Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Nucleic Acids Res ; 52(10): 5624-5642, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38554111

ABSTRACT

Gametocyte development of the Plasmodium parasite is a key step for transmission of the parasite. Male and female gametocytes are produced from a subpopulation of asexual blood-stage parasites, but the mechanisms that regulate the differentiation of sexual stages are still under investigation. In this study, we investigated the role of PbARID, a putative subunit of a SWI/SNF chromatin remodeling complex, in transcriptional regulation during the gametocyte development of P. berghei. PbARID expression starts in early gametocytes before the manifestation of male and female-specific features, and disruption of its gene results in the complete loss of gametocytes with detectable male features and the production of abnormal female gametocytes. ChIP-seq analysis of PbARID showed that it forms a complex with gSNF2, an ATPase subunit of the SWI/SNF chromatin remodeling complex, associating with the male cis-regulatory element, TGTCT. Further ChIP-seq of PbARID in gsnf2-knockout parasites revealed an association of PbARID with another cis-regulatory element, TGCACA. RIME and DNA-binding assays suggested that HDP1 is the transcription factor that recruits PbARID to the TGCACA motif. Our results indicated that PbARID could function in two chromatin remodeling events and paly essential roles in both male and female gametocyte development.


Subject(s)
Chromatin Assembly and Disassembly , Plasmodium berghei , Protozoan Proteins , Transcription Factors , Animals , Female , Male , Mice , Chromatin Assembly and Disassembly/genetics , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Genotype , Sequence Analysis, RNA , Chromatin/genetics , Chromatin/metabolism , Amino Acid Sequence , Sequence Analysis, Protein , Phylogeny , Transcriptome , Genome, Protozoan
2.
Proc Natl Acad Sci U S A ; 120(20): e2303432120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155862

ABSTRACT

Plasmodium parasites, the causative agents of malaria, possess a complex lifecycle; however, the mechanisms of gene regulation involved in the cell-type changes remain unknown. Here, we report that gametocyte sucrose nonfermentable 2 (gSNF2), an SNF2-like chromatin remodeling ATPase, plays an essential role in the differentiation of male gametocytes. Upon disruption of gSNF2, male gametocytes lost the capacity to develop into gametes. ChIP-seq analyses revealed that gSNF2 is widely recruited upstream of male-specific genes through a five-base, male-specific cis-acting element. In gSNF2-disrupted parasites, expression of over a hundred target genes was significantly decreased. ATAC-seq analysis demonstrated that decreased expression of these genes correlated with a decrease of the nucleosome-free region upstream of these genes. These results suggest that global changes induced in the chromatin landscape by gSNF2 are the initial step in male differentiation from early gametocytes. This study provides the possibility that chromatin remodeling is responsible for cell-type changes in the Plasmodium lifecycle.


Subject(s)
Malaria , Plasmodium , Male , Humans , Chromatin/genetics , Chromatin/metabolism , Plasmodium/genetics , Malaria/parasitology , Gene Expression Regulation , Cell Differentiation/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism
3.
PLoS Pathog ; 19(2): e1010890, 2023 02.
Article in English | MEDLINE | ID: mdl-36780562

ABSTRACT

Gametocyte development is a critical step in the life cycle of Plasmodium. Despite the number of studies on gametocyte development that have been conducted, the molecular mechanisms regulating this process remain to be fully understood. This study investigates the functional roles of two female-specific transcriptional regulators, PbAP2-FG2 and PbAP2R-2, in P. berghei. Knockout of pbap2-fg2 or pbap2r-2 impairs female gametocyte development, resulting in developmental arrest during ookinete development. ChIP-seq analyses of these two factors indicated their colocalization on the genome, suggesting that they function as a complex. These analyses also revealed that their target genes contained a variety of genes, including both male and female-enriched genes. Moreover, differential expression analyses showed that these target genes were upregulated through the disruption of pbap2-fg2 or pbap2r-2, indicating that these two factors function as a transcriptional repressor complex in female gametocytes. Formation of a complex between PbAP2-FG2 and PbAP2R-2 was confirmed by RIME, a method that combines ChIP and MS analysis. In addition, the analysis identified a chromatin regulator PbMORC as an interaction partner of PbAP2-FG2. Comparative target analysis between PbAP2-FG2 and PbAP2-G demonstrated a significant overlap between their target genes, suggesting that repression of early gametocyte genes activated by PbAP2-G is one of the key roles for this female transcriptional repressor complex. Our results indicate that the PbAP2-FG2-PbAP2R-2 complex-mediated repression of the target genes supports the female differentiation from early gametocytes.


Subject(s)
Plasmodium berghei , Protozoan Proteins , Plasmodium berghei/physiology , Protozoan Proteins/metabolism
4.
PLoS Pathog ; 18(8): e1010510, 2022 08.
Article in English | MEDLINE | ID: mdl-35947628

ABSTRACT

The sexual phase of Plasmodium represents a crucial step in malaria transmission, during which these parasites fertilize and form ookinetes to infect mosquitoes. Plasmodium development after fertilization is thought to proceed with female-stored mRNAs until the formation of a retort-form ookinete; thus, transcriptional activity in zygotes has previously been considered quiescent. In this study, we reveal the essential role of transcriptional activity in zygotes by investigating the function of a newly identified AP2 transcription factor, AP2-Z, in P. berghei. ap2-z was previously reported as a female transcriptional regulator gene whose disruption resulted in developmental arrest at the retort stage of ookinetes. In this study, although ap2-z was transcribed in females, we show that it was translationally repressed by the DOZI complex and translated after fertilization with peak expression at the zygote stage. ChIP-seq analysis of AP2-Z shows that it binds on specific DNA motifs, targeting the majority of genes known as an essential component of ookinetes, which largely overlap with the AP2-O targets, as well as genes that are unique among the targets of other sexual transcription factors. The results of this study also indicate the existence of a cascade of transcription factors, beginning with AP2-G, that proceeds from gametocytogenesis to ookinete formation.


Subject(s)
Malaria , Plasmodium berghei , Animals , Female , Malaria/genetics , Malaria/parasitology , Plasmodium berghei/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zygote/metabolism
6.
Nature ; 552(7683): 101-105, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29186116

ABSTRACT

Malaria is among the most serious infectious diseases affecting humans, accounting for approximately half a million deaths each year. Plasmodium falciparum causes most life-threatening cases of malaria. Acquired immunity to malaria is inefficient, even after repeated exposure to P. falciparum, but the immune regulatory mechanisms used by P. falciparum remain largely unknown. Here we show that P. falciparum uses immune inhibitory receptors to achieve immune evasion. RIFIN proteins are products of a polymorphic multigene family comprising approximately 150-200 genes per parasite genome that are expressed on the surface of infected erythrocytes. We found that a subset of RIFINs binds to either leucocyte immunoglobulin-like receptor B1 (LILRB1) or leucocyte-associated immunoglobulin-like receptor 1 (LAIR1). LILRB1-binding RIFINs inhibit activation of LILRB1-expressing B cells and natural killer (NK) cells. Furthermore, P. falciparum-infected erythrocytes isolated from patients with severe malaria were more likely to interact with LILRB1 than erythrocytes from patients with non-severe malaria, although an extended study with larger sample sizes is required to confirm this finding. Our results suggest that P. falciparum has acquired multiple RIFINs to evade the host immune system by targeting immune inhibitory receptors.


Subject(s)
Immune Evasion/immunology , Leukocyte Immunoglobulin-like Receptor B1/immunology , Membrane Proteins/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Receptors, Immunologic/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CHO Cells , Cricetulus , Erythrocytes/immunology , Erythrocytes/parasitology , HEK293 Cells , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Leukocyte Immunoglobulin-like Receptor B1/chemistry , Ligands , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Receptors, Immunologic/chemistry , Sample Size
7.
Arch Virol ; 167(1): 123-130, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34757503

ABSTRACT

Ticks are blood-sucking arthropods that transmit many pathogens, including arboviruses. Arboviruses transmitted by ticks are generally referred to as tick-borne viruses (TBVs). TBVs are known to cause diseases in humans, pets, and livestock. There is, however, very limited information on the occurrence and distribution of TBVs in sub-Saharan Africa. This study was designed to determine the presence and distribution of ticks infesting dogs and cattle in Ghana, as well as to identify the tick-borne or tick-associated viruses they harbour. A more diverse population of ticks was found to infest cattle (three genera) relative to those infesting dogs (one genus). Six phleboviruses and an orthonairovirus were detected in tick pools screened by RT-PCR. Subsequent sequence analysis revealed two distinct phleboviruses and the previously reported Odaw virus in ticks collected from dogs and a virus (16GH-T27) most closely related to four unclassified phleboviruses in ticks collected from cattle. The virus 16GH-T27 was considered a strain of Balambala tick virus (BTV) and named BTV strain 16GH-T27. Next-generation sequencing analysis of the BTV-positive tick pool detected only the L and S segments. Phylogenetic analysis revealed that BTV clustered with viruses previously defined as M-segment-deficient phleboviruses. The orthonairovirus detected in ticks collected from cattle was confirmed to be the medically important Dugbe virus. Furthermore, we discuss the importance of understanding the presence and distribution of ticks and TBVs in disease prevention and mitigation and the implications for public health. Our findings contribute to the knowledge pool on TBVs and tick-associated viruses.


Subject(s)
Phlebovirus , Tick-Borne Diseases , Ticks , Animals , Cattle , Dogs , Ghana/epidemiology , Phylogeny , Satellite Viruses , Tick-Borne Diseases/epidemiology , Tick-Borne Diseases/veterinary
8.
Mol Microbiol ; 113(1): 40-51, 2020 01.
Article in English | MEDLINE | ID: mdl-31231888

ABSTRACT

The malaria gametocyte, the gamete precursor, is the essential stage for malaria transmission to the mosquito vector. In the vertebrate host's blood, it develops into a mature male or female capable of transforming into a gamete in the mosquito blood meal. Despite the importance of this stage in the malaria life cycle, the genetic regulation of gametocyte development is poorly understood. In particular, transcription factors involved in sex-specific gene expression have not been identified. In this paper, we report that an AP2-family transcription factor, AP2-FG, is responsible for female-specific gene regulation. AP2-FG expression in Plasmodium berghei was observed exclusively in female gametocytes, in the beginning of 4-6 h before sexual dimorphism manifests in developing gametocytes. AP2-FG disruption resulted in the arrest of female maturation, but did not affect the development of males. Chromatin immunoprecipitation sequencing analysis suggested that AP2-FG directly regulates over 700 genes. Its targets include genes for female gametocyte-specific functions, such as gametogenesis, fertilization and zygote development. AP2-FG binding to target gene promoters was associated with a 10 bp sequence motif. These results indicate that AP2-FG plays a role in the differentiation of early gametocytes to mature females by governing a female-specific gene expression repertoire.


Subject(s)
Gametogenesis , Malaria/parasitology , Plasmodium berghei/genetics , Protozoan Proteins/metabolism , Transcription Factors/metabolism , Animals , Female , Gene Expression Regulation , Germ Cells/cytology , Life Cycle Stages , Male , Mice, Inbred BALB C , Plasmodium berghei/metabolism
9.
Biochem Biophys Res Commun ; 548: 167-173, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33647792

ABSTRACT

Plasmodium falciparum causes the most severe form of malaria. Acquired immunity against P. falciparum provides insufficient protection even after repeated infections. Therefore, P. falciparum parasites might exploit inhibitory receptors for immune evasion. P. falciparum RIFINs are products of a multigene family consisting of 150-200 genes. Previously, we demonstrated that some RIFINs downregulate the immune response through the leukocyte immunoglobulin-like receptor (LILR) family inhibitory receptor, LILRB1, and leukocyte-associated immunoglobulin-like receptor 1, LAIR1. In this study, we further analyzed the expression of inhibitory receptor ligands on P. falciparum-infected erythrocytes and found that P. falciparum-infected erythrocytes expressed ligands for another LILR family inhibitory receptor, LILRB2, that recognizes HLA class I molecules as a host ligand. Furthermore, we identified that a specific RIFIN was a ligand for LILRB2 by using a newly developed RIFIN expression library. In addition, the domain 3 of LILRB2 was involved in RIFIN binding, whereas the domains 1 and 2 of LILRB2 were involved in the binding to HLA class I molecules. These results suggest that inhibitory receptor LILRB2 is also targeted by RIFIN for immune evasion of P. falciparum similar to LILRB1 and LAIR1.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Receptors, Immunologic/metabolism , Animals , Erythrocytes/parasitology , Female , HEK293 Cells , Humans , Ligands , Malaria, Falciparum/parasitology , Membrane Glycoproteins/chemistry , Mice, Inbred BALB C , Protein Binding , Protein Domains , Receptors, Immunologic/chemistry
10.
Exp Parasitol ; 201: 67-77, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30904694

ABSTRACT

Recent evidence suggests that 1α,25-dihydroxyvitamin D3 (VD3), the active form of vitamin D, inhibits microbial proliferation. Previously, we used in vivo murine models to investigate the antimalarial activity of VD3 and confirmed potent antimalarial activity in the acute phase. This study aimed to clarify the mechanisms underlying the antimalarial activity of VD3 in vivo, particularly extensive inhibition of parasitemia in the acute phase, focusing on nitric oxide (NO), a potent antimalarial molecule. VD3 is a good NO inducer. When most Plasmodium chabaudi AS (PcAS)-infected mice treated with VD3 survived, NO was present in blood samples obtained from VD3-treated mice at a significantly higher rate at 2 and/or 3 days post-infection than that in vehicle-treated control mice. To verify the involvement of NO in the antimalarial activity of VD3, we used aminoguanidine (AG), an inducible NO synthase (iNOS) inhibitor, to abrogate the antimalarial activity of VD3. However, despite AG-induced reductions in NO levels, parasitemia remained inhibited during the acute phase, even in the presence of AG, and the antiplasmodial faculty of VD3 was not ablated. VD3-mediated antimalarial activity irrelevant of NO compelled us to consider another candidate. In a pilot experiment, we used cathelicidin (CAMP), an antimicrobial peptide, since it is known that VD3 induces CAMP synthesis. Serum CAMP levels increased on days 4 or 5 post-infection with or without VD3 administration, but experiments using exogenous CAMP did not display curative effects in PcAS-infected mice. The present study using VD3 to target the malarial parasite thus suggests a potential novel approach to treat malarial infections.


Subject(s)
Antimalarials/pharmacology , Cholecalciferol/pharmacology , Malaria/drug therapy , Plasmodium chabaudi/drug effects , Vitamin D/analogs & derivatives , Animals , Antimalarials/therapeutic use , Antimicrobial Cationic Peptides/biosynthesis , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/therapeutic use , Cholecalciferol/therapeutic use , Female , Guanidines/pharmacology , Malaria/mortality , Malaria/parasitology , Mice , Mice, Inbred BALB C , Nitric Oxide/biosynthesis , Nitric Oxide/pharmacology , Nitric Oxide/therapeutic use , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitrous Oxide/blood , Nitrous Oxide/metabolism , Parasitemia/drug therapy , Parasitemia/parasitology , Vitamin D/pharmacology , Vitamin D/therapeutic use , Cathelicidins
11.
Proc Natl Acad Sci U S A ; 112(41): 12824-9, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26417110

ABSTRACT

Gametocytes are nonreplicative sexual forms that mediate malaria transmission to a mosquito vector. They are generated from asexual blood-stage parasites that proliferate in the circulation. However, little is known about how this transition is genetically regulated. Here, we report that an Apetala2 (AP2) family transcription factor, AP2-G2, regulates this transition as a transcriptional repressor. Disruption of AP2-G2 in the rodent malaria parasite Plasmodium berghei did not prevent commitment to the sexual stage but did halt development before the appearance of sex-specific morphologies. ChIP-seq analysis revealed that AP2-G2 targeted ∼1,500 genes and recognized a five-base motif in their promoters. Most of these target genes are required for asexual proliferation of the parasites in the blood, suggesting that AP2-G2 blocks the program that precedes asexual replication to promote conversion to the sexual stage. Microarray analysis showed that the identified targets constituted ∼70% of the up-regulated genes in AP2-G2-depleted parasites, suggesting that AP2-G2 actually functions as a repressor in gametocytes. A promoter assay using a centromere plasmid demonstrated that the binding motif functions as a cis-acting negative regulatory element. These results suggest that global transcriptional repression, which occurs during the initial phase of gametocytogenesis, is an essential step in Plasmodium sexual development.


Subject(s)
Life Cycle Stages/physiology , Malaria/metabolism , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Response Elements/physiology , Transcription Factors/metabolism , Animals , Female , Malaria/genetics , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Transcription Factors/genetics
12.
J Gen Virol ; 98(11): 2876-2881, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29048274

ABSTRACT

During an entomological surveillance for arthropod-borne viruses in the Philippines, we isolated a previously unrecognized virus from female Armigeres spp. mosquitoes. Whole-genome sequencing, genetic characterization and phylogenetic analysis revealed that the isolated virus, designated Armigeres iflavirus (ArIFV), is a novel member of the iflaviruses (genus Iflavirus, family Iflaviridae) and phylogenetically related to Moku virus, Hubei odonate virus 4, slow bee paralysis virus and Graminella nigrifrons virus 1. To our knowledge, this is the first successful isolation of iflavirus from a dipteran insect. Spherical ArIFV particles of approximately 30 nm in diameter contained at least three major structural proteins. ArIFV multiplied to high titres (~109 p.f.u. ml-1) and formed clear plaques in a mosquito cell line, C6/36. Our findings provide new insights into the infection mechanism, genetic diversity and evolution of the Iflaviridae family.


Subject(s)
Culicidae/virology , Insect Viruses/classification , Insect Viruses/isolation & purification , RNA Viruses/classification , RNA Viruses/isolation & purification , Animals , Cell Line , Philippines , Viral Plaque Assay , Viral Structural Proteins/analysis , Virion/chemistry , Virion/ultrastructure
13.
PLoS Pathog ; 11(5): e1004905, 2015 May.
Article in English | MEDLINE | ID: mdl-26018192

ABSTRACT

Stage-specific transcription is a fundamental biological process in the life cycle of the Plasmodium parasite. Proteins containing the AP2 DNA-binding domain are responsible for stage-specific transcriptional regulation and belong to the only known family of transcription factors in Plasmodium parasites. Comprehensive identification of their target genes will advance our understanding of the molecular basis of stage-specific transcriptional regulation and stage-specific parasite development. AP2-O is an AP2 family transcription factor that is expressed in the mosquito midgut-invading stage, called the ookinete, and is essential for normal morphogenesis of this stage. In this study, we identified the genome-wide target genes of AP2-O by chromatin immunoprecipitation-sequencing and elucidate how this AP2 family transcription factor contributes to the formation of this motile stage. The analysis revealed that AP2-O binds specifically to the upstream genomic regions of more than 500 genes, suggesting that approximately 10% of the parasite genome is directly regulated by AP2-O. These genes are involved in distinct biological processes such as morphogenesis, locomotion, midgut penetration, protection against mosquito immunity and preparation for subsequent oocyst development. This direct and global regulation by AP2-O provides a model for gene regulation in Plasmodium parasites and may explain how these parasites manage to control their complex life cycle using a small number of sequence-specific AP2 transcription factors.


Subject(s)
Gene Expression Regulation , Genome, Protozoan , Malaria/genetics , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Transcription Factor AP-2/genetics , Amino Acid Sequence , Animals , Chromatin Immunoprecipitation , Female , High-Throughput Nucleotide Sequencing , Life Cycle Stages , Malaria/parasitology , Mice , Mice, Inbred BALB C , Microscopy, Immunoelectron , Molecular Sequence Data , Oocysts/growth & development , Oocysts/metabolism , Oocysts/parasitology , Plasmodium berghei/growth & development , Plasmodium berghei/isolation & purification , Protozoan Proteins/metabolism , RNA, Protozoan , Sequence Homology, Amino Acid , Transcription Factor AP-2/metabolism
14.
Malar J ; 15: 201, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068454

ABSTRACT

BACKGROUND: Plasmodium circumsporozoite protein (CSP) is a major surface antigen present in the sporozoite (Spz) stage of a malaria parasite. RTS, S vaccine, the most clinically advanced malaria vaccine, consists of a large portion of Plasmodium falciparum CSP (PfCSP). A highly infectious, recombinant rodent malaria, Plasmodium yoelii parasite bearing a full-length PfCSP, PfCSP/Py Spz, was needed as a tool to evaluate the role of PfCSP in mediating, protective, anti-malaria immunity in a mouse model. METHODS: A transgenic parasite, PfCSP/Py Spz, was generated by inserting a construct expressing the PfCSP at the locus of the P. yoelii CSP gene by double cross-over homologous recombination. Then the biological and protective properties of PfCSP/Py Spz were determined. RESULTS: This PfCSP/Py parasite produced up to 30,000 Spz in mosquito salivary glands, which is equal or even higher than the number of Spz produced by wild-type P. yoelii parasites. Five bites of PfCSP/Py-infected mosquitoes could induce blood infection in BALB/c mice. CONCLUSIONS: The current study has demonstrated a successful establishment of a transgenic P. yoelii parasite clone that is able to express a full-length PfCSP, PfCSP/Py parasite. Importantly, this PfCSP/Py parasite can be as infectious as the wild-type P. yoelii parasite both in mosquito vector and in mouse, a mammalian host. A new transgenic parasite that expresses a full-length PfCSP may become a useful tool for researchers to investigate immunity against PfCSP in a mouse model.


Subject(s)
Culicidae/parasitology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Female , Mice , Mice, Inbred BALB C , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/immunology , Plasmodium falciparum/genetics , Salivary Glands/parasitology , T-Lymphocytes/parasitology , Vaccines, Synthetic/immunology
15.
Genome Res ; 22(5): 985-92, 2012 May.
Article in English | MEDLINE | ID: mdl-22426943

ABSTRACT

The global spread of drug-resistant parasites is a serious problem for the treatment of malaria. Although identifying drug-resistance genes is crucial for the efforts against resistant parasites, an effective approach has not yet been developed. Here, we report a robust method for identifying resistance genes from parasites by using a Plasmodium artificial chromosome (PAC). Large genomic DNA fragments (10-50 kb) from the drug-resistant rodent malaria parasite Plasmodium berghei were ligated into the PAC and directly introduced into the drug-sensitive (i.e., wild-type) parasite by electroporation, resulting in a PAC library that encompassed the whole genomic sequence of the parasite. Subsequently, the transformed parasites that acquired resistance were selected by screening with the drug, and the resistance gene in the PAC was successfully identified. Furthermore, the drug-resistance gene was identified from a PAC library that was made from the pyrimethamine-resistant parasite Plasmodium chabaudi, further demonstrating the utility of our method. This method will promote the identification of resistance genes and contribute to the global fight against drug-resistant parasites.


Subject(s)
Drug Resistance/genetics , Genes, Protozoan , Plasmodium berghei/genetics , Plasmodium chabaudi/genetics , Animals , Antimalarials/pharmacology , Chromosomes, Artificial/genetics , Cloning, Molecular , Gene Library , Genetic Association Studies , Genome, Protozoan , Malaria/drug therapy , Malaria/parasitology , Plasmodium berghei/drug effects , Plasmodium chabaudi/drug effects , Pyrimethamine/pharmacology , Rats , Rats, Wistar , Transfection
16.
Mol Microbiol ; 87(1): 66-79, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23216750

ABSTRACT

The liver stage is the first stage of the malaria parasite that replicates in the vertebrate host. However, little is known about the interplay between the parasite liver stage and its host cell, the hepatocyte. In this study, we identified an exported protein that has a critical role in parasite development in host hepatocytes. Expressed sequence tag analysis of Plasmodium berghei liver-stage parasites indicated that transcripts encoding a protein with an N-terminal signal peptide, designated liver-specific protein 2 (LISP2), are highly expressed in this stage. Expression of LISP2 was first observed 24 h after infection and rapidly increased during the liver-stage schizogony. Immunofluorescent staining with anti-LSP2 antibodies showed that LISP2 was carried to the parasitophorous vacuole and subsequently transported to the cytoplasm and nucleus of host hepatocytes. Gene targeting experiments demonstrated that majority of the LISP2-mutant liver-stage parasites arrested their development during formation of merozoites. These results indicate that exported LISP2 is involved in parasite-host interactions required for the development of liver-stage parasites inside hepatocytes. This study demonstrated that mid-to-late liver-stage malarial parasites have a system for exporting proteins to the host cell as intraerythrocytic stages do and presumably to use the proteins to modify the host cell and improve the environment.


Subject(s)
Hepatocytes/metabolism , Hepatocytes/parasitology , Merozoites/growth & development , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Cytoplasm/metabolism , Expressed Sequence Tags , Hepatocytes/cytology , Host-Parasite Interactions , Humans , Liver/parasitology , Malaria/parasitology , Merozoites/pathology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Promoter Regions, Genetic , Protein Sorting Signals/genetics , Protein Transport , Protozoan Proteins/genetics
17.
Elife ; 122024 Jan 22.
Article in English | MEDLINE | ID: mdl-38252559

ABSTRACT

Gametocytes play key roles in the Plasmodium lifecycle. They are essential for sexual reproduction as precursors of the gametes. They also play an essential role in parasite transmission to mosquitoes. Elucidation of the gene regulation at this stage is essential for understanding these two processes at the molecular level and for developing new strategies to break the parasite lifecycle. We identified a novel Plasmodium transcription factor (TF), designated as a partner of AP2-FG or PFG. In this article, we report that this TF regulates the gene expression in female gametocytes in concert with another female-specific TF AP2-FG. Upon the disruption of PFG, majority of female-specific genes were significantly downregulated, and female gametocyte lost the ability to produce ookinetes. ChIP-seq analysis showed that it was located in the same position as AP2-FG, indicating that these two TFs form a complex. ChIP-seq analysis of PFG in AP2-FG-disrupted parasites and ChIP-seq analysis of AP2-FG in PFG-disrupted parasites demonstrated that PFG mediates the binding of AP2-FG to a ten-base motif and that AP2-FG binds another motif, GCTCA, in the absence of PFG. In promoter assays, this five-base motif was identified as another female-specific cis-acting element. Genes under the control of the two forms of AP2-FG, with or without PFG, partly overlapped; however, each form had target preferences. These results suggested that combinations of these two forms generate various expression patterns among the extensive genes expressed in female gametocytes.


Subject(s)
Culicidae , Plasmodium , Animals , Female , Transcription Factors/genetics , Plasmodium/genetics , Transcription Factor AP-2 , Biological Assay
18.
Biochem Biophys Res Commun ; 432(3): 504-8, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23402751

ABSTRACT

Protozoan parasites rely on purine nucleosides supplied by the host because they are unable to synthesise purine rings denovo. Nucleoside transporter 1 (NT1) and purine nucleoside phosphorylase (PNP) play an essential role in purine salvage in Plasmodium. It is unclear whether severe pathology, such as cerebral malaria (CM), develops in hosts infected with Plasmodium parasites that lack activity of NT1 or PNP. Plasmodium berghei (Pb) ANKA-infected mice show features similar to human CM, such as cerebral paralysis and cerebral haemorrhage. Therefore, Pb ANKA infection in mice is a good experimental model of CM. In this study, we generated pbnt1-disrupted Pb ANKA (Δpbnt1 parasites) and pbpnp-disrupted Pb ANKA (Δpbpnp parasites), and investigated the effect of pbnt1 or pbpnp disruption on the outcome of infection with Pb ANKA. We showed that the rapid increase of wild-type Pb ANKA (WT parasites) in mice early in infection was significantly inhibited by disruption of pbnt1. Moreover, Δpbnt1 parasite-infected mice showed neither cerebral paralysis nor cerebral haemorrhage, and all mice spontaneously recovered from infection. By contrast, mice infected with Δpbpnp parasites showed features similar to those of mice infected with WT parasites. In this study, we demonstrated that the high virulence of Pb ANKA in the asexual phase is suppressed by disruption of pbnt1 but not pbpnp.


Subject(s)
Malaria, Cerebral/parasitology , Nucleoside Transport Proteins/metabolism , Plasmodium berghei/pathogenicity , Protozoan Proteins/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Animals , Blood-Brain Barrier/parasitology , Disease Models, Animal , Female , Gene Deletion , Humans , Mice , Mice, Inbred C57BL , Nucleoside Transport Proteins/genetics , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Protozoan Proteins/genetics , Purine-Nucleoside Phosphorylase/genetics
19.
Mol Nutr Food Res ; 67(21): e2300185, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37706619

ABSTRACT

SCOPE: Malaria remains one of the most important infectious diseases in the world. Allyl isothiocyanate (AITC) is a main ingredient of traditional spice Wasabia japonica, which is reported to have anti-bacterial and antiparasitic activities. However, there is no information on effects of AITC against malaria. The present study investigates the anti-malarial activity of dietary AITC in vivo and that of AITC metabolites in vitro. METHODS AND RESULTS: The ad libitum administration of 35, 175, or 350 µM AITC-containing drinking water to ICR mice significantly inhibit the parasitemia induced after infection with Plasmodium berghei. On the other hand, after single oral administration of AITC (20 mg kg-1 body weight), N-acetyl-S-(N-allylthiocarbamoyl)-l-cysteine (NAC-AITC) as one of the AITC metabolites displays a serum Cmax of 11.4 µM at a Tmax of 0.5 h, but AITC is not detected at any time point. Moreover, NAC-AITC shows anti-malarial activity against Plasmodium falciparum in vitro, and its 50% inhibitory concentration (IC50 ) against parasitemia is 12.6 µM. CONCLUSIONS: These results indicate that orally administered AITC is metabolized to NAC-AITC and exerts anti-malarial activity against malaria parasites in blood, suggesting that the consumption of AITC-containing food stuffs such as cruciferous plants may prevent malaria.


Subject(s)
Antimalarials , Malaria , Mice , Animals , Antimalarials/pharmacology , Parasitemia/drug therapy , Mice, Inbred ICR , Isothiocyanates/pharmacology , Malaria/drug therapy
20.
mBio ; 14(1): e0251622, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36622145

ABSTRACT

Malaria transmission to humans begins with sporozoite infection of the liver. The elucidation of gene regulation during the sporozoite stage will promote the investigation of mechanisms of liver infection by this parasite and contribute to the development of strategies for preventing malaria transmission. AP2-Sp is a transcription factor (TF) essential for the formation of sporozoites or sporogony, which takes place in oocysts in the midguts of infected mosquitoes. To understand the role of this TF in the transcriptional regulatory system of this stage, we performed chromatin immunoprecipitation sequencing (ChIP-seq) analyses using whole mosquito midguts containing late oocysts as starting material and explored its genome-wide target genes. We identified 697 target genes, comprising those involved in distinct processes parasites experience during this stage, from sporogony to development into the liver stage and representing the majority of genes highly expressed in the sporozoite stage. These results suggest that AP2-Sp determines basal patterns of gene expression by targeting a broad range of genes directly. The ChIP-seq analyses also showed that AP2-Sp maintains its own expression by a transcriptional autoactivation mechanism (positive-feedback loop) and induces all TFs reported to be transcribed at this stage, including AP2-Sp2, AP2-Sp3, and SLARP. The results showed that AP2-Sp exists at the top of the transcriptional cascade of this stage and triggers the formation of this stage as a master regulator. IMPORTANCE The sporozoite stage plays a central role in malaria transmission from a mosquito to vertebrate host and is an important target for antimalarial strategies. AP2-Sp is a candidate master transcription factor for the sporozoite stage. However, study of its role in gene regulation has been hampered because of difficulties in performing genome-wide studies of gene regulation in this stage. Here, we conquered this problem and revealed that AP2-Sp has the following prominent features as a master transcription factor. First, it determines the repertory of gene expression during this stage. Second, it maintains its own expression through a transcriptional positive-feedback loop and induces all other transcription factors specifically expressed in this stage. This study represents a major breakthrough in fully understanding gene regulation in this important malarial stage.


Subject(s)
Malaria , Parasites , Animals , Humans , Sporozoites/physiology , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Malaria/parasitology , Gene Expression Regulation , Oocysts/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Parasites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL