Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 12(5): e12317, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32246897

ABSTRACT

A multi-person interview on the unrolling corona pandemic with Samuel Alizon, Akiko Iwasaki, Gerard Krause and Rino Rappuoli.


Subject(s)
Pandemics , Humans
2.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28947801

ABSTRACT

The relevance of genetic factors in conferring protection to severe malaria has been demonstrated, as in the case of sickle cell trait and G6PD deficiency 1 . However, it remains unknown whether environmental components, such as dietary or metabolic variations, can contribute to the outcome of infection 2 . Here, we show that administration of a high-fat diet to mice for a period as short as 4 days impairs Plasmodium liver infection by over 90%. Plasmodium sporozoites can successfully invade and initiate replication but die inside hepatocytes, thereby are unable to cause severe disease. Transcriptional analyses combined with genetic and chemical approaches reveal that this impairment of infection is mediated by oxidative stress. We show that reactive oxygen species, probably spawned from fatty acid ß-oxidation, directly impact Plasmodium survival inside hepatocytes, and parasite load can be rescued by exogenous administration of the antioxidant N-acetylcysteine or the ß-oxidation inhibitor etomoxir. Together, these data reveal that acute and transient dietary alterations markedly impact the establishment of a Plasmodium infection and disease outcome.


Subject(s)
Diet, High-Fat/methods , Host-Parasite Interactions/genetics , Malaria/diet therapy , Acetylcysteine/metabolism , Animals , Antioxidants/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Glucose Tolerance Test , Glucosephosphate Dehydrogenase Deficiency/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Liver/metabolism , Liver/parasitology , Liver Diseases/metabolism , Liver Diseases/parasitology , Macrophages/parasitology , Macrophages/pathology , Malaria/blood , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress , Parasite Load , Plasmodium berghei , Reactive Oxygen Species , Sickle Cell Trait/metabolism , Sporozoites/metabolism
3.
Malar J ; 15: 229, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27098483

ABSTRACT

BACKGROUND: Transmission of the malaria parasite Plasmodium falciparum from humans to the mosquito vector requires differentiation of a sub-population of asexual forms replicating within red blood cells into non-dividing male and female gametocytes. The nature of the molecular mechanism underlying this key differentiation event required for malaria transmission is not fully understood. METHODS: Whole genome sequencing was used to examine the genomic diversity of the gametocyte non-producing 3D7-derived lines F12 and A4. These lines were used in the recent detection of the PF3D7_1222600 locus (encoding PfAP2-G), which acts as a genetic master switch that triggers gametocyte development. RESULTS: The evolutionary changes from the 3D7 parental strain through its derivatives F12 (culture-passage derived cloned line) and A4 (transgenic cloned line) were identified. The genetic differences including the formation of chimeric var genes are presented. CONCLUSION: A genomics resource is provided for the further study of gametocytogenesis or other phenotypes using these parasite lines.


Subject(s)
Gametogenesis , Genome, Protozoan , Plasmodium falciparum/physiology , Polymorphism, Genetic , Plasmodium falciparum/genetics , Sequence Analysis, DNA
4.
Genome Biol ; 15(11): 493, 2014 Nov 03.
Article in English | MEDLINE | ID: mdl-25418785

ABSTRACT

BACKGROUND: Following fertilization, the early proteomes of metazoans are defined by the translation of stored but repressed transcripts; further embryonic development relies on de novo transcription of the zygotic genome. During sexual development of Plasmodium berghei, a rodent model for human malaria species including P. falciparum, the stability of repressed mRNAs requires the translational repressors DOZI and CITH. When these repressors are absent, Plasmodium zygote development and transmission to the mosquito vector is halted, as hundreds of transcripts become destabilized. However, which mRNAs are direct targets of these RNA binding proteins, and thus subject to translational repression, is unknown. RESULTS: We identify the maternal mRNA contribution to post-fertilization development of P. berghei using RNA immunoprecipitation and microarray analysis. We find that 731 mRNAs, approximately 50% of the transcriptome, are associated with DOZI and CITH, allowing zygote development to proceed in the absence of RNA polymerase II transcription. Using GFP-tagging, we validate the repression phenotype of selected genes and identify mRNAs relying on the 5' untranslated region for translational control. Gene deletion reveals a novel protein located in the ookinete crystalloid with an essential function for sporozoite development. CONCLUSIONS: Our study details for the first time the P. berghei maternal repressome. This mRNA population provides the developing ookinete with coding potential for key molecules required for life-cycle progression, and that are likely to be critical for the transmission of the malaria parasite from the rodent and the human host to the mosquito vector.


Subject(s)
Malaria, Falciparum/genetics , Plasmodium berghei/genetics , RNA-Binding Proteins/genetics , Transcriptome , Animals , Gene Expression Regulation, Developmental , Germ Cells/growth & development , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Microarray Analysis , Plasmodium berghei/growth & development , Plasmodium berghei/pathogenicity , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/biosynthesis , Zygote/growth & development
5.
EMBO Mol Med ; 6(1): 1, 2014 01.
Article in English | MEDLINE | ID: mdl-24408963
6.
Nat Med ; 20(1): 47-53, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24362933

ABSTRACT

Before they infect red blood cells and cause malaria, Plasmodium parasites undergo an obligate and clinically silent expansion phase in the liver that is supposedly undetected by the host. Here, we demonstrate the engagement of a type I interferon (IFN) response during Plasmodium replication in the liver. We identified Plasmodium RNA as a previously unrecognized pathogen-associated molecular pattern (PAMP) capable of activating a type I IFN response via the cytosolic pattern recognition receptor Mda5. This response, initiated by liver-resident cells through the adaptor molecule for cytosolic RNA sensors, Mavs, and the transcription factors Irf3 and Irf7, is propagated by hepatocytes in an interferon-α/ß receptor-dependent manner. This signaling pathway is critical for immune cell-mediated host resistance to liver-stage Plasmodium infection, which we find can be primed with other PAMPs, including hepatitis C virus RNA. Together, our results show that the liver has sensor mechanisms for Plasmodium that mediate a functional antiparasite response driven by type I IFN.


Subject(s)
Immunity, Innate/immunology , Interferon Type I/immunology , Liver/parasitology , Plasmodium/immunology , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , DEAD-box RNA Helicases/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Green Fluorescent Proteins , Immunohistochemistry , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon-Induced Helicase, IFIH1 , Liver/immunology , Luciferases , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Oligonucleotides/genetics , Plasmodium/genetics , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric
7.
PLoS One ; 7(2): e31623, 2012.
Article in English | MEDLINE | ID: mdl-22384044

ABSTRACT

The combination therapy of the Artemisinin-derivative Artemether (ART) with Lumefantrine (LM) (Coartem®) is an important malaria treatment regimen in many endemic countries. Resistance to Artemisinin has already been reported, and it is feared that LM resistance (LMR) could also evolve quickly. Therefore molecular markers which can be used to track Coartem® efficacy are urgently needed. Often, stable resistance arises from initial, unstable phenotypes that can be identified in vitro. Here we have used the Plasmodium falciparum multidrug resistant reference strain V1S to induce LMR in vitro by culturing the parasite under continuous drug pressure for 16 months. The initial IC(50) (inhibitory concentration that kills 50% of the parasite population) was 24 nM. The resulting resistant strain V1S(LM), obtained after culture for an estimated 166 cycles under LM pressure, grew steadily in 378 nM of LM, corresponding to 15 times the IC(50) of the parental strain. However, after two weeks of culturing V1S(LM) in drug-free medium, the IC(50) returned to that of the initial, parental strain V1S. This transient drug tolerance was associated with major changes in gene expression profiles: using the PFSANGER Affymetrix custom array, we identified 184 differentially expressed genes in V1S(LM). Among those are 18 known and putative transporters including the multidrug resistance gene 1 (pfmdr1), the multidrug resistance associated protein and the V-type H+ pumping pyrophosphatase 2 (pfvp2) as well as genes associated with fatty acid metabolism. In addition we detected a clear selective advantage provided by two genomic loci in parasites grown under LM drug pressure, suggesting that all, or some of those genes contribute to development of LM tolerance--they may prove useful as molecular markers to monitor P. falciparum LM susceptibility.


Subject(s)
Antimalarials/pharmacology , Ethanolamines/pharmacology , Fluorenes/pharmacology , Malaria/drug therapy , Plasmodium falciparum/genetics , Animals , Drug Design , Erythrocytes/drug effects , Erythrocytes/parasitology , Gene Expression Profiling , Humans , Inhibitory Concentration 50 , Likelihood Functions , Linear Models , Lumefantrine , Mutation , Oligonucleotide Array Sequence Analysis , Parasites , Phenotype , Polymerase Chain Reaction/methods , RNA, Messenger/metabolism
9.
PLoS Pathog ; 7(5): e1002046, 2011 May.
Article in English | MEDLINE | ID: mdl-21625527

ABSTRACT

Many eukaryotic developmental and cell fate decisions that are effected post-transcriptionally involve RNA binding proteins as regulators of translation of key mRNAs. In malaria parasites (Plasmodium spp.), the development of round, non-motile and replicating exo-erythrocytic liver stage forms from slender, motile and cell-cycle arrested sporozoites is believed to depend on environmental changes experienced during the transmission of the parasite from the mosquito vector to the vertebrate host. Here we identify a Plasmodium member of the RNA binding protein family PUF as a key regulator of this transformation. In the absence of Pumilio-2 (Puf2) sporozoites initiate EEF development inside mosquito salivary glands independently of the normal transmission-associated environmental cues. Puf2- sporozoites exhibit genome-wide transcriptional changes that result in loss of gliding motility, cell traversal ability and reduction in infectivity, and, moreover, trigger metamorphosis typical of early Plasmodium intra-hepatic development. These data demonstrate that Puf2 is a key player in regulating sporozoite developmental control, and imply that transformation of salivary gland-resident sporozoites into liver stage-like parasites is regulated by a post-transcriptional mechanism.


Subject(s)
Malaria/parasitology , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism , Sporozoites/growth & development , Animals , Anopheles/parasitology , Gene Expression Profiling , Liver/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microarray Analysis , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Plasmodium berghei/pathogenicity , Protein Processing, Post-Translational , Protozoan Proteins/genetics , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Salivary Glands/parasitology , Sporozoites/metabolism , Sporozoites/ultrastructure
10.
Nat Med ; 17(6): 732-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21572427

ABSTRACT

In regions of high rates of malaria transmission, mosquitoes repeatedly transmit liver-tropic Plasmodium sporozoites to individuals who already have blood-stage parasitemia. This manifests itself in semi-immune children (who have been exposed since birth to Plasmodium infection and as such show low levels of peripheral parasitemia but can still be infected) older than 5 years of age by concurrent carriage of different parasite genotypes at low asymptomatic parasitemias. Superinfection presents an increased risk of hyperparasitemia and death in less immune individuals but counterintuitively is not frequently observed in the young. Here we show in a mouse model that ongoing blood-stage infections, above a minimum threshold, impair the growth of subsequently inoculated sporozoites such that they become growth arrested in liver hepatocytes and fail to develop into blood-stage parasites. Inhibition of the liver-stage infection is mediated by the host iron regulatory hormone hepcidin, whose synthesis we found to be stimulated by blood-stage parasites in a density-dependent manner. We mathematically modeled this phenomenon and show how density-dependent protection against liver-stage malaria can shape the epidemiological patterns of age-related risk and the complexity of malaria infections seen in young children. The interaction between these two Plasmodium stages and host iron metabolism has relevance for the global efforts to reduce malaria transmission and for evaluation of iron supplementation programs in malaria-endemic regions.


Subject(s)
Host-Parasite Interactions/immunology , Malaria/immunology , Superinfection/immunology , Animals , Antimicrobial Cationic Peptides/physiology , Disease Progression , Hepcidins , Humans , Liver/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Plasmodium/immunology , Sporozoites/immunology
11.
PLoS Pathog ; 6(2): e1000767, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20169188

ABSTRACT

A universal feature of metazoan sexual development is the generation of oocyte P granules that withhold certain mRNA species from translation to provide coding potential for proteins during early post-fertilization development. Stabilisation of translationally quiescent mRNA pools in female Plasmodium gametocytes depends on the RNA helicase DOZI, but the molecular machinery involved in the silencing of transcripts in these protozoans is unknown. Using affinity purification coupled with mass-spectrometric analysis we identify a messenger ribonucleoprotein (mRNP) from Plasmodium berghei gametocytes defined by DOZI and the Sm-like factor CITH (homolog of worm CAR-I and fly Trailer Hitch). This mRNP includes 16 major factors, including proteins with homologies to components of metazoan P granules and archaeal proteins. Containing translationally silent transcripts, this mRNP integrates eIF4E and poly(A)-binding protein but excludes P body RNA degradation factors and translation-initiation promoting eIF4G. Gene deletion mutants of 2 core components of this mRNP (DOZI and CITH) are fertilization-competent, but zygotes fail to develop into ookinetes in a female gametocyte-mutant fashion. Through RNA-immunoprecipitation and global expression profiling of CITH-KO mutants we highlight CITH as a crucial repressor of maternally supplied mRNAs. Our data define Plasmodium P granules as an ancient mRNP whose protein core has remained evolutionarily conserved from single-cell organisms to germ cells of multi-cellular animals and stores translationally silent mRNAs that are critical for early post-fertilization development during the initial stages of mosquito infection. Therefore, translational repression may offer avenues as a target for the generation of transmission blocking strategies and contribute to limiting the spread of malaria.


Subject(s)
Gene Expression Regulation/physiology , Plasmodium berghei/physiology , Protozoan Proteins/physiology , RNA Interference/physiology , Animals , Blotting, Southern , Blotting, Western , Female , Flow Cytometry , Gene Expression , Gene Expression Profiling , Germ Cells , Immunoprecipitation , Phylogeny , RNA, Messenger/genetics , Ribonucleoproteins/physiology , Sexual Development , Zygote
12.
Bioinformatics ; 25(20): 2732-3, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19687029

ABSTRACT

UNLABELLED: Array-based comparative genomic hybridization (CGH) technology is used to discover and validate genomic structural variation, including copy number variants, insertions, deletions and other structural variants (SVs). The visualization and summarization of the array CGH data outputs, potentially across many samples, is an important process in the identification and analysis of SVs. We have developed a software tool for SV analysis using data from array CGH technologies, which is also amenable to short-read sequence data. AVAILABILITY AND IMPLEMENTATION: SnoopCGH is written in java and is available from http://snoopcgh.sourceforge.net/


Subject(s)
Comparative Genomic Hybridization/methods , Computational Biology/methods , Genome , Software , Comparative Genomic Hybridization/instrumentation , Computer Graphics , Gene Expression Profiling/methods , Genome, Bacterial , Oligonucleotide Array Sequence Analysis/methods , Plasmodium falciparum/genetics
13.
BMC Genomics ; 10: 353, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19653891

ABSTRACT

BACKGROUND: Gene copy number variation (CNV) is responsible for several important phenotypes of the malaria parasite Plasmodium falciparum, including drug resistance, loss of infected erythrocyte cytoadherence and alteration of receptor usage for erythrocyte invasion. Despite the known effects of CNV, little is known about its extent throughout the genome. RESULTS: We performed a whole-genome survey of CNV genes in P. falciparum using comparative genome hybridisation of a diverse set of 16 laboratory culture-adapted isolates to a custom designed high density Affymetrix GeneChip array. Overall, 186 genes showed hybridisation signals consistent with deletion or amplification in one or more isolate. There is a strong association of CNV with gene length, genomic location, and low orthology to genes in other Plasmodium species. Sub-telomeric regions of all chromosomes are strongly associated with CNV genes independent from members of previously described multigene families. However, approximately 40% of CNV genes were located in more central regions of the chromosomes. Among the previously undescribed CNV genes, several that are of potential phenotypic relevance are identified. CONCLUSION: CNV represents a major form of genetic variation within the P. falciparum genome; the distribution of gene features indicates the involvement of highly non-random mutational and selective processes. Additional studies should be directed at examining CNV in natural parasite populations to extend conclusions to clinical settings.


Subject(s)
Gene Dosage , Genome, Protozoan , Plasmodium falciparum/genetics , Animals , Comparative Genomic Hybridization , DNA, Protozoan/genetics , Gene Deletion , Oligonucleotide Array Sequence Analysis
14.
BMC Genomics ; 10: 270, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19534804

ABSTRACT

BACKGROUND: Plasmodium sporozoites migrate to the liver where they traverse several hepatocytes before invading the one inside which they will develop and multiply into thousands of merozoites. Although this constitutes an essential step of malaria infection, the requirements of Plasmodium parasites in liver cells and how they use the host cell for their own survival and development are poorly understood. RESULTS: To gain new insights into the molecular host-parasite interactions that take place during malaria liver infection, we have used high-throughput microarray technology to determine the transcriptional profile of P. berghei-infected hepatoma cells. The data analysis shows differential expression patterns for 1064 host genes starting at 6 h and up to 24 h post infection, with the largest proportion correlating specifically with the early stages of the infection process. A considerable proportion of those genes were also found to be modulated in liver cells collected from P. yoelii-infected mice 24 and 40 h after infection, strengthening the data obtained with the in vitro model and highlighting genes and pathways involved in the host response to rodent Plasmodium parasites. CONCLUSION: Our data reveal that host cell infection by Plasmodium sporozoites leads to a coordinated and sequential set of biological events, ranging from the initial stage of stress response up to the engagement of host metabolic processes and the maintenance of cell viability throughout infection.


Subject(s)
Gene Expression Profiling , Host-Parasite Interactions/genetics , Liver/metabolism , Malaria/genetics , Animals , Cell Line, Tumor , Hepatocytes/metabolism , Hepatocytes/parasitology , Liver/parasitology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Plasmodium berghei , Sporozoites , Time Factors
15.
PLoS Biol ; 7(4): e84, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19402747

ABSTRACT

Cytoadherance of Plasmodium falciparum-infected erythrocytes in the brain, organs and peripheral microvasculature is linked to morbidity and mortality associated with severe malaria. Parasite-derived P. falciparum Erythrocyte Membrane Protein 1 (PfEMP1) molecules displayed on the erythrocyte surface are responsible for cytoadherance and undergo antigenic variation in the course of an infection. Antigenic variation of PfEMP1 is achieved by in situ switching and mutually exclusive transcription of the var gene family, a process that is controlled by epigenetic mechanisms. Here we report characterisation of the P. falciparum silent information regulator's A and B (PfSir2A and PfSir2B) and their involvement in mutual exclusion and silencing of the var gene repertoire. Analysis of P. falciparum parasites lacking either PfSir2A or PfSir2B shows that these NAD(+)-dependent histone deacetylases are required for silencing of different var gene subsets classified by their conserved promoter type. We also demonstrate that in the absence of either of these molecules mutually exclusive expression of var genes breaks down. We show that var gene silencing originates within the promoter and PfSir2 paralogues are involved in cis spreading of silenced chromatin into adjacent regions. Furthermore, parasites lacking PfSir2A but not PfSir2B have considerably longer telomeric repeats, demonstrating a role for this molecule in telomeric end protection. This work highlights the pivotal but distinct role for both PfSir2 paralogues in epigenetic silencing of P. falciparum virulence genes and the control of pathogenicity of malaria infection.


Subject(s)
Antigenic Variation , Gene Silencing , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Sirtuins/physiology , Virulence/genetics , Animals , Antigens, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Cell Adhesion/genetics , Erythrocytes/metabolism , Erythrocytes/parasitology , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/physiopathology , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Plasmodium falciparum/metabolism , Promoter Regions, Genetic/physiology , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Protozoan Proteins/physiology , Telomere/metabolism , Transcriptional Activation , Virulence/immunology
16.
Proc Natl Acad Sci U S A ; 106(18): 7559-64, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19376968

ABSTRACT

We have cultured Plasmodium falciparum directly from the blood of infected individuals to examine patterns of mature-stage gene expression in patient isolates. Analysis of the transcriptome of P. falciparum is complicated by the highly periodic nature of gene expression because small variations in the stage of parasite development between samples can lead to an apparent difference in gene expression values. To address this issue, we have developed statistical likelihood-based methods to estimate cell cycle progression and commitment to asexual or sexual development lineages in our samples based on microscopy and gene expression patterns. In cases subsequently matched for temporal development, we find that transcriptional patterns in ex vivo culture display little variation across patients with diverse clinical profiles and closely resemble transcriptional profiles that occur in vitro. These statistical methods, available to the research community, assist in the design and interpretation of P. falciparum expression profiling experiments where it is difficult to separate true differential expression from cell-cycle dependent expression. We reanalyze an existing dataset of in vivo patient expression profiles and conclude that previously observed discrete variation is consistent with the commitment of a varying proportion of the parasite population to the sexual development lineage.


Subject(s)
Cell Cycle , Gene Expression Profiling , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Animals , Cell Cycle/genetics , Cells, Cultured , Humans
17.
J Mol Biol ; 389(3): 453-69, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19389407

ABSTRACT

The Plasmodium falciparum var multigene family encodes P. falciparum erythrocyte membrane protein 1, which is responsible for the pathogenic traits of antigenic variation and adhesion of infected erythrocytes to host receptors during malaria infection. Clonal antigenic variation of P. falciparum erythrocyte membrane protein 1 is controlled by the switching between exclusively transcribed var genes. The tremendous diversity of the var gene repertoire both within and between parasite strains is critical for the parasite's strategy of immune evasion. We show that ectopic recombination between var genes occurs during mitosis, providing P. falciparum with opportunities to diversify its var repertoire, even during the course of a single infection. We show that the regulation of the recombined var gene has been disrupted, resulting in its persistent activation although the regulation of most other var genes is unaffected. The var promoter and intron of the recombined var gene are not responsible for its atypically persistent activity, and we conclude that altered subtelomeric cis sequence is the most likely cause of the persistent activity of the recombined var gene.


Subject(s)
Antigenic Variation , Malaria, Falciparum/parasitology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/genetics , Animals , Introns , Malaria, Falciparum/immunology , Mitosis , Multigene Family , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Promoter Regions, Genetic , Recombination, Genetic , Transcription, Genetic
18.
PLoS One ; 4(1): e4285, 2009.
Article in English | MEDLINE | ID: mdl-19173007

ABSTRACT

The pir multigene family, found in the genomes of Plasmodium vivax, P. knowlesi and the rodent malaria species, encode variant antigens that could be targets of the immune response. Individual parasites of the rodent malaria Plasmodium yoelii, selected by micromanipulation, transcribe only 1 to 3 different pir (yir) suggesting tight transcriptional control at the level of individual cells. Using microarray and quantitative RT-PCR, we show that despite this very restricted transcription in a single cell, many yir genes are transcribed throughout the intra-erythrocytic asexual cycle. The timing and level of transcription differs between genes, with some being more highly transcribed in ring and trophozoite stages, whereas others are more highly transcribed in schizonts. Infection of immunodeficient mice with single infected erythrocytes results in populations of parasites each with transcriptional profiles different from that of the parent parasite population and from each other. This drift away from the original 'set' of transcribed genes does not appear to follow a preset pattern and "epigenetic memory" of the yir transcribed in the parent parasite can be rapidly lost. Thus, regulation of pir gene transcription may be different from that of the well-characterised multigene family, var, of Plasmodium falciparum.


Subject(s)
Epigenesis, Genetic , Gene Expression Profiling , Genes, Protozoan , Memory , Multigene Family , Plasmodium yoelii/genetics , Transcription, Genetic , Animals , Chromosomes/genetics , Clone Cells , Erythrocytes/parasitology , Female , Gene Expression Regulation , Genes, Switch , Malaria/genetics , Malaria/parasitology , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Schizonts/metabolism , Trophozoites/metabolism
19.
PLoS Pathog ; 4(11): e1000201, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18989463

ABSTRACT

Plasmodium sporozoites, the causative agent of malaria, are injected into their vertebrate host through the bite of an infected Anopheles mosquito, homing to the liver where they invade hepatocytes to proliferate and develop into merozoites that, upon reaching the bloodstream, give rise to the clinical phase of infection. To investigate how host cell signal transduction pathways affect hepatocyte infection, we used RNAi to systematically test the entire kinome and associated genes in human Huh7 hepatoma cells for their potential roles during infection by P. berghei sporozoites. The three-phase screen covered 727 genes, which were tested with a total of 2,307 individual siRNAs using an automated microscopy assay to quantify infection rates and qRT-PCR to assess silencing levels. Five protein kinases thereby emerged as top hits, all of which caused significant reductions in infection when silenced by RNAi. Follow-up validation experiments on one of these hits, PKCsigma (PKCzeta), confirmed the physiological relevance of our findings by reproducing the inhibitory effect on P. berghei infection in adult mice treated systemically with liposome-formulated PKCsigma-targeting siRNAs. Additional cell-based analyses using a pseudo-substrate inhibitor of PKCsigma added further RNAi-independent support, indicating a role for host PKCsigma on the invasion of hepatocytes by sporozoites. This study represents the first comprehensive, functional genomics-driven identification of novel host factors involved in Plasmodium sporozoite infection.


Subject(s)
Genome, Human , Malaria , Phosphotransferases/genetics , Plasmodium berghei/pathogenicity , Protein Kinase C , RNA, Small Interfering/pharmacology , Animals , Cell Line , Gene Silencing , Hepatocytes/enzymology , Hepatocytes/parasitology , Humans , Mice , Mice, Inbred C57BL , Signal Transduction , Sporozoites
20.
Microbes Infect ; 10(5): 462-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18468471

ABSTRACT

Rab genes encode a subgroup of small GTP-binding proteins within the ras super-family that regulate targeting and fusion of transport vesicles within the secretory and endocytic pathways. These genes are of particular interest in the protozoan phylum Apicomplexa, since a family of Rab GTPases has been described for Plasmodium and most putative secretory pathway proteins in Apicomplexa have conventional predicted signal peptides. Moreover, peptide motifs have now been identified within a large number of secreted Plasmodium proteins that direct their targeting to the red blood cell cytosol, the apicoplast, the food vacuole and Maurer's clefs; in contrast, motifs that direct proteins to secretory organelles (rhoptries, micronemes and microspheres) have yet to be defined. The nature of the vesicle in which these proteins are transported to their destinations remains unknown and morphological structures equivalent to the endoplasmic reticulum and trans-Golgi stacks typical of other eukaryotes cannot be visualised in Apicomplexa. Since Rab GTPases regulate vesicular traffic in all eukaryotes, and this traffic in intracellular parasites could regulate import of nutrient and drugs and export of antigens, host cell modulatory proteins and lactate we compare and contrast here the Rab families of Apicomplexa.


Subject(s)
Genome, Protozoan , Plasmodium falciparum/genetics , Theileria/genetics , rab GTP-Binding Proteins/genetics , Animals , DNA, Protozoan/analysis , Databases, Nucleic Acid , Gene Expression Profiling , Gene Order , Genes, Protozoan , Genomics , Malaria/parasitology , Multigene Family , Phylogeny , Sequence Homology, Nucleic Acid , T-Lymphocytes/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...