Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Elife ; 132024 Jun 26.
Article in English | MEDLINE | ID: mdl-38921824

ABSTRACT

While often undetected and untreated, persistent seasonal asymptomatic malaria infections remain a global public health problem. Despite the presence of parasites in the peripheral blood, no symptoms develop. Disease severity is correlated with the levels of infected red blood cells (iRBCs) adhering within blood vessels. Changes in iRBC adhesion capacity have been linked to seasonal asymptomatic malaria infections, however how this is occurring is still unknown. Here, we present evidence that RNA polymerase III (RNA Pol III) transcription in Plasmodium falciparum is downregulated in field isolates obtained from asymptomatic individuals during the dry season. Through experiments with in vitro cultured parasites, we have uncovered an RNA Pol III-dependent mechanism that controls pathogen proliferation and expression of a major virulence factor in response to external stimuli. Our findings establish a connection between P. falciparum cytoadhesion and a non-coding RNA family transcribed by Pol III. Additionally, we have identified P. falciparum Maf1 as a pivotal regulator of Pol III transcription, both for maintaining cellular homeostasis and for responding adaptively to external signals. These results introduce a novel perspective that contributes to our understanding of P. falciparum virulence. Furthermore, they establish a connection between this regulatory process and the occurrence of seasonal asymptomatic malaria infections.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , RNA Polymerase III , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Plasmodium falciparum/enzymology , Virulence , RNA Polymerase III/metabolism , RNA Polymerase III/genetics , Humans , Malaria, Falciparum/parasitology , Erythrocytes/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Virulence Factors/metabolism , Virulence Factors/genetics , Cell Adhesion , Gene Expression Regulation
2.
EMBO Rep ; 24(10): e57090, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37592911

ABSTRACT

The complex life cycle of the human malaria parasite, Plasmodium falciparum, is driven by specific transcriptional programs, but it is unclear how most genes are activated or silenced at specific times. There is an association between transcription and spatial organization; however, the molecular mechanisms behind genome organization are unclear. While P. falciparum lacks key genome-organizing proteins found in metazoans, it has all core components of the cohesin complex. To investigate the role of cohesin in P. falciparum, we functionally characterize the cohesin subunit Structural Maintenance of Chromosomes protein 3 (SMC3). SMC3 knockdown during early stages of the intraerythrocytic developmental cycle (IDC) upregulates a subset of genes involved in erythrocyte egress and invasion, which are normally expressed at later stages. ChIP-seq analyses reveal that during the IDC, SMC3 enrichment at the promoter regions of these genes inversely correlates with gene expression and chromatin accessibility. These data suggest that SMC3 binding contributes to the repression of specific genes until their appropriate time of expression, revealing a new mode of stage-specific gene repression in P. falciparum.

3.
ACS Infect Dis ; 9(6): 1257-1266, 2023 06 09.
Article in English | MEDLINE | ID: mdl-37216290

ABSTRACT

Malaria drug resistance is hampering the fight against the deadliest parasitic disease affecting over 200 million people worldwide. We recently developed quinoline-quinazoline-based inhibitors (as compound 70) as promising new antimalarials. Here, we aimed to investigate their mode of action by using thermal proteome profiling (TPP). The eukaryotic translation initiation factor 3 (EIF3i) subunit I was identified as the main target protein stabilized by compound 70 in Plasmodium falciparum. This protein has never been characterized in malaria parasites. P. falciparum parasite lines were generated expressing either a HA tag or an inducible knockdown of the PfEIF3i gene to further characterize the target protein. PfEIF3i was stabilized in the presence of compound 70 in a cellular thermal shift Western blot assay, pointing that PfEIF3i indeed interacts with quinoline-quinazoline-based inhibitors. In addition, PfEIF3i-inducible knockdown blocks intra-erythrocytic development in the trophozoite stage, indicating that it has a vital function. We show that PfEIF3i is mostly expressed in late intra-erythrocytic stages and localizes in the cytoplasm. Previous mass spectrometry reports show that PfEIF3i is expressed in all parasite life cycle stages. Further studies will explore the potential of PfEIF3i as a target for the design of new antimalarial drugs active all along the life cycle of the parasite.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Quinolines , Humans , Animals , Plasmodium falciparum/metabolism , Prokaryotic Initiation Factor-3/metabolism , Quinazolines/pharmacology , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/chemistry , Quinolines/pharmacology , Life Cycle Stages
4.
Nat Commun ; 13(1): 4123, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840625

ABSTRACT

Plasmodium vivax is the most widespread human malaria parasite. Due to the presence of extravascular reservoirs and relapsing infections from dormant liver stages, P. vivax is particularly difficult to control and eliminate. Experimental research is hampered by the inability to maintain P. vivax cultures in vitro, due to its tropism for immature red blood cells (RBCs). Here, we describe a new humanized mice model that can support efficient human erythropoiesis and maintain long-lasting multiplication of inoculated cryopreserved P. vivax parasites and their sexual differentiation, including in bone marrow. Mature gametocytes were transmitted to Anopheles mosquitoes, which led to the formation of salivary gland sporozoites. Importantly, blood-stage P. vivax parasites were maintained after the secondary transfer of fresh or frozen infected bone marrow cells to naïve chimeras. This model provides a unique tool for investigating, in vivo, the biology of intraerythrocytic P. vivax.


Subject(s)
Anopheles , Malaria, Vivax , Animals , Anopheles/parasitology , Humans , Malaria, Vivax/parasitology , Mice , Neoplasm Recurrence, Local , Plasmodium vivax , Sporozoites
5.
Br J Haematol ; 196(5): 1159-1169, 2022 03.
Article in English | MEDLINE | ID: mdl-34962643

ABSTRACT

COVID-19 has compelled scientists to better describe its pathophysiology to find new therapeutic approaches. While risk factors, such as older age, obesity, and diabetes mellitus, suggest a central role of endothelial cells (ECs), autopsies have revealed clots in the pulmonary microvasculature that are rich in neutrophils and DNA traps produced by these cells, called neutrophil extracellular traps (NETs.) Submicron extracellular vesicles, called microparticles (MPs), are described in several diseases as being involved in pro-inflammatory pathways. Therefore, in this study, we analyzed three patient groups: one for which intubation was not necessary, an intubated group, and one group after extubation. In the most severe group, the intubated group, platelet-derived MPs and endothelial cell (EC)-derived MPs exhibited increased concentration and size, when compared to uninfected controls. MPs of intubated COVID-19 patients triggered EC death and overexpression of two adhesion molecules: P-selectin and vascular cell adhesion molecule-1 (VCAM-1). Strikingly, neutrophil adhesion and NET production were increased following incubation with these ECs. Importantly, we also found that preincubation of these COVID-19 MPs with the phosphatidylserine capping endogenous protein, annexin A5, abolished cytotoxicity, P-selectin and VCAM-1 induction, all like increases in neutrophil adhesion and NET release. Taken together, our results reveal that MPs play a key role in COVID-19 pathophysiology and point to a potential therapeutic: annexin A5.


Subject(s)
COVID-19/immunology , Cell-Derived Microparticles/immunology , Endothelial Cells/immunology , Neutrophils/immunology , SARS-CoV-2/immunology , COVID-19/pathology , COVID-19/therapy , Cell Adhesion , Cell Death , Cell-Derived Microparticles/pathology , Cells, Cultured , Endothelial Cells/pathology , Extracellular Traps/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Intubation , Neutrophils/pathology , Phosphatidylserines/immunology
6.
J Med Chem ; 64(14): 10403-10417, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34185525

ABSTRACT

Epigenetic post-translational modifications are essential for human malaria parasite survival and progression through its life cycle. Here, we present new functionalized suberoylanilide hydroxamic acid (SAHA) derivatives that chemically combine the pan-histone deacetylase inhibitor SAHA with the DNA methyltransferase inhibitor procainamide. A three- or four-step chemical synthesis was designed starting from cheap raw materials. Compared to the single drugs, the combined molecules showed a superior activity in Plasmodium and a potent inhibition against human HDAC6, exerting no cytotoxicity in human cell lines. These new compounds are fully active in multidrug-resistant Plasmodium falciparum Cambodian isolates. They target transmission of the parasite by inducing irreversible morphological changes in gametocytes and inhibiting exflagellation. The compounds are slow-acting and have an additive antimalarial effect in combination with fast-acting epidrugs and dihydroartemisinin. The lead compound decreases parasitemia in mice in a severe malaria model. Taken together, this novel fused molecule offers an affordable alternative to current failing antimalarial therapy.


Subject(s)
Antimalarials/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Procainamide/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Hydroxamic Acids/chemistry , Molecular Structure , Procainamide/chemistry , Structure-Activity Relationship
7.
Mol Syst Biol ; 16(8): e9569, 2020 08.
Article in English | MEDLINE | ID: mdl-32816370

ABSTRACT

Mutually exclusive expression of the var multigene family is key to immune evasion and pathogenesis in Plasmodium falciparum, but few factors have been shown to play a direct role. We adapted a CRISPR-based proteomics approach to identify novel factors associated with var genes in their natural chromatin context. Catalytically inactive Cas9 ("dCas9") was targeted to var gene regulatory elements, immunoprecipitated, and analyzed with mass spectrometry. Known and novel factors were enriched including structural proteins, DNA helicases, and chromatin remodelers. Functional characterization of PfISWI, an evolutionarily divergent putative chromatin remodeler enriched at the var gene promoter, revealed a role in transcriptional activation. Proteomics of PfISWI identified several proteins enriched at the var gene promoter such as acetyl-CoA synthetase, a putative MORC protein, and an ApiAP2 transcription factor. These findings validate the CRISPR/dCas9 proteomics method and define a new var gene-associated chromatin complex. This study establishes a tool for targeted chromatin purification of unaltered genomic loci and identifies novel chromatin-associated factors potentially involved in transcriptional control and/or chromatin organization of virulence genes in the human malaria parasite.


Subject(s)
Adenosine Triphosphatases/metabolism , Plasmodium falciparum/pathogenicity , Proteomics/methods , Transcription Factors/metabolism , Virulence Factors/genetics , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , CRISPR-Cas Systems , Chromatin Immunoprecipitation Sequencing , Humans , Introns , Mass Spectrometry , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Promoter Regions, Genetic , Protein Interaction Maps , Virulence Factors/metabolism
8.
Blood ; 136(2): 247-256, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32285120

ABSTRACT

Microparticles (MPs) are submicron extracellular vesicles exposing phosphatidylserine (PS), detected at high concentration in the circulation of sickle cell anemia (SS) patients. Several groups studied the biological effects of MPs generated ex vivo. Here, we analyzed for the first time the impact of circulating MPs on endothelial cells (ECs) from 60 sickle cell disease (SCD) patients. MPs were collected from SCD patients and compared with MPs isolated from healthy individuals (AA). Other plasma MPs were purified from SS patients before and 2 years after the onset of hydroxyurea (HU) treatment or during a vaso-occlusive crisis and at steady-state. Compared with AA MPs, SS MPs increased EC ICAM-1 messenger RNA and protein levels, as well as neutrophil adhesion. We showed that ICAM-1 overexpression was primarily caused by MPs derived from erythrocytes, rather than from platelets, and that it was abolished by MP PS capping using annexin V. MPs from SS patients treated with HU were less efficient to induce a proinflammatory phenotype in ECs compared with MPs collected before therapy. In contrast, MPs released during crisis increased ICAM-1 and neutrophil adhesion levels, in a PS-dependent manner, compared with MPs collected at steady-state. Furthermore, neutrophil adhesion was abolished by a blocking anti-ICAM-1 antibody. Our study provides evidence that MPs play a key role in SCD pathophysiology by triggering a proinflammatory phenotype of ECs. We also uncover a new mode of action for HU and identify potential therapeutics: annexin V and anti-ICAM-1 antibodies.


Subject(s)
Anemia, Sickle Cell , Cell-Derived Microparticles/metabolism , Endothelium, Vascular/metabolism , Hydroxyurea/administration & dosage , Intercellular Adhesion Molecule-1/blood , RNA, Messenger/blood , Adolescent , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/pathology , Anemia, Sickle Cell/physiopathology , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Female , Humans , Inflammation/blood , Inflammation/drug therapy , Inflammation/pathology , Inflammation/physiopathology , Male
9.
PLoS Biol ; 17(6): e3000308, 2019 06.
Article in English | MEDLINE | ID: mdl-31181082

ABSTRACT

Plasmodium falciparum is the main cause of disease and death from malaria. P. falciparum virulence resides in the ability of infected erythrocytes (IEs) to sequester in various tissues through the interaction between members of the polymorphic P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesin family to various host receptors. Here, we investigated the effect of phosphorylation of variant surface antigen 2-CSA (VAR2CSA), a member of the PfEMP1 family associated to placental sequestration, on its capacity to adhere to chondroitin sulfate A (CSA) present on the placental syncytium. We showed that phosphatase treatment of IEs impairs cytoadhesion to CSA. MS analysis of recombinant VAR2CSA phosphosites prior to and after phosphatase treatment, as well as of native VAR2CSA expressed on IEs, identified critical phosphoresidues associated with CSA binding. Site-directed mutagenesis on recombinant VAR2CSA of 3 phosphoresidues localised within the CSA-binding region confirmed in vitro their functional importance. Furthermore, using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein-9 nuclease (CRISPR/Cas9), we generated a parasite line in which the phosphoresidue T934 is changed to alanine and showed that this mutation strongly impairs IEs cytoadhesion to CSA. Taken together, these results demonstrate that phosphorylation of the extracellular region of VAR2CSA plays a major role in IEs cytoadhesion to CSA and provide new molecular insights for strategies aiming to reduce the morbidity and mortality of PM.


Subject(s)
Antigens, Protozoan/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Animals , Antigenic Variation , Antigens, Protozoan/metabolism , Cell Culture Techniques , Cell Line , Erythrocytes/parasitology , Female , Humans , Malaria , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Parasites , Phosphorylation , Placenta , Plasmodium falciparum/genetics , Pregnancy , Protein Binding
10.
Genome Announc ; 6(5)2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29437092

ABSTRACT

Plasmodium falciparum is the species of human malaria parasite that causes the most severe form of the disease. Here, we used single-molecule real-time (SMRT) sequencing technology from Pacific Biosciences (PacBio) to sequence, assemble de novo, and annotate the genome of a P. falciparum NF54 clone.

11.
Sci Rep ; 7(1): 14042, 2017 10 25.
Article in English | MEDLINE | ID: mdl-29070841

ABSTRACT

Variegated surface antigen expression is key to chronic infection and pathogenesis of the human malaria parasite Plasmodium falciparum. This protozoan parasite expresses distinct surface molecules that are encoded by clonally variant gene families such as var, rif and stevor. The molecular mechanisms governing activation of individual members remain ill-defined. To investigate the molecular events of the initial transcriptional activation process we focused on a member of the apicomplexan ApiAP2 transcription factor family predicted to bind to the 5' upstream regions of the var gene family, AP2-exp (PF3D7_1466400). Viable AP2-exp mutant parasites rely on expressing no less than a short truncated protein including the N-terminal AP2 DNA-binding domain. RNA-seq analysis in mutant parasites revealed transcriptional changes in a subset of exported proteins encoded by clonally variant gene families. Upregulation of RIFINs and STEVORs was validated at the protein levels. In addition, morphological alterations were observed on the surface of the host cells infected by the mutants. This work points to a complex regulatory network of clonally variant gene families in which transcription of a subset of members is regulated by the same transcription factor. In addition, we highlight the importance of the non-DNA binding AP2 domain in functional gene regulation.


Subject(s)
Gene Expression Regulation , Plasmodium falciparum/genetics , Protozoan Proteins/physiology , Genes, Protozoan , Genetic Variation , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
12.
Cell Microbiol ; 17(8): 1205-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25703704

ABSTRACT

Plasmodium falciparum virulence is linked to its ability to sequester in post-capillary venules in the human host. Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) is the main variant surface antigen implicated in this process. Complete loss of parasite adhesion is linked to a large subtelomeric deletion on chromosome 9 in a number of laboratory strains such as D10 and T9-96. Similar to the cytoadherent reference line FCR3, D10 strain expresses PfEMP1 on the surface of parasitized erythrocytes, however without any detectable cytoadhesion. To investigate which of the deleted subtelomeric genes may be implicated in parasite adhesion, we selected 12 genes for D10 complementation studies that are predicted to code for proteins exported to the red blood cell. We identified a novel single copy gene (PF3D7_0936500) restricted to P. falciparum that restores adhesion to CD36, termed here virulence-associated protein 1 (Pfvap1). Protein knockdown and gene knockout experiments confirmed a role of PfVAP1 in the adhesion process in FCR3 parasites. PfVAP1 is co-exported with PfEMP1 into the host cell via vesicle-like structures called Maurer's clefts. This study identifies a novel highly conserved parasite molecule that contributes to parasite virulence possibly by assisting PfEMP1 to establish functional adhesion at the host cell surface.


Subject(s)
Cell Adhesion , Erythrocytes/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Erythrocytes/parasitology , Gene Knockdown Techniques , Gene Knockout Techniques , Genetic Complementation Test , Humans , Protein Binding , Sequence Deletion
13.
Nature ; 513(7518): 431-5, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25043062

ABSTRACT

Antigenic variation of the Plasmodium falciparum multicopy var gene family enables parasite evasion of immune destruction by host antibodies. Expression of a particular var subgroup, termed upsA, is linked to the obstruction of blood vessels in the brain and to the pathogenesis of human cerebral malaria. The mechanism determining upsA activation remains unknown. Here we show that an entirely new type of gene silencing mechanism involving an exonuclease-mediated degradation of nascent RNA controls the silencing of genes linked to severe malaria. We identify a novel chromatin-associated exoribonuclease, termed PfRNase II, that controls the silencing of upsA var genes by marking their transcription start site and intron-promoter regions leading to short-lived cryptic RNA. Parasites carrying a deficient PfRNase II gene produce full-length upsA var transcripts and intron-derived antisense long non-coding RNA. The presence of stable upsA var transcripts overcomes monoallelic expression, resulting in the simultaneous expression of both upsA and upsC type PfEMP1 proteins on the surface of individual infected red blood cells. In addition, we observe an inverse relationship between transcript levels of PfRNase II and upsA-type var genes in parasites from severe malaria patients, implying a crucial role of PfRNase II in severe malaria. Our results uncover a previously unknown type of post-transcriptional gene silencing mechanism in malaria parasites with repercussions for other organisms. Additionally, the identification of RNase II as a parasite protein controlling the expression of virulence genes involved in pathogenesis in patients with severe malaria may provide new strategies for reducing malaria mortality.


Subject(s)
Exoribonucleases/metabolism , Gene Silencing , Genes, Protozoan/genetics , Malaria, Cerebral/parasitology , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , RNA, Protozoan/metabolism , Alleles , Antigenic Variation/genetics , Chromatin/enzymology , Down-Regulation/genetics , Erythrocytes/parasitology , Exoribonucleases/deficiency , Exoribonucleases/genetics , Humans , Introns/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/pathogenicity , Promoter Regions, Genetic/genetics , Protozoan Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Protozoan/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Transcription Initiation Site , Virulence/genetics , Virulence Factors/genetics
14.
Eukaryot Cell ; 12(5): 697-702, 2013 May.
Article in English | MEDLINE | ID: mdl-23475702

ABSTRACT

The human malaria parasite Plasmodium falciparum modifies the erythrocyte it infects by exporting variant proteins to the host cell surface. The var gene family that codes for a large, variant adhesive surface protein called P. falciparum erythrocyte membrane protein 1 (PfEMP1) plays a particular role in this process, which is linked to pathogenesis and immune evasion. A single member of this gene family is highly transcribed while the other 59 members remain silenced. Importantly, var gene transcription occurs at a spatially restricted, but yet undefined, perinuclear site that is distinct from repressed var gene clusters. To advance our understanding of monoallelic expression, we investigated whether nuclear pores associate with the var gene expression site. To this end, we studied the nuclear pore organization during the asexual blood stage using a specific antibody directed against a subunit of the nuclear pore, P. falciparum Nup116 (PfNup116). Ring and schizont stage parasites showed highly polarized nuclear pore foci, whereas in trophozoite stage nuclear pores redistributed over the entire nuclear surface. Colocalization studies of var transcripts and anti-PfNup116 antibodies showed clear dissociation between nuclear pores and the var gene expression site in ring stage. Similar results were obtained for another differentially transcribed perinuclear gene family, the ribosomal DNA units. Furthermore, we show that in the poised state, the var gene locus is not physically linked to nuclear pores. Our results indicate that P. falciparum does form compartments of high transcriptional activity at the nuclear periphery which are, unlike the case in yeast, devoid of nuclear pores.


Subject(s)
DNA, Ribosomal/genetics , Nuclear Pore/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Cells, Cultured , DNA, Ribosomal/metabolism , Erythrocytes/parasitology , Gene Expression , Gene Expression Regulation , Genes, Protozoan , Humans , Nuclear Pore Complex Proteins/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/ultrastructure , Protein Transport , Protozoan Proteins/metabolism , Trophozoites/diagnostic imaging , Trophozoites/metabolism , Ultrasonography
15.
Nat Commun ; 4: 1530, 2013.
Article in English | MEDLINE | ID: mdl-23443558

ABSTRACT

The Plasmodium falciparum histone deacetylase Sir2a localizes at telomeric regions where it contributes to epigenetic silencing of clonally variant virulence genes. Apart from telomeres, PfSir2a also accumulates in the nucleolus, which harbours the developmentally regulated ribosomal RNA genes. Here we investigate the nucleolar function of PfSir2a and demonstrate that PfSir2a fine-tunes ribosomal RNA gene transcription. Using a parasite line in which PfSir2a has been disrupted, we observe that histones near the transcription start sites of all ribosomal RNA genes are hyperacetylated and that transcription of ribosomal RNA genes is upregulated. Complementation of the PfSir2a-disrupted parasites restores the ribosomal RNA levels, whereas PfSir2a overexpression in wild-type parasites decreases ribosomal RNA synthesis. Furthermore, we observe that PfSir2a modulation of ribosomal RNA synthesis is linked to an altered number of daughter merozoites and the parasite multiplication rate. These findings provide new insights into an epigenetic mechanism that controls malaria parasite proliferation and virulence.


Subject(s)
DNA, Ribosomal/genetics , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Transcription, Genetic , Animals , Animals, Genetically Modified , Erythrocytes/parasitology , Genetic Complementation Test , Humans , Merozoites/cytology , Merozoites/growth & development , Merozoites/ultrastructure , Mutation/genetics , Parasites/cytology , Parasites/growth & development , Parasites/ultrastructure , Plasmids/metabolism , Plasmodium falciparum/cytology , Plasmodium falciparum/ultrastructure
16.
Nucleic Acids Res ; 40(17): 8381-91, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22740656

ABSTRACT

Terminal deoxynucleotidyl transferase (TdT) is a DNA polymerase that increases the repertoire of antigen receptors by adding non-templated nucleotides (N-addition) to V(D)J recombination junctions. Despite extensive in vitro studies on TdT catalytic activity, the partners of TdT that enable N-addition remain to be defined. Using an intrachromosomal substrate, we show here that, in Chinese hamter ovary (CHO) cells, ectopic expression of TdT efficiently promotes N-additions at the junction of chromosomal double-strand breaks (DSBs) generated by the meganuclease I-SceI and that the size of the N-additions is comparable with that at V(D)J junctions. Importantly, no N-addition was observed in KU80- or XRCC4-deficient cells. These data show that, in a chromosomal context of non-lymphoid cells, TdT is actually able to promote N-addition at non-V(D)J DSBs, through a process that strictly requires the components of the canonical non-homologous end-joining pathway, KU80 and XRCC4.


Subject(s)
Antigens, Nuclear/physiology , DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA Nucleotidylexotransferase/metabolism , DNA-Binding Proteins/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Ku Autoantigen , Nucleotides/metabolism , V(D)J Recombination
17.
BMC Biol ; 10: 5, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22293287

ABSTRACT

BACKGROUND: Protein kinase CK2 is a pleiotropic serine/threonine protein kinase with hundreds of reported substrates, and plays an important role in a number of cellular processes. The cellular functions of Plasmodium falciparum CK2 (PfCK2) are unknown. The parasite's genome encodes one catalytic subunit, PfCK2α, which we have previously shown to be essential for completion of the asexual erythrocytic cycle, and two putative regulatory subunits, PfCK2ß1 and PfCK2ß2. RESULTS: We now show that the genes encoding both regulatory PfCK2 subunits (PfCK2ß1 and PfCK2ß2) cannot be disrupted. Using immunofluorescence and electron microscopy, we examined the intra-erythrocytic stages of transgenic parasite lines expressing hemagglutinin (HA)-tagged catalytic and regulatory subunits (HA-CK2α, HA-PfCK2ß1 or HA-PfCK2ß2), and localized all three subunits to both cytoplasmic and nuclear compartments of the parasite. The same transgenic parasite lines were used to purify PfCK2ß1- and PfCK2ß2-containing complexes, which were analyzed by mass spectrometry. The recovered proteins were unevenly distributed between various pathways, with a large proportion of components of the chromatin assembly pathway being present in both PfCK2ß1 and PfCK2ß2 precipitates, implicating PfCK2 in chromatin dynamics. We also found that chromatin-related substrates such as nucleosome assembly proteins (Naps), histones, and two members of the Alba family are phosphorylated by PfCK2α in vitro. CONCLUSIONS: Our reverse-genetics data show that each of the two regulatory PfCK2 subunits is required for completion of the asexual erythrocytic cycle. Our interactome study points to an implication of PfCK2 in many cellular pathways, with chromatin dynamics being identified as a major process regulated by PfCK2. This study paves the way for a kinome-wide interactomics-based approach to elucidate protein kinase function in malaria parasites.


Subject(s)
Casein Kinase II/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , Gene Expression Regulation , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Casein Kinase II/genetics , Hemagglutinins/chemistry , Histone Chaperones/metabolism , Histones/metabolism , Mass Spectrometry , Microscopy, Electron , Microscopy, Fluorescence , Phosphorylation , Plasmodium falciparum/enzymology , Plasmodium falciparum/growth & development
18.
Nucleic Acids Res ; 40(7): 3066-77, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22167473

ABSTRACT

In Plasmodium falciparum, perinuclear subtelomeric chromatin conveys monoallelic expression of virulence genes. However, proteins that directly bind to chromosome ends are poorly described. Here we identify a novel DNA/RNA-binding protein family that bears homology to the archaeal protein Alba (Acetylation lowers binding affinity). We isolated three of the four PfAlba paralogs as part of a molecular complex that is associated with the P. falciparum-specific TARE6 (Telomere-Associated Repetitive Elements 6) subtelomeric region and showed in electromobility shift assays (EMSAs) that the PfAlbas bind to TARE6 repeats. In early blood stages, the PfAlba proteins were enriched at the nuclear periphery and partially co-localized with PfSir2, a TARE6-associated histone deacetylase linked to the process of antigenic variation. The nuclear location changed at the onset of parasite proliferation (trophozoite-schizont), where the PfAlba proteins were also detectable in the cytoplasm in a punctate pattern. Using single-stranded RNA (ssRNA) probes in EMSAs, we found that PfAlbas bind to ssRNA, albeit with different binding preferences. We demonstrate for the first time in eukaryotes that Alba-like proteins bind to both DNA and RNA and that their intracellular location is developmentally regulated. Discovery of the PfAlbas may provide a link between the previously described subtelomeric non-coding RNA and the regulation of antigenic variation.


Subject(s)
DNA-Binding Proteins/metabolism , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism , Archaeal Proteins/chemistry , Cytoplasm/chemistry , DNA/chemistry , DNA/metabolism , DNA-Binding Proteins/analysis , DNA-Binding Proteins/chemistry , Dimerization , Plasmodium falciparum/growth & development , Plasmodium falciparum/ultrastructure , Protein Structure, Tertiary , Protozoan Proteins/analysis , Protozoan Proteins/chemistry , RNA/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/chemistry , Repetitive Sequences, Nucleic Acid , Telomere/chemistry
19.
Cell Host Microbe ; 10(5): 451-63, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-22100161

ABSTRACT

Many microbial pathogens, including the malaria parasite Plasmodium falciparum, vary surface protein expression to evade host immune responses. P. falciparium antigenic variation is linked to var gene family-encoded clonally variant surface protein expression. Mututally exclusive var gene expression is partially controlled by spatial positioning; silent genes are retained at distinct perinuclear sites and relocated to transcriptionally active locations for monoallelic expression. We show that var introns can control this process and that var intron addition relocalizes episomes from a random to a perinuclear position. This var intron-regulated nuclear tethering and repositioning is linked to an 18 bp nuclear protein-binding element that recruits an actin protein complex. Pharmacologically induced F-actin formation, which is restricted to the nuclear periphery, repositions intron-carrying episomes and var genes and disrupts mutually exclusive var gene expression. Thus, actin polymerization relocates var genes from a repressive to an active perinuclear compartment, which is crucial for P. falciparium phenotypic variation and pathogenesis.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Virulence Factors/metabolism , Actins/genetics , Cell Nucleus/genetics , Gene Expression Regulation , Humans , Introns , Plasmids/genetics , Plasmids/metabolism , Plasmodium falciparum/genetics , Protein Binding , Protein Transport , Protozoan Proteins/genetics , Virulence Factors/genetics
20.
J Immunol ; 185(8): 4777-82, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20861355

ABSTRACT

Somatic hypermutation (SHM) of Ig genes is the result of a two-phase process initiated by activation-induced cytidine deaminase, relying on two different strategies for the introduction of mutations at CG pairs (phase I) and at AT pairs (phase II). To explain the selectivity of phase II, two mechanisms were proposed: AT-selective error-prone DNA-polymerases, deoxyuridine triphosphate (dUTP) incorporation, or both. However, there has been no experimental evidence so far of the possible involvement of the latter. We have developed a ligation-anchored PCR method based on the formation of single-strand breaks at uracils. In this study, we show the presence of uracil in hypermutating VkOx genes in wild type (AID(+/+)) mice, demonstrating that dUTP incorporation via DNA polymerases could be a major mechanism in SHM. Thus, error-prone DNA polymerases would participate in SHM via low-fidelity replication and incorporation of dUTP.


Subject(s)
Deoxyuracil Nucleotides/genetics , Genes, Immunoglobulin , Oxazolone/analogs & derivatives , Somatic Hypermutation, Immunoglobulin/genetics , Animals , Immunoglobulin kappa-Chains/genetics , Mice , Mice, Inbred BALB C , Oxazolone/immunology , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...