telomere biology in aging and cancer: early history and

12
Genes Genet. Syst. (2017) 92, p. 107–118 Edited by Kenji Ichiyanagi * Corresponding author. E-mail: [email protected] DOI: http://doi.org/10.1266/ggs.17-00010 Telomere biology in aging and cancer: early history and perspectives Makoto T Hayashi * Department of Gene Mechanisms, Graduate School of Biostudies/The Hakubi Center for Advanced Research, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan (Received 16 March 2017, accepted 4 July 2017; J-STAGE Advance published date: 6 October 2017) The ends of eukaryotic linear chromosomes are protected from undesired enzy- matic activities by a nucleoprotein complex called the telomere. Expanding evidence indicates that telomeres have central functions in human aging and tumorigenesis. While it is undoubtedly important to follow current advances in telomere biology, it is also fruitful to be well informed in seminal historical studies for a comprehensive understanding of telomere biology, and for the anticipation of future directions. With this in mind, I here summarize the early history of telomere biology and current advances in the field, mostly focusing on mammalian studies relevant to aging and cancer. Key words: Telomere, Aging, Cancer, History INTRODUCTION All eukaryotes possess linear chromosomes, each of which must have two intrinsic ends. A natural end of a chromosome is structurally indistinguishable from an internal broken end, which causes immediate activation of the DNA damage checkpoint pathway. Once this path- way is activated, the cell cycle is halted until the dam- age is repaired mainly through the non-homologous end joining (NHEJ) or homologous recombination (HR) path- way. If the damage is excessive and cannot be repaired within a certain time range, the cell chooses to commit suicide (e.g., by apoptosis). Therefore, natural ends must be protected from activation of these pathways, and such a protective function is performed by a special nucleopro- tein complex called the telomere. The telomere is com- posed of repetitive DNA, which has a 3’ protruding end called the 3’-overhang, and proteins that specifically rec- ognize and assemble on the repetitive double- and single- stranded sequence. In mammalian systems, the protein complex is called shelterin, loss of which renders chromo- some ends vulnerable to enzymatic activities, resulting in activation of the DNA damage checkpoint pathway. Loss of telomeric repeats also leads to failure of shelterin bind- ing and end protection. Because DNA polymerase synthesizes DNA strictly in the 5’ to 3’ orientation, the end of the lagging strand cannot be fully replicated in each S phase of the cell cycle. On the other hand, the end of the leading strand is processed to regenerate the 3’-overhang following rep- lication. Thus, both the leading and lagging ends of replicated telomeres gradually shorten during each cell cycle unless they are replenished by a special reverse transcriptase called telomerase. Telomerase is highly expressed in unicellular eukaryotes as well as in the reproductive cells in multicellular organisms, whereas its activity is hardly detectable in normal somatic cells, which show a gradual shortening of telomeres in vitro and in vivo. Therefore, telomeres in the human soma eventually reach a critical length after a finite number of cell divisions and activate the DNA damage checkpoint, which causes irreversible cell cycle arrest (i.e., replicative senescence) and underlies cellular aging. In the absence of a functional checkpoint, cells keep dividing with ongo- ing telomere shortening, entering the next stage called crisis. In the crisis stage, chromosomes become unstable because of frequent end-to-end fusions, which are asso- ciated with massive cell death through mitotic catastro- phe. Eventually, rare survivors that acquire the telomere maintenance pathway may emerge, resulting in tumor generation. It is also well documented that mutations in a variety of genes involved in telomere maintenance cause diseases related to human aging. Thus, telomere biology is of great importance to our understanding of human aging and tumorigenesis. The recent technological expansion of molecular biol- ogy has greatly advanced the field, yet all of the current advances are based on seminal accomplishments by pio- neers in the field. In this review, I will summarize the early history (until the first decade of this century) of telomere biology with a focus on the mammalian system to see milestone discoveries in a new light; some of these

Upload: others

Post on 31-Dec-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Telomere biology in aging and cancer: early history and

107History of telomere biologyGenes Genet. Syst. (2017) 92, p. 107–118

Edited by Kenji Ichiyanagi* Corresponding author. E-mail: [email protected]: http://doi.org/10.1266/ggs.17-00010

Telomere biology in aging and cancer: early history and perspectives

Makoto T Hayashi*

Department of Gene Mechanisms, Graduate School of Biostudies/The Hakubi Center for Advanced Research, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan

(Received 16 March 2017, accepted 4 July 2017; J-STAGE Advance published date: 6 October 2017)

The ends of eukaryotic linear chromosomes are protected from undesired enzy-matic activities by a nucleoprotein complex called the telomere. Expanding evidence indicates that telomeres have central functions in human aging and tumorigenesis. While it is undoubtedly important to follow current advances in telomere biology, it is also fruitful to be well informed in seminal historical studies for a comprehensive understanding of telomere biology, and for the anticipation of future directions. With this in mind, I here summarize the early history of telomere biology and current advances in the field, mostly focusing on mammalian studies relevant to aging and cancer.

Key words: Telomere, Aging, Cancer, History

INTRODUCTION

All eukaryotes possess linear chromosomes, each of which must have two intrinsic ends. A natural end of a chromosome is structurally indistinguishable from an internal broken end, which causes immediate activation of the DNA damage checkpoint pathway. Once this path-way is activated, the cell cycle is halted until the dam-age is repaired mainly through the non-homologous end joining (NHEJ) or homologous recombination (HR) path-way. If the damage is excessive and cannot be repaired within a certain time range, the cell chooses to commit suicide (e.g., by apoptosis). Therefore, natural ends must be protected from activation of these pathways, and such a protective function is performed by a special nucleopro-tein complex called the telomere. The telomere is com-posed of repetitive DNA, which has a 3’ protruding end called the 3’-overhang, and proteins that specifically rec-ognize and assemble on the repetitive double- and single-stranded sequence. In mammalian systems, the protein complex is called shelterin, loss of which renders chromo-some ends vulnerable to enzymatic activities, resulting in activation of the DNA damage checkpoint pathway. Loss of telomeric repeats also leads to failure of shelterin bind-ing and end protection.

Because DNA polymerase synthesizes DNA strictly in the 5’ to 3’ orientation, the end of the lagging strand cannot be fully replicated in each S phase of the cell cycle. On the other hand, the end of the leading strand

is processed to regenerate the 3’-overhang following rep-lication. Thus, both the leading and lagging ends of replicated telomeres gradually shorten during each cell cycle unless they are replenished by a special reverse transcriptase called telomerase. Telomerase is highly expressed in unicellular eukaryotes as well as in the reproductive cells in multicellular organisms, whereas its activity is hardly detectable in normal somatic cells, which show a gradual shortening of telomeres in vitro and in vivo. Therefore, telomeres in the human soma eventually reach a critical length after a finite number of cell divisions and activate the DNA damage checkpoint, which causes irreversible cell cycle arrest (i.e., replicative senescence) and underlies cellular aging. In the absence of a functional checkpoint, cells keep dividing with ongo-ing telomere shortening, entering the next stage called crisis. In the crisis stage, chromosomes become unstable because of frequent end-to-end fusions, which are asso-ciated with massive cell death through mitotic catastro-phe. Eventually, rare survivors that acquire the telomere maintenance pathway may emerge, resulting in tumor generation. It is also well documented that mutations in a variety of genes involved in telomere maintenance cause diseases related to human aging. Thus, telomere biology is of great importance to our understanding of human aging and tumorigenesis.

The recent technological expansion of molecular biol-ogy has greatly advanced the field, yet all of the current advances are based on seminal accomplishments by pio-neers in the field. In this review, I will summarize the early history (until the first decade of this century) of telomere biology with a focus on the mammalian system to see milestone discoveries in a new light; some of these

Page 2: Telomere biology in aging and cancer: early history and

108 M. T HAYASHI

are visualized chronologically in Figs. 1 and 2. Besides the early history, recent advances in the field and per-spectives will also be briefly discussed.

A BRIEF HISTORY OF TELOMERE BIOLOGY

Beginning of the end story Everything has a begin-ning. The whole field of telomere biology was started by two Nobel Prize winners, Hermann Muller and Barbara McClintock, more than seven decades ago. Muller had studied chromosome instability, such as translocation, induced by ionizing radiation in Drosophila, which led him to find that natural chromosome ends never fuse to broken chromosome ends while broken ends often do so (Muller, 1938). He suggested the idea that natural chro-mosome ends are somehow protected against the repair process, which connects two broken ends, and named such a protective structure the “telomere”. A similar phenomenon was independently found by McClintock through her cytological studies using irradiated maize chromosomes (McClintock, 1931). She proposed the breakage-fusion-bridge (BFB) cycle hypothesis to explain chromosome instability: broken ends of two sister chro-matids fuse together, which generates a DNA bridge dur-ing anaphase in mitosis, eventually leading to breakage of the bridge. Such broken ends will fuse again in the next cell cycle, starting a multiple fusion and breakage cycle (McClintock, 1941). She could probably have seen how relevant the BFB cycle hypothesis is in cancer biol-ogy with her own eyes in her later years.

Their seminal studies were not revisited until the 1970s, when James Watson and Alexey Olovnikov proposed the “end replication problem” (Olovnikov, 1971, 1973; Watson, 1972). They independently pointed out that the unidi-rectionality of DNA polymerase should leave a gap at the very end of a chromosome replicated by lagging strand synthesis. This means that telomeres should get shorter and shorter upon their replication. Such telomere short-ening was proposed as the molecular basis of replicative senescence (or the Hayflick limit), an irreversible arrest of cellular growth following serial passage of cultured human cells, which was first observed by Leonard Hayflick (Hayflick and Moorhead, 1961).

Confirming this proposal at the molecular level, how-ever, had to await the next big bang in telomere biol-ogy, which was accomplished by the 2009 Nobel Prize winners, Jack Szostak, Elizabeth Blackburn and Carol Greider. Thanks to the development of Sanger sequenc-ing technology (Sanger and Coulson, 1975), Blackburn could successfully read telomeric sequences of the cili-ated protozoan Tetrahymena thermophila (Blackburn and Gall, 1978). This finding led Szostak to devise an artifi-cial minichromosome, capped with Tetrahymena telomeric sequences at its ends, which was successfully maintained in yeast (Szostak and Blackburn, 1982); this was the first

molecular evidence that a telomeric DNA sequence pos-sesses a protective function. The end replication problem and the structural and dynamic properties of telomeres implied the presence of enzymatic activity to counteract telomere shortening. Such an enzyme, a terminal trans-ferase that utilizes RNA as a template, was indeed identi-fied in Tetrahymena by Greider and Blackburn and named telomerase (Greider and Blackburn, 1985, 1987). Szostak then developed a system to identify mutants of yeast that are defective for telomere maintenance, and found that null mutants of a gene named EST1 (ever shorter telo-meres) show gradual telomere shortening and a senes-cence phenotype (Lundblad and Szostak, 1989). This series of elegant studies pioneered a new era of telomere biology (Blackburn et al., 2006) (Fig. 1).

Telomeric DNA meets protein After the telomeric DNA sequence of Tetrahymena had been published, sev-eral studies inferred the existence of telomere-binding proteins other than histones (Blackburn and Chiou, 1981; Gottschling and Cech, 1984), and such proteins were soon identified in another ciliated protozoan, Oxytricha nova (Gottschling and Zakian, 1986). Although the func-tion of the telomeric proteins was unclear at that time, it was proposed that “they act as stability elements or chromosome caps, thereby preventing fusion of telomeres” (Gottschling and Zakian, 1986). The first evidence that a telomeric protein protects chromosome ends came from two independent studies using budding yeast, in which David Shore’s and Virginia Zakian’s groups analyzed the function of the yeast telomere-binding protein Rap1 (Conrad et al., 1990; Lustig et al., 1990).

By this time, the concept of “checkpoints” that safe-guard the order of cell cycle events from intrinsic and extrinsic insults had been proposed by Leland Hartwell and Ted Weinert (Hartwell and Weinert, 1989). Among several cell cycle checkpoints, the DNA damage check-point is highly relevant to telomere end protection, since chromosome ends resemble DNA ends caused by DNA double-stranded breaks (DSBs), which cause a delay in DNA synthesis and were later linked to an increase of the “genome guardian”, p53 (Kastan et al., 1991). In the same year, Calvin Harley wrote that “when a certain length is reached on one or more telomeres in a divid-ing cell, a “checkpoint”, analogous to those regulating cell cycle events […] is signalled which evokes the Hayflick Limit: cells stop dividing” (Harley, 1991). Therefore, it was very tempting to speculate that telomeric proteins possess a protective function to prevent checkpoint acti-vation at the chromosome ends (de Lange, 1995).

Determination of the human telomeric DNA sequence (TTAGGG)n (Moyzis et al., 1988), which is conserved among vertebrate species (Meyne et al., 1989), allowed Titia de Lange and coworkers to perform electrophoretic mobility shift assays to identify a telomere-binding activ-

Page 3: Telomere biology in aging and cancer: early history and

109History of telomere biology

Telomeric proteins and structure

Telomere biology in aging and cancer

hTRF1 identified

Zhong et al. 1992

Checkpoint proposed

Hartwell & Weinert 1989

Progressive telomere shortening in vitro and in vivo

Harley et al. 1990Hastie et al. 1990

hTERT identified

Nakamura et al. 1997 Meyerson et al. 1997

Human telomericrepeat sequenced

Moyzis et al. 1988

hPOT1 identified

Baumann & Cech 2001

Shelterin/Telosomedefined

Liu et al. 2004 de Lange 2005

Function of hTRF1/hTRF2 revealed

van Steensel & de Lange 1997van Steensel et al. 1998

Telomerase activity associated with tumor cells

Kim et al. 1994

Normal human cells immortalized by telomerase

Nakayama et al. 1998Bodnar et al. 1998

Telomerase knockout mice

Blasco et al. 1997Lee et al. 1998

DKC1 associated with telomerase

Mitchell et al. 1999ALT defined

Bryan & Reddel 1997

t-loop identified

Griffith et al. 1999

3’-overhang identified

Makarov et al. 1997

hTRF2 identified

Bilaud et al. 1996, 1997Broccoli et al. 1997

Tumor growth suppressed by DN-hTERT

Hahn et al. 1999

200019901985 1995 2005

Fig. 2. The history of telomere biology in the aging and cancer fields from the late 1980s to the early 2000s. Milestone studies related to telomeric DNA and proteins (top) and telomere biology in aging and cancer (bottom) are aligned in chronological order.

Telomeric DNA and proteins

Aging and cancer

Absence of fusion between natural end and broken end

McClintock 1931

Telomere defined

Muller 1938

BFB cycle hypothesis proposed

McClintock 1941

Replicative senescence (Hayflick limit)

Hayflick & Moorhead 1961

Tetrahymena telomerase identified

Greider & Blackburn 1985

Telomere sequence of Tetrahymena

Blackburn & Gall 1978

Linear plasmid with Tetrahymenatelomeres maintained in yeast

Szostak & Blackburn 1982

End replication problem proposed

Olovnikov 1971, 1973 Watson 1972

Crisis defined

Jensen et al. 1964

RB1 gene identified

Friend et al. 1986

p53 protein identified

Deleo et al. 1979Linzer & Levine 1979

Lane & Crawford 1979Cultured human and hamster cells transformed by SV40

Koprowski et al. 1962Shein & Enders 1962

Rabson & Kirschstein 1962

1930 19801970196019501940

Fig. 1. The history of the telomere biology, aging, and cancer fields from the early 1930s to the late 1980s. Milestone studies related to telomeric DNA and proteins (top) and aging and cancer (bottom) are aligned in chronological order.

Page 4: Telomere biology in aging and cancer: early history and

110 M. T HAYASHI

ity (Zhong et al., 1992) and then clone the gene encoding a telomeric repeat binding factor, which was named TRF (for TTAGGG repeat binding factor; later renamed TRF1), from a HeLa cell extract (Chong et al., 1995). The pro-tein was shown to be specifically localized to telomeric DNA in vivo, and overexpression of full-length and trun-cated dominant-negative TRF1 resulted in shortening and elongation of telomeric DNA, respectively, providing the first evidence that a mammalian telomeric protein is also involved in telomere homeostasis (van Steensel and de Lange, 1997). In the meantime, Eric Gilson’s group raised antibodies against the DNA-binding motif of a yeast telomere-binding protein, Tbf1, to search for similar pro-teins in human cell extracts, and identified two telomere-binding proteins (Bilaud et al., 1996), one of which turned out to be TRF1 while the other was named TRF2 (Bilaud et al., 1997). TRF2 was independently identified on the basis of its sequence similarity to TRF1 by de Lange’s group at the same time (Broccoli et al., 1997), and soon shown to be central in telomere protection: overexpres-sion of truncated dominant-negative TRF2 caused chro-mosome end-to-end fusion in cultured human cells (van Steensel et al., 1998), and checkpoint kinase ATM (ataxia telangiectasia mutated)-dependent p53 activation, result-ing in apoptosis or senescence in a cell type-dependent manner (Karlseder et al., 1999). These seminal studies paved the way to elucidate the molecular function of telo-mere-binding proteins in a variety of species. The next component of the mammalian telomeric protein complex was identified in 1999 by a yeast two-hybrid screen and named a TRF1-interacting nuclear protein, TIN2 (Kim et al., 1999). Searches for TRF2-interacting proteins by a yeast two-hybrid screen and nanoelectrospray tandem mass spectrometry independently identified Rap1, the mammalian ortholog of yeast Rap1, in the following year (Li et al., 2000; Zhu et al., 2000).

Although TRF1 and TRF2 were identified as double-stranded DNA-binding factors, the 3’ end of mammalian telomeres had been shown to possess a single-stranded TTAGGG 3’-overhang (or G-overhang) (Makarov et al., 1997), which had originally been found in hypotricha cili-ates (Klobutcher et al., 1981). The telomeric protein in ciliates had been identified in 1986, as described above, which prompted biochemists to search for a similar sin-gle-stranded telomeric DNA-binding protein in higher eukaryotes. However, identification of such a protein had to await the availability of database mining. In 2001, Peter Baumann and Thomas Cech successfully identified a fission yeast single-stranded telomere-binding protein, Pot1, which has limited similarity to a portion of the telomere protein TEBP from Oxytricha and other ciliates (Baumann and Cech, 2001). A BLAST search with fission yeast Pot1 identified human POT1, which specifically binds to the single-stranded TTAGGG repeat (Baumann and Cech, 2001).

Identification of several telomeric proteins further accelerated the competition, resulting in reports of the same protein from three independent studies in 2004. de Lange and coworkers performed mass spectrometry of TRF2/TIN2-associated proteins and named a novel bind-ing partner POT1-interacting protein 1 (PIP1) based on its ability to interact with POT1 (Ye et al., 2004); Zhou Songyang’s group took a similar approach and identi-fied PTOP (POT1- and TIN2-organizing protein) (Liu et al., 2004b), while Susan Smith and colleagues utilized a yeast two-hybrid assay with TIN2 as a bait and reported TINT1 (TIN2 interacting protein 1) (Houghtaling et al., 2004). A single name, TPP1 (telomere protection pro-tein 1), was given later. Biochemical analysis of the six identified proteins (TRF1, TRF2, TIN2, RAP1, POT1 and TPP1) indicated that these components form a complex on telomeric chromatin, which has been named the telo-some (Liu et al., 2004a) or shelterin (de Lange, 2005), although the latter is more common in the current field.

During the period of telomere protein searches, telo-meric end structure was visualized by electron micros-copy, which demonstrated that telomeres can form a large duplex loop (called the t-loop) in vitro in a TRF2-dependent manner, and the t-loop can be isolated from cultured human cells (Griffith et al., 1999). The t-loop is currently the most widely accepted model for end pro-tection, proposing the integration of the 3’-overhang into double-stranded repeats with the aid of shelterin func-tion, especially provided by TRF2.

Telomeres are connected to aging and cancer At the same period when replicative senescence was reported in the early 1960s, another phenomenon that is highly relevant to tumorigenesis was described: transfor-mation of human cultured cells was characterized as the development of cytologically abnormal morphology follow-ing simian virus 40 (SV40) infection (Koprowski et al., 1962; Rabson and Kirschstein, 1962; Shein and Enders, 1962). Such transformed cells possess extended growth capacity compared to their untransformed counterparts, which is followed by massive degeneration of the trans-formed cultures. This is the crisis stage, and leads to almost complete loss of cellular viability; it is followed by recovery of the cultures, during which a viable minor sub-population occasionally outgrows in the culture (Girardi et al., 1965). By the end of the 1970s, p53 had been iden-tified as a protein exclusively expressed in transformed mouse cell lines (DeLeo et al., 1979) and co-immunopre-cipitated with SV40 large-T antigen (Lane and Crawford, 1979; Linzer and Levine, 1979). Ten years later, Bert Vogelstein’s and Arnold Levine’s groups had indepen-dently reported p53’s role as a tumor suppressor (Baker et al., 1989; Finlay et al., 1989). In the same period, the first tumor suppressor gene, Rb, which causes recessive mutations that predispose cells to retinoblastoma, was

Page 5: Telomere biology in aging and cancer: early history and

111History of telomere biology

identified (Friend et al., 1986), and shown to interact with the large-T antigen (DeCaprio et al., 1988). After the extensive research on replicative senescence and cri-sis, these two phenomena were linked to telomere biology as described briefly below (Fig. 2).

Soon after the identification of telomerase in Tetrahymena (Greider and Blackburn, 1985), human telomerase activity in HeLa cell crude extracts was reported (Morin, 1989). As predicted by the end rep-lication problem, telomeric DNA in human cells was shown to shorten gradually during serial passage until replicative senescence in primary culture (Harley et al., 1990), as well as in vivo aging in blood samples (Hastie et al., 1990). Such gradual telomere shortening was also reported in transformed human cells until cri-sis, after which post-recovery outgrown cell lines were shown to have gained telomerase activity (Counter et al., 1992). Telomere shortening was well explained by the finding that no normal somatic human tissues, other than ovaries and testes, are positive for telomerase activity (Kim et al., 1994). In stark contrast, more than 90% of cultured immortal cells representing a variety of human tissues and biopsies representing many tumor types were positive for telomerase activity (Kim et al., 1994). Sub-sequent studies identified telomerase activity in human stem and progenitor cells, such as hematopoietic progeni-tor cells and T lymphocytes (Chiu et al., 1996; Weng et al., 1996), albeit not enough to maintain telomere length fully in hematopoietic stem cells (Vaziri et al., 1994). Although the underlying mechanism was still unknown, these and other studies let Harley remodel Olovnikov’s “margi-notomy theory”, which predicted that critical deletion of chromosome termini eventually causes cell death (Harley, 1991; Olovnikov, 1973). The same concept was proposed by Woodring Wright and Jerry Shay as the two-stage mechanism hypothesis, in which gradual telomere short-ening somehow activates the checkpoint pathway (with p53 and Rb as key factors) to induce replicative senes-cence, while in the absence of functional p53 and Rb, cells keep dividing with progressive telomere shortening and enter the crisis stage (Wright and Shay, 1992). Recov-ery from crisis by spontaneous activation of telomerase consequently gives rise to tumorigenic cells. Therefore, telomere shortening was proposed as a molecular basis of anti-tumorigenic cell cycle arrest and massive cell death in senescence and crisis, respectively. Identification of a gene encoding the human telomerase catalytic subunit (hTERT; also known as hEST2) (Meyerson et al., 1997; Nakamura et al., 1997) enabled exogenous expression of hTERT in normal human fibroblasts and epithelial cells, which resulted in extensive elongation of telomeres and immortalization of the normal cells (Bodnar et al., 1998; Nakayama et al., 1998). Ectopic expression of hTERT in SV40 large-T- and oncogenic H-ras-transformed human fibroblasts and epithelial cells resulted in bypass of cri-

sis and tumorigenic conversion of these cells (Hahn et al., 1999a). Conversely, inhibition of telomerase activity by a dominant-negative DN-hTERT suppressed growth of human tumor-derived cell lines (Hahn et al., 1999b), demonstrating telomerase as a potential target of anti-cancer therapy. This experimental evidence established that telomere shortening underlies replicative senescence and crisis in human cultured somatic cells due to insuf-ficient telomerase expression, and that maintenance of telomere length is essential for unlimited cellular growth.

While telomerase activity was strongly associated with tumorigenesis, there was a striking exception. A number of immortalized human cell lines are capable of elongating telomeres without any detectable telomerase activity (Kim et al., 1994; Murnane et al., 1994; Bryan et al., 1995). These cells are characterized by very long and heterogeneous telomeres, maintained by a telomerase-independent mechanism referred to as ALT (alternative lengthening of telomeres) (Bryan and Reddel, 1997), which are also found in a subset of tumor-derived cell lines and tumors (Bryan et al., 1997). Roger Reddel and cowork-ers showed elegantly that exogenous DNA sequences integrated into telomeres of ALT cells are copied from the original telomere to another telomere, indicating an HR-based mechanism underlying ALT (Dunham et al., 2000). Although targeting telomerase by specific inhibi-tors had been a promising anti-cancer drug strategy, the finding of ALT raised a significant concern for therapeu-tic applications of telomerase inhibitors. Because ALT-positive tumors are capable of escaping the anti-growth effect of telomeraes inhibitors, it is of great importance to develop drugs that specifically target the ALT pathway.

The fact that patients suffering from ataxia telangiec-tasia, which is characterized by premature aging, predis-position to cancer, immunodeficiency, hypersensitivity to ionizing radiation, and neurological disorders, show an accelerated telomere shortening and increased telomere fusion phenotype suggested a causal link between telo-mere maintenance and aging-related disorders (Metcalfe et al., 1996; Xia et al., 1996). In 1999, direct evidence that causally links failure of telomere maintenance and diseases of aging humans was provided. Patients with the X-linked form of a human inherited disease, dyskera-tosis congenita, suffer from premature aging disorders caused by defects in highly regenerative tissues, attribut-able to mutations in the DKC1 gene that encodes the pro-tein dyskerin. Kathleen Collins and colleagues revealed that dyskerin, which had previously been proposed to be involved in ribosomal function, is required for telomerase RNA generation and consequently telomerase activity and telomere maintenance (Mitchell et al., 1999). This finding was the first step to explore the causal relation-ship between telomere dysfunction and human degenera-tive disorders. Subsequently, these findings and the fact that telomeres gradually shorten in human stem cells

Page 6: Telomere biology in aging and cancer: early history and

112 M. T HAYASHI

(Vaziri et al., 1994; Lansdorp, 1998), together with mouse genetics studies described below, led to the idea that degeneration of the self-renewal capacity of stem cells is partly due to a decline of telomere length or a change in telomere structure, consequently underlying individual aging (Van Zant and Liang, 2003; Bell and Van Zant, 2004; Sharpless and DePinho, 2004). Thus, by the early 2000s, diseases associated with aging and a physiological decline accompanied by aging had both been connected to telomere biology.

What mouse genetics taught us The telomere the-ory of aging and cancer was further strengthened by the development of mice lacking telomerase RNA component (mTR), which was identified in 1995 by Greider’s group (Blasco et al., 1995). Human telomerase RNA was also reported by Bryant Villeponteau and coworkers in the same year (Feng et al., 1995). In contrast to human tissues, most mouse tissues possess active telomerase (Prowse and Greider, 1995), and telomeres of laboratory-established mouse strains are kept very long (up to sev-eral hundred kb) (Kipling and Cooke, 1990). mTR–/–

mice were first reported by Greider and coworkers in 1997, which unexpectedly revealed that the first few generations of knockout animals are born successfully, suggesting that telomerase is not necessary for fertility or the maintenance of renewing tissues during develop-ment (Blasco et al., 1997). However, careful analysis of later-generation animals revealed that telomere repeats are lost gradually in each generation of mice, resulting in chromosome instability, such as aneuploidy and chromo-some end-to-end associations. Subsequent studies from Greider’s and Ronald DePinho’s groups revealed that telomerase is indeed required for successive maintenance of highly proliferative organs, such as testis, bone marrow and spleen, in later-generation mTR–/– mice, which show premature aging phenotypes at the organismal level, including skin lesions, alopecia and hair graying (Lee et al., 1998; Rudolph et al., 1999), supporting the notion that telomere shortening underlies the limited self-renewal capacity of stem cells in aged individuals.

Severe telomere shortening in late-generation mTR–/–

mice activates the p53-dependent cell cycle arrest pathway, and loss of both mTR and p53 synergisti-cally enhances malignant transformation (Chin et al., 1999). Such transformed cells in a p53–/– background often carry complex non-reciprocal translocations, a hall-mark of the BFB cycle during tumorigenesis (Artandi et al., 2000). These seminal studies using telomerase-deficient mice gave strong support to the telomere crisis model, named by Fuyuki Ishikawa, in which the two-stage model was extended to incorporate the BFB cycle as a driver of chromosome instability and consequently tumorigenesis during the crisis stage (de Lange, 1995; Ishikawa, 1997; Artandi and DePinho, 2000).

CURRENT PROGRESS AND FUTURE PERSPECTIVES

Telomeric proteins, transcripts and structures As described above, many important findings and concepts in telomere biology had been reported by the early 2000s.Although many other telomeric accessory factors have since been identified by proteomic analysis (Déjardin and Kingston, 2009; Lee et al., 2011; Grolimund et al., 2013; Bartocci et al., 2014) and the functions of these accessory factors are important in telomere maintenance (Arnoult and Karlseder, 2015), shelterin is still recognized as the only constitutive protein complex with a direct telomere-binding ability (Doksani and de Lange, 2014). Among shelterin components that directly bind to the telomere, TRF2 has a central role in protecting telomeric ends from activating the ATM-dependent DNA damage signaling pathway and from repair through classical NHEJ, while ATR-dependent signaling is repressed by POT1 (Denchi and de Lange, 2007). Another telomere-binding protein, TRF1, is also involved in suppression of ATR signaling through promoting telomeric semi-conservative replica-tion (Sfeir et al., 2009). The recent identification of TZAP, telomeric zinc finger-associated protein, as a TTAGGG-binding protein, however, surprised the field since it was reported to bind directly and preferentially to long dou-ble-stranded TTAGGG repeats (Li et al., 2017). Because TZAP competes with TRF2, negatively regulates telomere length and had been missed by conventional proteomics searches for shelterin-interacting proteins, it is an attrac-tive possibility that TZAP belongs to an unknown telo-meric complex.

Chromosome ends have long been thought to possess a suppressive structure against transcription because of their enrichment for heterochromatic marks (Blasco, 2007). Artificial insertion of a luciferase reporter into the subtelomere revealed 10 times less expression of the reporter compared to the same reporter in non-telomeric chromatin, suggesting that transcription is suppressed near telomeres, which has been named the telomere position effect (Baur et al., 2001). Despite this notion, a transcript that contains telomeric repeats was reported in 2007 by Joachim Lingner’s group and called telomeric repeat-containing RNA (TERRA) (Azzalin et al., 2007), proving for the first time that some subtelomeric loci are actively transcribed. Since its discovery, the TERRA non-coding transcript has been associated with proper telomere function through its interaction with vari-ous proteins, such as TRF1, TRF2, SUV39H1 and HP1 (Deng et al., 2009; López de Silanes et al., 2010; Scheibe et al., 2013; Porro et al., 2014). Interacting partners of TERRA also include telomerase RNA component, which suggests direct regulation of telomerase (Redon et al., 2010), although overexpression of TERRA does not affect telomerase activity in human cancer cells (Farnung et al.,

Page 7: Telomere biology in aging and cancer: early history and

113History of telomere biology

2012). TERRA can also form a DNA:RNA hybrid with telomeric DNA, called the R-loop, which has been impli-cated in telomere maintenance in ALT cells by regulating telomeric recombination (Arora et al., 2014). Because of its potential telomeric function in both telomerase-positive and telomerase-negative tumors, TERRA is considered as an attractive candidate therapeutic target (Cusanelli and Chartrand, 2015).

A number of studies clearly demonstrated that the telo-meric end has at least two states: a closed or capped state that protects chromosome ends from activating DNA dam-age checkpoints including the cell cycle arrest and damage repair pathways; and an open or uncapped state, in which both pathways are activated. Careful observation of telomere dysfunction-induced foci (TIFs), a colocalization of DNA damage marker proteins with telomeres (Takai et al., 2003), in multiple tumor cell lines indicated the existence of a third state, the intermediate state (Cesare et al., 2009) (Fig. 3). Many tumor cell lines as well as old normal human cells harbor unfused telomeres that are recognized as DNA damage, which indicates that these

intermediate-state telomeres activate the DNA damage checkpoint, yet are still protected from being subjected to repair processes (Cesare and Karlseder, 2012). The intermediate-state telomere is induced by extensive telo-mere shortening that prevents the telomere from form-ing the protective closed-state structure (e.g., the t-loop), and by partial depletion of TRF2 (e.g., by shRNA-based knockdown) that leads to a failure of closed-state forma-tion at an elongated telomere; on the other hand, the open state is a consequence of the complete loss of TRF2-binding sites, or deletion of TRF2 (e.g., by CRISPR/Cas9-based knockout) at an elongated telomere (Cesare et al., 2013). The intermediate-state telomere is proposed to induce replicative senescence without end-to-end fusion, while the open-state telomere is fused and underlies del-eterious chromosome instability and cell death during the telomere crisis stage.

The intermediate-state telomere can also be induced by a prolonged arrest in mitosis (Hayashi et al., 2012). The detailed mechanism is still unclear but such mitotic telo-mere deprotection requires Aurora B kinase activity, sug-

TRF1 TRF2 RAP1 TPP1TIN2 POT1Shelterin components

Open

Intermediate

Closed

NHEJCheckpoint

ONON

OFFON

OFFOFF

no TRF2

lessTRF2

enough repeats

and

enough TRF2

3’-overhang

3’

TTAGGG repeats

t-loop

Subtelomere

no repeats

fewer repeats

or

or

Fig. 3. Three-state model of telomere structure. The closed state is achieved when enough telomeric repeats and TRF2 are available to form the protective t-loop structure, in which the single-stranded 3’-overhang integrates into the double-stranded repeat locus. The closed state can suppress both ATM-mediated checkpoint signaling and the classical NHEJ pathway. It is not well understood how shelterin binds to the t-loop structure. When telomeric repeats shorten or TRF2 protein declines to a level that is insufficient to support the t-loop, the telomere end state becomes intermediate. Intermediate telomeres activate the ATM checkpoint but still hold enough TRF2 to suppress NHEJ. When almost all telomeric repeats or TRF2 are removed, the chromosome end state then becomes open, which resembles an internal DSB site and activates both checkpoint signaling and the NHEJ pathway. The intermediate state is relevant to telomeres in replicative senescence, while the open state is observed in the crisis stage. Each component of shelterin is highlighted at the bottom. TRF1 and TRF2 can bind to TIN2 independently of each other, forming subcomplexes.

Page 8: Telomere biology in aging and cancer: early history and

114 M. T HAYASHI

gesting an active pathway inducing the intermediate-state telomere during mitotic arrest. This partially explains a mechanism underlying cell death upon anti-tumor drug-induced mitotic arrest (Hayashi et al., 2015). The inter-mediate-state telomere may thus function as a biological sensor for cellular stresses, such as prolonged mitosis and cellular chronological aging (Cesare and Karlseder, 2012). What remains to be studied is whether the three-state model is relevant to tumorigenesis in vivo, and whether natural telomeres can take any additional states other than those described above.

Telomere crisis The consequences of chromosome end-to-end fusion have been well studied in the last decade. Although recent massive genome sequencing of malignant tumor cells indicated frequent duplications in opposite orientations, which is indicative of the BFB cycle (Campbell et al., 2010; Waddell et al., 2015), live-cell analysis of epithelial cells that harbor chromosome fusions demonstrated that an anaphase chromatin bridge formed between sister nuclei persists in the next cell cycle (Pampalona et al., 2012; Maciejowski et al., 2015). A persisting chromatin bridge results in either cytokinesis failure followed by tetraploidization (Pampalona et al., 2012), or resolution by enzymatic activities (Maciejowski et al., 2015). The latter was proposed to cause kataegis, a hypermutation pattern of clustered dC>dT and dC>dG changes at TpC sites, and chromothripsis, a catastrophic rearrangement of one or a few chromosome regions in a single event (Maciejowski and de Lange, 2017). These models, including the BFB cycle hypothesis, explain how a chromosome accumulates deleterious rearrangements during telomere crisis.

On the other hand, live-cell observation of human fibro-blast cells in telomere crisis indicated that cells often die during or after prolonged mitotic arrest. At least in fibroblasts, chromosome end-to-end fusion was shown to trigger mitotic arrest by an unknown mechanism, which leads to mitotic telomere deprotection and cell death dur-ing the same mitosis or the following cell cycle (Hayashi et al., 2015). Cell death following mitotic arrest is domi-nant in telomere crisis of fibroblasts, providing a novel insight about the mechanism of cell death during this catastrophic stage. It is worth noting that malignant tumors found in human patients are mostly of epithe-lial origin and fibroblast-derived sarcoma is very rare, suggesting distinct levels of resistance to tumorigenesis in these cell types. This may be explained by a strong preference for senescence in fibroblast cells compared to apoptosis in epithelial cells upon exposure to stress (Georgakopoulou et al., 2016). Another possibility that needs to be addressed, however, is that end-to-end fusion may follow a different process in each cell type, culminat-ing in either tumorigenesis or cell death during telomere crisis.

Telomeres, aging and disease It has been nearly 20 years since the first connection between telomere mainte-nance and dyskeratosis congenita was reported. Genetic diseases caused by malfunction of the telomere mainte-nance pathway have been increasingly reported and are collectively called the telomere syndromes (Armanios and Blackburn, 2012) or telomeropathies (Opresko and Shay, 2017). Mutations in genes known to function in telomere maintenance have now been identified in a variety of human genes involved in telomerase function (TR/TERC, TERT, DKC1, NOP10/NOLA3, NHP2/NOLA2, PARN, NAF1 and TCAB1/WRAP53) and telomere protection and replication (TIN2, RTEL1, POT1, CTC1, Apollo and TPP1) (Blackburn et al., 2015; Martínez and Blasco, 2017; Opresko and Shay, 2017). The cause of the telomere syn-dromes has been linked to the failure of stem cell replen-ishment, which requires a functional telomere mainte-nance pathway (Blackburn et al., 2015). What human telomere syndromes and mouse genetics have taught us is that telomere shortening caused by mutation in genes involved in telomere maintenance increases the risk of aging-related diseases and mortality.

Interestingly, mutations in the promoter of TERT, which increase hTERT expression only a few-fold, have been reported to cause familial and sporadic melanoma (Horn et al., 2013; Huang et al., 2013). This indicates that telomerase activity is highly regulated to be set in a certain range, otherwise the risks of aging diseases, including cancer incidence, increase. The field of telomere biology attracts public curiosity since it may have direct implications for human aging and life span. Because of this, however, careful studies will be required to dissect correlation and causality between telomere function and many aspects of human disease and health.

CONCLUDING REMARKS

As briefly discussed in the last section, the field of telo-mere biology will undoubtedly keep expanding to fulfil the demand of public interest in human health and dis-ease. Attaining a comprehensive understanding of the role of telomere dysfunction in aging and tumorigenesis will thus drive applied research, potentially leading to the development of effective prognostic indicators, improved therapeutic treatment, and extension of a healthy life span. As such, scientists are increasingly required to consider the impact of their work on the public. In this respect, learning how the seminal work of the pioneers has influenced current telomere biology research should help to understand the dynamics of the field in the near future. On the other hand, a number of leading stud-ies were the result of curiosity-driven basic research, which is an important lesson from history. Some break-throughs, such as the finding that Tetrahymena telomeres can protect the ends of minichromosome in evolutionarily

Page 9: Telomere biology in aging and cancer: early history and

115History of telomere biology

distant yeast cells, have even been the result of unex-pected findings. It is therefore of great importance to maintain both acknowledgement of the history of basic studies and a harmonized balance between basic and applied research for the sake of progress in science and human society.

I thank Jan Karlseder and Tomoichiro Miyoshi for critical read-ing of the manuscript, and Ian Smith for editing. This work is supported by grants from the Senri Life Science Foundation, the Uehara Memorial Foundation, the Daiichi Sankyo Foundation of Life Science, the Nakajima Foundation, a JSPS Grant-in-Aid for Young Scientists (A) (16H06176) and a JSPS Grant-in-Aid for Scientific Research on Innovative Areas (16H01406).

REFERENCES

Armanios, M., and Blackburn, E. H. (2012) The telomere syn-dromes. Nat. Rev. Genet. 13, 693–704.

Arnoult, N., and Karlseder, J. (2015) Complex interactions between the DNA-damage response and mammalian telo-meres. Nat. Struct. Mol. Biol. 22, 859–866.

Arora, R., Lee, Y., Wischnewski, H., Brun, C. M., Schwarz, T., and Azzalin, C. M. (2014) RNaseH1 regulates TERRA-telomeric DNA hybrids and telomere maintenance in ALT tumour cells. Nat. Commun. 5, 5220.

Artandi, S. E., Chang, S., Lee, S. L., Alson, S., Gottlieb, G. J., Chin, L., and DePinho, R. A. (2000) Telomere dysfunction promotes non-reciprocal translocations and epithelial can-cers in mice. Nature 406, 641–645.

Artandi, S. E., and DePinho, R. A. (2000) A critical role for telo-meres in suppressing and facilitating carcinogenesis. Curr. Opin. Genet. Dev. 10, 39–46.

Azzalin, C. M., Reichenbach, P., Khoriauli, L., Giulotto, E., and Lingner, J. (2007) Telomeric repeat containing RNA and RNA surveillance factors at mammalian chromosome ends. Science 318, 798–801.

Baker, S. J., Fearon, E. R., Nigro, J. M., Hamilton, S. R., Preisinger, A. C., Jessup, J. M., vanTuinen, P., Ledbetter, D. H., Barker, D. F., Nakamura, Y., et al. (1989) Chromosome 17 deletions and p53 gene mutations in colorectal carcinomas. Science 244, 217–221.

Bartocci, C., Diedrich, J. K., Ouzounov, I., Li, J., Piunti, A., Pasini, D., Yates III, J. R., and Lazzerini Denchi, E. (2014) Isolation of chromatin from dysfunctional telomeres reveals an important role for Ring1b in NHEJ-mediated chromo-some fusions. Cell Rep. 7, 1320–1332.

Baumann, P., and Cech, T. R. (2001) Pot1, the putative telomere end-binding protein in fission yeast and humans. Science 292, 1171–1175.

Baur, J. A., Zou, Y., Shay, J. W., and Wright, W. E. (2001) Telomere position effect in human cells. Science 292, 2075–2077.

Bell, D. R., and Van Zant, G. (2004) Stem cells, aging, and can-cer: inevitabilities and outcomes. Oncogene 23, 7290–7296.

Bilaud, T., Brun, C., Ancelin, K., Koering, C. E., Laroche, T., and Gilson, E. (1997) Telomeric localization of TRF2, a novel human telobox protein. Nat. Genet. 17, 236–239.

Bilaud, T., Koering, C. E., Binet-Brasselet, E., Ancelin, K., Pollice, A., Gasser, S. M., and Gilson, E. (1996) The telo-box, a Myb-related telomeric DNA binding motif found in proteins from yeast, plants and human. Nucleic Acids Res. 24, 1294–1303.

Blackburn, E. H., and Chiou, S. S. (1981) Non-nucleosomal pack-

aging of a tandemly repeated DNA sequence at termini of extrachromosomal DNA coding for rRNA in Tetrahy-mena. Proc. Nati. Acad. Sci. USA 78, 2263–2267.

Blackburn, E. H., Epel, E. S., and Lin, J. (2015) Human telomere biology: A contributory and interactive factor in aging, dis-ease risks, and protection. Science 350, 1193–1198.

Blackburn, E. H., and Gall, J. G. (1978) A tandemly repeated sequence at the termini of the extrachromosomal ribosomal RNA genes in Tetrahymena. J. Mol. Biol. 120, 33–53.

Blackburn, E. H., Greider, C. W., and Szostak, J. W. (2006) Telo-meres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging. Nat. Med. 12, 1133–1138.

Blasco, M. A. (2007) The epigenetic regulation of mammalian telomeres. Nat. Rev. Genet. 8, 299–309.

Blasco, M. A., Funk, W., Villeponteau, B., and Greider, C. W. (1995) Functional characterization and developmental regu-lation of mouse telomerase RNA. Science 269, 1267–1270.

Blasco, M. A., Lee, H. W., Hande, M. P., Samper, E., Lansdorp, P. M., DePinho, R. A., and Greider, C. W. (1997) Telomere shortening and tumor formation by mouse cells lacking telomerase RNA. Cell 91, 25–34.

Bodnar, A. G., Ouellette, M., Frolkis, M., Holt, S. E., Chiu, C. P., Morin, G. B., Harley, C. B., Shay, J. W., Lichtsteiner, S., and Wright, W. E. (1998) Extension of life-span by introduc-tion of telomerase into normal human cells. Science 279, 349–352.

Broccoli, D., Smogorzewska, A., Chong, L., and de Lange, T. (1997) Human telomeres contain two distinct Myb-related proteins, TRF1 and TRF2. Nat. Genet. 17, 231–235.

Bryan, T. M., Englezou, A., Dalla-Pozza, L., Dunham, M. A., and Reddel, R. R. (1997) Evidence for an alternative mechanism for maintaining telomere length in human tumors and tumor-derived cell lines. Nat. Med. 3, 1271–1274.

Bryan, T. M., Englezou, A., Gupta, J., Bacchetti, S., and Reddel, R. R. (1995) Telomere elongation in immortal human cells without detectable telomerase activity. EMBO J. 14, 4240–4248.

Bryan, T. M., and Reddel, R. R. (1997) Telomere dynamics and telomerase activity in in vitro immortalised human cells. Eur. J. Cancer 33, 767–773.

Campbell, P. J., Yachida, S., Mudie, L. J., Stephens, P. J., Pleasance, E. D., Stebbings, L. A., Morsberger, L. A., Latimer, C., McLaren, S., Lin, M.-L., et al. (2010) The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 467, 1109–1113.

Cesare, A. J., Hayashi, M. T., Crabbe, L., and Karlseder, J. (2013) The telomere deprotection response is functionally distinct from the genomic DNA damage response. Mol. Cell 51, 141–155.

Cesare, A. J., and Karlseder, J. (2012) A three-state model of telo-mere control over human proliferative boundaries. Curr. Opin. Cell Biol. 24, 731–738.

Cesare, A. J., Kaul, Z., Cohen, S. B., Napier, C. E., Pickett, H. A., Neumann, A. A., and Reddel, R. R. (2009) Spontane-ous occurrence of telomeric DNA damage response in the absence of chromosome fusions. Nat. Struct. Mol. Biol. 16, 1244–1251.

Chin, L., Artandi, S. E., Shen, Q., Tam, A., Lee, S. L., Gottlieb, G. J., Greider, C. W., and DePinho, R. A. (1999) p53 deficiency rescues the adverse effects of telomere loss and cooperates with telomere dysfunction to accelerate carcinogenesis. Cell 97, 527–538.

Chiu, C.-P., Dragowska, W., Kim, N. W., Vaziri, H., Yui, J., Thomas, T. E., Harley, C. B., and Lansdorp, P. M. (1996) Dif-

Page 10: Telomere biology in aging and cancer: early history and

116 M. T HAYASHI

ferential expression of telomerase activity in hematopoietic progenitors from adult human bone marrow. Stem Cells 14, 239–248.

Chong, L., Van Steensel, B., Broccoli, D., Erdjument-Bromage, H., Hanish, J., Tempst, P., and de Lange, T. (1995) A human telomeric protein. Science 270, 1663–1667.

Conrad, M. N., Wright, J. H., Wolf, A. J., and Zakian, V. A. (1990) RAP1 protein interacts with yeast telomeres in vivo: over-production alters telomere structure and decreases chromo-some stability. Cell 63, 739–750.

Counter, C. M., Avilion, A. A., LeFeuvre, C. E., Stewart, N. G., Greider, C. W., Harley, C. B., and Bacchetti, S. (1992) Telo-mere shortening associated with chromosome instability is arrested in immortal cells which express telomerase activ-ity. EMBO J. 11, 1921–1929.

Cusanelli, E., and Chartrand, P. (2015) Telomeric repeat-con-taining RNA TERRA: a noncoding RNA connecting telomere biology to genome integrity. Front. Genet. 6, 1–9.

de Lange, T. (1995) Telomere dynamics and genomic instability in human cancer. In Telomeres (eds.: Blackburn, E. H., and Greider, C. W.), pp. 265–293. Cold Spring Harbor Labora-tory Press, New York.

de Lange, T. (2005) Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev. 19, 2100–2110.

DeCaprio, J. A., Ludlow, J. W., Figge, J., Shew, J. Y., Huang, C. M., Lee, W. H., Marsilio, E., Paucha, E., and Livingston, D. M. (1988) SV40 large tumor antigen forms a specific com-plex with the product of the retinoblastoma susceptibility gene. Cell 54, 275–283.

Déjardin, J., and Kingston, R. E. (2009) Purification of proteins associated with specific genomic Loci. Cell 136, 175–186.

DeLeo, A. B., Jay, G., Appella, E., Dubois, G. C., Law, L. W., and Old, L. J. (1979) Detection of a transformation-related antigen in chemically induced sarcomas and other trans-formed cells of the mouse. Proc. Nat. Acad. Sci. USA 76, 2420–2424.

Denchi, E. L., and de Lange, T. (2007) Protection of telomeres through independent control of ATM and ATR by TRF2 and POT1. Nature 448, 1068–1071.

Deng, Z., Norseen, J., Wiedmer, A., Riethman, H., and Lieberman, P. M. (2009) TERRA RNA binding to TRF2 facilitates het-erochromatin formation and ORC recruitment at telo-meres. Mol. Cell 35, 403–413.

Doksani, Y., and de Lange, T. (2014) The role of double-strand break repair pathways at functional and dysfunctional telo-meres. Cold Spring Harb. Perspect. Biol. 6, 1–15.

Dunham, M. A., Neumann, A. A., Fasching, C. L., and Reddel, R. R. (2000) Telomere maintenance by recombination in human cells. Nat. Genet. 26, 447–450.

Farnung, B. O., Brun, C. M., Arora, R., Lorenzi, L. E., and Azzalin, C. M. (2012) Telomerase efficiently elongates highly transcribing telomeres in human cancer cells. PLoS One 7, e35714.

Feng, J., Funk, W. D., Wang, S. S., Weinrich, S. L., Avillion, A. A., Chiu, C. P., Adams, R. R., Chang, E., Allsopp, R. C., Yu, J., et al. (1995) The RNA component of human telomerase. Sci-ence 269, 1236–1241.

Finlay, C. A., Hinds, P. W., and Levine, A. J. (1989) The p53 proto-oncogene can act as a suppressor of transformation. Cell 57, 1083–1093.

Friend, S. H., Bernards, R., Rogelj, S., Weinberg, R. A., Rapaport, J. M., Albert, D. M., and Dryja, T. P. (1986) A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 323, 643–646.

Georgakopoulou, E., Evangelou, K., Havaki, S., Townsend, P., Kanavaros, P., and Gorgoulis, V. G. (2016) Apoptosis or senescence? Which exit route do epithelial cells and fibro-blasts preferentially follow? Mech. Ageing Dev. 156, 17–24.

Girardi, A. J., Jensen, F. C., and Koprowski, H (1965) SV40-induced transformation of human diploid cells: crisis and recovery. J. Cell. Comp. Physiol. 65, 69–84.

Gottschling, D. E., and Cech, T. R. (1984) Chromatin structure of the molecular ends of oxytricha macronuclear DNA: phased nucleosomes and a telomeric complex. Cell 38, 501–510.

Gottschling, D. E., and Zakian, V. A. (1986) Telomere proteins: specific recognition and protection of the natural termini of Oxytricha macronuclear DNA. Cell 47, 195–205.

Greider, C. W., and Blackburn, E. H. (1985) Identification of a specific telomere terminal transferase activity in Tetrahy-mena extracts. Cell 43, 405–413.

Greider, C. W., and Blackburn, E. H. (1987) The telomere ter-minal transferase of Tetrahymena is a ribonucleoprotein enzyme with two kinds of primer specificity. Cell 51, 887–898.

Griffith, J. D., Comeau, L., Rosenfield, S., Stansel, R. M., Bianchi, A., Moss, H., and de Lange, T. (1999) Mammalian telomeres end in a large duplex loop. Cell 97, 503–514.

Grolimund, L., Aeby, E., Hamelin, R., Armand, F., Chiappe, D., Moniatte, M., and Lingner, J. (2013) A quantitative telo-meric chromatin isolation protocol identifies different telo-meric states. Nat. Commun. 4, 1–12.

Hahn, W. C., Counter, C. M., Lundberg, A. S., Beijersbergen, R. L., Brooks, M. W., and Weinberg, R. A. (1999a) Creation of human tumour cells with defined genetic elements. Nature 400, 464–468.

Hahn, W. C., Stewart, S. A., Brooks, M. W., York, S. G., Eaton, E., Kurachi, A., Beijersbergen, R. L., Knoll, J. H., Meyerson, M., and Weinberg, R. A. (1999b) Inhibition of telomerase limits the growth of human cancer cells. Nat. Med. 5, 1164–1170.

Harley, C. B. (1991) Telomere loss: mitotic clock or genetic time bomb? Mutat. Res. 256, 271–282.

Harley, C. B., Futcher, A. B., and Greider, C. W. (1990) Telomeres shorten during ageing of human fibroblasts. Nature 345, 458–460.

Hartwell, L. H., and Weinert, T. A. (1989) Checkpoints: controls that ensure the order of cell cycle events. Science 246, 629–634.

Hastie, N. D., Dempster, M., Dunlop, M. G., Thompson, A. M., Green, D. K., and Allshire, R. C. (1990) Telomere reduction in human colorectal carcinoma and with ageing. Nature 346, 866–868.

Hayashi, M. T., Cesare, A. J., Fitzpatrick, J. A. J., Lazzerini-Denchi, E., and Karlseder, J. (2012) A telomere-dependent DNA damage checkpoint induced by prolonged mitotic arrest. Nat. Struct. Mol. Biol. 19, 387–394.

Hayashi, M. T., Cesare, A. J., Rivera, T., and Karlseder, J. (2015) Cell death during crisis is mediated by mitotic telomere deprotection. Nature 522, 492–496.

Hayflick, L., and Moorhead, P. S. (1961) The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621.

Horn, S., Figl, A., Rachakonda, P. S., Fischer, C., Sucker, A., Gast, A., Kadel, S., Moll, I., Nagore, E., Hemminki, K., et al. (2013) TERT promoter mutations in familial and sporadic mela-noma. Science 339, 959–961.

Houghtaling, B. R., Cuttonaro, L., Chang, W., and Smith, S. (2004) A dynamic molecular link between the telomere length regulator TRF1 and the chromosome end protector TRF2. Curr. Biol. 14, 1621–1631.

Huang, F. W., Hodis, E., Xu, M. J., Kryukov, G. V., Chin, L., and

Page 11: Telomere biology in aging and cancer: early history and

117History of telomere biology

Garraway, L. A. (2013) Highly recurrent TERT promoter mutations in human melanoma. Science 339, 957–959.

Ishikawa, F. (1997) Telomere crisis, the driving force in cancer cell evolution. Biochem. Biophys. Res. Commun. 230, 1–6.

Karlseder, J., Broccoli, D., Dai, Y., Hardy, S., and de Lange, T. (1999) p53- and ATM-dependent apoptosis induced by telo-meres lacking TRF2. Science 283, 1321–1325.

Kastan, M. B., Onyekwere, O., Sidransky, D., Vogelstein, B., and Craig, R. W. (1991) Participation of p53 protein in the cellu-lar response to DNA damage. Cancer Res. 51, 6304–6311.

Kim, N. W., Piatyszek, M. A., Prowse, K. R., Harley, C. B., West, M. D., Ho, P. L., Coviello, G. M., Wright, W. E., Weinrich, S. L., and Shay, J. W. (1994) Specific association of human telomerase activity with immortal cells and cancer. Science 266, 2011–2015.

Kim, S.-H., Kaminker, P., and Campisi, J. (1999) TIN2, a new regulator of telomere length in human cells. Nat. Genet. 23, 405–412.

Kipling, D., and Cooke, H. J. (1990) Hypervariable ultra-long telomeres in mice. Nature 347, 400–402.

Klobutcher, L. A., Swanton, M. T., Donini, P., and Prescott, D. M. (1981) All gene-sized DNA molecules in four species of hypo-trichs have the same terminal sequence and an unusual 3’ terminus. Proc. Nati. Acad. Sci. USA 78, 3015–3019.

Koprowski, H., Ponten, J. A., Jensen, F., Ravdin, R. G., Moorhead, P., and Saksela, E. (1962) Transformation of cultures of human tissue infected with simian virus SV40. J. Cell. Physiol. 59, 281–292.

Lane, D. P., and Crawford, L. V. (1979) T antigen is bound to a host protein in SV40-transformed cells. Nature 278, 261–263.

Lansdorp, P. M. (1998) Stem cell biology for the transfusion-ist. Vox Sang. 74, 91–94.

Lee, H. W., Blasco, M. A., Gottlieb, G. J., Horner, J. W., Greider, C. W., and DePinho, R. A. (1998) Essential role of mouse telom-erase in highly proliferative organs. Nature 392, 569–574.

Lee, O.-H., Kim, H., He, Q., Baek, H. J., Yang, D., Chen, L.-Y., Liang, J., Chae, H. K., Safari, A., Liu, D., et al. (2011) Genome-wide YFP fluorescence complementation screen identifies new regulators for telomere signaling in human cells. Mol. Cell. Proteomics 10, 1–11.

Li, B., Oestreich, S., and de Lange, T. (2000) Identification of human Rap1. Cell 101, 471–483.

Li, J. S. Z., Fusté, J. M., Simavorian, T., Bartocci, C., Tsai, J., Karlseder, J., and Denchi, E. L. (2017) TZAP: A telomere-associated protein involved in telomere length control. Sci-ence 355, 638–641.

Linzer, D. I., and Levine, A. J. (1979) Characterization of a 54K dalton cellular SV40 tumor antigen present in SV40-transformed cells and uninfected embryonal carcinoma cells. Cell 17, 43–52.

Liu, D., O’connor, M. S., Qin, J., and Songyang, Z. (2004a) Telo-some, a mammalian telomere-associated complex formed by multiple telomeric proteins. J. Biol. Chem. 279, 51338–51342.

Liu, D., Safari, A., O’connor, M. S., Chan, D. W., Laegeler, A., Qin, J., and Songyang, Z. (2004b) PTOP interacts with POT1 and regulates its localization to telomeres. Nat. Cell Biol. 6, 673–680.

López de Silanes, I., Stagno d’Alcontres, M., and Blasco, M. A. (2010) TERRA transcripts are bound by a complex array of RNA-binding proteins. Nat. Commun. 1, 1–10.

Lundblad, V., and Szostak, J. W. (1989) A mutant with a defect in telomere elongation leads to senescence in yeast. Cell 57, 633–643.

Lustig, A. J., Kurtz, S., and Shore, D. (1990) Involvement of the silencer and UAS binding protein RAP1 in regulation of telomere length. Science 250, 549–553.

Maciejowski, J., and de Lange, T. (2017) Telomeres in cancer: tumour suppression and genome instability. Nat. Rev. Mol. Cell Biol. 18, 175–186.

Maciejowski, J., Li, Y., Bosco, N., Campbell, P. J., and de Lange, T. (2015) Chromothripsis and kataegis induced by telomere crisis. Cell 163, 1641–1654.

Makarov, V. L., Hirose, Y., and Langmore, J. P. (1997) Long G tails at both ends of human chromosomes suggest a C strand degradation mechanism for telomere shortening. Cell 88, 657–666.

Martínez, P., and Blasco, M. A. (2017) Telomere-driven diseases and telomere-targeting therapies. J. Cell Biol. 216, 875–887.

McClintock, B. (1931) Cytological observations of deficiencies involving known genes, translocations and an inversion in Zea mays. Missouri Agricultural Exp. Station Res. Bull. 163, 1–30.

McClintock, B. (1941) The stability of broken ends of chromo-somes in Zea mays. Genetics 26, 234–282.

Metcalfe, J. A., Parkhill, J., Campbell, L., Stacey, M., Biggs, P., Byrd, P. J., and Taylor, A. M. R. (1996) Accelerated telomere shortening in ataxia telangiectasia. Nat. Genet. 13, 350–353.

Meyerson, M., Counter, C. M., Eaton, E. N., Ellisen, L. W., Steiner, P., Caddle, S. D., Ziaugra, L., Beijersbergen, R. L., Davidoff, M. J., Liu, Q., et al. (1997) hEST2, the putative human telomerase catalytic subunit gene, is up-regulated in tumor cells and during immortalization. Cell 90, 785–795.

Meyne, J., Ratliff, R. L., and MoYzIs, R. K. (1989) Conservation of the human telomere sequence (TTAGGG) n among verte-brates. Proc. Nati. Acad. Sci. USA 86, 7049–7053.

Mitchell, J. R., Wood, E., and Collins, K. (1999) A telomerase component is defective in the human disease dyskeratosis congenita. Nature 402, 551–555.

Morin, G. B. (1989) The human telomere terminal transferase enzyme is a ribonucleoprotein that synthesizes TTAGGG repeats. Cell 59, 521–529.

Moyzis, R. K., Buckingham, J. M., Cram, L. S., Dani, M., Deaven, L. L., Jones, M. D., Meyne, J., Ratliff, R. L., and Wu, J. R. (1988) A highly conserved repetitive DNA sequence, (TTAGGG)n, present at the telomeres of human chromo-somes. Proc. Nati. Acad. Sci. USA 85, 6622–6626.

Muller, H. J. (1938) The remaking of chromosomes. Collecting Net 13, 181–198.

Murnane, J. P., Sabatier, L., Marder, B. A., and Morgan, W. F. (1994) Telomere dynamics in an immortal human cell line. EMBO J. 13, 4953–4962.

Nakamura, T. M., Morin, G. B., Chapman, K. B., Weinrich, S. L., Andrews, W. H., Lingner, J., Harley, C. B., and Cech, T. R. (1997) Telomerase catalytic subunit homologs from fission yeast and human. Science 277, 955–959.

Nakayama, J., Tahara, H., Tahara, E., Saito, M., Ito, K., Nakamura, H., Nakanishi, T., Ide, T., and Ishikawa, F. (1998) Telomerase activation by hTRT in human normal fibroblasts and hepatocellular carcinomas. Nat. Genet. 18, 65–68.

Olovnikov, A. M. (1971) Principle of marginotomy in template synthesis of polynucleotides. Dokl. Akad. Nauk SSSR 201, 1496–1499.

Olovnikov, A. M. (1973) A theory of marginotomy: The incom-plete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenom-

Page 12: Telomere biology in aging and cancer: early history and

118 M. T HAYASHI

enon. J. Theor. Biol. 41, 181–190.Opresko, P. L., and Shay, J. W. (2017) Telomere-associated aging

disorders. Ageing Res. Rev. 33, 52–66.Pampalona, J., Frías, C., Genescà, A., and Tusell, L. (2012) Pro-

gressive telomere dysfunction causes cytokinesis failure and leads to the accumulation of polyploid cells. PLoS Genet. 8, 1–11.

Porro, A., Feuerhahn, S., Delafontaine, J., Riethman, H., Rougemont, J., and Lingner, J. (2014) Functional charac-terization of the TERRA transcriptome at damaged telo-meres. Nat. Commun. 5, 1–13.

Prowse, K. R., and Greider, C. W. (1995) Developmental and tissue-specific regulation of mouse telomerase and telomere length. Proc. Nati. Acad. Sci. USA 92, 4818–4822.

Rabson, A. S., and Kirschstein, R. L. (1962) Induction of malig-nancy in vitro in newborn hamster kidney tissue infected with simian vacuolating virus (SV40). Proc. Soc. Exp. Biol. Med. 111, 323–328.

Redon, S., Reichenbach, P., and Lingner, J. (2010) The non-coding RNA TERRA is a natural ligand and direct inhibitor of human telomerase. Nucleic Acids Res. 38, 5797–5806.

Rudolph, K. L., Chang, S., Lee, H. W., Blasco, M., Gottlieb, G. J., Greider, C., and DePinho, R. A. (1999) Longevity, stress response, and cancer in aging telomerase-deficient mice. Cell 96, 701–712.

Sanger, F., and Coulson, A. R. (1975) A rapid method for deter-mining sequences in DNA by primed synthesis with DNA polymerase. J. Mol. Biol. 94, 441–448.

Scheibe, M., Arnoult, N., Kappei, D., Buchholz, F., Decottignies, A., Butter, F., and Mann, M. (2013) Quantitative interaction screen of telomeric repeat-containing RNA reveals novel TERRA regulators. Genome Res. 23, 2149–2157.

Sfeir, A., Kosiyatrakul, S. T., Hockemeyer, D., MacRae, S. L., Karlseder, J., Schildkraut, C. L., and de Lange, T. (2009) Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138, 90–103.

Sharpless, N. E., and DePinho, R. A. (2004) Telomeres, stem cells, senescence, and cancer. J. Clin. Invest. 113, 160–168.

Shein, H. M., and Enders, J. F. (1962) Transformation induced by simian virus 40 in human renal cell cultures, I. Mor-phology and growth characteristics. Proc. Nati. Acad. Sci. USA 48, 1164–1172.

Szostak, J. W., and Blackburn, E. H. (1982) Cloning yeast telo-meres on linear plasmid vectors. Cell 29, 245–255.

Takai, H., Smogorzewska, A., and de Lange, T. (2003) DNA dam-age foci at dysfunctional telomeres. Curr. Biol. 13, 1549–1556.

van Steensel, B., and de Lange, T. (1997) Control of telomere length by the human telomeric protein TRF1. Nature 385, 740–743.

van Steensel, B., Smogorzewska, A., and de Lange, T. (1998) TRF2 protects human telomeres from end-to-end fusions. Cell 92, 401–413.

Van Zant, G., and Liang, Y. (2003) The role of stem cells in aging. Exp. Hematol. 31, 659–672.

Vaziri, H., Dragowska, W., Allsopp, R. C., Thomas, T. E., Harley, C. B., and Lansdorp, P. M. (1994) Evidence for a mitotic clock in human hematopoietic stem cells: loss of telomeric DNA with age. Proc. Nati. Acad. Sci. USA 91, 9857–9860.

Waddell, N., Pajic, M., Patch, A.-M., Chang, D. K., Kassahn, K. S., Bailey, P., Johns, A. L., Miller, D., Nones, K., Quek, K., et al. (2015) Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 518, 495–501.

Watson, J. D. (1972) Origin of concatemeric T7 DNA. Nat. New Biol. 239, 197–201.

Weng, N. P., Levine, B. L., June, C. H., and Hodes, R. J. (1996) Regulated expression of telomerase activity in human T lymphocyte development and activation. J. Exp. Med. 183, 2471–2479.

Wright, W. E., and Shay, J. W. (1992) The two-stage mechanism controlling cellular senescence and immortalization. Exp. Gerontol. 27, 383–389.

Xia, S. J., Shammas, M. A., and Shmookler Reis, R. J. (1996) Reduced telomere length in ataxia-telangiectasia fibro-blasts. Mutat. Res. 364, 1–11.

Ye, J. Z.-S., Hockemeyer, D., Krutchinsky, A. N., Loayza, D., Hooper, S. M., Chait, B. T., and de Lange, T. (2004) POT1-interacting protein PIP1: a telomere length regulator that recruits POT1 to the TIN2/TRF1 complex. Genes Dev. 18, 1649–1654.

Zhong, Z., Shiue, L., Kaplan, S., and de Lange, T. (1992) A mam-malian factor that binds telomeric TTAGGG repeats in vitro. Mol. Cell. Biol. 12, 4834–4843.

Zhu, X.-D., Küster, B., Mann, M., Petrini, J. H. J., and de Lange, T. (2000) Cell-cycle-regulated association of RAD50/MRE11/NBS1 with TRF2 and human telomeres. Nat. Genet. 25, 347–352.