Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Evolution ; 78(7): 1287-1301, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38581661

ABSTRACT

For parasites, robust proliferation within hosts is crucial for establishing the infection and creating opportunities for onward transmission. While faster proliferation enhances transmission rates, it is often assumed to curtail transmission duration by killing the host (virulence), a trade-off constraining parasite evolution. Yet in many diseases, including malaria, the preponderance of infections with mild or absent symptoms suggests that host mortality is not a sufficient constraint, raising the question of what restrains evolution toward faster proliferation. In malaria infections, the maximum rate of proliferation is determined by the burst size, the number of daughter parasites produced per infected red blood cell. Larger burst sizes should expand the pool of infected red blood cells that can be used to produce the specialized transmission forms needed to infect mosquitoes. We use a within-host model parameterized for rodent malaria parasites (Plasmodium chabaudi) to project the transmission consequences of burst size, focusing on initial acute infection where resource limitation and risk of host mortality are greatest. We find that resource limitation restricts evolution toward higher burst sizes below the level predicted by host mortality alone. Our results suggest resource limitation could represent a more general constraint than virulence-transmission trade-offs, preventing evolution towards faster proliferation.


Subject(s)
Malaria , Plasmodium chabaudi , Animals , Virulence , Plasmodium chabaudi/genetics , Plasmodium chabaudi/pathogenicity , Plasmodium chabaudi/physiology , Malaria/transmission , Malaria/parasitology , Malaria/prevention & control , Host-Parasite Interactions , Biological Evolution , Erythrocytes/parasitology , Models, Biological
2.
BMC Res Notes ; 16(1): 56, 2023 Apr 19.
Article in English | MEDLINE | ID: mdl-37076932

ABSTRACT

OBJECTIVE: To analyse the transcriptional profiles of the pir multigene family of Plasmodium chabaudi chabaudi in male and female gametocytes isolated from the blood of infected mice. RESULTS: Infected red blood cells containing female and male P. chabaudi gametocytes transcribe a distinct set of genes encoded by the multigene family pir. The overall patterns are similar to what has been observed in the close relative P. berghei, but here we show that gametocyte-associated pir genes are distinct from those involved in chronic blood-stage infection and highlight a male-associated pir gene which should be the focus of future studies.


Subject(s)
Malaria , Parasites , Plasmodium chabaudi , Male , Female , Animals , Mice , Plasmodium chabaudi/genetics , Malaria/parasitology
3.
Parasitol Int ; 91: 102623, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35803536

ABSTRACT

Malaria is a devastating disease that still claims over half a million lives every year, mostly in sub-Saharan Africa. One of the main barriers to malaria control is the evolution and propagation of drug-resistant mutant parasites. Knowing the genes and respective mutations responsible for drug resistance facilitates the design of drugs with novel modes of action and allows predicting and monitoring drug resistance in natural parasite populations in real-time. The best way to identify these mutations is to experimentally evolve resistance to the drug in question and then comparing the genomes of the drug-resistant mutants to that of the sensitive progenitor parasites. This simple evolutive concept was the starting point for the development of a paradigm over the years, based on the use of the rodent malaria parasite Plasmodium chabaudi to unravel the genetics of drug resistance in malaria. It involves the use of a cloned parasite isolate (P. chabaudi AS) whose genome is well characterized, to artificially select resistance to given drugs through serial passages in mice under slowly increasing drug pressure. The end resulting parasites are cloned and the genetic mutations are then discovered through Linkage Group Selection, a technique conceived by Prof. Richard Carter and his group, and/or Whole Genome Sequencing. The precise role of these mutations can then be interrogated in malaria parasites of humans through allelic replacement experiments and/or genotype-phenotype association studies in natural parasite populations. Using this paradigm, all the mutations underlying resistance to the most important antimalarial drugs were identified, most of which were pioneering and later shown to also play a role in drug resistance in natural infections of human malaria parasites. This supports the use of P. chabaudi a fast-track predictive model to identify candidate genetic markers of resistance to present and future antimalarial drugs and improving our understanding of the biology of resistance.


Subject(s)
Antimalarials , Malaria , Parasites , Plasmodium chabaudi , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Drug Resistance/genetics , Humans , Malaria/parasitology , Mice , Plasmodium chabaudi/genetics , Rodentia
4.
Malar J ; 20(1): 445, 2021 Nov 25.
Article in English | MEDLINE | ID: mdl-34823519

ABSTRACT

BACKGROUND: Plasmodium interspersed repeat (pir) is the largest multigene family in the genomes of most Plasmodium species. A variety of functions for the PIR proteins which they encode have been proposed, including antigenic variation, immune evasion, sequestration and rosetting. However, direct evidence for these is lacking. The repetitive nature of the family has made it difficult to determine function experimentally. However, there has been some success in using gene expression studies to suggest roles for some members in virulence and chronic infection. METHODS: Here pir gene expression was examined across the life cycle of Plasmodium berghei using publicly available RNAseq data-sets, and at high resolution in the intraerythrocytic development cycle using new data from Plasmodium chabaudi. RESULTS: Expression of pir genes is greatest in stages of the parasite which invade and reside in red blood cells. The marked exception is that liver merozoites and male gametocytes produce a very large number of pir gene transcripts, notably compared to female gametocytes, which produce relatively few. Within the asexual blood stages different subfamilies peak at different times, suggesting further functional distinctions. Representing a subfamily of its own, the highly conserved ancestral pir gene warrants further investigation due to its potential tractability for functional investigation. It is highly transcribed in multiple life cycle stages and across most studied Plasmodium species and thus is likely to play an important role in parasite biology. CONCLUSIONS: The identification of distinct expression patterns for different pir genes and subfamilies is likely to provide a basis for the design of future experiments to uncover their function.


Subject(s)
Gene Expression , Genes, Protozoan , Life Cycle Stages/genetics , Multigene Family , Plasmodium berghei/genetics , Plasmodium chabaudi/genetics
5.
Mol Biochem Parasitol ; 244: 111375, 2021 07.
Article in English | MEDLINE | ID: mdl-34023299

ABSTRACT

Malaria parasites exhibit a complex lifecycle, requiring extensive asexual replication in the liver and blood of the vertebrate host, and in the haemocoel of the insect vector. Yet, they must also undergo a single round of sexual reproduction, which occurs in the vector's midgut upon uptake of a blood meal. Sexual reproduction is obligate for infection of the vector and thus, is essential for onwards transmission to new hosts. Sex in malaria parasites involves several bottlenecks in parasite number, making the stages involved attractive targets for blocking disease transmission. Malaria parasites have evolved a suite of adaptations ("strategies") to maximise the success of sexual reproduction and transmission, which could undermine transmission-blocking interventions. Yet, understanding parasite strategies may also reveal novel opportunities for such interventions. Here, we outline how evolutionary and ecological theories, developed to explain reproductive strategies in multicellular taxa, can be applied to explain two reproductive strategies (conversion rate and sex ratio) expressed by malaria parasites within the vertebrate host.


Subject(s)
Gametogenesis , Life Cycle Stages/genetics , Malaria/parasitology , Plasmodium berghei/growth & development , Plasmodium chabaudi/growth & development , Plasmodium falciparum/growth & development , Plasmodium knowlesi/growth & development , Animals , Biological Coevolution , Culicidae/parasitology , Erythrocytes/parasitology , Female , Host-Parasite Interactions/genetics , Humans , Insect Vectors/parasitology , Liver/parasitology , Malaria/transmission , Male , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Plasmodium chabaudi/genetics , Plasmodium chabaudi/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium knowlesi/genetics , Plasmodium knowlesi/metabolism , Reproduction, Asexual , Sex Ratio
6.
Proc Natl Acad Sci U S A ; 117(50): 32098-32104, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33257570

ABSTRACT

The deadly symptoms of malaria occur as Plasmodium parasites replicate within blood cells. Members of several variant surface protein families are expressed on infected blood cell surfaces. Of these, the largest and most ubiquitous are the Plasmodium-interspersed repeat (PIR) proteins, with more than 1,000 variants in some genomes. Their functions are mysterious, but differential pir gene expression associates with acute or chronic infection in a mouse malaria model. The membership of the PIR superfamily, and whether the family includes Plasmodium falciparum variant surface proteins, such as RIFINs and STEVORs, is controversial. Here we reveal the structure of the extracellular domain of a PIR from Plasmodium chabaudi We use structure-guided sequence analysis and molecular modeling to show that this fold is found across PIR proteins from mouse- and human-infective malaria parasites. Moreover, we show that RIFINs and STEVORs are not PIRs. This study provides a structure-guided definition of the PIRs and a molecular framework to understand their evolution.


Subject(s)
Plasmodium chabaudi/ultrastructure , Protein Domains/immunology , Protozoan Proteins/ultrastructure , Repetitive Sequences, Amino Acid/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/ultrastructure , Circular Dichroism , Genome, Protozoan/genetics , Humans , Malaria/immunology , Malaria/virology , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/ultrastructure , Multigene Family/genetics , Multigene Family/immunology , Phylogeny , Plasmodium chabaudi/genetics , Plasmodium chabaudi/immunology , Protein Domains/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Repetitive Sequences, Amino Acid/genetics
7.
Nat Commun ; 11(1): 2763, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32488076

ABSTRACT

Malaria parasites complete their intra-erythrocytic developmental cycle (IDC) in multiples of 24 h suggesting a circadian basis, but the mechanism controlling this periodicity is unknown. Combining in vivo and in vitro approaches utilizing rodent and human malaria parasites, we reveal that: (i) 57% of Plasmodium chabaudi genes exhibit daily rhythms in transcription; (ii) 58% of these genes lose transcriptional rhythmicity when the IDC is out-of-synchrony with host rhythms; (iii) 6% of Plasmodium falciparum genes show 24 h rhythms in expression under free-running conditions; (iv) Serpentine receptor 10 (SR10) has a 24 h transcriptional rhythm and disrupting it in rodent malaria parasites shortens the IDC by 2-3 h; (v) Multiple processes including DNA replication, and the ubiquitin and proteasome pathways, are affected by loss of coordination with host rhythms and by disruption of SR10. Our results reveal malaria parasites are at least partly responsible for scheduling the IDC and coordinating their development with host daily rhythms.


Subject(s)
Circadian Rhythm/physiology , Erythropoiesis/physiology , Host-Parasite Interactions/physiology , Malaria/metabolism , Protozoan Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Secologanin Tryptamine Alkaloids/metabolism , Animals , Caenorhabditis elegans Proteins , Disease Models, Animal , Female , Gene Expression , Host-Parasite Interactions/genetics , Humans , Malaria/parasitology , Mice , Mice, Knockout , Plasmodium chabaudi/genetics , Plasmodium chabaudi/growth & development , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Rodentia , Transcriptome
8.
Science ; 368(6492): 746-753, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32409471

ABSTRACT

Malarial rhythmic fevers are the consequence of the synchronous bursting of red blood cells (RBCs) on completion of the malaria parasite asexual cell cycle. Here, we hypothesized that an intrinsic clock in the parasite Plasmodium chabaudi underlies the 24-hour-based rhythms of RBC bursting in mice. We show that parasite rhythms are flexible and lengthen to match the rhythms of hosts with long circadian periods. We also show that malaria rhythms persist even when host food intake is evenly spread across 24 hours, suggesting that host feeding cues are not required for synchrony. Moreover, we find that the parasite population remains synchronous and rhythmic even in an arrhythmic clock mutant host. Thus, we propose that parasite rhythms are generated by the parasite, possibly to anticipate its circadian environment.


Subject(s)
Circadian Rhythm/physiology , Fever/physiopathology , Fever/parasitology , Host-Parasite Interactions/physiology , Malaria/physiopathology , Malaria/parasitology , Plasmodium chabaudi/physiology , Animals , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Cues , Darkness , Eating , Erythrocytes/parasitology , Feeding Behavior , Gene Expression Regulation , Host-Parasite Interactions/genetics , Mice , Mice, Mutant Strains , Plasmodium chabaudi/genetics , Transcription, Genetic
9.
Malar J ; 19(1): 17, 2020 Jan 14.
Article in English | MEDLINE | ID: mdl-31937300

ABSTRACT

BACKGROUND: The intraerythrocytic development cycle (IDC) of the rodent malaria Plasmodium chabaudi is coordinated with host circadian rhythms. When this coordination is disrupted, parasites suffer a 50% reduction in both asexual stages and sexual stage gametocytes over the acute phase of infection. Reduced gametocyte density may not simply follow from a loss of asexuals because investment into gametocytes ("conversion rate") is a plastic trait; furthermore, the densities of both asexuals and gametocytes are highly dynamic during infection. Hence, the reasons for the reduction of gametocytes in infections that are out-of-synch with host circadian rhythms remain unclear. Here, two explanations are tested: first, whether out-of-synch parasites reduce their conversion rate to prioritize asexual replication via reproductive restraint; second, whether out-of-synch gametocytes experience elevated clearance by the host's circadian immune responses. METHODS: First, conversion rate data were analysed from a previous experiment comparing infections of P. chabaudi that were in-synch or 12 h out-of-synch with host circadian rhythms. Second, three new experiments examined whether the inflammatory cytokine TNF varies in its gametocytocidal efficacy according to host time-of-day and gametocyte age. RESULTS: There was no evidence that parasites reduce conversion or that their gametocytes become more vulnerable to TNF when out-of-synch with host circadian rhythms. CONCLUSIONS: The factors causing the reduction of gametocytes in out-of-synch infections remain mysterious. Candidates for future investigation include alternative rhythmic factors involved in innate immune responses and the rhythmicity in essential resources required for gametocyte development. Explaining why it matters for gametocytes to be synchronized to host circadian rhythms might suggest novel approaches to blocking transmission.


Subject(s)
Circadian Rhythm , Erythrocytes/parasitology , Malaria/parasitology , Plasmodium chabaudi/physiology , Tumor Necrosis Factor-alpha/administration & dosage , Animals , Circadian Rhythm/immunology , Female , Flow Cytometry , Gametogenesis/physiology , Linear Models , Malaria/blood , Malaria/immunology , Male , Merozoites/physiology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Plasmodium chabaudi/genetics , Plasmodium chabaudi/growth & development , Plasmodium chabaudi/immunology , Random Allocation , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
10.
Malar J ; 18(1): 222, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31262304

ABSTRACT

BACKGROUND: The ability of malaria (Plasmodium) parasites to adjust investment into sexual transmission stages versus asexually replicating stages is well known, but plasticity in other traits underpinning the replication rate of asexual stages in the blood has received less attention. Such traits include burst size (the number of merozoites produced per schizont), the duration of the asexual cycle, and invasion preference for different ages of red blood cell (RBC). METHODS: Here, plasticity [environment (E) effects] and genetic variation [genotype (G) effects] in traits relating to asexual replication rate are examined for 4 genotypes of the rodent malaria parasite Plasmodium chabaudi. An experiment tested whether asexual dynamics differ between parasites infecting control versus anaemic hosts, and whether variation in replication rate can be explained by differences in burst size, asexual cycle, and invasion rates. RESULTS: The within-host environment affected each trait to different extents but generally had similar impacts across genotypes. The dynamics of asexual densities exhibited a genotype by environment effect (G×E), in which one of the genotypes increased replication rate more than the others in anaemic hosts. Burst size and cycle duration varied between the genotypes (G), while burst size increased and cycle duration became longer in anaemic hosts (E). Variation in invasion rates of differently aged RBCs was not explained by environmental or genetic effects. Plasticity in burst size and genotype are the only traits making significant contributions to the increase in asexual densities observed in anaemic hosts, together explaining 46.4% of the variation in replication rate. CONCLUSIONS: That host anaemia induces several species of malaria parasites to alter conversion rate is well documented. Here, previously unknown plasticity in other traits underpinning asexual replication is revealed. These findings contribute to mounting evidence that malaria parasites deploy a suite of sophisticated strategies to maximize fitness by coping with, or exploiting the opportunities provided by, the variable within-host conditions experienced during infections. That genetic variation and genotype by environment interactions also shape these traits highlights their evolutionary potential. Asexual replication rate is a major determinant of virulence and so, understanding the evolution of virulence requires knowledge of the ecological (within-host environment) and genetic drivers of variation among parasites.


Subject(s)
Adaptation, Physiological/genetics , Gene-Environment Interaction , Genetic Variation/physiology , Plasmodium chabaudi/physiology , Reproduction, Asexual , Animals , Female , Mice , Mice, Inbred C57BL , Plasmodium chabaudi/genetics , Reproduction, Asexual/genetics
11.
Front Immunol ; 10: 3072, 2019.
Article in English | MEDLINE | ID: mdl-32038623

ABSTRACT

The distributions of human malaria parasite species overlap in most malarious regions of the world, and co-infections involving two or more malaria parasite species are common. Little is known about the consequences of interactions between species during co-infection for disease severity and parasite transmission success. Anti-malarial interventions can have disproportionate effects on malaria parasite species and may locally differentially reduce the number of species in circulation. Thus, it is important to have a clearer understanding of how the interactions between species affect disease and transmission dynamics. Controlled competition experiments using human malaria parasites are impossible, and thus we assessed the consequences of mixed-species infections on parasite fitness, disease severity, and transmission success using the rodent malaria parasite species Plasmodium chabaudi, Plasmodium yoelii, and Plasmodium vinckei. We compared the fitness of individual species within single species and co-infections in mice. We also assessed the disease severity of single vs. mixed infections in mice by measuring mortality rates, anemia, and weight loss. Finally, we compared the transmission success of parasites in single or mixed species infections by quantifying oocyst development in Anopheles stephensi mosquitoes. We found that co-infections of P. yoelii with either P. vinckei or P. chabaudi led to a dramatic increase in infection virulence, with 100% mortality observed in mixed species infections, compared to no mortality for P. yoelii and P. vinckei single infections, and 40% mortality for P. chabaudi single infections. The increased mortality in the mixed infections was associated with an inability to clear parasitaemia, with the non-P. yoelii parasite species persisting at higher parasite densities than in single infections. P. yoelii growth was suppressed in all mixed infections compared to single infections. Transmissibility of P. vinckei and P. chabaudi to mosquitoes was also reduced in the presence of P. yoelii in co-infections compared to single infections. The increased virulence of co-infections containing P. yoelii (reticulocyte restricted) and P. chabaudi or P. vinckei (predominantly normocyte restricted) may be due to parasite cell tropism and/or immune modulation of the host. We explain the reduction in transmission success of species in co-infections in terms of inter-species gamete incompatibility.


Subject(s)
Coinfection , Host-Parasite Interactions , Malaria/parasitology , Malaria/transmission , Mosquito Vectors/parasitology , Animals , Disease Models, Animal , Female , Host-Parasite Interactions/immunology , Malaria/immunology , Malaria/mortality , Mice , Parasite Load , Plasmodium chabaudi/classification , Plasmodium chabaudi/genetics , Plasmodium yoelii/classification , Plasmodium yoelii/genetics , Virulence
12.
Sci Rep ; 8(1): 2464, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29410428

ABSTRACT

Antimalarial drug resistance hampers effective malaria treatment. Critical SNPs in a particular, putative amino acid transporter were recently linked to chloroquine (CQ) resistance in malaria parasites. Here, we show that this conserved protein (PF3D7_0629500 in Plasmodium falciparum; AAT1 in P. chabaudi) is a structural homologue of the yeast amino acid transporter Tat2p, which is known to mediate quinine uptake and toxicity. Heterologous expression of PF3D7_0629500 in yeast produced CQ hypersensitivity, coincident with increased CQ uptake. PF3D7_0629500-expressing cultures were also sensitized to related antimalarials; amodiaquine, mefloquine and particularly quinine. Drug sensitivity was reversed by introducing a SNP linked to CQ resistance in the parasite. Like Tat2p, PF3D7_0629500-dependent quinine hypersensitivity was suppressible with tryptophan, consistent with a common transport mechanism. A four-fold increase in quinine uptake by PF3D7_0629500 expressing cells was abolished by the resistance SNP. The parasite protein localised primarily to the yeast plasma membrane. Its expression varied between cells and this heterogeneity was used to show that high-expressing cell subpopulations were the most drug sensitive. The results reveal that the PF3D7_0629500 protein can determine the level of sensitivity to several major quinine-related antimalarials through an amino acid-inhibitable drug transport function. The potential clinical relevance is discussed.


Subject(s)
Amino Acid Transport Systems/genetics , Antimalarials/pharmacology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae/drug effects , Amino Acid Transport Systems/metabolism , Amodiaquine/pharmacology , Animals , Biological Transport , Chloroquine/pharmacology , Conserved Sequence , Drug Resistance/genetics , Gene Expression , Humans , Mefloquine/pharmacology , Mutation , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/genetics , Plasmodium chabaudi/metabolism , Plasmodium falciparum/drug effects , Plasmodium falciparum/metabolism , Polymorphism, Single Nucleotide , Protozoan Proteins/metabolism , Quinine/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transgenes
13.
PLoS One ; 12(6): e0176533, 2017.
Article in English | MEDLINE | ID: mdl-28604779

ABSTRACT

Ubiquitination tags proteins for different functions within the cell. One of the most abundant and studied ubiquitin modification is the Lys48 polyubiquitin chain that modifies proteins for their destruction by proteasome. In Plasmodium is proposed that post-translational regulation is fundamental for parasite development during its complex life-cycle; thus, the objective of this work was to analyze the ubiquitination during Plasmodium chabaudi intraerythrocytic stages. Ubiquitinated proteins were detected during intraerythrocytic stages of Plasmodium chabaudi by immunofluorescent microscopy, bidimensional electrophoresis (2-DE) combined with immunoblotting and mass spectrometry. All the studied stages presented protein ubiquitination and Lys48 polyubiquitination with more abundance during the schizont stage. Three ubiquitinated proteins were identified for rings, five for trophozoites and twenty for schizonts. Only proteins detected with a specific anti- Lys48 polyubiquitin antibody were selected for Mass Spectrometry analysis and two of these identified proteins were selected in order to detect the specific amino acid residues where ubiquitin is placed. Ubiquitinated proteins during the ring and trophozoite stages were related with the invasion process and in schizont proteins were related with nucleic acid metabolism, glycolysis and protein biosynthesis. Most of the ubiquitin detection was during the schizont stage and the Lys48 polyubiquitination during this stage was related to proteins that are expected to be abundant during the trophozoite stage. The evidence that these Lys48 polyubiquitinated proteins are tagged for destruction by the proteasome complex suggests that this type of post-translational modification is important in the regulation of protein abundance during the life-cycle and may also contribute to the parasite cell-cycle progression.


Subject(s)
Erythrocytes/parasitology , Life Cycle Stages , Lysine/metabolism , Malaria/veterinary , Plasmodium chabaudi/growth & development , Plasmodium chabaudi/metabolism , Rodent Diseases/parasitology , Ubiquitination , Alternative Splicing , Animals , Gene Expression Regulation , Mass Spectrometry , Plasmodium chabaudi/genetics , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination/genetics
14.
Malar J ; 16(1): 185, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28468674

ABSTRACT

BACKGROUND: Parasite cytoadherence within the microvasculature of tissues and organs of infected individuals is implicated in the pathogenesis of several malaria syndromes. Multiple host receptors may mediate sequestration. The identity of the host receptor(s), or the parasite ligand(s) responsible for sequestration of Plasmodium species other than Plasmodium falciparum is largely unknown. The rodent malaria parasites may be useful to model interactions of parasite species, which lack the var genes with their respective hosts, as other multigene families are shared between the species. The role of the endothelial receptors ICAM-1 and CD36 in cytoadherence and in the development of pathology was investigated in a Plasmodium chabaudi infection in C57BL/6 mice lacking these receptors. The schizont membrane-associated cytoadherence (SMAC) protein of Plasmodium berghei has been shown to exhibit reduced CD36-associated cytoadherence in P. berghei ANKA-infected mice. METHODS: Parasite tissue sequestration and the development of acute stage pathology in P. chabaudi infections of mice lacking CD36 or ICAM-1, their respective wild type controls, and in infections with mutant P. chabaudi parasites lacking the smac gene were compared. Peripheral blood parasitaemia, red blood cell numbers and weight change were monitored throughout the courses of infection. Imaging of bioluminescent parasites in isolated tissues (spleen, lungs, liver, kidney and gut) was used to measure tissue parasite load. RESULTS: This study shows that neither the lack of CD36 nor the deletion of the smac gene from P. chabaudi significantly impacted on acute-stage pathology or parasite sequestration. By contrast, in the absence of ICAM-1, infected animals experience less anaemia and weight loss, reduced parasite accumulation in both spleen and liver and higher peripheral blood parasitaemia during acute stage malaria. The reduction in parasite tissue sequestration in infections of ICAM-1 null mice is maintained after mosquito transmission. CONCLUSIONS: These results indicate that ICAM-1-mediated cytoadherence is important in the P. chabaudi model of malaria and suggest that for rodent malarias, as for P. falciparum, there may be multiple host and parasite molecules involved in sequestration.


Subject(s)
CD36 Antigens/genetics , Intercellular Adhesion Molecule-1/genetics , Malaria/parasitology , Plasmodium chabaudi/physiology , Protozoan Proteins/genetics , Animals , CD36 Antigens/metabolism , Female , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Plasmodium chabaudi/genetics , Protozoan Proteins/metabolism
15.
Ecol Lett ; 19(9): 1041-50, 2016 09.
Article in English | MEDLINE | ID: mdl-27364562

ABSTRACT

A major challenge in disease ecology is to understand how co-infecting parasite species interact. We manipulate in vivo resources and immunity to explain interactions between two rodent malaria parasites, Plasmodium chabaudi and P. yoelii. These species have analogous resource-use strategies to the human parasites Plasmodium falciparum and P. vivax: P. chabaudi and P. falciparum infect red blood cells (RBC) of all ages (RBC generalist); P. yoelii and P. vivax preferentially infect young RBCs (RBC specialist). We find that: (1) recent infection with the RBC generalist facilitates the RBC specialist (P. yoelii density is enhanced ~10 fold). This occurs because the RBC generalist increases availability of the RBC specialist's preferred resource; (2) co-infections with the RBC generalist and RBC specialist are highly virulent; (3) and the presence of an RBC generalist in a host population can increase the prevalence of an RBC specialist. Thus, we show that resources shape how parasite species interact and have epidemiological consequences.


Subject(s)
Malaria/veterinary , Plasmodium chabaudi/physiology , Plasmodium yoelii/physiology , Rodent Diseases/epidemiology , Animals , Coinfection/epidemiology , Coinfection/parasitology , Coinfection/veterinary , Erythrocytes/parasitology , Genetic Fitness , Host-Parasite Interactions , Malaria/epidemiology , Malaria/parasitology , Male , Mice , Models, Biological , Plasmodium chabaudi/genetics , Plasmodium yoelii/genetics , Prevalence , Rodent Diseases/parasitology
16.
Sci Rep ; 6: 23449, 2016 Mar 21.
Article in English | MEDLINE | ID: mdl-26996203

ABSTRACT

Plasmodium multigene families play a central role in the pathogenesis of malaria. The Plasmodium interspersed repeat (pir) genes comprise the largest multigene family in many Plasmodium spp. However their function(s) remains unknown. Using the rodent model of malaria, Plasmodium chabaudi, we show that individual CIR proteins have differential localizations within infected red cell (iRBC), suggesting different functional roles in a blood-stage infection. Some CIRs appear to be located on the surface of iRBC and merozoites and are therefore well placed to interact with host molecules. In line with this hypothesis, we show for the first time that a subset of recombinant CIRs bind mouse RBCs suggesting a role for CIR in rosette formation and/or invasion. Together, our results unravel differences in subcellular localization and ability to bind mouse erythrocytes between the members of the cir family, which strongly suggest different functional roles in a blood-stage infection.


Subject(s)
Erythrocytes/parasitology , Malaria/parasitology , Plasmodium chabaudi/genetics , Plasmodium chabaudi/pathogenicity , Protozoan Proteins/physiology , Animals , Genes, Protozoan , Interspersed Repetitive Sequences , Malaria/genetics , Mice , Multigene Family , Protozoan Proteins/biosynthesis
17.
Am J Trop Med Hyg ; 92(6): 1214-21, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25940195

ABSTRACT

Resistant malaria parasites are frequently found in mixed infections with drug-sensitive parasites. Particularly early in the evolutionary process, the frequency of these resistant mutants can be extremely low and below the level of molecular detection. We tested whether the rarity of resistance in infections impacted the health outcomes of treatment failure and the potential for onward transmission of resistance. Mixed infections of different ratios of resistant and susceptible Plasmodium chabaudi parasites were inoculated in laboratory mice and dynamics tracked during the course of infection using highly sensitive genotype-specific quantitative polymerase chain reaction (qPCR). Frequencies of resistant parasites ranged from 10% to 0.003% at the onset of treatment. We found that the rarer the resistant parasites were, the lower the likelihood of their onward transmission, but the worse the treatment failure was in terms of parasite numbers and disease severity. Strikingly, drug resistant parasites had the biggest impact on health outcomes when they were too rare to be detected by any molecular methods currently available for field samples. Indeed, in the field, these treatment failures would not even have been attributed to resistance.


Subject(s)
Antimalarials/therapeutic use , Malaria/drug therapy , Plasmodium chabaudi/drug effects , Animals , Drug Resistance/genetics , Female , Genotype , Malaria/parasitology , Mice, Inbred C57BL , Plasmodium chabaudi/genetics , Polymerase Chain Reaction , Treatment Failure
18.
Trends Microbiol ; 23(3): 126-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25680587

ABSTRACT

The emergence of drug-resistant pathogens is often considered a canonical case of evolution by natural selection. Here we argue that the strength of selection can be a poor predictor of the rate of resistance emergence. It is possible for a resistant strain to be under negative selection and still emerge in an infection or spread in a population. Measuring the right parameters is a necessary first step toward the development of evidence-based resistance-management strategies. We argue that it is the absolute fitness of the resistant strains that matters most and that a primary determinant of the absolute fitness of a resistant strain is the ecological context in which it finds itself.


Subject(s)
Drug Resistance, Microbial/genetics , Drug Resistance/genetics , Evolution, Molecular , Genetic Fitness , Selection, Genetic , Mutation , Plasmodium chabaudi/drug effects , Plasmodium chabaudi/genetics
19.
Malar J ; 13: 115, 2014 Mar 26.
Article in English | MEDLINE | ID: mdl-24670151

ABSTRACT

BACKGROUND: Investment in the production of transmissible stages (gametocytes) and their sex ratio are malaria parasite traits that underpin mosquito infectivity and are therefore central to epidemiology. Malaria parasites adjust their levels of investment into gametocytes and sex ratio in response to changes in the in-host environment (including red blood cell resource availability, host immune responses, competition from con-specific genotypes in mixed infections, and drug treatment). This plasticity appears to be adaptive (strategic) because parasites prioritize investment (in sexual versus asexual stages and male versus female stages) in manners predicted to maximize fitness. However, the information, or 'cues' that parasites use to detect environmental changes and make appropriate decisions about investment into gametocytes and their sex ratio are unknown. METHODS: Single genotype Plasmodium chabaudi infections were exposed to 'cue' treatments consisting of intact or lysed uninfected red blood cells, lysed parasitized RBCs of the same clone or an unrelated clone, and an unmanipulated control. Infection dynamics (proportion of reticulocytes, red blood cell and asexual stage parasite densities) were monitored, and changes in gametocyte investment and sex ratio in response to cue treatments, applied either pre- or post-peak of infection were examined. RESULTS AND CONCLUSIONS: A significant reduction in gametocyte density was observed in response to the presence of lysed parasite material and a borderline significant increase in sex ratio (proportion of male gametocytes) upon exposure to lysed red blood cells (both uninfected and infected) was observed. Furthermore, the changes in gametocyte density and sex ratio in response to these cues depend on the age of infection. Demonstrating that variation in gametocyte investment and sex ratio observed during infections are a result of parasite strategies (rather than the footprint of host physiology), provides a foundation to investigate the fitness consequences of plasticity and explore whether drugs could be developed to trick parasites into making suboptimal decisions.


Subject(s)
Host-Parasite Interactions , Malaria/parasitology , Plasmodium chabaudi/physiology , Animals , Erythrocytes/parasitology , Male , Mice , Plasmodium chabaudi/genetics , Reproduction
20.
Malar J ; 12: 440, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24314024

ABSTRACT

BACKGROUND: A series of elegant experiments was recently published which demonstrated that transmission of malaria parasites through mosquitoes elicited an attenuated growth phenotype, whereby infections grew more slowly and reached peak parasitaemia at least five-fold lower than parasites which had not been mosquito transmitted. To assess the implications of these results it is essential to understand whether the attenuated infection phenotype is a general phenomenon across parasites genotypes and conditions. METHODS: Using previously published data, the impact of mosquito transmission on parasite growth rates and virulence of six Plasmodium chabaudi lines was analysed. RESULTS: The effect of mosquito transmission varied among strains, but did not lead to pronounced or consistent reductions in parasite growth rate. CONCLUSIONS: Mosquito-induced attenuated growth phenotype is sensitive to experimental conditions.


Subject(s)
Culicidae/parasitology , Malaria/parasitology , Plasmodium chabaudi/pathogenicity , Animals , Biomedical Research , Malaria/transmission , Mice , Phenotype , Plasmodium chabaudi/genetics , Plasmodium chabaudi/growth & development , Research Design , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...