Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Genome Res ; 34(3): 441-453, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38604731

ABSTRACT

Aneuploidy is widely observed in both unicellular and multicellular eukaryotes, usually associated with adaptation to stress conditions. Chromosomal duplication stability is a tradeoff between the fitness cost of having unbalanced gene copies and the potential fitness gained from increased dosage of specific advantageous genes. Trypanosomatids, a family of protozoans that include species that cause neglected tropical diseases, are a relevant group to study aneuploidies. Their life cycle has several stressors that could select for different patterns of chromosomal duplications and/or losses, and their nearly universal use of polycistronic transcription increases their reliance on gene expansion/contraction, as well as post-transcriptional control as mechanisms for gene expression regulation. By evaluating the data from 866 isolates covering seven trypanosomatid genera, we have revealed that aneuploidy tolerance is an ancestral characteristic of trypanosomatids but has a reduced occurrence in a specific monophyletic clade that has undergone large genomic reorganization and chromosomal fusions. We have also identified an ancient chromosomal duplication that was maintained across these parasite's speciation, named collectively as the trypanosomatid ancestral supernumerary chromosome (TASC). TASC has most genes in the same coding strand, is expressed as a disomic chromosome (even having four copies), and has increased potential for functional variation, but it purges highly deleterious mutations more efficiently than other chromosomes. The evidence of stringent control over gene expression in this chromosome suggests that these parasites have adapted to mitigate the fitness cost associated with this ancient chromosomal duplication.


Subject(s)
Aneuploidy , Chromosome Duplication , Gene Expression Regulation , Genome, Protozoan , Evolution, Molecular , Trypanosomatina/genetics , Phylogeny
2.
Proc Natl Acad Sci U S A ; 120(48): e2309306120, 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37988471

ABSTRACT

RNA-DNA hybrids are epigenetic features of all genomes that intersect with many processes, including transcription, telomere homeostasis, and centromere function. Increasing evidence suggests that RNA-DNA hybrids can provide two conflicting roles in the maintenance and transmission of genomes: They can be the triggers of DNA damage, leading to genome change, or can aid the DNA repair processes needed to respond to DNA lesions. Evasion of host immunity by African trypanosomes, such as Trypanosoma brucei, relies on targeted recombination of silent Variant Surface Glycoprotein (VSG) genes into a specialized telomeric locus that directs transcription of just one VSG from thousands. How such VSG recombination is targeted and initiated is unclear. Here, we show that a key enzyme of T. brucei homologous recombination, RAD51, interacts with RNA-DNA hybrids. In addition, we show that RNA-DNA hybrids display a genome-wide colocalization with DNA breaks and that this relationship is impaired by mutation of RAD51. Finally, we show that RAD51 acts to repair highly abundant, localised DNA breaks at the single transcribed VSG and that mutation of RAD51 alters RNA-DNA hybrid abundance at 70 bp repeats both around the transcribed VSG and across the silent VSG archive. This work reveals a widespread, generalised role for RNA-DNA hybrids in directing RAD51 activity during recombination and uncovers a specialised application of this interplay during targeted DNA break repair needed for the critical T. brucei immune evasion reaction of antigenic variation.


Subject(s)
Trypanosoma brucei brucei , R-Loop Structures , Antigenic Variation/genetics , DNA Breaks , DNA , RNA , Variant Surface Glycoproteins, Trypanosoma/genetics
3.
Nucleic Acids Res ; 51(20): 11123-11141, 2023 11 10.
Article in English | MEDLINE | ID: mdl-37843098

ABSTRACT

RNA-DNA hybrids are epigenetic features of genomes that provide a diverse and growing range of activities. Understanding of these functions has been informed by characterising the proteins that interact with the hybrids, but all such analyses have so far focused on mammals, meaning it is unclear if a similar spectrum of RNA-DNA hybrid interactors is found in other eukaryotes. The African trypanosome is a single-cell eukaryotic parasite of the Discoba grouping and displays substantial divergence in several aspects of core biology from its mammalian host. Here, we show that DNA-RNA hybrid immunoprecipitation coupled with mass spectrometry recovers 602 putative interactors in T. brucei mammal- and insect-infective cells, some providing activities also found in mammals and some lineage-specific. We demonstrate that loss of three factors, two putative helicases and a RAD51 paralogue, alters T. brucei nuclear RNA-DNA hybrid and DNA damage levels. Moreover, loss of each factor affects the operation of the parasite immune survival mechanism of antigenic variation. Thus, our work reveals the broad range of activities contributed by RNA-DNA hybrids to T. brucei biology, including new functions in host immune evasion as well as activities likely fundamental to eukaryotic genome function.


Subject(s)
Trypanosoma brucei brucei , Animals , Trypanosoma brucei brucei/metabolism , Immune Evasion/genetics , RNA/genetics , Antigens, Surface , Antigenic Variation/genetics , DNA/genetics , Mammals/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
4.
Trends Genet ; 37(1): 21-34, 2021 01.
Article in English | MEDLINE | ID: mdl-32993968

ABSTRACT

The genomes of all organisms are read throughout their growth and development, generating new copies during cell division and encoding the cellular activities dictated by the genome's content. However, genomes are not invariant information stores but are purposefully altered in minor and major ways, adapting cellular behaviour and driving evolution. Kinetoplastids are eukaryotic microbes that display a wide range of such read-write genome activities, in many cases affecting critical aspects of their biology, such as host adaptation. Here we discuss the range of read-write genome changes found in two well-studied kinetoplastid parasites, Trypanosoma brucei and Leishmania, focusing on recent work that suggests such adaptive genome variation is linked to novel strategies the parasites use to replicate their unconventional genomes.


Subject(s)
DNA Replication , DNA, Kinetoplast/genetics , Genome, Protozoan , Kinetoplastida/genetics , Leishmania/genetics , Trypanosoma brucei brucei/genetics , Animals
5.
J Eukaryot Microbiol ; 70(6): e12994, 2023.
Article in English | MEDLINE | ID: mdl-37548427

ABSTRACT

Selection and internalization of cargo via clathrin-mediated endocytosis requires adaptor protein complexes. One complex, AP-2, acts during cargo selection at the plasma membrane. African trypanosomes lack all components of the AP-2 complex, except for a recently identified orthologue of the AP-2-associated protein kinase 1, AAK1. In characterized eukaryotes, AAK1 phosphorylates the µ2 subunit of the AP-2 complex to enhance cargo recognition and uptake into clathrin-coated vesicles. Here, we show that kinetoplastids encode not one, but two AAK1 orthologues: one (AAK1L2) is absent from salivarian trypanosomes, while the other (AAK1L1) lacks important kinase-specific residues in a range of trypanosomes. These AAK1L1 and AAK1L2 novelties reinforce suggestions of functional divergence in endocytic uptake within salivarian trypanosomes. Despite this, we show that AAK1L1 null mutant Trypanosoma brucei, while viable, display slowed proliferation, morphological abnormalities including swelling of the flagellar pocket, and altered cargo uptake. In summary, our data suggest an unconventional role for a putative pseudokinase during endocytosis and/or vesicular trafficking in T. brucei, independent of AP-2.


Subject(s)
Parasites , Trypanosoma brucei brucei , Animals , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , Clathrin/metabolism , Parasites/metabolism , Endocytosis/physiology , Cell Membrane
6.
Exp Parasitol ; 255: 108639, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37918502

ABSTRACT

The subcellular localisation of Rad1, a subunit of the Leishmania major 9-1-1 complex, remains unexplored. Herein, we reveal that Rad1 localises predominantly to the nucleus. Upon hydroxyurea treatment, the diffuse nuclear localisation of Rad1 becomes more punctate, suggesting that Rad1 is responsive to replication stress. Moreover, Rad1 localisation correlates with cell cycle progression. In the majority of G1 to early S-phase cells, Rad1 localises predominantly to the nucleus. As cells progress from late-S phase to mitosis, Rad1 relocalizes to both the nucleus and the cytoplasm in ∼90 % of cells. This pattern of distribution is different from Rad9 and Hus1, which remain nuclear throughout the cell cycle, suggesting Leishmania Rad1 may regulate 9-1-1 activities and/or perform relevant functions outside the 9-1-1 complex.


Subject(s)
Cell Cycle Proteins , Leishmania major , Cell Cycle Proteins/genetics , Leishmania major/metabolism , Cell Cycle , DNA Damage
7.
Nucleic Acids Res ; 49(3): 1436-1454, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33450001

ABSTRACT

Homologous recombination dominates as the major form of DNA repair in Trypanosoma brucei, and is especially important for recombination of the subtelomeric variant surface glycoprotein during antigenic variation. RAD50, a component of the MRN complex (MRE11, RAD50, NBS1), is central to homologous recombination through facilitating resection and governing the DNA damage response. The function of RAD50 in trypanosomes is untested. Here we report that RAD50 and MRE11 are required for RAD51-dependent homologous recombination and phosphorylation of histone H2A following a DNA double strand break (DSB), but neither MRE11 nor RAD50 substantially influence DSB resection at a chromosome-internal locus. In addition, we reveal intrinsic separation-of-function between T. brucei RAD50 and MRE11, with only RAD50 suppressing DSB repair using donors with short stretches of homology at a subtelomeric locus, and only MRE11 directing DSB resection at the same locus. Finally, we show that loss of either MRE11 or RAD50 causes a greater diversity of expressed VSG variants following DSB repair. We conclude that MRN promotes stringent homologous recombination at subtelomeric loci and restrains antigenic variation.


Subject(s)
Antigenic Variation , DNA-Binding Proteins/physiology , MRE11 Homologue Protein/physiology , Protozoan Proteins/physiology , Recombinational DNA Repair , Trypanosoma brucei brucei/genetics , DNA Breaks, Double-Stranded , Trypanosoma brucei brucei/immunology
8.
PLoS Genet ; 16(7): e1008828, 2020 07.
Article in English | MEDLINE | ID: mdl-32609721

ABSTRACT

Homologous recombination (HR) has an intimate relationship with genome replication, both during repair of DNA lesions that might prevent DNA synthesis and in tackling stalls to the replication fork. Recent studies led us to ask if HR might have a more central role in replicating the genome of Leishmania, a eukaryotic parasite. Conflicting evidence has emerged regarding whether or not HR genes are essential, and genome-wide mapping has provided evidence for an unorthodox organisation of DNA replication initiation sites, termed origins. To answer this question, we have employed a combined CRISPR/Cas9 and DiCre approach to rapidly generate and assess the effect of conditional ablation of RAD51 and three RAD51-related proteins in Leishmania major. Using this approach, we demonstrate that loss of any of these HR factors is not immediately lethal but in each case growth slows with time and leads to DNA damage and accumulation of cells with aberrant DNA content. Despite these similarities, we show that only loss of RAD51 or RAD51-3 impairs DNA synthesis and causes elevated levels of genome-wide mutation. Furthermore, we show that these two HR factors act in distinct ways, since ablation of RAD51, but not RAD51-3, has a profound effect on DNA replication, causing loss of initiation at the major origins and increased DNA synthesis at subtelomeres. Our work clarifies questions regarding the importance of HR to survival of Leishmania and reveals an unanticipated, central role for RAD51 in the programme of genome replication in a microbial eukaryote.


Subject(s)
Homologous Recombination/genetics , Leishmania major/genetics , Leishmaniasis, Cutaneous/genetics , Rad51 Recombinase/genetics , CRISPR-Cas Systems/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA Replication/genetics , Gene Knockout Techniques , Genome/genetics , Humans , Leishmania major/pathogenicity , Leishmaniasis, Cutaneous/parasitology
9.
Nucleic Acids Res ; 48(17): 9660-9680, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32890403

ABSTRACT

Maintenance of genome integrity is critical to guarantee transfer of an intact genome from parent to offspring during cell division. DNA polymerases (Pols) provide roles in both replication of the genome and the repair of a wide range of lesions. Amongst replicative DNA Pols, translesion DNA Pols play a particular role: replication to bypass DNA damage. All cells express a range of translesion Pols, but little work has examined their function in parasites, including whether the enzymes might contribute to host-parasite interactions. Here, we describe a dual function of one putative translesion Pol in African trypanosomes, which we now name TbPolIE. Previously, we demonstrated that TbPolIE is associated with telomeric sequences and here we show that RNAi-mediated depletion of TbPolIE transcripts results in slowed growth, altered DNA content, changes in cell morphology, and increased sensitivity to DNA damaging agents. We also show that TbPolIE displays pronounced localization at the nuclear periphery, and that its depletion leads to chromosome segregation defects and increased levels of endogenous DNA damage. Finally, we demonstrate that TbPolIE depletion leads to deregulation of telomeric variant surface glycoprotein genes, linking the function of this putative translesion DNA polymerase to host immune evasion by antigenic variation.


Subject(s)
Antigenic Variation , DNA-Directed DNA Polymerase/metabolism , Telomere/genetics , Trypanosoma brucei brucei/genetics , Cell Line , Cell Nucleus/enzymology , Cell Nucleus/genetics , Chromosome Segregation , DNA Replication , DNA-Directed DNA Polymerase/genetics , Gene Expression Regulation , Genome, Protozoan , Humans , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA Interference , Telomere/metabolism , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/pathogenicity , Variant Surface Glycoproteins, Trypanosoma/genetics , DNA Polymerase theta
10.
Parasitology ; 148(10): 1223-1236, 2021 09.
Article in English | MEDLINE | ID: mdl-33678213

ABSTRACT

Kinetoplastid parasites are responsible for both human and animal diseases across the globe where they have a great impact on health and economic well-being. Many species and life cycle stages are difficult to study due to limitations in isolation and culture, as well as to their existence as heterogeneous populations in hosts and vectors. Single-cell transcriptomics (scRNA-seq) has the capacity to overcome many of these difficulties, and can be leveraged to disentangle heterogeneous populations, highlight genes crucial for propagation through the life cycle, and enable detailed analysis of host­parasite interactions. Here, we provide a review of studies that have applied scRNA-seq to protozoan parasites so far. In addition, we provide an overview of sample preparation and technology choice considerations when planning scRNA-seq experiments, as well as challenges faced when analysing the large amounts of data generated. Finally, we highlight areas of kinetoplastid research that could benefit from scRNA-seq technologies.


Subject(s)
Gene Expression Profiling , Kinetoplastida/genetics , Single-Cell Analysis , Host-Parasite Interactions , RNA-Seq
11.
Nucleic Acids Res ; 47(17): 9180-9197, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31350892

ABSTRACT

Ribonucleotides represent a threat to DNA genome stability and transmission. Two types of Ribonuclease H (RNase H) excise ribonucleotides when they form part of the DNA strand, or hydrolyse RNA when it base-pairs with DNA in structures termed R-loops. Loss of either RNase H is lethal in mammals, whereas yeast survives the absence of both enzymes. RNase H1 loss is tolerated by the parasite Trypanosoma brucei but no work has examined the function of RNase H2. Here we show that loss of T. brucei RNase H2 (TbRH2A) leads to growth and cell cycle arrest that is concomitant with accumulation of nuclear damage at sites of RNA polymerase (Pol) II transcription initiation, revealing a novel and critical role for RNase H2. Differential gene expression analysis reveals limited overall changes in RNA levels for RNA Pol II genes after TbRH2A loss, but increased perturbation of nucleotide metabolic genes. Finally, we show that TbRH2A loss causes R-loop and DNA damage accumulation in telomeric RNA Pol I transcription sites, also leading to altered gene expression. Thus, we demonstrate separation of function between two nuclear T. brucei RNase H enzymes during RNA Pol II transcription, but overlap in function during RNA Pol I-mediated gene expression during host immune evasion.


Subject(s)
Antigens, Protozoan/genetics , Genomic Instability/genetics , Ribonuclease H/genetics , Transcription Initiation, Genetic , Animals , Antigens, Protozoan/immunology , DNA/chemistry , DNA/genetics , DNA Damage/genetics , DNA Replication/genetics , Gene Expression Regulation/genetics , Humans , Nucleic Acid Conformation , RNA/chemistry , RNA/genetics , RNA Polymerase I/genetics , RNA Polymerase II/genetics , Ribonuclease H/chemistry , Ribonuclease H/immunology , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/immunology , Trypanosoma brucei brucei/pathogenicity
12.
PLoS Genet ; 14(12): e1007729, 2018 12.
Article in English | MEDLINE | ID: mdl-30543624

ABSTRACT

Switching of the Variant Surface Glycoprotein (VSG) in Trypanosoma brucei provides a crucial host immune evasion strategy that is catalysed both by transcription and recombination reactions, each operating within specialised telomeric VSG expression sites (ES). VSG switching is likely triggered by events focused on the single actively transcribed ES, from a repertoire of around 15, but the nature of such events is unclear. Here we show that RNA-DNA hybrids, called R-loops, form preferentially within sequences termed the 70 bp repeats in the actively transcribed ES, but spread throughout the active and inactive ES, in the absence of RNase H1, which degrades R-loops. Loss of RNase H1 also leads to increased levels of VSG coat switching and replication-associated genome damage, some of which accumulates within the active ES. This work indicates VSG ES architecture elicits R-loop formation, and that these RNA-DNA hybrids connect T. brucei immune evasion by transcription and recombination.


Subject(s)
Immune Evasion/genetics , Ribonuclease H/genetics , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/immunology , Variant Surface Glycoproteins, Trypanosoma/genetics , Variant Surface Glycoproteins, Trypanosoma/immunology , Animals , Antigenic Variation , DNA Damage , Genome, Protozoan , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Ribonuclease H/deficiency , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/immunology , Trypanosomiasis, African/parasitology
13.
BMC Genomics ; 21(1): 414, 2020 Jun 22.
Article in English | MEDLINE | ID: mdl-32571205

ABSTRACT

BACKGROUND: DNA replication in trypanosomatids operates in a uniquely challenging environment, since most of their genomes are constitutively transcribed. Trypanosoma cruzi, the etiological agent of Chagas disease, presents high variability in both chromosomes size and copy number among strains, though the underlying mechanisms are unknown. RESULTS: Here we have mapped sites of DNA replication initiation across the T. cruzi genome using Marker Frequency Analysis, which has previously only been deployed in two related trypanosomatids. The putative origins identified in T. cruzi show a notable enrichment of GC content, a preferential position at subtelomeric regions, coinciding with genes transcribed towards the telomeres, and a pronounced enrichment within coding DNA sequences, most notably in genes from the Dispersed Gene Family 1 (DGF-1). CONCLUSIONS: These findings suggest a scenario where collisions between DNA replication and transcription are frequent, leading to increased genetic variability, as seen by the increase SNP levels at chromosome subtelomeres and in DGF-1 genes containing putative origins.


Subject(s)
Polymorphism, Single Nucleotide , Replication Origin , Trypanosoma cruzi/genetics , Whole Genome Sequencing/methods , Animals , Base Composition , DNA Replication , DNA, Protozoan/genetics , High-Throughput Nucleotide Sequencing , Triatoma/parasitology , Trypanosoma cruzi/isolation & purification
14.
PLoS Pathog ; 14(11): e1007321, 2018 11.
Article in English | MEDLINE | ID: mdl-30440029

ABSTRACT

Antigenic variation by variant surface glycoprotein (VSG) coat switching in African trypanosomes is one of the most elaborate immune evasion strategies found among pathogens. Changes in the identity of the transcribed VSG gene, which is always flanked by 70-bp and telomeric repeats, can be achieved either by transcriptional or DNA recombination mechanisms. The major route of VSG switching is DNA recombination, which occurs in the bloodstream VSG expression site (ES), a multigenic site transcribed by RNA polymerase I. Recombinogenic VSG switching is frequently catalyzed by homologous recombination (HR), a reaction normally triggered by DNA breaks. However, a clear understanding of how such breaks arise-including whether there is a dedicated and ES-focused mechanism-is lacking. Here, we synthesize data emerging from recent studies that have proposed a range of mechanisms that could generate these breaks: action of a nuclease or nucleases; repetitive DNA, most notably the 70-bp repeats, providing an intra-ES source of instability; DNA breaks derived from the VSG-adjacent telomere; DNA breaks arising from high transcription levels at the active ES; and DNA lesions arising from replication-transcription conflicts in the ES. We discuss the evidence that underpins these switch-initiation models and consider what features and mechanisms might be shared or might allow the models to be tested further. Evaluation of all these models highlights that we still have much to learn about the earliest acting step in VSG switching, which may have the greatest potential for therapeutic intervention in order to undermine the key reaction used by trypanosomes for their survival and propagation in the mammalian host.


Subject(s)
Trypanosoma/immunology , Variant Surface Glycoproteins, Trypanosoma/genetics , Variant Surface Glycoproteins, Trypanosoma/immunology , Antigenic Variation/genetics , Antigenic Variation/physiology , DNA/metabolism , DNA Replication/immunology , Immune Evasion/genetics , Immune Evasion/immunology , Telomere/genetics , Transcription, Genetic/genetics , Trypanosoma/genetics , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/genetics , Trypanosomiasis, African/immunology
15.
Nucleic Acids Res ; 46(22): 11789-11805, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30304482

ABSTRACT

R-loops are stable RNA-DNA hybrids that have been implicated in transcription initiation and termination, as well as in telomere maintenance, chromatin formation, and genome replication and instability. RNA Polymerase (Pol) II transcription in the protozoan parasite Trypanosoma brucei is highly unusual: virtually all genes are co-transcribed from multigene transcription units, with mRNAs generated by linked trans-splicing and polyadenylation, and transcription initiation sites display no conserved promoter motifs. Here, we describe the genome-wide distribution of R-loops in wild type mammal-infective T. brucei and in mutants lacking RNase H1, revealing both conserved and diverged functions. Conserved localization was found at centromeres, rRNA genes and retrotransposon-associated genes. RNA Pol II transcription initiation sites also displayed R-loops, suggesting a broadly conserved role despite the lack of promoter conservation or transcription initiation regulation. However, the most abundant sites of R-loop enrichment were within the regions between coding sequences of the multigene transcription units, where the hybrids coincide with sites of polyadenylation and nucleosome-depletion. Thus, instead of functioning in transcription termination the most widespread localization of R-loops in T. brucei suggests a novel correlation with pre-mRNA processing. Finally, we find little evidence for correlation between R-loop localization and mapped sites of DNA replication initiation.


Subject(s)
Genome, Protozoan , Mutation , Trypanosoma brucei brucei/genetics , Binding Sites , Centromere , Chromatin/chemistry , Gene Expression Regulation , Nucleosomes , Polyadenylation , Promoter Regions, Genetic , Protein Domains , Protozoan Proteins/genetics , RNA Polymerase II/metabolism , RNA, Ribosomal/chemistry , Transcription Initiation Site , Transcription, Genetic , Variant Surface Glycoproteins, Trypanosoma/genetics
16.
Nucleic Acids Res ; 46(22): 11835-11846, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30380080

ABSTRACT

Leishmania species are protozoan parasites whose remarkably plastic genome limits the establishment of effective genetic manipulation and leishmaniasis treatment. The strategies used by Leishmania to maintain its genome while allowing variability are not fully understood. Here, we used DiCre-mediated conditional gene deletion to show that HUS1, a component of the 9-1-1 (RAD9-RAD1-HUS1) complex, is essential and is required for a G2/M checkpoint. By analyzing genome-wide instability in HUS1 ablated cells, HUS1 is shown to have a conserved role, by which it preserves genome stability and also a divergent role, by which it promotes genome variability. These roles of HUS1 are related to distinct patterns of formation and resolution of single-stranded DNA and γH2A, throughout the cell cycle. Our findings suggest that Leishmania 9-1-1 subunits have evolved to co-opt canonical genomic maintenance and genomic variation functions. Hence, this study reveals a pivotal function of HUS1 in balancing genome stability and transmission in Leishmania. These findings may be relevant to understanding the evolution of genome maintenance and plasticity in other pathogens and eukaryotes.


Subject(s)
Cell Cycle Proteins/genetics , DNA Repair Enzymes/genetics , Endonucleases/genetics , Genome, Protozoan , Leishmania major/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/metabolism , Computational Biology/methods , Culture Media/chemistry , DNA Repair Enzymes/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , Endonucleases/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Gene Deletion , Gene Expression Regulation , Genetic Engineering , Genetic Variation , Genomic Instability , Histones/genetics , Histones/metabolism , Leishmania major/metabolism , Whole Genome Sequencing
17.
PLoS Pathog ; 13(7): e1006477, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28742144

ABSTRACT

All cells are subject to structural damage that must be addressed for continued growth. A wide range of damage affects the genome, meaning multiple pathways have evolved to repair or bypass the resulting DNA lesions. Though many repair pathways are conserved, their presence or function can reflect the life style of individual organisms. To identify genome maintenance pathways in a divergent eukaryote and important parasite, Trypanosoma brucei, we performed RNAi screens to identify genes important for survival following exposure to the alkylating agent methyl methanesulphonate. Amongst a cohort of broadly conserved and, therefore, early evolved repair pathways, we reveal multiple activities not so far examined functionally in T. brucei, including DNA polymerases, DNA helicases and chromatin factors. In addition, the screens reveal Trypanosoma- or kinetoplastid-specific repair-associated activities. We also provide focused analyses of repair-associated protein kinases and show that loss of at least nine, and potentially as many as 30 protein kinases, including a nuclear aurora kinase, sensitises T. brucei to alkylation damage. Our results demonstrate the potential for synthetic lethal genome-wide screening of gene function in T. brucei and provide an evolutionary perspective on the repair pathways that underpin effective responses to damage, with particular relevance for related kinetoplastid pathogens. By revealing that a large number of diverse T. brucei protein kinases act in the response to damage, we expand the range of eukaryotic signalling factors implicated in genome maintenance activities.


Subject(s)
DNA Repair , Genome, Protozoan , Protein Kinases/genetics , Protozoan Proteins/genetics , RNA Interference , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/genetics , DNA Damage/drug effects , Evolution, Molecular , Methyl Methanesulfonate/analogs & derivatives , Methyl Methanesulfonate/toxicity , Mutagens/toxicity , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/drug effects
18.
Curr Genomics ; 19(2): 98-109, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29491738

ABSTRACT

INTRODUCTION: Understanding how the nuclear genome of kinetoplastid parasites is replicated received experimental stimulus from sequencing of the Leishmania major, Trypanosoma brucei and Trypanosoma cruzi genomes around 10 years ago. Gene annotations suggested key players in DNA replication initiation could not be found in these organisms, despite considerable conservation amongst characterised eukaryotes. Initial studies that indicated trypanosomatids might possess an archaeal-like Origin Recognition Complex (ORC), composed of only a single factor termed ORC1/CDC6, have been supplanted by the more recent identification of an ORC in T. brucei. However, the constituent subunits of T. brucei ORC are highly diverged relative to other eukaryotic ORCs and the activity of the complex appears subject to novel, positive regulation. The availability of whole genome sequences has also allowed the deployment of genome-wide strategies to map DNA replication dynamics, to date in T. brucei and Leishmania. ORC1/CDC6 binding and function in T. brucei displays pronounced overlap with the unconventional organisation of gene expression in the genome. Moreover, mapping of sites of replication initiation suggests pronounced differences in replication dynamics in Leishmania relative to T. brucei. CONCLUSION: Here we discuss what implications these emerging data may have for parasite and eukaryotic biology of DNA replication.

19.
Nucleic Acids Res ; 44(10): 4763-84, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26951375

ABSTRACT

Initiation of DNA replication depends upon recognition of genomic sites, termed origins, by AAA+ ATPases. In prokaryotes a single factor binds each origin, whereas in eukaryotes this role is played by a six-protein origin recognition complex (ORC). Why eukaryotes evolved a multisubunit initiator, and the roles of each component, remains unclear. In Trypanosoma brucei, an ancient unicellular eukaryote, only one ORC-related initiator, TbORC1/CDC6, has been identified by sequence homology. Here we show that three TbORC1/CDC6-interacting factors also act in T. brucei nuclear DNA replication and demonstrate that TbORC1/CDC6 interacts in a high molecular complex in which a diverged Orc4 homologue and one replicative helicase subunit can also be found. Analysing the subcellular localization of four TbORC1/CDC6-interacting factors during the cell cycle reveals that one factor, TbORC1B, is not a static constituent of ORC but displays S-phase restricted nuclear localization and expression, suggesting it positively regulates replication. This work shows that ORC architecture and regulation are diverged features of DNA replication initiation in T. brucei, providing new insight into this key stage of eukaryotic genome copying.


Subject(s)
DNA Replication , Origin Recognition Complex/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei/genetics , Cell Cycle , Cell Nucleus/genetics , Origin Recognition Complex/physiology , Protozoan Proteins/physiology , Trypanosoma brucei brucei/metabolism
20.
Mol Microbiol ; 101(6): 1054-68, 2016 09.
Article in English | MEDLINE | ID: mdl-27301589

ABSTRACT

The Rad9-Rad1-Hus1 (9-1-1) complex is a key component in the coordination of DNA damage sensing, cell cycle progression and DNA repair pathways in eukaryotic cells. This PCNA-related trimer is loaded onto RPA-coated single stranded DNA and interacts with ATR kinase to mediate effective checkpoint signaling to halt the cell cycle and to promote DNA repair. Beyond these core activities, mounting evidence suggests that a broader range of functions can be provided by 9-1-1 structural diversification. The protozoan parasite Leishmania is an early-branching eukaryote with a remarkably plastic genome, which hints at peculiar genome maintenance mechanisms. Here, we investigated the existence of homologs of the 9-1-1 complex subunits in L. major and found that LmRad9 and LmRad1 associate with chromatin in response to replication stress and form a complex in vivo with LmHus1. Similar to LmHus1, LmRad9 participates in telomere homeostasis and in the response to both replication stress and double strand breaks. However, LmRad9 and LmHus1-deficient cells present markedly opposite phenotypes, which suggest their functional compartmentalization. We show that some of the cellular pool of LmRad9 forms an alternative complex and that some of LmHus1 exists as a monomer. We propose that the diverse assembly of the Leishmania 9-1-1 subunits mediates functional compartmentalization, which has a direct impact on the response to genotoxic stress.


Subject(s)
Cell Cycle Proteins/metabolism , DNA Damage/physiology , Exonucleases/metabolism , Leishmania major/physiology , Cell Cycle Checkpoints/physiology , DNA-Binding Proteins/metabolism , Leishmania major/genetics , Leishmania major/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL