Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-24T16:57:20.174Z Has data issue: false hasContentIssue false

Diverse telomeres in trypanosomatids

Published online by Cambridge University Press:  22 February 2021

Ester Poláková
Affiliation:
Life Science Research Centre, Faculty of Science, University of Ostrava, 710 00Ostrava, Czech Republic
Kristína Záhonová
Affiliation:
Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05České Budějovice (Budweis), Czech Republic Faculty of Science, Charles University, BIOCEV, 252 50Vestec, Czech Republic
Amanda T. S. Albanaz
Affiliation:
Life Science Research Centre, Faculty of Science, University of Ostrava, 710 00Ostrava, Czech Republic
Anzhelika Butenko
Affiliation:
Life Science Research Centre, Faculty of Science, University of Ostrava, 710 00Ostrava, Czech Republic Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05České Budějovice (Budweis), Czech Republic
Julius Lukeš
Affiliation:
Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05České Budějovice (Budweis), Czech Republic Faculty of Sciences, University of South Bohemia, 370 05České Budějovice (Budweis), Czech Republic
Vyacheslav Yurchenko*
Affiliation:
Life Science Research Centre, Faculty of Science, University of Ostrava, 710 00Ostrava, Czech Republic Martsinovsky Institute of Medical Parasitology, Tropical and Vector Borne Diseases, Sechenov University, Moscow119435, Russia
*
Author for correspondence: Vyacheslav Yurchenko, E-mail: vyacheslav.yurchenko@osu.cz

Abstract

Telomeres are the ends of linear eukaryotic chromosomes facilitating the resolution of the ‘end replication and protection’ problems, associated with linearity. At the nucleotide level, telomeres typically represent stretches of tandemly arranged telomeric repeats, which vary in length and sequence among different groups of organisms. Recently, a composition of the telomere-associated protein complex has been scrutinized in Trypanosoma brucei. In this work, we subjected proteins from that list to a more detailed bioinformatic analysis and delineated a core set of 20 conserved proteins putatively associated with telomeres in trypanosomatids. Out of these, two proteins (Ku70 and Ku80) are conspicuously missing in representatives of the genus Blastocrithidia, yet telomeres in these species do not appear to be affected. In this work, based on the analysis of a large set of trypanosomatids widely different in their phylogenetic position and life strategies, we demonstrated that telomeres of trypanosomatids are diverse in length, even within groups of closely related species. Our analysis showed that the expression of two proteins predicted to be associated with telomeres (those encoding telomerase and telomere-associated hypothetical protein orthologous to Tb927.6.4330) may directly affect and account for the differences in telomere length within the species of the Leishmania mexicana complex.

Type
Research Article
Creative Commons
Creative Common License - CCCreative Common License - BYCreative Common License - NCCreative Common License - SA
This is an Open Access article, distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike licence (http://creativecommons.org/licenses/by-nc-sa/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the same Creative Commons licence is included and the original work is properly cited. The written permission of Cambridge University Press must be obtained for commercial re-use.
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press

Introduction

Trypanosomatidae is a family of protozoan parasites possessing a single large mitochondrion, which encompasses a network of catenated circular DNA molecules, the so-called kinetoplast or kDNA (Maslov et al., Reference Maslov, Opperdoes, Kostygov, Hashimi, Lukeš and Yurchenko2019). These species have been attracting research attention because of numerous unique or rare biochemical and molecular traits, such as trans-splicing and polycistronic transcription (Clayton, Reference Clayton2019; Michaeli, Reference Michaeli2011), mitochondrial RNA editing (Aphasizheva et al., Reference Aphasizheva, Alfonzo, Carnes, Cestari, Cruz-Reyes, Goringer, Hajduk, Lukeš, Madison-Antenucci, Maslov, McDermott, Ochsenreiter, Read, Salavati, Schnaufer, Schneider, Simpson, Stuart, Yurchenko, Zhou, Zíková, Zhang, Zimmer and Aphasizhev2020), presence of modified nucleotides (van Luenen et al., Reference van Luenen, Farris, Jan, Genest, Tripathi, Velds, Kerkhoven, Nieuwland, Haydock, Ramasamy, Vainio, Heidebrecht, Perrakis, Pagie, van Steensel, Myler and Borst2012) and unusual organelles (Szöör et al., Reference Szöör, Haanstra, Gualdrón-López and Michels2014; Docampo, Reference Docampo2016), or a bizarre variation of the nuclear genetic code (Záhonová et al., Reference Záhonová, Kostygov, Ševčíková, Yurchenko and Eliáš2016). Most of these flagellates are monoxenous (with one host in their life cycle) parasites restricted to invertebrates (Maslov et al., Reference Maslov, Votýpka, Yurchenko and Lukeš2013), while members of the genera Endotrypanum, Leishmania, Phytomonas, Porcisia and Trypanosoma have switched to dixeny (two-host life cycle) and infect vertebrates or plants in addition to invertebrates (Lukeš et al., Reference Lukeš, Butenko, Hashimi, Maslov, Votýpka and Yurchenko2018). It is established beyond a reasonable doubt that the dixenous species have evolved from the monoxenous ancestor(s) independently several times (Lukeš et al., Reference Lukeš, Skalický, Týč, Votýpka and Yurchenko2014). Notably, several Leishmania and Trypanosoma spp. are of medical importance, as they cause severe human diseases, and are fairly well-studied (Stuart et al., Reference Stuart, Brun, Croft, Fairlamb, Gurtler, McKerrow, Reed and Tarleton2008; Nussbaum et al., Reference Nussbaum, Honek, Cadmus and Efferth2010).

Telomeres typically represent repetitive physical ends of linear eukaryotic chromosomes, variable in length and sequence in different groups of organisms (Fulnečková et al., Reference Fulnečková, Ševčíková, Fajkus, Lukešová, Lukeš, Vlček, Lang, Kim, Eliáš and Sýkorová2013). Their main role is to protect chromosome ends from being recognized and processed as DNA double-strand breaks by the cellular repair machinery in order to prevent chromosomal end-to-end fusions (Pfeiffer and Lingner, Reference Pfeiffer and Lingner2013). Such shielding is provided by the telomere-associated protein complexes (Lewis and Wuttke, Reference Lewis and Wuttke2012) or specific complementary DNA structures, such as telomere loops (t-loops) facilitating the protection of chromosome ends (Tomáška et al., Reference Tomáška, Nosek, Kar, Willcox and Griffith2019). It is generally assumed that telomeres undergo gradual shortening with each round of cell division because of incomplete lagging strand synthesis of linear DNA templates by DNA polymerases, known as the ‘end replication problem’ (Olovnikov, Reference Olovnikov1973; Greider, Reference Greider1990; Hackett and Greider, Reference Hackett and Greider2002). In order to overcome this problem and, thus, prevent telomere shortening, cells engage a dedicated enzyme called telomerase (Greider and Blackburn, Reference Greider and Blackburn1985).

Telomeres of kinetoplastids share many traits with those of other eukaryotes. They have the canonical sequence (5′-ttaggg-3′) found in vertebrates, end with a t-loop, are associated with capping protein complexes and maintained by telomerases (Muñoz-Jordán et al., Reference Muñoz-Jordán, Cross, de Lange and Griffith2001; Conte and Cano, Reference Conte and Cano2005; Fulnečková et al., Reference Fulnečková, Ševčíková, Fajkus, Lukešová, Lukeš, Vlček, Lang, Kim, Eliáš and Sýkorová2013). Similar to the situation in other eukaryotic pathogens, genes encoding trypanosomatid virulence factors are often located in the sub-telomeric regions and their expression may be co-regulated with telomeres (Chiurillo et al., Reference Chiurillo, Cano, Da Silveira and Ramirez1999; Dobson et al., Reference Dobson, Scholtes, Myler, Turco and Beverley2006; Hovel-Miner et al., Reference Hovel-Miner, Boothroyd, Mugnier, Dreesen, Cross and Papavasiliou2012). Moreover, transposable elements are often found in association with telomeres (Pardue et al., Reference Pardue, Danilevskaya, Traverse and Lowenhaupt1997; Rahnama et al., Reference Rahnama, Novikova, Starnes, Zhang, Chen and Farman2020). In agreement with this, a sub-telomeric region of Leptomonas pyrrhocoris chromosome contains an integrated copy of an RNA-dependent RNA polymerase putatively originating from an RNA virus of the family Tombusviridae infecting this flagellate (Grybchuk et al., Reference Grybchuk, Akopyants, Kostygov, Konovalovas, Lye, Dobson, Zangger, Fasel, Butenko, Frolov, Votýpka, d'Avila-Levy, Kulich, Moravcová, Plevka, Rogozin, Serva, Lukeš, Beverley and Yurchenko2018), and possibly contributing to the retrotransposon translocation within the trypanosomatid genome. Telomeric regions of kinetoplastid chromosomes also possess several features distinguishing them from their counterpart in most of the other eukaryotes. For example, the telomeres of Trypanosoma brucei increase in length (by approximately 10 bp per generation) until they reach an equilibrium (Bernards et al., Reference Bernards, Michels, Lincke and Borst1983; Pays et al., Reference Pays, Laurent, Delinte, Van Meirvenne and Steinert1983; Horn et al., Reference Horn, Spence and Ingram2000). In trypanosomatids, a modified nucleobase, base J (β-D-gluco-pyranosyl-oxy-methyl-uracil) is involved in RNA polymerase II transcription termination and is preferentially localized to telomeres (Borst and van Leeuwen, Reference Borst and van Leeuwen1997; Genest et al., Reference Genest, Ter Riet, Cijsouw, van Luenen and Borst2007; van Luenen et al., Reference van Luenen, Farris, Jan, Genest, Tripathi, Velds, Kerkhoven, Nieuwland, Haydock, Ramasamy, Vainio, Heidebrecht, Perrakis, Pagie, van Steensel, Myler and Borst2012).

To the best of our knowledge, there has been very little systematic effort to analyse telomeres in trypanosomatids outside the medically relevant Trypanosoma and Leishmania spp. (Fu et al., Reference Fu, Perona-Wright and Barker1998; Fu and Barker, Reference Fu and Barker1998a, Reference Fu and Barker1998b; Chiurillo et al., Reference Chiurillo, Cano, Da Silveira and Ramirez1999, Reference Chiurillo, Peralta and Ramirez2002; Muñoz-Jordán et al., Reference Muñoz-Jordán, Cross, de Lange and Griffith2001; Janzen et al., Reference Janzen, Lander, Dreesen and Cross2004; Conte and Cano, Reference Conte and Cano2005; Genest and Borst, Reference Genest and Borst2007). Therefore, we decided to do that for a wide range of trypanosomatids with a special emphasis on largely neglected parasites of insects, which are not pathogenic to humans. We selected more than 20 proteins from a set of recently defined putative trypanosomatid telomere-associated proteins (Reis et al., Reference Reis, Schwebs, Dietz, Janzen and Butter2018) for more detailed in silico analyses. For most of these proteins, some functional information is available [Table 1; the TriTrypDB (Aslett et al., Reference Aslett, Aurrecoechea, Berriman, Brestelli, Brunk, Carrington, Depledge, Fischer, Gajria, Gao, Gardner, Gingle, Grant, Harb, Heiges, Hertz-Fowler, Houston, Innamorato, Iodice, Kissinger, Kraemer, Li, Logan, Miller, Mitra, Myler, Nayak, Pennington, Phan, Pinney, Ramasamy, Rogers, Roos, Ross, Sivam, Smith, Srinivasamoorthy, Stoeckert, Subramanian, Thibodeau, Tivey, Treatman, Velarde and Wang2010) gene IDs are used throughout the text]. The predicted telomere-associated complex appears to be a cohort of proteins with widely variable functions, from ribosome and proteasome subunits to telomerase and even DNA repair proteins (Boulton and Jackson, Reference Boulton and Jackson1998; Paugam et al., Reference Paugam, Bulteau, Dupouy-Camet, Creuzet and Friguet2003; Riha and Shippen, Reference Riha and Shippen2003; Janzen et al., Reference Janzen, Lander, Dreesen and Cross2004; Dreesen et al., Reference Dreesen, Li and Cross2005; Ruvinsky and Meyuhas, Reference Ruvinsky and Meyuhas2006; Chico et al., Reference Chico, Ciudad, Hsu, Lue and Larriba2011; Sandhu et al., Reference Sandhu, Sanford, Basu, Park, Pandya, Li and Chakrabarti2013; Nenarokova et al., Reference Nenarokova, Záhonová, Krasilnikova, Gahura, McCulloch, Ziková, Yurchenko and Lukeš2019). In this work, we analysed the evolutionary history of telomere-associated proteins in Kinetoplastea, performed a systematic analysis of telomere length variation among trypanosomatids on the dataset, which incorporates a wide range of understudied monoxenous members of the family Trypanosomatidae, and established a correlation between the level of transcription for several analysed telomere-associated proteins and the telomere length.

Table 1. Predicted telomere-associated protein complex composition in T. brucei

Materials and methods

In silico analyses

A putative set of telomere-associated proteins of T. brucei brucei TREU927 (Reis et al., Reference Reis, Schwebs, Dietz, Janzen and Butter2018) were used as queries for BLAST searches (Altschul et al., Reference Altschul, Gish, Miller, Myers and Lipman1990) against a dataset of annotated proteins of 64 trypanosomatids and the eubodonid Bodo saltans. First, BLASTp searches were performed with an E-value set to 1 and all the hits with an E-value not exceeding 10−15 were retained. If the respective sequence was not identified among annotated proteins, the searches were repeated with the tBLASTn algorithm against a database of genome sequences. In case no protein was identified in the genome, HMMER v.3.3 (Eddy, Reference Eddy2009), a more sensitive method for the identification of divergent homologues based on hidden Markov models was employed. Annotated proteins and assembled genome sequences were downloaded from the NCBI Genome (Sayers et al., Reference Sayers, Agarwala, Bolton, Brister, Canese, Clark, Connor, Fiorini, Funk, Hefferon, Holmes, Kim, Kimchi, Kitts, Lathrop, Lu, Madden, Marchler-Bauer, Phan, Schneider, Schoch, Pruitt and Ostell2019) and TriTrypDB v. 45/46 (Aslett et al., Reference Aslett, Aurrecoechea, Berriman, Brestelli, Brunk, Carrington, Depledge, Fischer, Gajria, Gao, Gardner, Gingle, Grant, Harb, Heiges, Hertz-Fowler, Houston, Innamorato, Iodice, Kissinger, Kraemer, Li, Logan, Miller, Mitra, Myler, Nayak, Pennington, Phan, Pinney, Ramasamy, Rogers, Roos, Ross, Sivam, Smith, Srinivasamoorthy, Stoeckert, Subramanian, Thibodeau, Tivey, Treatman, Velarde and Wang2010) databases. The validity of the hits was confirmed using reciprocal BLAST searches against T. brucei proteins and alignments including the query and all identified proteins, if necessary. The resulting gene presence/absence table and a cladogram manually written in a Newick format based on recent publications (Butenko et al., Reference Butenko, Kostygov, Sádlová, Kleschenko, Bečvář, Podešvová, Macedo, Žihala, Lukeš, Bates, Volf, Opperdoes and Yurchenko2019; Kostygov et al., Reference Kostygov, Grybchuk-Ieremenko, Malysheva, Frolov and Yurchenko2014, Reference Kostygov, Frolov, Malysheva, Ganyukova, Chistyakova, Tashyreva, Tesařová, Spodareva, Režnarová, Macedo, Butenko, d'Avila-Levy, Lukeš and Yurchenko2020; Kostygov and Yurchenko, Reference Kostygov and Yurchenko2017; Lukeš et al., Reference Lukeš, Butenko, Hashimi, Maslov, Votýpka and Yurchenko2018; Frolov et al., Reference Frolov, Malysheva, Ganyukova, Spodareva, Yurchenko and Kostygov2019; Kato et al., Reference Kato, Caceres, Seki, Silupu Garcia, Holguin Mauricci, Castro Martinez, Moreno Paico, Castro Muniz, Troyes Rivera, Villegas Briones, Guerrero Quincho, Sulca Jayo, Tineo Villafuerte, Manrique de Lara Estrada, Arias, Passara, Ruelas Llerena, Kubo, Tabbabi, Yamamoto and Hashiguchi2019) were used for Dollo parsimony analysis in the Count software (Csűrös, Reference Csűrös2010) and results were visualized in a graphical editor.

Trypanosomatid isolates and cultivation

Cultures of Crithidia expoeki (BJ08.175), C. fasciculata (Cf-Cl), C. fasciculata (ATCC20358), C. termophilla (ATCC30817), L. pyrrhocoris (H10), L. seymouri (ATCC30220), Novymonas esmeraldas (E262AT), Strigomonas oncopelti (TCC290E) and Zelonia costaricensis (15EC) were grown in BHI medium (Oxoid/Thermo Fisher Scientific, Basingstoke, UK) supplemented with 2 μg mL−1 Hemin (Sigma-Aldrich, St. Louis, USA) and 50 units mL−1 of Penicillin/Streptomycin (BioSera, Nuaillé, France) at 23°C. Cultures of Endotrypanum monterogeii (ATCC30507), Herpetomonas samuelpessoai, Leishmania (Leishmania) major (LV39), L. (L.) amazonensis (Josefa, LV78, LV79 and PH8), L. (L.) mexicana (M379), L. (Mundinia) martiniquensis (LEM2494), L. (M.) orientalis (PCM2), L. (Sauroleishmania) tarentolae (ParrotTarII), L. (Viannia) braziliensis (human2017), L. (V.) guyanensis (M4147), Phytomonas sp. (EM1), Porcisia deanei (TCC258) and P. hertigi (TCC260) were grown in M199 medium supplemented with 2 μg mL−1 Biopterin, 2 μg mL−1 Hemin (all Sigma-Aldrich), 25 mm HEPES (Lonza, Basel, Switzerland), 50 units mL−1 of Penicillin/Streptomycin (BioSera) and 10% fetal bovine serum (BioWest, Nuaillé, France) at 23°C. Cultures of Angomonas deanei (CT-IOC 044), A. desouzai (CT-IOC 109), Blastocrithidia sp. (p57), Blastocrithidia triatomae, Blechomonas ayalai (B08-376), Blechomonas pulexsimulantis (ATCC50186), Herpetomonas muscarum (MMO-01), Jaenimonas sp. (Finn-01.02), Kentomonas sorsogonicus (MF-08.02), Lafontella sp. (GMO-01), Vickermania ingenoplastis (CP21), Wallacemonas collosoma (ATCC30261) and W. rigidus (PL11) were maintained in SDM medium (BioWest) supplemented with 10% fetal bovine serum (BioWest) and 50 units mL−1 of Penicillin/Streptomycin (BioSera) at 23°C. In the cases of Lafontella and Endotrypanum, cultures were grown in a bi-phasic medium, overlaying blood agar. All species were validated by amplifying and sequencing the 18S rRNA gene as described previously (Kostygov et al., Reference Kostygov, Grybchuk-Ieremenko, Malysheva, Frolov and Yurchenko2014).

Quantification of transcription level of genes encoding telomeric proteins using RT-qPCR

RNA was isolated and transcript levels of the telomeric proteins were assessed by RT-qPCR as described previously (Záhonová et al., Reference Záhonová, Hadariová, Vacula, Yurchenko, Eliáš, Krajčovič and Vesteg2014; Kraeva et al., Reference Kraeva, Leštinová, Ishemgulova, Majerová, Butenko, Vaselek, Bespyatykh, Charyyeva, Spitzová, Kostygov, Lukeš, Volf, Votýpka and Yurchenko2019). Sequences of the specific primers for L. mexicana/amazonensis orthologues of T. brucei genes are listed in Supplementary Table 1. Expression values were normalized to those of 18S rRNA.

Southern blotting

The previously established terminal restriction fragment analysis of telomere lengths protocol was followed (Janzen et al., Reference Janzen, Lander, Dreesen and Cross2004). In brief, total genomic DNA from the log-phase grown cells was isolated and digested with AluI, HinfI and RsaI overnight. Restriction fragments were separated in 0.75% agarose gel, transferred to a ZetaProbe blotting membrane (Bio-Rad, Hercules, USA), probed with the DIG-labelled telomeric probe [CCCTAA]x25 in the DIG Easy Hyb buffer (Roche Diagnostics, Indianapolis, USA), and visualized with the DIG Luminescent Detection Kit (Roche Diagnostics). The probe was labelled by the Dioxigenin NT Labeling Kit (Jena Bioscience GmbH, Jena, Germany). Statistics of the telomere lengths were obtained with an online tool WALTER (Web-based Analyser of the Length of TElomeRes) (Lyčka et al., Reference Lyčka, Peška, Demko, Spyroglou, Kilar, Fajkus and Fojtová2021). For the loading and integrity control in the L. mexicana complex analysis, DNAs were processed as above, and the membrane was probed against a fragment of a gene encoding telomerase (LmxM.36.3930) (Supplementary Table 1).

Results and discussion

The core set of proteins putatively involved in telomere maintenance in kinetoplastids is conserved

To study the phylogenetic distribution of proteins predicted to be involved in telomere maintenance (Reis et al., Reference Reis, Schwebs, Dietz, Janzen and Butter2018), we analysed the presence/absence of the corresponding 24 genes in the available genomes of trypanosomatids and their close eubodonid relative, B. saltans (Table 2). Most of the studied proteins (20 of 24) are well conserved and we consider them as a core set putatively involved in telomere maintenance in kinetoplastids. It is worth noting that the telomere association and function in telomere maintenance has already been confirmed for some of these proteins, while some others have not been functionally characterized yet. Thus, the composition of the core set of proteins involved in telomere maintenance, as defined previously (Reis et al., Reference Reis, Schwebs, Dietz, Janzen and Butter2018) and discussed herein, should be taken with caution. Despite the fact that most of the respective genes are conserved across Kinetoplastea (Fig. 1, Table 2) and, thus appear to be present in the kinetoplastid common ancestor, we came across several interesting exceptions that are discussed in detail below.

Fig. 1. Gains and losses of genes encoding putative telomere-associated proteins in kinetoplastids.

Table 2. Presence of genes putatively involved in telomere maintenance in kinetoplastids

Species analysed by Southern blotting are shaded.

+, identified; empty, not identified; *, identified in strain UA301.

A set of three proteins (orthologues of T. brucei Tb927.3.1560, Tb927.9.5020 and Tb927.11.370) was acquired by the common ancestor of trypanosomatids upon the separation from bodonids (Fig. 1). One of them, Tb927.3.1560 [TIF-2, an orthologue of mammalian TINF2 (Jehi et al., Reference Jehi, Li, Sandhu, Ye, Benmerzouga, Zhang, Zhao and Li2014a; Reference Jehi, Wu and Li2014b)] was suggested to be essential, as it is involved in shelterin (a protein complex implicated in telomere protection) assembly and telomerase-mediated telomere length maintenance in other organisms (Walne et al., Reference Walne, Vulliamy, Beswick, Kirwan and Dokal2008; Frank et al., Reference Frank, Tran, Qu, Stohr, Segal and Xu2015). Yet, it is not present in bodonids and was secondarily lost in all other trypanosomatids outside of the genera Paratrypanosoma, Trypanosoma and Blechomonas (Fig. 1), raising a question of how do they cope with its absence or whether they replaced it with a functional analogue? Tb927.2.6100 is Trypanosoma-specific, confirming previous report (Beck et al., Reference Beck, Acestor, Schulfer, Anupama, Carnes, Panigrahi and Stuart2013). Surprisingly, this protein was shown to be specifically associated with kDNA, so its role in telomere maintenance, if any, remains to be elucidated by functional genetics approaches. Two proteins (orthologues of T. brucei Tb927.9.3930 and Tb927.9.4000) are present only in four species of the T. brucei group and may determine specific traits of these parasites.

An orthologue of Tb927.11.9870 (TelAP-1) is present in most species, but it is conspicuously absent from the representatives of two monoxenous groups (Blastocrithidia and Vickermania spp.) and most Phytomonas spp., plant pathogens with streamlined genomes (Porcel et al., Reference Porcel, Denoeud, Opperdoes, Noel, Madoui, Hammarton, Field, Da Silva, Couloux, Poulain, Katinka, Jabbari, Aury, Campbell, Cintron, Dickens, Docampo, Sturm, Koumandou, Fabre, Flegontov, Lukeš, Michaeli, Mottram, Szoor, Zilberstein, Bringaud, W and Dollet2014). While we cannot rule out a possibility that the protein is divergent beyond recognition by available bioinformatics tools, there may exist another component fulfilling the role of TelAP-1 in these species.

Of special attention is the absence of Tb927.3.5030 (Ku70) and Tb927.3.5030 (Ku80) orthologues in Blastocrithidia sp., which has a non-canonical nuclear genetic code with all three stop codons reassigned to encode amino acids (Záhonová et al., Reference Záhonová, Kostygov, Ševčíková, Yurchenko and Eliáš2016). It has been recently proposed that such an absence may lead to the accumulation of numerous insertions in many protein-coding genes of these organisms (Nenarokova et al., Reference Nenarokova, Záhonová, Krasilnikova, Gahura, McCulloch, Ziková, Yurchenko and Lukeš2019).

Trypanosomatid telomeres are variable in length

We performed a systematic screen of the telomere length across Trypanosomatidae by Southern blotting (Fig. 2, Table 3). Our analysis revealed that monoxenous Leishmaniinae (Kostygov and Yurchenko, Reference Kostygov and Yurchenko2017) of the genera Leptomonas, Novymonas and Zelonia have fairly short telomeres (weighted medians 900–1200 bp; hereafter only rounded weighted median data are compared in the text, see Table 3 for minimum and maximum values), while telomeres in analysed Crithidia spp. ranged from 400 bp in C. fasciculata Cf-C1 to 4300 bp in C. expoeki. These numbers correlate well with previous reports on telomere length in the selected representatives of the genera Crithidia, Leishmania and Trypanosoma (Genest et al., Reference Genest, Ter Riet, Cijsouw, van Luenen and Borst2007). Of note, the repertoire of genes implicated in telomere maintenance is identical in these flagellates (Table 2), so these differences can be explained by either the presence of other proteins involved in this process, or (more likely) differences in gene expression. Telomeres of Blechomonas, Herpetomonas, Jaenimonas and Wallacemonas spp. are 1300–2100 bp long. The endosymbiont-containing Strigomonadinae [Angomonas, Kentomonas and Strigomonas spp. (Votýpka et al., Reference Votýpka, Kostygov, Kraeva, Grybchuk-Ieremenko, Tesařová, Grybchuk, Lukeš and Yurchenko2014)] differ in telomere length, with S. oncopelti bearing the shortest chromosome ends of ~400 bp.

Fig. 2. Southern blotting analysis of telomere repeats in selected species of Trypanosomatidae. Marker sizes are indicated on the left. The vertical lines denote a composite image from the same blot. DNA integrity controls are presented in Supplementary Fig. 1 (left and middle panels).

Table 3. Telomere lengths (weighted median, minimum–maximum) in selected species of Trypanosomatidae

Representatives of three genera (Blastocrithidia, Leishmania and Trypanosoma) deserved special attention. Uniquely among trypanosomatids, Blastocrithidia spp. lack Ku proteins (Nenarokova et al., Reference Nenarokova, Záhonová, Krasilnikova, Gahura, McCulloch, Ziková, Yurchenko and Lukeš2019), yet their telomeres are of similar length to telomeres of other trypanosomatids (600 and 1500 bp in B. triatomae and Blastocrithidia sp., respectively), arguing that either Ku proteins are dispensable for the telomere length maintenance in these species, or their loss can be compensated by other factors. Telomere sizes vary in different Trypanosoma spp. represented by short telomeres in T. mega (400 bp) and substantially longer telomeres in T. brucei Lister 427 (3100–3400 bp). In contrast to the previous report (Dreesen and Cross, Reference Dreesen and Cross2008), we did not document differences in telomeres' length between the procyclic and bloodstream stages of T. brucei. However, both strains in our analysis have the same origin (Lister 427), while the abovementioned study compared Lister 427 and TREU927 strains. Similar to the cases discussed above, despite possessing the same repertoire of telomere-bound proteins, the distribution of telomere sizes in the LeishmaniaPorcisiaEndotrypanum clade (Espinosa et al., Reference Espinosa, Serrano, Camargo, Teixeira and Shaw2018) is wide, exemplified by two extreme cases of P. hertigi (5000 bp) and L. mexicana (400 bp, Fig. 2). Variable telomere length in Leishmania spp. (and possibly other Leishmaniinae) may be explained by the presence of a stress-sensitive telomere-proximal replication activity outside S phase of the cell cycle in these species (Damasceno et al., Reference Damasceno, Marques, Beraldi, Crouch, Lapsley, Obonaga, Tosi and McCulloch2020, Reference Damasceno, Marques, Black, Briggs and McCulloch2021).

RNA level of telomerase and several telomere-associated proteins correlates with telomere length in the species of L. mexicana complex

We analysed telomere length and expression of the core set of proteins putatively involved in telomere maintenance in closely related species forming the L. mexicana complex (Eresh et al., Reference Eresh, McCallum and Barker1994). Similar to the cases discussed above, telomeres in L. mexicana and four isolates of L. amazonensis greatly differed in length from ~400 bp in L. mexicana M379 to ~3400 bp in L. amazonensis LV79 (Fig. 3, Table 3, Supplementary Fig. 1). Such a wide range of telomere lengths correlated well with the expression of the Leishmania spp. telomerase (orthologue of Tb927.11.10190) and a telomere-associated hypothetical protein (orthologue of Tb927.6.4330). The higher expression of these proteins correlated with longer telomeres. The specific roles of these and other proteins remain to be further elucidated by functional studies.

Fig. 3. Transcript levels of telomere-associated proteins and telomere lengths in the species of L. mexicana complex. (A) Quantitative RT-PCR analysis of the core set of proteins implicated in telomere maintenance. Gene expression is presented as normalized means and standard deviations of three replicates. Data are presented in two graphs to account for differences in expression values. (B, C) Southern blotting analysis of telomere repeats (B) and telomerase-encoding gene (C, used as an additional DNA integrity control) in L. amazonensis LV78, LV79, PH8, Josefa and L. mexicana M379. Marker sizes are indicated on the left. DNA integrity controls are presented in Supplementary Fig. 1 (right panel).

Conclusions

The genome analysis has allowed us to identify a core set of 20 conserved proteins predicted to be responsible for telomere maintenance in trypanosomatids. Several proteins, previously identified in T. brucei pull-downs, are trypanosome-specific. Out of 20 proteins conserved in Trypanosomatidae, two (Ku70 and Ku80) are conspicuously missing in Blastocrithidia spp., yet telomeres in these species do not appear to be affected by their loss. We documented that telomeres of trypanosomatids are diverse in length, even within groups of closely related species. One such group is a complex of species, related to L. mexicana. Our analysis demonstrated that the expression of several telomere-associated proteins correlates with the documented differences in telomere length within species of the L. mexicana complex, which is indicative of a potential role these proteins may play in the telomere length maintenance.

Supplementary material

The supplementary material for this article can be found at https://doi.org/10.1017/S0031182021000378.

Acknowledgements

We thank Dr A Kostygov (University of Ostrava) for his advice on trypanosomatid cultivation, Dr P. Volf and Dr J. Sádlová (both Charles University) for providing Leishmania spp. isolates, Dr C. Janzen (University of Würzburg) for sharing the plasmid pSP73 bearing the telomeric probe, and Dr C. Janzen and Dr R. McCulloch (University of Glasgow) for specific advice on methodology.

Financial support

Computational resources were funded by the project ‘e-Infrastruktura CZ’ (e-INFRA LM2018140) provided within the program ‘Projects of Large Research, Development and Innovations Infrastructures’. This work was supported by the Grant Agency of Czech Republic (18-15962S and 20-07186S) and the European Regional Funds (project ‘Centre for Research of Pathogenicity and Virulence of Parasites’ CZ.02.1.01/16_019/0000759) to V.Y. and J.L., and the grant SGS/PrF/2021 from the University of Ostrava (to E.P. and A.T.S.A.). A.T.S.A. and E.P. were also supported by the Moravskoslezský kraji research initiative (RRC/02/2020) and City of Ostrava (0493/ZM1822/8), rescpectively. A part of this work on telomere diversity in Leishmania spp. was funded by the Russian Science Foundation (grant 19-15-00054) to V.Y. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Conflict of interest

None.

Ethical standards

Not applicable.

References

Altschul, SF, Gish, W, Miller, W, Myers, EW and Lipman, DJ (1990) Basic local alignment search tool. Journal of Molecular Biology 215, 403410.CrossRefGoogle ScholarPubMed
Aphasizheva, I, Alfonzo, J, Carnes, J, Cestari, I, Cruz-Reyes, J, Goringer, HU, Hajduk, S, Lukeš, J, Madison-Antenucci, S, Maslov, DA, McDermott, SM, Ochsenreiter, T, Read, LK, Salavati, R, Schnaufer, A, Schneider, A, Simpson, L, Stuart, K, Yurchenko, V, Zhou, ZH, Zíková, A, Zhang, L, Zimmer, S and Aphasizhev, R (2020) Lexis and grammar of mitochondrial RNA processing in trypanosomes. Trends in Parasitology 36, 337355.CrossRefGoogle ScholarPubMed
Aslett, M, Aurrecoechea, C, Berriman, M, Brestelli, J, Brunk, BP, Carrington, M, Depledge, DP, Fischer, S, Gajria, B, Gao, X, Gardner, MJ, Gingle, A, Grant, G, Harb, OS, Heiges, M, Hertz-Fowler, C, Houston, R, Innamorato, F, Iodice, J, Kissinger, JC, Kraemer, E, Li, W, Logan, FJ, Miller, JA, Mitra, S, Myler, PJ, Nayak, V, Pennington, C, Phan, I, Pinney, DF, Ramasamy, G, Rogers, MB, Roos, DS, Ross, C, Sivam, D, Smith, DF, Srinivasamoorthy, G, Stoeckert, CJ Jr., Subramanian, S, Thibodeau, R, Tivey, A, Treatman, C, Velarde, G and Wang, H (2010) TriTrypDB: a functional genomic resource for the Trypanosomatidae. Nucleic Acids Research 38, D457D462.CrossRefGoogle ScholarPubMed
Beck, K, Acestor, N, Schulfer, A, Anupama, A, Carnes, J, Panigrahi, AK and Stuart, K (2013) Trypanosoma brucei Tb927.2.6100 is an essential protein associated with kinetoplast DNA. Eukaryotic Cell 12, 970978.CrossRefGoogle ScholarPubMed
Bernards, A, Michels, PA, Lincke, CR and Borst, P (1983) Growth of chromosome ends in multiplying trypanosomes. Nature 303, 592597.CrossRefGoogle ScholarPubMed
Borst, P and van Leeuwen, F (1997) beta-D-glucosyl-hydroxymethyluracil, a novel base in African trypanosomes and other Kinetoplastida. Molecular and Biochemical Parasitology 90, 18.CrossRefGoogle ScholarPubMed
Boulton, SJ and Jackson, SP (1998) Components of the Ku-dependent non-homologous end-joining pathway are involved in telomeric length maintenance and telomeric silencing. EMBO Journal 17, 18191828.CrossRefGoogle ScholarPubMed
Butenko, A, Kostygov, AY, Sádlová, J, Kleschenko, Y, Bečvář, T, Podešvová, L, Macedo, DH, Žihala, D, Lukeš, J, Bates, PA, Volf, P, Opperdoes, FR and Yurchenko, V (2019) Comparative genomics of Leishmania (Mundinia). BMC Genomics 20, 726.CrossRefGoogle Scholar
Campelo, R, Diaz Lozano, I, Figarella, K, Osuna, A and Ramirez, JL (2015) Leishmania major telomerase TERT protein has a nuclear/mitochondrial eclipsed distribution that is affected by oxidative stress. Infection and Immunity 83, 5766.CrossRefGoogle Scholar
Chico, L, Ciudad, T, Hsu, M, Lue, NF and Larriba, G (2011) The Candida albicans Ku70 modulates telomere length and structure by regulating both telomerase and recombination. PLoS ONE 6, e23732.CrossRefGoogle Scholar
Chiurillo, MA, Cano, I, Da Silveira, JF and Ramirez, JL (1999) Organization of telomeric and sub-telomeric regions of chromosomes from the protozoan parasite Trypanosoma cruzi. Molecular and Biochemical Parasitology 100, 173183.CrossRefGoogle ScholarPubMed
Chiurillo, MA, Peralta, A and Ramirez, JL (2002) Comparative study of Trypanosoma rangeli and Trypanosoma cruzi telomeres. Molecular and Biochemical Parasitology 120, 305308.CrossRefGoogle ScholarPubMed
Clayton, C (2019) Regulation of gene expression in trypanosomatids: living with polycistronic transcription. Open Biology 9, 190072.CrossRefGoogle ScholarPubMed
Conte, FF and Cano, MI (2005) Genomic organization of telomeric and subtelomeric sequences of Leishmania (Leishmania) amazonensis. International Journal for Parasitology 35, 14351443.CrossRefGoogle ScholarPubMed
Csűrös, M (2010) Count: evolutionary analysis of phylogenetic profiles with parsimony and likelihood. Bioinformatics (Oxford, England) 26, 19101912.CrossRefGoogle ScholarPubMed
Damasceno, JD, Marques, CA, Beraldi, D, Crouch, K, Lapsley, C, Obonaga, R, Tosi, LR and McCulloch, R (2020) Genome duplication in Leishmania major relies on persistent subtelomeric DNA replication. eLife 9, e58030.CrossRefGoogle ScholarPubMed
Damasceno, JD, Marques, CA, Black, J, Briggs, E and McCulloch, R (2021) Read, write, adapt: challenges and opportunities during kinetoplastid genome replication. Trends in Genetics 37, 2134.CrossRefGoogle ScholarPubMed
de Lima, LP, Calderano, SG, da Silva, MS, de Araujo, CB, Vasconcelos, EJR, Iwai, LK, Pereira, CA, Fragoso, SP and Elias, MC (2019) Ortholog of the polymerase theta helicase domain modulates DNA replication in Trypanosoma cruzi. Scientific Reports 9, 2888.CrossRefGoogle ScholarPubMed
Dobson, DE, Scholtes, LD, Myler, PJ, Turco, SJ and Beverley, SM (2006) Genomic organization and expression of the expanded SCG/L/R gene family of Leishmania major: internal clusters and telomeric localization of SCGs mediating species-specific LPG modifications. Molecular and Biochemical Parasitology 146, 231241.CrossRefGoogle ScholarPubMed
Docampo, R (2016) The origin and evolution of the acidocalcisome and its interactions with other organelles. Molecular and Biochemical Parasitology 209, 39.CrossRefGoogle ScholarPubMed
Dreesen, O and Cross, GA (2006) Telomerase-independent stabilization of short telomeres in Trypanosoma brucei. Molecular and Cellular Biology 26, 49114919.CrossRefGoogle ScholarPubMed
Dreesen, O and Cross, GA (2008) Telomere length in Trypanosoma brucei. Experimental Parasitology 118, 103110.CrossRefGoogle ScholarPubMed
Dreesen, O, Li, B and Cross, GA (2005) Telomere structure and shortening in telomerase-deficient Trypanosoma brucei. Nucleic Acids Research 33, 45364543.CrossRefGoogle ScholarPubMed
Eddy, SR (2009) A new generation of homology search tools based on probabilistic inference. Genome Informatics 23, 205211.Google ScholarPubMed
Eresh, S, McCallum, SM and Barker, DC (1994) Identification and diagnosis of Leishmania mexicana complex isolates by polymerase chain reaction. Parasitology 109, 423433.CrossRefGoogle ScholarPubMed
Espinosa, OA, Serrano, MG, Camargo, EP, Teixeira, MM and Shaw, JJ (2018) An appraisal of the taxonomy and nomenclature of trypanosomatids presently classified as Leishmania and Endotrypanum. Parasitology 145, 430442.CrossRefGoogle ScholarPubMed
Estévez, AM (2008) The RNA-binding protein TbDRBD3 regulates the stability of a specific subset of mRNAs in trypanosomes. Nucleic Acids Research 36, 45734586.CrossRefGoogle ScholarPubMed
Fernández-Orgiler, A, Martinez-Jimenez, MI, Alonso, A, Alcolea, PJ, Requena, JM, Thomas, MC, Blanco, L and Larraga, V (2016) A putative Leishmania DNA polymerase theta protects the parasite against oxidative damage. Nucleic Acids Research 44, 48554870.CrossRefGoogle ScholarPubMed
Frank, AK, Tran, DC, Qu, RW, Stohr, BA, Segal, DJ and Xu, L (2015) The shelterin TIN2 subunit mediates recruitment of telomerase to telomeres. PLoS Genetics 11, e1005410.CrossRefGoogle ScholarPubMed
Frolov, AO, Malysheva, MN, Ganyukova, AI, Spodareva, VV, Yurchenko, V and Kostygov, AY (2019) Development of Phytomonas lipae sp. n. (Kinetoplastea: Trypanosomatidae) in the true bug Coreus marginatus (Heteroptera: Coreidae) and insights into the evolution of life cycles in the genus Phytomonas. PLoS ONE 14, e0214484.CrossRefGoogle Scholar
Fu, G and Barker, DC (1998a) Characterisation of Leishmania telomeres reveals unusual telomeric repeats and conserved telomere-associated sequence. Nucleic Acids Research 26, 21612167.CrossRefGoogle Scholar
Fu, G and Barker, DC (1998b) Rapid cloning of telomere-associated sequence using primer-tagged amplification. Biotechniques 24, 386390.CrossRefGoogle Scholar
Fu, G, Perona-Wright, G and Barker, DC (1998) Leishmania braziliensis: characterisation of a complex specific subtelomeric repeat sequence and its use in the detection of parasites. Experimental Parasitology 90, 236243.CrossRefGoogle ScholarPubMed
Fulnečková, J, Ševčíková, T, Fajkus, J, Lukešová, A, Lukeš, M, Vlček, Č, Lang, BF, Kim, E, Eliáš, M and Sýkorová, E (2013) A broad phylogenetic survey unveils the diversity and evolution of telomeres in eukaryotes. Genome Biology and Evolution 5, 468483.CrossRefGoogle ScholarPubMed
Genest, PA and Borst, P (2007) Analysis of telomere length variation in Leishmania over time. Molecular and Biochemical Parasitology 151, 213215.CrossRefGoogle ScholarPubMed
Genest, PA, Ter Riet, B, Cijsouw, T, van Luenen, HG and Borst, P (2007) Telomeric localization of the modified DNA base J in the genome of the protozoan parasite Leishmania. Nucleic Acids Research 35, 21162124.CrossRefGoogle ScholarPubMed
Glover, L, Hutchinson, S, Alsford, S and Horn, D (2016) VEX1 controls the allelic exclusion required for antigenic variation in trypanosomes. Proceedings of the National Academy of Sciences of the USA 113, 72257230.CrossRefGoogle ScholarPubMed
Glover, L, Marques, CA, Suska, O and Horn, D (2019) Persistent DNA damage foci and DNA replication with a broken chromosome in the African trypanosome. MBio 10, e01252–e01219.CrossRefGoogle ScholarPubMed
Greider, CW (1990) Telomeres, telomerase and senescence. Bioessays 12, 363369.CrossRefGoogle ScholarPubMed
Greider, CW and Blackburn, EH (1985) Identification of a specific telomere terminal transferase activity in Tetrahymena extracts. Cell 43, 405413.CrossRefGoogle ScholarPubMed
Grybchuk, D, Akopyants, NS, Kostygov, AY, Konovalovas, A, Lye, LF, Dobson, DE, Zangger, H, Fasel, N, Butenko, A, Frolov, AO, Votýpka, J, d'Avila-Levy, CM, Kulich, P, Moravcová, J, Plevka, P, Rogozin, IB, Serva, S, Lukeš, J, Beverley, SM and Yurchenko, V (2018) Viral discovery and diversity in trypanosomatid protozoa with a focus on relatives of the human parasite Leishmania. Proceedings of the National Academy of Sciences of the USA 115, E506E515.CrossRefGoogle ScholarPubMed
Hackett, JA and Greider, CW (2002) Balancing instability: dual roles for telomerase and telomere dysfunction in tumorigenesis. Oncogene 21, 619626.CrossRefGoogle ScholarPubMed
Hock, R, Furusawa, T, Ueda, T and Bustin, M (2007) HMG chromosomal proteins in development and disease. Trends in Cell Biology 17, 7279.CrossRefGoogle ScholarPubMed
Horn, D, Spence, C and Ingram, AK (2000) Telomere maintenance and length regulation in Trypanosoma brucei. EMBO Journal 19, 23322339.CrossRefGoogle ScholarPubMed
Hovel-Miner, GA, Boothroyd, CE, Mugnier, M, Dreesen, O, Cross, GA and Papavasiliou, FN (2012) Telomere length affects the frequency and mechanism of antigenic variation in Trypanosoma brucei. PLoS Pathogens 8, e1002900.CrossRefGoogle ScholarPubMed
Janzen, CJ, Lander, F, Dreesen, O and Cross, GA (2004) Telomere length regulation and transcriptional silencing in Ku80-deficient Trypanosoma brucei. Nucleic Acids Research 32, 65756584.CrossRefGoogle ScholarPubMed
Jehi, SE, Li, X, Sandhu, R, Ye, F, Benmerzouga, I, Zhang, M, Zhao, Y and Li, B (2014 a) Suppression of subtelomeric VSG switching by Trypanosoma brucei TRF requires its TTAGGG repeat-binding activity. Nucleic Acids Research 42, 1289912911.CrossRefGoogle ScholarPubMed
Jehi, SE, Wu, F and Li, B (2014 b) Trypanosoma brucei TIF2 suppresses VSG switching by maintaining subtelomere integrity. Cell Research 24, 870885.CrossRefGoogle ScholarPubMed
Jehi, SE, Nanavaty, V and Li, B (2016) Trypanosoma brucei TIF2 and TRF suppress VSG switching using overlapping and independent mechanisms. PLoS ONE 11, e0156746.CrossRefGoogle ScholarPubMed
Kato, H, Caceres, AG, Seki, C, Silupu Garcia, CR, Holguin Mauricci, C, Castro Martinez, SC, Moreno Paico, D, Castro Muniz, JL, Troyes Rivera, LD, Villegas Briones, ZI, Guerrero Quincho, S, Sulca Jayo, GL, Tineo Villafuerte, E, Manrique de Lara Estrada, C, Arias, FR, Passara, FS, Ruelas Llerena, N, Kubo, M, Tabbabi, A, Yamamoto, DS and Hashiguchi, Y (2019) Further insight into the geographic distribution of Leishmania species in Peru by cytochrome b and mannose phosphate isomerase gene analyses. PLoS Neglected Tropical Diseases 13, e0007496.CrossRefGoogle ScholarPubMed
Kostygov, AY and Yurchenko, V (2017) Revised classification of the subfamily Leishmaniinae (Trypanosomatidae). Folia Parasitologica 64, 020.CrossRefGoogle Scholar
Kostygov, AY, Grybchuk-Ieremenko, A, Malysheva, MN, Frolov, AO and Yurchenko, V (2014) Molecular revision of the genus Wallaceina. Protist 165, 594604.CrossRefGoogle ScholarPubMed
Kostygov, A, Frolov, AO, Malysheva, MN, Ganyukova, AI, Chistyakova, LV, Tashyreva, D, Tesařová, M, Spodareva, VV, Režnarová, J, Macedo, DH, Butenko, A, d'Avila-Levy, CM, Lukeš, J and Yurchenko, V (2020) Vickermania gen. nov., trypanosomatids that use two joined flagella to resist midgut peristaltic flow within the fly host. BMC Biology 18, 187.CrossRefGoogle ScholarPubMed
Kraeva, N, Leštinová, T, Ishemgulova, A, Majerová, K, Butenko, A, Vaselek, S, Bespyatykh, J, Charyyeva, A, Spitzová, T, Kostygov, AY, Lukeš, J, Volf, P, Votýpka, J and Yurchenko, V (2019) LmxM.22.0250-encoded dual specificity protein/lipid phosphatase impairs Leishmania mexicana virulence in vitro. Pathogens (Basel, Switzerland) 8, 241.Google ScholarPubMed
Kramer, S, Bannerman-Chukualim, B, Ellis, L, Boulden, EA, Kelly, S, Field, MC and Carrington, M (2013) Differential localization of the two T. brucei poly(A) binding proteins to the nucleus and RNP granules suggests binding to distinct mRNA pools. PLoS ONE 8, e54004.CrossRefGoogle Scholar
Leal, AZ, Schwebs, M, Briggs, E, Weisert, N, Reis, H, Lemgruber, L, Luko, K, Wilkes, J, Butter, F, McCulloch, R and Janzen, CJ (2020) Genome maintenance functions of a putative Trypanosoma brucei translesion DNA polymerase include telomere association and a role in antigenic variation. Nucleic Acids Research 48, 96609680.CrossRefGoogle Scholar
Lewis, KA and Wuttke, DS (2012) Telomerase and telomere-associated proteins: structural insights into mechanism and evolution. Structure (London, England: 1993) 20, 2839.CrossRefGoogle ScholarPubMed
Li, B, Espinal, A and Cross, GA (2005) Trypanosome telomeres are protected by a homologue of mammalian TRF2. Molecular and Cellular Biology 25, 50115021.CrossRefGoogle ScholarPubMed
Li, CH, Irmer, H, Gudjonsdottir-Planck, D, Freese, S, Salm, H, Haile, S, Estevez, AM and Clayton, C (2006) Roles of a Trypanosoma brucei 5′->3′ exoribonuclease homolog in mRNA degradation. RNA 12, 21712186.CrossRefGoogle ScholarPubMed
Lukeš, J, Skalický, T, Týč, J, Votýpka, J and Yurchenko, V (2014) Evolution of parasitism in kinetoplastid flagellates. Molecular and Biochemical Parasitology 195, 115122.CrossRefGoogle ScholarPubMed
Lukeš, J, Butenko, A, Hashimi, H, Maslov, DA, Votýpka, J and Yurchenko, V (2018) Trypanosomatids are much more than just trypanosomes: clues from the expanded family tree. Trends in Parasitology 34, 466480.CrossRefGoogle ScholarPubMed
Lyčka, M, Peška, V, Demko, M, Spyroglou, I, Kilar, A, Fajkus, J and Fojtová, M (2021) WALTER: an easy way to online evaluate telomere lengths from terminal restriction fragment analysis. BMC Bioinformatics (in press).CrossRefGoogle ScholarPubMed
Maslov, DA, Votýpka, J, Yurchenko, V and Lukeš, J (2013) Diversity and phylogeny of insect trypanosomatids: all that is hidden shall be revealed. Trends in Parasitology 29, 4352.CrossRefGoogle ScholarPubMed
Maslov, DA, Opperdoes, FR, Kostygov, AY, Hashimi, H, Lukeš, J and Yurchenko, V (2019) Recent advances in trypanosomatid research: genome organization, expression, metabolism, taxonomy and evolution. Parasitology 146, 127.CrossRefGoogle ScholarPubMed
Michaeli, S (2011) Trans-splicing in trypanosomes: machinery and its impact on the parasite transcriptome. Future Microbiology 6, 459474.CrossRefGoogle ScholarPubMed
Muñoz-Jordán, JL, Cross, GA, de Lange, T and Griffith, JD (2001) T-loops at trypanosome telomeres. EMBO Journal 20, 579588.CrossRefGoogle ScholarPubMed
Nanavaty, V, Sandhu, R, Jehi, SE, Pandya, UM and Li, B (2017) Trypanosoma brucei RAP1 maintains telomere and subtelomere integrity by suppressing TERRA and telomeric RNA:DNA hybrids. Nucleic Acids Research 45, 57855796.CrossRefGoogle ScholarPubMed
Nenarokova, A, Záhonová, K, Krasilnikova, M, Gahura, O, McCulloch, R, Ziková, A, Yurchenko, V and Lukeš, J (2019) Causes and effects of loss of classical nonhomologous end joining pathway in parasitic eukaryotes. MBio 10, e01541–e01519.CrossRefGoogle ScholarPubMed
Nussbaum, K, Honek, J, Cadmus, CM and Efferth, T (2010) Trypanosomatid parasites causing neglected diseases. Current Medicinal Chemistry 17, 15941617.CrossRefGoogle ScholarPubMed
Olovnikov, AM (1973) A theory of marginotomy. The incomplete copying of template margin in enzymic synthesis of polynucleotides and biological significance of the phenomenon. Journal of Theoretical Biology 41, 181190.CrossRefGoogle ScholarPubMed
Pardue, ML, Danilevskaya, ON, Traverse, KL and Lowenhaupt, K (1997) Evolutionary links between telomeres and transposable elements. Genetica 100, 7384.CrossRefGoogle ScholarPubMed
Paugam, A, Bulteau, AL, Dupouy-Camet, J, Creuzet, C and Friguet, B (2003) Characterization and role of protozoan parasite proteasomes. Trends in Parasitology 19, 5559.CrossRefGoogle ScholarPubMed
Pavani, RS, da Silva, MS, Fernandes, CA, Morini, FS, Araujo, CB, Fontes, MR, Sant'Anna, OA, Machado, CR, Cano, MI, Fragoso, SP and Elias, MC (2016) Replication protein A presents canonical functions and is also involved in the differentiation capacity of Trypanosoma cruzi. PLoS Neglected Tropical Diseases 10, e0005181.CrossRefGoogle ScholarPubMed
Pays, E, Laurent, M, Delinte, K, Van Meirvenne, N and Steinert, M (1983) Differential size variations between transcriptionally active and inactive telomeres of Trypanosoma brucei. Nucleic Acids Research 11, 81378147.CrossRefGoogle ScholarPubMed
Pfeiffer, V and Lingner, J (2013) Replication of telomeres and the regulation of telomerase. Cold Spring Harbor Perspectives in Biology 5, a010405.CrossRefGoogle ScholarPubMed
Porcel, BM, Denoeud, F, Opperdoes, FR, Noel, B, Madoui, M-A, Hammarton, TC, Field, MC, Da Silva, C, Couloux, A, Poulain, J, Katinka, M, Jabbari, K, Aury, J-M, Campbell, DA, Cintron, R, Dickens, NJ, Docampo, R, Sturm, NR, Koumandou, VL, Fabre, S, Flegontov, P, Lukeš, J, Michaeli, S, Mottram, JC, Szoor, B, Zilberstein, D, Bringaud, F, W, P and Dollet, M (2014) The streamlined genome of Phytomonas spp. relative to human pathogenic kinetoplastids reveals a parasite tailored for plants. PLoS Genetics 10, e1004007.CrossRefGoogle ScholarPubMed
Rahnama, M, Novikova, O, Starnes, JH, Zhang, S, Chen, L and Farman, ML (2020) Transposon-mediated telomere destabilization: a driver of genome evolution in the blast fungus. Nucleic Acids Research 48, 71977217.Google ScholarPubMed
Reis, H, Schwebs, M, Dietz, S, Janzen, CJ and Butter, F (2018) TelAP1 links telomere complexes with developmental expression site silencing in African trypanosomes. Nucleic Acids Research 46, 28202833.CrossRefGoogle ScholarPubMed
Riha, K and Shippen, DE (2003) Ku is required for telomeric C-rich strand maintenance but not for end-to-end chromosome fusions in Arabidopsis. Proceedings of the National Academy of Sciences of the USA 100, 611615.CrossRefGoogle Scholar
Rudd, SG, Glover, L, Jozwiakowski, SK, Horn, D and Doherty, AJ (2013) PPL2 translesion polymerase is essential for the completion of chromosomal DNA replication in the African trypanosome. Molecular Cell 52, 554565.CrossRefGoogle ScholarPubMed
Ruvinsky, I and Meyuhas, O (2006) Ribosomal protein S6 phosphorylation: from protein synthesis to cell size. Trends in Biochemical Sciences 31, 342348.CrossRefGoogle ScholarPubMed
Sandhu, R, Sanford, S, Basu, S, Park, M, Pandya, UM, Li, B and Chakrabarti, K (2013) A trans-spliced telomerase RNA dictates telomere synthesis in Trypanosoma brucei. Cell Research 23, 537551.CrossRefGoogle ScholarPubMed
Sayers, EW, Agarwala, R, Bolton, EE, Brister, JR, Canese, K, Clark, K, Connor, R, Fiorini, N, Funk, K, Hefferon, T, Holmes, JB, Kim, S, Kimchi, A, Kitts, PA, Lathrop, S, Lu, Z, Madden, TL, Marchler-Bauer, A, Phan, L, Schneider, VA, Schoch, CL, Pruitt, KD and Ostell, J (2019) Database resources of the National Center for Biotechnology Information. Nucleic Acids Research 47, D23D28.CrossRefGoogle ScholarPubMed
Stuart, K, Brun, R, Croft, S, Fairlamb, A, Gurtler, RE, McKerrow, J, Reed, S and Tarleton, R (2008) Kinetoplastids: related protozoan pathogens, different diseases. Journal of Clinical Investigation 118, 13011310.CrossRefGoogle ScholarPubMed
Szöör, B, Haanstra, JR, Gualdrón-López, M and Michels, PA (2014) Evolution, dynamics and specialized functions of glycosomes in metabolism and development of trypanosomatids. Current Opinion in Microbiology 22, 7987.CrossRefGoogle ScholarPubMed
Tomáška, L, Nosek, J, Kar, A, Willcox, S and Griffith, JD (2019) A new view of the t-loop junction: implications for self-primed telomere extension, expansion of disease-related nucleotide repeat blocks, and telomere evolution. Frontiers in Genetics 10, 792.CrossRefGoogle ScholarPubMed
van Luenen, HG, Farris, C, Jan, S, Genest, PA, Tripathi, P, Velds, A, Kerkhoven, RM, Nieuwland, M, Haydock, A, Ramasamy, G, Vainio, S, Heidebrecht, T, Perrakis, A, Pagie, L, van Steensel, B, Myler, PJ and Borst, P (2012) Glucosylated hydroxymethyluracil, DNA base J, prevents transcriptional readthrough in Leishmania. Cell 150, 909921.CrossRefGoogle ScholarPubMed
Votýpka, J, Kostygov, AY, Kraeva, N, Grybchuk-Ieremenko, A, Tesařová, M, Grybchuk, D, Lukeš, J and Yurchenko, V (2014) Kentomonas gen. n., a new genus of endosymbiont-containing trypanosomatids of Strigomonadinae subfam. n. Protist 165, 825838.CrossRefGoogle Scholar
Walne, AJ, Vulliamy, T, Beswick, R, Kirwan, M and Dokal, I (2008) TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes. Blood 112, 35943600.CrossRefGoogle ScholarPubMed
Yang, X, Figueiredo, LM, Espinal, A, Okubo, E and Li, B (2009) RAP1 is essential for silencing telomeric variant surface glycoprotein genes in Trypanosoma brucei. Cell 137, 99109.CrossRefGoogle ScholarPubMed
Záhonová, K, Hadariová, L, Vacula, R, Yurchenko, V, Eliáš, M, Krajčovič, J and Vesteg, M (2014) A small portion of plastid transcripts is polyadenylated in the flagellate Euglena gracilis. FEBS Letters 588, 783788.CrossRefGoogle ScholarPubMed
Záhonová, K, Kostygov, A, Ševčíková, T, Yurchenko, V and Eliáš, M (2016) An unprecedented non-canonical nuclear genetic code with all three termination codons reassigned as sense codons. Current Biology 26, 23642369.CrossRefGoogle ScholarPubMed
Zoltner, M, Krienitz, N, Field, MC and Kramer, S (2018) Comparative proteomics of the two T. brucei PABPs suggests that PABP2 controls bulk mRNA. PLoS Neglected Tropical Diseases 12, e0006679.CrossRefGoogle Scholar
Figure 0

Table 1. Predicted telomere-associated protein complex composition in T. brucei

Figure 1

Fig. 1. Gains and losses of genes encoding putative telomere-associated proteins in kinetoplastids.

Figure 2

Table 2. Presence of genes putatively involved in telomere maintenance in kinetoplastids

Figure 3

Fig. 2. Southern blotting analysis of telomere repeats in selected species of Trypanosomatidae. Marker sizes are indicated on the left. The vertical lines denote a composite image from the same blot. DNA integrity controls are presented in Supplementary Fig. 1 (left and middle panels).

Figure 4

Table 3. Telomere lengths (weighted median, minimum–maximum) in selected species of Trypanosomatidae

Figure 5

Fig. 3. Transcript levels of telomere-associated proteins and telomere lengths in the species of L. mexicana complex. (A) Quantitative RT-PCR analysis of the core set of proteins implicated in telomere maintenance. Gene expression is presented as normalized means and standard deviations of three replicates. Data are presented in two graphs to account for differences in expression values. (B, C) Southern blotting analysis of telomere repeats (B) and telomerase-encoding gene (C, used as an additional DNA integrity control) in L. amazonensis LV78, LV79, PH8, Josefa and L. mexicana M379. Marker sizes are indicated on the left. DNA integrity controls are presented in Supplementary Fig. 1 (right panel).

Supplementary material: Image

Poláková et al. supplementary material

Poláková et al. supplementary material

Download Poláková et al. supplementary material(Image)
Image 167.3 KB
Supplementary material: Image

Poláková et al. supplementary material

Figure S1

Download Poláková et al. supplementary material(Image)
Image 1.2 MB
Supplementary material: File

Poláková et al. supplementary material

Table S1

Download Poláková et al. supplementary material(File)
File 11.8 KB