Sie sind auf Seite 1von 22

View Online / Journal Homepage / Table of Contents for this issue

Chem Soc Rev Dynamic Article Links

Cite this: Chem. Soc. Rev., 2011, 40, 27192740

www.rsc.org/csr CRITICAL REVIEW


Chemistry in human telomere biology: structure, function and targeting
of telomere DNA/RNA
Yan Xu*
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

Received 5th October 2010


DOI: 10.1039/c0cs00134a

Telomeres are present at the ends of all eukaryotic chromosomes. Human telomeres play an important
Downloaded by Pennsylvania State University on 14 May 2012

role in critical processes underlying genome stability, cancer, and aging, and their importance was
recognized via the award of the 2009 Nobel Prize in Physiology or Medicine. Chemistry has made vast
and almost unparalleled contributions to telomere biology. This critical review highlights the
contributions of chemistry in human telomeres and summarizes the signicant development of human
telomere biology. First, I provide an overview of the advances in understanding of the structures and
functions of human telomeres. Second, I focus on the current eorts on developing various chemical
approaches to targeting human telomeres and telomerase for the treatment of cancer. Third, studies on
a newly discovered telomeric repeat-containing RNA are discussed in detail. Last, future challenges in
the eld are outlined, including perspectives of both chemistry and biology (412 references).

1. Introduction telomeres protect chromosomes from end-to-end fusion.13


The word telomere is derived from the Greek nouns telos
The telomere history was beginning to nd the special properties meaning end and meros meaning part. Though crucial for
of chromosome ends (Fig. 1). Landmark observations by many cellular functions, telomeres are subject to progressive
Muller and McClintock in the 1930s and 1940s indicated that decreases in length. Small DNA segments at each telomere cannot
be copied and are lost during DNA replication and cell division.
This puzzle was referred as the end replication problem.4
Division of Chemistry, Department of Medical Sciences, Faculty of
Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, Reaching the critical point of telomere shortening (the Hayick
889-1692, Japan. E-mail: xuyan@mkomi.rcast.u-tokyo.ac.jp, limit)57 leads cells to senescence or death, and it is assumed that
xuyan@fc.miyazaki-u.ac.jp; Fax: +81 0985-85-0993; this may serve as a molecular clock for cellular lifespan. The
Tel: +81 0985-85-0993 existence of a reverse transcriptase that could synthesize telomeric
DNA, discovered in 1985 by Greider and Blackburn, named after
telomerase, provided the experimental answer to this problem.8 A
Yan Xu earned his PhD at few years later, homologous recombination between telomeric
Tokyo Medical and Dental sequences was also shown to elongate telomeres by the alter-
University, where he studied
native lengthening of telomeres pathway (ALT).9
non-natural nucleic acids and
There are still other fundamental problems with the ends of
small molecules to stabilize
Z-form DNA and photo- linear chromosomes. Natural chromosome ends are recognized
chemical methods of probing as double-stranded breaks and get degraded by nucleases.1015
telomere G-quadruplex struc- Telomeres provide critical protection of chromosomes from
tures. Then he won a Japan such detriments. It has been suggested that the protective
Science and Technology Fellow- function of telomeres might depend on their state, e.g.,
ship at Kyoto University, whether they have capping or uncapping structures.1623
studying the structure and Telomeres have been likened to the aglets (tips) on the ends of
function of DNA by chemical shoelaces that keep them from fraying. Recent studies suggest
approaches. He became an that telomeres may form higher order structures to achieve the
Yan Xu assistant professor at The
capping function.2325 The contributions of both telomere
University of Tokyo in 2007.
Currently, he is an associate professor at the Faculty of DNA and various telomere-binding proteins to such structures
Medicine, University of Miyazaki. His group focuses on (i) have gained appreciation.2628 What are the structural features
understanding structure and function of human telomere DNA of telomeres and through which mechanisms do they ensure
and RNA, (ii) developing chemical approaches to targeting chromosome end protection? What is the molecular basis of
human telomere for discovering anticancer agents. the telomeric cap?

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2719
View Online

Fig. 1 Timeline of telomere studies.

Telomerase or its telomere DNA substrate was rst


Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

recognized as a unique and exciting anticancer target and has


been a major focus in cancer research since the mid-1990s.2946
The key advantages of targeting telomerase or telomeres
in comparison with most other cancer targets are their
Downloaded by Pennsylvania State University on 14 May 2012

relative universality, criticality, and specicity for cancer


cells.4760 Recently, telomere function has been directly
implicated in several other diseases such as dyskeratosis
congenita and idiopathic pulmonary brosis.6170 Chemical
approaches have been developed for targeting human telomeres
and telomerase for cancer therapy.7174 What is a structural
basis for the inhibition mechanism? What are the prospects for
the use of anti-telomerase and anti-telomere agents as drugs?
For a long time, telomeres have been considered to be
transcriptionally silent. A recent nding demonstrated that
telomere DNA is transcribed into telomeric repeat-containing
RNA (TERRA).75,76 Telomeric RNA, a newly emerging
player in telomere biology, may be a key component of
telomere machinery.77 Studies of telomeres have focused on
telomere DNA and its related proteins. The identication of
TERRA RNA in the chromosome end may change this Fig. 2 The green staining marks the telomeres.
situation. What are the structures and functions of human
telomeric RNA? How is telomeric RNA specically associated For example, duplex DNA can adopt a variety of sequence-
with telomeric DNA? dependent secondary structures that range from the canonical
The understanding of telomeres and telomerase is expected right-handed B form to the left-handed Z form.90,91 Triplex
to provide major insights into genome stability, cancer, and and tetraplex DNA structures are also known to exist.9193
telomere-related diseases. Fundamental knowledge from many Such local DNA conformations have been suggested to be
elds, including chemistry, structural biology, cell biology, biologically important in processes such as DNA replication,
and medicinal chemistry, have opened up the possibility of gene expression and regulation, and DNA damage repair.92,93
targeting telomerase, telomeres, and telomere-binding proteins For example, Z-DNA, a left-handed DNA conformation that
in therapeutic strategies against cancer and aging-related favors alternating GC sequences, is required for chromatin-
pathologies. Thousands of publications and numerous review dependent activation of the colony stimulating factor-1
articles have described signicant advances and key questions (CSF1) promoter.94
of human telomeres.7888 This review will highlight the Human telomeric DNA consists of a duplex region
contribution of chemistry to human telomeres, especially in composed of TTAGGG repeats, which terminates in a
the detailed chemical structure of telomere DNA at the atomic 100200-nucleotide (nt) G-rich single-stranded overhang.9597
level and the status of chemistry-based challenges of drug Fluorescent in situ hybridization showing human telomeres is
development. The intention of this article is not to shown in Fig. 2, indicating that telomeres are present at the
exhaustively cover the sphere of human telomere research, ends of chromosomes. Two structures of the 3 0 single-stranded
but to provide both insight and inspiration in the eld and overhang have been proposed: one suggests the formation of
stimulate further research to develop new chemical G-quadruplexes, while the other proposes the presence of a
approaches. t-loop.

2.1 G-quadruplex
2. Human telomere structure
G-quadruplexes are formed by the stacking of several
Besides encoding genetic information, DNA itself can take on G-tetrads and are constituted by four backbone strands
conformations other than the right-handed B-DNA structure.8991 in which several loops connect these strands (Fig. 3).

2720 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

(Fig. 4e), in which the interveningTTAbases that


occur between G-tetrad-forming guanines might form two
WatsonCrick base pairs linking two strands of opposite
polarity.
NMR spectroscopy by Phan et al. dened a dimeric
G-quadruplex formed by 16-mer d(GGGTTAGGGTTAGGGT)
and 6-mer d(TAGGGT) human telomeric sequences (Fig. 4f),
suggesting an association between the two telomere
sequences.129 This structure contains the (3+1) hybrid
G-quadruplex topology, in which three strands are oriented
in one direction and one is oriented in the opposite direction.
Fig. 3 Four guanines are arranged in a plane to form the G-tetrad by In 1993, an antiparallel G-quadruplex of telomeric sequence
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

hydrogen bonds. G-quadruplexes are formed by the stacking of several d[AGGG(TTAGGG)3] was observed by NMR in Na+ solution
G-tetrads. (Fig. 4g).130 NMR analysis by Patel et al. revealed an intra-
molecular basket-type structure in which the opposing GGG
Four guanines are arranged in a plane to form the G-tetrad by columns are antiparallel, with one diagonal and two lateral
Downloaded by Pennsylvania State University on 14 May 2012

hydrogen bonds and are stabilized by cations. The assembly of TTA loops and the syn/anti mixed guanines. On the other
guanine and its analog was discovered to form gels in 1910, hand, the same 22-mer sequence adopts a completely dierent,
and diraction studies by Gellert et al. in 1962 revealed the propeller-type structure in a crystal grown in the presence of
tetrameric arrangement between guanosine residues.98 A wider K+ ions (Fig. 4h).123 In this structure, four core GGGs are
interest in such structures has promoted two international parallel, with the three linking external loops positioned on the
meetings on this exciting topic.99101 G-quartet core exterior. Tan et al. suggested that the
A large number of dierent structures have been observed in molecular crowding condition was to stabilize the parallel
the human telomere.102111 The dierent G-quadruplex topologies G-quadruplex conformation.131,132
may be associated with some related aspects: the syn/anti In physiologically-relevant K+ solution, various four-
conformation of guanine residues, the relative orientation of repeat sequences show several very distinct folds. The
the G-quartet core, the types of linking loops, the sequences d[AGGG(TTAGGG)3], d[TAGGG(TTAGGG)3], and
and lengths of telomere DNA, and the nature of the associated d[TTAGGG(TTAGGG)3] sequences adopt a hybrid-type
metal cations.112118 G-quadruplex in which the loop types from the 5 0 end to the
Early NMR studies suggested that human telomeric 30 end are external, lateral, and lateral (Fig. 4i),133139 whereas the
sequences d(TTAGGG) and d(TTAGGGT) form a parallel d[TAGGG(TTAGGG)3TT] and d[TTAGGG(TTAGGG)3TT]
G-quadruplex in K+ solution with three G-tetrads in all anti- sequences form the hybrid-type G-quadruplex that diers in
guanines and lack of linking loops (Fig. 4a).119 Dimerization the order of loop arrangements,137,139,140 i.e., lateral, lateral,
of two such G-quadruplexes was demonstrated to occur and external from the 5 0 end (Fig. 4j). The sequence
through end-to-end stacking of 3 0 -terminal G-tetrads d[TAGGG(TTAGGG)3T] can interconvert the two orders of
(Fig. 4b).119,120 For the d(AGGGT) and d(TTAGGGT), a loop arrangements in K+ solution.111,139 In addition, the
novel A-tetrad was formed to stabilize the G-quadruplex, in d[AGGG(TTAGGG)3] sequence can fold into another anti-
which four adenines were held together in a plane by hydrogen parallel conformation, the so-called chair with the lateral
bonds.121,122 loops at the bottom of the G-quadruplex (Fig. 4k).133,141
The K+-stabilized crystal structure of d(AGGGTTAGGGT) Recently, an anti-parallel basket-type G-quadruplex formed
has been determined as shown in Fig. 4c,123 indicating a by the d[GGG(TTAGGG)3T] sequence in K+-containing
parallel G-quadruplex with two external loops abutting the solutions has been identied by NMR studies (Fig. 4l) that
sides of the G-tetrads and guanines in anti conformation. A is very dierent from the other known topologies, involving
TATA tetrad was observed in the dimer structure. In K+ only two G-tetrads together with a ve-nucleotide diagonal
solution, the same sequence adopts two dimeric parallel and loop and two lateral loops.142 The d[GGG(TTAG3)4] sequence
antiparallel G-quadruplexes, as reported by NMR studies.124 in K+ solutions was suggested for example to form a hybrid-
The parallel form is similar to the K+-stabilized crystal type G-quadruplex in which the extra TTAGG repeat was
structure observed in the crystalline state (Fig. 4c). The anti- used to generate an external loop (Fig. 4m).126 The conformational
parallel form has the opposing GGG columns with lateral diversity of human telomeric G-quadruplexes in K+ solution
loops on the ends of G-quadruplex and the syn/anti mixed conrms that a particular topology is not only associated with
guanines (Fig. 4d). Similarly, a human telomeric sequence some related external aspects but is also dependent on its own
d(GGGTTAGGG) was shown to form both antiparallel and sequence, the 5 0 and 3 0 end sequences in particular. Other
parallel bimolecular tetraplexes in K+-containing solutions.125,126 G-quadruplex folds have been proposed for human telomeric
In Na+ solution, the two similar sequences were suggested to sequences under dierent experimental conditions.125,126,143146
adopt the antiparallel-stranded G-quadruplexes.126,127 For Short human telomeric sequences as models for structural
d(TTAGGGTTAGGG), Na+ induces a tetrameric antiparallel studies of the telomere have provided important information
G-quadruplex (Fig. 4d), whereas K+ stabilizes the dimeric for understanding the structure of human telomeres. Little is
parallel and tetrameric antiparallel structures.128 The tetrameric known, however, about the structure of longer overhang
structure is made up of four strands with antiparallel orientation sequences of the telomere repeat. Most studies have focused

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2721
View Online
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A
Downloaded by Pennsylvania State University on 14 May 2012

Fig. 4 Schematic structures of human telomeric G-quadruplexes and i-motif. AFM image of higher-order telomeric DNA structures.

on individual G-quadruplexes formed by short telomeric structure was suggested to facilitate stacking of the 22-mer
DNA (especially 22-mer unimolecular G-quadruplexes). telomere units into higher-order packing structures (Fig. 4o).
However, it might be more biologically relevant to take into Because the 5 0 and 3 0 ends of the hybrid-type G-quadruplex
account the real length of the 3 0 -terminal single-stranded point in opposite directions, this structure should easily fold
overhang of human telomeric DNA in vivo (100200 nt) to into a higher-order packing structure by consecutive formation
directly reveal the structural features of long-telomeric-overhang of G-quadruplex units. End-to-end connection is simply
DNA, owing to its similarity in length to that of the intra- accomplished by a TTA linker to connect each 22-mer
cellular telomere. G-quadruplex structure. To gain structural information on
In 2002, Parkinson and Neidle et al. suggested that oligo- human single-stranded telomeric-overhang DNA, we visualized
merization of individual parallel G-quadruplexes can be simply the elongated telomere sequence by atomic force microscopy
performed to form a higher-order structure by inserting an (AFM).148 The AFM image clearly shows that the higher-
additional TTA loop to each 22-mer structure (Fig. 4n).123 A order G-quadruplex consists of blob-shaped protrusions
series of computational methods has been used to characterize arranged end-to-end and formed by adjacent G-quadruplex
the properties of the higher-order G-quadruplex with parallel- units (Fig. 4p).133,134 In further support of the dimeric
stranded topology, conrming that the overall multimer G-quadruplex, a study by molecular dynamics simulation
G-quadruplex becomes more stable with the addition of showed a stable dimeric G-quadruplex structure for human
further G-tetrads and that the TTA loop is the most exible telomeric DNA in which the interface between G-quadruplex
part of the model.147 The unique topology of the hybrid-type units is stabilized by specic stacking interactions of loop

2722 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

nucleotides (Fig. 4q).149 In other models, the G-quadruplexes present in one macronucleus.162 Although these approaches
formed by long telomeric DNAs are beads-on-a-string gave some structural information, the concentration of
structures (Fig. 4r).150 In this structure, two G-quadruplex G-quadruplexes in humans is too low to be detected in the
units are connected by one linker without a stacking inter- presence of only a few dozen chromosome ends (92 telomeres
action between the units and they can move relatively per cell in G1 of the cell cycle and twice as many in G2).
independently of each other. Nucleic acid structures are dicult to probe in vivo; in fact,
direct evidence for human telomeric DNA G-quadruplexes
2.2 G-quadruplex probe existing in cells has not yet been obtained. Therefore, a more
To gain a better understanding of telomere DNA conformations, eective chemical method of probing G-quadruplex structures
various studies have been performed using platinum cross- in living cells is desired.163
linking, FRET,125I-radioprobing, covalent ligation, click
reaction, sedimentation, and small molecule probes.151159 2.3 T-loop
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

We developed a photochemical method to distinguish between Using the puried telomeric fragments of human and mouse,
parallel and anti-parallel telomere G-quadruplexes.160 The t-loops were rst observed by electron microscopy in 1999.164
photoreactivity of iodouracil-containing human telomeric Telomeres have been shown to form a t-loop, a lasso-like
DNA is highly dependent on the DNA structure. A photo- structure in which the 3 0 -overhang invades the double-
Downloaded by Pennsylvania State University on 14 May 2012

reaction product, the 20 -deoxyribonolactone residue, is eectively stranded region of telomeric DNA (Fig. 6a). Although the
produced only in the diagonal loop of the anti-parallel atomic-level structural details of the loop are not known, the
G-quadruplex (Fig. 5a). As 5-halouracil-substituted DNA is G-rich 3 0 overhang must be incorporated into the double-
functional in living cell systems such as Escherichia coli, use of stranded telomeric region in a manner of base pairing with the
the photochemical reaction of 5-halouracil-containing DNA C-strand. The strand invasion takes place at a distance from
would provide a powerful tool to probe telomere G-quadruplex the terminal part of the duplex telomere region and, therefore,
conformations in living cells. results in a large duplex lariat structure. The circular parts of
In a study using platinum cross-linking, only antiparallel the t-loops range in size from very small (1 kb) to 425 kb.
G-quadruplex conformation was detected in both Na+- and A displacement (D) loop is involved in the t-loop in which the
K+-containing solutions.151 Single-molecule uorescence 3 0 overhang provides the primer for DNA synthesis and
energy transfer revealed that the antiparallel and parallel telomere extension (Fig. 6a). In addition to providing a
structures can coexist and interconvert under near-physiological mechanism for extension, the D-loop is a dynamic structure,
conditions.152,153 The conformational diversity of human telomeric which can be stabilized by branch migration to form a
G-quadruplexes is demonstrated by using 125I-radioprobing Holliday junction (HJ). In order to observe t-loops, it is
based on the comparison of the probability of breaks with necessary to introduce interstrand cross-links with psoralen
the distance from 125I to the corresponding nucleotides.141 and UV. Without the chemical cross-linking treatment that
Chemical ligation occurring at loop positions of unimolecular stabilizes the strand invasion, loops were observed at very low
G-quadruplexes is also used to explore the various solution frequency ratios (o5%).165,166
conformations of human telomere sequences.154 More recently,
a study utilizing double-nitroxide-modied (spin label) telomeric
sequences demonstrates the presence of a 1 : 1 mixture of the
parallel and antiparallel G-quadruplex structures in solution
by investigating the distance between the spin-labeled nucleo-
tides in the dierent G-quadruplexes (Fig. 5b).161
Using a single-chain antibody, Schatzel et al. suggested
that Stylonychia lemnae telomeric DNA forms an anti-parallel
G-quadruplex in vivo, in which about 2  108 telomeres are

Fig. 6 (a) t-loop. (b) A lariat model containing G-quadruplex.


Fig. 5 (a) A photochemical method was used to distinguish between (c) A segment containing 16-mer G-tracts can bind to the G-tract of
parallel and anti-parallel telomere G-quadruplexes. (b) The distance another segment of telomere DNA by forming G-quadruplex when the
between the spin-labeled nucleotides (orange arrows) in the dierent 3 0 -end invades the adjacent double-stranded segment of the telomere
G-quadruplexes was used to determine these conformations. to form the t-loop.

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2723
View Online

Thanks to the cross-linking reaction provided by chemistry,


the t-loop has been obtained as a possible telomere structure
by biologists. However, lack of evidence of their existence in
cells as truly biologically relevant structures remains a
challenging problem. Resolution of this question may have
to await the development of a chemical approach for t-loop
visualization.

2.4 Relationship between G-quadruplex and t-loop


Recently, a structural study suggested that the G-quadruplex
structure can stabilize a relatively large lariat structure Fig. 7 Human telomeres associate with the six-protein complex
(t-loop).167 The model revealed a possible relationship between shelterin.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

these two structures in which G-quadruplex formation induces


the 3 0 -overhang strand encroaching into the double-stranded
region (Fig. 6b and c). This model may provide conformational Protection of telomeres 1 (POT1) directly binds the 3 0 -single-
exibility for chromosome ends in response to environmental stranded overhang of the chromosome end in its
Downloaded by Pennsylvania State University on 14 May 2012

conditions such as protein binding. Furthermore, another N-terminus.177,178 The single-stranded DNA-binding domain
puzzle raised by the t-loop structure may be solved, as the in the N-terminus allows the proteins to bind to arrays of the
fact that it is stabilized by the 3 0 -overhang strand encroaching sequence TAGGGTTAG with great sequence specicity. It is
into the double-stranded region cannot explain the inhibition suggested that POT1 is able to disrupt the highly folded
of telomerase activity by a G-quadruplex stabilizing molecule, intramolecular G-quadruplex structures.179
as discussed in detail below. The present model indicates that TRF2- and TRF1-interacting nuclear protein 2 (TIN2)
not only strand encroachment but also G-quadruplex bridges TRF1 and TRF2 to the TPP1POT1 heterodimer,
formation could stabilize the t-loop structure,71,167 which therefore linking the duplex component of the telomeres to the
strikes a happy medium between the G-quadruplex and the single-stranded overhang.180184 TIN2 interacts with TRF1 at
t-loop (Fig. 6c). This may provide the exibility for responding its C-terminus and associates with a hinge domain of TRF2 at
to environmental conditions as protein binding. the N-terminus. Using a third protein interaction site located
The G-quadruplex confers an energetically preferable in its N-terminus, TIN2 binds to TPP1 and forms a complex.
conformation. However, these two structures of the 3 0 end Therefore, TIN2 binds to TRF1, TRF2, and TPP1 and
overhang might also co-exist. Nonetheless, the existence of a occupies a central position in shelterin.
folding of the G-strand overhang into a G-quadruplex has to Repressor/Activator protein 1 (Rap1) lacks DNA-
be considered most plausible in human cells based on the binding activity in human and is dependent on its interaction
available experimental evidence. with TRF2 for telomere binding.185,186 Rap1 forms a complex
with TRF2 by a C-terminal domain that mediates the
2.5 i-motif interaction with a short helical region in the hinge domain
It is known that the telomeric C-strand can also fold in a novel of TRF2.
structure, the i-motif, in which two parallel-stranded duplexes TIN2 and POT1 interacting protein 1 (TPP1) interacts with
associate in a head-to-tail orientation with intercalation POT1 and TIN2 by the POT1-binding domain and C-terminal
of the CC+ base pairs (Fig. 4s).168 This structure is stabilized of TIN2.187 The connection with TPP1 is critical for the
with a low pH through protonation of cytosines. It has association of POT1 with telomeres, providing a main way
been indicated that this conformation could participate in by which POT1 is recruited to telomeres.188,189
the dynamics of the telomeric DNA duplex and favor its Several telomeric DNA-binding proteins have been found
opening. to associate with telomere G-quadruplex. POT1 is known
to disrupt the telomeric G-quadruplex, allowing for
telomerase extension.179 Conversely, yeast Rap1 is believed
3. Telomere-binding proteins
to bind telomeric DNA to promote G-quadruplex
The TTAGGG repeats of human telomeres associate with formation,190 whereas human RaP1 is recruited to
the six-protein complex shelterin (Fig. 7). Three shelterin telomeres by TRF2. TRF2 has also been shown to bind and
proteins, TRF1, TRF2, and POT1, directly recognize promote the t-loops. Taken together, the shelterin
telomere DNA sequences, and are interconnected by three complex, despite consisting of only six proteins, has an
additional shelterin proteins, TIN2, TPP1, and Rap1. Each is immensely complex role in telomere regulation and protection
described below. and in the control of signaling cascades from the
Telomeric repeat-binding factors 1 and 2 (TRF1 and TRF2) chromosome ends.
directly bind the TTAGGG sequences in double-stranded In a recent study, biochemical purication of the telomeric
telomeric DNA by a C-terminal DNA-binding domain.169176 complexes showed that approximately 200 telomere-
TRF1 has acidic amino acids at its N-terminus, while associated proteins that interacted with and might inuence
TRF2 contains a Gly/Arg-rich domain as the basic domain. telomeric structure may exist in human cells.191 The great
They bind DNA as homodimers or oligomers by homotypic diversity of telomere components provides a hint about the
interactions in the TRF homology (TRFH) domain. presence of highly plastic organization of human telomeres.

2724 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

4. Functions of telomeres nuclease hydrolysis, suggesting that this superhelix structure


might be important for eectively packing telomeric DNA
Terminals of linear DNA are recognized as DNA strand into a protective capping state.
breaks, leading to activation of DNA damage signals. Similarly, The t-loop and G-quadruplex provide the cap states for the
the disruption of telomere maintenance results in chromosome telomere. However, these structures need to be exible, as the
end-to-end fusions and/or ends being recognized as DNA t-loop during DNA replication and the telomeres may adopt a
damage. The DNA damage machinery activates two independent more open state, allowing them to be extended by telomerase.
signaling pathways in cells: (i) the ATM (ataxia telangiectasia
mutated) kinase pathway and (ii) the ATR (ataxia telangiectasia
and Rad3-related) kinase pathway. In addition, double- 5. Telomere-related diseases
stranded break repair via the non-homologous end joining Several human diseases are associated with telomere
(NHEJ) or homologous recombination (HR) DNA repair shortening, such as heart disease, ulcerative colitis, liver
pathways results in inappropriate chromosomal end-to-end
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

cirrhosis, and atherosclerosis, and several premature aging


fusions. It was suggested that cells respond to dysfunctional syndromes.194197 A rst discovered disease, dyskeratosis
telomeres by undergoing senescence, apoptosis, or genomic congenita, has shorter telomeres and unstable telomerase
instability. Telomeres must perform the crucial job of because of mutations in components of the telomerase
Downloaded by Pennsylvania State University on 14 May 2012

protecting the chromosomes from DNA damage responses, complex.198200 Aplastic anemia has been reported to be
degradation, and fusion. This protection must include associated with mutations in the genes encoding aplastic
mechanisms that block the two kinase pathways at the anemia or telomere-binding protein.201,202 Recently, accelerated
telomeres and allow the telomeres to avoid both repair telomere shortening has also been found in some cases of
reactions. It is not clear how the 3 0 end overhang provides idiopathic pulmonary brosis.203 In addition, mouse models of
protection from the abnormal pathways, but the most widely telomerase deciency as well as telomeres shortening in human
accepted hypothesis is that the capping state in the chromo- disease have suggested that aging pathologies are associated
some ends serves to protect the telomere ends (Fig. 8). with Werner and Bloom syndrome and ATM syndrome,
The t-loop and G-quadruplex provide the possible cap further demonstrating that short telomeres contribute to the
states for telomere protection. Telomere loss leads to eventual pathobiology of these premature ageing diseases.204206
telomere uncapping, that is, disruption of the proper structure Telomere shortening and telomerase deciency associated
of the protective cap at the end of the telomere.1625 Telomere with these human diseases suggest that strategies of telomerase
loss leads to the disruption of the proper telomere structures, reactivation and telomere elongation might have therapeutic
that is, induction of eventual telomere uncapping at the end of potential for such premature aging syndromes.207 In a
the telomere.1625 With the perturbed structures, the linear previous study, it has been demonstrated that expression of
chromosome ends are identied by the cells DNA damage the catalytic subunit of human telomerase (hTERT) induces
surveillance machinery as DNA breaks, which can lead to telomerase activity and telomere elongation, resulting in a
many types of genome aberrations.192 greatly extended lifespan for a variety of human cell types.208
Although the t-loop provides a possible structure for Therapeutic agents that could be designed to stimulate the
telomere protection, many questions still remain to be alternative lengthening of the telomere pathway (ALT) would
answered. Binding of replication protein A (RPA) to the preferentially maintain telomere length. Several reports have
single-stranded region in the t-loop could lead to the suggested that small molecules can extend the lifespan of
activation of ATR kinase at the telomere.21 Consistently, a lymphocytes by activating telomerase, and such molecules
recent study showed that the 3 0 end overhang length, seemingly might potentially lead to treatments for these patients
sucient for t-loop formation, does not provide a protective (Fig. 9).207,209
structure for prevention of senescence in cells.193 Our recent
studies reveal that a higher-order DNA G-quadruplex
structure protects DNA double-stranded ends from being 6. Anticancer agents: chemical approaches to
recognized as double-stranded breaks and directs against target human telomeres and telomerase
Human telomerase is a cellular reverse transcriptase.8,210 It is
composed of two essential components: functional telomerase
RNA (hTR) which serve as templates for the addition of
telomeric repeats and the telomerase reverse transcriptase
catalytic subunit (hTERT).

Fig. 8 Telomeres are a lot like aglets on the ends of shoelaces that
keep them from fraying. A capping structure in the chromosome
ends serves to protect the telomere ends. An uncapping structure
results in the telomere dysfunction, which is likened to the loss of
aglets inducing the fraying. Fig. 9 Chemistry approaches provide molecule activators.

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2725
View Online

Telomerase was rst recognized as a unique and exciting telomere duplex DNA has been developed by conjugation of
anticancer target about 5 years after its discovery. Telomerase PyrroleImidazole polyamides and alkylating moieties.236
is activated in 8090% of human tumors of all cancer types,30
including cancer stem cells.211 No other tumor-associated gene 6.2 Nucleoside analogues
is as widely expressed as telomerase in cancers. This generality
Several nucleoside analogs are also reported to be promising
makes telomerase an important drug target for cancer.2960,212
agents with cell type specicity.237 These chemically synthesized
The fact that telomerase is low or undetectable in most normal
nucleosides act as chain-terminating inhibitors of reverse
cells provides tumor specicity to telomerase-based anticancer
transcriptases when they are incorporated into the DNA by
drugs, as this may reduce the undesired side eect of damaging
telomerase.238,239 Examples include azidothymidine (AZT)
normal cells. Thus, telomerase or its telomere DNA substrate
(Fig. 11b),240,241 6-thio-2 0 -deoxyguanosine 5 0 -triphosphate
presents a target with good selectivity for tumors over healthy
(TDG-TP),242 and L-enantiomers (L-dTTP and L-dGTP).243
tissue. Numerous approaches have been developed, including
20 ,30 -Dideoxyguanosine 50 -triphosphate, carbovir 50 -triphosphate,
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

the targeting of the two major components hTERT and


and D-carbocyclic-2 0 -deoxyguanosine 5 0 -triphosphate have also
hTR of telomerase, and the telomeres themselves (Fig. 10).
an inhibitory eect on telomerase.244
There are several late-stage trials in the pipeline.
Downloaded by Pennsylvania State University on 14 May 2012

6.3 Oligonucleotide derivatives


6.1 Small molecule compounds
It has been suggested that some chemical approaches have
A variety of small molecule compounds, including screened been directed to the 451-mer long human telomerase RNA
chemical libraries of reverse transcriptase inhibitors, have been (hTR), with special focus on the 11-mer template region
shown to directly interact with the hTERT component of (5 0 -CUAACCCUAAC-3 0 ). Antisense, ribozyme, or small
telomerase. One having a potent compound with good interfering RNA (siRNA) agents targeting hTR exhibit telomerase
activity, BIBR1532, led to telomerase inhibition and telomere inhibitory activity.244,245 One such oligonucleotide (GRN163L)
shortening (Fig. 11a).213215 Other catalytic inhibitors, epicatechin prevents hTR from forming an active complex with hTERT
(EGCG and MST312),216,217 isothiazolone (TMPI),218 and and has recently been entered into Phase I/II clinical
bisindole derivatives,219 are highly selective for telomerase. trials.246258 This 13-mer oligonucleotide is chemically
Nitrostyrene,220 quinoxaline,221 and rubromycin analogues222 modied through the use of a phosphorothioate (PS) linkage
have been shown to inhibit the activity of the telomerase
enzyme. A number of compounds, such as helenalin and
chrolactomycin,223,224 also showed activity against telomerase.
Other types of molecules, such as histone deacetylase (HDAC)
inhibitors,225,226 COX-2 inhibitors,227 and tyrosine kinase
inhibitors,228 have an inhibitory eect on telomeres and telomerase
as well as on a number of cellular processes.229,230 These
compounds also include telomerase-specic phosphorylation
inhibitors (bis-indolylmaleimide I and H-7),231 nonsteroidal anti-
inammatory drugs such as aspirin,232 and antioxidants such as
vitamin E.233 Thapsigargin inhibits telomerase by increasing
intracellular calcium concentrations.234
Several small compounds binding to the RNA/DNA
heteroduplex were identied to inhibit the enzyme by either Fig. 11 (a) Small molecule BIBR1532 interacts with the hTERT.
preventing strand dissociation or suciently distorting the (b) Nucleoside analogue AZT. (c) Oligonucleotide derivative
substrate.235 Recently, an approach for alkylating human GRN163L. (d) Telomerase siRNA.

Fig. 10 Telomerase or its telomere DNA substrate presents a target for cancer. Numerous approaches have been developed, including the
targeting of the two major components hTERT and hTR of telomerase, and the telomeres themselves.

2726 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

to prevent enzymatic degradation and by linking a lipid 6.5 Structure-based agents


palmitate moiety to its end to enhance cellular uptake
Folding of telomeric DNA into G-quadruplexes inhibits
(Fig. 11c).
telomerase by locking the single-stranded telomeric substrate
Chemical modication on nucleic acid-based drugs has
into an inactive conformation that is no longer recognized, nor
provided several agents with potential anticancer activity.
elongated, by the enzyme.290 Telomerase has been shown to
These modied molecules bind RNA sequences with improved
function in telomere protection as well as in telomere
selectivity, enhanced ecacy, and improved pharmacological
elongation.291,292 The inability of telomerase to access the
properties.
telomere is able to activate telomere length-independent
2 0 -O-methyl (2 0 -O-Me) and 2 0 -O-(2-methoxyethyl) (20 -MOE)
damage signal causing immediate cell arrest or death.
modied RNAs can eciently and sequence-selectively inhibit
G-quadruplex formation also blocks the single-stranded binding
telomerase, causing telomeres to shorten and cell proliferation
protein hPOT1 to telomere,293,294 leading to a disruption
to decrease.259,260 2 0 ,5 0 -Oligoadenylate oligonucleotides
between telomere and shelterin complex. Recently, it was also
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

directed against the template region of hTR also eciently


suggested that G-quadruplexes can cause the deletion of
inhibit telomerase.261,262 An oligonucleotide, 20 ,5 0 -oligoadenylate
telomere lagging strand, resulting in a dominant factor in
(2-5A-anti-hTR), inhibits human telomerase by specically
the telomere shortening.295,296 Complete replication of
and eectively degrading the targeted RNA.263265 Combining
telomeric ends requires unwinding higher-order structures of
Downloaded by Pennsylvania State University on 14 May 2012

2-5A-anti-hTR with adenoviral transfer of the p53 gene has a


G-rich telomeric DNA. RecQ, BLM, and WRN helicases also
signicantly greater antitumor eect for malignant gliomas.266
cooperate with POT1 in the absence of G-quadruplex ligands
Other chemically modied derivatives, including 2 0 -deoxy-,
to unwind telomere structures of G-quadruplexes.297 In such a
hydroxy-, methoxy-, and uoro-N3 0 P5 0 phosphoramidates,
normal condition, G-quadruplexes function as capping
have been tested as potential therapeutic agents against
structures. The G-quadruplex stabilization by ligands inhibits
telomerase in vitro.267
the unwinding of G-quadruplex sequences. It was suggested
In addition to oligonucleotides directed against the TERT
that deletions could result from failure to resolve higher-order
mRNA, a ribozyme targeting downstream from the 5 0 end of
structures of G-rich DNA such as G-quadruplex.78 Dierential
hTERT mRNA exhibits telomerase inhibitory activity.268
activation of the DNA damage machinery should be involved
Ribozyme cleavage of the RNA component of human
in the deletion mechanism in the presence of G-quadruplex
telomerase also inhibits telomerase activity.269271 A study
ligands.
showed that telomerase siRNAs targeted against two
G-quadruplex structures represent a new class of molecular
regions of the human telomerases cause ecient inhibition of
targets for DNA-interactive compounds that may be useful to
telomerase expression (Fig. 11d). Telomerase activity in
target telomeres.298300 A large number of ligands reportedly
cells transfected with siRNAs was reduced 77  3% after
interact with G-quadruplexes and act as inhibitors of telomerase
1 day.272
activity. NMR and X-ray studies were performed to obtain
Peptide nucleic acids (PNAs) are modied analogs of DNA
detailed information on the interaction of G-quadruplex DNA
or RNA that possess a neutral amide backbone and bind to
with small molecules.
complementary nucleic acids with very high anity. Introduction
of complementary PNAs to telomerase RNA eectively 6.6 Pentacyclic acridinium (RHPS4)
inhibited telomerase activity, shortened telomeres, and
arrested cancer cell proliferation.273277 A G-quartet end-stacking binding mode was observed in the
NMR structure of a complex between a pentacyclic acridinium
(RHPS4) ligand and the parallel human telomere d(TTAGGGT)4
6.4 Peptides and proteins G-quadruplex (Fig. 12a and b).301,302 The p-system of the drug
Various chemically synthesized peptides as telomerase-based overlaps primarily with two bases of each G-tetrad by stacking
drugs were reported. Telomerase is over-expressed in nearly all interactions with the G-tetrads. Although several exceptions to
cancers, making it an immune target. Multiple approaches to this structural feature are known, the requirements shown by
active telomerase immunotherapy have led to many products most such ligands seem to be a planar aromatic chromophore
in clinical development.278281 These products involve use of and substituents with terminal basic groups. Several studies
synthetic TERT peptides or genetic components to prime suggest that the ligand inhibits telomerase and induces
antigen-presenting cells. The most advanced products, telomere uncapping and damage.302311
GV1001 and GRNVAC1, were designed to stimulate immune
6.7 Acridine ligand (BRACO-19)
responses to cancer cells. GV1001, a synthetic peptide containing
the 16 amino-acid residues of hTERT protein, is currently in Neidle et al. reported a crystal structure of a complex between
phase II and III trials in non-small cell lung cancer and a bimolecular human telomeric G-quadruplex of sequence
pancreatic cancer,282 and GRNVAC1 is in a phase II trial.283 d(TAGGGTTAGGGT) and a 3,6,9-trisubstituted acridine
Other approaches of telomerase inhibition include that ligand (BRACO-19) (Fig. 13a and b).312 BRACO-19 is a drug
telomerase genes downregulate at the promoter level by the molecule sandwiched between two bimolecular quadruplexes
use of a dominant negative hTERT gene delivery,284 active to form a biological unit, such that the G-quadruplexes are
telomerase assembly inhibits by interfering with the molecular uniquely stacked 5 0 3 0 . A number of such trisubstituted
chaperones p23 and Hsp90,285287 and telomerase hTR gene acridine analogs, with a variety of side-chain modications
modiers by expressing mutant telomerases.288,289 and stereoisomer variations, have exhibited strong

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2727
View Online

Fig. 12 (a) NMR structure of the complex of RHPS4 (in green)


bound to the tetramolecular G-quadruplex. (b) RHPS4.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

Fig. 14 (a) NMR structure of the complex of MMQ1 (in green)


bound to the (d[T2AG3T])4 tetramolecular G-quadruplex. (b) MMQ1.
Downloaded by Pennsylvania State University on 14 May 2012

(c) Cyclo-bis-quinacridine BOQ1.

in the dimeric structure and the steric hindrance of the


acrocyclic scaold that impedes duplex binding.

6.9 Naphthalene diimide (ND)


A series of tri- and tetrasubstituted naphthalene diimides (ND)
are exceptional G-quadruplex binding agents (Fig. 15), with
melting temperature (Tm) values for the human intramolecular
G-quadruplex that are at least two-fold greater than those of
the acridine compound BRACO19, consistent with telomerase
inhibition.333,334 Both crystalline structures suggested that the
Fig. 13 (a) X-Ray structure of the complex of BRACO-19 inter- and intramolecular G-quadruplex structures formed with
(in green) bound to the bimolecular human telomere G-quadruplex. a naphthalene diimide ligand exhibited ligand-G-quadruplex
(b) BRACO-19. (c) Disubstituted triazole-linked compounds. end stacking (Fig. 15a and b).335 This arrangement of ligands
and human telomeric G-quadruplexes results in a double
G-quadruplex binding with accompanying telomerase inhibitory sandwich, in which there are two ligand molecules stacked on
activity.313325 Recently, a series of disubstituted triazole- each other to form the boundary between the G-quadruplexes.
linked acridine compounds was shown to inhibit the activity Ligands are also bound to the external TTA loop by stacking-
of the telomerase enzyme that was found to have selectivity for type interactions. Based on the G-quadruplex recognizing
human telomeric G-quadruplexes (Fig. 13c).326 Both properties of the ND moiety, G-quadruplex ligand/alkylating
lead compounds also have selective inhibitory eects on the dual-functioned molecules were developed by tethering alkylating
proliferation of cancer cell lines rather than on normal agents to the ND core (Fig. 15c).336 It is showed that the
cell lines. alkylation process occurred preferentially on the G-quadruplex
structure in comparison to other DNA conformations.
6.8 Quinacridine analogue (MMQ1)
Teulade-Fichou, Mergny and coworkers reported that
quinacridine analogues have the ability to stabilize
G-quadruplexes.327330 An NMR structure reveals that the
dipropylamino analog MMQ1, a crescent-shaped molecule, is
likely to maximize the overlap with the guanines of the
accessible G-quartet (Fig. 14a and b).331 The NOE cross-peaks
of NMR suggest that the ligand stacks over G-quartets, with
one molecule located at the 5 0 end and the other at the 3 0 end.
In addition to the p-stacking interactions between the ligand
and the G-quartet, the cationic side chains are clearly directed
toward the negatively charged G-quadruplex. A cyclo-
bis-quinacridine BOQ1 with two polyammonium linker
bridges binds signicantly more strongly to G-quadruplexes Fig. 15 (a) X-Ray structure of the complex of ND (in green) bound
than to the monomer compounds (Fig. 14c).332 This selectivity to the d[TAGGG(TTAGGG)3] intramolecular G-quadruplex. (b) ND.
is attributed to the enhancement of the ligand aromatic surface (c) QM5, a G-quadruplex ligand/alkylating dual-functioned molecule.

2728 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

Fig. 16 (a) X-Ray crystal structure of TMPyP4 (in green) bound to


Downloaded by Pennsylvania State University on 14 May 2012

the bimolecular human telomere G-quadruplex. (b) TMPyP4.


(c) Se2SAP. (d) Mn(III)-coordinated porphyrin.

Fig. 17 (a)(f) Telomestatin and the related compounds.


6.10 Porphyrin analogue (TMPyP4)
(g) Cyclic oligoamide. (h) Cyclic oxazole-based tripeptide.
Porphyrin analogs are well established binding agents for
G-quadruplexes. Hurley et al. have extensively studied the
L1H1-7OTD (more planar analogue),365 and 6OTD
tetra-N-methyl-4-pyridyl porphyrin (TMPyP4), demonstrating
(dimerisation)366 were shown to interact strongly with
that the ligand has a high anity for G-quadruplex and
G-quadruplex DNA (Fig. 17df), with a fair quadruplex-
eciently inhibits telomerase (Fig. 16).337345 An X-ray crystal
over-duplex DNA selectivity. Other series of macrocyclic
structure has been imaged of TMPyP4 bound to the bimolecular
molecules, cyclic oligoamide,367 cyclic oxazole-based
human telomere G-quadruplex d(TAGGGTTAGGG)
tripeptide (Fig. 17g and h),368,369 and cyclo[n]pyrroles370 have
(Fig. 16a and b).346 An alternative binding mode indicated
been investigated as G-quadruplex binding ligands.
that porphyrin molecules bind by stacking on the TTA
Several metal-coordinated ligands have been reported as
nucleotides but without any direct contact with the G-quartets.
potent telomerase inhibitors. The cationic or highly polarized
The nucleotides in base pairs are involved in the complex,
nature of these ligands is believed to have a major contribution
either as part of the external loop structure or at the 5 0 region
on the interaction with G-quadruplex DNA. The functionalized
of the stacked G-quadruplex. To enhance the binding selectivity
nickel(II)salphen complexes are excellent G-quadruplex DNA
of G-quadruplex, several porphyrin-like ligands such as
stabilizers (Fig. 18a).371 The Ni2+ ion is possibly playing an
diselenosapphyrin Se2SAP (Fig. 16c),347 Mn(III)-coordinated
important role for the stability, replacing one of the metal ions
porphyrin (Fig. 16d),348 and tetrakis-(diisopropylguanidinio)
in the channel of the G-quadruplex. A series of platinum(II)
zinc phthalocyanine Zn-DIGP349 have been tested for their
complexes containing dipyridophenazine (dppz) have been
ability to interact with the human telomeric G-quadruplex.
shown to inhibit telomerase activity (Fig. 18b).372 The cationic
These ligands were shown to bind strongly and selectively to
Pt(II) complexes containing an extended p-surface are stacked
G-quadruplexes. Mn(III)-porphyrin targets the human
on the ends of the G-quadruplex. The large p-aromatic
telomere G-quadruplex by four orders of magnitude over
surfaces and positive charges near the center of these mole-
duplex DNA.
cules increase anity to G-quadruplex. A self-assembled
platinum molecular square [Pt(en)(4,4 0 -dipyridyl)]4 ligand that
6.11 Other interactive ligands
is potentially compatible with these features is an ecient
A natural product telomestatin targets G-quadruplexes with G-quadruplex binder and telomerase inhibitor (Fig. 18c).373
high specicity. Telomestatin is one of the most exciting A family of bisquinolinium-substituted derivatives containing
G-quadruplex ligands extensively studied (Fig. 17a),350359 triazines- or pyridine-based core was developed to inhibit
although there are no structural characterizations of the human telomerase activity at low doses (Fig. 19a).374 Furthermore,
binding mode (NMR, X-ray crystallography). It is likely that replacement of the central cores by a phenanthroline unit
a perfect shape adaptation between the macrocycle molecule showed to enhance the binding selectivity for intramolecular
and a G-quartet and the hydrophobic forces contribute human telomere G-quadruplexes (Fig. 19b).375
to stacking interactions. Its outstanding selectivity for Recently, Balasubramanian and coworkers developed an
G-quadruplex and highly promising biological activity encourage engineered zinc nger protein that binds with high specicity
chemists to synthesize a novel class of chemically challenging to the intramolecular G-quadruplex formed by the human
telomestatin-like compounds. HXDV354360362 and HXDL363 telomeric sequence, and that inhibits the activity of the enzyme
for example have been found to greatly stabilize the telomerase in vitro.376 A study from the same laboratory
G-quadruplex structure (Fig. 17b and c). S2A2-6OTD,364 reported that a polyamine-conjugated anthracene ligand

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2729
View Online

Fig. 18 (a) Nickel(II)salphen ligands. (b) Platinum(II)-dppz.


(c) [Pt(en)(4,4 0 -dipyridyl)]4 ligand.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A
Downloaded by Pennsylvania State University on 14 May 2012

Fig. 19 (a) Bisquinolinium-substituted triazine 12459.


(b) Phenanthroline-connected bisquinoline. (c) Polyamine-conjugated
anthracene. (d) Chiral cyclic helicene M1.

targeted the central cation channel of the human telomeric Fig. 20 (a) PIPER and peryleneEDTAFe(II) ligand. (b) Human
G-quadruplex, a specic structural element in the structure telomere sequence specic cleavage by G-quadruplex-forming
(Fig. 19c).377 More recently, a chiral cyclic helicene was DNAEDTP complex. The structures of EDTP and BrG are shown.
(c) Representation of photo-cross-linking of human telomeric DNA
suggested to be sandwiched within a chiral-cleft site that was
by a photocontrolled 6-mer oligonucleotide. Structure of a 6-mer
selectively formed by two human telomere G-quadruplexes,
oligonucleotide with a photo-cross-linking reagent, psoralen, at the
exhibiting potent inhibitory activity against telomerase 5 0 -end.
(Fig. 19d).378
syn-preferring 8-bromoguanosine (BrG) in the DNAEDTP
6.12 Several novel methods probe were performed to form a highly stable G-quadruplex
structure between the probe and target sequence.133,136
The perylene diimide, N,N 0 -bis[2-(1-piperidino)-ethyl]3,4,9,10- Based on the same strategy, a small photocontrolled 6-mer
perylenetetracarboxylic diimide (PIPER), has been reported to oligonucleotide was employed to eectively inhibit telomerase
be selective for human telomere G-quadruplex structures.379 (Fig. 20c).382 Psoralen was selected as a photo-cross-linking
A peryleneEDTAFe(II) ligand as a DNA cleaving unit was reagent because of its reaction selectivity with pyrimidine
bound to the perylene core of PIPER (Fig. 20a). The conjugate bases. An oligonucleotide bearing psoralen produces a
was shown to selectively cleave G-quadruplex DNA in the photoadduct with the telomere target upon irradiation by
presence of dithiothreitol (DTT).380 G-quadruplex formation. Doses of both psoralen oligonucleotide
A structure based approach was developed to sequence- and light irradiation trigger death of cancer cells. Because the
specic cleaving of human telomeric DNA by G-quadruplex main eect on the target is controllable by switching the
formation (Fig. 20b).381 As mentioned above, a dimeric light on/o, the small, photocontrolled oligonucleotide can
G-quadruplex can be formed by the three-repeat (16mer) minimize side eects by irradiation of only cancer cells, which
and single-repeat (6-mer) human telomeric sequences. This alleviates the main problem with present-day anticancer
dimeric G-quadruplex structure suggests a way of how a agents.
segment of three G-tracts could bind to a remote G-tract. A
N,N,N 0 ,N 0 -ethylenediamine-tetramethylene-phosphonic acid
(EDTP) as the metal binding group was conjugated to the
7. Telomere RNA: a newly emerging player
16-mer oligonucleotide. The oligonucleotide binds to the
target of human telomere DNA by G-quadruplex formation For a long time, telomeres have been considered transcriptionally
and causes a sequence-specic strand break. Formation of a silent, and that their DNA was not transcribed into strands of
stable G-quadruplex between the DNAEDTP probe and the RNA. A recent nding demonstrated that telomere DNA is
target sequence is a key step in the cleavage reaction. For this transcribed into telomeric repeat-containing RNA (referred to
purpose, substitutions of dG in the syn conformation with a as TERRA) in mammalian cells.383,384 TERRA molecules

2730 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

were detected in dierent human and rodent cell lines and RNA sequence r(GGGUUAGGGU) in K+ solution, which
contained mainly UUAGGG repeats of heterogeneous length. revealed that two parallel G-quadruplex units assemble via
These ndings raise the crucial question of how TERRA RNA 5 0 5 0 stacking (Fig. 21d).390 This also explains why a 23-mer
is specically associated with chromosome ends. The existence telomeric RNA sequence in K+ solution gives unresolved
of TERRA RNA may reveal a new level of regulation and NMR spectra in the imino proton region. Recently, electro-
protection of chromosome ends that could facilitate valuable spray ionization mass spectrometry (ESI-MS) shows that the
insight into fundamental biological processes such as cancer dimers in the longer sequences are stabilized by stacking of
and aging.385387 Revealing the structure and function of two G-quadruplex subunits (Fig. 21e).393 These observations
telomere RNA will be essential for understanding telomere are consistent with the models for arrangements of
biology and telomere-related diseases. G-quadruplexes in long human telomeric RNA sequences,
such as a model of alternate-direction stacking of
G-quadruplex blocks (Fig. 21f).111,390,394
7.1 Telomere RNA structure
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

It has been shown by CD, NMR, MS, and X-ray crystallo-


To dene the structural features of human telomere RNA graphy that G-rich telomeric RNA sequences are able to form
sequence, we examined the conformation of the 12-mer human parallel G-quadruplex structures in Na+ solution as well as in
telomere RNA sequence r(UAGGGUUAGGGU) in the K+ conditions. However, whether TERRA RNA G-quadruplexes
Downloaded by Pennsylvania State University on 14 May 2012

presence of Na+. Multi-method approaches including exist in living cells is unknown. Very recently, we developed a
circular dichroism (CD), NMR, and gel electrophoresis have chemical method to investigate the structural features of
demonstrated that the telomere RNA forms a parallel dimeric human TERRA RNA in living cells.395 Pyrene was designed
G-quadruplex with external loops similar to the crystalline and as a light-switching probe. The advantage of the distance
NMR structure of human telomeric DNA (Fig. 21a).388,389 dependence of excimer formation with pyrene can be used as
The dimeric RNA G-quadruplex formation was directly a unique excimer signaling device for detecting G-quadruplex
observed by MALDI-TOFMS, suggesting that the RNA structures (Fig. 21g). When the pyrene-labeled probe is free in
G-quadruplex remains stable even in the gas phase. A human solution without G-quadruplex formation, the pyrene
24-mer telomere RNA sequence r(UUAGGG)4 was also molecules are spatially separated and only the monomer
suggested by CD spectroscopy to form a parallel G-quadruplex emission peaks are observed. Formation of G-quadruplex
(Fig. 21b).389 The telomere RNA G-quadruplex was found to brings the pyrene molecules at the 5 0 and/or 3 0 ends into close
induce a strong RNase resistance for UUAGGG repeats proximity, allowing the formation of an excimer. The excimer
telomere RNA. This nding agrees with the observation possesses broad red-shifted emission, in contrast with the
that repeat RNA exists in cells at higher levels and enriches pyrene monomer. The change in emission color serves as a
810-fold in puried polyadenylated RNA compared to total way to rapidly probe the G-quadruplex structure, and the
RNA. The telomere RNA G-quadruplex that resists telomere excimer uorescence intensity can be used for sensitive
RNA degradation may oer a concentration supply of telomere monitoring of G-quadruplex formation. An excimer uorescence
RNA to participate in essential biological processes. of pyrene (green) is observed in living cells, providing in vivo
An NMR-based solution structure suggested a same evidence of the presence of the G-quadruplex in human
folding topology for the 12-mer human telomeric RNA TERRA RNA (Fig. 20h). Co-localized images between probe
r(UAGGGUUAGGGU) sequence in K+ solution and nuclear DNA clearly show that the TERRA RNA
390
(Fig. 21a). Terminal U bases stack on the top and the G-quadruplex was restricted to cell nuclei. Furthermore,
bottom of the G-tetrad core, respectively. The K+-stabilized TERRA RNA G-quadruplex foci colocalized with TRF2 foci,
crystal structure of r(UAGGGUUAGGGU) has been indicating that the TERRA RNA G-quadruplex is a common
determined to be a parallel G-quadruplex,391 suggesting that structural component of mammalian telomeres. Importantly,
2 0 -OH hydroxyl groups in the RNA quadruplex play a the TERRA RNA G-quadruplex was also detected at the ends
signicant role in redening the hydration structure in the of metaphase chromosomes from cells consistent with
grooves and the hydrogen bonding networks. previous observations that accumulation of TERRA RNA is
More recently, we reported the structural features of human not only in nuclei but also at telomeres.
telomere RNA r(UAGGGU) in the presence of K+ and Na+.
It has been demonstrated that a novel U-tetrad is formed at
7.2 Discovery of a telomere DNARNA G-quadruplex by
the 3 0 end of a parallel human telomeric RNA G-quadruplex
click chemistry
(Fig. 21c).392 The U-tetrad dramatically stabilizes the human
telomeric RNA G-quadruplex structure, leading to an increase This discovery of telomere RNA raises the crucial question of
in Tm of 29 1C. We noted that four uridine residues located at how telomeric RNA is specically associated with telomeric
the end of the G-quadruple are more favorable for U-tetrad DNA in terms of chromosome-end regulation and protection.
formation than the dimer telomere RNA G-quadruplex, in A possible association between telomere RNA and telomere
which only two uridine residues are positioned at the ends. The DNA may be through the formation of an intermolecular
U-tetrad-stabilized telomeric RNA G-quadruplex structure DNARNA G-quadruplex. It is technically dicult to study the
adds considerably to our understanding of the diversity of DNARNA hybrid G-quadruplex structure by traditional
RNA G-quadruplex architectures. methods, such as NMR spectroscopy and X-ray crystallography,
Telomeric RNA sequences seem to be structural since the DNA G-quadruplex, RNA G-quadruplex, and
polymorphisms. This concurs with the NMR study of a 10-mer DNARNA hybrid G-quadruplex may coexist as a mixture

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2731
View Online
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A
Downloaded by Pennsylvania State University on 14 May 2012

Fig. 21 (a)(f) Structures of TERRA RNA G-quadruplexes. (g) Schematic for detection of RNA G-quadruplex formation in living cells using a
dual-pyrene probe. (h) Green signals correspond to the probe (green). Cell nuclei were stained with SYTO 25 dye (red). Costaining using antibodies
anti-human TRF2 (in red). Chromosome DNA was stained by propidium iodide (red). (i) Schematic depiction of the detection of the DNARNA
G-quadruplex. The use of 5 0 -azido-labeled DNA and 5 0 -alkyne-labeled RNA may result in a mixture of three types of G-quadruplexes. Only the
DNARNA G-quadruplex brings the alkyne and the azido group into close proximity to give the product of an azidealkyne cycloaddition. (j) and
(k) Higher-order telomeric DNA G-quadruplex, telomeric DNARNA G-quadruplex, TERRA RNA G-quadruplex, and/or t-loop may be
formed at the ends of chromosomes to provide a protective eect.

2732 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

in solution. Click chemistry has helped overcome this diculty Recent studies indicated that siRNA-mediated depletion
(Fig. 21i).396 The click reaction can trap a particular species or of telomeric RNA caused an increase in telomere dys-
produce a snapshot of the various inter-converting structures function-induced foci and aberrations in metaphase
that are present in a complex solution. The detected telomeres.401,402 Conversely, increased levels of telomeric
azido-alkyne cycloaddition product between the 5 0 -alkyne- RNA observed to be associated with thermal shock, may help
labeled RNA and the 50 -azido DNA identies that a DNARNA protect telomeres against stress-induced damage.384 Recently,
hybrid G-quadruplex structure can be formed from human some proteins have been suggested to be associated with
telomeric DNA and RNA sequences.396 telomeric RNA,403 such as TRF2, which was found to recruit
telomeric RNA to telomeric DNA.401
7.3 Telomere RNA function
DNARNA hybrid G-quadruplex structures could inhibit 8. Conclusions and future prospects
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

telomerase activity by locking telomeric substrate DNA into


8.1 Structure and function
an inactive conformation that is no longer recognized or
elongated by the telomerase, consistent with the nding that Telomere biology has come a long way in the past 70 years,
telomerase activity is dramatically reduced in the presence from the observation that chromosomes require protection, to
Downloaded by Pennsylvania State University on 14 May 2012

of increased concentrations of telomere RNA in vitro. the award of the 2009 Nobel Prize for the discovery of
Alternatively, through the use of in vitro reconstituted telomerase and the eects of telomere shortening on cells.
telomerase and synthetic TERRA molecules, TERRA RNA Functional telomeres have been experimentally implicated in a
has been shown to directly pair with the template region of number of molecular cell processes. Molecular components of
hTR as a telomerase ligand and natural direct inhibitor of telomeres, including terminal double- and single-stranded
human telomerase.397 Lingner et al. proposed three telomeric DNA, protein complex shelterin, and telomerase
possible modes for telomerase sequestration by TERRA. form physical states associated with functional capping and
The released TERRA from the telomere may bind and uncapping.
inhibit telomere-proximal telomerase molecules. Telomere- Telomeric DNA itself has been suggested to form a t-loop
bound TERRA may also bind and sequester telomerase and and/or G-quadruplex as a possible cap state. However, key
prevent its access to the telomere. Alternatively, TERRA questions include whether such structures occur in living cells
may bind to telomeric chromatin bound telomerase and and, furthermore, what their biological function might be. To
prevent it from accessing the telomere. Genetic experiments date, whether the two structures exist in living cells is
in Saccharomyces cerevisiae provided evidence that TERRA unknown. Chemistry is expected to oer a powerful approach
regulates telomerase in vivo. TERRA RNA acts as a potent to answer this question.
competitive inhibitor for telomeric DNA in addition to the Like t-loops, G-quadruplexes shelter the 3 0 end of the
intermolecular G-quadruplex mode. G-overhang and, although this provides a capping function,
Formation of an intermolecular G-quadruplex by telomeric such structures would need to be resolved by telomere
RNA and DNA provides a protective eect. Human telomeric synthesis during replication and telomerase extension. The
RNA inhibits chromosome end fusions and has an eect on two capping structures need not be mutually exclusive, and
the inhibition of cellular senescence when TRF2 is inhibited by could represent dierent functional states. Capping structures
inducing a dominant negative allele of TRF2 (TRF2DBD) thus may be dynamic in response to cell cycle changes.
(Xu, Y., Ito, K., Katada, H., K, Komiyama, M., submitted What is the molecular basis of the telomeric cap and how do
for publication). Human telomeric RNA causes a signicant t-loops and G-quadruplexes contribute to end protection? It is
increase in the clonogenic capacity of cells after exposure of an not clear what the primary signal is that detects uncapped
oligonucleotide homologous to the telomere 3 0 -single stranded telomeres, nor is it known how the signal gets translated and
overhang (T-oligo), which activates DNA damage signals amplied throughout the cell.
and induces cell senescence.398,399 Telomeric DNARNA What is the role of the many shelterin accessory factors?
G-quadruplexes should avoid the exposure of homologous Although our understanding of the individual roles of shelter-
single-stranded terminal DNA because it blocks the access of a in components has improved greatly, much remains to be
key DNA damage regulator(s) to the single stranded DNA. discovered about the transcriptional, translational, and post-
Consistent with higher-order telomeric DNA G-quadruplex translational regulation of these components. Telomeric over-
structures, the telomeric DNARNA G-quadruplex and hang DNA has been suggested to form the higher-order DNA
TERRA RNA G-quadruplexes may be formed at the terminus structures containing consecutive G-quadruplexes. Whether
of this superhelix structure to provide a protective shelterin components and other proteins bind to such
eect (Fig. 21j and k). The consecutive formation of RNA superhelix structures remains to be elucidated.
G-quadruplexes by long TERRA RNA is consistent with a In addition, mechanisms of telomere replication remain
very recent result that TERRA molecules have an poorly dened. It has been suggested that telomere replication
approximate length of 200 bases.400 In contrast, the lack of could be regulated by the higher order structures of telomeres.
an uncapping structure at the 3 0 -termini of chromosome It is necessary to remove the t-loop and/or G-quadruple
ends leads to exposure of the single-stranded terminal DNA structures that form on the telomere strands, suggesting a
and is recognized by a key DNA damage regulator(s) (ATM), stalling of replication forks at telomeres. Some proteins, such
which activates various DNA damage signals. as RecQ, BLM, WRN and POT1, could be required to resolve

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2733
View Online

G-quadruples.297 The passage of the replication fork through There is a clear need to revisit the structural and functional
the telomeric repeats may restart to avoid loss of telomeric mechanisms of telomeres accompanying telomere RNA
DNA, allowing telomerase and perhaps other enzymes access participation. Telomere RNA may be related to telomere
to telomeres. DNA by formation of an intermolecular G-quadruplex,
providing a protective eect. Alternatively, dimerization may
8.2 Drug design occur on the telomeric RNA G-quadruplex and DNA
G-quadruplex, similar to the dimeric structure formed by
Functional telomeres have been implicated in cancer
both telomere DNA G-quadruplex and telomere RNA
progression and aging-related diseases. Although telomere- or
G-quadruplex units. Stabilization of such structures by small
telomerase-based inhibitors work well in vitro and in tissue
molecules can lead to telomerase inhibition. This would
culture, they have yet to pass clinical trials. For successful
transform telomere RNA into a potential target for anti-
cancer treatment, several obstacles still have to be overcome.
cancer agents directed against telomeres.
First, for agents targeting telomere maintenance, a telomerase-
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

Telomere RNA, a new player to provide another possible


independent, recombination-based mechanism (ALT) is
answer, may be appropriate to solve the old problem of the
already known to exist in cells.404 Telomeres are typically long
end replication problem. In normal cells, telomere RNA may
and heterogeneous in size in ALT cells. Some studies have
act as a template for telomere elongation synthesis by reverse
investigated potential targets to block recombination-based
Downloaded by Pennsylvania State University on 14 May 2012

transcriptase. Support for this hypothesis comes from the


telomere maintenance.405 In future, a combination of
nding that telomere RNAs are signicantly downregulated
ALT-based drugs and telomerase therapies might be eective.
in various types of human cancers compared to normal tissues.
Second, many gaps might remain in our understanding of
In normal cells, the reverse transcription pathway may be
the complexities of telomere regulation and protection
involved in the maintenance of telomere length, whereas
specicity in telomere-associated DNA repair pathways induced
telomerase is the important enzyme responsible for telomere
by drug treatments. It is clear that DNA repair systems
length maintenance in cancer cells. As indicated by Blasco
eectively counteract the DNA damage in intrachromosomal
et al., the lower telomere RNA production associated with
regions. The number of DNA damage events in a single
carcinogenesis may favor telomerase-mediated telomere
human cell range from 104106 per day, requiring
elongation in cancer cells.384
approximately 10161018 repair events per day in an adult
The recently identied TERRA RNAs that have been
human (1012 cells).406 It is not surprising that strong DNA
shown to interact with TRF2, and the transcription of
repair systems that are further enhanced by participation of
TERRA RNA is bound by a complex set of proteins.403
the telomere-associated proteins can successfully overcome
Whether these proteins or other factors interact with the
DNA damage-induced telomere dysfunction. Chemistry
telomeric RNA G-quadruplex and the DNARNA
should make major contributions in the elucidation of the
G-quadruplex to produce the protective eects remains to be
mechanisms of damage or repair recognition and provides a
discovered. Although TERRA RNA was suggested to not be
combinational approach to both the complex systems and the
translated into proteins, the possibility of its rapid transit
telomere.
through the cytoplasm cannot be excluded. If TERRA RNA
Finally, the structure-based design of telomerase inhibitors
may be translated, the (Arg-Val)n and (Gly-Leu)n poly-
necessitates molecular understanding of the range of
peptides would possibly be generated. Interestingly, the two
G-quadruplex topologies. The conformational diversity of
polypeptides contain similar motifs of G-quadruplex RNA
human telomeric G-quadruplexes introduces design diculties
and RNA-binding domains, arginine, and glycine-rich region
in the specic ligands. A recent example showed a multiple
(RGG box).409,410
target molecule from the higher-order DNA structures
Another possibility is that the process of transcription is
containing consecutive G-quadruplexes. Although any novel
more important than the TERRA RNA itself, similar to
cancer therapeutic always makes arguments, the pre-clinical
several non-coding RNA in that not the RNA itself but the
and emerging clinical experimental evidence for telomerase or
transcription process plays a key role.411 In addition, telomere
telomeres as a target for cancer therapy is encouraging, and
DNA, TERRA RNA, and related-proteins may form TDRP
the targeting of telomere maintenance mechanisms continues
bodies (telomeric DNA, RNA and proteins referred to as
to be an exciting prospect in future cancer strategies.
TDRP), similar to P bodies in mRNA surveillance and mRNA
In contrast, the fact that telomere shortening and telomerase
decay, RNA-mediated silencing and translational control.412
deciency are associated with these human diseases suggested
. . .to understand an end is to understand a beginning of all
that telomerase reactivation and telomere elongation could be
things. . . chemistry ought to make a beginning. . .
an eective way to treat these human disorders. Small circles
of telomeric DNA are being used as templates for rolling-
circle mechanisms of telomerase-independent telomere
elongation.407,408 Chemical approaches can be used to Acknowledgements
engineer articial telomeres on human chromosomes.
This work was partially supported by a Grant-in-Aid for
Scientic Research (B) from the Ministry of Education,
8.3 Telomere RNA
Science, Sports, Culture, and Technology, Japan (22350071),
The nding of telomere RNA molecules opens new doors to and by the Global Centers of Excellence Program for
better understanding of the essential biological role of telomeres. Chemistry Innovation.

2734 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

References 49 L. H. Hurley, Nat. Rev. Cancer, 2002, 2, 188.


50 E. M. Rezler, D. J. Bearss and L. H. Hurley, Curr. Opin.
1 H. J. Muller, Collect. Net, 1938, 13, 181. Pharmacol., 2002, 2, 415.
2 B. McClintock, Proc. Natl. Acad. Sci. U. S. A., 1939, 25, 405. 51 D. Cairns, R. J. Anderson, P. J. Perry and T. C. Jenkins,
3 B. McClintock, Genetics, 1941, 26, 282. Curr. Pharm. Des., 2002, 8, 2491.
4 A. M. Olovnikov, Dokl. Akad. Nauk SSSR, 1971, 201, 1496. 52 S. Huard and C. Autexier, Curr. Med. Chem.: Anti-Cancer
5 L. Hayick and P. S Moorhead, Exp. Cell Res., 1961, 25, 585. Agents, 2002, 2, 577.
6 L. Hayick, Exp. Cell Res., 1961, 37, 614. 53 E. M. Rezler and D. J. Bearss, Annu. Rev. Pharmacol. Toxicol.,
7 A. M. Olovnikov, J. Theor. Biol., 1973, 41, 181. 2003, 43, 359.
8 C. W. Greider and E. H. Blackburn, Cell (Cambridge, Mass.), 54 G. Saretzki, Cancer Lett. (Shannon, Irel.), 2003, 194, 209.
1985, 43, 405. 55 J. F. Riou, Curr. Med. Chem.: Anti-Cancer Agents, 2004, 4, 439.
9 M. A. Dunham, A. A. Neumann, C. L. Fasching and 56 J. W. Shay and W. E. Wright, Cancer Cell, 2005, 7, 1.
R. R. Reddel, Nat. Genet., 2000, 26, 447. 57 F. Pendino, I. Tarkanyi, C. Dudognon, J. Hillion, M. Lanotte,
10 F. dAdda di Fagagna, P. M. Reaper, L. Clay-Farrace, H. Fiegler, J. Aradi and E. Segal-Bendirdjian, Curr. Cancer Drug Targets,
P. Carr, T. Von Zglinicki, G. Saretzki, N. P. Carter and 2006, 6, 147.
S. P. Jackson, Nature, 2003, 426, 194.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

58 J. W. Shay and W. E. Wright, Nat. Rev. Drug Discovery, 2006, 5,


11 H. Takai, A. Smogorzewska and T. de Lange, Curr. Biol., 2003, 577.
13, 1549. 59 A. De Cian, L. Lacroix, C. Douarre, N. Temime-Smaali,
12 K. Riha, M. L. Heacock and D. E. Shippen, Annu. Rev. Genet., C. Trentesaux, J. F. Riou and J. L. Mergny, Biochimie, 2008,
2006, 40, 237. 90, 131.
Downloaded by Pennsylvania State University on 14 May 2012

13 D. Churikov, C. Wei and C. M. Price, Mol. Cell. Biol., 2006, 26, 60 C. B. Harley, Nat. Rev. Cancer, 2008, 8, 167.
6971. 61 T. Vulliamy, A. Marrone, F. Goldman, A. Dearlove, M. Bessler,
14 T. G. B. Celli and T. de Lange, Nat. Cell Biol., 2005, 7, 712. P. J. Mason and I. Dokal, Nature, 2001, 413, 432.
15 C. M. Azzalin and J. Lingner, Nature, 2007, 448, 1001. 62 J. M. Wong and K. Collins, Lancet, 2003, 362, 983.
16 E. H. Blackburn, Nature, 2001, 408, 53. 63 M. A. Blasco, Nat. Rev. Genet., 2005, 6, 611.
17 E. H. Blackburn, Cell (Cambridge, Mass.), 2001, 106, 661. 64 K. Collins, Nat. Rev. Mol. Cell Biol., 2006, 7, 484.
18 T. R. Cech, Cell (Cambridge, Mass.), 2004, 116, 273. 65 S. E. Artandi, N. Engl. J. Med., 2006, 355, 1195.
19 M. A. Blasco, Nat. Rev. Genet., 2005, 6, 611. 66 T. Finkel, M. Serrano and M. A. Blasco, Nature, 2007, 448, 767.
20 J. Karlseder and R. E. Verdun, Nature, 2007, 447, 924. 67 M. A. Blasco, Nat. Chem. Biol., 2007, 10, 640.
21 T. de Lange, Science, 2009, 326, 948. 68 T. R. Calado and N. S. Young, Blood, 2008, 111, 4446.
22 R. OSullivan and J. Karlseder, Nat. Rev. Mol. Cell Biol., 2010, 69 L. S. M. Wong, H. Oeseburg, R. A. de Boer, W. H. van Gilst,
11, 171. D. J. van Veldhuisen and P. van der Harst, Eur. J. Heart Failure,
23 N. Maizels, Nat. Struct. Mol. Biol., 2006, 13, 10551059. 2008, 10, 1049.
24 T. de Lange, Nat. Rev. Mol. Cell Biol., 2004, 5, 323329. 70 M. Armanios, Annu. Rev. Genomics Hum. Genet., 2009, 10, 45.
25 D. Rhodes and R. Giraldo, Curr. Opin. Struct. Biol., 1995, 5, 311. 71 D. J. Patel, A. T. Phan and V. Kuryavyi, Nucleic Acids Res., 2007,
26 W. Palm and T. de Lange, Annu. Rev. Genet., 2008, 42, 301. 35, 7429.
27 T. de Lange, Genes Dev., 2005, 19, 2100. 72 S. Balasubramanian and S. Neidle, Curr. Opin. Chem. Biol., 2009,
28 M.-J. Giraud-Panis, S. Pisano, A. Poulet, M.-H. Le Du and 13, 345.
E. Gilson, FEBS Lett., 2010, 10, 3785. 73 S. Neidle, Curr. Opin. Chem. Biol., 2009, 19, 239.
29 N. D. Hastie, M. Dempster, M. G. Dunlop, A. M. Thompson, 74 S. Neidle, FEBS J., 2010, 277, 1118.
D. K. Green and R. C. Allshire, Nature, 1990, 346, 866. 75 C. M. Azzalin, P. Reichenbach, L. Khoriauli and J. Lingner,
30 N. W. Kim, M. A. Piatyszek, K. R. Prowse, C. B. Harley, Science, 2007, 318, 798.
M. D. West, P. L. Ho, G. M. Coviello, W. E. Wright, 76 S. Schoefter and M. A. Blasco, Nat. Cell Biol., 2008, 10, 228.
S. L. Weinrich and J. W. Shay, Science, 1994, 266, 2011. 77 S. Feuerhahn, N. Iglesias, A. Panza, A. Porro and J. Lingner,
31 G. B. Morin, J. Natl. Cancer Inst., 1995, 87, 859. FEBS Lett., 2010, 584, 3812.
32 C. B. Harley, W. Andrews, C. P. Chiu, J. Feng, W. Funk, 78 P. M. Lansdorp, Trends Biochem. Sci., 2005, 30, 388.
F. Gaeta, K. Hirsch, N. W. Kim, M. Kozlowski, S. S. Wang, 79 I. Joseph, D. Jia and A. J. Lustig, Trends Biochem. Sci., 2006, 31,
S. L. Weinrich, M. D. West, A. Avilion, S. Le, C. W. Greider and 114.
B. Villonponteau, Proc. Am. Assoc. Cancer Res., 1995, 36, 671. 80 N. Hug and J. Lingner, Chromosoma, 2006, 115, 413.
33 E. Raymond, D. Sun, S. F. Chen, B. Windle and D. D. Von Ho, 81 S. A. Stewart and R. A. Weinberg, Annu. Rev. Cell Dev. Biol.,
Curr. Opin. Biotechnol., 1996, 7, 583. 2006, 22, 531.
34 E. Raymond, S. Faivre, V. Dieras and D. Von Ho, Bull. Cancer, 82 R. E. Verdun and J. Karlseder, Nature, 2007, 447, 924.
1997, 84, 1123. 83 M. A. Blasco, Nat. Rev. Genet., 2007, 8, 299.
35 J. W. Shay and A. F. Gazdar, J. Clin. Pathol., 1997, 50, 106. 84 G. Cristofari, K. Sikora and J. Lingner, ACS Chem. Biol., 2007, 2,
36 A. W. Lloyd, Drug Discovery Today, 1998, 3, 523. 155.
37 A. E. Pitts and D. R. Corey, Drug Discovery Today, 1999, 4, 155. 85 Y. Deng, S. S. Chan and S. Chang, Nat. Rev. Cancer, 2008, 8, 450.
38 P. J. Perry and T. C. Jenkins, Expert Opin. Invest. Drugs, 1999, 8, 86 D. R. Corey, Chem. Biol., 2009, 16, 219.
1981. 87 E. Gilson and E. Segal-Bendirdjian, Biochimie, 2010, 92, 321.
39 M. N. Helder, S. de Jong, E. G. E. de Vries and A. G. J. 88 E. Sahin and R. A. DePinho, Nature, 2010, 464, 520.
van der Zee, Drug Resist. Updates, 1999, 2, 104. 89 A. A. Travers, Annu. Rev. Biochem., 1989, 58, 427.
40 W. C. Hahn, S. A. Stewart, M. W. Brooks, Y. S. Gork, E. Eaton, 90 A. H. Wang, G. J. Quigley, F. J. Kolpak, J. L. Crawford,
A. Kurachi, R. L. Beijersbergen, J. H. Knoll, M. Meyerson and J. H. van Boom, G. van der Marel and A. Rich, Nature, 1979,
R. A. Weinberg, Nat. Med. (N. Y.), 1999, 5, 1164. 282, 680.
41 P. T. Rowley and M. Tabler, Anticancer Res., 2000, 20, 4419. 91 DNA Structure and Function, ed. R. R. Sinden, Academic Press,
42 L. R. Kelland, Anti-Cancer Drugs, 2000, 11, 503. New York, 1994.
43 R. Hodes, Proc. Natl. Acad. Sci. U. S. A., 2001, 98, 7649. 92 A. Rich and S. Zhang, Nat. Rev. Genet., 2003, 4, 566.
44 L. K. White, W. E. Wright and J. W. Shay, Trends Biotechnol., 93 J. A. Hackett, D. M. Feldser and C. W. Greider, Cell, 2001, 106,
2001, 19, 114. 275.
45 J. W. Shay, Y. Zou, E. Hiyama and W. E. Wright, Hum. Mol. 94 R. Liu, H. Liu, X. Chen, M. Kirby, P. O. Brown and K. Zhao,
Genet., 2001, 10, 677. Cell, 2001, 106, 309.
46 P. J. Perry and T. C. Jenkins, Mini-Rev. Med. Chem., 2001, 1, 31. 95 R. K. Moyzis, J. M. Buckingham, L. S. Cram, M. Dani,
47 S. Neidle and G. Parkinson, Nat. Rev. Drug Discovery, 2002, 1, L. L. Deaven, M. D. Jones, J. Meyne, R. L. Ratli and
383. J.-R. Wu, Proc. Natl. Acad. Sci. U. S. A., 1988, 85, 6622.
48 J.-L. Mergny, J.-F. Riou, P. Mailliet, M. P. Teulade-Fichou and 96 W. E. Wright, V. M. Tesmer, K. E. Human, S. D. Levene and
E. Gilson, Nucleic Acids Res., 2002, 30, 839. J. W. Shay, Genes Dev., 1997, 11, 2801.

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2735
View Online

97 H. Riethman, Annu. Rev. Genomics Hum. Genet., 2008, 9, 1. 142 K. W. Lim, S. Amrane, S. Bouaziz, W. Xu, Y. Mu, D. J. Patel,
98 M. Gellert, M. N. Lipsett and D. R. Davies, Proc. Natl. Acad. Sci. K. N. Luu and A. T. Phan, J. Am. Chem. Soc., 2009, 131, 4301.
U. S. A., 1962, 48, 2013. 143 I. M. Pedroso, L. F. Duarte, G. Yanez, K. Burkewitz and
99 P. Bates, J. L. Mergny and D. Yang, EMBO Rep., 2007, 8, T. M. Fletcher, Biopolymers, 2007, 87, 74.
1003. 144 T. I. Gaynutdinov, R. D. Neumann and I. G. Panyutin,
100 W. D. Wilson and H. Sugiyama, ACS Chem. Biol., 2007, 2, 589. Nucleic Acids Res., 2008, 36, 4079.
101 B. S. Herbert, J. L. Huppert, F. B. Johnson, A. N. Lane and 145 D. Renciuk, I. Kejnovska, P. Skolakova, K. Bednarova,
A. T. Phan, Biochimie, 2009, 91, 1059. J. Motlova and M. Vorlickova, Nucleic Acids Res., 2009, 37, 6625.
102 W. Guschlbauer, J. F. Chantot and D. Thiele, J. Biomol. Struct. 146 J. Kypr, I. Kejnovska, D. Renciuk and M. Vorlickova, Nucleic
Dyn., 1990, 8, 491. Acids Res., 2009, 37, 1713.
103 J. R. Williamson, Annu. Rev. Biophys. Biomol. Struct., 1994, 23, 147 S. Haider, G. N. Parkinson and S. Neidle, Biophys. J., 2008, 95,
703. 296.
104 T. Simonsson, Biol. Chem., 2001, 382, 621. 148 Y. Xu, T. Ishizuka, K. Kurabayashi and M. Komiyama,
105 A. N. Lane, J. B. Chaires, R. D. Gray and J. O. Trent, Angew. Chem., Int. Ed., 2009, 48, 7833.
Nucleic Acids Res., 2008, 36, 5482. 149 L. Petraccone, J. O. Trent and J. B. Chaires, J. Am. Chem. Soc.,
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

106 D. E. Gilbert and J. Feigon, Curr. Opin. Struct. Biol., 1999, 9, 305. 2008, 130, 16530.
107 A. T. Phan, V. Kuryavyi and D. J. Patel, Curr. Opin. Struct. Biol., 150 H. Q. Yu, D. Miyoshi and N. Sugimoto, J. Am. Chem. Soc., 2006,
2006, 16, 288. 128, 15461.
108 S. Burge, G. N. Parkinson, P. Hazel, A. K. Todd and S. Neidle, 151 S. Redon, S. Bombard, M.-A. Elizondo-Riojas and
Nucleic Acids Res., 2006, 34, 5402. J.-C. Chottard, Nucleic Acids Res., 2003, 31, 1605.
Downloaded by Pennsylvania State University on 14 May 2012

109 J. L. Huppert, Chem. Soc. Rev., 2008, 37, 1375. 152 L. Ying, J. J. Green, H. Li, D. Klenerman and
110 J. L. Mergny, A. De Cian, A. Ghelab, B. Sacca and L. Lacroix, S. Balasubramanian, Proc. Natl. Acad. Sci. U. S. A., 2003, 100,
Nucleic Acids Res., 2005, 33, 81. 14629.
111 A. T. Phan, FEBS J., 2010, 277, 1107. 153 J. Y. Lee, B. Okumus, D. S. Kim and T. Ha, Proc. Natl. Acad.
112 J. R. Williamson, Curr. Opin. Struct. Biol., 1993, 3, 357. Sci. U. S. A., 2005, 102, 18938.
113 D. E. Gilbert and J. Feigon, Curr. Opin. Struct. Biol., 1999, 9, 305. 154 J. Qi and R. H. Shafer, Nucleic Acids Res., 2005, 33, 3185.
114 H. Arthanari and P. H. Bolton, Chem. Biol., 2001, 8, 221. 155 J. Li, J. J. Correia, L. Wang, J. O. Trent and J. B. Chaires,
115 S. Neidle and G. N. Parkinson, Curr. Opin. Struct. Biol., 2003, 13, Nucleic Acids Res., 2005, 33, 4649.
275. 156 M. Tera, K. Iida, K. Ikebukuro, H. Seimiya, K. Shin-ya and
116 J. T. Davis, Angew. Chem., Int. Ed., 2004, 43, 668. K. Nagasawa, Org. Biomol. Chem., 2010, 8, 2749.
117 A. Wlodarczyk, P. Grzybowski, A. Patkowski and A. Dobek, 157 L. Xu, D. Zhang, J. Huang, M. Deng, M. Zhang and X. Zhou,
J. Phys. Chem., 2005, 109, 594. Chem. Commun., 2010, 46, 743.
118 D. Miyoshi, A. Nakao and N. Sugimoto, Nucleic Acids Res., 158 S. Shi, J. Zhao, X. T. Geng, T. M. Yao, H. L. Huang, T. L. Liu,
2003, 31, 1156. L. F. Zheng, Z. H. Li, D. J. Yang and L. N. Ji, Dalton Trans.,
119 Y. Wang and D. J. Patel, Biochemistry, 1992, 31, 8112. 2010, 39, 2490.
120 Y. Kato, T. Ohyama, H. Mita and Y. Yamamoto, J. Am. Chem. 159 H. J. Yu, X. H. Wang, M. L. Fu, J. S. Ren and X. G. Qu,
Soc., 2005, 127, 9980. Nucleic Acids Res., 2008, 36, 5695.
121 P. K. Patel, A. S. R. Koti and R. V. Hosur, Nucleic Acids Res., 160 Y. Xu and H. Sugiyama, J. Am. Chem. Soc., 2004, 126, 6274.
1991, 27, 3836. 161 V. Singh, M. Azarkh, T. E. Exner, J. S. Hartig and M. Drescher,
122 E. Gavathiotis and M. S. Searle, Org. Biomol. Chem., 2003, 1, Angew. Chem., Int. Ed., 2009, 48, 9728.
1650. 162 C. Schatzel, I. Berger, J. Postberg, J. Hanes, H. J. Lipps and
123 G. N. Parkinson, M. P. Lee and S. Neidle, Nature, 2002, 417, 876. A. Pluckthun, Proc. Natl. Acad. Sci. U. S. A., 2001, 98, 8572.
124 A. T. Phan and D. J. Patel, J. Am. Chem. Soc., 2003, 125, 15021. 163 H. J. Lipps and D. Rhodes, Trends Cell Biol., 2003, 28, 632.
125 P. Balagurumoorthy and S. K. Brahmachari, J. Biol. Chem., 164 J. D. Grith, L. Comeau, S. Roseneld, R. M. Stansel,
1994, 269, 21858. A. Bianchi, H. Moss and T. de Lange, Cell (Cambridge, Mass.),
126 M. Vorlickova, J. Chladkova, I. Kejnovska, M. Fialova and 1999, 97, 503.
J. Kypr, Nucleic Acids Res., 2005, 33, 5851. 165 R. M. Stansel, T. de Lange and J. D. Grith, EMBO J., 2001, 20,
127 I. N. Rujan, J. C. Meleney and P. H. Bolton, Nucleic Acids Res., E5532.
2005, 33, 2022. 166 C. Wei and M. Price, Cell. Mol. Life Sci., 2003, 60, 2283.
128 M. Kaushik, A. Bansal, S. Saxena and S. Kukreti, Biochemistry, 167 Y. Xu, H. Sato, Y. Sannohe, K. I. Shinohara and H. Sugiyama,
2007, 46, 7119. J. Am. Chem. Soc., 2008, 130, 16470.
129 N. Zhang, A. T. Phan and D. J. Patel, J. Am. Chem. Soc., 2005, 168 A. T. Phan and J. L. Mergny, Nucleic Acids Res., 2002, 30, 4618.
127, 17277. 169 Z. Zhong, L. Shiue, S. Kaplan and T. de Lange, Mol. Cell. Biol.,
130 Y. Wang and D. J. Patel, Structure (London), 1993, 1, 263. 1992, 12, 4834.
131 Y. Xue, Z. Y. Kan, Q. Wang, Y. Yao, J. Liu, Y. H. Hao and 170 A. Bianchi, S. Smith, L. Chong, P. Elias and T. de Lange,
Z. Tan, J. Am. Chem. Soc., 2007, 129, 11185. EMBO J., 1997, 16, 1785.
132 D. Miyoshi and N. Sugimoto, Biochimie, 2008, 90, 1040. 171 T. Bilaud, C. Brun, K. Ancelin, C. E. Koering, T. Laroche and
133 Y. Xu, Y. Noguchi and H. Sugiyama, Bioorg. Med. Chem., 2006, E. Gilson, Nat. Genet., 1997, 17, 236.
14, 5584. 172 D. Broccoli, A. Smogorzewska, L. Chong and T. de Lange,
134 A. Ambrus, D. Chen, J. X. Dai, T. Bialis, R. A. Jones and Nat. Genet., 1997, 17, 231.
D. Z. Yang, Nucleic Acids Res., 2006, 34, 2723. 173 Y. Chen, Y. Yang, M. van Overbeek, J. R. Donigian, P. Baciu,
135 K. N. Luu, A. T. Phan, V. Kuryavyi, L. Lacroix and D. J. Patel, T. de Lange and M. Lei, Science, 2008, 319, 1092.
J. Am. Chem. Soc., 2006, 128, 9963. 174 R. Court, L. Chapman, L. Fairall and D. Rhodes, EMBO Rep.,
136 A. Matsugami, Y. Xu, Y. Noguchi, H. Sugiyama and 2005, 6, 39.
M. Katahira, FEBS J., 2007, 274, 3545. 175 L. Fairall, L. Chapman, H. Moss, T. de Lange and D. Rhodes,
137 A. T. Phan, K. N. Luu and D. J. Patel, Nucleic Acids Res., 2006, Mol. Cell, 2001, 8, 351.
34, 5715. 176 T. Nishikawa, H. Okamura, A. Nagadoi, P. Konig, D. Rhodes
138 J. Dai, C. Punchihewa, A. Ambrus, D. Chen, R. A. Jones and and Y. Nishimura, Structure (London), 2001, 9, 1237.
D. Z. Yang, Nucleic Acids Res., 2007, 35, 2240. 177 P. Baumann and T. R. Cech, Science, 2001, 292, 117.
139 A. T. Phan, V. Kuryavyi, K. N. Luu and D. J. Patel, Nucleic 178 M. Lei, E. R. Podell and T. R. Cech, Nat. Struct. Mol. Biol., 2004,
Acids Res., 2007, 35, 6517. 11, 1223.
140 J. Dai, M. Carver, C. Punchihewa, R. A. Jones and D. Z. Yang, 179 A. J. Zaug, E. R. Podell and T. R. Cech, Proc. Natl. Acad. Sci.
Nucleic Acids Res., 2007, 35, 4927. U. S. A., 2005, 102, 10864.
141 Y. He, R. D. Neumann and I. G. Panyutin, Nucleic Acids Res., 180 B. R. Houghtaling, L. Cuttonaro, W. Chang and S. Smith,
2004, 32, 5359. Curr. Biol., 2004, 14, 1621.

2736 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

181 S. H. Kim, C. Beausejour, A. R. Davalos, P. Kaminker, S. J. Heo 211 M. M. Ho, A. V. Nq, S. Lam and J. Y. Hung, Cancer Res., 2007,
and J. Campisi, J. Biol. Chem., 2004, 279, 43799. 67, 4827.
182 S. H. Kim, P. Kaminker and J. Campisi, Nat. Genet., 1999, 23, 212 C. Autexier and N. F. Lue, Annu. Rev. Biochem., 2006, 75, 493.
405. 213 K. Damm, U. Hemmann, P. Garin-Chesa, N. Hauel,
183 J. Z. Ye, J. R. Donigian, M. Van Overbeek, D. Loayza, Y. Luo, I. Kaumann, H. Priepke, C. Niestroj, C. Daiber, B. Enenkel,
A. N. Krutchinsky, B. T. Chait and T. de Lange, J. Biol. Chem., B. Guilliard, I. Lauritsch, E. Muller, E. Pascolo, G. Sauter,
2004, 279, 47264. M. Pantic, U. M. Martens, C. Wenz, J. Lingner, N. Kraut,
184 J. Z. Ye, D. Hockemeyer, A. N. Krutchinsky, D. Loayza, W. J. Rettig and A. Schnapp, EMBO J., 2001, 20, 6958.
S. M. Hooper, B. T. Chait and T. de Lange, Genes Dev., 2004, 214 E. Pascolo, C. Wenz, J. Lingner, N. Hauel, H. Priepke,
18, 1649. I. Kaumann, P. GarinChesa, W. J. Rettig, K. Damm and
185 B. Li, S. Oestreich and T. de Lange, Cell (Cambridge, Mass.), A. Schnapp, J. Biol. Chem., 2002, 277, 15566.
2000, 101, 471. 215 H. El Daly, M. Kull, S. Zimmermann, M. Pantic, C. F. Waller
186 X. D. Zhu, B. Kuster, M. Mann, J. H. Petrini and T. de Lange, and U. M. Martens, Blood, 2005, 105, 1742.
Nat. Genet., 2000, 25, 347. 216 I. Naasani, H. Seimiya and T. Tsuruo, Biochem. Biophys. Res.
187 D. Liu, A. Safari, M. S. OConnor, D. W. Chan, A. Laegeler, Commun., 1998, 249, 391.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

Q. Jun and S. Y. Zhou, Nat. Cell Biol., 2004, 6, 673. 217 H. Seimiya, T. Ohhara, T. Suzuki, I. Naasani, T. Shimazaki,
188 D. Hockemeyer, W. Palm, T. Else, J. P. Daniels, K. K. Takai, K. Tsuchiya and T. Tsuruo, Mol. Cancer Ther., 2002, 1, 657.
J. Z. Ye, C. E. Keegan, T. de Lange and G. D. Hammer, 218 N. Hayakawa, K. Nozawa, A. Ogawa, N. Kato, K. Yoshida,
Nat. Struct. Mol. Biol., 2007, 14, 754. K. Akamatsu, M. Tsuchiya, A. Nagasaka and S. Yoshida,
189 D. Loayza and T. de Lange, Nature, 2003, 424, 1013. Biochemistry, 1999, 38, 11501.
Downloaded by Pennsylvania State University on 14 May 2012

190 R. Giraldo, M. Suzuki, L. Chapman and D. Rhodes, Proc. Natl. 219 S. Sasaki, T. Ehara, I. Sakata, Y. Fujino, N. Harada, J. Kimura,
Acad. Sci. U. S. A., 1994, 91, 7658. H. Nakamura and M. Maeda, Bioorg. Med. Chem. Lett., 2001,
191 J. Dejardin and R. E. Kingston, Cell (Cambridge, Mass.), 2009, 11, 583.
136, 175. 220 J. H. Kim, G. E. Lee, J. E. Lee and I. K. Chung, Mol. Pharmacol.,
192 A. Lechel, A. Satyanarayana, Z. Ju, R. R. Plentz, S. Schaetzlein, 2003, 63, 1117.
C. Rudolph, L. Wilkens, S. U. Wiemann, G. Saretzki, 221 J. H. Kim, G. E. Lee, S. W. Kim and I. K. Chung, Biochem. J.,
N. P. Malek, M. P. Manns, J. Buer and K. L. Rudolph, 2003, 373, 523.
EMBO Rep., 2005, 6, 275. 222 T. Ueno, H. Takahashi, M. Oda, M. Mizunuma, A. Yokoyama,
193 S. A. Stewart, I. Ben-Porath, V. J. Carey, B. F. OConnor, Y. Goto, Y. Mizushina, K. Sakaguchi and H. Hayashi,
W. C. Hahn and R. A. Weinberg, Nat. Genet., 2003, 33, 492. Biochemistry, 2000, 39, 5995.
194 H. Oh, S. C. Wang, A. Prahash, M. Sano, C. S. Moravec, 223 P. R. Huang, Y. M. Yeh and T. C. V. Wang, Cancer Lett.
G. E. Taet, L. H. Michael, K. A. Youker, M. L. Entman and (Shannon, Irel.), 2005, 227, 169.
M. D. Schneider, Proc. Natl. Acad. Sci. U. S. A., 2003, 100, 5378. 224 R. Nakai, H. Ishida, A. Asai, H. Ogawa, Y. Yamamoto,
195 J. N. OSullivan, M. P. Bronner, T. A. Brentnall, J. C. Finley, H. Kawasaki, S. Akinaga, T. Mizukami and Y. Yamashita,
W. T. Shen, S. Emerson, M. J. Emond, K. A. Gollahon, Chem. Biol., 2006, 13, 183.
A. H. Moskovitz, D. A. Crispin, J. D. Potter and 225 P. Wu, L. Meng, H. Wang, J. F. Zhou, G. Xu, S. X. Wang, L. Xi,
P. S. Rabinovitch, Nat. Genet., 2002, 32, 280. G. Chen, B. B. Wang, T. Zhu, Y. P. Lu and D. Ma, Biochem.
196 S. U. Wiemann, A. Satyanarayana, M. Tsahuridu, Biophys. Res. Commun., 2005, 335, 36.
H. L. Tillmann, L. Zender, J. Klempnauer, P. Flemming, 226 H. J. Woo, S. J. Lee, B. T. Choi, Y. M. Park and Y. H. Choi,
S. Franco, M. A. Blasco, M. P. Manns and K. L. Rudolph, Exp. Mol. Pathol., 2007, 82, 77.
FASEB J., 2002, 16, 935. 227 K. Hasegawa, Y. Ohashi, K. Ishikawa, A. Yasue, R. Kato,
197 N. J. Samani, R. Boultby, R. Butler, J. R. Thompson and Y. Achiwa, E. Nishio and Y. Udagawa, Int. J. Oncol., 2005, 26,
A. H. Goodall, Lancet, 2001, 358, 472. 1419.
198 J. R. Mitchell, E. Wood and K. Collins, Nature, 1999, 402, 228 H. Ouchi, H. Ishiguro, N. Ikeda, M. Hori, Y. Kubota and
551. H. Uemura, Int. J. Urol., 2005, 12, 73.
199 T. Vulliamy, A. Marrone, F. Goldman, A. Dearlove, M. Bessler, 229 B. Zhu, L. H. Zhang, Y. M. Zhao, J. R. Cui and S. J. Strada,
P. J. Mason and I. Dokal, Nature, 2001, 413, 432. J. Cell. Biochem., 2006, 97, 166.
200 T. Vulliamy, A. Marrone, R. Szydlo, A. Walne, P. J. Mason and 230 C. M. Wang, Z. J. Jia and R. L. Zheng, Planta Med., 2007, 73,
I. Dokal, Nat. Genet., 2004, 36, 447. 180.
201 H. Yamaguchi, R. T. Calado, H. Ly, S. Kajigaya, 231 W. C. Ku, A. J. Cheng and T. C. V. Wang, Biochem. Biophys.
G. M. Baerlocher, S. J. Chanock, P. M. Lansdorp and Res. Commun., 1997, 241, 730.
N. S. Young, N. Engl. J. Med., 2005, 352, 1413. 232 H. He, H. H. X. Xia, J. DeWang, Q. Gu, M. C. M. Lin, B. Zou,
202 A. Marrone, D. Stevens, T. Vulliamy, I. Dokal and P. J. Mason, S. K. Lam, A. O. O. Chan, M. F. Yuen, H. F. Kung and B. C. Y.
Blood, 2004, 104, 3936. Wong, Cancer (N. Y.), 2006, 106, 1243.
203 J. K. Alder, J. J. Chen, L. Lancaster, S. Dano, S. C. Su, 233 A. O. O. Chan, M. F. Yuen, H. F. Kung and B. C. Y. Wong,
J. D. Cogan, I. Vulto, M. Xie, X. Qi, R. M. Tuder, Cancer (N. Y.), 2006, 106, 1243.
J. A. Phillips, 3rd, P. M. Lansdorp, J. E. Loyd and 234 Y. Bermudez, S. Ahmadi, N. E. Lowell and P. A. Kruk, Cancer
M. Y. Armanios, Proc. Natl. Acad. Sci. U. S. A., 2008, 105, Detect. Prev., 2007, 31, 119.
13051. 235 C. West, R. Francis and S. H. Friedman, Bioorg. Chem., 2001, 29,
204 S. Chang, A. S. Multani, N. G. Cabrera, M. L. Naylor, P. Laud, 107.
D. Lombard, S. Pathak, L. Guarente and R. A. DePinho, 236 S. Sasaki, T. Bando, M. Minoshima, T. Shimizu, K. Shinohara,
Nat. Genet., 2004, 36, 877. T. Takaoka and H. Sugiyama, J. Am. Chem. Soc., 2006, 128, 12162.
205 K. K. Wong, R. S. Maser, R. M. Bachoo, J. Menon, 237 J. Murakami, N. Nagai, K. Shigemasa and K. Ohama, Eur. J.
D. R. Carrasco, Y. Gu, F. W. Alt and R. A. DePinho, Nature, Cancer, 1999, 35, 1027.
2003, 421, 643. 238 C. Strahl and E. H. Blackburn, Nucleic Acids Res., 1994, 22, 893.
206 S. Franco, H. J. van de Vrugt, P. Fernandez, M. Aracil, F. Arwert 239 O. A. Olivero, Environ. Mol. Mutagen., 2007, 48, 215.
and M. A. Blasco, Blood, 2004, 104, 3927. 240 D. E. Gomez, A. Kassim and O. A. Olivero, Int. J. Oncol., 1995,
207 C. K. Garcia, W. E. Wright and J. W. Shay, Nucleic Acids Res., 7, 1057.
2007, 35, 7406. 241 A. S. Multani, C. Furlong and S. Pathak, Int. J. Oncol., 1998, 13,
208 A. G. Bodnar, M. Ouellette, M. Frolkis, S. E. Holt, C. P. Chiu, 923.
G. B. Morin, C. B. Harley, J. W. Shay, S. Lichtsteiner and 242 T. M. Fletcher, B. E. Cathers, K. S. Ravikumar, B. M. Mamiya
W. E. Wright, Science, 1998, 279, 349. and S. M. Kerwin, Bioorg. Chem., 2001, 29, 36.
209 R. B. Eros, Exp. Gerontol., 2007, 42, 416. 243 T. Yamaguchi, R. Yamada, A. Tomikawa, K. Shudo, M. Saito,
210 S. B. Cohen, M. E. Graham, G. O. Lovrecz, N. Bache, F. Ishikawa and M. Saneyoshi, Biochem. Biophys. Res. Commun.,
P. J. Robinson and R. R. Reddel, Science, 2007, 315, 1850. 2000, 279, 475.

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2737
View Online

244 X. D. Gao and Y. R. Chen, Asian J. Androl., 2007, 9, 697. 274 S. E. Hamilton, A. E. Pitts, R. R. Katipally, X. Y. Jia,
245 X. D. Gao and Y. R. Chen, Chin. Med. J. (Beijing, Engl. Ed.), J. P. Rutter, B. A. Davies, J. W. Shay, W. E. Wright and
2007, 120, 755. D. R. Corey, Biochemistry, 1997, 36, 11873.
246 T. M. Fletcher, B. E. Cathers, K. S. Ravikumar, B. M. Mamiya 275 M. A. Shammas, C. G. Simmons, D. R. Corey and R. J. S. Reis,
and S. M. Kerwin, Bioorg. Chem., 2001, 29, 36. Oncogene, 1999, 18, 6191.
247 A. Asai, Y. Oshima, Y. Yamamoto, T. Uochi, H. Kusaka, 276 S. E. Hamilton, C. G. Simmons, I. S. Kathiriya and D. R. Corey,
S. Akinaga, Y. Yamashita, K. Pongracz, R. Pruzan, Chem. Biol., 1999, 6, 343.
E. Wunder, M. Piatyszek, S. H. Li, A. C. Chin, C. B. Harley 277 J. G. Harrison, C. Frier, R. Laurant, R. Dennis, K. D. Raney and
and S. Gryaznov, Cancer Res., 2003, 63, 3931. S. Balasubramanian, Bioorg. Med. Chem. Lett., 1999, 9, 1273.
248 M. Akiyama, T. Hideshima, M. A. Shammas, T. Hayashi, 278 X. Cortez-Gonzalez and M. Zanetti, J. Transl. Med., 2007, 5,
M. Hamasaki, Y. T. Tai, P. Richardson, S. Gryaznov, 12.
N. C. Munshi and K. C. Anderson, Cancer Res., 2003, 63, 6187. 279 S. L. Bernhardt, M. K. Gjertsen, S. Trachsel, M. Moller,
249 T. Ozawa, S. M. Gryaznov, L. J. Hu, K. Pongracz, R. A. Santos, J. A. Eriksen, M. Meo, T. Buanes and G. Gaudernack, Br. J.
A. W. Bollen, K. R. Lamborn and D. F. Deen, Neurooncology Cancer, 2006, 95, 1474.
(Amsterdam), 2004, 6, 218. 280 S. M. Domchek, A. Recio, R. Mick, C. E. Clark, E. L. Carpenter,
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

250 E. S. Wang, K. D. Wu, A. C. Chin, S. ChenKiang, K. Pongracz, K. R. Fox, A. DeMichele, L. M. Schuchter, M. S. Leibowitz,
S. Gryaznov and M. A. S. Moore, Blood, 2004, 103, 258. M. H. Wexler, A. B. Vance, G. L. Beatty, E. Veloso,
251 Z. G. Dikmen, G. C. Gellert, S. Jackson, S. Gryaznov, M. D. Feldman and R. H. Vonderheide, Cancer Res., 2007, 67,
R. Tressler, P. Dogan, W. E. Wright and J. W. Shay, 10546.
Cancer Res., 2005, 65, 7866. 281 I. Bolonaki, A. Kotsakis, E. Papadimitraki, D. Aggouraki,
Downloaded by Pennsylvania State University on 14 May 2012

252 B. S. Herbert, G. C. Gellert, A. Hochreiter, K. Pongracz, G. Konsolakis, A. Vagia, C. Christophylakis, I. Nikoloudi,


W. E. Wright, D. Zielinska, A. C. Chin, C. B. Harley, E. Magganas, A. Galanis, P. Cordopatis, K. Kosmatopoulos,
J. W. Shay and S. M. Gryaznov, Oncogene, 2005, 24, 5262. V. Georgoulias and D. Mavroudis, J. Clin. Oncol., 2007, 25, 2727.
253 M. W. Djojosubroto, A. C. Chin, N. Go, S. Schaetzlein, 282 P. F. Brunsvig, S. Aamdal, M. K. Gjertsen, G. Kvalheim,
M. P. Manns, S. Gryaznov, C. B. Harley and K. L. Rudolph, C. J. Markowski-Grimsrud, I. Sve, M. Dyrhaug, S. Trachsel,
Hepatology (Philadelphia), 2005, 42, 1127. M. Moller, J. A. Eriksen and G. Gaudernack, Cancer Immunol.
254 G. C. Gellert, Z. G. Dikmen, W. E. Wright, S. Gryaznov and Immunother., 2006, 55, 1553.
J. W. Shay, Breast Cancer Res. Treat., 2006, 96, 73. 283 Z. Su, J. Dannull, B. K. Yang, P. Dahm, D. Coleman, D. Yancey,
255 A. E. Hochreiter, H. Xiao, E. M. Goldblatt, S. M. Gryaznov, S. Sichi, D. Niedzwiecki, D. Boczkowski, E. Gilboa and
K. D. Miller, S. Badve, G. W. Sledge and B. S. Herbert, J. Vieweg, J. Immunol., 2005, 174, 3798.
Clin. Cancer Res., 2006, 12, 3184. 284 W. C. Hahn, S. A. Stewart, M. W. Brooks, S. G. York, E. Eaton,
256 S. R. Jackson, C. H. Zhu, V. Paulson, L. Watkins, Z. G. Dikmen, A. Kurachi, R. L. Beijersbergen, J. H. M. Knoll, M. Meyerson
S. M. Gryaznov, W. E. Wright and J. W. Shay, Cancer Res., 2007, and R. A. Weinberg, Nat. Med. (N. Y.), 1999, 5, 164.
67, 1121. 285 S. E. Holt, D. L. Aisner, J. Baur, V. M. Tesmer, M. Dy,
257 J. Gomez-Millan, E. M. Goldblatt, S. M. Gryaznov, M. Ouellette, J. B. Trager, G. B. Morin, D. O. Toft,
M. S. Mendonca and B. S. Herbert, Int. J. Radiat. Oncol., Biol., J. W. Shay, W. E. Wright and M. A. White, Genes Dev., 1999,
Phys., 2007, 67, 897. 13, 817.
258 Z. G. Dikmen, W. E. Wright, J. W. Shay and S. M. Gryaznov, 286 L. Neckers, Curr. Top. Med. Chem., 2006, 6, 1163.
J. Cell. Biochem., 2007, 104, 444. 287 O. A. Toogun, W. Zeiger and B. C. Freeman, Proc. Natl. Acad.
259 A. E. Pitts and D. R. Corey, Proc. Natl. Acad. Sci. U. S. A., 1998, Sci. U. S. A., 2007, 104, 5765.
95, 11549. 288 M. M. Kim, M. A. Rivera, I. L. Botchkina, R. Shalaby,
260 A. N. Elayadi, A. Demieville, E. V. Wancewicz, B. P. Monia and A. D. Thor and E. H. Blackburn, Proc. Natl. Acad. Sci.
D. R. Corey, Nucleic Acids Res., 2001, 29, 1683. U. S. A., 2001, 98, 7982.
261 S. Kondo, Y. Kondo, G. Li, R. H. Silverman and J. K. Cowell, 289 C. Guiducci, M. A. Cerone and S. Bacchetti, Oncogene, 2001, 20,
Oncogene, 1998, 16, 3323. 714.
262 D. M. Kushner, J. M. Jayashree, B. Bandyopadhyay, H. Cramer, 290 A. M. Zahler, J. R. Williamson, T. R. Cech and D. M. Prescott,
D. W. Leaman, A. W. Kennedy, R. H. Silverman and Nature, 1991, 350, 718.
J. K. Cowell, Gynecol. Oncol., 2000, 76, 183. 291 S. W. Chan and E. H. Blackburn, Mol. Cell, 2003, 11, 1379.
263 Y. Kondo, S. Koga, T. Komata and S. Kondo, Oncogene, 2000, 292 K. Masutomi, R. Possemato, J. M. Wong, J. L. Currier,
19, 2205. Z. Tothova, J. B. Manola, S. Ganesan, P. M. Lansdorp,
264 S. Mukai, Y. Kondo, S. Koga, T. Komata, B. P. Barna and K. Collins and W. C. Hahn, Proc. Natl. Acad. Sci. U. S. A.,
S. Kondo, Cancer Res., 2000, 60, 4461. 2005, 102, 8222.
265 S. Koga, Y. Kondo, T. Komata and S. Kondo, Gene Ther., 2001, 293 P. Baumann and T. R. Cech, Science, 2001, 292, 1171.
8, 654. 294 H. He, A. S. Multani, W. Cosme-Blanco, H. Tahara, J. Ma,
266 T. Komata1, Y. Kondo, S. Koga, S. C. Ko, L. W. K. Chung and S. Pathak, Y. Deng and S. Chang, EMBO J., 2006, 25, 5180.
S. Kondo, Gene Ther., 2000, 7, 2071. 295 L. Crabbe, R. E. Verdun, C. I. Haggblom and J. Karlseder,
267 S. M. Gryaznov, K. Pongracz, T. Matray, R. Schultz, R. Pruzan, Science, 2004, 306, 1951.
J. Aimi, A. Chin, C. Harley, B. Shea-Herbert, J. Shay, Y. Oshima, 296 H. Ding, M. Schertzer, X. Wu, M. Gertsenstein, S. Selig,
A. Asai and Y. Yamashita, Nucleosides, Nucleotides Nucleic M. Kammori, R. Pourvali, S. Poon, I. Vulto, E. Chavez,
Acids, 2001, 20, 401. P. P. Tam, A. Nagy and P. M. Lansdorp, Cell, 2004, 117, 873.
268 Y. Yokoyama, Y. Takahashi, A. Shinohara, X. Y. Wan, 297 P. L. Opresko, P. A. Mason, E. R. Podell, M. Lei, I. D.
S. Takahashi, K. Niwa and T. Tamaya, Biochem. Biophys. Res. Hickson, T. R. Cech and V. A. Bohr, J. Biol. Chem., 2005, 280,
Commun., 2000, 273, 316. 32069.
269 Y. Kanazawa, K. Ohkawa, K. Ueda, E. Mita, T. Takehara, 298 D. Monchaud and M. P. Teulade-Fichou, Org. Biomol. Chem.,
Y. Sasaki, A. Kasahara and N. Hayashi, Biochem. Biophys. 2008, 6, 627.
Res. Commun., 1996, 225, 570. 299 S. N Georgiades, N. H. Abd Karim, K. Suntharalingam and
270 Y. Yokoyama, Y. Takahashi, A. Shinohara, Z. L. Lian, R. Vilar, Angew. Chem., Int. Ed., 2010, 49, 4020.
X. Y. Wan, Niwa and T. Tamaya, Cancer Res., 1998, 58, 5406. 300 T. M. Ou, Y. J. Lu, J. H. Tan, Z. S. Huang, K. Y. Wong and
271 M. Folini, G. Colella, R. Villa, S. Lualdi, M. G. Daidone and L. Q. Gu, ChemMedChem, 2008, 3, 690.
N. Zaaroni, J. Invest. Dermatol., 2000, 114, 259. 301 E. Gavathiotis, R. A. Heald, M. F. G. Stevens and M. S. Searle,
272 M. A. Shammas, H. Koley, R. B. Batchu, R. C. Bertheau, J. Mol. Biol., 2003, 334, 25.
A. Protopopov, N. C. Munshi and R. K. Goyal, Mol. Cancer, 302 E. Gavathiotis, R. A. Heald, M. F. G. Stevens and M. S. Searle,
2005, 4, 24. Angew. Chem., Int. Ed., 2001, 40, 4749.
273 J. C. Norton, M. A. Piatyszek, W. E. Wright, J. W. Shay and 303 S. M. Gowan, R. Heald, M. F. G. Stevens and L. R. Kelland,
D. R. Corey, Nat. Biotechnol., 1996, 14, 615. Mol. Pharmacol., 2001, 60, 981.

2738 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011
View Online

304 C. Leonetti, S. Amodei, C. DAngelo, A. Rizzo, B. Benassi, 331 C. Hounsou, L. Guittat, D. Monchaud, M. Jourdan, N. Saettel,
A. Antonelli, R. Elli, M. F. G. Stevens, M. DIncalci, G. Zupi J.-L. Mergny and M.-P. Teulade-Fichou, ChemMedChem, 2007,
and A. Biroccio, Mol. Pharmacol., 2004, 66, 1138. 2, 655.
305 R. A. Heald, C. Modi, J. C. Cookson, I. Hutchinson, 332 M.-P. Teulade-Fichou, C. Carrasco, L. Guittat, C. Bailly,
C. A. Laughton, S. M. Gowan, L. R. Kelland and M. F. G. P. Alberti, J.-L. Mergny, A. David, J.-M. Lehn and
Stevens, J. Med. Chem., 2005, 45, 590. W. D. Wilson, J. Am. Chem. Soc., 2003, 125, 4732.
306 R. A. Heald and M. F. G. Stevens, Org. Biomol. Chem., 2003, 1, 333 C. Sissi, L. Lucatello, K. A. Paul, D. J. Maloney, M. B. Boxer,
3377. M. V. Camarasa, G. Pezzoni, E. Menta and M. Palumbo,
307 J. Stanslas, D. J. Hagan, M. J. Ellis, C. Turner, J. Carmichael, Bioorg. Med. Chem., 2007, 15, 555.
W. Ward, T. R. Hammonds and M. F. G. Stevens, J. Med. 334 F. Cuenca, O. Greciano, M. Gunaratnam, S. Haider, D. Munnur,
Chem., 2000, 43, 1563. R. Nanjunda, W. D. Wilso and S. Neidle, Bioorg. Med. Chem.
308 J. C. Cookson, R. A. Heald and M. F. G. Stevens, J. Med. Chem., Lett., 2008, 18, 1668.
2005, 48, 7198. 335 G. N. Parkinson, F. Cuenca and S. Neidle, J. Mol. Biol., 2008,
309 J. C. Cookson, F. Dai, V. Smith, R. A. Heald, C. A. Laughton, 381, 1145.
M. F. G. Stevens and A. M. Burger, Mol. Pharmacol., 2005, 68, 336 M. Di Antonio, F. Doria, S. N. Richter, C. Bertipaglia, M. Mella,
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

1551. C. Sissi, M. Palumbo and M. Freccero, J. Am. Chem. Soc., 2009,


310 P. Phatak, J. C. Cookson, F. Dai, V. Smith, R. B. Gartenhaus, 131, 13132.
M. F. G. Stevens and A. M. Burger, Br. J. Cancer, 2007, 96, 337 R. T. Wheelhouse, D. Sun, H. Han, F. X. Han and L. H. Hurley,
1223. J. Am. Chem. Soc., 1998, 120, 3261.
311 E. Salvati, C. Leonetti, A. Rizzo, M. Scarsella, M. Mottolese, 338 F. Xiaoguang, Huan, R. T. Wheelhouse and L. H. Hurley, J. Am.
Downloaded by Pennsylvania State University on 14 May 2012

R. Galati, I. Sperduti, M. F. Stevens, M. DIncalci, M. Blasco, Chem. Soc., 1999, 121, 3561.
G. Chiorino, S. Bauwens, B. Horard, E. Gilson, A. Stoppacciaro, 339 E. Izbicka, R. T. Wheelhouse, E. Raymond, K. K. Davidson,
G. Zupi and A. Biroccio, J. Clin. Invest., 2007, 117, 3236. R. A. Lawrence, D. Sun, B. E. Windle, L. H. Hurley and
312 N. H. Campbell, G. N. Parkinson, A. P. Reszka and S. Neidle, D. D. Von Ho, Cancer Res., 1999, 59, 639.
J. Am. Chem. Soc., 2008, 130, 6722. 340 D. F. Shi, R. T. Wheelhouse, D. Sun and L. H. Hurley, J. Med.
313 M. A. Read, A. A. Wood, J. R. Harrison, S. M. Gowan, Chem., 2001, 44, 4509.
L. R. Kelland, H. S. Dosanjh and S. Neidle, J. Med. Chem., 341 L. C. Grand, H. Han, R. M. Munoz, S. Weitman, D. D. Von
1999, 42, 4538. Ho, L. H. Hurley and D. J. Bearss, Mol. Cancer Ther., 2002, 1,
314 M. Read, R. J. Harrison, B. Romagnoli, F. A. Tanious, 566.
S. H. Gowan, A. P. Reszka, W. D. Wilson, L. R. Kelland and 342 M.-Y. Kim, M. Gleason-Guzman, E. Izbicka, D. Nishioka and
S. Neidle, Proc. Natl. Acad. Sci. U. S. A., 2001, 98, 4844. L. H. Hurley, Cancer Res., 2003, 63, 3247.
315 R. J. Harrison, S. M. Gowan, L. R. Kelland and S. Neidle, 343 M. W. Freyer, R. Buscaglia, K. Kaplan, D. Cashman,
Bioorg. Med. Chem. Lett., 1999, 9, 2463. L. H. Hurley and E. A. Lewis, Biophys. J., 2007, 92, 2007.
316 R. J. Harrison, J. Cuesta, G. Chessari, M. A. Read, S. K. Basra, 344 L. Martino, B. Pagano, I. Fotticchia, S. Neidle and C. Giancola,
A. P. Reszka, J. Morrell, S. M. Gowan, C. M. Incles, J. Phys. Chem. B, 2009, 113, 14779.
F. A. Tanious, W. D. Wilson, L. R. Kelland and S. Neidle, 345 I. M. Dixon, F. Lopez, A. M. Tejera, J. P. Esteve, M. A. Blasco,
J. Med. Chem., 2003, 46, 4463. G. Pratviel and B. Meunier, J. Am. Chem. Soc., 2007, 129, 1502.
317 S. Taetz, C. Baldes, T. E. Murdter, E. Kleideiter, K. Piotrowska, 346 G. N. Parkinson, R. Ghosh and S. Neidle, Biochemistry, 2007, 46,
U. Bock, E. Haltner-Ukomadu, J. Mueller, H. Huwer, 2390.
U. F. Schaefer, U. Klotz and C. M. Lehr, Pharm. Res., 2006, 347 J. Seenisamy, S. Bashyam, V. Gokhale, H. Vankayalapati,
23, 1031. D. Sun, A. SiddiquiJain, N. Streiner, K. Shinya, E. White,
318 A. M. Burger, F. Dai, C. M. Schultes, A. P. Reszka, M. J. Moore, W. D. Wilson and L. H. Hurley, J. Am. Chem. Soc., 2005, 127,
J. A. Double and S. Neidle, Cancer Res., 2005, 65, 1489. 2944.
319 C. M. Schultes, B. Guyen, J. Cuesta and S. Neidle, Bioorg. Med. 348 I. M. Dixon, F. Lopez, A. M. Tejera, J. P. Esteve, M. A. Blasco,
Chem. Lett., 2004, 14, 4347. G. Pratviel and B. Meunier, J. Am. Chem. Soc., 2007, 129, 1502.
320 M. J. B. Moore, C. M. Schultes, J. Cuesta, F. Cuenca, 349 J. Alzeer, B. R. Vummidi, P. J. Roth and N. W. Luedtke,
M. Gunaratnam, F. A. Tanious, W. D. Wilson and S. Neidle, Angew. Chem., Int. Ed., 2009, 48, 9362.
J. Med. Chem., 2006, 49, 582. 350 K. Shin-ya, K. Wierzba, K. Matsuo, T. Ohtani, Y. Yamada,
321 S. M. Gowan, J. R. Harrison, L. Patterson, M. Valenti, K. Furihata, Y. Hayakawa and H. Seto, J. Am. Chem. Soc., 2001,
M. A. Read, S. Neidle and L. R. Kelland, Mol. Pharmacol., 123, 1262.
2002, 61, 1154. 351 M.-Y. Kim, H. Vankayalapati, K. Shin-ya, K. Wierzba and
322 C. M. Incles, C. M. Schultes, L. R. Kelland and S. Neidle, L. H. Hurley, J. Am. Chem. Soc., 2002, 124, 2098.
Mol. Pharmacol., 2003, 64, 1101. 352 M.-Y. Kim, M. Gleason-Guzman, E. Izbicka, D. Nishioka and
323 C. M. Incles, C. M. Schultes, H. Kempski, H. Koehler, L. H. Hurley, Cancer Res., 2003, 63, 3247.
L. R. Kelland and S. Neidle, Mol. Cancer Ther., 2004, 3, 1201. 353 T. Tauchi, K. Shin-Ya, G. Sashida, M. Sumi, A. Nakajima,
324 M. Gunaratnam, O. Greciano, C. Martins, A. P. Reszka, T. Shimamoto, J. H. Ohyashiki and K. Ohyashiki, Oncogene,
C. M. Schultes, H. Morjani, J.-F. Riou and S. Neidle, 2003, 22, 5338.
Biochem. Pharmacol., 2007, 74, 679. 354 D. Gomez, R. Paterski, T. Lemarteleur, K. Shinya, J. L. Mergny
325 C. Martins, M. Gunaratnam, J. Stuart, V. Makwana, and J. F. Riou, J. Biol. Chem., 2004, 279, 41487.
O. Greciano, A. P. Reszka, L. R. Kelland and S. Neidle, 355 E. M. Rezler, J. Seenisamy, S. Bashyam, M. Y. Kim, E. White,
Bioorg. Med. Chem. Lett., 2007, 17, 2293. W. D. Wilson and L. H. Hurley, J. Am. Chem. Soc., 2005, 127,
326 S. Sparapani, S. M. Haider, F. Doria, M. Gunaratnam and 9439.
S. Neidle, J. Am. Chem. Soc., 2010, 132, 12263. 356 D. Gomez, T. Wenner, B. Brassart, C. Douarre,
327 J.-L. Mergny, L. Lacroix, M.-P. Teulade-Fichou, C. Hounsou, M. F. ODonohue, V. El Khoury, K. Shin-Ya, H. Morjani,
L. Guittat, M. Hoarau, P. B. Arimondo, J.-P. Vigneron, C. Trentesaux and J. F. Riou, J. Biol. Chem., 2006, 281, 38721.
J.-M. Lehn, J.-F. Riou, T. Garestier and C. Helene, Proc. Natl. 357 D. Gomez, M. F. ODonohue, T. Wenner, C. Douarre,
Acad. Sci. U. S. A., 2001, 98, 3062. J. Macadre, P. Koebel, M. J. Giraud-Panis, H. Kaplan,
328 C. Allain, D. Monchaud and M.-P. Teulade-Fichou, J. Am. A. Kolkes, K. Shin-ya and J. F. Riou, Cancer Res., 2006, 66,
Chem. Soc., 2006, 128, 11890. 6908.
329 V. Gabelica, E. S. Baker, M.-P. Teulade-Fichou, E. De Pauw and 358 H. Tahara, K. Shinya, H. Seimiya, H. Yamada, T. Tsuruo and
M. T. Bowers, J. Am. Chem. Soc., 2007, 129, 895. T. Ide, Oncogene, 2006, 25, 1955.
330 J.-L. Mergny, L. Lacroix, M.-P. Teulade-Fichou, C. Hounsou, 359 T. Tauchi, K. Shin-Ya, G. Sashida, M. Sumi, S. Okabe,
L. Guittat, M. Hoarau, P. B. Arimondo, J.-P. Vigneron, J. H. Ohyashiki and K. Ohyashiki, Oncogene, 2006, 25, 5719.
J.-M. Lehn, J.-F. Riou, T. Garestier and C. Helene, Proc. Natl. 360 G. S. Minhas, D. S. Pilch, J. E. Kerrigan, E. J. Lavoie and
Acad. Sci. U. S. A., 2001, 98, 3062. J. E. Rice, Bioorg. Med. Chem. Lett., 2006, 16, 3891.

This journal is c The Royal Society of Chemistry 2011 Chem. Soc. Rev., 2011, 40, 27192740 2739
View Online

361 M. Tera, Y. Sohtome, H. Ishizuka, T. Doi, M. Takagi, K. Shin-ya 385 B. Luke and J. Lingner, EMBO J., 2009, 28, 2503.
and K. Nagasawa, Heterocycles, 2006, 69, 505. 386 S. Schoefter and M. A. Blasco, EMBO J., 2009, 28, 2323.
362 C. M. Barbieri, A. R. Srinivasan, S. G. Rzuczek, J. E. Rice, 387 S. Feuerhahn, N. Iglesias, A. Panza, A. Porro and J. Lingner,
E. J. LaVoie and D. S. Pilch, Nucleic Acids Res., 2007, 35, FEBS Lett., 2010, 84, 3812.
3272. 388 Y. Xu, K. Kaminaga and M. Komiyama, Nucleic Acids Symp.
363 S. G. Rzuczek, D. S. Pilch, E. J. LaVoie and J. E. Rice, Bioorg. Ser. (Oxf)., 2008, 52, 175.
Med. Chem. Lett., 2008, 18, 913. 389 Y. Xu, K. Kaminaga and M. Komiyama, J. Am. Chem. Soc.,
364 M. Tera, Y. Sohtome, H. Ishizuka, T. Doi, M. Takagi, K. Shin-ya 2008, 130, 11179.
and K. Nagasawa, Heterocycles, 2006, 69, 505. 390 H. Martadinata and A. T. Phan, J. Am. Chem. Soc., 2009, 131,
365 M. Tera, K. Iida, H. Ishizuka, M. Takagi, M. Suganuma, T. Doi, 2570.
K. Shin-ya and K. Nagasawa, ChemBioChem, 2009, 10, 431. 391 G. W. Collie, S. M. Haider, S. Neidle and G. N. Parkinson,
366 K. Iida, M. Tera, T. Hirokawa, K. Shin-ya and K. Nagasawa, Nucleic Acids Res., 2010, 38, 5569.
Chem. Commun., 2009, 6481. 392 Y. Xu, T. Ishizuka, T. Kimura and M. Komiyama, J. Am. Chem.
367 P. S. Shirude, E. R. Gillies, S. Ladame, F. Godde, K. Shin-ya, Soc., 2010, 132, 7231.
I. Huc and S. Balasubramanian, J. Am. Chem. Soc., 2007, 129, 393 G. W. Collie, G. N. Parkinson, S. Neidle, F. Rosu, E. De Pauw
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0CS00134A

11890. and V. Gabelica, J. Am. Chem. Soc., 2010, 132, 9328.


368 K. Jantos, R. Rodriguez, S. Ladame, P. S. Shirude and 394 A. Randall and J. D. Grith, J. Biol. Chem., 2009, 284, 13980.
S. Balasubramanian, J. Am. Chem. Soc., 2006, 128, 13662. 395 Y. Xu, Y. Suzuki, K. Ito and M. Komiyama, Proc. Natl. Acad.
369 T. K. Chakraborty, A. Arora, S. Roy, N. Kumar and S. Maiti, Sci. U. S. A., 2010, 107, 14579.
J. Med. Chem., 2007, 50, 5539. 396 Y. Xu, Y. Suzuki and M. Komiyama, Angew. Chem., Int. Ed.,
Downloaded by Pennsylvania State University on 14 May 2012

370 E. S. Baker, J. T. Lee, J. L. Sessler and M. T. Bowers, J. Am. 2009, 48, 3281.
Chem. Soc., 2006, 128, 2641. 397 S. Redon, P. Reichenbach and J. Lingner, Nucleic Acids Res.,
371 J. E. Reed, A. A. Arnal, S. Neidle and R. Vilar, J. Am. Chem. 2010, 38, 5797.
Soc., 2006, 128, 5992. 398 G. Z. Li, M. S. Eller, R. Firoozabadi and B. A. Gilchrest, Proc.
372 D. L. Ma, C. M. Che and S. C. Yan, J. Am. Chem. Soc., 2009, Natl. Acad. Sci. U. S. A., 2003, 100, 527.
131, 1835. 399 G. Z. Li, M. S. Eller, K. Hanna and B. A. Gilchrest, Exp. Cell
373 R. Kieltyka, P. Englebienne, J. Fakhoury, C. Autexier, Res., 2004, 301, 189.
N. Moitessier and H. F. Sleiman, J. Am. Chem. Soc., 2008, 130, 400 A. Porro, S. Feuerhahn, P. Reichenbach and J. Lingner,
10040. Mol. Cell. Biol., 2010, 30, 4808.
374 J. F. Riou, L. Guittat, P. Mailliet, A. Laoui, E. Renou, 401 Z. Deng, J. Norseen, A. Wiedmer, H. Riethman and
O. Petitgenet, F. Megnin-Chanet, C. Helene and J. L. Mergny, P. M. Lieberman, Mol. Cell, 2009, 35, 403.
Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 2672. 402 C. Caslini, J. A. Connelly, A. Serna, D. Broccoli and J. L. Hess,
375 A. De Cian, E. DeLemos, J.-L. Mergny, M.-P. Teulade-Fichou Mol. Cell. Biol., 2009, 29, 4519.
and D. Monchaud, J. Am. Chem. Soc., 2007, 129, 1856. 403 I. L. de Silanes, M. S. dAlcontres and M. A. Blasco,
376 S. Ladame, J. A. Schouten, J. Roldan, J. E. Redman, S. Neidle Nat. Commun., 2010, 1, 33.
and S. Balasubramanian, Biochemistry, 2006, 45, 1393. 404 A. J. Cesare and R. R. Reddel, Nat. Rev. Genet., 2010, 11, 319.
377 R. Rodriguez, G. D. Pantos, D. P. Goncalves, J. K. Sanders and 405 Z. H. Zhong, W. Q. Jiang, A. J . Cesare, A. A. Neumann,
S. Balasubramanian, Angew. Chem., Int. Ed., 2007, 46, 5405. R. Wadhwa and R. R. Reddel, J. Biol. Chem., 2007, 282, 29314.
378 K. Shinohara, Y. Sannohe, S. Kaieda, K. Tanaka, H. Osuga, 406 O. D. Scharer, Angew. Chem., Int. Ed., 2003, 42, 2946.
H. Tahara, Y. Xu, T. Kawase, T. Bando and H. Sugiyam, 407 S. Natarajan and M. J. McEachern, Mol. Cell. Biol., 2002, 22,
J. Am. Chem. Soc., 2010, 132, 3778. 4512.
379 O. Y. Fedoro, M. Salazar, H. Han, V. V. Chemeris, 408 U. M. Lindstrom, R. A Chandrasekaran, L. Orbai,
S. M. Kerwin and L. H. Hurley, Biochemistry, 1998, 37, 12367. S. A. Helquist, G. P. Miller, E. Oroudjev, H. G. Hansma and
380 W. Tuntiwechapikul, J. T. Lee and M. Salazar, J. Am. Chem. E. T. Kool, Proc. Natl. Acad. Sci. U. S. A., 2002, 99, 15953.
Soc., 2001, 123, 5606. 409 H. Siomi, M. C. Siomi, R. L. Nussbaum and G. Dreyfuss,
381 Y. Xu, Y. Suzuki, T. Lonnberg and M. Komiyama, J. Am. Chem. Cell (Cambridge, Mass.), 1993, 2, 291.
Soc., 2009, 131, 2871. 410 J. C. Darnell, K. B. Jensen, P. Jin, V. Brown, S. T. Warren and
382 Y. Xu, K. Ito and M. Komiyama, J. Am. Chem. Soc., 2010, 132, R. B. Darnell, Cell (Cambridge, Mass.), 2001, 107, 489.
631. 411 J. A. Martens, L. Laprade and F. Winston, Nature, 2004, 429,
383 C. M. Azzalin, P. Reichenbach, L. Khoriauli and J. Lingner, 571.
Science, 2007, 318, 798. 412 A. Eulalio, I. Behm-Ansmant and E. Izaurralde, Nat. Rev. Mol.
384 S. Schoefter and M. A. Blasco, Nat. Cell Biol., 2008, 10, 228. Cell Biol., 2007, 8, 9.

2740 Chem. Soc. Rev., 2011, 40, 27192740 This journal is c The Royal Society of Chemistry 2011

Das könnte Ihnen auch gefallen