Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
mBio ; 15(5): e0314023, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38530030

ABSTRACT

The Plasmodium falciparum merozoite surface protein MSPDBL2 is a polymorphic antigen targeted by acquired immune responses, and normally expressed in only a minority of mature schizonts. The potential relationship of MSPDBL2 to sexual commitment is examined, as variable mspdbl2 transcript levels and proportions of MSPDBL2-positive mature schizonts in clinical isolates have previously correlated with levels of many sexual stage parasite gene transcripts, although not with the master regulator ap2-g. It is demonstrated that conditional overexpression of the gametocyte development protein GDV1, which promotes sexual commitment, also substantially increases the proportion of MSPDBL2-positive schizonts in culture. Conversely, truncation of the gdv1 gene is shown to prevent any expression of MSPDBL2. However, across diverse P. falciparum cultured lines, the variable proportions of MSPDBL2 positivity in schizonts do not correlate significantly with variable gametocyte conversion rates, indicating it is not involved in sexual commitment. Confirming this, examining a line with endogenous hemagglutinin-tagged AP2-G showed that the individual schizonts expressing MSPDBL2 are mostly different from those expressing AP2-G. Using a selection-linked integration system, modified P. falciparum lines were engineered to express an intact or disrupted version of MSPDBL2, showing the protein is not required for sexual commitment or early gametocyte development. Asexual parasite multiplication rates were also not affected by expression of either intact or disrupted MSPDBL2 in a majority of schizonts. Occurring alongside sexual commitment, the role of the discrete MSPDBL2-positive schizont subpopulation requires further investigation in natural infections where it is under immune selection. IMPORTANCE: Malaria parasites in the blood are remarkably variable, able to switch antigenic targets so they may survive within humans who have already developed specific immune responses. This is one of the challenges in developing vaccines against malaria. MSPDBL2 is a target of naturally acquired immunity expressed in minority proportions of schizonts, the end stages of each 2-day replication cycle in red blood cells which contain merozoites prepared to invade new red blood cells. Results show that the proportion of schizonts expressing MSPDBL2 is positively controlled by the expression of the regulatory gametocyte development protein GDV1. It was previously known that expression of GDV1 leads to increased expression of AP2-G which causes parasites to switch to sexual development, so a surprising finding here is that MSPDBL2-positive parasites are mostly distinct from those that express AP2-G. This discrete antigenic subpopulation of mostly asexual parasites is regulated alongside sexually committed parasites, potentially enabling survival under stress conditions.


Subject(s)
Antigens, Protozoan , Plasmodium falciparum , Protozoan Proteins , Schizonts , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , Schizonts/metabolism , Schizonts/immunology , Schizonts/genetics , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/immunology , Gene Expression Regulation , Erythrocytes/parasitology
2.
PLoS Pathog ; 19(3): e1011281, 2023 03.
Article in English | MEDLINE | ID: mdl-37000891

ABSTRACT

During the blood stage of a malaria infection, malaria parasites export both soluble and membrane proteins into the erythrocytes in which they reside. Exported proteins are trafficked via the parasite endoplasmic reticulum and secretory pathway, before being exported across the parasitophorous vacuole membrane into the erythrocyte. Transport across the parasitophorous vacuole membrane requires protein unfolding, and in the case of membrane proteins, extraction from the parasite plasma membrane. We show that trafficking of the exported Plasmodium protein, Pf332, differs from that of canonical eukaryotic soluble-secreted and transmembrane proteins. Pf332 is initially ER-targeted by an internal hydrophobic sequence that unlike a signal peptide, is not proteolytically removed, and unlike a transmembrane segment, does not span the ER membrane. Rather, both termini of the hydrophobic sequence enter the ER lumen and the ER-lumenal species is a productive intermediate for protein export. Furthermore, we show in intact cells, that two other exported membrane proteins, SBP1 and MAHRP2, assume a lumenal topology within the parasite secretory pathway. Although the addition of a C-terminal ER-retention sequence, recognised by the lumenal domain of the KDEL receptor, does not completely block export of SBP1 and MAHRP2, it does enhance their retention in the parasite ER. This indicates that a sub-population of each protein adopts an ER-lumenal state that is an intermediate in the export process. Overall, this suggests that although many exported proteins traverse the parasite secretory pathway as typical soluble or membrane proteins, some exported proteins that are ER-targeted by a transmembrane segment-like, internal, non-cleaved hydrophobic segment, do not integrate into the ER membrane, and form an ER-lumenal species that is a productive export intermediate. This represents a novel means, not seen in typical membrane proteins found in model systems, by which exported transmembrane-like proteins can be targeted and trafficked within the lumen of the secretory pathway.


Subject(s)
Malaria , Plasmodium , Humans , Protein Transport , Protozoan Proteins/metabolism , Plasmodium/metabolism , Endoplasmic Reticulum/metabolism , Erythrocytes/parasitology , Malaria/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism
3.
mBio ; 13(4): e0194822, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35950755

ABSTRACT

The merozoite surface protein MSPDBL2 of Plasmodium falciparum is under strong balancing selection and is a target of naturally acquired antibodies. Remarkably, MSPDBL2 is expressed in only a minority of mature schizonts of any cultured parasite line, and mspdbl2 gene transcription increases in response to overexpression of the gametocyte development inducer GDV1, so it is important to understand its natural expression. Here, MSPDBL2 in mature schizonts was analyzed in the first ex vivo culture cycle of 96 clinical isolates from 4 populations with various levels of infection endemicity in different West African countries, by immunofluorescence microscopy with antibodies against a conserved region of the protein. In most isolates, less than 1% of mature schizonts were positive for MSPDBL2, but the frequency distribution was highly skewed, as nine isolates had more than 3% schizonts positive and one had 73% positive. To investigate whether the expression of other gene loci correlated with MSPDBL2 expression, whole-transcriptome sequencing was performed on schizont-enriched material from 17 of the isolates with a wide range of proportions of schizonts positive. Transcripts of particular genes were highly significantly positively correlated with MSPDBL2 positivity in schizonts as well as with mspdbl2 gene transcript levels, showing overrepresentation of genes implicated previously as involved in gametocytogenesis but not including the gametocytogenesis master regulator ap2-g. Single-cell transcriptome analysis of a laboratory-adapted clone showed that most individual parasites expressing mspdbl2 did not express ap2-g, consistent with MSPDBL2 marking a developmental subpopulation that is distinct but likely to co-occur alongside sexual commitment. IMPORTANCE These findings contribute to understanding malaria parasite antigenic and developmental variation, focusing on the merozoite surface protein encoded by the single locus under strongest balancing selection. Analyzing the initial ex vivo generation of parasites grown from a wide sample of clinical infections, we show a unique and highly skewed pattern of natural expression frequencies of MSPDBL2, distinct from that of any other antigen. Bulk transcriptome analysis of a range of clinical isolates showed significant overrepresentation of sexual development genes among those positively correlated with MSPDBL2 protein and mspdbl2 gene expression, indicating the MSPDBL2-positive subpopulation to be often coincident with parasites developing sexually in preparation for transmission. Single-cell transcriptome data confirm the absence of a direct correlation with the ap2-g master regulator of sexual development, indicating that the MSPDBL2-positive subpopulation has a separate function in asexual survival and replication under conditions that promote terminal sexual differentiation.


Subject(s)
Malaria, Falciparum , Parasites , Animals , Malaria, Falciparum/parasitology , Membrane Proteins/genetics , Merozoites , Parasites/genetics , Plasmodium falciparum , Protozoan Proteins/metabolism , Schizonts/genetics , Transcriptome
4.
Sci Rep ; 10(1): 245, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937828

ABSTRACT

The central role that erythrocyte invasion plays in Plasmodium falciparum survival and reproduction makes this process an attractive target for therapeutic or vaccine development. However, multiple invasion-related genes with complementary and overlapping functions afford the parasite the plasticity to vary ligands used for invasion, leading to phenotypic variation and immune evasion. Overcoming the challenge posed by redundant ligands requires a deeper understanding of conditions that select for variant phenotypes and the molecular mediators. While host factors including receptor heterogeneity and acquired immune responses may drive parasite phenotypic variation, we have previously shown that host-independent changes in invasion phenotype can be achieved by continuous culturing of the W2mef and Dd2 P. falciparum strains in moving suspension as opposed to static conditions. Here, we have used a highly biologically replicated whole transcriptome sequencing approach to identify the molecular signatures of variation associated with the phenotype switch. The data show increased expression of particular invasion-related genes in switched parasites, as well as a large number of genes encoding proteins that are either exported or form part of the export machinery. The genes with most markedly increased expression included members of the erythrocyte binding antigens (EBA), reticulocyte binding homologues (RH), surface associated interspersed proteins (SURFIN), exported protein family 1 (EPF1) and Plasmodium Helical Interspersed Sub-Telomeric (PHIST) gene families. The data indicate changes in expression of a repertoire of genes not previously associated with erythrocyte invasion phenotypes, suggesting the possibility that moving suspension culture may also select for other traits.


Subject(s)
Erythrocytes/parasitology , Gene Expression Profiling , Phenotype , Plasmodium falciparum/physiology , Epigenesis, Genetic , Humans
5.
Biochem J ; 477(2): 525-540, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31942933

ABSTRACT

Subtilisin-like serine peptidases (subtilases) play important roles in the life cycle of many organisms, including the protozoan parasites that are the causative agent of malaria, Plasmodium spp. As with other peptidases, subtilase proteolytic activity has to be tightly regulated in order to prevent potentially deleterious uncontrolled protein degradation. Maturation of most subtilases requires the presence of an N-terminal propeptide that facilitates folding of the catalytic domain. Following its proteolytic cleavage, the propeptide acts as a transient, tightly bound inhibitor until its eventual complete removal to generate active protease. Here we report the identification of a stand-alone malaria parasite propeptide-like protein, called SUB1-ProM, encoded by a conserved gene that lies in a highly syntenic locus adjacent to three of the four subtilisin-like genes in the Plasmodium genome. Template-based modelling and ab initio structure prediction showed that the SUB1-ProM core structure is most similar to the X-ray crystal structure of the propeptide of SUB1, an essential parasite subtilase that is discharged into the parasitophorous vacuole (PV) to trigger parasite release (egress) from infected host cells. Recombinant Plasmodium falciparum SUB1-ProM was found to be a fast-binding, potent inhibitor of P. falciparum SUB1, but not of the only other essential blood-stage parasite subtilase, SUB2, or of other proteases examined. Mass-spectrometry and immunofluorescence showed that SUB1-ProM is expressed in the PV of blood stage P. falciparum, where it may act as an endogenous inhibitor to regulate SUB1 activity in the parasite.


Subject(s)
Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Serine Proteases/chemistry , Subtilisin/genetics , Amino Acid Sequence/genetics , Animals , Erythrocytes/parasitology , Genome/genetics , Humans , Life Cycle Stages/genetics , Malaria, Falciparum/enzymology , Malaria, Falciparum/parasitology , Peptide Hydrolases/chemistry , Peptide Hydrolases/genetics , Plasmodium falciparum/pathogenicity , Proteolysis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Serine Proteases/genetics , Subtilisin/chemistry , Vacuoles/parasitology
6.
BMC Genomics ; 19(1): 894, 2018 Dec 10.
Article in English | MEDLINE | ID: mdl-30526479

ABSTRACT

BACKGROUND: Malaria parasites are genetically polymorphic and phenotypically plastic. In studying transcriptome variation among parasites from different infections, it is challenging to overcome potentially confounding technical and biological variation between samples. We investigate variation in the major human parasite Plasmodium falciparum, generating RNA-seq data on multiple independent replicate sample preparations of merozoite-containing intra-erythrocytic schizonts from a panel of clinical isolates and from long-term laboratory-adapted clones, with a goal of robustly identifying differentially expressed genes. RESULTS: Analysis of biological sample replicates shows that increased numbers improve the true discovery rate of differentially expressed genes, and that six independent replicates of each parasite line allowed identification of most differences that could be detected with larger numbers. For highly expressed genes, focusing on the top quartile at schizont stages, there was more power to detect differences. Comparing cultured clinical isolates and laboratory-adapted clones, genes more highly expressed in the laboratory-adapted clones include those encoding an AP2 transcription factor (PF3D7_0420300), a ubiquitin-binding protein and two putative methyl transferases. In contrast, higher expression in clinical isolates was seen for the merozoite surface protein gene dblmsp2, proposed to be a marker of schizonts forming merozoites committed to sexual differentiation. Variable expression was extremely strongly, but not exclusively, associated with genes known to be targeted by Heterochromatin Protein 1. Clinical isolates show variable expression of several known merozoite invasion ligands, as well as other genes for which new RT-qPCR assays validate the quantitation and allow characterisation in samples with more limited material. Expression levels of these genes vary among schizont preparations of different clinical isolates in the first ex vivo cycle in patient erythrocytes, but mean levels are similar to those in continuously cultured clinical isolates. CONCLUSIONS: Analysis of multiple biological sample replicates greatly improves identification of genes variably expressed between different cultured parasite lines. Clinical isolates recently established in culture show differences from long-term adapted clones in transcript levels of particular genes, and are suitable for analyses requiring biological replicates to understand parasite phenotypes and variable expression likely to be relevant in nature.


Subject(s)
Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Schizonts/genetics , Transcriptome/genetics , Adolescent , Animals , Child , Child, Preschool , Gene Expression Profiling , Humans , Parasites/isolation & purification , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schizonts/isolation & purification
7.
Sci Rep ; 8(1): 15763, 2018 10 25.
Article in English | MEDLINE | ID: mdl-30361631

ABSTRACT

Parasites infect hosts in widely varying environments, encountering diverse challenges for adaptation. To identify malaria parasite genes under locally divergent selection across a large endemic region with a wide spectrum of transmission intensity, genome sequences were obtained from 284 clinical Plasmodium falciparum infections from four newly sampled locations in Senegal, The Gambia, Mali and Guinea. Combining these with previous data from seven other sites in West Africa enabled a multi-population analysis to identify discrete loci under varying local selection. A genome-wide scan showed the most exceptional geographical divergence to be at the early gametocyte gene locus gdv1 which is essential for parasite sexual development and transmission. We identified a major structural dimorphism with alternative 1.5 kb and 1.0 kb sequence deletions at different positions of the 3'-intergenic region, in tight linkage disequilibrium with the most highly differentiated single nucleotide polymorphism, one of the alleles being very frequent in Senegal and The Gambia but rare in the other locations. Long non-coding RNA transcripts were previously shown to include the entire antisense of the gdv1 coding sequence and the portion of the intergenic region with allelic deletions, suggesting adaptive regulation of parasite sexual development and transmission in response to local conditions.


Subject(s)
Genetic Loci , Malaria, Falciparum/parasitology , Metagenomics/methods , Parasites/genetics , Selection, Genetic , Sexual Development/genetics , Africa, Western , Alleles , Animals , Base Sequence , Gene Frequency/genetics , Genetic Variation , Genome , Geography , Haplotypes/genetics , Homozygote , Humans , Malaria, Falciparum/genetics , Plasmodium falciparum/genetics , Polymorphism, Single Nucleotide/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Sci Rep ; 7(1): 6436, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743888

ABSTRACT

It is important to understand intrinsic variation in asexual blood stage multiplication rates of the most virulent human malaria parasite, Plasmodium falciparum. Here, multiplication rates of long-term laboratory adapted parasite clones and new clinical isolates were measured, using a newly standardised assay of growth from low starting density in replicate parallel cultures with erythrocytes from multiple different donors, across multiple cycles. Multiplication rates of long-term established clones were between 7.6 and 10.5 fold per 48 hours, with clone Dd2 having a higher rate than others (clones 3D7, HB3 and D10). Parasite clone-specific growth was then analysed in co-culture assays with all possible heterologous pairwise combinations. This showed that co-culture of different parasites did not affect their replication rates, indicating that there were no suppressive interactions operating between parasites. Multiplication rates of eleven new clinical isolates were measured after a few weeks of culture, and showed a spectrum of replication rates between 2.3 and 6.0 fold per 48 hours, the entire range being lower than for the long-term laboratory adapted clones. Multiplication rate estimates remained stable over time for several isolates tested repeatedly up to three months after culture initiation, indicating considerable persistence of this important trait variation.


Subject(s)
Malaria, Falciparum/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Coculture Techniques , Erythrocytes/parasitology , Humans , Microsatellite Repeats , Plasmodium falciparum/isolation & purification
9.
PLoS One ; 10(4): e0121786, 2015.
Article in English | MEDLINE | ID: mdl-25849462

ABSTRACT

The malaria parasite exports hundreds of proteins into its host cell. The majority of exported proteins contain a Host-Targeting motif (also known as a Plasmodium export element) that directs them for export. Prior to export, the Host-Targeting motif is cleaved by the endoplasmic reticulum-resident protease Plasmepsin V and the newly generated N-terminus is N-α-acetylated by an unidentified enzyme. The cleaved, N-α-acetylated protein is trafficked to the parasitophorous vacuole, where it is translocated across the vacuole membrane. It is clear that cleavage and N-α-acetylation of the Host-Targeting motif occur at the endoplasmic reticulum, and it has been proposed that Host-Targeting motif cleavage and N-α-acetylation occur either on the luminal or cytosolic side of the endoplasmic reticulum membrane. Here, we use self-associating 'split' fragments of GFP to determine the topology of Plasmepsin V in the endoplasmic reticulum membrane; we show that the catalytic protease domain of Plasmepsin V faces the endoplasmic reticulum lumen. These data support a model in which the Host-Targeting motif is cleaved and N-α-acetylated in the endoplasmic reticulum lumen. Furthermore, these findings suggest that cytosolic N-α-acetyltransferases are unlikely to be candidates for the N-α-acetyltransferase of Host-Targeting motif-containing exported proteins.


Subject(s)
Aspartic Acid Endopeptidases/metabolism , Endoplasmic Reticulum/enzymology , Intracellular Membranes/enzymology , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Vacuoles/enzymology , Acetylation , Amino Acid Motifs , Aspartic Acid Endopeptidases/genetics , Endoplasmic Reticulum/genetics , Plasmodium falciparum/genetics , Protein Transport/physiology , Protozoan Proteins/genetics , Vacuoles/genetics
10.
Mol Biochem Parasitol ; 196(1): 29-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25106850

ABSTRACT

During blood-stage infection, malaria parasites export numerous proteins to the host erythrocyte. The Poly-Helical Interspersed Sub-Telomeric (PHIST) proteins are an exported family that share a common 'PRESAN' domain, and include numerous members in Plasmodium falciparum, Plasmodium vivax and Plasmodium knowlesi. In P. falciparum, PHIST proteins have been implicated in protein trafficking and intercellular communication. A number of PHIST proteins are essential for parasite survival. Here, we identify nine members of the PHISTb sub-class of PHIST proteins, including one protein known to be essential for parasite survival, that localise to the erythrocyte periphery. These proteins have solubility characteristics consistent with their association with the erythrocyte cytoskeleton. Together, an extended PRESAN domain, comprising the PRESAN domain and preceding sequence, form a novel targeting-domain that is sufficient to localise a protein to the erythrocyte periphery. We validate the role of this domain in RESA, thus identifying a cytoskeleton-binding domain in RESA that functions independently of its known spectrin-binding domain. Our data suggest that some PHISTb proteins may act as cross-linkers of the erythrocyte cytoskeleton. We also show for the first time that peripherally-localised PHISTb proteins are encoded in genomes of P. knowlesi and vivax indicating a conserved role for the extended PRESAN domain of these proteins in targeting to the erythrocyte periphery.


Subject(s)
Erythrocytes/metabolism , Erythrocytes/parasitology , Plasmodium falciparum/metabolism , Protein Transport , Protozoan Proteins/metabolism , Cytoskeleton/metabolism , Plasmodium falciparum/genetics , Plasmodium knowlesi/genetics , Plasmodium knowlesi/metabolism , Plasmodium vivax/genetics , Plasmodium vivax/metabolism , Protein Binding , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Solubility
11.
Mol Microbiol ; 87(4): 835-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23279267

ABSTRACT

The malaria parasite exports proteins across its plasma membrane and a surrounding parasitophorous vacuole membrane, into its host erythrocyte. Most exported proteins contain a Host Targeting motif (HT motif) that targets them for export. In the parasite secretory pathway, the HT motif is cleaved by the protease plasmepsin V, but the role of the newly generated N-terminal sequence in protein export is unclear. Using a model protein that is cleaved by an exogenous viral protease, we show that the new N-terminal sequence, normally generated by plasmepsin V cleavage, is sufficient to target a protein for export, and that cleavage by plasmepsin V is not coupled directly to the transfer of a protein to the next component in the export pathway. Mutation of the fourth and fifth positions of the HT motif, as well as amino acids further downstream, block or affect the efficiency of protein export indicating that this region is necessary for efficient export. We also show that the fifth position of the HT motif is important for plasmepsin V cleavage. Our results indicate that plasmepsin V cleavage is required to generate a new N-terminal sequence that is necessary and sufficient to mediate protein export by the malaria parasite.


Subject(s)
Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Aspartic Acid Endopeptidases/metabolism , Host-Parasite Interactions , Humans , Malaria, Falciparum/enzymology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Protein Processing, Post-Translational , Protein Transport , Protozoan Proteins/genetics
12.
Protist ; 163(5): 755-66, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22503086

ABSTRACT

The mitochondrial electron transport chain is essential to Plasmodium and is the target of the antimalarial drug atovaquone. The mitochondrial genomes of Plasmodium sp. are the most reduced known, and the majority of mitochondrial proteins are encoded in the nucleus and imported into the mitochondrion post-translationally. Many organisms have signalling pathways between the mitochondria and the nucleus to regulate the expression of nuclear-encoded mitochondrially-targeted proteins, for example in response to mitochondrial dysfunction. We have studied the transcript profiles of synchronous Plasmodium falciparum treated with an LD(50) concentration of the complex III inhibitor antimycin A, to investigate whether such pathways exist in the parasite. There was a broad perturbation of gene expression. The differentially expressed genes were enriched for transcripts encoding proteins involved in invasion, stress response, nucleotide biosynthesis and respiration. Some effects were attributable to a delay in the gene expression phase of drug-treated parasites. However, our data indicated regulation of mitochondrial stress response genes and genes involved in pyrimidine biosynthesis, implying the existence of a signalling pathway from the mitochondrion to the nucleus.


Subject(s)
Antimalarials/pharmacology , Antimycin A/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Gene Expression Profiling , Mitochondria/drug effects , Plasmodium falciparum/drug effects , Mitochondria/enzymology , Plasmodium falciparum/enzymology , Signal Transduction , Stress, Physiological
13.
Mol Biochem Parasitol ; 179(1): 37-41, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21620902

ABSTRACT

The antimalarial activity of the antibiotic thiostrepton has long been attributed to inhibition of apicoplast protein synthesis through binding of apicoplast ribosomal RNA. However, the kinetics of parasite death upon thiostrepton treatment differ from those seen for other inhibitors of apicoplast housekeeping functions. We have analysed global changes in gene expression of the malaria parasite, Plasmodium falciparum, in an attempt to shed light on the responses of the parasite to this drug. Our results indicate a delay in gene expression profiles of thiostrepton-treated parasites. A small number of genes appear to be regulated outside of this trend; our data suggest a response from genes encoding components of the mitochondrial translational machinery, while little response is seen from genes encoding apicoplast-targeted proteins. Our findings are consistent with an effect of thiostrepton on mitochondrial protein synthesis, and thus warrant a re-evaluation of the target of thiostrepton in Plasmodium. They also provide some suggestion of mitochondrion-nucleus signalling in the parasite.


Subject(s)
Antimalarials/metabolism , Gene Expression Profiling , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Thiostrepton/metabolism , Mitochondria/drug effects , Mitochondria/genetics , Mitochondrial Proteins/biosynthesis
14.
Philos Trans R Soc Lond B Biol Sci ; 365(1541): 785-97, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20124345

ABSTRACT

Protist mitochondrial genomes show a very wide range of gene content, ranging from three genes for respiratory chain components in Apicomplexa and dinoflagellates to nearly 100 genes in Reclinomonas americana. In many organisms the rRNA genes are fragmented, although still functional. Some protist mitochondria encode a full set of tRNAs, while others rely on imported molecules. There is similarly a wide variation in mitochondrial genome organization, even among closely related groups. Mitochondrial gene expression and control are generally poorly characterized. Transcription probably relies on a 'viral-type' RNA polymerase, although a 'bacterial-type' enzyme may be involved in some cases. Transcripts are heavily edited in many lineages. The chloroplast genome generally shows less variation in gene content and organization, although greatly reduced genomes are found in dinoflagellate algae and non-photosynthetic organisms. Genes in the former are located on small plasmids in contrast to the larger molecules found elsewhere. Control of gene expression in chloroplasts involves transcriptional and post-transcriptional regulation. Redox poise and the ATP/ADP ratio are likely to be important determinants. Some protists have an additional extranuclear genome, the nucleomorph, which is a remnant nucleus. Nucleomorphs of two separate lineages have a number of features in common.


Subject(s)
Biological Evolution , Genome , Organelles/genetics , Gene Expression , Genome, Chloroplast , Genome, Mitochondrial , RNA/genetics , Symbiosis/genetics
15.
BMC Microbiol ; 6: 45, 2006 May 18.
Article in English | MEDLINE | ID: mdl-16707026

ABSTRACT

BACKGROUND: Giardia intestinalis is a parasitic protozoan and major cause of diarrhoeal disease. Disease transmission is dependent on the ability of the parasite to differentiate back and forth between an intestine-colonising trophozoite and an environmentally-resistant infective cyst. Our current understanding of the intracellular signalling mechanisms that regulate parasite replication and differentiation is limited, yet such information could suggest new methods of disease control. Phosphoinositide-3 kinase (PI3K) signalling pathways have a central involvement in many vital eukaryotic processes, such as regulation of cell growth, intracellular membrane trafficking and cell motility. Here we present evidence for the existence of functional PI3K intracellular signalling pathways in G. intestinalis. RESULTS: We have identified and characterised two genes, Gipi3k1 and Gipi3k2, which encode putative PI3Ks. Both genes are expressed in trophozoites and encysting cells, suggesting a possible role of GiPI3K1 and GiPI3K2 in regulating giardial growth and differentiation. Extensive nucleotide and amino acid sequence characterisation predicts that both encoded PI3Ks are functional as indicated by the presence of highly conserved structural domains and essential catalytic residues. The inhibitory effect of the PI3K inhibitor LY294002 on trophozoite proliferation also supports their functionality. Phylogenetic analysis supports the identity of GiPI3K1 as a Class I isoform and GiPI3K2 as a Class III isoform. In addition, giardial genes encoding putative homologues of phosphoinositide-metabolising enzymes such as PTEN, MTM, PIPkin and PI 5-phosphatase as well as downstream effectors with phosphoinositide-binding domains have been identified, placing GiPI3K1 and GiPI3K2 in a broader signalling context. Compared with twenty-six PI3Ks from other organisms, GiPI3K1 and GiPI3K2 are unique in that they contain large insertions within their highly conserved kinase domains. The function of these insertions is unknown; however we show here that they are not intron-derived and would probably not hinder substrate binding. These insertions may represent a plausible drug target. CONCLUSION: G. intestinalis encodes and expresses two putative PI3Ks, at least one of which appears to be required during normal parasite proliferation. The identification of Class I and Class III but not Class II isoforms suggests that both extracellularly-initiated signalling (Class I-regulated) and intracellular vesicle trafficking (Class III-regulated) might be controlled by PI3Ks in G. intestinalis. The presence of genes encoding putative homologues of phosphoinositide-metabolising enzymes and downstream effectors in the G. intestinalis genome further suggests that the overall architecture of PI3K signalling may be comparable with pathways present in other better-studied organisms.


Subject(s)
Giardia lamblia/enzymology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Chromones/pharmacology , Computational Biology , Enzyme Inhibitors/pharmacology , Gene Expression , Giardia lamblia/genetics , Giardia lamblia/growth & development , Molecular Sequence Data , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Structure, Tertiary , Protozoan Proteins/antagonists & inhibitors , Sequence Alignment , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...