Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 744
Filter
1.
Nat Commun ; 15(1): 4697, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824128

ABSTRACT

Differentiation of male gametocytes into flagellated fertile male gametes relies on the assembly of axoneme, a major component of male development for mosquito transmission of the malaria parasite. RNA-binding protein (RBP)-mediated post-transcriptional regulation of mRNA plays important roles in eukaryotic sexual development, including the development of female Plasmodium. However, the role of RBP in defining the Plasmodium male transcriptome and its function in male gametogenesis remains incompletely understood. Here, we performed genome-wide screening for gender-specific RBPs and identified an undescribed male-specific RBP gene Rbpm1 in the Plasmodium. RBPm1 is localized in the nucleus of male gametocytes. RBPm1-deficient parasites fail to assemble the axoneme for male gametogenesis and thus mosquito transmission. RBPm1 interacts with the spliceosome E complex and regulates the splicing initiation of certain introns in a group of 26 axonemal genes. RBPm1 deficiency results in intron retention and protein loss of these axonemal genes. Intron deletion restores axonemal protein expression and partially rectifies axonemal defects in RBPm1-null gametocytes. Further splicing assays in both reporter and endogenous genes exhibit stringent recognition of the axonemal introns by RBPm1. The splicing activator RBPm1 and its target introns constitute an axonemal intron splicing program in the post-transcriptional regulation essential for Plasmodium male development.


Subject(s)
Axoneme , Introns , Protozoan Proteins , RNA Splicing , RNA-Binding Proteins , Introns/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Animals , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Male , Axoneme/metabolism , Female , Gametogenesis/genetics , Spliceosomes/metabolism , Spliceosomes/genetics , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Malaria/parasitology , Plasmodium/genetics , Plasmodium/metabolism
2.
ACS Infect Dis ; 10(6): 1904-1913, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38752809

ABSTRACT

Malaria parasites have a complex life cycle and undergo replication and population expansion within vertebrate hosts and mosquito vectors. These developmental transitions rely on changes in gene expression and chromatin reorganization that result in the activation and silencing of stage-specific genes. The ApiAp2 family of DNA-binding proteins plays an important role in regulating gene expression in malaria parasites. Here, we characterized the ApiAp2 protein in Plasmodium berghei, which we termed Ap2-D. In silico analysis revealed that Ap2-D has three beta-sheets followed by a helix at the C-terminus for DNA binding. Using gene tagging with 3XHA-mCherry, we found that Ap2-D is expressed in Plasmodium blood stages and is present in the parasite cytoplasm and nucleus. Surprisingly, our gene deletion study revealed a completely dispensable role for Ap2-D in the entirety of the P. berghei life cycle. Ap2-D KO parasites were found to grow in the blood successfully and progress through the mosquito midgut and salivary glands. Sporozoites isolated from mosquito salivary glands were infective for hepatocytes and achieved similar patency as WT in mice. We emphasize the importance of genetic validation of antimalarial drug targets before progressing them to drug discovery.


Subject(s)
Life Cycle Stages , Plasmodium berghei , Protozoan Proteins , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Animals , Mice , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Malaria/parasitology , Transcription Factors/genetics , Transcription Factors/metabolism , Sporozoites/growth & development , Sporozoites/metabolism , Sporozoites/physiology , Salivary Glands/parasitology , Mosquito Vectors/parasitology , Female , Anopheles/parasitology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Hepatocytes/parasitology
3.
Mol Microbiol ; 121(6): 1095-1111, 2024 06.
Article in English | MEDLINE | ID: mdl-38574236

ABSTRACT

The protozoan parasite Plasmodium, the causative agent of malaria, undergoes an obligatory stage of intra-hepatic development before initiating a blood-stage infection. Productive invasion of hepatocytes involves the formation of a parasitophorous vacuole (PV) generated by the invagination of the host cell plasma membrane. Surrounded by the PV membrane (PVM), the parasite undergoes extensive replication. During intracellular development in the hepatocyte, the parasites provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterized by a long-lasting association of the autophagy marker protein, and ATG8 family member, LC3B with the PVM. LC3B localization at the PVM does not follow the canonical autophagy pathway since upstream events specific to canonical autophagy are dispensable. Here, we describe that LC3B localization at the PVM of Plasmodium parasites requires the V-ATPase and its interaction with ATG16L1. The WD40 domain of ATG16L1 is crucial for its recruitment to the PVM. Thus, we provide new mechanistic insight into the previously described PAAR response targeting Plasmodium liver stage parasites.


Subject(s)
Autophagy-Related Proteins , Autophagy , Hepatocytes , Liver , Microtubule-Associated Proteins , Plasmodium berghei , Vacuolar Proton-Translocating ATPases , Vacuoles , Vacuoles/metabolism , Vacuoles/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium berghei/enzymology , Animals , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Liver/parasitology , Mice , Hepatocytes/parasitology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Malaria/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans
4.
Curr Opin Microbiol ; 79: 102469, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574448

ABSTRACT

Malaria blood stage parasites commit to either one of two distinct cellular fates while developing within erythrocytes of their mammalian host: they either undergo another round of asexual replication or they differentiate into nonreplicative transmissible gametocytes. Depending on the state of infection, either path may support or impair the ultimate goal of human-to-human transmission via the mosquito vector. Malaria parasites therefore evolved strategies to control investments into asexual proliferation versus gametocyte formation. Recent work provided fascinating molecular insight into shared and unique mechanisms underlying the control and environmental modulation of sexual commitment in the two most widely studied malaria parasite species, Plasmodium falciparum and P. berghei. With this review, we aim at placing these findings into a comparative mechanistic context.


Subject(s)
Plasmodium berghei , Plasmodium falciparum , Plasmodium falciparum/physiology , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Animals , Humans , Plasmodium berghei/physiology , Plasmodium berghei/growth & development , Plasmodium berghei/genetics , Malaria/parasitology , Malaria/transmission , Erythrocytes/parasitology
5.
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
6.
Mol Microbiol ; 121(5): 940-953, 2024 05.
Article in English | MEDLINE | ID: mdl-38419272

ABSTRACT

Plasmodium is an obligate intracellular parasite that requires intense lipid synthesis for membrane biogenesis and survival. One of the principal membrane components is oleic acid, which is needed to maintain the membrane's biophysical properties and fluidity. The malaria parasite can modify fatty acids, and stearoyl-CoA Δ9-desaturase (Scd) is an enzyme that catalyzes the synthesis of oleic acid by desaturation of stearic acid. Scd is dispensable in P. falciparum blood stages; however, its role in mosquito and liver stages remains unknown. We show that P. berghei Scd localizes to the ER in the blood and liver stages. Disruption of Scd in the rodent malaria parasite P. berghei did not affect parasite blood stage propagation, mosquito stage development, or early liver-stage development. However, when Scd KO sporozoites were inoculated intravenously or by mosquito bite into mice, they failed to initiate blood-stage infection. Immunofluorescence analysis revealed that organelle biogenesis was impaired and merozoite formation was abolished, which initiates blood-stage infections. Genetic complementation of the KO parasites restored merozoite formation to a level similar to that of WT parasites. Mice immunized with Scd KO sporozoites confer long-lasting sterile protection against infectious sporozoite challenge. Thus, the Scd KO parasite is an appealing candidate for inducing protective pre-erythrocytic immunity and hence its utility as a GAP.


Subject(s)
Malaria , Merozoites , Organelle Biogenesis , Plasmodium berghei , Sporozoites , Stearoyl-CoA Desaturase , Animals , Female , Mice , Anopheles/parasitology , Endoplasmic Reticulum/metabolism , Liver/parasitology , Malaria/parasitology , Merozoites/growth & development , Merozoites/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium berghei/enzymology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Sporozoites/growth & development , Sporozoites/metabolism , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics
7.
mSphere ; 8(4): e0058722, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37272704

ABSTRACT

During invasion, Plasmodium parasites secrete proteins from rhoptry and microneme apical end organelles, which have crucial roles in attaching to and invading target cells. A sporozoite stage-specific gene silencing system revealed that rhoptry neck protein 2 (RON2), RON4, and RON5 are important for sporozoite invasion of mosquito salivary glands. Here, we further investigated the roles of RON4 during sporozoite infection of the liver in vivo. Following intravenous inoculation of RON4-knockdown sporozoites into mice, we demonstrated that sporozoite RON4 has multiple functions during sporozoite traversal of sinusoidal cells and infection of hepatocytes. In vitro infection experiments using a hepatoma cell line revealed that secreted RON4 is involved in sporozoite adhesion to hepatocytes and has an important role in the early steps of hepatocyte infection. In addition, in vitro motility assays indicated that RON4 is required for sporozoite attachment to the substrate and the onset of migration. These findings indicate that RON4 is crucial for sporozoite migration toward and invasion of hepatocytes via attachment ability and motility.IMPORTANCEMalarial parasite transmission to mammals is established when sporozoites are inoculated by mosquitoes and migrate through the bloodstream to infect hepatocytes. Many aspects of the molecular mechanisms underpinning migration and cellular invasion remain largely unelucidated. By applying a sporozoite stage-specific gene silencing system in the rodent malarial parasite, Plasmodium berghei, we demonstrated that rhoptry neck protein 4 (RON4) is crucial for sporozoite infection of the liver in vivo. Combined with in vitro investigations, it was revealed that RON4 functions during a crossing of the sinusoidal cell layer and invading hepatocytes, at an early stage of liver infection, by mediating the sporozoite capacity for adhesion and the onset of motility. Since RON4 is also expressed in Plasmodium merozoites and Toxoplasma tachyzoites, our findings contribute to understanding the conserved invasion mechanisms of Apicomplexa parasites.


Subject(s)
Malaria , Plasmodium berghei , Sporozoites , Animals , Mice , Plasmodium berghei/growth & development , Plasmodium berghei/physiology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria/metabolism , Malaria/parasitology , Malaria/pathology , Sporozoites/physiology , Protozoan Proteins/metabolism , Hepatocytes/metabolism , Hepatocytes/parasitology , Hepatocytes/pathology
8.
J Immunol ; 208(5): 1292-1304, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35131868

ABSTRACT

Pathogen-specific CD8 T cells face the problem of finding rare cells that present their cognate Ag either in the lymph node or in infected tissue. Although quantitative details of T cell movement strategies in some tissues such as lymph nodes or skin have been relatively well characterized, we still lack quantitative understanding of T cell movement in many other important tissues, such as the spleen, lung, liver, and gut. We developed a protocol to generate stable numbers of liver-located CD8 T cells, used intravital microscopy to record movement patterns of CD8 T cells in livers of live mice, and analyzed these and previously published data using well-established statistical and computational methods. We show that, in most of our experiments, Plasmodium-specific liver-localized CD8 T cells perform correlated random walks characterized by transiently superdiffusive displacement with persistence times of 10-15 min that exceed those observed for T cells in lymph nodes. Liver-localized CD8 T cells typically crawl on the luminal side of liver sinusoids (i.e., are in the blood); simulating T cell movement in digital structures derived from the liver sinusoids illustrates that liver structure alone is sufficient to explain the relatively long superdiffusive displacement of T cells. In experiments when CD8 T cells in the liver poorly attach to the sinusoids (e.g., 1 wk after immunization with radiation-attenuated Plasmodium sporozoites), T cells also undergo Lévy flights: large displacements occurring due to cells detaching from the endothelium, floating with the blood flow, and reattaching at another location. Our analysis thus provides quantitative details of movement patterns of liver-localized CD8 T cells and illustrates how structural and physiological details of the tissue may impact T cell movement patterns.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Movement/physiology , Liver/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Animals , Capillaries/cytology , Cellular Microenvironment/physiology , Liver/blood supply , Malaria/pathology , Mice , Plasmodium berghei/growth & development , Sporozoites/growth & development , Sporozoites/immunology , Vaccination
9.
Front Immunol ; 12: 758052, 2021.
Article in English | MEDLINE | ID: mdl-34899708

ABSTRACT

Hepatopathy is frequently observed in patients with severe malaria but its pathogenesis remains unclear. Galectins are evolutionarily conserved glycan-binding proteins with pleiotropic roles in innate and adaptive immune responses, and exhibit pivotal roles during Plasmodium spp. infection. Here, we analyzed the impact of blockage of galectin-receptor interactions by treatment with alpha (α)-lactose on liver immunopathology during the erythrocytic stage of malaria in mice infected with Plasmodium berghei ANKA (PbANKA). Our results found that compared with PbANKA-infected mice (malarial mice), blockage of galectin-receptor interactions led to decreased host survival rate and increased peripheral blood parasitemia; exacerbated liver pathology, increased numbers of CD68+ macrophages and apoptotic cells, and increased parasite burden in the livers on days 5 and 7 post infection (p.i.) as well as increased mRNA expression levels of galectin-9 (Gal-9) and its receptor, the T cell immunoglobulin domain and mucin domain protein 3 (Tim-3), interferon (IFN)α, IFNγ, and the triggering receptor expressed on myeloid cells (TREM)-1 in the livers or spleens of PbANKA-infected mice on day 7 p.i. Observed by transmission electron microscopy, the peritoneal macrophages isolated from malarial mice with α-lactose treatment had more pseudopodia than those from malarial mice. Measured by using quantitative real-time reverse transcription-polymerase chain reaction assay, the mRNA expression levels of Gal-9, IFNα, IFNß, IFNγ, and TREM-1 were increased in the peritoneal macrophages isolated from malarial mice with α-lactose treatment in comparison of those from malarial mice. Furthermore, significant positive correlations existed between the mRNA levels of Gal-9 and Tim-3/IFNγ/TREM-1 in both the livers and the peritoneal macrophages, and between Gal-9 and Tim-3/TREM-1 in the spleens of malarial mice; significant positive correlations existed between the mRNA levels of Gal-9 and IFNγ in the livers and between Gal-9 and IFNα in the peritoneal macrophages from malarial mice treated with α-lactose. Our data suggest a potential role of galectin-receptor interactions in limiting liver inflammatory response and parasite proliferation by down-regulating the expressions of IFNα, IFNγ, and TREM-1 during PbANKA infection.


Subject(s)
Erythrocytes/parasitology , Galectins/physiology , Liver/pathology , Malaria/pathology , Parasitemia/pathology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Female , Galectins/antagonists & inhibitors , Hepatitis A Virus Cellular Receptor 2/antagonists & inhibitors , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Lactose/pharmacology , Lactose/toxicity , Liver/parasitology , Lung/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/ultrastructure , Malaria/blood , Mice , Plasmodium berghei/growth & development , Pseudopodia/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Triggering Receptor Expressed on Myeloid Cells-1/biosynthesis , Triggering Receptor Expressed on Myeloid Cells-1/genetics
10.
Biochem Pharmacol ; 194: 114834, 2021 12.
Article in English | MEDLINE | ID: mdl-34774530

ABSTRACT

Malaria, which is caused by protozoa of the genus Plasmodium, remains a major endemic public health problem worldwide. Since artemisinin combination therapies are used as a first-line treatment in all endemic regions, the emergence of parasites resistant to these regimens has become a serious problem. Differentiation-inducing factor 1 (DIF-1) is a chlorinated alkylphenone originally found in the cellular slime mold Dictyostelium discoideum. DIF-1 and its derivatives exhibit a range of biological activities. In the present study, we investigated the effects of 41 DIF derivatives on the growth of Plasmodium falciparum in vitro using four laboratory strains and 12 field isolates. Micromolar concentrations of several DIF derivatives strongly suppressed the growth of the four laboratory strains, including strains that exhibited resistance to chloroquine and artemisinin, as well as strains that were susceptible to these drugs. In addition, DIF-1(+2), the most potent derivative, strongly suppressed the growth of 12 field isolates. We also examined the effects of DIF-1(+2) on the activity of the rodent malarial parasite Plasmodium berghei in mice. Intraperitoneal administration of DIF-1(+2) over 4 days (50 or 70 mg/kg/day) significantly suppressed the growth of the parasite in the blood with no apparent adverse effects, and a dose of 70 mg/kg/day significantly prolonged animal survival. These results suggest that DIF derivatives, such as DIF-1(+2), could serve as new lead compounds for the development of antimalarial agents.


Subject(s)
Antimalarials/pharmacology , Dictyostelium , Hexanones/pharmacology , Parasites/growth & development , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , 3T3-L1 Cells , Animals , Female , Humans , Mice , Mice, Inbred BALB C , Parasites/drug effects , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects
11.
Parasitol Int ; 84: 102403, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34119684

ABSTRACT

The transcription factor (TF) AP2-G is essential for gametocytogenesis in the malaria parasite; however, it remains unclear if AP2-G determines commitment to sexual stage development fate in the schizont stage, or whether AP2-G directly initiates sexual stage differentiation and development beginning in the late-trophozoite stage. In this study, we addressed this issue by investigating the expression profile of AP2-G and determining genome-wide target genes in Plasmodium berghei. Fluorescence microscopy showed that AP2-G expression was first observed in the parasite 12 h after erythrocyte invasion and peaked at 18 h when sexual features were first manifested in early gametocytes. Expression of AP2-G decreased with manifestation of sex-specific features. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) was performed at peak AP2-G expression and identified over 1000 binding sites in the genome. The main binding motif of the TF predicted from the binding sites was GTACNY. Predicted targets contained a number of genes related to protein biogenesis, suggesting that AP2-G plays a role in establishing a cellular basis required for sexual differentiation. AP2-G binding sites also existed upstream of gametocyte-specific TFs, namely AP2-G2, AP2-FG, and AP2-G itself. Furthermore, the target contained two AP2 TF-related genes. Disruption of these genes resulted in the arrest of ookinete development. These results suggest another role of AP2-G: activating a transcriptional cascade to promote conversion into early gametocytes. Taken together, AP2-G is involved not in establishing sexual commitment of schizonts, but rather in triggering the initiation of differentiation and the early development of gametocytes in the late trophozoite stage.


Subject(s)
Malaria/metabolism , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , Schizonts/physiology , Animals , Gametogenesis , Mice , Mice, Inbred BALB C , Plasmodium berghei/growth & development , Rats , Rats, Wistar , Schizonts/growth & development
12.
Parasit Vectors ; 14(1): 241, 2021 May 07.
Article in English | MEDLINE | ID: mdl-33962671

ABSTRACT

BACKGROUND: Transmission-blocking vaccine (TBV) is a promising strategy for malaria elimination. It is hypothesized that mixing or fusing two antigens targeting different stages of sexual development may provide higher transmission-blocking activity than these antigens used individually. METHODS: A chimeric protein composed of fragments of Pbg37 and PSOP25 was designed and expressed the recombinant protein in Escherichia coli Rosetta-gami B (DE3). After immunizing mice with individual recombinant proteins Pbg37 and PSOP25, mixed proteins (Pbg37+PSOP25), or the fusion protein (Pbg37-PSOP25), the antibody titers of individual sera were analyzed by ELISA. IFA and Western blot were performed to test the reactivity of the antisera with the native proteins in the parasite. The transmission-blocking activity of the different immunization schemes was assessed using in vitro and in vivo assays. RESULTS: When Pbg37 and PSOP25 were co-administered in a mixture or as a fusion protein, they elicited similar antibody responses in mice as single antigens without causing immunological interference with each other. Antibodies against the mixed or fused antigens recognized the target proteins in the gametocyte, gamete, zygote, and ookinete stages. The mixed proteins or the fusion protein induced antibodies with significantly stronger transmission-reducing activities in vitro and in vivo than individual antigens. CONCLUSIONS: There was no immunological interference between Pbg37 and PSOP25. The bivalent vaccines, which expand the portion of the sexual development during which the transmission-blocking antibodies act, produced significantly stronger transmission-reducing activities than single antigens. Altogether, these data provide the theoretical basis for the development of combination TBVs targeting different sexual stages.


Subject(s)
Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Protozoan Proteins/administration & dosage , Vaccines, Combined/administration & dosage , Animals , Antibodies, Protozoan/blood , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Immunization , Malaria/blood , Malaria/parasitology , Malaria/transmission , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccines, Combined/genetics , Vaccines, Combined/immunology
13.
Mol Biochem Parasitol ; 244: 111375, 2021 07.
Article in English | MEDLINE | ID: mdl-34023299

ABSTRACT

Malaria parasites exhibit a complex lifecycle, requiring extensive asexual replication in the liver and blood of the vertebrate host, and in the haemocoel of the insect vector. Yet, they must also undergo a single round of sexual reproduction, which occurs in the vector's midgut upon uptake of a blood meal. Sexual reproduction is obligate for infection of the vector and thus, is essential for onwards transmission to new hosts. Sex in malaria parasites involves several bottlenecks in parasite number, making the stages involved attractive targets for blocking disease transmission. Malaria parasites have evolved a suite of adaptations ("strategies") to maximise the success of sexual reproduction and transmission, which could undermine transmission-blocking interventions. Yet, understanding parasite strategies may also reveal novel opportunities for such interventions. Here, we outline how evolutionary and ecological theories, developed to explain reproductive strategies in multicellular taxa, can be applied to explain two reproductive strategies (conversion rate and sex ratio) expressed by malaria parasites within the vertebrate host.


Subject(s)
Gametogenesis , Life Cycle Stages/genetics , Malaria/parasitology , Plasmodium berghei/growth & development , Plasmodium chabaudi/growth & development , Plasmodium falciparum/growth & development , Plasmodium knowlesi/growth & development , Animals , Biological Coevolution , Culicidae/parasitology , Erythrocytes/parasitology , Female , Host-Parasite Interactions/genetics , Humans , Insect Vectors/parasitology , Liver/parasitology , Malaria/transmission , Male , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Plasmodium chabaudi/genetics , Plasmodium chabaudi/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Plasmodium knowlesi/genetics , Plasmodium knowlesi/metabolism , Reproduction, Asexual , Sex Ratio
14.
mSphere ; 6(2)2021 03 31.
Article in English | MEDLINE | ID: mdl-33789941

ABSTRACT

Mosquitoes may feed multiple times during their life span in addition to those times needed to acquire and transmit malaria. To determine the impact of subsequent blood feeding on parasite development in Anopheles gambiae, we examined Plasmodium parasite infection with or without an additional noninfected blood meal. We found that an additional blood meal significantly reduced Plasmodium berghei immature oocyst numbers, yet had no effect on the human parasite Plasmodium falciparum These observations were reproduced when mosquitoes were fed an artificial protein meal, suggesting that parasite losses are independent of blood ingestion. We found that feeding with either a blood or protein meal compromises midgut basal lamina integrity as a result of the physical distention of the midgut, enabling the recognition and lysis of immature P. berghei oocysts by mosquito complement. Moreover, we demonstrate that additional feeding promotes P. falciparum oocyst growth, suggesting that human malaria parasites exploit host resources provided with blood feeding to accelerate their growth. This is in contrast to experiments with P. berghei, where the size of surviving oocysts is independent of an additional blood meal. Together, these data demonstrate distinct differences in Plasmodium species in evading immune detection and utilizing host resources at the oocyst stage, representing an additional, yet unexplored component of vectorial capacity that has important implications for the transmission of malaria.IMPORTANCE Mosquitoes must blood feed multiple times to acquire and transmit malaria. However, the impact of an additional mosquito blood meal following malaria parasite infection has not been closely examined. Here, we demonstrate that additional feeding affects mosquito vector competence; namely, additional feeding significantly limits Plasmodium berghei infection, yet has no effect on infection of the human parasite P. falciparum Our experiments support that these killing responses are mediated by the physical distension of the midgut and by temporary damage to the midgut basal lamina that exposes immature P. berghei oocysts to mosquito complement, while human malaria parasites are able to evade these killing mechanisms. In addition, we provide evidence that additional feeding promotes P. falciparum oocyst growth. This is in contrast to P. berghei, where oocyst size is independent of an additional blood meal. This suggests that human malaria parasites are able to exploit host resources provided by an additional feeding to accelerate their growth. In summary, our data highlight distinct differences in malaria parasite species in evading immune recognition and adapting to mosquito blood feeding. These observations have important, yet previously unexplored, implications for the impact of multiple blood meals on the transmission of malaria.


Subject(s)
Anopheles/parasitology , Feeding Behavior , Host-Parasite Interactions , Plasmodium/growth & development , Plasmodium/immunology , Animals , Anopheles/physiology , Blood , Female , Immune Evasion , Malaria/parasitology , Malaria/transmission , Meals , Mice , Mosquito Vectors/parasitology , Oocysts/growth & development , Oocysts/immunology , Plasmodium/classification , Plasmodium berghei/growth & development , Plasmodium berghei/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology
15.
Biochem J ; 478(9): 1705-1732, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33843972

ABSTRACT

Autophagy, a lysosome-dependent degradative process, does not appear to be a major degradative process in malaria parasites and has a limited repertoire of genes. To better understand the autophagy process, we investigated Plasmodium falciparum Atg18 (PfAtg18), a PROPPIN family protein, whose members like S. cerevisiae Atg18 (ScAtg18) and human WIPI2 bind PI3P and play an essential role in autophagosome formation. Wild type and mutant PfAtg18 were expressed in P. falciparum and assessed for localization, the effect of various inhibitors and antimalarials on PfAtg18 localization, and identification of PfAtg18-interacting proteins. PfAtg18 is expressed in asexual erythrocytic stages and localized to the food vacuole, which was also observed with other Plasmodium Atg18 proteins, indicating that food vacuole localization is likely a shared feature. Interaction of PfAtg18 with the food vacuole-associated PI3P is essential for localization, as PfAtg18 mutants of PI3P-binding motifs neither bound PI3P nor localized to the food vacuole. Interestingly, wild type ScAtg18 interacted with PI3P, but its expression in P. falciparum showed complete cytoplasmic localization, indicating additional requirement for food vacuole localization. The food vacuole multi-drug resistance protein 1 (MDR1) was consistently identified in the immunoprecipitates of PfAtg18 and P. berghei Atg18, and also interacted with PfAtg18. In contrast with PfAtg18, ScAtg18 did not interact with MDR1, which, in addition to PI3P, could play a critical role in localization of PfAtg18. Chloroquine and amodiaquine caused cytoplasmic localization of PfAtg18, suggesting that these target PfAtg18 transport pathway. Thus, PI3P and MDR1 are critical mediators of PfAtg18 localization.


Subject(s)
Autophagy-Related Proteins/genetics , Multidrug Resistance-Associated Proteins/genetics , Phosphatidylinositol Phosphates/metabolism , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Vacuoles/metabolism , Amodiaquine/pharmacology , Animals , Antimalarials/pharmacology , Autophagy/genetics , Autophagy-Related Proteins/metabolism , Biological Transport , Chloroquine/pharmacology , Erythrocytes/drug effects , Erythrocytes/parasitology , Gene Expression Regulation , Humans , Malaria/parasitology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Multidrug Resistance-Associated Proteins/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protein Binding , Protozoan Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Vacuoles/drug effects
16.
Cell Rep ; 35(3): 108992, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882310

ABSTRACT

Plant-nectar-derived sugar is the major energy source for mosquitoes, but its influence on vector competence for malaria parasites remains unclear. Here, we show that Plasmodium berghei infection of Anopheles stephensi results in global metabolome changes, with the most significant impact on glucose metabolism. Feeding on glucose or trehalose (the main hemolymph sugars) renders the mosquito more susceptible to Plasmodium infection by alkalizing the mosquito midgut. The glucose/trehalose diets promote proliferation of a commensal bacterium, Asaia bogorensis, that remodels glucose metabolism in a way that increases midgut pH, thereby promoting Plasmodium gametogenesis. We also demonstrate that the sugar composition from different natural plant nectars influences A. bogorensis growth, resulting in a greater permissiveness to Plasmodium. Altogether, our results demonstrate that dietary glucose is an important determinant of mosquito vector competency for Plasmodium, further highlighting a key role for mosquito-microbiota interactions in regulating the development of the malaria parasite.


Subject(s)
Acetobacteraceae/metabolism , Anopheles/metabolism , Glucose/pharmacology , Metabolome , Mosquito Vectors/metabolism , Trehalose/pharmacology , Acetobacteraceae/growth & development , Animals , Anopheles/drug effects , Anopheles/microbiology , Anopheles/parasitology , Digestive System/microbiology , Digestive System/parasitology , Female , Gametogenesis/drug effects , Gametogenesis/genetics , Gene Expression Regulation , Glucose/metabolism , Host-Pathogen Interactions/genetics , Hydrogen-Ion Concentration , Life Cycle Stages/drug effects , Life Cycle Stages/genetics , Malaria/parasitology , Microbiota/genetics , Mosquito Vectors/drug effects , Mosquito Vectors/microbiology , Mosquito Vectors/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Symbiosis/genetics , Trehalose/metabolism
17.
DNA Repair (Amst) ; 101: 103078, 2021 05.
Article in English | MEDLINE | ID: mdl-33711786

ABSTRACT

Mitochondrion is an essential organelle in malaria parasite and its DNA must be maintained for optimal function during its complex life cycle. Base excision repair is one of the major pathways by which this is achieved. Apurinic/apyrimidinic (AP) endonucleases are important components of this pathway as they create a nick at the 5'-phosphodiester bond in the AP site and generate free 5'-phosphate and 3'-hydroxyl groups. Two class II AP endonucleases (Apn1 and Ape1) have been annotated in the Plasmodium berghei genome. Using reverse genetic approaches, we provide direct evidence that Apn1 is exclusively localized to the mitochondria of P. berghei. Surprisingly, our gene deletion study revealed a completely dispensable role of Apn1 for the entirety of the P. berghei life cycle. Apn1- parasites were found to successfully grow in the blood. They were transmitted normally to the mosquito midguts and salivary glands. Sporozoites obtained from the salivary glands were infective and achieved similar patency as WT. Our results help emphasize the non-availability of this enzyme as a plausible drug target. We also emphasize the importance of genetic validation of antimalarial drug targets before furthering them down the drug discovery pipeline.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Life Cycle Stages , Mitochondria/enzymology , Plasmodium berghei/enzymology , Amino Acid Sequence , DNA Damage , DNA Repair , DNA, Mitochondrial/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Models, Molecular , Phylogeny , Plasmodium berghei/growth & development , Protein Conformation , Sequence Alignment , Sequence Analysis, Protein
18.
Biochem Biophys Res Commun ; 549: 61-66, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33667710

ABSTRACT

The glyoxalase system is a ubiquitous detoxification pathway of methylglyoxal, a cytotoxic byproduct of glycolysis. Actively proliferating cells, such as cancer cells, depend on their energy metabolism for glycolysis. Therefore, the glyoxalase system has been evaluated as a target of anticancer drugs. The malaria sporozoite, which is the infective stage of the malaria parasite, actively proliferates and produces thousands of merozoites within 2-3 days in hepatocytes. This is the first step of infection in mammalian hosts. The glyoxalase system appears to play an important role in this active proliferation stage of the malaria parasite in hepatocytes. In this study, we aimed to dissect the role of the glyoxalase system in malaria parasite proliferation in hepatocytes to examine its potential as a target of malaria prevention using a reverse genetics approach. The malaria parasite possesses a glyoxalase system, comprised of glyoxalases and GloI-like protein, in the cytosol and apicoplast. We generated cytosolic glyoxalase II (cgloII) knockout, apicoplast targeted glyoxalase gloII (tgloII) knockout, and cgloII and tgloII double-knockout parasites and performed their phenotypic analysis. We did not observe any defects in the cgloII or tgloII knockout parasites. In contrast, we observed approximately 90% inhibition of the liver-stage proliferation of cgloII and tgloII double-knockout parasites in vivo. These findings suggest that although the glyoxalase system is dispensable, it plays an important role in parasite proliferation in hepatocytes. Additionally, the results indicate a complementary relationship between the cytosolic and apicoplast glyoxalase pathways. We expect that the parasite utilizes a system similar to that observed in cancer cells to enable its rapid proliferation in hepatocytes; this process could be targeted in the development of novel strategies to prevent malaria.


Subject(s)
Lactoylglutathione Lyase/metabolism , Life Cycle Stages , Liver/parasitology , Metabolic Networks and Pathways , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Animals , Female , Gene Knockout Techniques , Malaria/parasitology , Malaria/pathology , Mice, Inbred BALB C , Mice, Inbred ICR , Parasites/metabolism
19.
Sci Rep ; 11(1): 4127, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619283

ABSTRACT

In the malaria-causing parasite's life cycle, Plasmodium sporozoites must travel from the midgut of a mosquito to the salivary glands before they can infect a mammalian host. However, only a fraction of sporozoites complete the journey. Since salivary gland invasion is required for transmission of sporozoites, insights at the molecular level can contribute to strategies for malaria prevention. Recent advances in single-cell RNA sequencing provide an opportunity to assess sporozoite heterogeneity at a resolution unattainable by bulk RNA sequencing methods. In this study, we use a droplet-based single-cell RNA sequencing workflow to analyze the transcriptomes of over 8000 Plasmodium berghei sporozoites derived from the midguts and salivary glands of Anopheles stephensi mosquitoes. The detection of known marker genes confirms the successful capture and sequencing of samples composed of a mixed population of sporozoites. Using data integration, clustering, and trajectory analyses, we reveal differences in gene expression profiles of individual sporozoites, and identify both annotated and unannotated markers associated with sporozoite development. Our work highlights the utility of a high-throughput workflow for the transcriptomic profiling of Plasmodium sporozoites, and provides new insights into gene usage during the parasite's development in the mosquito.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Plasmodium berghei/genetics , Single-Cell Analysis , Sporozoites/genetics , Transcriptome , Computational Biology/methods , Gene Expression Profiling/methods , Gene Ontology , Genetic Heterogeneity , Malaria/parasitology , Organ Specificity/genetics , Plasmodium berghei/growth & development , Single-Cell Analysis/methods , Sporozoites/growth & development
20.
Cell Microbiol ; 23(5): e13321, 2021 05.
Article in English | MEDLINE | ID: mdl-33600048

ABSTRACT

The circumsporozoite protein, CSP, is the major surface protein of Plasmodium sporozoites, the form of malaria parasites transmitted by mosquitoes. CSP is involved in sporozoite formation within and egress from oocysts, entry into mosquito salivary glands and mammalian liver as well as migration in the skin. Yet, how CSP facilitates sporozoite formation, oocyst egress and hepatocyte specific invasion is still not fully understood. Here, we aimed at generating a series of parasites expressing full-length versions of CSP with internally inserted green fluorescent protein between known domains at the endogenous csp locus. This enabled the investigation of sporozoite formation in living oocysts. GFP insertion after the signal peptide leads to cleavage of GFP before the fusion protein reached the plasma membrane while insertion of GFP before or after the TSR domain prevented sporozoite egress and liver invasion. These data suggest different strategies for obtaining mature salivary gland sporozoites that express GFP-CSP fusions.


Subject(s)
Anopheles/parasitology , Malaria/parasitology , Oocysts/physiology , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , Sporozoites/growth & development , Animals , Cell Membrane/metabolism , Green Fluorescent Proteins , Mice, Inbred C57BL , Microtubules/ultrastructure , Movement , Plasmodium berghei/metabolism , Plasmodium berghei/ultrastructure , Protein Transport , Protozoan Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sporozoites/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL