Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(1): 105586, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38141766

ABSTRACT

About 247 million cases of malaria occurred in 2021 with Plasmodium falciparum accounting for the majority of 619,000 deaths. In the absence of a widely available vaccine, chemotherapy remains crucial to prevent, treat, and contain the disease. The efficacy of several drugs currently used in the clinic is likely to suffer from the emergence of resistant parasites. A global effort to identify lead compounds led to several initiatives such as the Medicine for Malaria Ventures (MMV), a repository of compounds showing promising efficacy in killing the parasite in cell-based assays. Here, we used mass spectrometry coupled with cellular thermal shift assay to identify putative protein targets of MMV000848, a compound with an in vitro EC50 of 0.5 µM against the parasite. Thermal shift assays showed a strong increase of P. falciparum purine nucleoside phosphorylase (PfPNP) melting temperature by up to 15 °C upon incubation with MMV000848. Binding and enzymatic assays returned a KD of 1.52 ± 0.495 µM and an IC50 value of 21.5 ± 2.36 µM. The inhibition is competitive with respect to the substrate, as confirmed by a cocrystal structure of PfPNP bound with MMV000848 at the active site, determined at 1.85 Å resolution. In contrast to transition states inhibitors, MMV000848 specifically inhibits the parasite enzyme but not the human ortholog. An isobologram analysis shows subadditivity with immucillin H and with quinine respectively, suggesting overlapping modes of action between these compounds. These results point to PfPNP as a promising antimalarial target and suggest avenues to improve inhibitor potency.


Subject(s)
Antimalarials , Plasmodium falciparum , Purine-Nucleoside Phosphorylase , Antimalarials/chemistry , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Purine-Nucleoside Phosphorylase/chemistry , Quinine/chemistry , Mass Spectrometry , Protein Binding
2.
PLoS Pathog ; 19(1): e1011118, 2023 01.
Article in English | MEDLINE | ID: mdl-36696458

ABSTRACT

Resistance of the human malaria parasites, Plasmodium falciparum, to artemisinins is now fully established in Southeast Asia and is gradually emerging in Sub-Saharan Africa. Although nonsynonymous SNPs in the pfk13 Kelch-repeat propeller (KREP) domain are clearly associated with artemisinin resistance, their functional relevance requires cooperation with other genetic factors/alterations of the P. falciparum genome, collectively referred to as genetic background. Here we provide experimental evidence that P. falciparum cyclophilin 19B (PfCYP19B) may represent one putative factor in this genetic background, contributing to artemisinin resistance via its increased expression. We show that overexpression of PfCYP19B in vitro drives limited but significant resistance to not only artemisinin but also piperaquine, an important partner drug in artemisinin-based combination therapies. We showed that PfCYP19B acts as a negative regulator of the integrated stress response (ISR) pathway by modulating levels of phosphorylated eIF2α (eIF2α-P). Curiously, artemisinin and piperaquine affect eIF2α-P in an inverse direction that in both cases can be modulated by PfCYP19B towards resistance. Here we also provide evidence that the upregulation of PfCYP19B in the drug-resistant parasites appears to be maintained by a short tandem repeat (SRT) sequence polymorphism in the gene's promoter region. These results support a model that artemisinin (and other drugs) resistance mechanisms are complex genetic traits being contributed to by altered expression of multiple genes driven by genetic polymorphism at their promoter regions.


Subject(s)
Antimalarials , Drug Resistance , Malaria, Falciparum , Plasmodium falciparum , Humans , Antimalarials/pharmacology , Cyclophilins/genetics , Cyclophilins/metabolism , Drug Resistance/genetics , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Microsatellite Repeats , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Up-Regulation
3.
J Integr Plant Biol ; 65(6): 1442-1466, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36807520

ABSTRACT

Plants accumulate a vast array of secondary metabolites, which constitute a natural resource for pharmaceuticals. Oldenlandia corymbosa belongs to the Rubiaceae family, and has been used in traditional medicine to treat different diseases, including cancer. However, the active metabolites of the plant, their biosynthetic pathway and mode of action in cancer are unknown. To fill these gaps, we exposed this plant to eight different stress conditions and combined different omics data capturing gene expression, metabolic profiles, and anti-cancer activity. Our results show that O. corymbosa extracts are active against breast cancer cell lines and that ursolic acid is responsible for this activity. Moreover, we assembled a high-quality genome and uncovered two genes involved in the biosynthesis of ursolic acid. Finally, we also revealed that ursolic acid causes mitotic catastrophe in cancer cells and identified three high-confidence protein binding targets by Cellular Thermal Shift Assay (CETSA) and reverse docking. Altogether, these results constitute a valuable resource to further characterize the biosynthesis of active metabolites in the Oldenlandia group, while the mode of action of ursolic acid will allow us to further develop this valuable compound.


Subject(s)
Oldenlandia , Oldenlandia/chemistry , Transcriptome , Metabolomics , Genomics , Ursolic Acid
4.
Article in English | MEDLINE | ID: mdl-32071059

ABSTRACT

We report a systematic, cellular phenotype-based antimalarial screening of the Medicines for Malaria Venture Pathogen Box collection, which facilitated the identification of specific blockers of late-stage intraerythrocytic development of Plasmodium falciparum First, from standard growth inhibition assays, we identified 173 molecules with antimalarial activity (50% effective concentration [EC50] ≤ 10 µM), which included 62 additional molecules over previously known antimalarial candidates from the Pathogen Box. We identified 90 molecules with EC50 of ≤1 µM, which had significant effect on the ring-trophozoite transition, while 9 molecules inhibited the trophozoite-schizont transition and 21 molecules inhibited the schizont-ring transition (with ≥50% parasites failing to proceed to the next stage) at 1 µM. We therefore rescreened all 173 molecules and validated hits in microscopy to prioritize 12 hits as selective blockers of the schizont-ring transition. Seven of these molecules inhibited the calcium ionophore-induced egress of Toxoplasma gondii, a related apicomplexan parasite, suggesting that the inhibitors may be acting via a conserved mechanism which could be further exploited for target identification studies. We demonstrate that two molecules, MMV020670 and MMV026356, identified as schizont inhibitors in our screens, induce the fragmentation of DNA in merozoites, thereby impairing their ability to egress and invade. Further mechanistic studies would facilitate the therapeutic exploitation of these molecules as broadly active inhibitors targeting late-stage development and egress of apicomplexan parasites relevant to human health.


Subject(s)
Antimalarials/pharmacology , Drug Evaluation, Preclinical/methods , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , DNA Fragmentation/drug effects , Humans , Merozoites/drug effects , Parasitic Sensitivity Tests , Schizonts/drug effects , Trophozoites/drug effects
5.
PLoS Pathog ; 14(3): e1006930, 2018 03.
Article in English | MEDLINE | ID: mdl-29538461

ABSTRACT

Due to their remarkable parasitocidal activity, artemisinins represent the key components of first-line therapies against Plasmodium falciparum malaria. However, the decline in efficacy of artemisinin-based drugs jeopardizes global efforts to control and ultimately eradicate the disease. To better understand the resistance phenotype, artemisinin-resistant parasite lines were derived from two clones of the 3D7 strain of P. falciparum using a selection regimen that mimics how parasites interact with the drug within patients. This long term in vitro selection induced profound stage-specific resistance to artemisinin and its relative compounds. Chemosensitivity and transcriptional profiling of artemisinin-resistant parasites indicate that enhanced adaptive responses against oxidative stress and protein damage are associated with decreased artemisinin susceptibility. This corroborates our previous findings implicating these cellular functions in artemisinin resistance in natural infections. Genomic characterization of the two derived parasite lines revealed a spectrum of sequence and copy number polymorphisms that could play a role in regulating artemisinin response, but did not include mutations in pfk13, the main marker of artemisinin resistance in Southeast Asia. Taken together, here we present a functional in vitro model of artemisinin resistance that is underlined by a new set of genetic polymorphisms as potential genetic markers.


Subject(s)
Artemisinins/pharmacology , Drug Resistance/genetics , Genetic Markers , Malaria, Falciparum/parasitology , Oxidative Stress , Polymorphism, Genetic , Protozoan Proteins/metabolism , Antimalarials/pharmacology , Gene Expression Profiling , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/genetics , Phenotype , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protozoan Proteins/genetics
6.
Malar J ; 19(1): 363, 2020 Oct 09.
Article in English | MEDLINE | ID: mdl-33036628

ABSTRACT

BACKGROUND: Sequencing technology advancements opened new opportunities to use transcriptomics for studying malaria pathology and epidemiology. Even though in recent years the study of whole parasite transcriptome proved to be essential in understanding parasite biology there is no compiled up-to-date reference protocol for the efficient generation of transcriptome data from growing number of samples. Here, a comprehensive methodology on how to preserve, extract, amplify, and sequence full-length mRNA transcripts from Plasmodium-infected blood samples is presented that can be fully streamlined for high-throughput studies. RESULTS: The utility of various commercially available RNA-preserving reagents in a range of storage conditions was evaluated. Similarly, several RNA extraction protocols were compared and the one most suitable method for the extraction of high-quality total RNA from low-parasitaemia and low-volume blood samples was established. Furthermore, the criteria needed to evaluate the quality and integrity of Plasmodium RNA in the presence of human RNA was updated. Optimization of SMART-seq2 amplification method to better suit AT-rich Plasmodium falciparum RNA samples allowed us to generate high-quality transcriptomes from as little as 10 ng of total RNA and a lower parasitaemia limit of 0.05%. Finally, a modified method for depletion of unwanted human haemoglobin transcripts using in vitro CRISPR-Cas9 treatment was designed, thus improving parasite transcriptome coverage in low parasitaemia samples. To prove the functionality of the pipeline for both laboratory and field strains, the highest  2-hour resolution RNA-seq transcriptome for P. falciparum 3D7 intraerythrocytic life cycle available to  date was generated, and the entire protocol was applied to create the largest transcriptome data from Southeast Asian field isolates. CONCLUSIONS: Overall, the presented methodology is an inclusive pipeline for generation of good quality transcriptomic data from a diverse range of Plasmodium-infected blood samples with varying parasitaemia and RNA inputs. The flexibility of this pipeline to be adapted to robotic handling will facilitate both small and large-scale future transcriptomic studies in the field of malaria.


Subject(s)
Blood/parasitology , Gene Expression Profiling/methods , Plasmodium falciparum/genetics , RNA, Protozoan/analysis , Specimen Handling/methods , High-Throughput Nucleotide Sequencing , Humans , Malaria, Falciparum/physiopathology , Plasmodium falciparum/isolation & purification
7.
BMC Genomics ; 19(1): 372, 2018 May 21.
Article in English | MEDLINE | ID: mdl-29783949

ABSTRACT

BACKGROUND: Gene copy number variants (CNVs), which consist of deletions and amplifications of single or sets of contiguous genes, contribute to the great diversity in the Plasmodium falciparum genome. In vitro studies in the laboratory have revealed their important role in parasite fitness phenotypes such as red cell invasion, transmissibility and cytoadherence. Studies of natural parasite populations indicate that CNVs are also common in the field and thus may facilitate adaptation of the parasite to its local environment. RESULTS: In a survey of 183 fresh field isolates from three populations in Eastern Africa with different malaria transmission intensities, we identified 94 CNV loci using microarrays. All CNVs had low population frequencies (minor allele frequency < 5%) but each parasite isolate carried an average of 8 CNVs. Nine CNVs showed high levels of population differentiation (FST > 0.3) and nine exhibited significant clines in population frequency across a gradient in transmission intensity. The clearest example of this was a large deletion on chromosome 9 previously reported only in laboratory-adapted isolates. This deletion was present in 33% of isolates from a population with low and highly seasonal malaria transmission, and in < 9% of isolates from populations with higher transmission. Subsets of CNVs were strongly correlated in their population frequencies, implying co-selection. CONCLUSIONS: These results support the hypothesis that CNVs are the target of selection in natural populations of P. falciparum. Their environment-specific patterns observed here imply an important role for them in conferring adaptability to the parasite thus enabling it to persist in its highly diverse ecological environment.


Subject(s)
DNA Copy Number Variations , Plasmodium falciparum/genetics , Adaptation, Physiological/genetics , Africa, Eastern , Child , Child, Preschool , Chromosome Deletion , Female , Humans , Infant , Male , Plasmodium falciparum/physiology
8.
PLoS Biol ; 13(4): e1002132, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25901609

ABSTRACT

Successful control of falciparum malaria depends greatly on treatment with artemisinin combination therapies. Thus, reports that resistance to artemisinins (ARTs) has emerged, and that the prevalence of this resistance is increasing, are alarming. ART resistance has recently been linked to mutations in the K13 propeller protein. We undertook a detailed kinetic analysis of the drug responses of K13 wild-type and mutant isolates of Plasmodium falciparum sourced from a region in Cambodia (Pailin). We demonstrate that ART treatment induces growth retardation and an accumulation of ubiquitinated proteins, indicative of a cellular stress response that engages the ubiquitin/proteasome system. We show that resistant parasites exhibit lower levels of ubiquitinated proteins and delayed onset of cell death, indicating an enhanced cell stress response. We found that the stress response can be targeted by inhibiting the proteasome. Accordingly, clinically used proteasome inhibitors strongly synergize ART activity against both sensitive and resistant parasites, including isogenic lines expressing mutant or wild-type K13. Synergy is also observed against Plasmodium berghei in vivo. We developed a detailed model of parasite responses that enables us to infer, for the first time, in vivo parasite clearance profiles from in vitro assessments of ART sensitivity. We provide evidence that the clinical marker of resistance (delayed parasite clearance) is an indirect measure of drug efficacy because of the persistence of unviable parasites with unchanged morphology in the circulation, and we suggest alternative approaches for the direct measurement of viability. Our model predicts that extending current three-day ART treatment courses to four days, or splitting the doses, will efficiently clear resistant parasite infections. This work provides a rationale for improving the detection of ART resistance in the field and for treatment strategies that can be employed in areas with ART resistance.


Subject(s)
Artemisinins/pharmacology , Plasmodium falciparum/physiology , Stress, Physiological , Animals , Dose-Response Relationship, Drug , Drug Resistance , Genome, Protozoan , Mutation , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics
9.
Clin Infect Dis ; 63(6): 784-791, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27313266

ABSTRACT

BACKGROUND: Deployment of mefloquine-artesunate (MAS3) on the Thailand-Myanmar border has led to a sustained reduction in falciparum malaria, although antimalarial efficacy has declined substantially in recent years. The role of Plasmodium falciparum K13 mutations (a marker of artemisinin resistance) in reducing treatment efficacy remains controversial. METHODS: Between 2003 and 2013, we studied the efficacy of MAS3 in 1005 patients with uncomplicated P. falciparum malaria in relation to molecular markers of resistance. RESULTS: Polymerase chain reaction (PCR)-adjusted cure rates declined from 100% in 2003 to 81.1% in 2013 as the proportions of isolates with multiple Pfmdr1 copies doubled from 32.4% to 64.7% and those with K13 mutations increased from 6.7% to 83.4%. K13 mutations conferring moderate artemisinin resistance (notably E252Q) predominated initially but were later overtaken by propeller mutations associated with slower parasite clearance (notably C580Y). Those infected with both multiple Pfmdr1 copy number and a K13 propeller mutation were 14 times more likely to fail treatment. The PCR-adjusted cure rate was 57.8% (95% confidence interval [CI], 45.4, 68.3) compared with 97.8% (95% CI, 93.3, 99.3) in patients with K13 wild type and Pfmdr1 single copy. K13 propeller mutation alone was a strong risk factor for recrudescence (P = .009). The combined population attributable fraction of recrudescence associated with K13 mutation and Pfmdr1 amplification was 82%. CONCLUSIONS: The increasing prevalence of K13 mutations was the decisive factor for the recent and rapid decline in efficacy of artemisinin-based combination (MAS3) on the Thailand-Myanmar border.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum/drug therapy , Mefloquine , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemisinins/pharmacology , Artemisinins/therapeutic use , Drug Resistance , Female , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Male , Mefloquine/pharmacology , Mefloquine/therapeutic use , Myanmar/epidemiology , Plasmodium falciparum/pathogenicity , Prospective Studies , Thailand/epidemiology
10.
Mol Microbiol ; 91(5): 918-34, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24372851

ABSTRACT

Drug resistance in Plasmodium falciparum remains a challenge for the malaria eradication programmes around the world. With the emergence of artemisinin resistance, the efficacy of the partner drugs in the artemisinin combination therapies (ACT) that include quinoline-based drugs is becoming critical. So far only few resistance markers have been identified from which only two transmembrane transporters namely PfMDR1 (an ATP-binding cassette transporter) and PfCRT (a drug-metabolite transporter) have been experimentally verified. Another P. falciparum transporter, the ATP-binding cassette containing multidrug resistance-associated protein (PfMRP2) represents an additional possible factor of drug resistance in P. falciparum. In this study, we identified a parasite clone that is derived from the 3D7 P. falciparum strain and shows increased resistance to chloroquine, mefloquine and quinine through the trophozoite and schizont stages. We demonstrate that the resistance phenotype is caused by a 4.1 kb deletion in the 5' upstream region of the pfmrp2 gene that leads to an alteration in the pfmrp2 transcription and thus increased level of PfMRP2 protein. These results also suggest the importance of putative promoter elements in regulation of gene expression during the P. falciparum intra-erythrocytic developmental cycle and the potential of genetic polymorphisms within these regions to underlie drug resistance.


Subject(s)
Drug Resistance/genetics , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Polymorphism, Genetic , Promoter Regions, Genetic/genetics , Protozoan Proteins/genetics , Quinolines/pharmacology , Antimalarials/pharmacology , Base Pairing/genetics , Base Sequence , Clone Cells , Drug Resistance/drug effects , Gene Expression Regulation/drug effects , Genome, Protozoan/genetics , Intracellular Space/drug effects , Intracellular Space/metabolism , Molecular Sequence Data , Polymerase Chain Reaction , Protein Transport/drug effects , Protozoan Proteins/metabolism , Sequence Analysis, DNA , Sequence Deletion/genetics , Transcription, Genetic/drug effects , Transcriptome/genetics
11.
Genome Res ; 22(5): 925-38, 2012 May.
Article in English | MEDLINE | ID: mdl-22415456

ABSTRACT

Malaria genetic variation has been extensively characterized, but the level of epigenetic plasticity remains largely unexplored. Here we provide a comprehensive characterization of transcriptional variation in the most lethal malaria parasite, Plasmodium falciparum, based on highly accurate transcriptional analysis of isogenic parasite lines grown under homogeneous conditions. This analysis revealed extensive transcriptional heterogeneity within genetically homogeneous clonal parasite populations. We show that clonally variant expression controlled at the epigenetic level is an intrinsic property of specific genes and gene families, the majority of which participate in host-parasite interactions. Intrinsic transcriptional variability is not restricted to genes involved in immune evasion, but also affects genes linked to lipid metabolism, protein folding, erythrocyte remodeling, or transcriptional regulation, among others, indicating that epigenetic variation results in both antigenic and functional variation. We observed a general association between heterochromatin marks and clonally variant expression, extending previous observations for specific genes to essentially all variantly expressed gene families. These results suggest that phenotypic variation of functionally unrelated P. falciparum gene families is mediated by a common mechanism based on reversible formation of H3K9me3-based heterochromatin. In changing environments, diversity confers fitness to a population. Our results support the idea that P. falciparum uses a bet-hedging strategy, as an alternative to directed transcriptional responses, to adapt to common fluctuations in its environment. Consistent with this idea, we found that transcriptionally different isogenic parasite lines markedly differed in their survival to heat-shock mimicking febrile episodes and adapted to periodic heat-shock with a pattern consistent with natural selection of pre-existing parasites.


Subject(s)
Epigenesis, Genetic , Genes, Protozoan , Plasmodium falciparum/genetics , Transcriptome , Adaptation, Physiological/genetics , Culture Techniques , Gene Expression Profiling , Heat-Shock Response , Heterochromatin/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/physiology , Transcription, Genetic , Trophozoites/growth & development , Trophozoites/metabolism , Trophozoites/physiology
12.
PLoS Pathog ; 9(2): e1003170, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23468622

ABSTRACT

Epigenetic mechanisms are emerging as one of the major factors of the dynamics of gene expression in the human malaria parasite, Plasmodium falciparum. To elucidate the role of chromatin remodeling in transcriptional regulation associated with the progression of the P. falciparum intraerythrocytic development cycle (IDC), we mapped the temporal pattern of chromosomal association with histone H3 and H4 modifications using ChIP-on-chip. Here, we have generated a broad integrative epigenomic map of twelve histone modifications during the P. falciparum IDC including H4K5ac, H4K8ac, H4K12ac, H4K16ac, H3K9ac, H3K14ac, H3K56ac, H4K20me1, H4K20me3, H3K4me3, H3K79me3 and H4R3me2. While some modifications were found to be associated with the vast majority of the genome and their occupancy was constant, others showed more specific and highly dynamic distribution. Importantly, eight modifications displaying tight correlations with transcript levels showed differential affinity to distinct genomic regions with H4K8ac occupying predominantly promoter regions while others occurred at the 5' ends of coding sequences. The promoter occupancy of H4K8ac remained unchanged when ectopically inserted at a different locus, indicating the presence of specific DNA elements that recruit histone modifying enzymes regardless of their broad chromatin environment. In addition, we showed the presence of multivalent domains on the genome carrying more than one histone mark, highlighting the importance of combinatorial effects on transcription. Overall, our work portrays a substantial association between chromosomal locations of various epigenetic markers, transcriptional activity and global stage-specific transitions in the epigenome.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Life Cycle Stages/physiology , Plasmodium falciparum/genetics , Animals , Genome, Protozoan , Histones/genetics , Real-Time Polymerase Chain Reaction , Time Factors , Transcription, Genetic
13.
Proc Natl Acad Sci U S A ; 109(26): E1772-81, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22619330

ABSTRACT

Cerebral malaria is the most deadly manifestation of infection with Plasmodium falciparum. The pathology of cerebral malaria is characterized by the accumulation of infected erythrocytes (IEs) in the microvasculature of the brain caused by parasite adhesins on the surface of IEs binding to human receptors on microvascular endothelial cells. The parasite and host molecules involved in this interaction are unknown. We selected three P. falciparum strains (HB3, 3D7, and IT/FCR3) for binding to a human brain endothelial cell line (HBEC-5i). The whole transcriptome of isogenic pairs of selected and unselected parasites was analyzed using a variant surface antigen-supplemented microarray chip. After selection, the most highly and consistently up-regulated genes were a subset of group A-like var genes (HB3var3, 3D7_PFD0020c, ITvar7, and ITvar19) that showed 11- to >100-fold increased transcription levels. These var genes encode P. falciparum erythrocyte membrane protein (PfEMP)1 variants with distinct N-terminal domain types (domain cassette 8 or domain cassette 13). Antibodies to HB3var3 and PFD0020c recognized the surface of live IEs and blocked binding to HBEC-5i, thereby confirming the adhesive function of these variants. The clinical in vivo relevance of the HBEC-selected parasites was supported by significantly higher surface recognition of HBEC-selected parasites compared with unselected parasites by antibodies from young African children suffering cerebral malaria (Mann-Whitney test, P = 0.029) but not by antibodies from controls with uncomplicated malaria (Mann-Whitney test, P = 0.58). This work describes a binding phenotype for virulence-associated group A P. falciparum erythrocyte membrane protein 1 variants and identifies targets for interventions to treat or prevent cerebral malaria.


Subject(s)
Brain/blood supply , Endothelium, Vascular/parasitology , Plasmodium falciparum/genetics , Plasmodium/genetics , Protozoan Proteins/genetics , Animals , Brain/parasitology , Humans , Ligands , Transcription, Genetic , Transcriptome , Up-Regulation
14.
BMC Genomics ; 15: 959, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25373614

ABSTRACT

BACKGROUND: Over the course of its intraerythrocytic developmental cycle (IDC), the malaria parasite Plasmodium falciparum tightly orchestrates the rise and fall of transcript levels for hundreds of genes. Considerable debate has focused on the relative importance of transcriptional versus post-transcriptional processes in the regulation of transcript levels. Enzymatically active forms of RNAPII in other organisms have been associated with phosphorylation on the serines at positions 2 and 5 of the heptad repeats within the C-terminal domain (CTD) of RNAPII. We reasoned that insight into the contribution of transcriptional mechanisms to gene expression in P. falciparum could be obtained by comparing the presence of enzymatically active forms of RNAPII at multiple genes with the abundance of their associated transcripts. RESULTS: We exploited the phosphorylation state of the CTD to detect enzymatically active forms of RNAPII at most P. falciparum genes across the IDC. We raised highly specific monoclonal antibodies against three forms of the parasite CTD, namely unphosphorylated, Ser5-P and Ser2/5-P, and used these in ChIP-on-chip type experiments to map the genome-wide occupancy of RNAPII. Our data reveal that the IDC is divided into early and late phases of RNAPII occupancy evident from simple bi-phasic RNAPII binding profiles. By comparison to mRNA abundance, we identified sub-sets of genes with high occupancy by enzymatically active forms of RNAPII and relatively low transcript levels and vice versa. We further show that the presence of active and repressive histone modifications correlates with RNAPII occupancy over the IDC. CONCLUSIONS: The simple early/late occupancy by RNAPII cannot account for the complex dynamics of mRNA accumulation over the IDC, suggesting a major role for mechanisms acting downstream of RNAPII occupancy in the control of gene expression in this parasite.


Subject(s)
Genome, Protozoan , Malaria, Falciparum/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , RNA Polymerase II/metabolism , Antibodies, Monoclonal/pharmacology , Binding Sites/genetics , Chromatin Immunoprecipitation , Cluster Analysis , Computational Biology , Erythrocytes/parasitology , Gene Dosage , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Annotation , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Protein Subunits/antagonists & inhibitors , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/chemistry , RNA, Messenger/genetics , Transcription, Genetic , Transcriptional Activation
15.
Mol Microbiol ; 87(6): 1167-82, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23373537

ABSTRACT

Histone variants are important components of eukaryotic chromatin and can alter chromatin structure to confer specialized functions. H2B variant histones are rare in nature but have evolved independently in the phyla Apicomplexa and Trypanasomatida. Here, we investigate the apicomplexan-specific Plasmodium falciparum histone variant Pf H2B.Z and show that within nucleosomes Pf H2B.Z dimerizes with the H2A variant Pf H2A.Z and that Pf H2B.Z and Pf H2A.Z occupancy correlates in the subset of genes examined. These double-variant nucleosomes also carry common markers of euchromatin like H3K4me3 and histone acetylation. Pf H2B.Z levels are elevated in intergenic regions across the genome, except in the var multigene family, where Pf H2A.Z/Pf H2B.Z double-variant nucleosomes are only enriched in the promoter of the single active var copy and this enrichment is developmentally regulated. Importantly, this pattern seems to be specific for var genes and does not apply to other heterochromatic gene families involved in red blood cell invasion which are also subject to clonal expression. Thus, Pf H2A.Z/Pf H2B.Z double-variant nucleosomes appear to have a highly specific function in the regulation of P. falciparum virulence.


Subject(s)
DNA, Intergenic , Histones/metabolism , Nucleosomes/metabolism , Promoter Regions, Genetic , Protozoan Proteins/biosynthesis , Gene Expression Regulation , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Binding , Protein Multimerization , Virulence
16.
Mol Cell Proteomics ; 11(2): M111.010645, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22023809

ABSTRACT

Differential expression of ligands in the human malaria parasite Plasmodium falciparum enables it to recognize different receptors on the erythrocyte surface, thereby providing alternative invasion pathways. Switching of invasion from using sialated to nonsialated erythrocyte receptors has been linked to the transcriptional activation of a single parasite ligand. We have used quantitative proteomics to show that in addition to this single known change, there are a significant number of changes in the expression of merozoite proteins that are regulated independent of transcription during invasion pathway switching. These results demonstrate a so far unrecognized mechanism by which the malaria parasite is able to adapt to variations in the host cell environment by post-transcriptional regulation.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Merozoites/metabolism , Merozoites/parasitology , Plasmodium falciparum/pathogenicity , Proteomics , Protozoan Proteins/metabolism , Biomarkers/metabolism , Blotting, Western , Cells, Cultured , Erythrocytes/metabolism , Gene Expression Profiling , Humans , L-Lactate Dehydrogenase/metabolism , Malaria, Falciparum/genetics , Malaria, Falciparum/metabolism , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/metabolism , Oligonucleotide Array Sequence Analysis , Peptide Fragments/analysis , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , RNA Processing, Post-Transcriptional , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Life Sci Alliance ; 7(3)2024 Mar.
Article in English | MEDLINE | ID: mdl-38158220

ABSTRACT

The malaria parasite Plasmodium vivax remains a major global public health challenge, and no vaccine is approved for use in humans. Here, we assessed whether P. vivax strain-transcendent immunity can be achieved by repeated infection in Aotus monkeys. Sterile immunity was achieved after two homologous infections, whereas subsequent heterologous challenge provided only partial protection. IgG levels based on P. vivax lysate ELISA and protein microarray increased with repeated infections and correlated with the level of homologous protection. Parasite transcriptional profiles provided no evidence of major antigenic switching upon homologous or heterologous challenge. However, we observed significant sequence diversity and transcriptional differences in the P. vivax core gene repertoire between the two strains used in the study, suggesting that partial protection upon heterologous challenge is due to molecular differences between strains rather than immune evasion by antigenic switching. Our study demonstrates that sterile immunity against P. vivax can be achieved by repeated homologous blood stage infection in Aotus monkeys, thus providing a benchmark to test the efficacy of candidate blood stage P. vivax malaria vaccines.


Subject(s)
Malaria Vaccines , Malaria, Vivax , Malaria , Animals , Humans , Malaria, Vivax/prevention & control , Malaria, Vivax/parasitology , Aotidae , Haplorhini
18.
Cell Chem Biol ; 31(4): 743-759.e8, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38593807

ABSTRACT

Identification of new druggable protein targets remains the key challenge in the current antimalarial development efforts. Here we used mass-spectrometry-based cellular thermal shift assay (MS-CETSA) to identify potential targets of several antimalarials and drug candidates. We found that falcilysin (FLN) is a common binding partner for several drug candidates such as MK-4815, MMV000848, and MMV665806 but also interacts with quinoline drugs such as chloroquine and mefloquine. Enzymatic assays showed that these compounds can inhibit FLN proteolytic activity. Their interaction with FLN was explored systematically by isothermal titration calorimetry and X-ray crystallography, revealing a shared hydrophobic pocket in the catalytic chamber of the enzyme. Characterization of transgenic cell lines with lowered FLN expression demonstrated statistically significant increases in susceptibility toward MK-4815, MMV000848, and several quinolines. Importantly, the hydrophobic pocket of FLN appears amenable to inhibition and the structures reported here can guide the development of novel drugs against malaria.


Subject(s)
Antimalarials , Malaria , Methylamines , Quinolines , Humans , Antimalarials/chemistry , Malaria/drug therapy , Phenols/therapeutic use , Quinolines/pharmacology , Quinolines/metabolism , Drug Development
19.
Mol Microbiol ; 84(2): 243-59, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22435676

ABSTRACT

The Plasmodium falciparum genome is equipped with several subtelomeric gene families that are implicated in parasite virulence and immune evasion. Members of these families are uniformly positioned within heterochromatic domains and are thus subject to variegated expression. The best-studied example is that of the var family encoding the major parasite virulence factor P. falciparum erythrocyte membrane protein 1 (PfEMP1). PfEMP1 undergoes antigenic variation through switches in mutually exclusive var gene transcription. var promoters function as crucial regulatory elements in the underlying epigenetic control strategy. Here, we analysed promoters of upsA, upsB and upsC var, rifA1-type rif, stevor, phist and pfmc-2tm genes and investigated their role in endogenous gene transcription by comparative genome-wide expression profiling of transgenic parasite lines. We find that the three major var promoter types are functionally equal and play an essential role in singular gene choice. Unlike var promoters, promoters of non-var families are not silenced by default, and transcription of non-var families is not subject to the same mode of mutually exclusive transcription as has been observed for var genes. Our findings identified a differential logic in the regulation of var and other subtelomeric virulence gene families, which will have important implications for our understanding and future analyses of phenotypic variation in malaria parasites.


Subject(s)
Gene Expression Regulation , Genes, Protozoan , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Virulence Factors/biosynthesis , Virulence Factors/genetics , Gene Expression Profiling , Promoter Regions, Genetic , Transcription, Genetic
20.
Mol Cell Proteomics ; 10(8): M110.006411, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21558492

ABSTRACT

Studies of the Plasmodium falciparum transcriptome have shown that the tightly controlled progression of the parasite through the intra-erythrocytic developmental cycle (IDC) is accompanied by a continuous gene expression cascade in which most expressed genes exhibit a single transcriptional peak. Because the biochemical and cellular functions of most genes are mediated by the encoded proteins, understanding the relationship between mRNA and protein levels is crucial for inferring biological activity from transcriptional gene expression data. Although studies on other organisms show that <50% of protein abundance variation may be attributable to corresponding mRNA levels, the situation in Plasmodium is further complicated by the dynamic nature of the cyclic gene expression cascade. In this study, we simultaneously determined mRNA and protein abundance profiles for P. falciparum parasites during the IDC at 2-hour resolution based on oligonucleotide microarrays and two-dimensional differential gel electrophoresis protein gels. We find that most proteins are represented by more than one isoform, presumably because of post-translational modifications. Like transcripts, most proteins exhibit cyclic abundance profiles with one peak during the IDC, whereas the presence of functionally related proteins is highly correlated. In contrast, the abundance of most parasite proteins peaks significantly later (median 11 h) than the corresponding transcripts and often decreases slowly in the second half of the IDC. Computational modeling indicates that the considerable and varied incongruence between transcript and protein abundance may largely be caused by the dynamics of translation and protein degradation. Furthermore, we present cyclic abundance profiles also for parasite-associated human proteins and confirm the presence of five human proteins with a potential role in antioxidant defense within the parasites. Together, our data provide fundamental insights into transcript-protein relationships in P. falciparum that are important for the correct interpretation of transcriptional data and that may facilitate the improvement and development of malaria diagnostics and drug therapy.


Subject(s)
Erythrocytes/parasitology , Host-Parasite Interactions , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Aryldialkylphosphatase/genetics , Aryldialkylphosphatase/metabolism , Catalase/genetics , Catalase/metabolism , Cell Culture Techniques , Erythrocytes/enzymology , Erythrocytes/metabolism , Humans , Lactoferrin/genetics , Lactoferrin/metabolism , Malaria, Falciparum/parasitology , Models, Biological , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proteome/genetics , Proteome/metabolism , Protozoan Proteins/genetics , Spores, Protozoan/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL