Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 4401, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37479728

ABSTRACT

African trypanosomes are dixenous eukaryotic parasites that impose a significant human and veterinary disease burden on sub-Saharan Africa. Diversity between species and life-cycle stages is concomitant with distinct host and tissue tropisms within this group. Here, the spatial proteomes of two African trypanosome species, Trypanosoma brucei and Trypanosoma congolense, are mapped across two life-stages. The four resulting datasets provide evidence of expression of approximately 5500 proteins per cell-type. Over 2500 proteins per cell-type are classified to specific subcellular compartments, providing four comprehensive spatial proteomes. Comparative analysis reveals key routes of parasitic adaptation to different biological niches and provides insight into the molecular basis for diversity within and between these pathogen species.


Subject(s)
Trypanosoma brucei brucei , Trypanosoma congolense , Trypanosomiasis, African , Tsetse Flies , Humans , Animals , Trypanosomiasis, African/parasitology , Tsetse Flies/parasitology , Proteome , Proteomics
2.
Cell Surf ; 8: 100078, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35647418

ABSTRACT

East Coast Fever is a devastating African cattle disease caused by the apicomplexan parasite, Theileria parva. Little is known about the cell surface, and few proteins have been identified. Here, we take an in silico approach to identify novel cell surface proteins, and predict the structure of four key proteins.

4.
Nat Commun ; 11(1): 1326, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165615

ABSTRACT

Persistent pathogens have evolved to avoid elimination by the mammalian immune system including mechanisms to evade complement. Infections with African trypanosomes can persist for years and cause human and animal disease throughout sub-Saharan Africa. It is not known how trypanosomes limit the action of the alternative complement pathway. Here we identify an African trypanosome receptor for mammalian factor H, a negative regulator of the alternative pathway. Structural studies show how the receptor binds ligand, leaving inhibitory domains of factor H free to inactivate complement C3b deposited on the trypanosome surface. Receptor expression is highest in developmental stages transmitted to the tsetse fly vector and those exposed to blood meals in the tsetse gut. Receptor gene deletion reduced tsetse infection, identifying this receptor as a virulence factor for transmission. This demonstrates how a pathogen evolved a molecular mechanism to increase transmission to an insect vector by exploitation of a mammalian complement regulator.


Subject(s)
Complement Factor H/metabolism , Trypanosoma/physiology , Tsetse Flies/parasitology , Animals , Antibodies, Monoclonal/metabolism , CHO Cells , Cattle , Cell Membrane/metabolism , Complement C3b/metabolism , Complement Factor H/chemistry , Cricetinae , Cricetulus , Mice, Inbred BALB C , Parasitemia/blood , Protein Binding , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Up-Regulation
5.
Elife ; 92020 03 26.
Article in English | MEDLINE | ID: mdl-32213288

ABSTRACT

The sleeping sickness parasite, Trypanosoma brucei, uses quorum sensing (QS) to balance proliferation and transmission potential in the mammal bloodstream. A signal transduction cascade regulates this process, a component of which is a divergent member of the DYRK family of protein kinases, TbDYRK. Phylogenetic and mutational analysis in combination with activity and phenotypic assays revealed that TbDYRK exhibits a pre-activated conformation and an atypical HxY activation loop motif, unlike DYRK kinases in other eukaryotes. Phosphoproteomic comparison of TbDYRK null mutants with wild-type parasites identified molecules that operate on both the inhibitory 'slender retainer' and activatory 'stumpy inducer' arms of the QS control pathway. One of these molecules, the RNA-regulator TbZC3H20, regulates parasite QS, this being dependent on the integrity of its TbDYRK phosphorylation site. This analysis reveals fundamental differences to conventional DYRK family regulation and links trypanosome environmental sensing, signal transduction and developmental gene expression in a coherent pathway.


Subject(s)
Gene Expression Regulation, Developmental , Protein Serine-Threonine Kinases/physiology , Protein-Tyrosine Kinases/physiology , Quorum Sensing/physiology , Trypanosoma brucei brucei/physiology , Amino Acid Motifs , Cell Differentiation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Signal Transduction/physiology , Transcription, Genetic , Trypanosoma brucei brucei/genetics , Dyrk Kinases
6.
PLoS Negl Trop Dis ; 13(5): e0007373, 2019 05.
Article in English | MEDLINE | ID: mdl-31120889

ABSTRACT

Infections of humans and livestock with African trypanosomes are treated with drugs introduced decades ago that are not always fully effective and often have severe side effects. Here, the trypanosome haptoglobin-haemoglobin receptor (HpHbR) has been exploited as a route of uptake for an antibody-drug conjugate (ADC) that is completely effective against Trypanosoma brucei in the standard mouse model of infection. Recombinant human anti-HpHbR monoclonal antibodies were isolated and shown to be internalised in a receptor-dependent manner. Antibodies were conjugated to a pyrrolobenzodiazepine (PBD) toxin and killed T. brucei in vitro at picomolar concentrations. A single therapeutic dose (0.25 mg/kg) of a HpHbR antibody-PBD conjugate completely cured a T. brucei mouse infection within 2 days with no re-emergence of infection over a subsequent time course of 77 days. These experiments provide a demonstration of how ADCs can be exploited to treat protozoal diseases that desperately require new therapeutics.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antiprotozoal Agents/administration & dosage , Benzodiazepines/administration & dosage , Pyrroles/administration & dosage , Trypanosomiasis, African/drug therapy , Animals , Antibodies, Monoclonal/chemistry , Antiprotozoal Agents/chemistry , Benzodiazepines/chemistry , Female , Humans , Mice , Mice, Inbred BALB C , Pyrroles/chemistry , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/parasitology
7.
PLoS Pathog ; 14(7): e1007195, 2018 07.
Article in English | MEDLINE | ID: mdl-30020996

ABSTRACT

The sleeping sickness parasite Trypanosoma brucei has a complex life cycle, alternating between a mammalian host and the tsetse fly vector. A tightly controlled developmental programme ensures parasite transmission between hosts as well as survival within them and involves strict regulation of mitochondrial activities. In the glucose-rich bloodstream, the replicative 'slender' stage is thought to produce ATP exclusively via glycolysis and uses the mitochondrial F1FO-ATP synthase as an ATP hydrolysis-driven proton pump to generate the mitochondrial membrane potential (ΔΨm). The 'procyclic' stage in the glucose-poor tsetse midgut depends on mitochondrial catabolism of amino acids for energy production, which involves oxidative phosphorylation with ATP production via the F1FO-ATP synthase. Both modes of the F1FO enzyme critically depend on FO subunit a, which is encoded in the parasite's mitochondrial DNA (kinetoplast or kDNA). Comparatively little is known about mitochondrial function and the role of kDNA in non-replicative 'stumpy' bloodstream forms, a developmental stage essential for disease transmission. Here we show that the L262P mutation in the nuclear-encoded F1 subunit γ that permits survival of 'slender' bloodstream forms lacking kDNA ('akinetoplastic' forms), via FO-independent generation of ΔΨm, also permits their differentiation into stumpy forms. However, these akinetoplastic stumpy cells lack a ΔΨm and have a reduced lifespan in vitro and in mice, which significantly alters the within-host dynamics of the parasite. We further show that generation of ΔΨm in stumpy parasites and their ability to use α-ketoglutarate to sustain viability depend on F1-ATPase activity. Surprisingly, however, loss of ΔΨm does not reduce stumpy life span. We conclude that the L262P γ subunit mutation does not enable FO-independent generation of ΔΨm in stumpy cells, most likely as a consequence of mitochondrial ATP production in these cells. In addition, kDNA-encoded genes other than FO subunit a are important for stumpy form viability.


Subject(s)
DNA, Mitochondrial , Trypanosoma brucei brucei/metabolism , Trypanosoma brucei brucei/pathogenicity , Trypanosomiasis, African/metabolism , Trypanosomiasis, African/transmission , Animals , DNA, Kinetoplast/metabolism , Host-Parasite Interactions/physiology , Mice
8.
PLoS Pathog ; 14(6): e1007145, 2018 06.
Article in English | MEDLINE | ID: mdl-29940034

ABSTRACT

Trypanosoma brucei, the agents of African trypanosomiasis, undergo density-dependent differentiation in the mammalian bloodstream to prepare for transmission by tsetse flies. This involves the generation of cell-cycle arrested, quiescent, stumpy forms from proliferative slender forms. The signalling pathway responsible for the quorum sensing response has been catalogued using a genome-wide selective screen, providing a compendium of signalling protein kinases phosphatases, RNA binding proteins and hypothetical proteins. However, the ordering of these components is unknown. To piece together these components to provide a description of how stumpy formation arises we have used an extragenic suppression approach. This exploited a combinatorial gene knockout and overexpression strategy to assess whether the loss of developmental competence in null mutants of pathway components could be compensated by ectopic expression of other components. We have created null mutants for three genes in the stumpy induction factor signalling pathway (RBP7, YAK, MEKK1) and evaluated complementation by expression of RBP7, NEK17, PP1-6, or inducible gene silencing of the proposed differentiation inhibitor TbTOR4. This indicated that the signalling pathway is non-linear. Phosphoproteomic analysis focused on one pathway component, a putative MEKK, identified molecules with altered expression and phosphorylation profiles in MEKK1 null mutants, including another component in the pathway, NEK17. Our data provide a first molecular dissection of multiple components in a signal transduction cascade in trypanosomes.


Subject(s)
Blood/parasitology , Protozoan Proteins/metabolism , Quorum Sensing , RNA-Binding Proteins/metabolism , Signal Transduction , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/parasitology , Animals , Cell Differentiation , Genome , Mice , Phosphorylation , Protozoan Proteins/genetics , RNA-Binding Proteins/genetics , Trypanosoma brucei brucei/genetics
9.
PLoS Pathog ; 13(1): e1006055, 2017 01.
Article in English | MEDLINE | ID: mdl-28125726

ABSTRACT

African trypanosomes have complex life cycles comprising at least ten developmental forms, variously adapted to different niches in their tsetse fly vector and their mammalian hosts. Unlike many other protozoan pathogens, they are always extracellular and have evolved intricate surface coats that allow them to obtain nutrients while also protecting them from the immune defenses of either insects or mammals. The acquisition of macromolecular nutrients requires receptors that function within the context of these surface coats. The best understood of these is the haptoglobin-hemoglobin receptor (HpHbR) of Trypanosoma brucei, which is used by the mammalian bloodstream form of the parasite, allowing heme acquisition. However, in some primates it also provides an uptake route for trypanolytic factor-1, a mediator of innate immunity against trypanosome infection. Recent studies have shown that during the evolution of African trypanosome species the receptor has diversified in function from a hemoglobin receptor predominantly expressed in the tsetse fly to a haptoglobin-hemoglobin receptor predominantly expressed in the mammalian bloodstream. Structural and functional studies of homologous receptors from different trypanosome species have allowed us to propose an evolutionary history for how one receptor has adapted to different roles in different trypanosome species. They also highlight the challenges that a receptor faces in operating on the complex trypanosome surface and show how these challenges can be met.


Subject(s)
Immunity, Innate , Protozoan Proteins/genetics , Receptors, Cell Surface/genetics , Trypanosoma brucei brucei/immunology , Trypanosomiasis, African/immunology , Tsetse Flies/parasitology , Animals , Biological Evolution , Humans , Life Cycle Stages , Models, Molecular , Primates , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , Trypanosomiasis, African/parasitology
10.
Elife ; 52016 04 15.
Article in English | MEDLINE | ID: mdl-27083048

ABSTRACT

The haptoglobin-haemoglobin receptor of the African trypanosome species, Trypanosoma brucei, is expressed when the parasite is in the bloodstream of the mammalian host, allowing it to acquire haem through the uptake of haptoglobin-haemoglobin complexes. Here we show that in Trypanosoma congolense this receptor is instead expressed in the epimastigote developmental stage that occurs in the tsetse fly, where it acts as a haemoglobin receptor. We also present the structure of the T. congolense receptor in complex with haemoglobin. This allows us to propose an evolutionary history for this receptor, charting the structural and cellular changes that took place as it adapted from a role in the insect to a new role in the mammalian host.


Subject(s)
Evolution, Molecular , Hemoglobins/chemistry , Hemoglobins/metabolism , Protozoan Proteins/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Trypanosoma congolense/genetics , Animals , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Receptors, Cell Surface/genetics , Tsetse Flies/parasitology
11.
PLoS Pathog ; 11(12): e1005259, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26719972

ABSTRACT

Variations on the statement "the variant surface glycoprotein (VSG) coat that covers the external face of the mammalian bloodstream form of Trypanosoma brucei acts a physical barrier" appear regularly in research articles and reviews. The concept of the impenetrable VSG coat is an attractive one, as it provides a clear model for understanding how a trypanosome population persists; each successive VSG protects the plasma membrane and is immunologically distinct from previous VSGs. What is the evidence that the VSG coat is an impenetrable barrier, and how do antibodies and other extracellular proteins interact with it? In this review, the nature of the extracellular surface of the bloodstream form trypanosome is described, and past experiments that investigated binding of antibodies and lectins to trypanosomes are analysed using knowledge of VSG sequence and structure that was unavailable when the experiments were performed. Epitopes for some VSG monoclonal antibodies are mapped as far as possible from previous experimental data, onto models of VSG structures. The binding of lectins to some, but not to other, VSGs is revisited with more recent knowledge of the location and nature of N-linked oligosaccharides. The conclusions are: (i) Much of the variation observed in earlier experiments can be explained by the identity of the individual VSGs. (ii) Much of an individual VSG is accessible to antibodies, and the barrier that prevents access to the cell surface is probably at the base of the VSG N-terminal domain, approximately 5 nm from the plasma membrane. This second conclusion highlights a gap in our understanding of how the VSG coat works, as several plasma membrane proteins with large extracellular domains are very unlikely to be hidden from host antibodies by VSG.


Subject(s)
Trypanosomiasis, African/immunology , Variant Surface Glycoproteins, Trypanosoma/chemistry , Variant Surface Glycoproteins, Trypanosoma/immunology , Variant Surface Glycoproteins, Trypanosoma/metabolism , Host-Parasite Interactions , Humans , Protein Conformation , Trypanosoma brucei brucei
12.
Elife ; 3: e05553, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25497229

ABSTRACT

The haptoglobin-haemoglobin receptor (HpHbR) of African trypanosomes allows acquisition of haem and provides an uptake route for trypanolytic factor-1, a mediator of innate immunity against trypanosome infection. In this study, we report the structure of Trypanosoma brucei HpHbR in complex with human haptoglobin-haemoglobin (HpHb), revealing an elongated ligand-binding site that extends along its membrane distal half. This contacts haptoglobin and the ß-subunit of haemoglobin, showing how the receptor selectively binds HpHb over individual components. Lateral mobility of the glycosylphosphatidylinositol-anchored HpHbR, and a ∼50° kink in the receptor, allows two receptors to simultaneously bind one HpHb dimer. Indeed, trypanosomes take up dimeric HpHb at significantly lower concentrations than monomeric HpHb, due to increased ligand avidity that comes from bivalent binding. The structure therefore reveals the molecular basis for ligand and innate immunity factor uptake by trypanosomes and identifies adaptations that allow efficient ligand uptake in the context of the complex trypanosome cell surface.


Subject(s)
Haptoglobins/chemistry , Hemoglobins/chemistry , Lipoproteins, HDL/metabolism , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Trypanosoma brucei brucei/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biological Transport , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Knockout Techniques , Haptoglobins/genetics , Haptoglobins/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sf9 Cells , Spodoptera , Trypanosoma brucei brucei/chemistry
13.
Eukaryot Cell ; 13(3): 412-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24442893

ABSTRACT

In the bloodstream of mammalian hosts, the sleeping sickness parasite, Trypanosoma brucei, exists as a proliferative slender form or a nonproliferative, transmissible, stumpy form. The transition between these developmental forms is controlled by a density-dependent mechanism that is important for the parasite's infection dynamics, immune evasion via ordered antigenic variation, and disease transmissibility. However, stumpy formation has been lost in most laboratory-adapted trypanosome lines, generating monomorphic parasites that proliferate uncontrolled as slender forms in vitro and in vivo. Nonetheless, these forms are readily amenable to cell culture and high-throughput screening for trypanocidal lead compounds. Here, we have developed and exploited a high-throughput screen for developmental phenotypes using a transgenic monomorphic cell line expressing a reporter under the regulation of gene control signals from the stumpy-specific molecule PAD1. Using a whole-cell fluorescence-based assay to screen over 6,000 small molecules from a kinase-focused compound library, small molecules able to activate stumpy-specific gene expression and proliferation arrest were assayed in a rapid assay format. Independent follow-up validation identified one hit able to induce modest, yet specific, changes in mRNA expression indicative of a partial differentiation to stumpy forms in monomorphs. Further, in pleomorphs this compound induced a stumpy-like phenotype, entailing growth arrest, morphological changes, PAD1 expression, and enhanced differentiation to procyclic forms. This not only provides a potential tool compound for the further understanding of stumpy formation but also demonstrates the use of high-throughput screening in the identification of compounds able to induce specific phenotypes, such as differentiation, in African trypanosomes.


Subject(s)
High-Throughput Screening Assays , Phenotype , Protozoan Proteins/genetics , Small Molecule Libraries/pharmacology , Trypanosoma brucei brucei/genetics , Protozoan Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/pathogenicity , Virulence/genetics
14.
Nature ; 505(7485): 681-685, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24336212

ABSTRACT

The protozoan parasites Trypanosoma brucei spp. cause important human and livestock diseases in sub-Saharan Africa. In mammalian blood, two developmental forms of the parasite exist: proliferative 'slender' forms and arrested 'stumpy' forms that are responsible for transmission to tsetse flies. The slender to stumpy differentiation is a density-dependent response that resembles quorum sensing in microbial systems and is crucial for the parasite life cycle, ensuring both infection chronicity and disease transmission. This response is triggered by an elusive 'stumpy induction factor' (SIF) whose intracellular signalling pathway is also uncharacterized. Laboratory-adapted (monomorphic) trypanosome strains respond inefficiently to SIF but can generate forms with stumpy characteristics when exposed to cell-permeable cAMP and AMP analogues. Exploiting this, we have used a genome-wide RNA interference library screen to identify the signalling components driving stumpy formation. In separate screens, monomorphic parasites were exposed to 8-(4-chlorophenylthio)-cAMP (pCPT-cAMP) or 8-pCPT-2'-O-methyl-5'-AMP to select cells that were unresponsive to these signals and hence remained proliferative. Genome-wide Ion Torrent based RNAi target sequencing identified cohorts of genes implicated in each step of the signalling pathway, from purine metabolism, through signal transducers (kinases, phosphatases) to gene expression regulators. Genes at each step were independently validated in cells naturally capable of stumpy formation, confirming their role in density sensing in vivo. The putative RNA-binding protein, RBP7, was required for normal quorum sensing and promoted cell-cycle arrest and transmission competence when overexpressed. This study reveals that quorum sensing signalling in trypanosomes shares similarities to fundamental quiescence pathways in eukaryotic cells, its components providing targets for quorum-sensing interference-based therapeutics.


Subject(s)
Genome/genetics , Quorum Sensing/genetics , Signal Transduction/genetics , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism , Animals , Cell Differentiation , Cyclic AMP/metabolism , G1 Phase , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation , Protein Kinases/genetics , RNA Interference , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Reproducibility of Results , Trypanosoma brucei brucei/enzymology , Trypanosoma brucei brucei/growth & development
15.
Article in English | MEDLINE | ID: mdl-24294594

ABSTRACT

African trypanosomes are sustained in the bloodstream of their mammalian hosts by their extreme capacity for antigenic variation. However, for life cycle progression, trypanosomes also must generate transmission stages called stumpy forms that are pre-adapted to survive when taken up during the bloodmeal of the disease vector, tsetse flies. These stumpy forms are rather different to the proliferative slender forms that maintain the bloodstream parasitaemia. Firstly, they are non proliferative and morphologically distinct, secondly, they show particular sensitivity to environmental cues that signal entry to the tsetse fly and, thirdly, they are relatively robust such that they survive the changes in temperature, pH and proteolytic environment encountered within the tsetse midgut. These characteristics require regulated changes in gene expression to pre-adapt the parasite and the use of environmental sensing mechanisms, both of which allow the rapid initiation of differentiation to tsetse midgut procyclic forms upon transmission. Interestingly, the generation of stumpy forms is also regulated and periodic in the mammalian blood, this being governed by a density-sensing mechanism whereby a parasite-derived signal drives cell cycle arrest and cellular development both to optimize transmission and to prevent uncontrolled parasite multiplication overwhelming the host. In this review we detail recent developments in our understanding of the molecular mechanisms that underpin the production of stumpy forms in the mammalian bloodstream and their signal perception pathways both in the mammalian bloodstream and upon entry into the tsetse fly. These discoveries are discussed in the context of conserved eukaryotic signaling and differentiation mechanisms. Further, their potential to act as targets for therapeutic strategies that disrupt parasite development either in the mammalian bloodstream or upon their transmission to tsetse flies is also discussed.


Subject(s)
Adaptation, Physiological , Blood/parasitology , Gene Expression Regulation , Trypanosoma/physiology , Tsetse Flies/parasitology , Animals , Humans , Mammals , Trypanosoma/genetics , Trypanosoma/growth & development
16.
Mol Biochem Parasitol ; 190(2): 60-2, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23835071

ABSTRACT

African trypanosomes differentiate between various developmental stages both in mammalian hosts and their tsetse vector to adapt to and survive in the different environments they encounter. In the bloodstream, trypanosomes naturally exist as either proliferative slender-forms or non-proliferative stumpy-forms, the latter being responsible for both prolonged infection and transmission. However, most trypanosome studies are carried out on laboratory-adapted monomorphic cell lines, incapable of differentiating to stumpy-forms or completing the life cycle through the tsetse fly. Partly, this has been due to the inefficiency of transfection of pleomorphic strains which have retained the ability to generate stumpy-forms. Recently, Amaxa Nucleofector® technology was shown to increase transfection efficiency for monomorphic bloodstream forms. Using this technology we have optimised a similar method for pleomorphic bloodstream form transfection, generating transfection efficiencies of 10(-7)-10(-6). This permits routine genetic manipulation of pleomorphic lines, which have the most biological relevance for trypanosomes in the field.


Subject(s)
Transfection/methods , Transformation, Genetic , Trypanosoma brucei brucei/genetics , Genomic Instability
17.
PLoS One ; 8(6): e67069, 2013.
Article in English | MEDLINE | ID: mdl-23840587

ABSTRACT

The gene expression of Trypanosoma brucei has been examined extensively in the blood of mammalian hosts and in forms found in the midgut of its arthropod vector, the tsetse fly. However, trypanosomes also undergo development within the mammalian bloodstream as they progress from morphologically 'slender forms' to transmissible 'stumpy forms' through morphological intermediates. This transition is temporally progressive within the first wave of parasitaemia such that gene expression can be monitored in relatively pure slender and stumpy populations as well as during the progression between these extremes. The development also represents the progression of cells from translationally active forms adapted for proliferation in the host to translationally quiescent forms, adapted for transmission. We have used metabolic labelling to quantitate translational activity in slender forms, stumpy forms and in forms undergoing early differentiation to procyclic forms in vitro. Thereafter we have examined the cohort of total mRNAs that are enriched throughout development in the mammalian bloodstream (slender, intermediate and stumpy forms), irrespective of strain, revealing those that exhibit consistent developmental regulation rather than sample specific changes. Transcripts that cosediment with polysomes in stumpy forms and slender forms have also been enriched to identify transcripts that escape translational repression prior to transmission. Combined, the expression and polysomal association of transcripts as trypanosomes undergo development in the mammalian bloodstream have been defined, providing a resource for trypanosome researchers. This facilitates the identification of those that undergo developmental regulation in the bloodstream and therefore those likely to have a role in the survival and capacity for transmission of stumpy forms.


Subject(s)
Host-Pathogen Interactions/genetics , Mammals/parasitology , Polyribosomes/genetics , RNA, Protozoan/genetics , RNA, Protozoan/metabolism , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/genetics , Animals , Blood/parasitology , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trypanosoma brucei brucei/physiology , Tsetse Flies/physiology
18.
Nucleic Acids Res ; 40(16): 7705-17, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22684509

ABSTRACT

Trypanosomatid parasites provide an extreme model for the posttranscriptional control of eukaryotic gene expression. However, most analysis of their differential gene regulation has focussed on comparisons between life-cycle stages that exist in the blood of mammalian hosts and tsetse flies, the parasite's vector. These environments differ acutely in their temperature, and nutritional, metabolic and molecular composition. In the bloodstream, however, a more exquisitely regulated developmental step occurs: the production of transmissible stumpy forms from proliferative slender forms. This transition occurs in the relatively homogenous bloodstream environment, with stumpy-specific gene expression being repressed until accumulation of a proposed parasite-derived signal, stumpy induction factor. Here, we have dissected the regulatory signals that repress the expression of the stumpy-specific surface transporter PAD1 in slender forms. Using transgenic parasites capable of stumpy formation we show that PAD1-repression is mediated by its 3'-untranslated region. Dissection of this region in monomorphic slender forms and pleomorphic slender and stumpy forms has revealed that two regulatory regions co-operate to repress PAD1 expression, this being alleviated on exposure to SIF in pleomorphs or cAMP analogues that act as stumpy induction factor mimics in monomorphs. These studies identify elements that regulate trypanosome gene expression during development in their mammalian host.


Subject(s)
3' Untranslated Regions , Gene Expression Regulation, Developmental , Membrane Transport Proteins/genetics , Protozoan Proteins/genetics , Regulatory Sequences, Ribonucleic Acid , Trypanosoma brucei brucei/genetics , Animals , Gene Silencing , Mice , Polyadenylation , Trypanosoma brucei brucei/growth & development , Trypanosoma brucei brucei/metabolism
19.
Nat Rev Microbiol ; 10(6): 431-8, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22543519

ABSTRACT

During their life cycle, trypanosomes must overcome conflicting demands to ensure their survival and transmission. First, they must evade immunity without overwhelming the host. Second, they must generate and maintain transmission stages at sufficient levels to allow passage into their tsetse vector. Finally, they must rapidly commit to onward development when they enter the tsetse fly. On the basis of recent quantification and modelling of Trypanosoma brucei infection dynamics, we propose that the interplay between immune evasion and development achieves both infection chronicity and transmissibility. Moreover, we suggest that a novel form of bistable regulation ensures developmental commitment on entry into the tsetse fly midgut.


Subject(s)
Immune Evasion , Malaria/transmission , Trypanosoma brucei brucei/pathogenicity , Animals , Chronic Disease , Humans , Trypanosoma brucei brucei/immunology , Tsetse Flies/parasitology
20.
PLoS Pathog ; 7(10): e1002340, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22046137

ABSTRACT

Trypanosomatid parasites are notorious for the human diseases they cause throughout Africa and South America. However, non-pathogenic trypanosomatids are also found worldwide, infecting a wide range of hosts. One example is Trypanosoma (Megatrypanum) theileri, a ubiquitous protozoan commensal of bovids, which is distributed globally. Exploiting knowledge of pathogenic trypanosomatids, we have developed Trypanosoma theileri as a novel vehicle to deliver vaccine antigens and other proteins to cattle. Conditions for the growth and transfection of T. theileri have been optimised and expressed heterologous proteins targeted for secretion or specific localisation at the cell interior or surface using trafficking signals from Trypanosoma brucei. In cattle, the engineered vehicle could establish in the context of a pre-existing natural T. theileri population, was maintained long-term and generated specific immune responses to an expressed Babesia antigen at protective levels. Building on several decades of basic research into trypanosomatid pathogens, Trypanosoma theileri offers significant potential to target multiple infections, including major cattle-borne zoonoses such as Escherichia coli, Salmonella spp., Brucella abortus and Mycobacterium spp. It also has the potential to deliver therapeutics to cattle, including the lytic factor that protects humans from cattle trypanosomiasis. This could alleviate poverty by protecting indigenous African cattle from African trypanosomiasis.


Subject(s)
Cattle Diseases/immunology , Parasitic Diseases, Animal/immunology , Trypanosoma/immunology , Trypanosomiasis, Bovine/immunology , Vaccination/veterinary , Zoonoses , Animals , Cattle , Cattle Diseases/prevention & control , Cells, Cultured , Trypanosoma/genetics , Trypanosoma/pathogenicity , Trypanosomiasis, Bovine/parasitology , Vaccines, Synthetic/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...