amelioration of the cell’s basic antioxidant machinery. Key Words: mesenchymal stem cells, telomerase, oxidative stress, genomic damage Abbreviations: MSCs, human Mesenchymal Stem Sells; ASCs, Adipose Stem Cells; WJ, Wharton’s Jelly; hTERT, catalytic subunit of human telomerase; ROS, Reactive Oxygen Species; SAC, Spindle Assembly Checkpoint. Introduction Several studies have demonstrated the therapeutic po- tential of human mesenchymal stem cells (MSCs).Their self-renewal ability and multilineage differentiation (stemness) have made them a rather attractive candi- date for cell based therapies. In fact, clinical trials are currently taking place using MSCs as the main ther- apeutic agent for different human diseases [1]. Even though MSCs can be easily isolated from bone marrow, adipose tissue, the umbilical cord and other tissues, their expansion in vitro is not free of concerns [2,3]. It is ac- tually well documented that MSCs reachreplicative senescence rather fast in culture conditions [4,5] which impairs their regenerative potential [6,7]. Replicative senescence is a common feature to all somatic cells and most stem cells; after a period of rig- orous proliferation the rate of cell division drops until cells reach a state where they are still alive but unable to divide further.The phenomenon is accompanied by a number of characteristic changes, such as increase in size and flattening, nuclear changes as well as several changes in gene and protein expression patterns [8,9]. The telomere shortening hypothesis is currently the best explanation for the limited proliferation potential of the cells.Telomeres are hexanucleotide repeats which extend Correspondence: Varvara Trachana, PhD, Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece. E-mail: vtrachana@med.uth.gr (Received 20 February 2017; accepted 25 March 2017) ISSN 1465-3249 Copyright © 2017 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.jcyt.2017.03.078 Cytotherapy, 2017; 19: 808–820
  • 2. over several thousand base pairs (bp) in length, forming functional “caps” at the end of the chromosomes.These repeats due to the “end-replication-problem” are sub- jected to erosion with each round of DNA replication. Telomeric DNA is synthesized by a specialized reverse transcriptase called telomerase [10].When telomerase is not present or is expressed at very low levels, pro- gressive shortening of the telomeres is taking place in every cell division until the telomeres reach a crucial length. At this point cellular senescence is triggered by activating the DNA damage checkpoint that prevents cells from further cycling [11]. For MSCs the proliferation limit determined by the length of their telomeres bears another obstacle: the age of the donor of the cells, since it has been shown to be correlated with the initial telomeres length, and therefore the time that MSCs can be main- tained in culture [12]. The initial telomere length of the donor’s MSCs could therefore be a limiting factor of the MSCs therapeutic potential since it will affect the survival and integration ability of the trans- planted cells to the adult tissue [13–15]. In fact there is increasing evidence that the age of the donor affects several properties of MSCs [16]. On the other hand, it has been demonstrated that the erosion of the telomeres is not only due to the way DNA is replicated, but it can also be accelerated or even induced by environmental factors, such as the oxida- tive stress (OS) [17,18]. Different human cells subjected to long-term exposure to mild OS have shown to undergo accelerated telomere erosion that triggers pre- mature senescence [17]. This phenomenon has been collectively termed ‘stress induced premature senes- cence’ (SIPS) [19]. It was also reported that hydrogen peroxide (H2O2) can cause predominant DNA damage at the 5΄ site of 5′-GGG-3′ in the telomere sequence. Furthermore, H2O2 induced the formation of 8-oxo- 7,8-dihydro-2′-deoxyguanosine (8-oxo-dG), an oxidized derivative of deoxyguanosine and major product of DNA oxidation [20], in telomere sequences more efficient- ly than that in non-telomere sequences [21]. Different groups, however, have reported that SIPS could also be independent of telomeric damage [22]. Despite the controversies on the matter, it is widely acknowl- edged that the progressive oxidative damage to the macromolecules, and especially to DNA, can induce cellular senescence [23]. Even though many different groups have studied the involvement of OS in MSCs senescence in culture [24–26], the exact underlying mo- lecular mechanism remains unclear. Interestingly, it has been recently reported that telomerase has also non-canonical or extratelomeric functions, that are involved in processes such resis- tance to stress and especially to oxidative stress. In fact, Borras et al. [27] showed that glutathione, a physio- logical antioxidant regulates telomerase activity, highlighting therefore the interplay between telomerase and the cellular oxidative status. Moreover, emerg- ing data demonstrate that telomerase could be directly involved in the reduction of reactive oxygen species (ROS) generation and therefore decreased oxidative DNA damage [28–31]. In order to investigate the antioxidant properties of telomerase, and their implication in the DNA damage driven decline in the cellular proliferation po- tential, in the present study we analyzed the oxidative stress-induced DNA damage response of MSCs ge- netically modified with the catalytic subunit of human telomerase (hTERT). In the analysis we included adipose derived MSCs (ASCs) from adult individu- als as well as umbilical cord’s Wharton Jelly derived cells (WJ-MSCs), in an attempt to determine possi- ble differences that could be attributed to the donor’s initial telomeres length. We believe that this ap- proach provides insights regarding the recently proposed antioxidant role for telomerase, which could contribute in developing strategies that will over- come the senescence-associated impairment of the regenerative potential of MSCs. Materials and methods Construction of the hTERT transposon An entry clone encoding the human telomerase reverse transcriptase catalytic subunit (hTERT), (GeneID: 7015, clone ID: IOH36343) was shuttled into a pT2- CAGGS-EYFP-GW plasmid [32] using LR clonase (Life Technologies), according to manufacturer’s in- structions. The reaction mixture was used for transformation of Mach1 E. coli cells and transformants were selected on agar plates supplemented with 100 µg/ ml Ampicillin. Plasmid isolation was performed using NucleoSpin plasmid isolation kit (Macherey-Nagel). The identity of pT2-CAGGS-EYFP-hTERT plasmid was verified by BsrGI restriction digestion. Generation of hTERT mesenchymal stem cells MSCs were enzymatically isolated from adipose tissue (ASCs) of healthy donors (four different individu- als, n = 4, age range of 49–60 years) by lipoaspiration. Informed consent was given for the collection of samples from all donors and the collection was per- formed in accordance with established guidelines. For the isolation of ASCs from the adipose tissue, tissues were treated with collagenase (2.7 mg/ml) and hyal- uronidase (0.7 mg/ml) solution for 1h at 37°C followed by incubation with trypsin (2.5%). Cell suspension was diluted with equal volume of PBS, passed through a sterile 0.2 µm filter and centrifuged at 500g for 30 min at room temperature (RT).We also isolated and used MSCs from theWharton Jelly of umbilical cords (WJ Telomerase protects against genomic instability 809
  • 3. MSCs) from term-gestation newborns after birth (two different individuals, n = 2), having obtained consent from the parents. The isolating method used in our laboratory was previously described [33]. After isolation ASCs and WJ MSCs were diluted in Dulbecco’s modified Eagle’s medium (DMEM, Sigma-Aldrich, Saint Louis, MO, USA) supple- mented with 10% fetal bovine serum (FBS, Sigma- Aldrich), penicillin (100 IU/mL) and streptomycin (100 µg/mL) and placed in a 6-well plates. 2x105 cells at passage 4 were grown to 70–80% confluency and then nucleofected with 7.5 µg DNA encoding Sleep- ing Beauty 100X transposase [34], pT2-CAGGS- YFP-hTERT and pT2-SV40-neoR transposon plasmids (1:8:1 ratio) using a Nucleofector device (Lonzabio). hTERT ASCs and hTERTWJ MSCs were selected with 100 µg/ml G418. All following analy- ses were performed in four individual cultures of ASCs (four control ASCs and four hTERT ASCs) and in two individual cultures ofWJ MSCs (two controlWJ- MSCs and two hTERT WJ-MSCs. Culture conditions Cells were cultured under standard cell-culture in DMEM (supplemented with 10% FBS, penicillin (100 IU/mL) and streptomycin (100 µg/mL)) in a hu- midified incubator set to 37°C and 5% CO2 and 20% O2. Medium was changed twice a week and cells were passed when reached confluency. Cell number was de- termined in duplicates using an hemocytometer. For exposure to hydrogen peroxide (H2O2), 200.000 cells (hTERT or control MSCs) were seeded per well on a 6 well plate and at 65–70% confluence cells were exposed to 300µΜ H2O2 for 30 min in serum-free medium. The medium was then replaced with fresh complete medium and cells were normally cultured for 48 h (recovery time) in order to recover from the stress. Antibodies Mouse monoclonal anti-γH2AX (05–636; phosphor S139, clone JBW301), and anti-53BP1 (clone BP13,) were from Millipore (MA, USA). Anti- 8-hydroxy- 2’- deoxyguanosine (8-oxo-dG, Clone 2E2), was purchased from Trevigen. For the western blots anti- hTERT (NB120-32020) was from Novus and anti- β-actin antibody (sc-47778) from Santa Cruz. Secondary antibodies for confocal microscopy, Alexa Fluor 594 anti-mouse IgG (A11005) and AlexaFluor 488 anti-mouse IgG (A11001 were obtained from Mo- lecular Probes (Invitrogen). qRT-PCR Total RNA was purified from hTERT or control MSCs using the NucleoSpin RNA kit (Macherey-Nagel), according to manufacturer’s instructions. cDNA gen- eration and qRT-PCR reactions were performed using the KAPA SYBR Fast 1-step Green qRT-PCR kit (KAPA Biosystems) in a Rotor-Gene 6000 operat- ing system. Each analysis was performed in triplicates. The correct size of amplified qRT-PCR products was verified by electrophoresis in a 2% agarose gel. Western blot analysis hTERT or control MSCs were lysed in 1% SDS in PBS containing Protease Inhibitors Cocktail Set III (Calbiochem) and Benzonase (Novagen). Protein con- centrations were determined by the Bradford method with bovine serum albumin as standard (Bio-Rad Laboratories, CA, USA). Samples(<20 µg total protein) were analyzed by SDS-PAGE and transferred onto ni- trocellulose membranes. Membranes were blocked with 5% weight per volume (w/v) non-fat dry milk in PBS/ 0.1%Tween20 probed with the appropriate antibodies. Secondary antibodies conjugated with alkaline- phosphatase were detected using nitro blue tetrazolium chloride/5-bromo-4-chloro-3-indolyl phosphate p-toluidine salt (NBT/BCIP) substrate (Applichem). Equal protein loading was verified by reprobing each membrane with the antibody against β-actin. Quantification of Telomerase activity Real Time-PCR based telomeric repeat amplification protocol (TRAP) assay was performed according to the manufacturer’s protocol (TeloTAGGGTelomerase PCR ELIZAPLUS-Roche Applied Science, Indianapolis, IN, USA). In brief, 2 × 105 hTERT or control MSCs were lysed in 200 µl lysis buffer and the cell lysate was centrifuged at 16,000 g for 20 min at 4°C. Cell extract (0.5 µg of total protein) was used for PCR reaction. The amplification product (3 µl) was transferred for the hybridization, and the ELISA assay was performed in triplicates, in which two served as controls (one con- tained a negative control, and a second containing the high telomerase activity positive control). Telomerase activity of each sample was calculated as a percentage of the relative telomerase activity of the positive control cells. Flow cytometry For phenotypic characterization, hTERT and control MSCs were stained with phycoerythrin (PE)- conjugated antibodies against hemopoietic (CD34, 45) or mesenchymal (CD29, 73, 90, 105 and 146) stem cell markers. Unstained cells or cells stained with IgG isotype antibodies were used as negative controls. Cells were measured in a Cytomics FC500 flow cytom- eter (Beckman Coulter) using the CXP2.2 software. 810 V.Trachana et al.
  • 4. Multilineage differentiation For osteogenic or adipogenic differentiation, hTERT and control MSCs were grown to 90% confluency and cultured for 28 days either in StemPro Osteogenesis or StemPro Adipogenesis medium (Life Technolo- gies). Differentiation of cells into osteocytes or adipocytes was monitored by Alizarin Red or Oil Red staining, respectively. Immunofluorescence Immunofluorescence experiments were performed as previously described [35]. Briefly, MSCs were grown on coverslips and subjected to the above described oxi- dative treatment or left to reach the desired passage and then fixed in 4% paraformaldehyde. Fixed samples were incubated with primary antibodies of interest (γ- H2AX, 53BP1 or 8-oxo-dG) and the appropriate secondary antibodies. Coverslips were embedded in 10 µl of Vectashield mounting medium for fluores- cence with 4,6-diamidino-2-phenylindole (DAPI,Vector Laboratories, CA, USA) to visualize the nuclei, and analyzed on ZEISS Axio Imager Z2 fluorescent microscope. Images were captured on confocal microscope ZEISS LSM780 and ZEN 2011 program. For calculations of damaged nuclei at least 5 ran- domly selected fields were analyzed for each culture condition. Cells with >5 γ-H2AX, 53BP1 or 8-oxo- dG stained foci in their nuclei were counted as positive for DNA damage by a single observer blinded to treat- ment regimen. Measurement of antioxidant enzymes activity Total superoxide dismutase (SOD) and catalase ac- tivities were measured in MSCs cell lysates using commercially available kits (Superoxide Dismutase Assay Kit 706002, Catalase Assay Kit 707002 Cayman Chemical, Ann Arbor, MI) according to the manu- facturer’s recommendations. Statistical analysis For the statistical analysis of the results the GraphPad Prism 5 software (GraphPad Software, San Diego, Cal- ifornia USA) was used. P values less than 0.05 were considered significant and the significance is ex- pressed with asterisks: * = P < 0.05; ** = P < 0.01; *** = P < 0.001. Results are reported as mean ± stan- dard error (means ± S.Ε.) unless otherwise stated. Results Genetically modified MSCs have increased expression of hTERT and enhanced telomerase activity Ectopic expression of hTERT in hTERT ASCs was confirmed by qRT-PCR analysis (Figure 1A).The iden- tity of amplified hTERT and actin beta (ACTB) housekeeping gene fragments was verified by agarose electrophoresis (Figure 1B). Production of recombi- nant yellow fluorescent protein (YFP)-TERT was evaluated by Western blotting. A protein band with a molecular weight of ~160 kDa was detected in hTERT ASCs corresponding toYFP-TERT, whereas endog- enous hTERT was detected in all samples at the expected molecular weight (127 kDa) (Figure 1C). These results verify successful overexpression of hTERT in ASCs. hTERT WJ-MSCs showed similarly increased mRNA levels of the telomerase catalytic subunit and production of the recombinant YFP-TERT protein (Supplementary Figure S1A and B respectively). Finally, realTime-PCR basedTRAP assay (Roche Applied Science) was performed according to the man- ufacturer’s protocol.The assay confirmed that ASCs genetically modified with the hTERT (ASCs1-ASCS4) Figure 1. Genetically modified ASCs have increased messenger RNA (mRNA) levels of hTERT and produce recombinant YFP-TERT protein. (A) mRNA levels of control and hTERT-overexpressing ASCs normalized to housekeeping β-actin (ACTB) gene. Values showed are the means ± SE. *P < 0.05 versus control ASCs. (B) qRT-PCR–amplified fragments analyzed in a 2% agarose electrophoresis. (C)Western blot analysis for hTERT in control and hTERT-ASCs.The analysis was performed in all control and hTERT ASC individual cultures and a characteristic image is demonstrated. Molecular weight markers are run in the middle lane. β-actin was used as a loading control. Telomerase protects against genomic instability 811
  • 5. have increased telomerase activity when compared to the control cells (Figure 2). MSCs overexpressing hTERT retain their mesenchymal properties hTERT MSCs derived from adipose tissue were further characterized in order to verify that the genetic mod- ification does not affect their mesenchymal properties. hTERT ASCs were plastic-adherent and had a similar fibroblastic morphology as control cells. Flow cytometry of hTERT ASCs showed that these cells do not express the hemopoietic CD34 marker, whereas they express typical mesenchymal stem cell CD markers 29, 73, 90, 105 and 146 at similar levels to control ASCs (Table I). Similarly, hTERTWJ-MSCs do not express the CD34 marker and express the typical mesenchy- mal CD29 and CD105 markers (Supplementary Table SI). The multilineage differentiation potential of hTERT ASCs was also assessed. Cells were differentiated towards adipocytes and osteocytes, as indicated by Oil Red and Alizarin Red staining, respectively.At early pas- sages (p ≤ 8) both control as well as genetically modified ASCs were able to differentiate towards both adipocytes and osteocytes (data not shown). As shown in Figure 3, control ASCs at passage 10 were able to differentiate towards adipocytes but not osteocytes, whereas hTERT ASCs of the same passage retained their osteogenic dif- ferentiation capacity (Figure 3A, B). Moreover, as recently demonstrated (in a paral- lel work done by members of our team [36],) control WJ-MSCs maintain their differentiation ability towards adipocytes and osteocytes even at high passage (passage 40). Here, we demonstrate that hTERT overexpression did not affect this differentiation ability ofWJ-MSCs, as indicated in Figure S1C. The above data prove that the genetic modifica- tion did not alter the mesenchymal properties of adipose derived or WJ-derived cells. It also implies that the overexpression of hTERT may contribute in retain- ing their mesenchymal properties for longer (at higher passages). hTERT overexpression offers protection against DNA damage accumulation during long-term culture MSCs were kept in culture until their proliferation rate dropped to less than 2 cumulative population doublings per 4 weeks. At this stage control MSCs had the char- acteristic senescent-like morphology (large, flattened vacuolated cells), suggesting that they have reached their proliferation limit. In accordance with previously re- ported data [37], control ASCs reached this state at passage 20 ± 2.3, whereas hTERT ASCs at did not seem to enter this state even at high passages (passage > 30). Similarly, control WJ-MSCs reached their prolifera- tion limit at passage 22 ± 1.3, whereas hTERT overexpressing WJ-MSCs did not show any reduction in their duplication time until at least passage 30. Our observations, together with those previously re- ported [37], reveal the beneficial effect of hTERT overexpression on the growth potential of MSCs, which, based on our results, is not limited to adipose-derived cells but apply to MSCs from different sources. In an attempt to explain the amelioration in the growth potential of hTERT overexpressing cells and given the importance of DNA damage accumulation in limiting cellular proliferation capacity [38], we analysed the DNA damage in the nuclei of growing cells. We evaluated the amount of DNA damage in control ASCs as well hTERT ASCs at early passage (p6), middle passage (p12) and high passage (p20).The pres- ence of 8-oxo-7,8-dihydro-2′-deoxyguanosine (8-oxo- dG), major product of DNA oxidation as mentioned earlier, was assessed. 8-oxo-dG lesions could lead to reduced efficiency of the base excision repair process leading, in turn, to persistent DNA single strand breaks Figure 2. ASCs genetically modified with hTERT have increased telomerase activity. RT-PCR–basedTRAP assay was used to measure telomerase activity in hTERT ASCs at passage 7. Telomerase ac- tivity of the hTERT ASCs is depicted as fold change relative to the activity of the control ASCs of the same passage.The analysis was performed in triplicate with four control ASCs and four hTERT ASCs. Values showed are the means ± SE. Table I. Flow cytometry analysis results for CD stem cell markers in control ASCs and hTERT-overexpressing ASCs (hTERT ASCs) at passage 7. Stem cell marker hTERT ASCs passage 7 Control ASCs passage 7 CD34 0% 2% CD29 99.6% 99.8% CD73 99.4% 99.7% CD90 99.3% 99.3% CD105 99.7% 97.5% CD146 96.4% 93.8% 812 V.Trachana et al.
  • 6. (SSB) intermediates; the latter could then act as double strand breaks (DSB)-prone sites in subsequent rounds of DNA replication [39]. DSB activate ataxia- telangiectasia mutated/ataxia telangiectasia and Rad3- related serine/threonine-protein kinases (ATM/ATR) that bind to the DNA and induce phosphorylation of many downstream proteins, such as H2A.X and 53BP1, in order to both mark the location and initiate repair of the damage. In order to evaluate DSB formation in long-term cultures of MSCs, we therefore assessed control ASCs as well as hTERT ASCs of different pas- sages (p6, p12 and p20) for the presence of the phosphorylated form (on Ser139) of the histone variant H2AX (γH2AX) and the 53BP1 protein. Figure 4A shows that growing ASCs accumulate DSB as well as oxidative lesions, as indicated by the increased per- centage of cells positive for γΗ2ΑΧ/ 53BP1 (i, ii) and 8-oxo-dG staining (iii), respectively. All nuclei bearing >5 foci (8-oxo-dG, γΗ2ΑΧ or 53BP1) were counted as positive. Importantly, as depicted in the same figure (Figure 4A), our result demonstrate that cells geneti- cally modified with the catalytic subunit of hTERT accumulate significantly less damage than the control cells in all passages tested. Figure 4B shows confocal microscopy characteristic images of foci positive for γΗ2ΑΧ, 53BP1 and 8-oxo-dG in the nuclei of control as well hTERT overexpressing cells. It is obvious that hTERT overexpressing cells accumulate a lot less DNA damage associated foci in their nuclei at high passage (p = 20).The latter suggest that the elevated activity of telomerase protects cells against oxidative lesions and DSB accumulation which could explain hTERT cells’ enhanced proliferation capacity. Ectopic expression of hTERT results in ameliorated response to exogenous oxidative stress-induced DNA damage The protection acquired by hTERT overexpressing cells against DNA lesions accumulating during growth of ASCs, prompt us to investigate whether similar pro- tection occurs after exposure to acute external oxidative stress.Therefore, control ASCs as well as hTERT ASCs were treated with H2O2 for 30 min in order to evaluate DNA damage accumulation. One hour after treat- ment nuclei with >5 foci (for all markers) were counted as positive. As Figure 5 shows, both control ASCs as well as hTERT ASCs at passage 6 (early), passage 12 (middle) and passage 20 (high) treated with H2O2, ac- cumulate similarly high amounts of DNA damage associated foci (Figure 5A–C, p(6/12/20) + H2O2). Both control ASCs and hTERT ASCs at early and middle Figure 3. ASCs that overexpress hTERT retain their differentiation potential. (A, B) Passage 10 ASCs and hTERT ASCs cultured under standard conditions (control medium) or under conditions to induce adipogenic/osteogenic differentiation (differentiation medium). Under differentiation conditions, cells began to accumulate lipid droplets and acquired a foamy appearance with the characteristic staining for oil red, typically seen in adipocytes (A), or exhibited calcium deposition, with the characteristic staining for alizarin red, which is typically seen in osteocytes (B) (Scale bar: 10 µmol/L). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) Telomerase protects against genomic instability 813
  • 7. passage were, however, able to, at least partially, repair the damage when left to recover in fresh medium for 48 h (p6/12 + H2O2 recovery 48h), but not at high passage (p20 + H2O2 recovery 48h). This ability was found to be enhanced in hTERT ASCs, since signifi- cantly less amount of DNA damage was measured in these cells 48 h after the oxidative treatment at passages 6 and 12, than in control ASCs. The latter was grad- ually diminished as cells reached higher passages (p20). Overall these results suggest that ASCs of early and middle passage genetically modified with the cata- lytic subunit of hTERT have enhanced ability to repair the externally provoked oxidative stress-induced DNA lesions. A B Figure 4. hTERT overexpression in ASCs offers protection against DNA damage accumulation in long-term culture. (A) Percentage of control ASCs and hTERT-overexpressing ASCs (hTERT ASCs) with DNA damage measured by immunofluorescence using specific an- tibodies against γH2AX (i), 53BP1 (ii) or 8-oxo-dG (iii) at the indicated passages. Nuclei bearing >5 foci stained with 8-oxo-dG, γΗ2ΑΧ or 53BP1 antibody were counted as positive. Values shown are the means ± SE. *P < 0.05, **P < 0.01 and ***P < 0.001 versus control ASCs in the passage indicated. (B) Characteristic images of control and hTERT ASCs at low (passage 6) and high passage (passage 20) with DNA damage evaluated by confocal microscope and after staining with γH2AX, 53BP1 or 8-oxo-dG antibody (green). Nuclei are stained with DAPI (blue). Scale bar 20 µm. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) 814 V.Trachana et al.
  • 8. Figure 5. hTERT overexpression in ASCs offers protection against external oxidative insult. Percentage of control ASCs and hTERT- overexpressing ASCs (hTERT ASCS) with DNA damage measured by IF using γH2AX (A), 53BP1 (B) or 8-oxo-dG (C) antibodies at low (passage 6), middle (passage 12) and high (passage 20) passages after treatment with 300 µmol/L H2O2 for 30 min (passage 6/12/ 20 + H2O2) and after 48 h of recovery time (passage 6/12/20 + H2O2 recovery 48 h). Nuclei bearing >5 foci stained with 8-oxo-dG, γΗ2ΑΧ or 53BP1 antibody were counted as positive. Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in each passage indicated. Telomerase protects against genomic instability 815
  • 9. Enhanced TERT activity protects against oxidative stress provoked nuclei malformations It has been suggested that oxidative stress could lead to aberrant nucleus morphology, such as giant nuclei, donut-shaped nuclei, micronuclei or multilobular nuclei, which is also considered a characteristic of se- nescent cells [40]. Indeed, as shown in Figure 6A both control as well as hTERT ASCs demonstrate an in- crease in such malformations (i.e mitotic defects) in passage 20 (senescence reaching) as compared to low passage 6. Nevertheless, hTETR overexpressing ASCs had significantly less nucleus defects than control cells in both time points tested (p6, p20). Figure 6B shows characteristic nuclei aberration of high passage 20 control ASCs, such as giant nucleus, donut-shaped nucleus, micronucleus or multilobular nucleus (Figure 6B, i, ii, iii, iv respectively). Additionally, we tested whether the externally pro- voked oxidative stress could lead to an increase of such nuclei malformations. Both control ASCs and hTERT ASCs at low (p6) and high (p20) passage treated with 300 µΜ Η2Ο2 for 30 min and left to recover for 48 h, were DAPI-stained in order to evaluate their nuclear morphology. Firstly, as indicated in Figure 6A, both control ASCs and hTERT showed an increase in nucleus malformations in both passages tested after treatment with H2O2 (p6/20 + H2O2 recovery 48h) in comparison with the same passages without treat- ment (p6/20). However, significantly less aberrant nuclei were present in hTERT ASCs 48 after treat- ment with Η2Ο2 at both low and high passages in comparison with the control cells. The latter indi- cates that the hTERT overexpression was able protect against oxidative stress-induced nuclei malforma- tions, which, together with the previous observations, suggests an overall ameliorated antioxidant response. Genetically modified MSCs with hTERT demonstrate increased activity of the basic antioxidant enzymes Superoxide dismutases (SOD) are metaloenzymes that catalyze the dismutation of the superoxide anion to mo- lecular oxygen and H2O2 and thus form a crucial part of the cellular antioxidant defense mechanism [41]. Catalase (CAT) is a ubiquitous antioxidant enzyme in- volved in the detoxification of H2O2 by catalyzing its conversion to molecular oxygen and water [42]. In an A B Figure 6. hTERT-overexpressing ASCs show resistance to aberrant nuclei formation. (A) Percentage of cells with mitotic defects (*giant nuclei, donut-shaped nuclei, micronuclei and multilobular nuclei) evaluated using DAPI staining of control ASCs and hTERT- overexpressing ASCs (hTERT ASCs) at low (passage 6) and high passage (passage 20). Same nuclei aberrations were assessed at passages 6 and 20 after the cells were treated with 300 µmol/L Η2Ο2 for 30 min and left to recover for 48 h (passage 6/20 + H2O2 recovery 48 h). Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in the passage indicated. (B) Characteristic images of DAPI-stained (blue) giant nuclei (arrow in (i)), donut-shaped nuclei (ii), micronucleus (arrow in (iii)) and multilobular nuclei (iv) of control ASCs at high passage (passage 20) (Scale bar 25 µm). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.) 816 V.Trachana et al.
  • 10. attempt to explain the amelioration of the oxidative re- sponse observed in hTERT overexpressing cells we analysed the basic antioxidant capacity of ASCs. For this reason, we measured the activities of both enzymes (SOD and CAT) using commercially available kits (Cayman, Chemical, Ann Arbor, MI). As shown in Figure 7A ASCs genetically modified with hTERT (hTERT ASCs) have similar activity of SOD with control cells at low passage (p6). At high passage (p20) however, control cells show a decrease in their SOD activity whereas hTERT overexpressing cells main- tain significantly higher levels, similar to the levels of the low passage. Regarding CAT activity, we observed that both control as well as hTERT overexpressing cells have increased activity at higher passages (Figure 7B, p20), which is in accordance with previously reported data. In specific, it was reported that senescent cul- tures of human lung fibroblasts have elevated H2O2 generation which stimulate cells to increase their cata- lase activity [43]. Besides confirming that, we report here, that hTERT ASCs have significantly higher CAT activity levels than control cells, both at passage 6 as well as in passage 20 (Figure 7B). The above results reveal an enhancement of the basic antioxidant enzy- matic defense of the cells due to hTERT overexpression. Discussion Endogenous oxidants generated by normal cellular oxi- dative metabolism are a major obstacle to overcome in cell based therapies that require ex vivo long-term cell culture. The DNA damage that oxidative stress (OS) provokes could lead to cellular senescence and therefore limit the number of cells with the self- renewal and multilineage differentiation ability (stemness) that is required for successful transplan- tation. Besides DNA damage, it has been reported that OS -either normally occurred due to metabolism or through exposure to external oxidants- leads to ab- errant nuclear morphology. It was demonstrated that a significant number of senescent or progeroid human fibroblasts exhibit nuclei malformations, such as multilobbular nuclei and one or multiple micronu- clei that are attributed to mitotic slippage [40]. Mitotic slippage is a known consequence of a dysfunctional spindle assembly checkpoint (SAC) [44] and OS was proposed to be a major cause of its impairment [45]. Moreover, it was recently demonstrated that, exog- enously induced OS is able to promote aneuploidy in MSCs [37] which could also be attributed to an im- paired SAC. Actually, it was proposed that OS could affect the functionality of the SAC by regulating the expression of two key SAC mediators, BubR1 and Mad2 [46]. All the above suggest that OS provokes genomic instability, i.e. DNA damage, nuclear defor- mations and aneuploidy that reduce the stemness of MSCs in culture and limit transplantation success. On the other hand, accumulating recent evi- dence link the catalytic subunit of telomerase to functions other than telomere length maintenance. As a matter of fact, one of these extratellomeric func- tions of telomerase that attracted a lot of attention recently is its implication in the antioxidant defense mechanism(s) of the cell [47]. The precise molecu- lar mechanism of telomerase’s telomere independent functions is becoming an important-still mostly unanswered-question. In order to shed light onto these issues, in the present study we genetically modified human mesenchymal stem cells derived from different sources (adult adipose tissue and umbilical cord’s Wharton’s Jelly) with the cata- lytic subunit of telomerase (hTERT) in order to elucidate its contribution in overcoming endogenously occurring or externally provoked OS. Firstly, we showed Figure 7. ASCs genetically modified with hTERT have higher basic antioxidant enzymes activities. Analysis of the SOD (A) and CAT (B) enzymatic activity of control ASCs and ASCS overexpressing hTERT (hTERT ASCs) at low (passage 6) and high (passage 20) pas- sages. Values shown are the means ± SE. *P < 0.05 and **P < 0.01 versus control ASCs in the passage indicated. Telomerase protects against genomic instability 817
  • 11. that genetic modification with hTERT did not alter the mesenchymal properties of adipose orWharton’s Jelly derived cells, in agreement with previously reported data on human bone marrow stromal cells [48] and mouse adipose derived MSCs [49]. Furthermore, we report that overexpression of hTERT in ASCs may contrib- ute in retaining their mesenchymal properties for longer, an observation which is in agreement with previous results [50]. Importantly, we proved that the genetic modification of MSCs with hTERT results in de- creased DNA damage accumulation, that occurs either as a result of long term culture or due to acute oxida- tive external insult. It has been suggested that hTERT overexpression ameliorates the DNA damage re- sponse by either accelerating DNA repair kinetics through increased levels of intracellular deoxynucleotides (dNTP) and ribonucleotides (NTPs) [51], or by at- tracting DNA repair proteins to the sites of DNA damage [52]. Even though the hTERT participation in DNA damage repair is a rather attractive notion and some supportive data do exist, the exact mechanism remains elusive. Based on our data it is, at this point, safer to hypothesize that the protection against DNA damage accumulation results from an hTERT driven enhancement of the antioxidant cellular response. In support, recent studies showed that the telomerase lo- calizes to subcellular locations other than the nucleus, such as the mitochondria. hTERT was shown to shuttle dynamically from the nucleus to the mitochondria, where it decreases mitochondrial reactive oxygen species (ROS) generation [28–31]. Furthermore, strong evi- dence has been provided that increased hTERT expression not only reduces the total basal ROS levels, but also significantly antagonizes the increase in cel- lular ROS in response to exogenous ROS triggers (H2O2) [53]. We also showed that MSCs genetically modified with hTERT exhibit resistance to nuclei malforma- tions that arise either through prolonged culture or due to H2O2 treatment. Nuclei malformations, as men- tioned above, could be the result of an OS-induced SAC impairment.The resistance against OS-induced aberrant nuclei formation demonstrated here is a novel indication of telomerase’s ability to counteract OS and promote cell survival. We know that in order to resist ROS, the cells use enzymatic antioxidants, with superoxide dismutase (SOD) and catalase (CAT) being the major ones [54]. Our data demonstrate that hTERT overexpression results in increased SOD and CAT activities. It was previously reported that hTERT modified MSC dem- onstrated elevated protein levels of the principal protein scavenger of mitochondrial superoxide, MnSOD (SOD2), suggesting that hTERT overexpression could be a key regulator of the metabolic status [37]. Here we demonstrate that hTERT overexpression could directly lead to an enhancement of the antioxidant ma- chinery of the cell by elevating the activity of the key enzymes. In support, one previous study has shown that mouse embryonic fibroblasts isolated from telomerase-deficient mice, have decreased catalase ac- tivity which results in redox imbalance in these mice [55]. Finally, it has been reported that the donor’s initial telomeres length might be a critical restriction for the maintenance of the MSCs in prolonged cell culture [56]. Following transplantation, the telomere length might limit the therapeutic potential of MSCs, by de- creasing either the duration of cell survival in the tissue or their ability to integrate in the adult tissue [57,58]. Here, we prove that both adult adipose derived stem cells as well as umbilical cord’sWharton’s Jelly derived cells genetically modified with hTERT have an en- hanced growth potential that could be attributed to their improved antioxidant capacity. Therefore, this genetic manipulation implemented by us, and others [48,59], could overcome the obstacle of shorter initial telomere length of the adult stem cells. Understand- ing the restrains of this strategy we believe that synthetic or natural compounds that activate the telomerase might be a better approach. As a matter of fact, Tichon et al. [60] demon- strated that two novel compounds, designated AGS- 499 and AGS-500, when administrated to MSCs increased the average telomere length, preserved genome integrity and allowed MSCs to differentiate into various lineage. The authors also demonstrated that these effects were telomerase depended. Our study confirms these previously reported results but also demonstrates further novel effects of the hTERT overexpression. We showed that hTERT enhance- ment, via direct genetic manipulation, confers higher growth potential to the cells, most probably due to the amelioration of the cellular response to the OS that either occurs normally during cellular senescence or it is exogenously provoked. Moreover, we demon- strated that this ameliorated OS response of the hTERT overexpressing cells comes from direct en- hancement of the cell’s basic enzymatic antioxidant machinery, which in turn protects against OS-induced genomic damage. In other words, our study pro- vides novel data in support of the recently proposed antioxidant role of telomerase. Besides that, we believe that our analysis offers a comprehensive evaluation of the outcomes of an augmented telomerase activity in human MSCs that might help overcome the adult stem cell-based therapies limitations. Acknowledgments This work was funded by a SYNERGASIA project (11ΣΥΝ_1_1112). Authors wish to thank Dr. Z. Ivics 818 V.Trachana et al.
  • 12. and Dr. Z. Izsvák for kindly providing the Sleeping Beauty 100X transposon system, A. Kalkavouri and A. Ntoga for technical assistance and Dr. E. Panteris for his help with the microscopy techniques. Disclosure of interests:The authors declare no com- mercial or financial conflict of interest. References [1] Wei X, Yang X, Han Z, Qu F, Shao L, Shi Y. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 2013;34:747–54. doi:10.1038/aps.2013.50. [2] Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell 2008;2:313–19. doi:10.1016/j.stem.2008.03.002. [3] Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ, et al. Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng 2001;7:211– 28. doi:10.1089/107632701300062859. [4] Baxter MA, Wynn RF, Jowitt SN, Wraith JE, Fairbairn LJ, Bellantuono I. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 2004;22:675–82. doi:10.1634/stemcells.22-5 -675. [5] Brandl A, Meyer M, Bechmann V, Nerlich M, Angele P. Oxidative stress induces senescence in human mesenchymal stem cells. Exp Cell Res 2011;317:1541–7. doi:10.1016/ j.yexcr.2011.02.015. [6] Gu Y, Li T, Ding Y, Sun L, Tu T, Zhu W, et al. Changes in mesenchymal stem cells following long-term culture in vitro. Mol Med Rep 2016;13:5207–15. doi:10.3892/mmr.2016 .5169. [7] Kasper G, Mao L, Geissler S, Draycheva A, Trippens J, Kühnisch J, et al. Insights into mesenchymal stem cell aging: involvement of antioxidant defense and actin cytoskeleton. Stem Cells 2009;27:1288–97. doi:10.1002/stem.49. [8] Campisi J. The biology of replicative senescence. Eur J Cancer A 1997;33:703–9. doi:10.1016/S0959-8049(96)00058-5. [9] Cristofalo VJ, Lorenzini A, Allen RG, Torres C, Tresini M. Replicative senescence: a critical review. Mech Ageing Dev 2004;125:827–48. doi:10.1016/j.mad.2004.07.010. [10] Collins K, Mitchell JR. Telomerase in the human organism. Oncogene 2002;21:564–79. doi:10.1038/sj/onc/. [11] Smogorzewska A, de Lange T. Different telomere damage signaling pathways in human and mouse cells. EMBO J 2002;21:4338–48. doi:10.1093/emboj/cdf433. [12] Siegel G, Kluba T, Hermanutz-Klein U, Bieback K, Northoff H, Schafer R. Phenotype, donor age and gender affect function of human bone marrow-derived mesenchymal stromal cells. BMC Med 2013;11:146. doi:10.1186/1741-7015-11-146. [13] Huang K, Zhou DH, Huang SL, Liang SH. [Age-related biological characteristics of human bone marrow mesenchymal stem cells from different age donors]. Zhongguo ShiYan Xue Ye Xue Za Zhi 2005;13:1049–53. [14] Stenderup K, Justesen J, Clausen C, Kassem M. Aging is associated with decreased maximal life span and accelerated senescence of bone marrow stromal cells. Bone 2003;33:919– 26. [15] Stolzing A, Jones E, McGonagle D, Scutt A. Age-related changes in human bone marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech Ageing Dev 2008;129:163–73. doi:10.1016/j.mad.2007.12.002. [16] Kalaszczynska I, Ferdyn K. Wharton’s jelly derived mesenchymal stem cells: future of regenerative medicine? Recent findings and clinical significance. Biomed Res Int 2015;2015:430847. doi:10.1155/2015/430847. [17] Von Zglinicki T. Oxidative stress shortens telomeres. Trends Biochem Sci 2002;27:339–44. doi:10.1016/S0968 -0004(02)02110-2. [18] Houben JMJ, Moonen HJJ, van Schooten FJ, Hageman GJ. Telomere length assessment: biomarker of chronic oxidative stress? Free Radic Biol Med 2008;44:235–46. doi:10.1016/ j.freeradbiomed.2007.10.001. [19] Toussaint O, Medrano EE, von Zglinicki T. Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp Gerontol 2000;35:927–45. [20] Fraga CG, Shigenaga MK, Park JW, Degan P, Ames BN. Oxidative damage to DNA during aging: 8-hydroxy-2’- deoxyguanosine in rat organ DNA and urine. Proc Natl Acad Sci USA 1990;87:4533–7. doi:10.1073/pnas.87.12 .4533. [21] Oikawa S, Kawanishi S. Site-specific DNA damage at GGG sequence by oxidative stress may accelerate telomere shortening. FEBS Lett 1999;453:365–8. doi:10.1016/S0014 -5793(99)00748-6. [22] Chen QM, Prowse KR, Tu VC, Purdom S, Linskens MH. Uncoupling the senescent phenotype from telomere shortening in hydrogen peroxide-treated fibroblasts. Exp Cell Res 2001;265:294–303. doi:10.1006/excr.2001.5182. [23] Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature 2000;408:239–47. doi:10.1038/ 35041687. [24] Choi KM, Seo YK, Yoon HH, Song KY, Kwon SY, Lee HS, et al. Effect of ascorbic acid on bone marrow-derived mesenchymal stem cell proliferation and differentiation. J Biosci Bioeng 2008;105:586–94. doi:10.1263/jbb.105.586. [25] Lin TM, Tsai JL, Lin SD, Lai CS, Chang CC. Accelerated growth and prolonged lifespan of adipose tissue-derived human mesenchymal stem cells in a medium using reduced calcium and antioxidants. Stem Cells Dev 2005;14:92–102. doi:10.1089/scd.2005.14.92. [26] Tsai CC, Chen YJ, Yew TL, Chen LL, Wang JY, Chiu CH, et al. Hypoxia inhibits senescence and maintains mesenchymal stem cell properties through down-regulation of E2A-p21 by HIF-TWIST. Blood 2010;117:459–69. doi:10.1182/blood -2010-05-287508. [27] Borrás C, Esteve JM, Viña JB, Sastre J, Viña J, Pallardó FV. Glutathione regulates telomerase activity in 3T3 fibroblasts. J Biol Chem 2004;279:34332–5. doi:10.1074/ jbc.M402425200. [28] Ahmed S, Passos JF, Birket MJ, Beckmann T, Brings S, Peters H, et al. Telomerase does not counteract telomere shortening but protects mitochondrial function under oxidative stress. J Cell Sci 2008;121:1046–53. doi:10.1242/jcs.019372. [29] Singhapol C, Pal D, Czapiewski R, Porika M, Nelson G, Saretzki GC. Mitochondrial telomerase protects cancer cells from nuclear DNA damage and apoptosis. PLoS ONE 2013;8:e52989. doi:10.1371/journal.pone.0052989. [30] Haendeler J, Drose S, Buchner N, Jakob S, Altschmied J, Goy C, et al. Mitochondrial telomerase reverse transcriptase binds to and protects mitochondrial DNA and function from damage. Arterioscler Thromb Vasc Biol 2009;29:929–35. doi:10.1161/ATVBAHA.109.185546. [31] Saretzki G. Telomerase, mitochondria and oxidative stress. Exp Gerontol 2009;44:485–92. doi:10.1016/j.exger.2009.05.004. [32] Petrakis S, Rasko T, Mates L, Ivics Z, Izsvak Z, Kouzi-Koliakou K, et al. Gateway-compatible transposon vector to genetically modify human embryonic kidney and adipose-derived stromal cells. Biotechnol J 2012;7:891–7. doi:10.1002/biot.201100471. Telomerase protects against genomic instability 819
  • 13. [33] Tsagias N, Koliakos I, Karagiannis V, Eleftheriadou M, Koliakos GG. Isolation of mesenchymal stem cells using the total length of umbilical cord for transplantation purposes. Transfus Med 2011;21:253–61. doi:10.1111/ j.1365-3148.2011.01076.x. [34] Mates L, Chuah MK, Belay E, Jerchow B, Manoj N, Acosta-Sanchez A, et al. Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates. Nat Genet 2009;41:753–61. doi:10.1038/ng.343. [35] Tsolou A, Nelson G, Trachana V, Chondrogianni N, Saretzki G, Von Zglinicki T, et al. The 19S proteasome subunit Rpn7 stabilizes DNA damage foci upon genotoxic insult. IUBMB Life 2012;64:432–42. doi:10.1002/iub.1018. [36] Kapetanou M, Chondrogianni N, Petrakis S, Koliakos G, Gonos ES. Proteasome activation enhances stemness and lifespan of human mesenchymal stem cells. Free Radic Biol Med 2017;103:226–35. doi:10.1016/j.freeradbiomed.2016 .12.035. [37] Estrada JC, Torres Y, Benguría A, Dopazo A, Roche E, Carrera-Quintanar L, et al. Human mesenchymal stem cell-replicative senescence and oxidative stress are closely linked to aneuploidy. Cell Death Dis 2013;4:e691. doi:10.1038/ cddis.2013.211. [38] Chen JH, Hales CN, Ozanne SE. DNA damage, cellular senescence and organismal ageing: causal or correlative? Nucleic Acids Res 2007;35:7417–28. doi:10.1093/nar/ gkm681. [39] Eccles LJ, O’Neill P, Lomax ME. Delayed repair of radiation induced clustered DNA damage: friend or foe? Mutat Res 2011;711:134–41. doi:10.1016/j.mrfmmm.2010.11.003. [40] Ohshima S. Abnormal mitosis in hypertetraploid cells causes aberrant nuclear morphology in association with H2O2- induced premature senescence. Cytometry A 2008;73:808–15. doi:10.1002/cyto.a.20604. [41] Zelko IN, Mariani TJ, Folz RJ. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med 2002;33:337– 49. doi:10.1016/S0891-5849(02)00905-X. [42] Radi R, Turrens JF, Chang LY, Bush KM, Crapo JD, Freeman BA. Detection of catalase in rat heart mitochondria. J Biol Chem 1991;266:22028–34. [43] Allen RC, Tresini M, Keogh BP, Doggett DL, Cristofalo VJ. Differences in electron transport potential, antioxidant defenses, and oxidant generation in young and senescent fetal lung fibroblasts (WI-38). J Cell Physiol 1999;180:114–22. doi:10.1002/(SICI)1097-4652(199907)180:1<114::AID- JCP13>3.0.CO;2-0. [44] Trachana V, Van Wely KHM, Guerrero AA, Fütterer A, Martinez-A C. Dido disruption leads to centrosome amplification and mitotic checkpoint defects compromising chromosome stability. Proc Natl Acad Sci USA 2007;104: 2691–6. doi:10.1073/pnas.0611132104. [45] D’Angiolella V, Santarpia C, Grieco D. Oxidative stress overrides the spindle checkpoint. Cell Cycle 2007;6:576–9. doi:10.4161/cc.6.5.3934. [46] Ikawa-Yoshida A, Ando K, Oki E, Saeki H, Kumashiro R, Taketani K, et al. Contribution of BubR1 to oxidative stress- induced aneuploidy in p53-deficient cells. Cancer Med 2013;2:447–56. doi:10.1002/cam4.101. [47] Saretzki G. Extra-telomeric functions of human telomerase: cancer, mitochondria and oxidative stress. Curr Pharm Des 2014;20:6386–403. doi:10.2174/1381612820666140630 095606. [48] Bocker W, Yin Z, Drosse I, Haasters F, Rossmann O, Wierer M, et al. Introducing a single-cell-derived human mesenchymal stem cell line expressing hTERT after lentiviral gene transfer. J Cell Mol Med 2008;12:1347–59. doi:10.1111/j.1582 -4934.2008.00299.x. [49] Madonna R, Taylor DA, Geng YJ, De Caterina R, Shelat H, Perin EC, et al. Transplantation of mesenchymal cells rejuvenated by the overexpression of telomerase and myocardin promotes revascularization and tissue repair in a murine model of hindlimb ischemia. Circ Res 2013;113:902–14. doi:10.1161/CIRCRESAHA.113.301690. [50] Kang SK, Putnam L, Dufour J, Ylostalo J, Jung JS, Bunnell BA. Expression of telomerase extends the lifespan and enhances osteogenic differentiation of adipose tissue-derived stromal cells. Stem Cells 2004;22:1356–72. doi:10.1634/ stemcells.2004-0023. [51] Sharma GG, Gupta A, Wang H, Scherthan H, Dhar S, Gandhi V, et al. hTERT associates with human telomeres and enhances genomic stability and DNA repair. Oncogene 2003;22:131–46. doi:10.1038/sj.onc.1206063. [52] Shin KH, Kang MK, Dicterow E, Kameta A, Baluda MA, Park NH. Introduction of human telomerase reverse transcriptase to normal human fibroblasts enhances DNA repair capacity. Clin Cancer Res 2004;10:2551–60. [53] Indran IR, Hande MP, Pervaiz S. hTERT overexpression alleviates intracellular ROS production, improves mitochondrial function, and inhibits ROS-mediated apoptosis in cancer cells. Cancer Res 2010;71:266–76. doi:10.1158/0008-5472.CAN- 10-1588. [54] Caradonna F, Mauro M. Role of the antioxidant defence system and telomerase in arsenic-induced genomic instability. Mutagenesis 2016;31:661–7. doi:10.1093/mutage/gew034. [55] Perez-Rivero G, Ruiz-Torres MP, Diez-Marques ML, Canela A, Lopez-Novoa JM, Rodriguez-Puyol M, et al. Telomerase deficiency promotes oxidative stress by reducing catalase activity. Free Radic Biol Med 2008;45:1243–51. doi:10.1016/ j.freeradbiomed.2008.07.017. [56] Bonab MM, Alimoghaddam K, Talebian F, Ghaffari SH, Ghavamzadeh A, Nikbin B. Aging of mesenchymal stem cell in vitro. BMC Cell Biol 2006;7:14. doi:10.1186/1471-2121 -7-14. [57] Ju Z, Jiang H, Jaworski M, Rathinam C, Gompf A, Klein C, et al. Telomere dysfunction induces environmental alterations limiting hematopoietic stem cell function and engraftment. Nat Med 2007;13:742–7. doi:10.1038/nm1578. [58] Mangerini R, Lanino E, Terranova P, Faraci M, Pistillo MP, Gaetani GF, et al. Telomere length of donors influences granulocyte recovery in children after hematopoietic stem cell transplantation. Ann Hematol 2009;88:1029–31. doi:10.1007/ s00277-009-0712-z. [59] Nakahara H, Misawa H, Hayashi T, Kondo E, Yuasa T, Kubota Y, et al. Bone repair by transplantation of hTERT-immortalized human mesenchymal stem cells in mice. Transplantation 2009;88:346–53. doi:10.1097/TP .0b013e3181ae5ba2. [60] Tichon A, Eitan E, Kurkalli BG, Braiman A, Gazit A, Slavin S, et al. Oxidative stress protection by novel telomerase activators in mesenchymal stem cells derived from healthy and diseased individuals. Curr Mol Med 2013;13:1010–22. Appendix: Supplementary material Supplementary data to this article can be found online at doi:10.1016/j.jcyt.2017.03.078. 820 V.Trachana et al.