Sei sulla pagina 1di 9

E XP E RI ME N T AL C ELL RE S EA RC H 319 (2013) 133–141

Available online at www.sciencedirect.com

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

Research Article

Telomeres—structure, function, and regulation

Weisi Lua, Yi Zhangb, Dan Liub, Zhou Songyanga,b,c,n, Ma Wanb,nn


a
State Key Laboratory for Biocontrol, SYSU, Guangzhou, PR China
b
Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston,
TX 77030, USA
c
Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA

article information abstract

Article Chronology: In mammals, maintenance of the linear chromosome ends (or telomeres) involves faithful
Received 16 August 2012 replication of genetic materials and protection against DNA damage signals, to ensure genome
Accepted 13 September 2012 stability and integrity. These tasks are carried out by the telomerase holoenzyme and a unique
Available online 21 September 2012 nucleoprotein structure in which an array of telomere-associated proteins bind to telomeric
Keywords: DNA to form special protein/DNA complexes. The telomerase complex, which is comprised of
Telomere telomeric reverse transcriptase (TERT), telomeric RNA component (TERC), and other assistant
Telomerase factors, is responsible for adding telomeric repeats to the ends of chromosomes. Without proper
Telosome telomere maintenance, telomere length will shorten with successive round of DNA replication
Telomere maintenance due to the so-called end replication problem. Aberrant regulation of telomeric proteins and/or
End protection telomerase may lead to abnormalities that can result in diseases such as dyskeratosis congenita
Extra-telomeric (DC) and cancers. Understanding the mechanisms that regulate telomere homeostasis and the
Telomere dysfunction factors that contribute to telomere dysfunction should aid us in developing diagnostic and
therapeutic tools for these diseases.
& 2012 Elsevier Inc. All rights reserved.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Telomere structure and maintenance . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Telomeric DNA and G-quadruplex . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 134
Telomerase and its regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 135
Telomere dysfunction and the p53/p16 pathways. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Telomeres and pluripotency. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
Telomere interactome—protein–protein interacting networks at telomeres . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
TRF1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 136
TRF2 and RAP1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
POT1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137

n
Corresponding author at: Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
nn
Corresponding author.
E-mail addresses: songyang@bcm.edu (Z. Songyang), mw6@bcm.edu (M. Wan).

0014-4827/$ - see front matter & 2012 Elsevier Inc. All rights reserved.
http://dx.doi.org/10.1016/j.yexcr.2012.09.005
134 E XP E RI ME N T AL C E L L RE S E AR CH 319 (2013) 133–141

TPP1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
TIN2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 137
Extra-telomeric function of telomere proteins. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 138

Introduction homologous end joining (NHEJ; Fig. 1) [13]. Indeed, genetic and
epigenetic aberrance in telomere-associated proteins, telomerase,
and epigenetic enzymes can result in abnormal telomere
In the 1930s, by comparing X-ray generated break ends of
length, telomere damage induced foci (TIF), telomere end–end
chromosomes to natural ones in fly and maize, Muller and
fusion, and ultimately cell cycle arrest, senescence, and genome
McClintock concluded that natural chromosome ends could
instability [13,14]. In humans, telomere dysfunction has been
shield chromosomes from rearrangement/fusion events that
implicated in bone marrow failure syndromes, leukemia, and
often occurred between intra-chromosomal breaks [1,2]. Later
cancer development [9,10,15–17]. Understanding the mechan-
studies in Tetrahymena and yeast showed that the chromosome
isms by which telomeric DNA, telomerase, and telomere-
ends were protected by tandem repeats of hexanucleotide units
interacting proteins function together will undoubtedly facilitate
that could function across species, suggesting evolutionary and
the development of diagnostic and therapeutic tools for
functional conservation [3–6]. In the mid-1980s, the seminal
telomere-relevant diseases. In this review, we will focus mainly
work from Blackburn and Greider, which demonstrated the
on the achievement and progress made in telomere biology by
existence of an enzymatic activity within cell extracts that added
Chinese scientists.
tandem hexanucleotides to natural chromosome ends, eventually
led to the discovery of telomerase [7]. And the past decade has
seen leaps and bounds in our understanding of the various Telomere structure and maintenance
components in controlling telomere homeostasis [8–12].
Telomere maintenance requires the telomerase and a network Telomeric DNA and G-quadruplex
of telomere-associated proteins, and necessitates the inhibition Telomeres are comprised of repeat sequences and bound by
of various DNA-damage response (DDR) signals that include ATM multiple telomeric interacting proteins. In mammalian cells,
and ATR pathways, homologous recombination (HR), and non- telomere DNA contains double-stranded tandem repeats of

Fig. 1 – Canonical and extra-telomeric function of telomere-binding proteins. The six-protein telosome complex and telomerase
holoenzyme are depicted in the small box. In the nucleus, the telosome and telomerase complex maintain telomere length and
protect telomere ends. Several telomere proteins also participate in non-telomeric pathways in the cytoplasm.
EX P ER IM E NTA L CE LL R ES E A RC H 319 (2013) 133–141 135

TTAGGG followed by terminal 30 G-rich single-stranded over- providing a novel tool for telomerase studies [26]. This study also
hangs. Telomere DNA is thought to adopt the T-loop structure, revealed that the length of G overhangs of lagging strands
where the telomere end folds back on itself and the 30 G strand appeared longer than those of leading strands in human BJ cells
overhang invades into the double-stranded DNA (the so-called D- [26]. It should be noted that the telomerase is thought to be
loop) [8]. preferentially recruited to the shortest telomeres for length
Because of its enriched G content, the single-stranded telomere maintenance [27]. Recent work from Zhao et al. challenges this
G overhangs can form G-quadruplexes, where each G base serves model by showing that telomerase extends most chromosome
as both donor and an acceptor for hydrogen bond formation. ends during each S phase and uncouples from subsequent
In humans, telomeric G-quadruplex structures have been impli- C-strand fill-in process in human cancer cells. These findings
cated in telomere protection, suppression of recombination, and suggest step-wise telomere maintenance pathways and may
inhibition of telomerase-dependent telomere extension [18]. Tan open up a new avenue of research for anti-telomerase therapeu-
and colleagues from Institute of Zoology at Chinese Academy tics [28].
of Sciences have been systematically exploring the impact of
different physical and chemical conditions on the stability of the Telomerase and its regulation
telomeric G-quadruplex. For instance, they have observed that The telomerase is a unique ribonucleoprotein complex that
human telomere DNA forms a parallel-stranded conformation consists of the telomerase reverse transcriptase (TERT), and a
under molecular crowding conditions that mimic physiological telomerase RNA component (TERC) that serves as the template
environment [19]. Using different approaches, including a real- for telomere extension during de novo addition of TTAGGG
time fluorescence assay, the group provides evidence that the repeats onto chromosome ends (Fig. 1) [29]. While TERC expres-
unwinding of intramolecular telomere G-quadruplex structures sion is ubiquitous, TERT expression appears highly regulated. For
by BLM and other RecQ family of helicases would be more example, telomerase is expressed in stem cell compartments and
difficult than unwinding of duplexes, supporting the role of in embryonic stem cells, but TERT expression and telomerase
G-quadruplexes in telomere protection [20]. activity are often very low or undetectable in somatic cells [30].
Previous work has demonstrated that four TTAGGG repeats can In contrast, in most cancer cells (85–90%) telomerase activity
fold into a G-quadruplex and influence telomere elongation appears elevated [31–33]. In mice, deletion of either TERC or
in vitro, suggesting that G-quadruplexes are poor substrates for TERT leads to telomere shortening, genomic instability, aneu-
the telomerase [21]. By DMS footprinting and exonuclease ploidy, telomeric fusion, and aging-related phenotypes [34,35]. In
hydrolysis, the Tan group showed that the G-quadruplex pre- comparison, overexpression of TERT can dramatically increase
ferentially forms at the very 30 end of telomeres where it may the life span of mice in the context of overexpressing tumor
inhibit telomerase accessibility for telomere extension [22]. End suppressor genes such as p53, p16, and p19 [36]. Therefore,
processing enzymes such as telomerase and helicases likely telomerase dysfunction may result in defects in various highly
require a free single-stranded 30 telomere end. By analyzing the proliferative cells/tissues and ultimately lead to degenerative
size of the 30 tails required for different end reactions, Tan and diseases, such as dyskeratosis congenita (DC).
his colleagues have demonstrated that a minimal tail of 6, 8, and Following the cloning and complete sequencing of human
12 nt is required respectively for telomere G-quadruplex TERT (hTERT) [37], extensive efforts have shifted to studying
unwinding, telomere extension by telomerase, and the alterna- how TERT expression is regulated. With a focus on the effects of a
tive lengthening of telomere (ALT) mechanism [23]. Due to the variety of factors on telomerase regulation, Liu and colleagues
farthest 30 distal end of telomere DNA leaving a tail of r5 nt, have found that the C-terminus of p53 interacts with the
their study also suggests that telomere G-quadruplexes may N-terminus of human telomerase-associated protein 1 (hTEP1)
regulate the aforementioned end reactions at chromosomal ends and inhibits telomerase activity in vitro [38]. In agreement with
[23]. Taken together, telomere G-quadruplex structures regulate this finding, p53 has been shown to interact with SP1 and inhibit
the action of multiple enzymes, including the telomerase, at the its targeting to the hTERT promoter for transactivation [39]. They
very end of chromosomes for telomere length regulation and end also showed that nerve growth factor (NGF)-induced inhibition
protection, and may therefore serve as a potential drug target for of telomerase occurred in a MAP kinase signaling-dependent
aging and cancer therapies. manner in PC12 phenochromocytoma cells [40], and the TGF-
In yeast, the telomerase subunit Est1p has recently been beta signaling downstream transcription factor Smad3 can be
shown to be capable of converting single-stranded telomeric recruited to the TERT promoter for transcription repression [41].
G-rich DNA into a G-quadruplex structure in vitro in a magnesium- Further studies have demonstrated that bone morphogenetic
dependent manner, and required for telomerase-mediated telo- protein-7 (BMP7) induces Smad3 phosphorylation, nuclear trans-
mere protection [24,25]. In these studies by the Zhou group, Est1p location, and hTERT gene repression, resulting in telomere short-
mutants can disrupt G-quadruplex in vitro and lead to telomere ening, tumor growth arrest, senescence, and cell death [42,43].
shortening and cellular senescence in vivo, suggesting a positive More recently, the Liu group discovered that, under oxidative
role for G-quadruplex in telomere length regulation [24]. It is clear stress, glyceraldehyde 3-phosphate dehydrogenase (GAPDH)
that further studies are required to tease out the functional could interact with TERC and lead to the inhibition of telomerase
differences of G-quadruplex in regulating telomerase and telomere activity, telomere shortening, thereby inducing cancer cell senes-
length between humans and other organisms. cence [44].
The length of 30 G-overhang is critical for end processing and Epigenetic modification of histones can modulate chromatin
telomerase regulation. Taking advantage of a double-strand structure and accessibility of the transcriptional machinery to
specific nuclease (DSN), Zhao et al. developed an approach to regulatory regions of target genes. Based on reporter assays and
measure the exact size of single-stranded 30 G overhangs, thus results from using the histone deacetylase inhibitor trichostatin
136 E XP E RI ME N T AL C E L L RE S E AR CH 319 (2013) 133–141

A (TSA), histone deacetylation has been suggested to repress self-renewal of iPSCs [62]. Moreover, telomerase re-activation
hTERT expression by modulating histone acetylation on the TERT appears to occur earlier than pluripotent genes (Oct4 and Nanog)
promoter in human cells [45]. In addition, numerous transcrip- during reprogramming, suggesting that telomere regulation is
tion factors, such as c-MYC, SP1, MAD1, and HIF-2a, have been both a critical step for generating pluripotency and essential for
shown to recruit either histone acetyltransferases (HATs) or maintaining stem cell-based tissue homeostasis [62].
histone deacetylases (HDACs) to the promoter of TERT to control
its expression [46–48]. Interestingly, the Xu group showed that
histone H3K4 methyltranferase SMYD3 and histone demethylase Telomere interactome—protein–protein interacting
LSD1 together with HDACs could directly activate and repress networks at telomeres
TERT expression, respectively [49,50]. Furthermore, MAD1 has
been shown to recruit RBP2 (H3K4 demethylase) to TERT Telomere maintenance relies on a vast network of protein
promoter for its repression [51]. Recent work from Xu and complexes at the telomeres. Central to this process is a six-
colleagues also indicates that MAP kinase cascade-mediated protein complex (Fig. 1), known as the telosome or shelterin
phosphorylation of histone H3 Ser10 at the TERT promoter [63,64]. The telosome is composed of the telomeric repeat-
triggers telomerase transcriptional activation in proliferating binding protein 1 and 2 (TRF1 and TRF2), the TRF1-interacting
cells [52]. Additionally, Song et al. also showed that the systemic protein 2 (TIN2), RAP1, protection of telomeres 1 (POT1), and
environment and stem cell niche are major reasons for the TPP1 [63,64]. TRF1 and TRF2 share a common domain structure
impairment of lymphopoiesis in aging telomerase knockout mice consisting of an N-terminal TRFH domain, and a C-terminal
[53], suggesting additional avenues for anti-aging therapy in the SANT/Myb domain with high binding specificity for the half site
context of telomere dysfunction. Taken together, these studies 50 -YTAGGGTTR-30 in telomeric double-stranded DNA (dsDNA)
have highlighted unique possibilities of exploring factors from [64]. The two N-terminal OB-folds of POT1 are highly specific for
extracellular environment, intracellular signaling cascades, tran- the 50 -TAGGGTTAG-30 sequence of single-stranded G-overhangs
scription factors, and epigenetic components for anti-aging and [65]. TIN2 serves as a hub to bridge various telosome compo-
anti-cancer therapies. nents by directly binding to TRF1, TRF2, and the TPP1/POT1
heterodimer [11]. TPP1 directly interacts with TIN2 and POT1 as
Telomere dysfunction and the p53/p16 pathways well as the telomerase [11,66]. Mammalian RAP1 is targeted to
Telomere dysfunction could lead to accumulated damage at the telomeric DNA by directly interacting with TRF2 [11].
telomeres and cellular senescence and death. In fact, telomere Together, the six core proteins function as the platform that
damage is a key factor in cellular senescence [54]. p53 and p16- recruits players from diverse pathways to the telomeres for
mediated signaling cascades are two of the critical pathways maintenance and protection. This is evident in a recent study
implicated in telomere dysfunction and cellular senescence. In from our group where we carried out a BiFC-based genome-wide
particular, work from the groups of Tong and Zhang has been screen for interaction partners of the six core telomere proteins
instrumental to our understanding in these widely studied areas. in live human cells [12]. We identified 4300 proteins, including
They have carried out pioneer work to reveal connections numerous novel telomere-associated proteins. Thanks to the
between p16/p21/p53 regulation, telomere function, aging, and unique properties of the BiFC strategy, a number of weak/
cancer progression [55–58]. The group has recently shown that transient telosome interaction proteins, such as protein kinases
HDTIC compounds from the traditional Chinese medicine Astra- and ubiquitin E3 ligases, were identified [12]. These findings
galus membranaceous can prevent telomere shortening, enhance underscore the complexity of the telomere maintenance network
DNA repair ability, and delay senescence through attenuating and the amount of work still needed to understand these
oxidative stress, suggesting new mechanisms and agents for anti- pathways.
aging therapy [59]. In support of these findings, the Blasco group
has also shown that TA-65, a small molecule from A. membranaceous, TRF1
is capable of activating the telomerase, elongating telomeres,
inhibiting DNA damage, and extending tumor-free life-span of The first double-stranded telomere DNA binding protein identi-
mice [60]. fied [67], TRF1 acts as a negative regulator for telomere length
[68]. Homozygous deletion of TRF1 in mice led to embryonic
Telomeres and pluripotency lethality around the blastocyst stage with severe growth defects
Pluripotent cells such as embryonic stem cells (ESCs) and accompanied by apoptosis [69]. The absence of telomerase could
induced pluripotent cells (iPSCs) hold unrivaled promise for not rescue this phenotype, suggesting essential function of TRF1
regenerative medicine, and have been the subjects of intense that may be independent of telomere length regulation [69].
studies, including those that focus on the role of telomeres in Through crystal structure analysis, Lei and colleagues demon-
developmental pluripotency. For instance, the Liu group from strated that TRF1 could recognize TIN2 through its TRFH domain,
Nan Kai University showed that ESCs with long telomeres and the TRFH domain of TRF1 (not TRF2) interacts with PinX1
exhibited bona fide developmental pluripotency, in terms of [70]. Their more recent work further revealed that the atypical
generating complete ESC pups and germline-competent chimeras GTPase domain of Fbx4 could serve as a substrate-binding motif
[61]. These observations suggest that functional telomeres are for the SCF E3 ligase complex and bind to a globular domain of
essential for maintaining pluripotency in ESCs/iPSCs, and possi- TRF1 for its ubiquitination and degradation. These findings
ble markers for evaluating pluripotency. Using iPSCs derived provide clues to the underlying mechanism for controlling
from telomerase-deficient cells, the group went on to demon- TRF1, whose expression is tightly regulated, as well as telomere
strate that telomerase plays a critical role in reprogramming and homeostasis [71].
EX P ER IM E NTA L CE LL R ES E A RC H 319 (2013) 133–141 137

TRF2 and RAP1 elongation [84–86]. In contrast to human POT1, mice have two
Pot1 orthologs with distinct function—Pot1a and Pot1b [85,86].
Originally identified through homology search [72], TRF2 has Loss-of-function studies for Pot1a and/or Pot1b in mice and in
emerged as a critical player in telomere maintenance— cells have revealed that Pot1a mainly represses the ATR-
negatively regulating telomere length [73] and participating in mediated DNA damage response, while Pot1b regulates 50 -end
telomere end protection. These activities are carried out at least resection [85,86]. Human POT1 is thought to combine the
in part through the multitude of factors that interact with TRF2, features of mouse POT1a and POT1b [87]. A recent study on
such as Apollo, ERCC1/XRF, the DNA repair MRN complex, WRN, mouse Pot1b indicates that it can inhibit excessive 50 -resection
and FEN1 [70,74]. Using biochemical approaches, our group and by interacting with the CST (Ctc1/Stn1/Ten1) complex during
the Lei group discovered that the TRFH domain within human 30 -overhang generation [88]. These observations have led to the
TRF2 could recognize [Y/F]XL peptides with the consensus motif current model, where POT1 may exclude RPA, major ssDNA
of YYHKYRLSPL [70,75]. A number of novel TRF2 TRFH interactors binding proteins, for G-overhang binding and thereby inhibit
were identified based on the binding motif, including phospha- ATR-mediated DNA damage signals. In support of this model, the
tase nuclear targeting subunit (PNUTS) and microcephalin 1 Zou group recently demonstrated that hnRNPA1, TERRA (telo-
(MCPH1) [75]. We provided evidence that PNUTS and MCPH1 meric repeat-containing RNA), and POT1 could act together to
could localize to telomeres, and respectively control telomere displace RPA from telomeric ssDNA after DNA replication, and
length and DNA damage responses at the telomeres [75]. In promote telomere end protection [89].
addition, DDX39 (DEAD-box RNA helicase) has been reported to
directly interact with TRF2 via the FXLXP motif and depletion of TPP1
DDX39 leads to induction of TIF (telomere dysfunction-induced
foci), adding another pathway that connects TRF2 to DDR Our group first identified TPP1 (known as PTOP then) through
inhibition [76]. It is clear that TRF2 acts as a hub to recruit large-scale affinity purification and found it to interact with both
various factors for telomere regulation. One such example is the TIN2 and POT1 [90]. Work from us and other groups has since
ATM-mediated NHEJ pathway. TRF2-deficient MEFs show mas- shown that human TPP1 binds to the c-terminus of POT1 and is
sive end-to-end fusions mediated by the NHEJ pathway that lead required for POT1 telomere localization [90–92]. TPP1 turns out
to severe proliferation defects [77]. Similar to TRF1, homozygous to be the mammalian homolog of Oxytricha nova TEBPb and
inactivation of TRF2 in mice is embryonic lethal. Unlike TRF1, contains an N-terminal OB fold [93]. Indeed, structural analysis
however, the TRF2 lethality cannot be rescued by p53 abrogation, revealed the similarities between human TPP1 OB fold and that
indicating that different mechanisms are utilized by TRF1 and of TEBPb, suggesting that the TPP1/POT1 heterodimer is the
TRF2 to ensure survival during embryonic development [77]. homolog of TEBPa/b heterodimer [66]. As is the case for TEBPa/b,
RAP1 appears highly conserved, with a RCT domain, a BRCT TPP1 directly interacts with POT1 and enhances POT1 affinity for
domain, and Myb domain(s) [8]. Unlike its budding yeast telomeric ssDNA [66,93]. Furthermore, TPP1 directly interacts
counterpart that can directly bind telomeres through its Myb with the telomerase for its recruitment to telomeres [66,93]. For
domain, mammalian RAP1 lacks telomere-binding capacity and example, decreased telomerase binding to telomeres and telo-
depends on its interacting partner TRF2 for telomere localization mere shortening have been detected in TPP1 null MEFs and mice
[8,78]. Through large-scale affinity purification and mass spec- [91]. Moreover, TPP1-deficient MEFs are unable to be repro-
trometry, our group identified numerous DNA repair proteins grammed to iPSCs, a process that is dependent on telomerase
(Rad50, Mre11, PARP1, and Ku86/Ku70) in the RAP1/TRF2 com- activity [91]. Notably, in acd mice which are spontaneously
plex [79]. In fact, recent studies have shown that the DNA repair occurring mutants that are haploid-insufficient for mouse TPP1,
factor BTBD12 complexes with TRF2/RAP1 and facilitates Holli- growth defects are apparent in various highly proliferative and
day junction processing and DNA damage response, suggesting telomerase-positive tissues, such as hair follicle stem cell com-
its potential role at repressing HR at telomeres [80]. In contrast partments [94]. Using the zebrafish model, our group has also
to TRF1 and TRF2, RAP1-deficient mice are viable, although with shown that depletion of zfTPP1 leads to multiple abnormalities
increased telomere recombination and fragility [81]. in embryogenesis, including neural death, heart malformation,
caudal defect, and extensive apoptosis, suggesting functional
POT1 conservation of this protein [95]. Moreover, TPP1 has also been
shown to interact with OBFC1/Stn1, an OB-fold protein that
POT1 was originally cloned based on sequence homology to the directly binds to ss-telomeric DNA, for telomere length control
single-stranded G-rich DNA-binding protein in ciliated protozoa [96]. In fact, a recent study revealed that the OBFC1/Stn1-
[82]. It was later revealed by Lei et al. that the N-terminus of containing CST complex is involved in 50 -end resection for 30 -
yeast Pot1p (Pot1pN) could adopt an oligonucleotide/oligosac- overhang generation and depletion of OBFC1/Stn1 leads to
charide-binding (OB) fold with two protruding loops that form a telomere elongation [88,97]. Taken together, TPP1 is essential
clamp for ssDNA binding [83]. In comparison, the two OB folds of for both telomere end protection and length regulation, through
human POT1 (hPOT1) recognize the telomeric single-stranded repressing ATR-mediated DNA damage signaling and modulating
DNA (ssDNA) decamer TTAGGGTTAG, with the N-terminal OB telomerase-dependent telomere elongation.
fold of hPOT1 binding the first six nucleotides, while the second
OB fold binds and protects the 30 end of ssDNA [65]. TIN2
POT1 has been shown to protect telomere ends from ATR-
dependent DNA damage response, control 50 -end resection at By directly interacting with TRF1, TRF2, and TPP1 [11], TIN2 acts as
telomere termini, and regulate telomerase-dependent telomere the central component in the telosome complex [98]. Here, TIN2
138 E XP E RI ME N T AL C E L L RE S E AR CH 319 (2013) 133–141

disruption results in significantly decreased telomere localization of convergence of diverse fields, we are gaining a more compre-
all telosome components, accumulated RPA binding to telomere hensive understanding of the pathways and players involved in
termini, and increased ATR-mediated DNA damage responses, which ensuring telomere integrity. In particular, work from China and
also mirror the phenotypes in POT1a/1b double knockout mice [99]. Chinese scientists has contributed tremendously to the advance-
TIN2 deletion in mice leads to embryonic lethality, and data from ment of telomere-related research. Further work on the mechan-
TIN2-deficient cells further support a role of TIN2 in facilitating isms of telomerase regulation, telomere-binding protein
TRF2-dependent inhibition of ATM-mediated DDR, and in telomerase function, and the interplay between telomeres and other cellular
recruitment and telomere length regulation [99]. To date, TIN2 is the compartments should greatly facilitate our understanding of
only telosome component with identified mutations in human telomere-relevant diseases and our search for appropriate diag-
diseases. In patients with dyskeratosis congenita (DC), dysfunction nostic and therapeutic tools.
in TIN2-dependent telomere length control and TPP1-mediated
telomerase recruitment may be manifested. Expression of TIN2 with
missense mutations found in DC patients could recapitulate the
Acknowledgment
telomere shortening phenotype observed in patients [100], making
TIN2 a possible target for diagnostic and therapeutic studies.
We would like to apologize to those whose work cannot be cited
here due to space limitations. This work is supported by National
Basic Research Program (973 Program 2010CB945400 and
Extra-telomeric function of telomere proteins
2012CB911200), NSFC 91019020, NCI CA133249, NIGMS
GM081627, GM095599, and the Welch Foundation Q-1673. We
Non-canonical function of telomerase and telomere proteins has
would also like to acknowledge the support of the Dan L. Duncan
been garnering increasing attention lately. Accumulating evi-
Cancer Center (P30CA125123), and the Administrative and
dence suggests a broader role for these proteins outside the
Genome-wide RNAi Screens Cores (IDDRC P30HD024064).
telomeres (Fig. 1). For example, human TIN2, TPP1, and POT1 can
localize and interact in the cytoplasm [101]. And TRF2 has been
implicated in regulating the proliferation and differentiation of references
neural tumor and stem cells [27,102,103]. In fact, TRF2 appears
to play an important role in homologous recombination (HR)
repair of double-strand breaks at non-telomeric regions [1] B. McClintock, H.E. Hill, The cytological identification of the
[104,105]. Furthermore, extra-telomeric RAP1 binding sites chromosome associated with the R–G linkage group in Zea mays,
Genetics 16 (1931) 175–190.
appeared enriched in subtelomeric regions and in genes deregu-
[2] E.H. Blackburn, C.W. Greider, J.W. Szostak, Telomeres and
lated as a result of RAP1 deletion in mouse [81]. Recently, human telomerase: the path from maize, tetrahymena and yeast to
RAP1 was found to associate with ikB kinases in the cytoplasm human cancer and aging, Nat. Med. 12 (2006) 1133–1138.
and act as a crucial regulator of NF-kB modulated gene expres- [3] M.C. Yao, E. Blackburn, J.G. Gall, Amplification of the rRNA genes
sion [107]. Our genome-wide ChIP-seq data on human RAP1 and in tetrahymena, Cold Spring Harb. Symp. Quant. Biol. 43 (Pt 2)
TRF2 indicate that in addition to telomere sequences, telomere (1979) 1293–1296.
[4] M.C. Yao, E. Blackburn, J. Gall, Tandemly repeated C-C-C-C-A-A
proteins can also target interstitial sites, possibly to regulate
hexanucleotide of tetrahymena rDNA is present elsewhere in
transcription [106]. Another group reached similar conclusions
the genome and may be related to the alteration of the somatic
through their studies on TRF1 and TRF2 [108]. It has also been genome, J Cell. Biol. 90 (1981) 515–520.
reported that the telomerase contains mitochondria targeting [5] E.H. Blackburn, J.G. Gall, A tandemly repeated sequence at the
signals, and exogenously expressed telomerase can localize to termini of the extrachromosomal ribosomal RNA genes in
the mitochondria and regulate apoptotic responses to oxidative tetrahymena, J. Mol. Biol. 120 (1978) 33–53.
stress [109–111]. [6] J.W. Szostak, E.H. Blackburn, Cloning yeast telomeres on linear
plasmid vectors, Cell 29 (1982) 245–255.
Recently, our group has demonstrated that the N-terminus of
[7] C.W. Greider, E.H. Blackburn, Identification of a specific telo-
TIN2 harbors mitochondrial localization signals, which can target
mere terminal transferase activity in tetrahymena extracts, Cell
endogenous TIN2 to the mitochondria [112]. Knocking down 43 (1985) 405–413.
TIN2 by RNAi led to metabolic changes such as enhanced oxygen [8] W. Palm, T. de Lange, How shelterin protects mammalian
consumption and mitochondrial ATP synthesis, implicating TIN2 telomeres, Annu. Rev. Genet. 42 (2008) 301–334.
in metabolic control. These results point to a novel connection [9] P. Martinez, M.A. Blasco, Role of shelterin in cancer and aging,
between telomeric proteins and metabolism, and the possibility Aging Cell 9 (2010) 653–666.
[10] S.E. Artandi, R.A. DePinho, Telomeres and telomerase in cancer,
that TIN2 represents a new factor that may regulate the switch
Carcinogenesis 31 (2010) 9–18.
between aerobic glycolysis and oxidative phosphorylation. [11] H. Xin, D. Liu, Z. Songyang, The telosome/shelterin complex and
Further studies may provide additional evidence to link other its functions, Genome Biol. 9 (2008) 232.
telomere-associated proteins in addition to TIN2 to metabolism [12] O.H. Lee, H. Kim, Q. He, H.J. Baek, D. Yang, L.Y. Chen, J. Liang,
and other extra-telomeric function. H.K. Chae, A. Safari, D. Liu, Z. Songyang, Genome-wide YFP
fluorescence complementation screen identifies new regulators
for telomere signaling in human cells, Mol. Cell Proteomics 10
(2011) M110001628.
Conclusion [13] T. de Lange, How telomeres solve the end-protection problem,
Science 326 (2009) 948–952.
The field has come a long way since the discovery of telomeres [14] M.A. Blasco, The epigenetic regulation of mammalian telomeres,
and telomerase. With more sophisticated techniques and the Nat. Rev. Genet. 8 (2007) 299–309.
EX P ER IM E NTA L CE LL R ES E A RC H 319 (2013) 133–141 139

[15] R.T. Calado, N.S. Young, Telomere diseases, N. Engl. J. Med. 361 [37] Y.S. Cong, J. Wen, S. Bacchetti, The human telomerase catalytic
(2009) 2353–2365. subunit hTERT: organization of the gene and characterization of
[16] R.T. Calado, N.S. Young, Telomere maintenance and human bone the promoter, Hum. Mol. Genet. 8 (1999) 137–142.
marrow failure, Blood 111 (2008) 4446–4455. [38] H. Li, Y. Cao, M.C. Berndt, J.W. Funder, J.P. Liu, Molecular
[17] P.J. Mason, M. Bessler, The genetics of dyskeratosis congenita, interactions between telomerase and the tumor suppressor
Cancer Genet. 204 (2011) 635–645. protein p53 in vitro, Oncogene 18 (1999) 6785–6794.
[18] H.J. Lipps, D. Rhodes, G-quadruplex structures: in vivo evidence [39] D. Xu, Q. Wang, A. Gruber, M. Bjorkholm, Z. Chen, A. Zaid,
and function, Trends Cell Biol. 19 (2009) 414–422. G. Selivanova, C. Peterson, K.G. Wiman, P. Pisa, Downregulation
[19] Y. Xue, Z.Y. Kan, Q. Wang, Y. Yao, J. Liu, Y.H. Hao, Z. Tan, Human of telomerase reverse transcriptase mRNA expression by wild
telomeric DNA forms parallel-stranded intramolecular G- type p53 in human tumor cells, Oncogene 19 (2000) 5123–5133.
quadruplex in Kþ solution under molecular crowding condition, [40] H. Li, A.R. Pinto, W. Duan, J. Li, B.H. Toh, J.P. Liu, Telomerase
J. Am. Chem. Soc. 129 (2007) 11185–11191. down-regulation does not mediate PC12 pheochromocytoma
[20] J.Q. Liu, C.Y. Chen, Y. Xue, Y.H. Hao, Z. Tan, G-quadruplex hinders cell differentiation induced by NGF, but requires MAP kinase
translocation of BLM helicase on DNA: a real-time fluorescence signalling, J. Neurochem. 95 (2005) 891–901.
spectroscopic unwinding study and comparison with duplex [41] H. Li, D. Xu, J. Li, M.C. Berndt, J.P. Liu, Transforming growth
substrates, J. Am. Chem. Soc. 132 (2010) 10521–10527. factor beta suppresses human telomerase reverse transcriptase
[21] A.M. Zahler, J.R. Williamson, T.R. Cech, D.M. Prescott, Inhibition (hTERT) by Smad3 interactions with c-Myc and the hTERT gene,
of telomerase by G-quartet DNA structures, Nature 350 (1991) J. Biol. Chem. 281 (2006) 25588–25600.
718–720. [42] L. Cassar, H. Li, A.R. Pinto, C. Nicholls, S. Bayne, J.P. Liu, Bone
[22] J. Tang, Z.Y. Kan, Y. Yao, Q. Wang, Y.H. Hao, Z. Tan, G-quadruplex morphogenetic protein-7 inhibits telomerase activity, telomere
preferentially forms at the very 30 end of vertebrate telomeric maintenance, and cervical tumor growth, Cancer Res. 68 (2008)
DNA, Nucleic Acids Res. 36 (2008) 1200–1208. 9157–9166.
[23] Q. Wang, J.Q. Liu, Z. Chen, K.W. Zheng, C.Y. Chen, Y.H. Hao, [43] L. Cassar, C. Nicholls, A.R. Pinto, H. Li, J.P. Liu, Bone morphoge-
Z. Tan, G-quadruplex formation at the 30 end of telomere DNA netic protein-7 induces telomerase inhibition, telomere short-
inhibits its extension by telomerase, polymerase and unwinding ening, breast cancer cell senescence, and death via Smad3,
by helicase, Nucleic Acids Res. 39 (2011) 6229–6237. FASEB J. 23 (2009) 1880–1892.
[24] M.L. Zhang, X.J. Tong, X.H. Fu, B.O. Zhou, J. Wang, X.H. Liao, Q.J. Li, [44] C. Nicholls, A.R. Pinto, H. Li, L. Li, L. Wang, R. Simpson, J.P. Liu,
N. Shen, J. Ding, J.Q. Zhou, Yeast telomerase subunit Est1p has Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) induces
guanine quadruplex-promoting activity that is required for telo- cancer cell senescence by interacting with telomerase RNA
mere elongation, Nat. Struct. Mol. Biol. 17 (2010) 202–209. component, Proc. Natl. Acad. Sci. USA (2012).
[25] X.J. Tong, Q.J. Li, Y.M. Duan, N.N. Liu, M.L. Zhang, J.Q. Zhou, Est1 [45] Y.S. Cong, S. Bacchetti, Histone deacetylation is involved in the
protects telomeres and inhibits subtelomeric y0 -element transcriptional repression of hTERT in normal human cells,
recombination, Mol. Cell Biol. 31 (2011) 1263–1274. J. Biol. Chem. 275 (2000) 35665–35668.
[26] Y. Zhao, H. Hoshiyama, J.W. Shay, W.E. Wright, Quantitative [46] D. Xu, N. Popov, M. Hou, Q. Wang, M. Bjorkholm, A. Gruber,
telomeric overhang determination using a double-strand spe- A.R. Menkel, M. Henriksson, Switch from Myc/Max to Mad1/
cific nuclease, Nucleic Acids Res. 36 (2008) e14. Max binding and decrease in histone acetylation at the telo-
[27] A. Bianchi, D. Shore, How telomerase reaches its end: mechan- merase reverse transcriptase promoter during differentiation of
ism of telomerase regulation by the telomeric complex, Mol. HL60 cells, Proc. Natl. Acad. Sci. USA 98 (2001) 3826–3831.
Cell 31 (2008) 153–165. [47] M. Hou, X. Wang, N. Popov, A. Zhang, X. Zhao, R. Zhou,
[28] Y. Zhao, A.J. Sfeir, Y. Zou, C.M. Buseman, T.T. Chow, J.W. Shay, A. Zetterberg, M. Bjorkholm, M. Henriksson, A. Gruber, D. Xu,
W.E. Wright, Telomere extension occurs at most chromosome The histone deacetylase inhibitor trichostatin A derepresses the
ends and is uncoupled from fill-in in human cancer cells, Cell telomerase reverse transcriptase (hTERT) gene in human cells,
138 (2009) 463–475. Exp. Cell Res. 274 (2002) 25–34.
[29] T.M. Nakamura, G.B. Morin, K.B. Chapman, S.L. Weinrich, [48] F. Lou, X. Chen, M. Jalink, Q. Zhu, N. Ge, S. Zhao, X. Fang, Y. Fan,
W.H. Andrews, J. Lingner, C.B. Harley, T.R. Cech, Telomerase M. Bjorkholm, Z. Liu, D. Xu, The opposing effect of hypoxia-
catalytic subunit homologs from fission yeast and human, inducible factor-2alpha on expression of telomerase reverse
Science 277 (1997) 955–959. transcriptase, Mol. Cancer Res. 5 (2007) 793–800.
[30] M.A. Blasco, Telomeres and human disease: ageing, cancer and [49] C. Liu, X. Fang, Z. Ge, M. Jalink, S. Kyo, M. Bjorkholm, A. Gruber,
beyond, Nat. Rev. Genet. 6 (2005) 611–622. J. Sjoberg, D. Xu, The telomerase reverse transcriptase (hTERT)
[31] J.W. Shay, S. Bacchetti, A survey of telomerase activity in human gene is a direct target of the histone methyltransferase SMYD3,
cancer, Eur. J. Cancer 33 (1997) 787–791. Cancer Res. 67 (2007) 2626–2631.
[32] J.W. Shay, W.E. Wright, Telomerase activity in human cancer, [50] Q. Zhu, C. Liu, Z. Ge, X. Fang, X. Zhang, K. Straat, M. Bjorkholm,
Curr. Opin. Oncol. 8 (1996) 66–71. D. Xu, Lysine-specific demethylase 1 (LSD1) is required for the
[33] A.J. Cesare, R.R. Reddel, Alternative lengthening of telomeres: transcriptional repression of the telomerase reverse transcrip-
models, mechanisms and implications, Nat. Rev. Genet. 11 tase (hTERT) gene, PLOS ONE 3 (2008) e1446.
(2010) 319–330. [51] Z. Ge, W. Li, N. Wang, C. Liu, Q. Zhu, M. Bjorkholm, A. Gruber, D.
[34] M.A. Blasco, M. Rizen, C.W. Greider, D. Hanahan, Differential Xu, Chromatin remodeling: recruitment of histone demethylase
regulation of telomerase activity and telomerase RNA during RBP2 by Mad1 for transcriptional repression of a Myc target gene,
multi-stage tumorigenesis, Nat. Genet. 12 (1996) 200–204. telomerase reverse transcriptase, FASEB J. 24 (2010) 579–586.
[35] Y. Liu, B.E. Snow, M.P. Hande, D. Yeung, N.J. Erdmann, [52] Z. Ge, C. Liu, M. Bjorkholm, A. Gruber, D. Xu, Mitogen-activated
A. Wakeham, A. Itie, D.P. Siderovski, P.M. Lansdorp, M.O. protein kinase cascade-mediated histone H3 phosphorylation is
Robinson, L. Harrington, The telomerase reverse transcriptase is critical for telomerase reverse transcriptase expression/telo-
limiting and necessary for telomerase function in vivo, Curr. merase activation induced by proliferation, Mol. Cell Biol. 26
Biol. 10 (2000) 1459–1462. (2006) 230–237.
[36] A. Tomas-Loba, I. Flores, P.J. Fernandez-Marcos, M.L. Cayuela, [53] Z. Song, J. Wang, L.M. Guachalla, G. Terszowski, H.R. Rodewald,
A. Maraver, A. Tejera, C. Borras, A. Matheu, P. Klatt, J.M. Flores, Z. Ju, K.L. Rudolph, Alterations of the systemic environment are
J. Vina, M. Serrano, M.A. Blasco, Telomerase reverse transcriptase the primary cause of impaired B and T lymphopoiesis in
delays aging in cancer-resistant mice, Cell 135 (2008) 609–622. telomere-dysfunctional mice, Blood 115 (2010) 1481–1489.
140 E XP E RI ME N T AL C E L L RE S E AR CH 319 (2013) 133–141

[54] F. d’Adda di Fagagna, P.M. Reaper, L. Clay-Farrace, H. Fiegler, [72] T. Bilaud, C. Brun, K. Ancelin, C.E. Koering, T. Laroche, E. Gilson,
P. Carr, T. Von Zglinicki, G. Saretzki, N.P. Carter, S.P. Jackson, Telomeric localization of TRF2, a novel human telobox protein,
A DNA damage checkpoint response in telomere-initiated Nat. Genet. 17 (1997) 236–239.
senescence, Nature 426 (2003) 194–198. [73] A. Smogorzewska, B. van Steensel, A. Bianchi, S. Oelmann,
[55] X. Zhang, Z. Chen, Y. Chen, T. Tong, Delivering antisense M.R. Schaefer, G. Schnapp, T. de Lange, Control of human telomere
telomerase RNA by a hybrid adenovirus/adeno-associated virus length by TRF1 and TRF2, Mol. Cell Biol. 20 (2000) 1659–1668.
significantly suppresses the malignant phenotype and enhances [74] R. Diotti, D. Loayza, Shelterin complex and associated factors at
cell apoptosis of human breast cancer cells, Oncogene 22 (2003) human telomeres, Nucleus 2 (2011) 119–135.
2405–2416. [75] H. Kim, O.H. Lee, H. Xin, L.Y. Chen, J. Qin, H.K. Chae, S.Y. Lin,
[56] J. Duan, Z. Chen, P. Liu, Z. Zhang, T. Tong, Wild-type p16INK4a A. Safari, D. Liu, Z. Songyang, TRF2 functions as a protein hub
suppresses cell growth, telomerase activity and DNA repair in and regulates telomere maintenance by recognizing specific
peptide motifs, Nat. Struct. Mol. Biol. 16 (2009) 372–379.
human breast cancer MCF-7 cells, Int. J. Oncol. 24 (2004)
[76] H.H. Yoo, I.K. Chung, Requirement of DDX39 DEAD box RNA
1597–1605.
helicase for genome integrity and telomere protection, Aging
[57] J. Duan, Z. Zhang, T. Tong, Irreversible cellular senescence
Cell 10 (2011) 557–571.
induced by prolonged exposure to H2O2 involves DNA-damage-
[77] G.B. Celli, T. de Lange, DNA processing is not required for ATM-
and-repair genes and telomere shortening, Int. J. Biochem. Cell
mediated telomere damage response after TRF2 deletion, Nat.
Biol. 37 (2005) 1407–1420.
Cell Biol. 7 (2005) 712–718.
[58] N. Li, Q. Li, X. Cao, G. Zhao, L. Xue, T. Tong, The tumor suppressor
[78] B. Li, S. Oestreich, T. de Lange, Identification of human Rap1:
p33ING1b upregulates p16INK4a expression and induces cel-
implications for telomere evolution, Cell 101 (2000) 471–483.
lular senescence, FEBS Lett. 585 (2011) 3106–3112. [79] M.S. O’Connor, A. Safari, D. Liu, J. Qin, Z. Songyang, The human
[59] P. Wang, Z. Zhang, Y. Sun, X. Liu, T. Tong, The two isomers of Rap1 protein complex and modulation of telomere length,
HDTIC compounds from Astragali Radix slow down telomere J. Biol. Chem. 279 (2004) 28585–28591.
shortening rate via attenuating oxidative stress and increasing [80] J.M. Svendsen, A. Smogorzewska, M.E. Sowa, B.C. O’Connell,
DNA repair ability in human fetal lung diploid fibroblast cells, S.P. Gygi, S.J. Elledge, J.W. Harper, Mammalian BTBD12/SLX4
DNA Cell Biol. 29 (2010) 33–39. assembles a Holliday junction resolvase and is required for DNA
[60] B.B. de Jesus, K. Schneeberger, E. Vera, A. Tejera, C.B. Harley, repair, Cell 138 (2009) 63–77.
M.A. Blasco, The telomerase activator TA-65 elongates short [81] P. Martinez, M. Thanasoula, A.R. Carlos, G. Gomez-Lopez,
telomeres and increases health span of adult/old mice without A.M. Tejera, S. Schoeftner, O. Dominguez, D.G. Pisano,
increasing cancer incidence, Aging Cell 10 (2011) 604–621. M. Tarsounas, M.A. Blasco, Mammalian Rap1 controls telomere
[61] J. Huang, F. Wang, M. Okuka, N. Liu, G. Ji, X. Ye, B. Zuo, M. Li, function and gene expression through binding to telomeric and
P. Liang, W.W. Ge, J.C. Tsibris, D.L. Keefe, L. Liu, Association of extratelomeric sites, Nat. Cell Biol. 12 (2010) 768–780.
telomere length with authentic pluripotency of ES/iPS cells, Cell [82] P. Baumann, T.R. Cech, Pot1, the putative telomere end-binding
Res. 21 (2011) 779–792. protein in fission yeast and humans, Science 292 (2001)
[62] F. Wang, Y. Yin, X. Ye, K. Liu, H. Zhu, L. Wang, M. Chiourea, 1171–1175.
M. Okuka, G. Ji, J. Dan, B. Zuo, M. Li, Q. Zhang, N. Liu, L. Chen, X. [83] M. Lei, E.R. Podell, P. Baumann, T.R. Cech, DNA self-recognition
Pan, S. Gagos, D.L. Keefe, L. Liu, Molecular insights into the in the structure of Pot1 bound to telomeric single-stranded
heterogeneity of telomere reprogramming in induced pluripo- DNA, Nature 426 (2003) 198–203.
tent stem cells, Cell Res. 22 (2012) 757–768. [84] H. He, A.S. Multani, W. Cosme-Blanco, H. Tahara, J. Ma, S. Pathak,
[63] D. Liu, M.S. O’Connor, J. Qin, Z. Songyang, Telosome, a mam- Y. Deng, S. Chang, POT1b protects telomeres from end-to-end
malian telomere-associated complex formed by multiple telo- chromosomal fusions and aberrant homologous recombination,
meric proteins, J. Biol. Chem. 279 (2004) 51338–51342. EMBO J. 25 (2006) 5180–5190.
[64] T. de Lange, Shelterin: the protein complex that shapes and [85] D. Hockemeyer, J.P. Daniels, H. Takai, T. de Lange, Recent
safeguards human telomeres, Genes Dev. 19 (2005) 2100–2110. expansion of the telomeric complex in rodents: two distinct
[65] M. Lei, E.R. Podell, T.R. Cech, Structure of human POT1 bound to POT1 proteins protect mouse telomeres, Cell 126 (2006) 63–77.
[86] L. Wu, A.S. Multani, H. He, W. Cosme-Blanco, Y. Deng, J.M. Deng,
telomeric single-stranded DNA provides a model for chromo-
O. Bachilo, S. Pathak, H. Tahara, S.M. Bailey, R.R. Behringer,
some end-protection, Nat. Struct. Mol. Biol. 11 (2004)
S. Chang, Pot1 deficiency initiates DNA damage checkpoint
1223–1229.
activation and aberrant homologous recombination at telo-
[66] F. Wang, E.R. Podell, A.J. Zaug, Y. Yang, P. Baciu, T.R. Cech, M. Lei,
meres, Cell 126 (2006) 49–62.
The POT1–TPP1 telomere complex is a telomerase processivity
[87] W. Palm, D. Hockemeyer, T. Kibe, T. de Lange, Functional
factor, Nature 445 (2007) 506–510.
dissection of human and mouse POT1 proteins, Mol. Cell Biol. 29
[67] Z. Zhong, L. Shiue, S. Kaplan, T. de Lange, A mammalian factor
(2009) 471–482.
that binds telomeric TTAGGG repeats in vitro, Mol. Cell Biol. 12
[88] P. Wu, H. Takai, T. de Lange, Telomeric 30 overhangs derive from
(1992) 4834–4843. resection by Exo1 and Apollo and fill-in by POT1b-associated
[68] B. van Steensel, T. de Lange, Control of telomere length by the
CST, Cell 150 (2012) 39–52.
human telomeric protein TRF1, Nature 385 (1997) 740–743. [89] R.L. Flynn, R.C. Centore, R.J. O’Sullivan, R. Rai, A. Tse,
[69] J. Karlseder, L. Kachatrian, H. Takai, K. Mercer, S. Hingorani, Z. Songyang, S. Chang, J. Karlseder, L. Zou, TERRA and hnRNPA1
T. Jacks, T. de Lange, Targeted deletion reveals an essential orchestrate an RPA-to-POT1 switch on telomeric single-
function for the telomere length regulator Trf1, Mol. Cell Biol. 23 stranded DNA, Nature 471 (2011) 532–536.
(2003) 6533–6541. [90] D. Liu, A. Safari, M.S. O’Connor, D.W. Chan, A. Laegeler, J. Qin,
[70] Y. Chen, Y. Yang, M. van Overbeek, J.R. Donigian, P. Baciu, Z. Songyang, PTOP interacts with POT1 and regulates its
T. de Lange, M. Lei, A shared docking motif in TRF1 and TRF2 localization to telomeres, Nat. Cell Biol. 6 (2004) 673–680.
used for differential recruitment of telomeric proteins, Science [91] A.M. Tejera, M. Stagno d’Alcontres, M. Thanasoula, R.M. Marion,
319 (2008) 1092–1096. P. Martinez, C. Liao, J.M. Flores, M. Tarsounas, M.A. Blasco, TPP1
[71] Z. Zeng, W. Wang, Y. Yang, Y. Chen, X. Yang, J.A. Diehl, X. Liu, is required for TERT recruitment, telomere elongation during
M. Lei, Structural basis of selective ubiquitination of TRF1 by nuclear reprogramming, and normal skin development in mice,
SCFFbx4, Dev. Cell 18 (2010) 214–225. Dev. Cell 18 (2010) 775–789.
EX P ER IM E NTA L CE LL R ES E A RC H 319 (2013) 133–141 141

[92] T. Kibe, G.A. Osawa, C.E. Keegan, T. de Lange, Telomere protec- associated with senescence in proliferating neural cells and
tion by TPP1 is mediated by POT1a and POT1b, Mol. Cell Biol. 30 differentiation of neurons, J. Neurochem. 97 (2006) 567–581.
(2010) 1059–1066. [104] Z. Mao, A. Seluanov, Y. Jiang, V. Gorbunova, TRF2 is required for
[93] H. Xin, D. Liu, M. Wan, A. Safari, H. Kim, W. Sun, M.S. O’Connor, repair of nontelomeric DNA double-strand breaks by homo-
Z. Songyang, TPP1 is a homologue of ciliate TEBP-beta and logous recombination, Proc. Natl. Acad. Sci. USA 104 (2007)
interacts with POT1 to recruit telomerase, Nature 445 (2007) 13068–13073.
559–562. [105] P.S. Bradshaw, D.J. Stavropoulos, M.S. Meyn, Human telomeric
[94] C.E. Keegan, J.E. Hutz, T. Else, M. Adamska, S.P. Shah, A.E. Kent, protein TRF2 associates with genomic double-strand breaks as
J.M. Howes, W.G. Beamer, G.D. Hammer, Urogenital and caudal an early response to DNA damage, Nat. Genet. 37 (2005)
dysgenesis in adrenocortical dysplasia (acd) mice is caused by a 193–197 (Epub 2005 Jan 2023).
splicing mutation in a novel telomeric regulator, Hum. Mol. [106] D. Yang, Y. Xiong, H. Kim, Q. He, Y. Li, R. Chen, Z. Songyang,
Genet. 14 (2005) 113–123. Human telomeric proteins occupy selective interstitial sites,
[95] Y. Xie, D. Yang, Q. He, Z. Songyang, Zebrafish as a model system Cell Res. (2011).
to study the physiological function of telomeric protein TPP1, [107] H. Teo, S. Ghosh, H. Luesch, A. Ghosh, E.T. Wong, N. Malik,
A. Orth, P. de Jesus, A.S. Perry, J.D. Oliver, N.L. Tran, L.J. Speiser,
PLOS ONE 6 (2011) e16440.
M. Wong, E. Saez, P. Schultz, S.K. Chanda, I.M. Verma,
[96] M. Wan, J. Qin, Z. Songyang, D. Liu, OB fold-containing protein 1
V. Tergaonkar, Telomere-independent Rap1 is an IKK adaptor
(OBFC1), a human homolog of yeast Stn1, associates with TPP1
and regulates NF-kappaB-dependent gene expression, Nat. Cell
and is implicated in telomere length regulation, J. Biol. Chem.
Biol. 12 (2010) 758–767.
284 (2009) 26725–26731.
[108] T. Simonet, L.E. Zaragosi, C. Philippe, K. Lebrigand, C. Schouteden,
[97] L.Y. Chen, S. Redon, J. Lingner, The human CST complex is a
A. Augereau, S. Bauwens, J. Ye, M. Santagostino, E. Giulotto,
terminator of telomerase activity, Nature (2012).
F. Magdinier, B. Horard, P. Barbry, R. Waldmann, E. Gilson, The
[98] M.S. O’Connor, A. Safari, H. Xin, D. Liu, Z. Songyang, A critical
human TTAGGG repeat factors 1 and 2 bind to a subset of
role for TPP1 and TIN2 interaction in high-order telomeric
interstitial telomeric sequences and satellite repeats, Cell Res.
complex assembly, Proc. Natl. Acad. Sci. USA 103 (2006)
(2011).
11874–11879.
[109] O.A. Kovalenko, M.J. Caron, P. Ulema, C. Medrano, A.P. Thomas,
[99] K.K. Takai, T. Kibe, J.R. Donigian, D. Frescas, T. de Lange,
M. Kimura, M.G. Bonini, U. Herbig, J.H. Santos, A mutant
Telomere protection by TPP1/POT1 requires tethering to TIN2, telomerase defective in nuclear-cytoplasmic shuttling fails to
Mol. Cell 44 (2011) 647–659. immortalize cells and is associated with mitochondrial
[100] D. Yang, Q. He, H. Kim, W. Ma, Z. Songyang, TIN2 protein dysfunction, Aging Cell 9 (2010) 203–219.
dyskeratosis congenita missense mutants are defective in [110] J.H. Santos, J.N. Meyer, B. Van Houten, Mitochondrial localiza-
association with telomerase, J. Biol. Chem. 286 (2011) tion of telomerase as a determinant for hydrogen peroxide-
23022–23030. induced mitochondrial DNA damage and apoptosis, Hum. Mol.
[101] L.Y. Chen, D. Liu, Z. Songyang, Telomere maintenance through Genet. 15 (2006) 1757–1768.
spatial control of telomeric proteins, Mol. Cell Biol. 27 (2007) [111] J.H. Santos, J.N. Meyer, M. Skorvaga, L.A. Annab, B. Van Houten,
5898–5909. Mitochondrial hTERT exacerbates free-radical-mediated
[102] P. Zhang, M.J. Pazin, C.M. Schwartz, K.G. Becker, R.P. Wersto, mtDNA damage, Aging Cell 3 (2004) 399–411.
C.M. Dilley, M.P. Mattson, Nontelomeric TRF2–REST interaction [112] L.Y. Chen, Y. Zhang, Q. Zhang, H. Li, Z. Luo, H. Fang, S.H. Kim,
modulates neuronal gene silencing and fate of tumor and stem L. Qin, P. Yotnda, J. Xu, B.P. Tu, Y. Bai, Z. Songyang, Mitochon-
cells, Curr. Biol. 18 (2008) 1489–1494. drial localization of telomeric protein TIN2 links telomere
[103] P. Zhang, K. Furukawa, P.L. Opresko, X. Xu, V.A. Bohr, regulation to metabolic control, Mol. Cell (2012) (Epub ahead
M.P. Mattson, TRF2 dysfunction elicits DNA damage responses of print).

Potrebbero piacerti anche