Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 19(12): e1011807, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38051755

ABSTRACT

Malaria is caused by the rapid proliferation of Plasmodium parasites in patients and disease severity correlates with the number of infected red blood cells in circulation. Parasite multiplication within red blood cells is called schizogony and occurs through an atypical multinucleated cell division mode. The mechanisms regulating the number of daughter cells produced by a single progenitor are poorly understood. We investigated underlying regulatory principles by quantifying nuclear multiplication dynamics in Plasmodium falciparum and knowlesi using super-resolution time-lapse microscopy. This confirmed that the number of daughter cells was consistent with a model in which a counter mechanism regulates multiplication yet incompatible with a timer mechanism. P. falciparum cell volume at the start of nuclear division correlated with the final number of daughter cells. As schizogony progressed, the nucleocytoplasmic volume ratio, which has been found to be constant in all eukaryotes characterized so far, increased significantly, possibly to accommodate the exponentially multiplying nuclei. Depleting nutrients by dilution of culture medium caused parasites to produce fewer merozoites and reduced proliferation but did not affect cell volume or total nuclear volume at the end of schizogony. Our findings suggest that the counter mechanism implicated in malaria parasite proliferation integrates extracellular resource status to modify progeny number during blood stage infection.


Subject(s)
Malaria, Falciparum , Malaria , Parasites , Animals , Humans , Parasites/physiology , Malaria, Falciparum/parasitology , Malaria/parasitology , Plasmodium falciparum/physiology , Merozoites/physiology , Erythrocytes/parasitology
2.
PLoS Pathog ; 19(12): e1011899, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38150475

ABSTRACT

Centrins are small calcium-binding proteins that have a variety of roles and are universally associated with eukaryotic centrosomes. Rapid proliferation of the malaria-causing parasite Plasmodium falciparum in the human blood depends on a particularly divergent and acentriolar centrosome, which incorporates several essential centrins. Their precise mode of action, however, remains unclear. In this study calcium-inducible liquid-liquid phase separation is revealed as an evolutionarily conserved principle of assembly for multiple centrins from P. falciparum and other species. Furthermore, the disordered N-terminus and calcium-binding motifs are defined as essential features for reversible biomolecular condensation, and we demonstrate that certain centrins can form co-condensates. In vivo analysis using live cell STED microscopy shows liquid-like dynamics of centrosomal centrin. Additionally, implementation of an inducible protein overexpression system reveals concentration-dependent formation of extra-centrosomal centrin assemblies with condensate-like properties. The timing of foci formation and dissolution suggests that centrin assembly is regulated. This study thereby provides a new model for centrin accumulation at eukaryotic centrosomes.


Subject(s)
Calcium , Parasites , Animals , Humans , Calcium/metabolism , Parasites/metabolism , Calcium-Binding Proteins/metabolism , Centrosome/metabolism
3.
mBio ; 14(4): e0077923, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37345936

ABSTRACT

Plasmodium falciparum proliferates through schizogony in the clinically relevant blood stage of infection. During schizogony, consecutive rounds of DNA replication and nuclear division give rise to multinucleated stages before cellularization occurs. Although these nuclei reside in a shared cytoplasm, DNA replication and nuclear division occur asynchronously. Here, by mapping the proteomic context of the S-phase-promoting kinase PfCRK4, we show that it has a dual role for nuclear-cycle progression: PfCRK4 orchestrates not only DNA replication, but in parallel also the rearrangement of intranuclear microtubules from hemispindles into early mitotic spindles. Live-cell imaging of a reporter parasite showed that these microtubule rearrangements coincide with the onset of DNA replication. Together, our data render PfCRK4 a key factor for nuclear-cycle progression, linking entry into S-phase with the initiation of mitotic events. In part, such links may compensate for the absence of canonical cell cycle checkpoints in P. falciparum. IMPORTANCE The human malaria parasite Plasmodium falciparum proliferates in erythrocytes through schizogony, forming multinucleated stages before cellularization occurs. In marked contrast to the pattern of proliferation seen in most model organisms, P. falciparum nuclei multiply asynchronously despite residing in a shared cytoplasm. This divergent mode of replication is, thus, a good target for therapeutic interventions. To exploit this potential, we investigated a key regulator of the parasite's unusual cell cycle, the kinase PfCRK4 and found that this kinase regulated not only DNA replication but also in parallel the rearrangement of nuclear microtubules into early mitotic spindles. Since canonical cell cycle checkpoints have not been described in P. falciparum parasites, linking entry into S-phase and the initiation of mitotic events via a kinase, may be an alternative means to exert control, which is typically achieved by checkpoints.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Proteomics , Cell Division , Cell Cycle , S Phase , Malaria, Falciparum/parasitology , Erythrocytes/parasitology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
4.
PLoS Pathog ; 19(5): e1011325, 2023 05.
Article in English | MEDLINE | ID: mdl-37130129

ABSTRACT

Malaria-causing parasites achieve rapid proliferation in human blood through multiple rounds of asynchronous nuclear division followed by daughter cell formation. Nuclear divisions critically depend on the centriolar plaque, which organizes intranuclear spindle microtubules. The centriolar plaque consists of an extranuclear compartment, which is connected via a nuclear pore-like structure to a chromatin-free intranuclear compartment. Composition and function of this non-canonical centrosome remain largely elusive. Centrins, which reside in the extranuclear part, are among the very few centrosomal proteins conserved in Plasmodium falciparum. Here we identify a novel centrin-interacting centriolar plaque protein. Conditional knock down of this Sfi1-like protein (PfSlp) caused a growth delay in blood stages, which correlated with a reduced number of daughter cells. Surprisingly, intranuclear tubulin abundance was significantly increased, which raises the hypothesis that the centriolar plaque might be implicated in regulating tubulin levels. Disruption of tubulin homeostasis caused excess microtubules and aberrant mitotic spindles. Time-lapse microscopy revealed that this prevented or delayed mitotic spindle extension but did not significantly interfere with DNA replication. Our study thereby identifies a novel extranuclear centriolar plaque factor and establishes a functional link to the intranuclear compartment of this divergent eukaryotic centrosome.


Subject(s)
Microtubules , Protozoan Proteins , Tubulin , Centrosome/metabolism , Homeostasis , Microtubules/metabolism , Tubulin/genetics , Plasmodium falciparum , Protozoan Proteins/genetics
5.
PLoS Pathog ; 19(3): e1011157, 2023 03.
Article in English | MEDLINE | ID: mdl-36862652

ABSTRACT

Malaria remains a significant threat to global health, and despite concerted efforts to curb the disease, malaria-related morbidity and mortality increased in recent years. Malaria is caused by unicellular eukaryotes of the genus Plasmodium, and all clinical manifestations occur during asexual proliferation of the parasite inside host erythrocytes. In the blood stage, Plasmodium proliferates through an unusual cell cycle mode called schizogony. Contrary to most studied eukaryotes, which divide by binary fission, the parasite undergoes several rounds of DNA replication and nuclear division that are not directly followed by cytokinesis, resulting in multinucleated cells. Moreover, despite sharing a common cytoplasm, these nuclei multiply asynchronously. Schizogony challenges our current models of cell cycle regulation and, at the same time, offers targets for therapeutic interventions. Over the recent years, the adaptation of advanced molecular and cell biological techniques have given us deeper insight how DNA replication, nuclear division, and cytokinesis are coordinated. Here, we review our current understanding of the chronological events that characterize the unusual cell division cycle of P. falciparum in the clinically relevant blood stage of infection.


Subject(s)
Malaria, Falciparum , Parasites , Plasmodium , Animals , Cell Division , Cell Cycle , Cytokinesis , Eukaryota
6.
Methods Mol Biol ; 2470: 425-433, 2022.
Article in English | MEDLINE | ID: mdl-35881363

ABSTRACT

Immunofluorescence labeling enables the detection and characterization of various parasite proteins presented on the surface of the infected red blood cell. Several approaches for immunofluorescence detection of red blood cell surface-presented proteins of Plasmodium spp. have been successfully established and published over the years. However, finding the right approach depends on the scientific question, and different protocols have different advantages. Here, we discuss some aspects that should be considered and present an easily applicable protocol for labeling parasite surface antigens, which subsequently can be analyzed by immunofluorescence microscopy (or flow cytometry).


Subject(s)
Erythrocytes , Plasmodium falciparum , Antigens, Surface/metabolism , Erythrocytes/metabolism , Membrane Proteins/metabolism , Microscopy, Fluorescence/methods , Plasmodium falciparum/metabolism , Staining and Labeling
7.
Sci Adv ; 8(13): eabj5362, 2022 04.
Article in English | MEDLINE | ID: mdl-35353560

ABSTRACT

Malaria-causing parasites proliferate within erythrocytes through schizogony, forming multinucleated stages before cellularization. Nuclear multiplication does not follow a strict geometric 2n progression, and each proliferative cycle produces a variable number of progeny. Here, by tracking nuclei and DNA replication, we show that individual nuclei replicate their DNA at different times, despite residing in a shared cytoplasm. Extrapolating from experimental data using mathematical modeling, we provide strong indication that a limiting factor exists, which slows down the nuclear multiplication rate. Consistent with this prediction, our data show that temporally overlapping DNA replication events were significantly slower than partially overlapping or nonoverlapping events. Our findings suggest the existence of evolutionary pressure that selects for asynchronous DNA replication, balancing available resources with rapid pathogen proliferation.


Subject(s)
Cell Nucleus , Plasmodium falciparum , Cell Division , DNA Replication , Erythrocytes/parasitology , Plasmodium falciparum/genetics
8.
PLoS Pathog ; 17(10): e1009969, 2021 10.
Article in English | MEDLINE | ID: mdl-34614006

ABSTRACT

The pathology associated with malaria infection is largely due to the ability of infected human RBCs to adhere to a number of receptors on endothelial cells within tissues and organs. This phenomenon is driven by the export of parasite-encoded proteins to the host cell, the exact function of many of which is still unknown. Here we inactivate the function of one of these exported proteins, PFA66, a member of the J-domain protein family. Although parasites lacking this protein were still able to grow in cell culture, we observed severe defects in normal host cell modification, including aberrant morphology of surface knobs, disrupted presentation of the cytoadherence molecule PfEMP1, and a total lack of cytoadherence, despite the presence of the knob associated protein KAHRP. Complementation assays demonstrate that an intact J-domain is required for recovery to a wild-type phenotype and suggest that PFA66 functions in concert with a HSP70 to carry out host cell modification. Strikingly, this HSP70 is likely to be of host origin. ATPase assays on recombinant protein verify a functional interaction between PFA66 and residual host cell HSP70. Taken together, our data reveal a role for PFA66 in host cell modification, strongly implicate human HSP70s as being essential in this process and uncover a new KAHRP-independent molecular factor required for correct knob biogenesis.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Host-Parasite Interactions/physiology , Malaria, Falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Plasmodium falciparum/metabolism , Virulence
9.
Life Sci Alliance ; 4(11)2021 11.
Article in English | MEDLINE | ID: mdl-34535568

ABSTRACT

Proliferation of Plasmodium falciparum in red blood cells is the cause of malaria and is underpinned by an unconventional cell division mode, called schizogony. Contrary to model organisms, P. falciparum replicates by multiple rounds of nuclear divisions that are not interrupted by cytokinesis. Organization and dynamics of critical nuclear division factors remain poorly understood. Centriolar plaques, the centrosomes of P. falciparum, serve as microtubule organizing centers and have an acentriolar, amorphous structure. The small size of parasite nuclei has precluded detailed analysis of intranuclear microtubule organization by classical fluorescence microscopy. We apply recently developed super-resolution and time-lapse imaging protocols to describe microtubule reconfiguration during schizogony. Analysis of centrin, nuclear pore, and microtubule positioning reveals two distinct compartments of the centriolar plaque. Whereas centrin is extranuclear, we confirm by correlative light and electron tomography that microtubules are nucleated in a previously unknown and extended intranuclear compartment, which is devoid of chromatin but protein-dense. This study generates a working model for an unconventional centrosome and enables a better understanding about the diversity of eukaryotic cell division.


Subject(s)
Centrosome/physiology , Intranuclear Space/metabolism , Microtubules/metabolism , Cell Division/physiology , Cell Line , Centrosome/metabolism , Chromatin , Cytokinesis , Humans , Microtubule-Organizing Center/physiology , Microtubules/physiology , Nuclear Pore , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism
10.
Front Cell Infect Microbiol ; 11: 658616, 2021.
Article in English | MEDLINE | ID: mdl-34026661

ABSTRACT

Regulating the number of progeny generated by replicative cell cycles is critical for any organism to best adapt to its environment. Classically, the decision whether to divide further is made after cell division is completed by cytokinesis and can be triggered by intrinsic or extrinsic factors. Contrarily, cell cycles of some species, such as the malaria-causing parasites, go through multinucleated cell stages. Hence, their number of progeny is determined prior to the completion of cell division. This should fundamentally affect how the process is regulated and raises questions about advantages and challenges of multinucleation in eukaryotes. Throughout their life cycle Plasmodium spp. parasites undergo four phases of extensive proliferation, which differ over three orders of magnitude in the amount of daughter cells that are produced by a single progenitor. Even during the asexual blood stage proliferation parasites can produce very variable numbers of progeny within one replicative cycle. Here, we review the few factors that have been shown to affect those numbers. We further provide a comparative quantification of merozoite numbers in several P. knowlesi and P. falciparum parasite strains, and we discuss the general processes that may regulate progeny number in the context of host-parasite interactions. Finally, we provide a perspective of the critical knowledge gaps hindering our understanding of the molecular mechanisms underlying this exciting and atypical mode of parasite multiplication.


Subject(s)
Malaria, Falciparum , Parasites , Animals , Cytokinesis , Erythrocytes , Merozoites , Plasmodium falciparum
11.
Commun Biol ; 4(1): 600, 2021 05 20.
Article in English | MEDLINE | ID: mdl-34017052

ABSTRACT

The eukaryotic signal recognition particle (SRP) contains an Alu domain, which docks into the factor binding site of translating ribosomes and confers translation retardation. The canonical Alu domain consists of the SRP9/14 protein heterodimer and a tRNA-like folded Alu RNA that adopts a strictly 'closed' conformation involving a loop-loop pseudoknot. Here, we study the structure of the Alu domain from Plasmodium falciparum (PfAlu), a divergent apicomplexan protozoan that causes human malaria. Using NMR, SAXS and cryo-EM analyses, we show that, in contrast to its prokaryotic and eukaryotic counterparts, the PfAlu domain adopts an 'open' Y-shaped conformation. We show that cytoplasmic P. falciparum ribosomes are non-discriminative and recognize both the open PfAlu and closed human Alu domains with nanomolar affinity. In contrast, human ribosomes do not provide high affinity binding sites for either of the Alu domains. Our analyses extend the structural database of Alu domains to the protozoan species and reveal species-specific differences in the recognition of SRP Alu domains by ribosomes.


Subject(s)
Alu Elements , Plasmodium falciparum/metabolism , Ribosomes/metabolism , Signal Recognition Particle/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Models, Molecular , Nucleic Acid Conformation , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Ribosomes/genetics , Scattering, Small Angle
12.
PLoS Biol ; 19(3): e3001105, 2021 03.
Article in English | MEDLINE | ID: mdl-33705378

ABSTRACT

Apicomplexan parasites are defined by complex apical structures, which are necessary for interaction with incredibly diverse host cells. Two studies now amend a long-standing paradigm by showing conservation of an essential ring structure in the entire phylum.


Subject(s)
Apicomplexa , Parasites , Animals , Cytoskeleton
13.
mBio ; 11(1)2020 01 21.
Article in English | MEDLINE | ID: mdl-31964736

ABSTRACT

The human malaria parasite Plasmodium falciparum uses mutually exclusive expression of the PfEMP1-encoding var gene family to evade the host immune system. Despite progress in the molecular understanding of the default silencing mechanism, the activation mechanism of the uniquely expressed var member remains elusive. A GC-rich noncoding RNA (ncRNA) gene family has coevolved with Plasmodium species that express var genes. Here, we show that this ncRNA family is transcribed in a clonally variant manner, with predominant transcription of a single member occurring when the ncRNA is located adjacent to and upstream of an active var gene. We developed a specific CRISPR interference (CRISPRi) strategy that allowed for the transcriptional repression of all GC-rich members. A lack of GC-rich ncRNA transcription led to the downregulation of the entire var gene family in ring-stage parasites. Strikingly, in mature blood-stage parasites, the GC-rich ncRNA CRISPRi affected the transcription patterns of other clonally variant gene families, including the downregulation of all Pfmc-2TM members. We provide evidence for the key role of GC-rich ncRNA transcription in var gene activation and discovered a molecular link between the transcriptional control of various clonally variant multigene families involved in parasite virulence. This work opens new avenues for elucidating the molecular processes that control immune evasion and pathogenesis in P. falciparumIMPORTANCEPlasmodium falciparum is the deadliest malaria parasite species, accounting for the vast majority of disease cases and deaths. The virulence of this parasite is reliant upon the mutually exclusive expression of cytoadherence proteins encoded by the 60-member var gene family. Antigenic variation of this multigene family serves as an immune evasion mechanism, ultimately leading to chronic infection and pathogenesis. Understanding the regulation mechanism of antigenic variation is key to developing new therapeutic and control strategies. Our study uncovers a novel layer in the epigenetic regulation of transcription of this family of virulence genes by means of a multigene-targeting CRISPR interference approach.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , GC Rich Sequence , Multigene Family , Plasmodium falciparum/genetics , RNA, Untranslated/genetics , Antigenic Variation/genetics , Gene Expression Regulation , Malaria, Falciparum/parasitology , Nucleic Acid Conformation , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , RNA, Untranslated/chemistry , Transcription, Genetic , Virulence
14.
Mol Biochem Parasitol ; 229: 47-52, 2019 04.
Article in English | MEDLINE | ID: mdl-30831155

ABSTRACT

Immunofluorescence staining is the key technique for visualizing organization of endogenous cellular structures in single cells. Labeling and imaging of blood stage Plasmodium falciparum has always been challenging since it is a small intracellular parasite. A widely-used standard for parasite immunofluorescence is fixation in suspension with addition of minute amounts of glutaraldehyde to the paraformaldehyde-based solution. While this maintains red blood cell integrity, it has been postulated that antigenicity of the parasite proteins was, if at all, only slightly reduced. Here we show the deleterious effect that even these small quantities of glutaraldehyde can have on immunofluorescence staining quality and present an alternative cell seeding protocol that allows fixation with only paraformaldehyde. The highly improved signal intensity and staining efficiency enabled us to carry out RescueSTED nanoscopy on microtubules and nuclear pores and describe their organization in greater detail throughout the blood stage cycle.


Subject(s)
Erythrocytes/chemistry , Erythrocytes/parasitology , Fluorescent Antibody Technique/methods , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , Fluorescent Dyes/chemistry , Humans , Nanotechnology , Staining and Labeling
15.
Nature ; 563(7729): 121-125, 2018 11.
Article in English | MEDLINE | ID: mdl-30333624

ABSTRACT

Many evolutionarily distant pathogenic organisms have evolved similar survival strategies to evade the immune responses of their hosts. These include antigenic variation, through which an infecting organism prevents clearance by periodically altering the identity of proteins that are visible to the immune system of the host1. Antigenic variation requires large reservoirs of immunologically diverse antigen genes, which are often generated through homologous recombination, as well as mechanisms to ensure the expression of one or very few antigens at any given time. Both homologous recombination and gene expression are affected by three-dimensional genome architecture and local DNA accessibility2,3. Factors that link three-dimensional genome architecture, local chromatin conformation and antigenic variation have, to our knowledge, not yet been identified in any organism. One of the major obstacles to studying the role of genome architecture in antigenic variation has been the highly repetitive nature and heterozygosity of antigen-gene arrays, which has precluded complete genome assembly in many pathogens. Here we report the de novo haplotype-specific assembly and scaffolding of the long antigen-gene arrays of the model protozoan parasite Trypanosoma brucei, using long-read sequencing technology and conserved features of chromosome folding4. Genome-wide chromosome conformation capture (Hi-C) reveals a distinct partitioning of the genome, with antigen-encoding subtelomeric regions that are folded into distinct, highly compact compartments. In addition, we performed a range of analyses-Hi-C, fluorescence in situ hybridization, assays for transposase-accessible chromatin using sequencing and single-cell RNA sequencing-that showed that deletion of the histone variants H3.V and H4.V increases antigen-gene clustering, DNA accessibility across sites of antigen expression and switching of the expressed antigen isoform, via homologous recombination. Our analyses identify histone variants as a molecular link between global genome architecture, local chromatin conformation and antigenic variation.


Subject(s)
Antigenic Variation/genetics , Chromatin/genetics , Chromatin/metabolism , DNA, Protozoan/metabolism , Genome/genetics , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/immunology , DNA, Protozoan/genetics , Haplotypes/genetics , Histones/deficiency , Histones/genetics , Multigene Family/genetics , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Variant Surface Glycoproteins, Trypanosoma/biosynthesis , Variant Surface Glycoproteins, Trypanosoma/genetics
16.
Cell Rep ; 22(11): 2951-2963, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29539423

ABSTRACT

Heterochromatin plays a central role in the process of immune evasion, pathogenesis, and transmission of the malaria parasite Plasmodium falciparum during blood stage infection. Here, we use ChIP sequencing to demonstrate that sporozoites from mosquito salivary glands expand heterochromatin at subtelomeric regions to silence blood-stage-specific genes. Our data also revealed that heterochromatin enrichment is predictive of the transcription status of clonally variant genes members that mediate cytoadhesion in blood stage parasites. A specific member (here called NF54varsporo) of the var gene family remains euchromatic, and the resultant PfEMP1 (NF54_SpzPfEMP1) is expressed at the sporozoite surface. NF54_SpzPfEMP1-specific antibodies efficiently block hepatocyte infection in a strain-specific manner. Furthermore, human volunteers immunized with infective sporozoites developed antibodies against NF54_SpzPfEMP1. Overall, we show that the epigenetic signature of var genes is reset in mosquito stages. Moreover, the identification of a strain-specific sporozoite PfEMP1 is highly relevant for vaccine design based on sporozoites.


Subject(s)
Hepatocytes/immunology , Protozoan Proteins/metabolism , Sporozoites/immunology , Animals
17.
mBio ; 8(4)2017 07 11.
Article in English | MEDLINE | ID: mdl-28698275

ABSTRACT

Plasmodium falciparum relies on monoallelic expression of 1 of 60 var virulence genes for antigenic variation and host immune evasion. Each var gene contains a conserved intron which has been implicated in previous studies in both activation and repression of transcription via several epigenetic mechanisms, including interaction with the var promoter, production of long noncoding RNAs (lncRNAs), and localization to repressive perinuclear sites. However, functional studies have relied primarily on artificial expression constructs. Using the recently developed P. falciparum clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system, we directly deleted the var2csa P. falciparum 3D7_1200600 (Pf3D7_1200600) endogenous intron, resulting in an intronless var gene in a natural, marker-free chromosomal context. Deletion of the var2csa intron resulted in an upregulation of transcription of the var2csa gene in ring-stage parasites and subsequent expression of the PfEMP1 protein in late-stage parasites. Intron deletion did not affect the normal temporal regulation and subsequent transcriptional silencing of the var gene in trophozoites but did result in increased rates of var gene switching in some mutant clones. Transcriptional repression of the intronless var2csa gene could be achieved via long-term culture or panning with the CD36 receptor, after which reactivation was possible with chondroitin sulfate A (CSA) panning. These data suggest that the var2csa intron is not required for silencing or activation in ring-stage parasites but point to a subtle role in regulation of switching within the var gene family.IMPORTANCEPlasmodium falciparum is the most virulent species of malaria parasite, causing high rates of morbidity and mortality in those infected. Chronic infection depends on an immune evasion mechanism termed antigenic variation, which in turn relies on monoallelic expression of 1 of ~60 var genes. Understanding antigenic variation and the transcriptional regulation of monoallelic expression is important for developing drugs and/or vaccines. The var gene family encodes the antigenic surface proteins that decorate infected erythrocytes. Until recently, studying the underlying genetic elements that regulate monoallelic expression in P. falciparum was difficult, and most studies relied on artificial systems such as episomal reporter genes. Our study was the first to use CRISPR/Cas9 genome editing for the functional study of an important, conserved genetic element of var genes-the intron-in an endogenous, episome-free manner. Our findings shed light on the role of the var gene intron in transcriptional regulation of monoallelic expression.


Subject(s)
Antigens, Protozoan/genetics , CRISPR-Cas Systems , Gene Silencing , Introns , Plasmodium falciparum/genetics , Transcriptional Activation , Antigenic Variation , Gene Editing , Gene Expression Regulation , Protozoan Proteins/genetics
18.
Nucleic Acids Res ; 44(20): 9710-9718, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27466391

ABSTRACT

Monoallelic expression of the var multigene family enables immune evasion of the malaria parasite Plasmodium falciparum in its human host. At a given time only a single member of the 60-member var gene family is expressed at a discrete perinuclear region called the 'var expression site'. However, the mechanism of var gene counting remains ill-defined. We hypothesize that activation factors associating specifically with the expression site play a key role in this process. Here, we investigate the role of a GC-rich non-coding RNA (ncRNA) gene family composed of 15 highly homologous members. GC-rich genes are positioned adjacent to var genes in chromosome-central gene clusters but are absent near subtelomeric var genes. Fluorescence in situ hybridization demonstrates that GC-rich ncRNA localizes to the perinuclear expression site of central and subtelomeric var genes in trans. Importantly, overexpression of distinct GC-rich ncRNA members disrupts the gene counting process at the single cell level and results in activation of a specific subset of var genes in distinct clones. We identify the first trans-acting factor targeted to the elusive perinuclear var expression site and open up new avenues to investigate ncRNA function in antigenic variation of malaria and other protozoan pathogens.


Subject(s)
Base Composition , Gene Expression Regulation , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA, Untranslated/genetics , Transcriptional Activation , Base Sequence , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Microscopy, Fluorescence , Nucleic Acid Conformation , Plasmodium falciparum/metabolism , RNA, Untranslated/chemistry
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...