Vous êtes sur la page 1sur 7

Cellular Immunology 289 (2014) 6369

Contents lists available at ScienceDirect

Cellular Immunology
journal homepage: www.elsevier.com/locate/ycimm

Review

Telomeres and telomerase in T cells of tumor immunity


Yaqin Qian, Lili Yang , Shui Cao
Department of Immunology, Tianjin Cancer Institute & Hospital, Tianjin Medical University, Tianjin, China
National Clinical Research Center of Cancer, China
Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
Research Center of Lung Cancer, Tianjin, China

a r t i c l e

i n f o

Article history:
Received 7 October 2013
Accepted 24 March 2014
Available online 1 April 2014
Keywords:
Telomere
Telomerase
T cells
hTERT overexpression
Immunotherapy

a b s t r a c t
Telomeres are specic nucleoprotein structures at the end of a eukaryotic chromosomes characterized by
repeats of the sequence TTAGGG and regulated by the enzyme telomerase which prevents their degradation, loss, rearrangement and end-to-end fusion. During activation, T lymphocytes actively divide, albeit
through only a nite number of cell divisions due to shortening of telomeres. However, studies have demonstrated that human telomerase reverse transcriptase (hTERT), thought to be the major component regulating telomerase activity, can enhance the proliferation of T cells when overexpressed. There are many
treatments for cancers, most of which are targeting the telomere and telomerase of tumor cells. However,
the hTERT-transduced T cells improve their potential for proliferation, making them an appropriate cell
resource for tumor adoptive immunotherapy, a procedure whereby T cells are isolated from patients,
expanded ex vivo and eventually delivered back into the patients, provides a new approach for tumor
therapy through improved overall survival rates in cancer patients. In this review, we will focus on the
telomerase activity in T cells, the regulation of telomerase activity, and hTERT-transduced T cells used
in adoptive immunotherapy for cancer.
2014 Elsevier Inc. All rights reserved.

1. Introduction
Telomeres are specialized structures at the end of linear chromosomes characterized by repeats of the sequence TTAGGG [1].
They, and their associated proteins, are folded into a telomere loop
structure called a T loop, which may provide a general scaffold for
both the protection and replication of telomeres [2,3] and as such,
are required to maintain chromosomal integrity and prevent endto-end fusions of chromosomes. Somatic cells have a limited number of cell divisions and, when telomeric ends become too short,
DNA damage signals from telomeres can send signals to stop division and induce apoptosis through a process termed cellular senescence or replicative senescence [4,5]. This is partly because in
normal cells, the DNA replication machinery is unable to completely duplicate the telomeric DNA so that telomeres are shortened with each round of cell division [6]. These events are well
exemplied in the immune system where evidence show that telomeres are associated with the proliferation of leukocytes [7]. In
Corresponding authors at: Department of Immunology, Tianjin Cancer Institute
& Hospital, Tianjin Medical University, Huanhuxi Road, Tiyuanbei, Hexi District,
Tianjin 300060, China. Fax: +86 022 23537796.
E-mail addresses: yanglili1012@gmail.com (L. Yang), caoshui@yahoo.com
(S. Cao).
http://dx.doi.org/10.1016/j.cellimm.2014.03.009
0008-8749/ 2014 Elsevier Inc. All rights reserved.

this review, we will focus on telomere function and telomerase


activity in T cells, the regulation of telomerase activity, the proliferation of T cells with human telomerase reverse transcriptase
(hTERT) overexpression, and the hTERT-transduced T cells used
in adoptive immunotherapy.

2. Telomere biology
Telomeres are characterized by the presence of multiple repeats
of a short DNA sequence ending in a 30 single-stranded overhang
which forms structures called T-loops [8] and G quadruplexes
[9]. There are variable numbers of repeats including 1015 kb in
humans and 2050 kb in certain mouse and rat species. Telomere
structure is provided by binding shelterin proteins to telomeric repeats: TRF1, TRF2, and Pot1, which play an essential role in the
recruitment of telomerase to telomere and telomere length regulation [6,10,11]. The three other shelterin proteins, TIN2, TPP1, RAP1,
bind to telomeres through indirect proteinprotein interactions via
TRF1, TRF2, and Pot1. Apart from shelterins, other proteins like
TEN1, Pinx1, which are not telomere specic, are also present at
telomeres and carry out important functions there [11]. Physically,
telomeres are the cap at the end of the linear chromosomes of
eukaryotes, thus preventing nucleic acid ends from being

64

Y. Qian et al. / Cellular Immunology 289 (2014) 6369

recognized by DNA-damage responses that can lead to chromosomal fusions and/or chromosomal instability and serve as a primer for the replication of DNA ends during cell division [12]. The
human telomerase complex is bound to the 30 single-stranded overhang of telomere, a ribonucleoprotein comprising hTERT, human
telomerase RNA (hTR) and related proteins, which prevent the
telomeres from degradation, loss, re-arrangement or end-to-end
fusion [6]. In this complex the most important components for telomerase regulation are hTERT, which provides the active site for
catalysis and hTR which provides a template boundary element
that limits the extent of reverse transcription for DNA synthesis.
The proliferative capacity of primary human cells in tissue culture is limited to 5070 population doublings and the telomere
length in humans is about 510 kb which has been suggested to
decrease in length with age [13]. When telomeres of cells reach
critical length, a growing fraction of the cells will enter into the
stage of cellular senescence characterized by loss of cell proliferation [14,15]. Cancer cells instead maintain their telomere length
mainly through the activation of telomerase or, alternatively,
through the use of a homologous recombination-mediated mechanism of telomere elongation known as alternative lengthening of
telomeres. The presence of this other lengthening mechanism
compensates for the lack of telomerase activity in the cell where
telomerase is not ubiquitous or complement its activity [16]. Similarly, telomere maintenance is involved in the induction of dyskeratosis congenita, which displays the features of genetic
anticipation (a phenomenon whereby the symptoms of a genetic
disorder become apparent at an earlier age as it is passed on to
the next generation) with increased severity and earlier onset in
successive generations [17].
Telomerase activity in normal cells is only detected in cells with
proliferative potential, such as germ line and hematopoietic cells
including activated lymphocytes, while most human somatic cells
lack telomerase activity and only somatic stem and progenitor
cells express it at a low level. [18]. Conversely, numerous cancer
cell types up-regulate telomerase to maintain telomeres length
preventing replicative senescence or apoptosis [19,20], which
considering the fact that cancer cells also have a high telomerase
expression and have a concomitant high enzymatic activity make
it a useful molecular marker in cancer diagnosis and prognosis
[21,22]. Telomere dysfunction has also been implicated in bone
marrow failure syndromes, leukemia and cancer development
[2325] and shortening occurs in a variety of human tissues and
organs during aging, including peripheral blood cells and renal
cortex among others [13,18,26]. These studies conrm the tight
relationship between telomeres and aging, cancer, autoimmune
diseases.

cells, which may derive from CD28+CD8+ T cells, exhibiting shorter


telomeres than the parental cells from the same donors. Furthermore, these T cells have a correspondingly lower capacity for cell
division in vitro before they reach senescence [32,33]. Regulatory
CD4+CD25+ T cells (Tregs) are important regulators helping cause
an inefcient immune response against tumor cells which have
also been found to have shortened telomere length, indicating that
induction of TERT in vivo helps prevent replication senescence. In
vitro, TERT activity was readily inducible, but was not sufcient
to prevent further telomere shortening [34,35]. In tumor-inltrating lymphocytes (TILs), clonotypes with longer telomeres are able
to persist and mediate anti-tumor effects and hence their length
and ability to reactivate TERT activity help determine the life span
and anti-tumor activity of these cells [35,36].
Initial studies of telomerase activity in normal peripheral blood
T cells indicated little or no detectable activity [19]. However, other
studies showed instead that telomerase can be expressed at high
levels in T cells and tested the possibility that lymphocytes might
use the expression of telomerase as an adaptive strategy for the
extension of their replicative capacity [37]. This discrepancy
maybe accounted by the fact that telomerase activity differs in tissues with intermediate levels found in tonsil T cells, while a low to
undetectable level of telomerase activity is detected in peripheral
blood T lymphocytes [38]. In mouse model systems, CD4+T cells
are induced to express telomerase in an antigen-specic manner
by in vivo antigen challenge [39] and inammatory cytokines,
including IL-2, IL-7 and IL-15, were shown to up-regulate TERT
expression in human T cells [35]. These ndings indicate that the
expression of telomerase is induced in activated T cells and can
act to sustain telomere length and replicative capacity.
Recent studies demonstrate that in rheumatoid arthritis (RA)
naive CD4+ T cells have shorter telomeres compared to healthy
subjects, and a reduced capacity to up-regulate telomerase activity,
due to insufcient induction of hTERT [40,41]. Conversely, ectopic
hTERT expression on T cells can restore apoptosis resistance. Telomere shortening, dysfunction and fusion also contribute to disease
progression in chronic lymphocytic leukemia (CLL) where telomere
length in T cells is signicantly shorter for the ZAP-70+/CD38+ CLL
patient samples than the ZAP-70/CD38, and where both naive
and memory T-cells from ZAP-70+/CD38+ CLL patients exhibit a signicantly shorter average telomere length [42,43]. Even in the premalignant condition Myelodysplastic syndromes (MDS), T cells
show shortened telomeres compared to controls and, furthermore,
have proliferative defects contributing to the impaired inducible
telomerase activity combined with a deciency in naive T cells hindering their regeneration and contributing to the accumulation of
senescent cells [28].

3. Telomere and telomerase in T cells

4. Regulation of telomerase activity

T lymphocytes have a limited replicative life span, just like most


somatic human cells, until they reach the terminally differentiated
state and then enter into a replicative senescence phase due to progressive telomere loss with age [27]. It has been investigated that
telomere length of naive T cells is longer than memory CD4+ T cells
from the same individual [28,29] through the study of human T cell
proliferation in vitro by the periodic addition of IL-2 and antigen
stimulation, mitogens, or a combination of agonistic antibodies
such as anti-CD3 and anti-CD28. When naive T cells and memory
T cells are grown this way, only naive T cells are capable of substantially more extensive divisions than memory T cells before
reaching replicative senescence (Fig. 1 describes the presence or
absence of telomere and telomerase in the development of T cells)
[30,31]. Similarly, a difference in telomere length is also observed
when comparing human CD8+ T cell subsets with CD28CD8+ T

Telomerase is a ribonucleoprotein complex composed of two


main subunits: hTERT and hTR component that serves as a template for replication and binds the 30 overhang of telomeres. When
the hTERT gene is silenced, the telomerase activity is repressed
indicating that the major determinant of enzyme activity is the
transcriptional regulation of the catalytic subunit hTERT [44].
Although hTR is also necessary for the re-establishment of telomerase activity, recent studies suggest that only hTERT is needed to
restore telomerase activity in telomerase-negative normal cells,
such as epithelial cells and human broblasts and that ectopic
expression of hTERT, combined with activated oncogenes, results
in tumorigenesis [45]. The regulatory regions of hTERT and hTR
contain numerous binding sites for a variety of transcription
factors, including both activators and repressors, and signaling
pathways participating in the regulation of hTERT [Table 1].

Y. Qian et al. / Cellular Immunology 289 (2014) 6369

65

Fig. 1. The presence or absence of telomere and telomerase marks cells for different cell fates. The naive T cells have longer telomere length and higher telomerase activity
than memory T cells. During the development of T cells, highly differentiated memory T-cells have shorter telomeres and lost the ability to up-regulate telomerase activity.
Subsequent encounters with antigens make these cells enter replicative senescence. When stimulated by anti-CD3/CD28, they show telomerase activity again. The number of
pluses reects the relative levels of telomere length and telomerase activity (Refs. [2931]).

Table 1
An illustration of some transcriptional factors involved in the regulation of telomerase
(for hTERT and hTR).
Factor

Function

References

hTERT expression
Sp1
NFAT1
c-myc
Estrogen
P53
P73
PRb
Mad1
WT1
TGFb
CTCF
TLA1

Activator
Activator
Activator
Activator
Repressor
Repressor
Repressor
Repressor
Repressor
Repressor
Repressor
Repressor

[48]
[54]
[44]
[49]
[46]
[46]
[47]
[51]
[49]
[57]
[52]
[55]

hTR expression
Sp1
HIF-1
Sp3

Activator
Activator
Repressor

[67]
[67]
[67]

Fig. 2 describes the transcriptional regulation of the hTERT core


promoter.
The core promoter region of hTERT is a major target of enzyme
regulation with the hTERT gene located on the short arm of human
chromosome 5 (5p15.33), more than 2 Mb away from the telomere, spans 330 bp upstream of the translational start site and
37 bp of exon 2. The regulatory region has abundant transcriptional binding sites that bind a variety of transcription factors
including both activators and repressors of hTERT. P53 and P73
down-regulate hTERT expression through binding to the Sp1/Sp3
complex [46] while the Rb protein, complexes with E2F, recruits
a histone deacetylase (HDAC) protein to the chromatin suppressing
DNA synthesis [47]. Transcription factor Sp1 binds with GC-boxes
to activate hTERT transcription [48] while CCCTC-binding factor
(CTCF) is thought to be the factor that can negatively regulate
the transcription of hTERT. Oncogenes, such as c-Myc, often form
complexes with Max protein and bind to the E-box in the hTERT
promoter to up-regulate transcription. Otherwise, binding of
WT1 (Wilms tumor suppressor) to the hTERT promoter negatively
regulates hTERT expression. The hTERT promoter contains two
estrogen responsive elements, thus estrogen binds to the

Fig. 2. Transcriptional regulation of the hTERT core promoter. Some regulators of hTERT transcription are summarized in the site of hTERT core promoter. The repressors
which down-regulate hTERT transcription through the binding sites such as P53, P73, Wilms tumor suppressor (WT1), Myeloid zinc nger (MZF), Transforming growth
factor-b (TGF-b), CCCTC-binding factor (CTCF), Mad, Rb protein are shown. Similarly, the activators which up-regulate hTERT transcription such as Nuclear Factor-kappa B
(NF-jB), Sp1, c-myc, estrogen are summarized in the core promoter. Major regulatory pathways exert either repressive, as in the case of the TGF- b pathway, or activated, as is
Wnt pathway. Abbreviations: ERE, estrogen responsive element; TCF4, T cell factor 4.

66

Y. Qian et al. / Cellular Immunology 289 (2014) 6369

estrogen-response element in the hTERT promoter region and activates hTERT transcription while promoter and histone methylation
also play a key role in its regulation [44,4952], a characteristic of
most cancers and a novel biomarker for the diagnosis of leptomeningeal metastases [53]. Five putative nuclear factors of activated T
cells (NFAT)-binding sites are identied in the hTERT promoter and
are known to be activated by overexpressed NFAT1, which may
have a functional synergy with Sp1 in hTERT transcriptional regulation [54]. However, TAL1 (T-cell acute lymphoblastic leukemia 1)
is a negative regulator of the hTERT promoter and its overexpression leads to a decrease of hTERT expression and reduces telomerase activity [55].
There are several pathways that interfere with hTERT regulation: PI3K/ataxia telangiectasia mutated (Akt) pathway, mitogenactivated protein kinase (MAPK) pathway [56], TGF-b signaling
pathway [57] and the NF-kB pathway [58]. The PI3K/Akt signaling
cascade is also a central regulator of cell proliferation, growth and
apoptosis and when active it in turn mediates the phosphorylation of hTERT thereby enhancing telomerase activity [59]. In
response to environmental and genotoxic stress as well as activation of ataxia teleangiectasia mutated (ATM), the MAPK pathway,
an indirect stimulator or repressor of the hTERT promoter, is triggered and occupies a key role in cell proliferation and survival
[60]. Activation of JNK in ovarian surface epithelial cells induced
telomerase activity by binding with the hTERT promoter [61].
The TGF-b signaling pathway is initiated by ligand binding to
the TGF-b receptor, activating Smad transcription factor (SMAD3),
which is bound to Max protein, resulting in suppression of the
transcription of hTERT [57]. When NF-kB was activated it translocates into the nucleus where it binds to the region of the hTERT
promoter inducing the up-regulation of hTERTs expression [58].
Similarly, last year, Hoffmeyer et al. [62] showed that the Wnt
pathway component b-catenin can regulate the expression of
the telomerase catalytic subunit TERT and that furthermore hTERT
is a target of Wnt/b-catenin. In vivo, b-catenin expression
increases TERT transcription. b-catenin can bind to the TERT
promoter, always together with T cell factor 4 (TCF4) and induce
the TERT expression whether in mouse embryonic stem cells or
human cancer cells [63]. This nding puts Wnt upstream of TERT
and provides a foundation for linking telomerase levels and
self-renewal [64].
Another level of regulation of this enzyme is through alternative
splicing. hTERT is subjected to numerous alternative splicing
events, but the regulation and the function of these splice variants
are obscure. The major splice variant (termed a+b- or b-deletion) is
highly expressed in stem and cancer cells. In a breast cancer cell
panel, overexpressed b-deletion protein competed for binding to
hTR and thereby inhibited the endogenous telomerase activity
and protected breast cancer cells from cisplatin-induced apoptosis
[65].
hTR, which increases in abundance in tumor cells, indicates that
hTR abundance contributes to telomerase regulation [44]. In
mouse models, it has been found that deciency for mouse TR
(mTR) also shows defects in telomere elongation [66], which indicates the essential requirement of hTR for telomerase activity.
Studies about the regulation of hTR show that transcription of
hTR is activated by Sp1 and HIF-1 while repressed by Sp3. The
MAPK signaling pathways may be involved in the regulation of
hTR transcription via binding certain proteins. Furthermore, hTR
expression in telomerase-positive cell lines is associated with
hyperacetylation of H3 and H4 and methylation of Lys4 H3
[67,68]. Since hTR and hTERT are signicant parts of telomerase,
the regulation of hTR and hTERT expression are vital to the transcription of telomerase. Research on these mechanisms may provide an alternative method to activate or repress telomerase
activity.

5. hTERT overexpression enhances the proliferation of T cells


The investigation about the effects of hTERT overexpression on
cell survival shows that hTERT overexpression in different types of
cells can protect cells against apoptosis and replicative senescence,
whether theyre normal or cancerous. There are also indications
that nuclear hTERT is essential for cell survival during oxidative
stress as its knockdown sensitizes cells to apoptosis through
chemotherapeutic agents [56]. Replicative senescence in various
human cell types, including broblasts and endothelial cells, can
be prevented by the ectopic expression of hTERT. Its gene, transformed into various human cells, results in the extension of their
replicative life span without inducing changes associated with
transformation [69,70].
Human T lymphocytes display a limited life span- about 3050
population doublings (PD) when cultured in vitro [27], telomeres
shortened with each round of cell division with age [6]. It was
found that the mean telomere length declined with age in both
granulocytes and lymphocytes, the loss of telomeric DNA with
age in T lymphocytes exceeds that in other cell types [71]. It has
been proven that rhadinoviral vector-mediated hTERT transduction of primary T cells exhibit telomerase reverse-transcriptase
(RT) activity and most of the parental T-cell properties [72].
hTERT-transduced T cells provide a proliferative advantage for
extending the replicative life span of CD4+ T cells. However, they
cannot prevent the overall loss of telomeric DNA, which means
overexpression of hTERT in CD4+ T cells does not prevent eventual
chromosomal abnormalities and replicative senescence. Nevertheless, the telomere loss in hTERT-transduced CD4+ T cells were less
due to PD, and the hTERT-transduced CD4+ T cells have shorter
telomeres at senescence compared to the controls [73]. Moreover,
the hTERT-transduced CD8+ T cells extend their proliferative life
span, retain their cytotoxic properties and do not present any signs
of apoptosis or senescence after more than 170 PDs. Overexpression of telomerase activity, progressive telomere shortening and nal senescence are also observed in hTERT overexpression of CD8+
T cells [74]. However, other observations indicate that hTERTtransduced cytotoxic T lymphocytes (CTLs) clone shows a decreased functional activity during prolonged culture [75]. Another
study shows hTERT-transduced cells including CD4+ helper and
regulatory T cells are more resistant to oxidative stress, which
causes preferential DNA damage in telomeres, and that they do
not alter antigen specicity or phenotype during the long-term
culture [76].
Investigation of the molecular mechanisms regulating the proliferation of T cells, which overexpress hTERT, shows that a significant decline in the number of PDs was repetitively observed in late
passage hTERT-transduced CD8+ T cells with extended life span
(REF). Additionally, they accumulate the cyclin-dependent inhibitors p16Ink4aand p21Cip1 that largely have been associated with
in vitro growth arrest independently of their telomere status [77].
Moreover, overexpression of ubiquitin carboxyl-terminalhydrolase
L1 (UCHL1) is observed in hTERT-transduced CD8+ T cells with extended life span. This protein plays an important role in many cellular processes, including DNA repair, cell cycle regulation, antigen
presentation, signal transduction, vesicular trafc, cell differentiation, stress response and apoptosis [78]. Furthermore, hTERT overexpression can enhance the proliferation of T cells and extend
their life span which make them ideal candidates as cell sources
for adoptive immunotherapy, especially for the cancer therapy.
6. Improve the proliferation of T cells: a new adoptive
immunotherapy for tumors
The use of lymphokine-activated killer (LAK) cells or TILs for
adoptive immunotherapy has achieved remarkable response rates

Y. Qian et al. / Cellular Immunology 289 (2014) 6369

in cancer patients with immunotherapies based on the adoptive


transfer of TILs are the best available treatment for patients with
metastatic melanoma [79,80]. In order to enhance the antigen-specic activation of tumor-reactive T cells that are applied in adoptive cell therapy, the cells are designed by special TCR and scFv
receptors to redirect T cells specically to the tumor. It is generally
known that CD8+ T cytotoxic lymphocytes play the major role in
the anti-tumor effect, while the CD4+ T cells are considered as providers of additional stimuli. However, CD4+ T cells recognize antigens presented by MHC class II molecules and can be directly
cytotoxic to MHC class II-expressing tumor target cells which
makes the addition of antigen-specic helper cells to CD8+ effectors a tool to increase the rate of clinical success of adoptive immunotherapies [81].
TILs proliferate extensively in the days after transfer, but undergo rapid decreases in telomere length making cells with longer
telomeres better candidates to persist and mediate anti-tumor effects [36]. For adoptive cell transfer TILs should be cultured ex vivo.
However, it is inevitable that upon prolonged periods of culturing
and expansion in vitro the cells ultimately enter into a phase of replicative senescence, which results from TERT deciency and consecutive progressive loss of telomere DNA. As is known, it is
difcult to isolate sufcient numbers of tumor-specic T cells from
most malignancies. Therefore, to make sure to get the appropriate
number of the cells, it is necessary to nd methods to enhance the
proliferation of T cells and protect them from telomere loss. As we
have discussed, human T-cell clones overexpressing TERT have
identical growth rates compared with their un-transduced counterparts and have no differences in their gene expression patterns
[74]. One approach involves the overexpression TERT in patients T
cells (hTERT-transduced T cells), which is sufcient to protect T
cells from senescence and up-regulate the potential of proliferation
with the caveat that these cells cannot prevent eventual chromosomal abnormalities and replicative senescence [73]. However,
these cells do not appear to develop genetic changes which are
characteristic of the malignant transformation.
In summary, the development of adoptive immunotherapy continues rapidly armed with the knowledge that ectopic telomerase
expression can rescue human antigen-specic T cells from senescence, from which TERT-immortalized T cell lines and clones overexpressing telomerase have been developed providing an
invaluable source for cancer immunotherapy [35]. As a way to apply adoptive immunotherapy effectively, especially for specialized
anti-tumor T cells, hTERT may be useful as a tool to circumvent the
limitations of other currently available therapeutic T-cell applications. We believe that hTERT-transduced TILs enhance the
potential of proliferation of these cells compared with their nontransduced counterparts. Therefore, it is reasonable that they can
improve anti-tumor function and hence provide an approach for
adoptive immunotherapy. However, although T cells with hTERT
overexpression provide a novel immunotherapy approach, there
are other problems preventing the use of this adoptive immunotherapy approach. hTERT has been proposed as a prototype
Table 2
The advantages and disadvantages of adoptive immunotherapy against tumor with
hTERT-transduced T cells.
Advantages
Proliferative advantage
No difference in gene expression pattern
Functions as usual
Disadvantages
Genomic instability
Recognized by CTL
Eventually become transformed
Replicative senescence

67

universal tumor antigen and serves as an ideal target for


anti-tumor therapeutics. Thus there is a conjecture that the
hTERT-transduced T cells will also be recognized by CTLs depending on hTERT expression level. hTERT overexpression may cause
genomic instability in transduced cells. Will the hTERT-transduced
T cells eventually become transformed? Table 2 displays the
advantages and disadvantages of adoptive immunotherapy with
hTERT-transduced T cells. This potential issue may limit the possibility of applying these cells for immunotherapy. For this reason, in
order to apply this approach to the clinic successfully, further
researches will be required.
Acknowledgments
This work was supported by a grant from National Basic Research Program of China (973 program) (No. 2012CB9333004)
and Science and Technology Development Fund Project Afliated
with the Education Department of Tianjin (No. 20120110). John
E. Anderson, M.D. from Johns Hopkins University and Wei Sheng,
M.D. from Immunology Program at the H. Lee Moftt Cancer Center; Tampa, FL, USA provided their assistant for checking grammar
mistakes throughout the text.
References
[1] R.K. Moyzis, J.M. Buckingham, L.S. Cram, M. Dani, L.L. Deaven, M.D. Jones, J.
Meyne, R.L. Ratliff, J.R. Wu, A highly conserved repetitive DNA sequence,
(TTAGGG)n, present at the telomeres of human chromosomes, Proc. Natl. Acad.
Sci. U.S.A. 85 (1988) 66226626.
[2] J.D. Grifth, L. Comeau, S. Roseneld, R.M. Stansel, A. Bianchi, H. Moss, T. de
Lange, Mammalian telomeres end in a large duplex loop, Cell 97 (1999) 503
514.
[3] A. Smogorzewska, T. de Lange, Regulation of telomerase by telomeric proteins,
Annu. Rev. Biochem. 73 (2004) 177208.
[4] J. Campisi, Cancer and ageing: rival demons?, Nat Rev. Cancer 3 (2003) 339
349.
[5] F. dAdda di Fagagna, P.M. Reaper, L. Clay-Farrace, H. Fiegler, P. Carr, T. Von
Zglinicki, G. Saretzki, N.P. Carter, S.P. Jackson, A DNA damage checkpoint
response in telomere-initiated senescence, Nature 426 (2003) 194198.
[6] C.M. Raynaud, L. Sabatier, O. Philipot, K.A. Olaussen, J.C. Soria, Telomere length,
telomeric proteins and genomic instability during the multistep carcinogenic
process, Crit. Rev. Oncol. Hematol. 66 (2008) 99117.
[7] C.P. Chiu, C.B. Harley, Replicative senescence and cell immortality: the role of
telomeres and telomerase, Proc. Soc. Exp. Biol. Med. 214 (1997) 99106.
[8] R.J. Wellinger, D. Sen, The DNA structures at the ends of eukaryotic
chromosomes, Eur. J. Cancer 33 (1997) 735749.
[9] J.R. Williamson, M.K. Raghuraman, T.R. Cech, Monovalent cation-induced
structure of telomeric DNA: the G-quartet model, Cell 59 (1989) 871880.
[10] W. Palm, T. de Lange, How shelterin protects mammalian telomeres, Annu.
Rev. Genet. 42 (2008) 301334.
[11] J. Nandakumar, T.R. Cech, Finding the end: recruitment of telomerase to
telomeres, Nat. Rev. Mol. Cell Biol. 14 (2013) 6982.
[12] M.P. Hande, E. Samper, P. Lansdorp, M.A. Blasco, Telomere length dynamics
and chromosomal instability in cells derived from telomerase null mice, J. Cell
Biol. 144 (1999) 589601.
[13] M.Z. Levy, R.C. Allsopp, A.B. Futcher, C.W. Greider, C.B. Harley, Telomere endreplication problem and cell aging, J. Mol. Biol. 225 (1992) 951960.
[14] R.C. Allsopp, H. Vaziri, C. Patterson, S. Goldstein, E.V. Younglai, A.B. Futcher,
C.W. Greider, C.B. Harley, Telomere length predicts replicative capacity of
human broblasts, Proc. Natl. Acad. Sci. U.S.A. 89 (1992) 1011410118.
[15] C.B. Harley, A.B. Futcher, C.W. Greider, Telomeres shorten during ageing of
human broblasts, Nature 345 (1990) 458460.
[16] D. Conomos, H.A. Pickett, R.R. Reddel, Alternative lengthening of telomeres:
remodeling the telomere architecture, Front. Oncol. 3 (2013) 27.
[17] P.J. Mason, M. Bessler, The genetics of dyskeratosis congenita, Cancer Genet.
204 (2011) 635645.
[18] M.W. Djojosubroto, Y.S. Choi, H.W. Lee, K.L. Rudolph, Telomeres and
telomerase in aging, regeneration and cancer, Mol. Cells 15 (2003) 164175.
[19] C.M. Counter, J. Gupta, C.B. Harley, B. Leber, S. Bacchetti, Telomerase activity in
normal leukocytes and in hematologic malignancies, Blood 85 (1995) 2315
2320.
[20] M. Engelhardt, K. Mackenzie, P. Drullinsky, R.T. Silver, M.A. Moore, Telomerase
activity and telomere length in acute and chronic leukemia, pre- and postex vivo culture, Cancer Res. 60 (2000) 610617.
[21] J. Albanell, F. Lonardo, V. Rusch, M. Engelhardt, J. Langenfeld, W. Han, D.
Klimstra, E. Venkatraman, M.A. Moore, E. Dmitrovsky, High telomerase activity
in primary lung cancers: association with increased cell proliferation rates and
advanced pathologic stage, J. Natl. Cancer Inst. 89 (1997) 16091615.

68

Y. Qian et al. / Cellular Immunology 289 (2014) 6369

[22] Y.H. Uen, S.R. Lin, D.C. Wu, Y.C. Su, J.Y. Wu, T.L. Cheng, C.W. Chi, J.Y. Wang,
Prognostic signicance of multiple molecular markers for patients with stage
II colorectal cancer undergoing curative resection, Ann. Surg. 246 (2007)
10401046.
[23] R.T. Calado, N.S. Young, Telomere maintenance and human bone marrow
failure, Blood 111 (2008) 44464455.
[24] S.E. Artandi, R.A. DePinho, Telomeres and telomerase in cancer, Carcinogenesis
31 (2010) 918.
[25] C. Gnes, K.L. Rudolph, The role of telomeres in stem cells and cancer, Cell 152
(2013) 390393.
[26] C.M. Kong, X.W. Lee, X. Wang, Telomere shortening in human diseases, FEBS J.
280 (2013) 31803193.
[27] R.B. Effros, G. Pawelec, Replicative senescence of T cells: does the Hayick
Limit lead to immune exhaustion?, Immunol Today 18 (1997) 450454.
[28] L. Yang, A. Mailloux, D.E. Rollison, J.S. Painter, J. Maciejewski, R.L. Paquette, T.P.
Loughran, K. McGraw, H. Makishima, R. Radhakrishnan, S. Wei, X. Ren, R.
Komrokji, A.F. List, P.K. Epling-Burnette, Naive T-cells in myelodysplastic
syndrome display intrinsic human telomerase reverse transcriptase (hTERT)
deciency, Leukemia 27 (2013) 897906.
[29] M.A. Moro-Garcia, R. Alonso-Arias, C. Lopez-Larrea, Molecular mechanisms
involved in the aging of the T-cell immune response, Curr. Genomics 13 (2012)
589602.
[30] N. Rufer, W. Dragowska, G. Thornbury, E. Roosnek, P.M. Lansdorp, Telomere
length dynamics in human lymphocyte subpopulations measured by ow
cytometry, Nat. Biotechnol. 16 (1998) 743747.
[31] N.P. Weng, B.L. Levine, C.H. June, R.J. Hodes, Human naive and memory T
lymphocytes differ in telomeric length and replicative potential, Proc. Natl.
Acad. Sci. U.S.A. 92 (1995) 1109111094.
[32] J. Monteiro, F. Batliwalla, H. Ostrer, P.K. Gregersen, Shortened telomeres in
clonally expanded CD28-CD8+ T cells imply a replicative history that is
distinct from their CD28+CD8+ counterparts, J. Immunol. 156 (1996) 3587
3590.
[33] R.J. Hodes, K.S. Hathcock, N.P. Weng, Telomeres in T and B cells, Nat. Rev.
Immunol. 2 (2002) 699706.
[34] D. Wolf, H. Rumpold, C. Koppelstatter, G.A. Gastl, M. Steurer, G. Mayer, E.
Gunsilius, H. Tilg, A.M. Wolf, Telomere length of in vivo expanded CD4(+)CD25
(+) regulatory T-cells is preserved in cancer patients, Cancer Immunol.
Immunother. 55 (2006) 11981208.
[35] E.V. Barsov, Telomerase and primary T cells: biology and immortalization for
adoptive immunotherapy, Immunotherapy 3 (2011) 407421.
[36] X. Shen, J. Zhou, K.S. Hathcock, P. Robbins, D.J. Powell Jr., S.A. Rosenberg, R.J.
Hodes, Persistence of tumor inltrating lymphocytes in adoptive
immunotherapy correlates with telomere length, J. Immunother. 30 (2007)
123129.
[37] N.P. Weng, K.S. Hathcock, R.J. Hodes, Regulation of telomere length and
telomerase in T and B cells: a mechanism for maintaining replicative potential,
Immunity 9 (1998) 151157.
[38] N.P. Weng, B.L. Levine, C.H. June, R.J. Hodes, Regulated expression of
telomerase activity in human T lymphocyte development and activation, J.
Exp. Med. 183 (1996) 24712479.
[39] K.S. Hathcock, N.P. Weng, R. Merica, M.K. Jenkins, R. Hodes, Cutting edge:
antigen-dependent regulation of telomerase activity in murine T cells, J.
Immunol. 160 (1998) 57025706.
[40] H. Fujii, L. Shao, I. Colmegna, J.J. Goronzy, C.M. Weyand, Telomerase
insufciency in rheumatoid arthritis, Proc. Natl. Acad. Sci. U.S.A. 106 (2009)
43604365.
[41] M. Tamayo, A. Mosquera, J.I. Rego, J.L. Fernandez-Sueiro, F.J. Blanco, J.L.
Fernandez, Differing patterns of peripheral blood leukocyte telomere length in
rheumatologic diseases, Mutat. Res. 683 (2010) 6873.
[42] T.T. Lin, B.T. Letsolo, R.E. Jones, J. Rowson, G. Pratt, S. Hewamana, C. Fegan, C.
Pepper, D.M. Baird, Telomere dysfunction and fusion during the progression of
chronic lymphocytic leukemia: evidence for a telomere crisis, Blood 116
(2010) 18991907.
[43] A. Rth, D. de Beer, H. Nckel, L. Sellmann, U. Dhrsen, J. Drig, G.M.
Baerlocher, Signicantly shorter telomeres in T-cells of patients with ZAP-70+/
CD38+chronic lymphocytic leukaemia, Br. J. Haematol. 143 (2008) 383386.
[44] C. Cifuentes-Rojas, D.E. Shippen, Telomerase regulation, Mutat. Res. Fundam.
Mol. Mech. Mutagen. 730 (2012) 2027.
[45] D.L. Qi, T. Ohhira, C. Fujisaki, T. Inoue, T. Ohta, M. Osaki, E. Ohshiro, T. Seko, S.
Aoki, M. Oshimura, H. Kugoh, Identication of PITX1 as a TERT suppressor gene
located on human chromosome 5, Mol. Cell. Biol. 31 (2011) 16241636.
[46] A. Cukusic, N. Skrobot Vidacek, M. Sopta, I. Rubelj, Telomerase regulation at the
crossroads of cell fate, Cytogenet. Genome Res. 122 (2008) 263272.
[47] C. Wang, X. Hou, S. Mohapatra, Y. Ma, W.D. Cress, W.J. Pledger, J. Chen,
Activation of p27Kip1 Expression by E2F1. A negative feedback mechanism, J.
Biol. Chem. 280 (2005) 1233912343.
[48] S. Kyo, M. Takakura, T. Taira, T. Kanaya, H. Itoh, M. Yutsudo, H. Ariga, M.
Inoue, Sp1 cooperates with c-Myc to activate transcription of the human
telomerase reverse transcriptase gene (hTERT), Nucleic Acids Res. 28 (2000)
669677.
[49] M. Daniel, G.W. Peek, T.O. Tollefsbol, Regulation of the human catalytic
subunit of telomerase (hTERT), Gene 498 (2012) 135146.
[50] N. Kikuno, H. Shiina, S. Urakami, K. Kawamoto, H. Hirata, Y. Tanaka, S. Majid,
M. Igawa, R. Dahiya, Genistein mediated histone acetylation and
demethylation activates tumor suppressor genes in prostate cancer cells, Int.
J. Cancer 123 (2008) 552560.

[51] M. Gladych, A. Wojtyla, B. Rubis, Human telomerase expression regulation,


Biochem. Cell Biol. 89 (2011) 359376.
[52] S. Kyo, M. Takakura, T. Fujiwara, M. Inoue, Understanding and exploiting
hTERT promoter regulation for diagnosis and treatment of human cancers,
Cancer Sci. 99 (2008) 15281538.
[53] S. Bougel, B. Lhermitte, G. Gallagher, J.C. de Flaugergues, R.C. Janzer, J.
Benhattar, Methylation of the hTERT promoter: a novel cancer biomarker for
leptomeningeal metastasis detection in cerebrospinal uids, Clin. Cancer Res.
19 (2013) 22162223.
[54] A. Chebel, J.P. Rouault, I. Urbanowicz, L. Baseggio, W.W. Chien, G. Salles, M.
Ffrench, Transcriptional activation of hTERT, the human telomerase reverse
transcriptase, by nuclear factor of activated T cells, J. Biol. Chem. 284 (2009)
3572535734.
[55] J.M. Terme, V. Mocquet, A.S. Kuhlmann, L. Zane, F. Mortreux, E. Wattel, M. Duc
Dodon, P. Jalinot, Inhibition of the hTERT promoter by the proto-oncogenic
protein TAL1, Leukemia 23 (2009) 20812089.
[56] E. Lamy, V. Goetz, M. Erlacher, C. Herz, V. Mersch-Sundermann,
HTERT: another brick in the wall of cancer cells, Mutat. Res. 752 (2013)
119128.
[57] S. Wu, A. Hultquist, P. Hydbring, C. Cetinkaya, F. Oberg, L.G. Larsson, TGF-beta
enforces senescence in Myc-transformed hematopoietic tumor cells through
induction of Mad1 and repression of Myc activity, Exp. Cell Res. 315 (2009)
30993111.
[58] D. Bai, L. Ueno, P.K. Vogt, Akt-mediated regulation of NFkappaB and the
essentialness of NFkappaB for the oncogenicity of PI3K and Akt, Int. J. Cancer
125 (2009) 28632870.
[59] Y.S. Cong, W.E. Wright, J.W. Shay, Human telomerase and its regulation,
Microbiol. Mol. Biol. Rev. 66 (2002) 407425 (table of contents).
[60] E.F. Wagner, A.R. Nebreda, Signal integration by JNK and p38 MAPK pathways
in cancer development, Nat. Rev. Cancer 9 (2009) 537549.
[61] M.Y. Alfonso-De Matte, H. Yang, M.S. Evans, J.Q. Cheng, P.A. Kruk, Telomerase is
regulated by c-Jun NH2-terminal kinase in ovarian surface epithelial cells,
Cancer Res. 62 (2002) 45754578.
[62] K. Hoffmeyer, A. Raggioli, S. Rudloff, R. Anton, A. Hierholzer, I. Del Valle, K.
Hein, R. Vogt, R. Kemler, Wnt/-catenin signaling regulates telomerase in stem
cells and cancer cells, Science 336 (2012) 15491554.
[63] M.A. Blasco, The epigenetic regulation of mammalian telomeres, Nat. Rev.
Genet. 8 (2007) 299309.
[64] C.W. Greider, Wnt regulates TERTputting the horse before the cart, Science
336 (2012) 15191520.
[65] I. Listerman, J. Sun, F.S. Gazzaniga, J.L. Lukas, E.H. Blackburn, The major reverse
transcriptase-incompetent splice variant of the human telomerase protein
inhibits telomerase activity but protects from apoptosis, Cancer Res. 73 (2013)
28172828.
[66] Y.J. Chiang, M.T. Hemann, K.S. Hathcock, L. Tessarollo, L. Feigenbaum, W.C.
Hahn, R.J. Hodes, Expression of telomerase RNA template, but not telomerase
reverse transcriptase, is limiting for telomere length maintenance in vivo, Mol.
Cell. Biol. 24 (2004) 70247031.
[67] C.J. Cairney, W.N. Keith, Telomerase redened: integrated regulation of hTR
and hTERT for telomere maintenance and telomerase activity, Biochimie 90
(2008) 1323.
[68] S.P. Atkinson, S.F. Hoare, R.M. Glasspool, W.N. Keith, Lack of telomerase gene
expression in alternative lengthening of telomere cells is associated with
chromatin remodeling of the hTR and hTERT gene promoters, Cancer Res. 65
(2005) 75857590.
[69] S. Shitara, M. Kakeda, K. Nagata, M. Hiratsuka, A. Sano, K. Osawa, A. Okazaki,
M. Katoh, Y. Kazuki, M. Oshimura, K. Tomizuka, Telomerase-mediated
life-span extension of human primary broblasts by human articial
chromosome (HAC) vector, Biochem. Biophys. Res. Commun. 369 (2008)
807811.
[70] M.K. Kang, N.H. Park, Extension of cell life span using exogenous telomerase,
Methods Mol. Biol. 371 (2007) 151165.
[71] N. Rufer, T.H. Brummendorf, S. Kolvraa, C. Bischoff, K. Christensen, L.
Wadsworth, M. Schulzer, P.M. Lansdorp, Telomere uorescence
measurements in granulocytes and T lymphocyte subsets point to a high
turnover of hematopoietic stem cells and memory T cells in early childhood, J.
Exp. Med. 190 (1999) 157167.
[72] T. Toptan, A. Ensser, H. Fickenscher, Rhadinovirus vector-derived human
telomerase reverse transcriptase expression in primary T cells, Gene Ther. 17
(2010) 653661.
[73] A. Roth, Telomere loss, senescence, and genetic instability in CD4+ T
lymphocytes overexpressing hTERT, Blood 106 (2005) 4350.
[74] N. Rufer, M. Migliaccio, J. Antonchuk, R.K. Humphries, E. Roosnek, P.M.
Lansdorp, Transfer of the human telomerase reverse transcriptase (TERT) gene
into T lymphocytes results in extension of replicative potential, Blood 98
(2001) 597603.
[75] M.W. Schreurs, M.A. Hermsen, R.I. Geltink, K.B. Scholten, A.A. Brink, E.W.
Kueter, M. Tijssen, C.J. Meijer, B. Ylstra, G.A. Meijer, E. Hooijberg, Genomic
stability and functional activity may be lost in telomerase-transduced human
CD8+ T lymphocytes, Blood 106 (2005) 26632670.
[76] R.M. Luiten, Ectopic hTERT expression extends the life span of human CD4+
helper and regulatory T-cell clones and confers resistance to oxidative stressinduced apoptosis, Blood 101 (2003) 45124519.
[77] O. Menzel, M. Migliaccio, D.R. Goldstein, S. Dahoun, M. Delorenzi, N. Rufer,
Mechanisms regulating the proliferative potential of human CD8+ T
lymphocytes overexpressing telomerase, J. Immunol. 177 (2006) 36573668.

Y. Qian et al. / Cellular Immunology 289 (2014) 6369


[78] L. Thadikkaran, O. Menzel, J.D. Tissot, N. Rufer, Proteomic and transcriptomic
analysis of human CD8(+) T lymphocytes over-expressing telomerase,
Proteomics Clin. Appl. 1 (2007) 299311.
[79] L.J. Berry, M. Moeller, P.K. Darcy, Adoptive immunotherapy for cancer: the next
generation of gene-engineered immune cells, Tissue Antigens 74 (2009) 277
289.

69

[80] A.F. Lee, P.A. Sieling, D.J. Lee, Immune correlates of melanoma survival in
adoptive cell therapy, Oncoimmunology 2 (2013) e22889.
[81] P. Muranski, N.P. Restifo, Adoptive immunotherapy of cancer using CD4(+) T
cells, Curr. Opin. Immunol. 21 (2009) 200208.

Vous aimerez peut-être aussi