Vous êtes sur la page 1sur 9

Fungal Genetics and Biology 45 (2008) S54–S62

Contents lists available at ScienceDirect

Fungal Genetics and Biology


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

Review

Predicted elements of telomere organization and function in Ustilago maydis


Patricia Sánchez-Alonso b, Plinio Guzman a,*
a
Departamento de Ingeniería Genética de Plantas, Cinvestav Campus Guanajuato, Apdo. Postal 629, Irapuato, Gto. 36821, Mexico
b
Centro de Investigaciones Microbiológicas, Instituto de Ciencias, BUAP Av. San Claudio y 18 sur, Edif. 76 3er piso, C.U. Puebla, Pue. 72570, Mexico

a r t i c l e i n f o a b s t r a c t

Article history: Telomeres are specialized caps of nucleoprotein complexes located at the chromosome termini. They
Received 4 December 2007 consist of short DNA repeats and of an assortment of associated proteins whose function is currently
Accepted 11 April 2008 under intense investigation in model systems. These specialized structures protect the linear ends of
Available online 29 May 2008
eukaryotic chromosomes against DNA repair and degradation activities, and serve as the substrate for tel-
omerase, the ribonucleoprotein complex that synthesises the telomere repeats. The pivotal role of the
Keywords: telomeres in the maintenance of cell viability in several model eukaryotes, including humans, greatly
Basidiomycete
promoted research in telomere biology. Studies on telomere structure and function in fungi other than
Fungi
Chromosome organization
model systems are limited to providing information on the telomeric repeat sequences. Here, we have
Telomere-associated sequences summarized the current knowledge on the organization of chromosome ends and on the proteins partic-
Telomerase ipating in telomere function in model systems including recent information obtained for filamentous
RecQ helicase fungi. We also describe Ustilago maydis genes that are potential homologs of proteins known from other
Contingency genes systems to participate in telomere biology.
Telosome Ó 2008 Elsevier Inc. All rights reserved.
Shelterin
Epigenetic silencing
DNA-damage response

1. Introduction sequence. A less common mechanism to maintain telomeres is


known as ALT (alternative lengthening of telomeres), and involves
Every eukaryotic chromosome is assembled from a DNA mole- homologous recombination between telomeric repeats in order to
cule associated with proteins that compact the DNA to be stored extend the chromosomal ends (Reddel, 2003).
in the nucleus of the cell. An array of tens or hundreds of copies The discovery of telomerase ignited research on telomere biol-
of a distinct simple DNA sequence is tandemly repeated at the ogy (Blackburn et al., 2006). Soon it was evident a critical role that
two ends of the DNA molecule, this assembly is known as the telo- these protective caps at chromosome-ends had in the maintenance
mere. In most organisms, except in Drosophila where chromo- of cell viability in various model eukaryotes and it was clear that
some-ends specific retrotransposons were recruited to carry out the regulation of telomere length is a complex task for the cell.
telomere functions, the synthesis, composition and functioning of Most cells’ telomeres shorten in every round of replication. Since
telomeres share various conserved features among all eukaryotic very short telomeres result in cell cycle arrest, telomere length is
organisms (Gilson and Geli, 2007; Villasante et al., 2007). thought to be a rate-limiting factor for lifespan and cell survival
Current knowledge of telomere biology comes from the pioneer- (Bodnar et al., 1998; Vaziri and Benchimol, 1998).
ing work on protozoans and Saccharomyces cerevisiae and subse- The telomerase is a highly regulated enzyme whose expression
quently on mammalian cells (Greider, 1998). In the late seventies, and activity are strictly regulated in the metazoans as well as in
Elizabeth H. Blackburn discovered that Tetrahymena telomeres con- unicellular eukaryotes (Cech, 2004; Greider and Blackburn,
sisted of a short DNA sequence motif that was repeated several times 2004). The capability of cells to replicate has been associated with
at the chromosomal end (Blackburn and Gall, 1978). This pattern the functioning of this enzyme and with the extension of the telo-
was then found conserved throughout eukaryotes. The telomerase, meres. Telomerase activity is practically undetectable in the major-
a polymerase involved in the synthesis of the telomere repeats was ity of human somatic cells, while most carcinogenic tumor cells,
identified a few years later (Lingner et al., 1997). Telomerase is a germ cells, and stem cells show significant telomerase activity
specialized reverse transcriptase that includes a RNA component (Kim et al., 1994; Wright et al., 1996). Thus, this sort of evidence
required as the template for synthesis of the telomeric repeated correlates telomerase activity with malignant cellular transforma-
tion (Sharpless and DePinho, 2007). Conversely, the absence of tel-
* Corresponding author. Fax: +52 462 6245849. omerase activity in mesenchymal stem cells of mouse hinders
E-mail address: pguzman@ira.cinvestav.mx (P. Guzman). cellular differentiation (Liu et al., 2004).

1087-1845/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.fgb.2008.04.009
P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62 S55

Telomere-associated proteins, known as the telosome in S. cere- Table 1


visiae and shelterin in humans, initially thought only to be involved Telomere repeat motifs in fungi

in telomere protection and telomerase access, have pivotal roles in Organism Telomere repeat Reference
regulating telomere length and replication (Karlseder, 2006). For Aspergillus nidulans TTAGGG a
instance, the shelterin complex, protects the telomere and is also Pneumocystis carinii TTAGGG b
required for telomere length control, recruiting and releasing telo- Metarrhizium anisopliae TTAGGG c
merase from telomere 30 overhangs (de Lange, 2005). POT1 and Magnaporthe grisea TTAGGG d
Neurospora crassa TTAGGG e
TPP1, two components of this complex, are accessory proteins that Cladosporium fulvum TTAGGG f
operate as a telomerase processing factor (Wang et al., 2007; Xin Glomus intraradices TTAGGG g
et al., 2007). In S. cerevisiae the evolutionarily conserved helicase Beauveria bassiana TTAGGG h
Pif1p regulates the amount of telomerase at chromosomal-ends Pestalotiopsis microspora TTAGGG i
Botrytis cinerea TTAGGG j
by displacing the enzyme from the ends while in Schizosaccharo-
Fusarium oxysporum TTAGGG k
myces pombe, Taz1 protein is required for DNA replication across Ustilago maydis TTAGGG l
the telomeres (Boule et al., 2005; Miller et al., 2006). Saccharomyces cerevisiae TG1–3 m
Adjacent to the telomeric repeats are the subtelomeric regions Schizosaccharomyces pombe TTAC(A)(C)G(1–8) n
that often consist of middle repeated sequences that may harbor Kluyveromyces lactis GGTGTACGGATTT GATTAGGTATGT o
Candida albicans GGTGTACGGATGT CTAACTTCTT p
genes. The chromatin structure of these regions has features of het-
Aspergillus oryzae TTAGGGTCAACA q
erochromatin and the potential to silence adjacent genes, a phe-
nomenon known as TPE (telomere position effect). Epigenetic (a) Bhattacharyya and Blackburn (1997), (b) Underwood et al. (1996), (c) Inglis et al.
(2005), (d) Farman and Leong (1995), (e) Schechtman (1990), (f) Coleman et al.
tags, such as histone hypoacetylation and DNA methylation of telo-
(1993), (g) Hijri et al. (2007), (h) Padmavathi et al. (2003), (i) Levis et al. (1997)and
meric and subtelomeric chromatin have also been observed; the Long et al. (1998), (j) Levis et al. (1997), (k) Powell and Kistler (1990), (l) Guzman
presence of such epigenetic tags correlates with telomere length and Sanchez (1994), (m) Shampay et al. (1984), (n) Matsumoto et al. (1987), (o)
regulation (Ottaviani et al., 2008). McEachern and Blackburn (1995) and McEachern and Hicks (1993), (p) McEachern
and Hicks (1993), (q) Kusumoto et al. (2003).
In contrast to the vast knowledge of telomere biology in model
organisms, our current information about the structure and func-
tion of telomeres in filamentous fungi is scarce. Ustilago maydis Adjacent to the telomere repeats are middle repetitive sequences
possess several advantages for genetic manipulation compared to that are usually found at several chromosomal ends and are highly
other fungal systems and has served as a model to study several polymorphic in length; these regions are commonly known as subt-
basic biological phenomena at the molecular level. The fact that elomeric regions or telomere-associated sequences (TAS). Saccharo-
it is able to grow as yeast-like cells and as hyphae and that fungal myces cerevisiae TAS harbors two main types of sequences known
mycelium can proliferate within tumors of maize tissue opens new as X and Y0 which may function as a buffer at chromosomal ends
opportunities to analyze telomere function during this peculiar (Zakian and Blanton, 1988). Saccharomyces cerevisiae telomerase
lifestyle. In this review we analyze structural features of the mutants undergo telomere attrition and senescence-related pheno-
telomeric region in U. maydis and other fungi and describe genes types. Survivor of telomerase mutants display rearrangements and
involved in well establish processes related to telomere function, amplifications within TAS, rendering chromosomes stable (Chen
correlating them with putative U. maydis homologs. This type of et al., 2001). Ustilago maydis TAS located adjacent to telomeric re-
data analysis through comparative genomics is useful to advance peats consist of two main basic classes of sequences, UTASa and
hypothesis aimed to find links between telomere function and UTASb. UTASa is highly conserved and is mostly located at chromo-
pathogenesis and/or development in fungi that can be later somal ends, whereas UTASb is less conserved and is found inter-
approached experimentally. spersed throughout the genome. Such sequences were identified
in five clones isolated from a chromosome end-enriched library of U.
2. Basic structural features of telomeric regions maydis DNA. UTASa encodes truncated versions of a helicase gene
(Sanchez-Alonso and Guzman, 1998).
The telomeric repeat in most eukaryotes consist of a short tan- In genome sequencing projects, telomere repeats and adjacent
demly repeated G-rich sequence. The G-rich strand protrudes as a TAS are rarely present or assembled within the chromosomal con-
single-strand tail whose length varies in a species-specific manner; tigs. Bioinformatic tools and direct cloning of chromosomal ends
in metazoans this single-stranded tail is usually longer and dis- are required to assemble the ends. TERMINUS (Telomeric End-Read
places the G-rich repeats at the double-stranded portion of the Mining IN Unassembled Sequences) is a tool that has been devel-
telomere. This arrangement is a half Holliday structure that con- oped to map telomere regions on complete genomes by assembling
tributes to telomere maintenance, and triggers the formation of a telomeric reads into chromosome end contigs (Li et al., 2005).
dynamically stable loop structure known as the T-loop (Griffith Using TERMINUS, 45 of the 46 U. maydis chromosome ends were
et al., 1999). In yeast, the T-loop is absent, nevertheless the telo- mapped; the assemblies of U. maydis telomere regions as well as
mere folds over subtelomeric regions, without invasion or base assemblies from other fungi are located at http://fungus.
pairing of DNA sequences; this folding is required for epigenetic kbrin.uky.edu/cgi-bin/gbrowse/U_maydis_broad_tel_100k.
control of regional silencing and telomere lengthening (de Bruin Magnaporthe oryzae is one of the few fungi to have all its chro-
et al., 2001). mosome ends sequenced from the telomeric repeats to the unique
In most filamentous fungi the basic telomere unit is 50 -TTAGG sequences within each chromosome (Rehmeyer et al., 2006). The
G-30 . This motif is conceivably the ancestral telomere repeat in fourteen chromosome ends were sequenced from fosmid clones
metazoans since lower metazoans such as corals, the group of containing telomeric DNA; three independent clones per end were
organisms that span the gap between the fungi and higher metazo- characterized. Eleven of the 14 ends contain highly conserved
ans, contain this repeat at their chromosomal ends (Sinclair et al., sequences encoding truncated versions of a helicase gene, one
2007; Traut et al., 2007). Indeed, this telomeric repeated hexamer chromosome end has rDNA connected to the telomere repeats,
is found in U. maydis and in filamentous fungi in general (see Table and in the two remaining chromosome ends, unique sequences
1). Telomere repeats show differences in some yeasts, the telomere are found adjacent to the telomere repeats. Transposon insertions
unit is longer in Candida sp. and Kluyveromyces lactis and shows an of various types are also common in M. oryzae chromosome ends
irregular repeat structure in S. cerevisiae and S. pombe (see Table 1). (Rehmeyer et al., 2006).
S56 P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62

3. RecQ helicase genes linked to telomeres The function of these genes is unknown, although a member of this
family was identified as a putative transcription factor in a yeast
A remarkable feature found at several fungal chromosomal ends one-hybrid screen (van het Hoog et al., 2007). In K. lactis 30% of
is the presence of highly conserved sequences that encode RecQ genes located at chromosome ends encode proteins predicted to
helicases, located very close to the telomere repeats. Helicase be extracellular or plasma membrane proteins. Other genes encode
genes are generally found at S. cerevisiae chromosome ends. They FLO-like proteins, which also show a telomere-associated location
are encoded on the Y0 elements that become amplified in survivor in S. cerevisiae (Fairhead and Dujon, 2006). Pathogenic fungi may
cells of telomerase negative mutants (Chen et al., 2001). In S. pom- encode families of well characterized or putative virulence factors
be sequences encoding RecQ helicases are located at the four chro- at chromosome ends. Almost two dozen genes encoding adhesins
mosome ends of two of its three chromosomes. Expression of such are located at telomeric regions in Candida glabrata (De Las Peñas
helicases increases in survivor populations of telomerase mutants. et al., 2003). Likewise, in Pneumocystis carinii clusters of major sur-
Likewise, overexpression of a helicase domain in S. pombe telome- face antigen genes are predicted to occur at every chromosome end
rase negative mutants allows a quicker recovery after crisis, sug- (Keely et al., 2005).
gesting a role in telomere maintenance under this particular Little is known about the presence of contingency genes or
condition (Mandell et al., 2005). avirulence genes at chromosome ends of phytopathogenic fungi,
RecQ helicase genes associated with chromosome ends were nevertheless genes with avirulence function in some pathogenic
first described in U. maydis (Sanchez-Alonso and Guzman, 1998). isolates, have been found near telomere ends (Farman, 2007).
They were then found in a similar location in M. grisea (Gao Although this observation hints at the possibility that phytopatho-
et al., 2002), in M. oryzae (Rehmeyer et al., 2006), Metarhizium ani- genic fungi can use a strategy to amplify genes participating in host
sopliae (Inglis et al., 2005) and S. pombe (Mandell et al., 2005). evasion at chromosome termini, the number of avirulence genes in
Apart from the work of telomere-linked RecQ helicases in S. pombe this location is not significantly represented (Farman, 2007). In U.
and the distant helicase encoded in the Y0 element of S. cerevisiae, maydis two gene families associated with chromosome ends has
little is known about the role of such sequences. The location of been predicted (Kamper et al., 2006). Most members of this family
RecQ helicase genes close to the chromosome ends may corre- are located within 14 kb at the ends of the chromosomal contigs
spond to a primitive mechanism for chromosomal stabilization in (see Fig. 1). The function of such families is unknown, although
the absence of telomerase. A meaningful correlation is the fact that they are known to be Ustilago-specific genes, mostly pseudogenes,
WRN, a protein responsible for the Werner Syndrome in humans displaying a high degree of conservation between them (Kamper
and Sgs1p in S. cerevisiae encode RecQ helicases. Although, WRN et al., 2006).
and Sgs1p are not telomere-linked, they are required for telomere
metabolism. WRN interacts with TRF1, TRF2 and POT1 that are 5. Telomerase components in Ustilago maydis
components of the telosome/shelterin complex and Ku70/80 a het-
erodimer which has a role in telomere maintenance (Multani and 5.1. Reverse transcriptase catalytic subunit
Chang, 2007).
The central components of the telomerase enzyme are the re-
4. Gene families encoded at chromosome ends verse transcriptase catalytic subunit (TERT) and the RNA template
that is associated non-covalently to the catalytic subunit (Greider
In several fungi and protists genes encoding proteins involved and Blackburn, 1989). Additional protein components include
in interactions with the environment often increase in number EST1a and EST1b and the dyskerin (dyskeratosis congenital) in
near telomeres. These genes, termed ‘contingency genes’, often dis- mammals, and Est1, Est3 and SM7 proteins in S. cerevisiae (Smog-
play allelic diversity and in most cases are dispensable for survival, orzewska and de Lange, 2004). The TERT subunit contains at least
nevertheless they grant possibilities to confront new environ- four domains. The first is RT, comprising seven conserved motifs (1,
ments. In the human parasites Plasmodium falciparum and Trypan- 2, A, B0 , C, D, E), that correspond to the reverse transcriptase
osoma brucei, families of antigenic surface proteins that induce the domain which catalyzes the synthesis of the G-rich strand from
immune system response are encoded in telomeric regions. This is the RNA template moiety; RT is present in all characterized TERTs.
proposed to be part of a mechanism developed to amplify and Next, is the RNA-binding domain that spans three conserved
diversify surface antigen genes which may hinder host recognition motifs, CP, QFP, and T, and is required for specific binding of TERT
(Barry et al., 2003). to the telomerase RNA subunit. The third domain, the N-terminal
Saccharomyces cerevisiae, depending on its niche, encodes genes domain, may have a conserved region known as GQ which is only
near telomeres that display variation in gene amplification and/or needed for in vivo activity. Finally, the C terminal domain which
expression. Some of these are the PAU genes that represent the appears to be involved in processivity and recruitment of the telo-
largest multigene family in S. cerevisiae with 23 members, most merase to the telomere (Autexier and Lue, 2006). Gene UM00761.1
of them located near the telomeres. PAU genes are regulated by is likely to encode the catalytic core of the U. maydis telomerase
anaerobiosis, and expression analysis showed that some are in- since it contains these four conserved domains including all the
duced during alcoholic fermentation and are negatively regulated motifs present in known TERTs (Sanchez-Alonso et al., unpub-
by oxygen (Rachidi et al., 2000). Others are the MAL and MEL genes lished). Fig. 2A shows a phylogeny comparing known TERTs and
that are required for maltose and alpha-galactosidase fermenta- possible homologs obtained from fungal genomes following BLAST
tion, respectively, and are advantageous in strains used in the bak- analysis at available databases. Ustilago maydis TERT is found in the
ing and brewing industries (Gibson et al., 1997; Teunissen and same clade as predicted TERTs from other filamentous fungi, and
Steensma, 1995). Likewise, the FLO genes that encode cell-wall gly- more specifically shares a branch with Coprinopsis cinerea, another
coproteins participate in the regulation of cellular adhesion (Halme basidiomycete.
et al., 2004).
The gene content at chromosome ends has also been examined 5.2. Dyskerin
in other fungi. The TLO family of C. albicans consists of 15 members,
14 of which are located at chromosome ends. This family is unique Dyskerin is basically, a small-nucleolar ribonucleoprotein
in C. albicans and members are present on every chromosome. (snoRNP) with pseudouridine synthase activity that associates
P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62 S57

Fig. 1. Location of two telomere-associated protein families in U. maydis. Boxes represent left and right ends from the assembled chromosomes where members of Families
#1 and #2 are encoded. ORFs were taken from previously inferred coordinates (see Supplementary Table S2 in Kamper et al. (2006). They are displayed as black boxes for
Family #1 and gray boxes for Family #2. Beginning at the chromosome ends, a 1kb scale is shown at the bottom of the Figure.

Fig. 2. Phylogenetic trees of (A) TERT and (B) Dyskerin proteins. Each tree includes representative proteins from fungi, plants, animals and protists. (A) The phylogenetic tree
is based on the alignment for T, 1, 2, A, B0 , C, D, and E motifs from the RNA-binding and the RT domains of the TERT catalytic core. Accession numbers are as follows:
Saccharomyces cerevisiae (NP_013422.1), Candida albicans (XP_717626.1), Schizosaccharomyces pombe (AAC49802), Ustilago maydis (XP_756908.1), Coprinopsis cinerea
(EAU85360.1), Cryptococcus neoformans (XP_775503.1), Botryotinia fuckeliana (XP_001561236.1), Magnaporthe grisea (XP_363691.1), Gibberella zeae (XP_381218.1), Aspergillus
fumigatus (XP_749051), Aspergillus clavatus (XP_001273297.1), Zea mays (AAX21528.1), Oryza sativa (AAK35007.1), Arabidopsis thaliana (NP_197187.1), Vitis vinifera
(CAO40853.1), Populus trichocarpa (gw1.III.1219.1), Xenopus laevis (AF212299_1), Mus musculus (NP_033380.1), Canis familiaris (NP_001026800.1), Homo sapiens
(NP_937983.2), Tetrahymena termophila (O77448), Euplotes aediculatus (O00939), Oxytricha trifallax (O76332). (B) Phylogenetic tree is based on TrueB_N/PUA pseudouridine
synthases similar to DKC or Cbf5. Sequences were delimited to the three conserved overlapping-regions of dyskerin: the DKCLD N-terminal domain, tRNA pseudouridine
synthases B region, and human dyskerin-like pseudouridine synthase region. Accession numbers are as follows: Ustilago maydis (XP_756832.1), Coprinopsis cinerea
(EAU92842.1), Criptococcus neoformans (XP_570199), Emericella nidulans (O43100.1), Gibberella zeae (XP_387382), Magnaporthe grisea (XP_367607.2), Saccharomyces cerev-
isiae (P33322), Candida albicans (O43101), Schizosaccharomyces pombe (O14007), Arabidopsis thaliana (Q9LD90), Vitis vinifera (CAO15636), Hordeum vulgare (CAB85492), Oryza
sativa (AAG46137), Chlamydomonas reinhardtii (EDP05982), Homo sapiens (NP_001354), Canis familiaris (XP_549382), Mus musculus (NP_001025478), Gallus gallus
(NP_001026286), Xenopus laevis (AAI30043), Drosophila melanogaster (AAD19897), Caenorhabdithis elegans (O17919), Tetrahymena termophila (XP_001012103). Trees were
created using the neighbor-joining method with a bootstrap value of 1000 replicates and plotted using NJplot.
S58 P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62

early with the telomerase catalytic core during the assembly of Candida albicans (Hsu et al., 2007). In contrast to the other telome-
TERT with the RNA template. Dyskerin, comprises several proteins rase components, sequence conservation of Est1 is limited among
that along with the template RNA form a complex (dyskerin eukaryotes. BLAST sequence searches in databases identified the
complex) involved in telomerase performance and stability locus UM04177.1 as the putative U. maydis Est1 gene (Sanchez-
(Smogorzewska and de Lange, 2004). Mass spectrometric sequenc- Alonso et al., unpublished).
ing-analysis of purified human telomerase indicates that the telo-
merase consists of telomerase reverse transcriptase, telomerase 6. Predicted U. maydis telomere-associated proteins
RNA, and dyskerin (Cohen et al., 2007). In mammals, the Dyskerin
complex includes four proteins, NAP57/Dyskerin, NOP10, NHP2 Telomere-associated proteins define another class of compo-
and GAR1. These are encoded by highly conserved genes in eukary- nents of telomeres. These proteins are essential for the organiza-
otes. In S. cerevisiae, the four possible components are Gar1p, Cbf5p tion and homeostasis of the ribonucleoprotein structure of the
(the ortholog of dyskerin), Nhp2p, and Nop10p (Henras et al., telomere. Telomere-associated proteins have important functions
2004). Fig. 2B shows a phylogeny comparing known dyskerins in processes such as the formation of the telomere cap, replication
and possible homologs obtained from several genomes including control by the telomerase pathway, alternative lengthening based
fungi after BLAST searches. The probable dyskerin gene in U. may- on homologous recombination and in nuclear membrane binding,
dis is UM00685.1, and it is found in the same clade as other among others. Several of the telomere-associated proteins are con-
predicted fungal dyskerins, particularly with dyskerins from the served among eukaryotes (Smogorzewska and de Lange, 2004). We
basidiomycetes C. cinerea and Cryptococcus (Sanchez-Alonso searched the U. maydis predicted proteome for possible homologs
et al., unpublished). of conserved telomere-associated proteins. Table 2. lists the pro-
teins that were found along with the predicted U. maydis genes.
5.3. Est1 protein
6.1. Telosome/shelterin complex components
A third component of the telomerase is the Est1 protein. Est1 is
a non-catalytic protein required for proper telomerase function Telomere maintenance relies on a multi-subunit complex
(Lendvay et al., 1996). In S. cerevisiae, Est1 assists in the recruit- known as telosome/shelterin. This complex is formed by proteins
ment of the telomerase to short chromosome termini which are with related functions in metazoan and unicellular eukaryotes.
preferred substrates for this enzyme (Bianchi and Shore, 2007). The function and location of the telosome/shelterin complex is re-
Est1 is also required for telomerase assembly and activation in stricted to the chromosome termini (Smogorzewska and de Lange,

Table 2
Predicted loci for telomere-associated proteins in Ustilago maydis

Ustilago maydis locus Telomere-associated Best alignment hit to Score and E-value Effect on telomeres Refs.
protein currently characterized
homologs (accession)
UM05117.1 POT1 Gallus gallus POT1 homolog S = 48.5 E = 5.6  106 Components of the telosome/shelterin a, b
(NP_996875) complex. Protection of telomere, telomere
UM02326.1 TRF1 Muntiacus reevesi TRF1 S = 55.8 E = 2  105 length regulation
(AAU43270.1)
UM05756.1 Ku80 Aspergillus sojae Ku80 S = 207 E = 2  1051 Components of the Ku DNA-damage response c
(BAE78504) heterodimer. Protection at telomere,
UM05148.1 Ku70 Aspergillus oryzae Ku70 S = 225 E = 1  1056 recognition and binding of DNA DBS. Involved
(BAE78501) in NHEJ recombination
UM01085.1 Rad50 Mus musculus Rad50 S = 501 E = 1  10139 Components of the MRN/MRX complex. d, e
(NP_033038) Generation of G-overhangs. Prevention of re-
UM04704.1 Mre11 Coprinopsis cinerea Mre11 S = 575 E=4  10162 replication. Required for ALT mechanism
homolog (Q9UVN9)
UM01003.1 Nbs1 Mus musculus Nbs1 S = 52.8 E = 2  104
(NP_038780)
UM02874.1 RecQ-like helicases Schizosaccharomyces pombe S = 567 E = 2  10159 Maintaining genomic stability. Resolution of f, g, h
Rqh1 (NP_593092.1) telomere recombination intermediates where
replication stall, and at an interface between
DNA replication and DNA repair in
dysfunctional telomeres
UM01110.1 ATR, ATM Homo sapiens ATR S = 806 E = 0.0 Signal transducer, and component of telomere i, j, k
(CAA70298) capping functions Controls cellular response to
DSB
UM00963.1 SIR2family of proteins Mus musculus Sirt1 S = 280 E = 2  1073 Histone deacetylase. Spreading of silencing l
(NP_062786.1)
79
UM058920.1 Gallus gallus Sirt1 S = 297 E = 9  10
(NP_001017414)
UM06360.1 Nph2 Homo sapiens Nph2 S = 183 E = 3  1045 Dyskerin complex components along with m, n
(NP_004999) DKC1. Involved in telomerase catalytic core
UM04573.1 Gar1 Homo sapiens Gar1 S = 101 E = 3  1020 and RNP biogenesis
(NP_061856)
UM03354.1 Nop1 Saccharomyces cerevisiae S = 901 E = 4  1017
Nop1 (2AQA_A)

Protein BLASTs were performed by using BLOSUM62 matrix and default gap costs. (a) Baumann and Cech (2001); (b) de Lange (2005); (c) Ribes-Zamora et al. (2007); (d) Chai
et al. (2006); (e) Larrivee et al. (2004); (f) Opresko et al. (2004), (g) Li et al. (2004); (h) Eller et al. (2006); (i) Lavin and Kozlov (2007), (j) Sabourin et al. (2007); (k) Hector et al.
(2007); (l) Kaeberlein (2007); (m) Cohen et al. (2007), (n) Henras et al. (2004).
P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62 S59

2004). These proteins have important roles, such as determining silenced chromatin (Kaeberlein, 2007). Sir2-related proteins are
the structure and stability of T-loops, regulating the access of telo- broadly named sirtuins (North and Verdin, 2004). A potential
merase to telomeric ssDNA and safeguarding chromosome ends to homolog of Sir2 is predicted in U. maydis (see Table 2).
prevent recognition as double-strand breaks (Griffith et al., 1999). Telomere length has a major effect on the epigenetic status of
The yeast telosome complex includes the Rap1 and Cdc13 proteins, chromosome ends. Acetylation and methylation of histones are
which interact directly with double-stranded and single-stranded part of the mechanism to regulate the length of telomere repeats
telomeric DNA, respectively, and also factors recruited by Rap1. which is important for maintenance of telomere architecture. Loss
Those factors are the silent information regulators Sir2, Sir3 and of telomeric repeats lead to a loss of heterochromatic architecture
Sir4, and the RAP1 interacting proteins Rif1 and Rif2. In humans at chromosome termini. Thus, histone modifications are required
the shelterin complex consists of six proteins: POT1, TRF1, TRF2, to maintain a stable chromatin structure and for the counting of
TIN1, Rap1 and TPP1 (Blasco, 2007; de Lange, 2005). Information telomere repeats (Blasco, 2007).
on the functions of these components and their presence in U. may-
dis is presented below. 6.2. DNA-damage response

6.1.1. Protection of the chromosome termini DNA double-strand breaks (DSBs) are recognized by DNA-dam-
A key component required for the protection of chromosome age response (DDR) factors. A basic role of telomeres is to prevent
ends and the regulation of telomere length in S. pombe and mam- chromosome ends from being recognized as DSB. Mre11/Rad50/
mals is POT1 (Protection Of Telomeres) (Baumann and Cech, 2001). Nbs1 (MRN) in mammals and Mre11/Rad50/Xrs2 (MRX) in S. cere-
POT1 is a single-stranded telomere DNA binding protein ortholo- visiae are complexes involved early in DSB repair. They are widely
gous to yeast Cdc13. Cdc13 is enriched at the single-stranded do- conserved in evolution, and their components have been reported
main of telomeres at late S-phase together with the essential in many metazoans and unicellular eukaryotes (Chai et al., 2006;
endonuclease Dna2, which is thought to generate the long TG-rich Larrivee et al., 2004). MRN/MRX also function in S-phase check-
single-strand overhangs of telomeric DNA that are also present at point activation, in prevention of DNA re-replication and in the
late S-phase (Tomita et al., 2004). Cdc13 recruits telomerase possi- maintenance of telomere integrity (Lee et al., 2007; Olson et al.,
bly through a direct interaction with Est1 (Bianchi et al., 2004). In 2007). Indeed several functions for MRN/MRX at telomeres have
humans, Pot1 interacts with TIN2, and with TPP1, the homolog of been described. In mammalian cells, after conventional DNA syn-
ciliate TEBP beta (Xin et al., 2007). TPP1 regulates the levels of thesis, telomeric DNA resembles DSB and MRN/MRX complexes re-
POT1 and TIN2 both within the nucleus and in the cytoplasm, since cruited by TRF2 participate in the generation of 30 single-strand
it contains a functional nuclear export signal, which aids nuclear overhangs by C-strand resection at the S-phase of the cell cycle
traffic of these shelterin complex proteins (Chen et al., 2007). (Zhu et al., 2000). In yeast, knock-out of MRX components resulted
TIN2 protects TRF1 from ADP-ribosylation by tankyrase (a poly- in a shortening of G-overhangs (Larrivee et al., 2004; Zhong et al.,
adenosine diphosphate–ribose polymerase). Poly-ADP ribosylation 2007) and depletion of the human MRN complex, affects the route
of TRF1 leads to its dissociation of double-strand telomeric DNA of alternate lengthening (Larrivee et al., 2004; Zhong et al., 2007).
and destruction by ubiquitination mediated proteolysis (Ye and The MRE/MRX complex is likely to be present in U. maydis since
de Lange, 2004). POT1 is highly conserved throughout evolution homologs of Mre11, Rad50 and Nbs1 are predicted to be encoded
and candidate genes can be predicted in several organisms from in the genome (see Table 2).
mammals and plants to unicellular eukaryotes, whereas TPP1 Members of the RecQ family of DNA helicases participate in
and TIN2 are much less well conserved and cannot be readily iden- telomere metabolism by interacting with proteins involved in the
tified in available databases (Baumann and Cech, 2001). maintenance of the chromosome termini as well as by setting up
Of the six proteins of the shelterin complex, TRF1 and TRF2 rec- of the DNA-damage response at the telomere. The RecQ family of
ognize and bind to double-stranded telomeric repeats. Similar to DNA helicases comprises a highly conserved group of enzymes that
Rap1 from S. cerevisiae, and TAZ1 from S. pombe, TRF1 and TRF2 exhibits homology with, and receives its name from, the RecQ heli-
have a SANT/myb-like domain that recognizes double-stranded case of Escherichia coli (Irino et al., 1986). Mutations in RecQ heli-
telomere sequences. This binding is essential for telomere length case genes cause genome instability, hyper-recombination, and
control. In mammalian cells TRF2 interacts with POT1 and with hypersensitivity to DNA-damaging agents. In S. cerevisiae the
hRAP1 (Hug and Lingner, 2006). On the other hand, TRF1, is a neg- recombination based ALT pathway require the RecQ homolog
ative telomere-length regulator, and its binding to the double- Sgs1p to allow Type II survivors to rise in telomerase-negative mu-
strand telomere repeat is controlled by tankyrases 1 and 2 and tants (Huang et al., 2001; Johnson et al., 2001). In humans there are
by TIN2. Access of telomerase could be possible through the disso- five helicase RecQ homologs, mutations in three of them trigger
ciation and ubiquitin-mediated proteolysis of TRF1 (Chang et al., syndromes associated with genome instability and cancer (Opresko
2003). Mutations in either Rap1, TAZ1, TRF1 or TRF2 lead to a telo- et al., 2004). Such is the case of Werner (WRN) that initiates the
mere elongation phenotype (Hug and Lingner, 2006). A possible DNA-damage response pathway when telomeres are disrupted
homolog of TRF is also predicted in U. maydis (see Table 2). (Eller et al., 2006). In addition WRN interacts with TRF1, TRF2
and POT1, components of the shelterin complex, and with the Ku
6.1.2. Epigenetic silencing at chromosome termini complex (Li et al., 2004; Opresko et al., 2004). The Ku complex is
A conserved feature of telomeres is the heterochromatization of a DNA-damage response heterodimer that has a dual function of
telomere-associated regions that results in a silencing effect recognizing and protecting the chromosome termini and recogniz-
known as telomere position effect or TPE. Silencing is a reversible ing non-homologous-end-joining (NHEJ) when DNA double-strand
state of transcriptional repression that initiates with heterochro- breaks occurred. The dual activities of the Ku heterodimer, which
matization at specific sequences and then expands to adjacent consists of Ku70 and Ku80, relies in the different affinities defined
regions. This function of telomeres is essential for chromosome by alpha helices located on each surface of the two sides of the
maintenance. The telosome/shelterin complex and the siRNA complex. Ku70 binds to the DNA end, which is required for NHEJ,
machinery mediate telomeric silencing (Blasco, 2007; Smog- and Ku80 binds to the telomere and is found in the telomere het-
orzewska and de Lange, 2004). In S. cerevisiae, Sir2/Sir3 complex erochromatin (Ribes-Zamora et al., 2007). Homologs of a WRN
is recruited by Rap1 or Ku to the telomere. Sir2, a histone deacetyl- RecQ helicase and the Ku complex (Ku70 and Ku80) may be pres-
ase type III, removes acetyl residues from histones to spread the ent in the U. maydis genome (see Table 2).
S60 P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62

The ATM protein kinases family also has an effect on telo- A locus with strong similarity to ATM kinases is present in the
mere homeostasis. ATM and ATR kinases are members of the U. maydis genome (see Table 2).
phosphatidyl inositol 3-kinase-like family of signal transducers
that are conserved among living organisms. The human ATM 7. Closing remarks
(ataxia telangiectasia mutated) and ATR (ataxia telangiectasia re-
lated) kinases and their yeast homologs Tel1p and Mec1p ki- Telomeres and telomere-associated sequences are active
nases respectively, function upstream in the checkpoint regions required not only for maintenance of chromosomal integ-
pathway for DNA damage repair response (Lavin and Kozlov, rity but often also as a reservoir of genetic information valuable
2007). They are positive telomere length regulators that act in for environmental adaptation. In this review, we have summarized
response to telomere shortening. Tel1p preferentially associates general features of telomere biology and suggested candidate U.
to short telomeres and this association is stimulated by Mre11 maydis genes that are potentially involved in telomere metabolism.
(Hector et al., 2007; Sabourin et al., 2007). A substrate of Tel1p Although, genome sequencing projects are invaluable resources
and Mec1p is the single-strand binding protein Cdc13. Thus, that reveal the DNA sequence at the chromosome ends, sequences
phosphorylation of Cdc13p together with the MRX complex are at the tip of the chromosome are often not present within the chro-
important for the maintenance of chromosome termini regulat- mosomal contigs. Furthermore, telomeric regions are enriched for
ing telomerase recruitment in the telomere (Tseng et al., 2006). DNA repetitive elements that make the assembly of the sequences

Fig. 3. A model for telomere organization and function in U. maydis. The structure and associated components of the chromosome ends from (A) S. cerevisiae, (B) S. pombe and
(C) U. maydis are shown. (A) and (B) are based on published findings (Blasco, 2007; Eller et al., 2006; Hug and Lingner, 2006; Lavin and Kozlov, 2007; Lee et al., 2007); in (C)
the model was deduced based on the predicted proteins shown in Table 2.
P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62 S61

a difficult task. Thus, to have an accurate vision of the chromosome Chai, W., Sfeir, A.J., Hoshiyama, H., Shay, J.W., Wright, W.E., 2006. The involvement
of the Mre11/Rad50/Nbs1 complex in the generation of G-overhangs at human
ends it is often necessary to make use of strategies to directly clone
telomeres. EMBO Rep. 7, 225–230.
the tip of the chromosome up to the single copy sequences. M. ory- Chang, W., Dynek, J.N., Smith, S., 2003. TRF1 is degraded by ubiquitin-mediated
zae is the only filamentous fungus to have all its chromosome ends proteolysis after release from telomeres. Genes Dev. 17, 1328–1333.
revealed. A common feature found adjacent to the telomere re- Chen, L.Y., Liu, D., Songyang, Z., 2007. Telomere maintenance through spatial control
of telomeric proteins. Mol. Cell. Biol. 27, 5898–5909.
peats is the occurrence of sequences encoding RecQ helicases. Chen, Q., Ijpma, A., Greider, C.W., 2001. Two survivor pathways that allow growth in
Complete and truncated versions of RecQ helicases are often pres- the absence of telomerase are generated by distinct telomere recombination
ent at several chromosome ends. This feature may be unique to events. Mol. Cell Biol. 21, 1819–1827.
Cohen, S.B., Graham, M.E., Lovrecz, G.O., Bache, N., Robinson, P.J., Reddel, R.R., 2007.
fungal genomes and has been detected in 10 additional fungal gen- Protein composition of catalytically active human telomerase from immortal
omes. Species-specific genes are also common at chromosome cells. Science 315, 1850–1853.
ends. Indeed, U. maydis encodes two such gene families (see Coleman, M.J., McHale, M.T., Arnau, J., Watson, A., Oliver, R.P., 1993. Cloning and
characterisation of telomeric DNA from Cladosporium fulvum. Gene 132, 67–
Fig. 1). The identity and function of such genes has not been estab- 73.
lished and a comparative analysis in various independent U. maydis de Bruin, D., Zaman, Z., Liberatore, R.A., Ptashne, M., 2001. Telomere looping permits
isolates may be useful to understand the nature of these gene gene activation by a downstream UAS in yeast. Nature 409, 109–113.
de Lange, T., 2005. Shelterin: the protein complex that shapes and safeguards
families. human telomeres. Genes Dev. 19, 2100–2110.
Bioinformatic analysis revealed potential U. maydis homologs De Las Peñas, A., Pan, S.J., Castano, I., Alder, J., Cregg, R., Cormack, B.P., 2003.
involved in telomere metabolism. Two genes of the telomerase Virulence-related surface glycoproteins in the yeast pathogen Candida glabrata
are encoded in subtelomeric clusters and subject to RAP1- and SIR-dependent
complex were readily identified, genes encoding the catalytic sub-
transcriptional silencing. Genes Dev. 17, 2245–2258.
unit and dyskerin, suggesting that the mechanism previously de- Eller, M.S., Liao, X., Liu, S., Hanna, K., Backvall, H., Opresko, P.L., Bohr, V.A., Gilchrest,
scribed in model systems for telomere synthesis is conserved in B.A., 2006. A role for WRN in telomere-based DNA damage responses. Proc. Natl.
U. maydis. Likewise, genes encoding the telomere-associated pro- Acad. Sci. USA 103, 15073–15078.
Fairhead, C., Dujon, B., 2006. Structure of Kluyveromyces lactis subtelomeres:
teins POT1 and TRF1 or the MRN/MRX and Ku complexes are likely duplications and gene content. FEMS Yeast Res. 6, 428–441.
to be conserved in U. maydis. These findings indicate that the Farman, M.L., 2007. Telomeres in the rice blast fungus Magnaporthe oryzae: the
essential functions of the telomere, as well as the recognition of world of the end as we know it. FEMS Microbiol. Lett. 273, 125–132.
Farman, M.L., Leong, S.A., 1995. Genetic and physical mapping of telomeres in the
DNA double-strand breaks by the DNA damage repair complex, rice blast fungus, Magnaporthe grisea. Genetics 140, 479–492.
are also conserved in U. maydis. A model for telomere organization Gao, W., Khang, C.H., Park, S.Y., Lee, Y.H., Kang, S., 2002. Evolution and organization
and function in this fungus is shown in Fig. 3. of a highly dynamic, subtelomeric helicase gene family in the rice blast fungus
Magnaporthe grisea. Genetics 162, 103–112.
In summary, little information has been generated on the struc- Gibson, A.W., Wojciechowicz, L.A., Danzi, S.E., Zhang, B., Kim, J.H., Hu, Z., Michels,
ture and function of telomeres in filamentous fungi. Complete C.A., 1997. Constitutive mutations of the Saccharomyces cerevisiae MAL-
analysis of full chromosome termini lies ahead for fungal genomes. activator genes MAL23, MAL43, MAL63, and mal64. Genetics 146, 1287–1298.
Gilson, E., Geli, V., 2007. How telomeres are replicated. Nat. Rev. Mol. Cell. Biol. 8,
The assumption that the telomere-associated region is an impor- 825–838.
tant reservoir of gene families encoding contingency-like genes Greider, C.W., 1998. Telomeres and senescence: the history, the experiment, the
must be confirmed. Likewise, the role of the telomerase complex future. Curr. Biol. 8, R178–181.
Greider, C.W., Blackburn, E.H., 1989. A telomeric sequence in the RNA of
during the cell cycle and the life cycle of fungi and particularly in
Tetrahymena telomerase required for telomere repeat synthesis. Nature 337,
dimorphic fungi will have to be assessed. 331–337.
Greider, C.W., Blackburn, E.H., 2004. Tracking telomerase. Cell 116, S83–S86. 81 p
following S86.
Acknowledgments Griffith, J.D., Comeau, L., Rosenfield, S., Stansel, R.M., Bianchi, A., Moss, H., de Lange,
T., 1999. Mammalian telomeres end in a large duplex loop. Cell 97, 503–514.
We thank June K. Simpson and Gabriela Olmedo for reviews and Guzman, P.A., Sanchez, J.G., 1994. Characterization of telomeric regions from
Ustilago maydis. Microbiology 140 (Pt 3), 551–557.
comments on the manuscript. Halme, A., Bumgarner, S., Styles, C., Fink, G.R., 2004. Genetic and epigenetic
This work is supported by grants from CONACyT, Mexico. regulation of the FLO gene family generates cell-surface variation in yeast. Cell
116, 405–415.
Hector, R.E., Shtofman, R.L., Ray, A., Chen, B.R., Nyun, T., Berkner, K.L., Runge, K.W.,
References 2007. Tel1p preferentially associates with short telomeres to stimulate their
elongation. Mol. Cell 27, 851–858.
Autexier, C., Lue, N.F., 2006. The structure and function of telomerase reverse Henras, A.K., Capeyrou, R., Henry, Y., Caizergues-Ferrer, M., 2004. Cbf5p, the
transcriptase. Annu. Rev. Biochem. 75, 493–517. putative pseudouridine synthase of H/ACA-type snoRNPs, can form a complex
Barry, J.D., Ginger, M.L., Burton, P., McCulloch, R., 2003. Why are parasite with Gar1p and Nop10p in absence of Nhp2p and box H/ACA snoRNAs. RNA 10,
contingency genes often associated with telomeres? Int. J. Parasitol. 33, 1704–1712.
29–45. Hijri, M., Niculita, H., Sanders, I.R., 2007. Molecular characterization of chromosome
Baumann, P., Cech, T.R., 2001. Pot1, the putative telomere end-binding protein in termini of the arbuscular mycorrhizal fungus Glomus intraradices
fission yeast and humans. Science 292, 1171–1175. (Glomeromycota). Fungal Genet. Biol. 44, 1380–1386.
Bhattacharyya, A., Blackburn, E.H., 1997. Aspergillus nidulans maintains short Hsu, M., Yu, E.Y., Singh, S.M., Lue, N.F., 2007. Mutual dependence of Candida albicans
telomeres throughout development. Nucleic Acids Res. 25, 1426–1431. Est1p and Est3p in telomerase assembly and activation. Eukaryot. Cell 6, 1330–
Bianchi, A., Negrini, S., Shore, D., 2004. Delivery of yeast telomerase to a DNA break 1338.
depends on the recruitment functions of Cdc13 and Est1. Mol. Cell 16, 139–146. Huang, P., Pryde, F.E., Lester, D., Maddison, R.L., Borts, R.H., Hickson, I.D., Louis, E.J.,
Bianchi, A., Shore, D., 2007. Increased association of telomerase with short 2001. SGS1 is required for telomere elongation in the absence of telomerase.
telomeres in yeast. Genes Dev. 21, 1726–1730. Curr. Biol. 11, 125–129.
Blackburn, E.H., Gall, J.G., 1978. A tandemly repeated sequence at the termini of the Hug, N., Lingner, J., 2006. Telomere length homeostasis. Chromosoma 115, 413–425.
extrachromosomal ribosomal RNA genes in Tetrahymena. J. Mol. Biol. 120, 33– Inglis, P.W., Rigden, D.J., Mello, L.V., Louis, E.J., Valadares-Inglis, M.C., 2005.
53. Monomorphic subtelomeric DNA in the filamentous fungus, Metarhizium
Blackburn, E.H., Greider, C.W., Szostak, J.W., 2006. Telomeres and telomerase: the anisopliae, contains a RecQ helicase-like gene. Mol. Genet. Genomics 274, 79–
path from maize, Tetrahymena and yeast to human cancer and aging. Nat. Med. 90.
12, 1133–1138. Irino, N., Nakayama, K., Nakayama, H., 1986. The recQ gene of Escherichia coli K12:
Blasco, M.A., 2007. The epigenetic regulation of mammalian telomeres. Nat. Rev. primary structure and evidence for SOS regulation. Mol. Gen. Genet. 205, 298–
Genet. 8, 299–309. 304.
Bodnar, A.G., Ouellette, M., Frolkis, M., Holt, S.E., Chiu, C.P., Morin, G.B., Harley, C.B., Johnson, F.B., Marciniak, R.A., McVey, M., Stewart, S.A., Hahn, W.C., Guarente, L.,
Shay, J.W., Lichtsteiner, S., Wright, W.E., 1998. Extension of life-span by 2001. The Saccharomyces cerevisiae WRN homolog Sgs1p participates in
introduction of telomerase into normal human cells. Science 279, 349–352. telomere maintenance in cells lacking telomerase. EMBO J. 20, 905–913.
Boule, J.B., Vega, L.R., Zakian, V.A., 2005. The yeast Pif1p helicase removes Kaeberlein, M., 2007. Molecular basis of ageing. EMBO Rep. 8, 907–911.
telomerase from telomeric DNA. Nature 438, 57–61. Kamper, J., Kahmann, R., Bolker, M., Ma, L.J., Brefort, T., Saville, B.J., Banuett, F.,
Cech, T.R., 2004. Beginning to understand the end of the chromosome. Cell 116, Kronstad, J.W., Gold, S.E., Muller, O., Perlin, M.H., Wosten, H.A., de Vries, R., Ruiz-
273–279. Herrera, J., Reynaga-Pena, C.G., Snetselaar, K., McCann, M., Perez-Martin, J.,
S62 P. Sánchez-Alonso, P. Guzman / Fungal Genetics and Biology 45 (2008) S54–S62

Feldbrugge, Basse, C.W., Steinberg, G., Ibeas, J.I., Holloman, W., Guzman, P., Padmavathi, J., UmaDevi, K., Rao, C.U., Reddy, N.N., 2003. Telomere fingerprinting
Farman, M., Stajich, J.E., Sentandreu, R., Gonzalez-Prieto, J.M., Kennell, J.C., for assessing chromosome number, isolate typing and recombination in the
Molina, L., Schirawski, J., Mendoza-Mendoza, A., Greilinger, D., Munch, K., entomopathogen Beauveria bassiana. Mycol. Res. 107, 572–580.
Rossel, N., Scherer, M., Vranes, M., Ladendorf, O., Vincon, V., Fuchs, U., Sandrock, Powell, W.A., Kistler, H.C., 1990. In vivo rearrangement of foreign DNA by Fusarium
B., Meng, S., Ho, E.C., Cahill, M.J., Boyce, K.J., Klose, J., Klosterman, S.J., Deelstra, oxysporum produces linear self-replicating plasmids. J. Bacteriol. 172, 3163–
H.J., Ortiz-Castellanos, L., Li, W., Sanchez-Alonso, P., Schreier, P.H., Hauser-Hahn, 3171.
I., Vaupel, M., Koopmann, E., Friedrich, G., Voss, H., Schluter, T., Margolis, J., Platt, Rachidi, N., Martinez, M.J., Barre, P., Blondin, B., 2000. Saccharomyces cerevisiae PAU
D., Swimmer, C., Gnirke, A., Chen, F., Vysotskaia, V., Mannhaupt, G., Guldener, U., genes are induced by anaerobiosis. Mol. Microbiol. 35, 1421–1430.
Munsterkotter, M., Haase, D., Oesterheld, M., Mewes, H.W., Mauceli, E.W., Reddel, R.R., 2003. Alternative lengthening of telomeres, telomerase, and cancer.
DeCaprio, D., Wade, C.M., Butler, J., Young, S., Jaffe, D.B., Calvo, S., Nusbaum, C., Cancer Lett. 194, 155–162.
Galagan, J., Birren, B.W., . Insights from the genome of the biotrophic fungal Rehmeyer, C., Li, W., Kusaba, M., Kim, Y.S., Brown, D., Staben, C., Dean, R., Farman,
plant pathogen Ustilago maydis. Nature 444, 97–101. M., 2006. Organization of chromosome ends in the rice blast fungus,
Karlseder, J., 2006. Telomeric proteins: clearing the way for the replication fork. Nat. Magnaporthe oryzae. Nucleic Acids Res. 34, 4685–4701.
Struct. Mol. Biol. 13, 386–387. Ribes-Zamora, A., Mihalek, I., Lichtarge, O., Bertuch, A.A., 2007. Distinct faces of the
Keely, S.P., Renauld, H., Wakefield, A.E., Cushion, M.T., Smulian, A.G., Fosker, N., Ku heterodimer mediate DNA repair and telomeric functions. Nat. Struct. Mol.
Fraser, A., Harris, D., Murphy, L., Price, C., Quail, M.A., Seeger, K., Sharp, S., Tindal, Biol. 14, 301–307.
C.J., Warren, T., Zuiderwijk, E., Barrell, B.G., Stringer, J.R., Hall, N., 2005. Gene Sabourin, M., Tuzon, C.T., Zakian, V.A., 2007. Telomerase and Tel1p preferentially
arrays at Pneumocystis carinii telomeres. Genetics 170, 1589–1600. associate with short telomeres in S. cerevisiae. Mol. Cell. 27, 550–561.
Kim, N.W., Piatyszek, M.A., Prowse, K.R., Harley, C.B., West, M.D., Ho, P.L., Coviello, Sanchez-Alonso, P., Guzman, P., 1998. Organization of chromosome ends in
G.M., Wright, W.E., Weinrich, S.L., Shay, J.W., 1994. Specific association of Ustilago maydis. RecQ-like helicase motifs at telomeric regions. Genetics
human telomerase activity with immortal cells and cancer. Science 266, 2011– 148, 1043–1054.
2015. Schechtman, M.G., 1990. Characterization of telomere DNA from Neurospora crassa.
Kusumoto, K.I., Suzuki, S., Kashiwagi, Y., 2003. Telomeric repeat sequence of Gene 88, 159–165.
Aspergillus oryzae consists of dodeca-nucleotides. Appl. Microbiol. Biotechnol. Shampay, J., Szostak, J.W., Blackburn, E.H., 1984. DNA sequences of telomeres
61, 247–251. maintained in yeast. Nature 310, 154–157.
Larrivee, M., LeBel, C., Wellinger, R.J., 2004. The generation of proper constitutive G- Sharpless, N.E., DePinho, R.A., 2007. Cancer biology: gone but not forgotten. Nature
tails on yeast telomeres is dependent on the MRX complex. Genes Dev. 18, 445, 606–607.
1391–1396. Sinclair, C.S., Richmond, R.H., Ostrander, G.K., 2007. Characterization of the telomere
Lavin, M.F., Kozlov, S., 2007. ATM activation and DNA damage response. Cell Cycle 6, regions of scleractinian coral, Acropora surculosa. Genetica 129, 227–233.
931–942. Smogorzewska, A., de Lange, T., 2004. Regulation of telomerase by telomeric
Lee, A.Y., Liu, E., Wu, X., 2007. The Mre11/Rad50/Nbs1 complex plays an important proteins. Annu. Rev. Biochem. 73, 177–208.
role in the prevention of DNA rereplication in mammalian cells. J. Biol. Chem. Teunissen, A.W., Steensma, H.Y., 1995. Review: the dominant flocculation genes of
282, 32243–32255. Saccharomyces cerevisiae constitute a new subtelomeric gene family. Yeast 11,
Lendvay, T.S., Morris, D.K., Sah, J., Balasubramanian, B., Lundblad, V., 1996. 1001–1013.
Senescence mutants of Saccharomyces cerevisiae with a defect in telomere Tomita, K., Kibe, T., Kang, H.Y., Seo, Y.S., Uritani, M., Ushimaru, T., Ueno, M., 2004.
replication identify three additional EST genes. Genetics 144, 1399–1412. Fission yeast Dna2 is required for generation of the telomeric single-strand
Levis, C., Giraud, T., Dutertre, M., Fortini, D., Brygoo, Y., 1997. Telomeric DNA of overhang. Mol. Cell. Biol. 24, 9557–9567.
Botrytis cinerea: a useful tool for strain identification. FEMS Microbiol. Lett. 157, Traut, W., Szczepanowski, M., Vitkova, M., Opitz, C., Marec, F., Zrzavy, J., 2007. The
267–272. telomere repeat motif of basal Metazoa. Chromosome Res. 15, 371–382.
Li, B., Navarro, S., Kasahara, N., Comai, L., 2004. Identification and biochemical Tseng, S.F., Lin, J.J., Teng, S.C., 2006. The telomerase-recruitment domain of the
characterization of a Werner’s syndrome protein complex with Ku70/80 and telomere binding protein Cdc13 is regulated by Mec1p/Tel1p- dependent
poly(ADP-ribose) polymerase-1. J. Biol. Chem. 279, 13659–13667. phosphorylation. Nucleic Acids Res. 34, 6327–6336.
Li, W., Rehmeyer, C.J., Staben, C., Farman, M.L., 2005. TERMINUS—Telomeric End- Underwood, A.P., Louis, E.J., Borts, R.H., Stringer, J.R., Wakefield, A.E., 1996.
Read Mining IN Unassembled Sequences. Bioinformatics 21, 1695–1698. Pneumocystis carinii telomere repeats are composed of TTAGGG and the
Lingner, J., Hughes, T.R., Shevchenko, A., Mann, M., Lundblad, V., Cech, T.R., 1997. subtelomericsequence contains a gene encoding the major surface glycoprotein.
Reverse transcriptase motifs in the catalytic subunit of telomerase. Exp. Cell Mol. Microbiol. 19, 273–281.
Res. 294, 1–8. van het Hoog, M., Rast, T.J., Martchenko, M., Grindle, S., Dignard, D., Hogues, H.,
Liu, L., DiGirolamo, C.M., Navarro, P.A., Blasco, M.A., Keefe, D.L., 2004. Telomerase Cuomo, C., Berriman, M., Scherer, S., Magee, B.B., Whiteway, M., Chibana, H.,
deficiency impairs differentiation of mesenchymal stem cells. Exp. Cell Res. 294, Nantel, A., Magee, P.T., 2007. Assembly of the Candida albicans genome into
1–8. sixteen supercontigs aligned on the eight chromosomes. Genome Biol. 8, R52.
Long, D.M., Smidansky, E.D., Archer, A.J., Strobel, G.A., 1998. In vivo addition of Vaziri, H., Benchimol, S., 1998. Reconstitution of telomerase activity in normal
telomeric repeats to foreign DNA generates extrachromosomal DNAs in the human cells leads to elongation of telomeres and extended replicative life span.
taxol-producing fungus Pestalotiopsis microspora. Fungal Genet. Biol. 24, 335– Curr. Biol. 8, 279–282.
344. Villasante, A., Abad, J.P., Planello, R., Mendez-Lago, M., Celniker, S.E., de Pablos, B.,
Mandell, J.G., Goodrich, K.J., Bahler, J., Cech, T.R., 2005. Expression of a RecQ helicase 2007. Drosophila telomeric retrotransposons derived from an ancestral element
homolog affects progression through crisis in fission yeast lacking telomerase. J. that was recruited to replace telomerase. Genome Res. 17, 1909–1918.
Biol. Chem. 280, 5249–5257. Wang, F., Podell, E.R., Zaug, A.J., Yang, Y., Baciu, P., Cech, T.R., Lei, M., 2007. The
Matsumoto, T., Fukui, K., Niwa, O., Sugawara, N., Szostak, J.W., Yanagida, M., 1987. POT1-TPP1 telomere complex is a telomerase processivity factor. Nature 445,
Identification of healed terminal DNA fragments in linear minichromosomes of 506–510.
Schizosaccharomyces pombe. Mol. Cell. Biol. 7, 4424–4430. Wright, W.E., Piatyszek, M.A., Rainey, W.E., Byrd, W., Shay, J.W., 1996. Telomerase
McEachern, M.J., Blackburn, E.H., 1995. Runaway telomere elongation caused by activity in human germline and embryonic tissues and cells. Dev. Genet. 18,
telomerase RNA gene mutations. Nature 376, 403–409. 173–179.
McEachern, M.J., Hicks, J.B., 1993. Unusually large telomeric repeats in the yeast Xin, H., Liu, D., Wan, M., Safari, A., Kim, H., Sun, W., O’Connor, M.S., Songyang, Z.,
Candida albicans. Mol. Cell. Biol. 13, 551–560. 2007. TPP1 is a homologue of ciliate TEBP-beta and interacts with POT1
Miller, K.M., Rog, O., Cooper, J.P., 2006. Semi-conservative DNA replication through torecruit telomerase. Nature 445, 559–562.
telomeres requires Taz1. Nature 440, 824–828. Ye, J.Z., de Lange, T., 2004. TIN2 is a tankyrase 1 PARP modulator in the TRF1
Multani, A.S., Chang, S., 2007. WRN at telomeres: implications for aging and cancer. telomere length control complex. Nat. Genet. 36, 618–623.
J. Cell Sci. 120, 713–721. Zakian, V.A., Blanton, H.M., 1988. Distribution of telomere-associated sequences
North, B.J., Verdin, E., 2004. Sirtuins: Sir2-related NAD-dependent protein on natural chromosomes in Saccharomyces cerevisiae. Mol. Cell. Biol. 8,
deacetylases. Genome Biol. 5, 224. 2257–2260.
Olson, E., Nievera, C.J., Liu, E., Lee, A.Y., Chen, L., Wu, X., 2007. The Mre11 complex Zhong, Z.H., Jiang, W.Q., Cesare, A.J., Neumann, A.A., Wadhwa, R., Reddel, R.R., 2007.
mediates the S-phase checkpoint through an interaction with replication Disruption of telomere maintenance by depletion of the MRE11/RAD50/NBS1
protein A. Mol. Cell. Biol. 27, 6053–6067. complex in cells that use alternative lengthening of telomeres. J. Biol. Chem.
Opresko, P.L., Cheng, W.H., Bohr, V.A., 2004. Junction of RecQ helicase biochemistry 282, 29314–29322.
and human disease. J. Biol. Chem. 279, 18099–18102. Zhu, X.D., Kuster, B., Mann, M., Petrini, J.H., de Lange, T., 2000. Cell-cycle- regulated
Ottaviani, A., Gilson, E., Magdinier, F., 2008. Telomeric position effect: from the association of RAD50/MRE11/NBS1 with TRF2 and human telomeres. Nat.
yeast paradigm to human pathologies? Biochimie 90, 93–107. Genet. 25, 347–352.

Vous aimerez peut-être aussi