Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
J Cell Sci ; 137(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38832798

ABSTRACT

Plasmodium sporozoites are the infective forms of the malaria parasite in the mosquito and vertebrate host. Gliding motility allows sporozoites to migrate and invade mosquito salivary glands and mammalian hosts. Motility and invasion are powered by an actin-myosin motor complex linked to the glideosome, which contains glideosome-associated proteins (GAPs), MyoA and the myosin A tail-interacting protein (MTIP). However, the role of several proteins involved in gliding motility remains unknown. We identified that the S14 gene is upregulated in sporozoite from transcriptome data of Plasmodium yoelii and further confirmed its transcription in P. berghei sporozoites using real-time PCR. C-terminal 3×HA-mCherry tagging revealed that S14 is expressed and localized on the inner membrane complex of the sporozoites. We disrupted S14 in P. berghei and demonstrated that it is essential for sporozoite gliding motility, and salivary gland and hepatocyte invasion. The gliding and invasion-deficient S14 knockout sporozoites showed normal expression and organization of inner membrane complex and surface proteins. Taken together, our data show that S14 plays a role in the function of the glideosome and is essential for malaria transmission.


Subject(s)
Malaria , Plasmodium berghei , Protozoan Proteins , Sporozoites , Sporozoites/metabolism , Plasmodium berghei/metabolism , Plasmodium berghei/genetics , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Animals , Mice , Malaria/parasitology , Salivary Glands/parasitology , Salivary Glands/metabolism , Anopheles/parasitology
2.
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
3.
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
4.
Mol Microbiol ; 121(6): 1095-1111, 2024 Jun.
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
5.
PLoS Pathog ; 20(2): e1012008, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38354186

ABSTRACT

Leucine-rich repeat (LRR) proteins are commonly involved in innate immunity of animals and plants, including for pattern recognition of pathogen-derived elicitors. The Anopheles secreted LRR proteins APL1C and LRIM1 are required for malaria ookinete killing in conjunction with the complement-like TEP1 protein. However, the mechanism of parasite immune recognition by the mosquito remains unclear, although it is known that TEP1 lacks inherent binding specificity. Here, we find that APL1C and LRIM1 bind specifically to Plasmodium berghei ookinetes, even after depletion of TEP1 transcript and protein, consistent with a role for the LRR proteins in pathogen recognition. Moreover, APL1C does not bind to ookinetes of the human malaria parasite Plasmodium falciparum, and is not required for killing of this parasite, which correlates LRR binding specificity and immune protection. Most of the live P. berghei ookinetes that migrated into the extracellular space exposed to mosquito hemolymph, and almost all dead ookinetes, are bound by APL1C, thus associating LRR protein binding with parasite killing. We also find that APL1C binds to the surface of P. berghei sporozoites released from oocysts into the mosquito hemocoel and forms a potent barrier limiting salivary gland invasion and mosquito infectivity. Pathogen binding by APL1C provides the first functional explanation for the long-known requirement of APL1C for P. berghei ookinete killing in the mosquito midgut. We propose that secreted mosquito LRR proteins are required for pathogen discrimination and orientation of immune effector activity, potentially as functional counterparts of the immunoglobulin-based receptors used by vertebrates for antigen recognition.


Subject(s)
Anopheles , Malaria , Animals , Humans , Leucine-Rich Repeat Proteins , Anopheles/parasitology , Sporozoites/metabolism , Proteins/metabolism , Plasmodium berghei/metabolism
6.
Mol Microbiol ; 121(3): 565-577, 2024 03.
Article in English | MEDLINE | ID: mdl-38396332

ABSTRACT

Plasmodium sporozoites are the highly motile and invasive forms of the malaria parasite transmitted by mosquitoes. Sporozoites form within oocysts at the midgut wall of the mosquito, egress from oocysts and enter salivary glands prior to transmission. The GPI-anchored major surface protein, the circumsporozoite protein (CSP) is important for Plasmodium sporozoite formation, egress, migration and invasion. To visualize CSP, we previously generated full-length versions of CSP internally tagged with the green fluorescent protein, GFP. However, while these allowed for imaging of sporogony in oocysts, sporozoites failed to egress. Here, we explore different strategies to overcome this block in egress and obtain salivary gland resident sporozoites that express CSP-GFP. Replacing the N-terminal and repeat region with GFP did not allow sporozoite formation. Lowering expression of CSP-GFP at the endogenous locus allowed sporozoite formation but did not overcome egress block. Crossing of CSP-GFP expressing parasites that are blocked in egress with wild-type parasites yielded a small fraction of parasites that entered salivary glands and expressed various levels of CSP-GFP. Expressing CSP-GFP constructs from a silent chromosome region from promoters that are active only post salivary gland invasion yielded normal numbers of fluorescent salivary gland sporozoites, albeit with low levels of fluorescence. We also show that lowering CSP expression by 50% allowed egress from oocysts but not salivary gland entry. In conclusion, Plasmodium berghei parasites with normal CSP expression tolerate a certain level of CSP-GFP without disruption of oocyst egress and salivary gland invasion.


Subject(s)
Anopheles , Sporozoites , Animals , Sporozoites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Anopheles/parasitology , Oocysts , Plasmodium berghei/genetics , Plasmodium berghei/metabolism
7.
mBio ; 15(4): e0023224, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38411954

ABSTRACT

Neddylation is a type of posttranslational modification known to regulate a wide range of cellular processes by covalently conjugating the ubiquitin-like protein Nedd8 to target proteins at lysine residues. However, the role of neddylation in malaria parasites has not been determined. Here, for the first time, we showed that neddylation plays an essential role in malaria transmission in Plasmodium berghei. We found that disruption of Nedd8 did not affect blood-stage propagation, gametocyte development, gamete formation, or zygote formation while abolishing the formation of ookinetes and further transmission of the parasites in mosquitoes. These phenotypic defects in Nedd8 knockout parasites were complemented by reintroducing the gene that restored mosquito transmission to wild-type levels. Our data establish the role of P. berghei Nedd8 in malaria parasite transmission.IMPORTANCENeddylation is a process by which Nedd8 is covalently attached to target proteins through three-step enzymatic cascades. The attachment of Nedd8 residues results in a range of diverse functions, such as cell cycle regulation, metabolism, immunity, and tumorigenesis. The potential neddylation substrates are cullin (CUL) family members, which are implicated in controlling the cell cycle. Cullin neddylation leads to the activation of cullin-RING ubiquitin ligases, which regulate a myriad of biological processes through target-specific ubiquitylation. Neddylation possibly regulates meiosis in zygotes, which subsequently develop into ookinetes. Our findings point to an essential function of this neddylation pathway and highlight its possible importance in designing novel intervention strategies.


Subject(s)
Plasmodium berghei , Ubiquitins , Animals , Ubiquitins/genetics , Ubiquitins/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Cullin Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
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)
Liver , Malaria , Merozoites , Organelle Biogenesis , Plasmodium berghei , Sporozoites , Stearoyl-CoA Desaturase , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium berghei/enzymology , Animals , Mice , Liver/parasitology , Merozoites/growth & development , Merozoites/metabolism , Malaria/parasitology , Stearoyl-CoA Desaturase/metabolism , Stearoyl-CoA Desaturase/genetics , Sporozoites/growth & development , Sporozoites/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Anopheles/parasitology , Female , Endoplasmic Reticulum/metabolism
9.
Mol Microbiol ; 121(4): 767-780, 2024 04.
Article in English | MEDLINE | ID: mdl-38238886

ABSTRACT

Endoplasmic reticulum (ER) plays a pivotal role in the regulation of stress responses in multiple eukaryotic cells. However, little is known about the effector mechanisms that regulate stress responses in ER of the malaria parasite. Herein, we aimed to identify the importance of a transmembrane protein 33 (TMEM33)-domain-containing protein in life cycle of the rodent malaria parasite Plasmodium berghei. TMEM33 is an ER membrane-resident protein that is involved in regulating stress responses in various eukaryotic cells. A C-terminal tagged TMEM33 was localized in the ER throughout the blood and mosquito stages of development. Targeted deletion of TMEM33 confirmed its importance for asexual blood stages and ookinete development, in addition to its essential role for sporozoite infectivity in the mammalian host. Pilot scale analysis shows that the loss of TMEM33 results in the initiation of ER stress response and induction of autophagy. Our findings conclude an important role of TMEM33 in the development of all life cycle stages of the malaria parasite, which indicates its potential as an antimalarial target.


Subject(s)
Malaria , Plasmodium berghei , Animals , Endoplasmic Reticulum/metabolism , Life Cycle Stages , Malaria/parasitology , Membrane Proteins/metabolism , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism
10.
Mol Microbiol ; 121(3): 394-412, 2024 03.
Article in English | MEDLINE | ID: mdl-37314965

ABSTRACT

Plasmodium parasites, the eukaryotic pathogens that cause malaria, feature three distinct invasive forms tailored to the host environment they must navigate and invade for life cycle progression. One conserved feature of these invasive forms is the micronemes, apically oriented secretory organelles involved in egress, motility, adhesion, and invasion. Here we investigate the role of GPI-anchored micronemal antigen (GAMA), which shows a micronemal localization in all zoite forms of the rodent-infecting species Plasmodium berghei. ∆GAMA parasites are severely defective for invasion of the mosquito midgut. Once formed, oocysts develop normally, however, sporozoites are unable to egress and exhibit defective motility. Epitope-tagging of GAMA revealed tight temporal expression late during sporogony and showed that GAMA is shed during sporozoite gliding motility in a similar manner to circumsporozoite protein. Complementation of P. berghei knockout parasites with full-length P. falciparum GAMA partially restored infectivity to mosquitoes, indicating conservation of function across Plasmodium species. A suite of parasites with GAMA expressed under the promoters of CTRP, CAP380, and TRAP, further confirmed the involvement of GAMA in midgut infection, motility, and vertebrate infection. These data show GAMA's involvement in sporozoite motility, egress, and invasion, implicating GAMA as a regulator of microneme function.


Subject(s)
Culicidae , Parasites , Animals , Culicidae/metabolism , Culicidae/parasitology , Parasites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Oocysts , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Sporozoites/metabolism
11.
Int J Parasitol ; 54(2): 99-107, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37774810

ABSTRACT

The successful completion of gamete fertilization is essential for malaria parasite transmission, and this process can be targeted by intervention strategies. In this study, we identified a conserved gene (PBANKA_0813300) in the rodent malaria parasite Plasmodium berghei, which encodes a protein of 54 kDa (designated as Pbs54). Localization studies indicated that Pbs54 is associated with the plasma membranes of gametes and ookinetes. Functional studies by gene disruption showed that the Δpbs54 parasites had no defect in asexual proliferation, gametocyte development, or gametogenesis. However, the interactions between male and female gametes were significantly decreased compared with wild-type parasites. The Δpbs54 lines did not show a further reduction in zygote and ookinete numbers during in vitro culture, indicating that the defects were probably restricted to gamete fertilization. Consistent with this finding, mosquitoes fed on Δpbs54-infected mice showed a 30.1% reduction in infection prevalence and a 74.7% reduction in oocyst intensity. Cross-fertilization assay indicated that both male and female gametes were impaired in the Δpbs54 parasites. To evaluate its transmission-blocking potential, we obtained polyclonal antibodies from mice immunized with the recombinant Pbs54 (rPbs54) protein. In vitro assays showed that anti-rPbs54 sera inhibited ookinete formation by 42.7%. Our experiments identified Pbs54 as a fertility factor required for mosquito transmission and a novel candidate for a malaria transmission-blocking vaccine.


Subject(s)
Culicidae , Malaria Vaccines , Malaria , Animals , Female , Male , Mice , Antibodies, Protozoan , Fertilization , Germ Cells , Malaria/prevention & control , Membrane Proteins/genetics , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Recombinant Proteins
12.
Sci Rep ; 13(1): 22222, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38097730

ABSTRACT

Plasmodium oocysts develop on the abluminal side of the mosquito midgut in relatively small numbers. Oocysts possess an extracellular cell wall-the capsule-to protect them from the insect's haemolymph environment. To further maximise transmission, each oocyst generates hundreds of sporozoites through an asexual multiplication step called sporogony. Completion of transmission requires sporozoite egress from the capsule (excystation), but this process remains poorly understood. In this study, we fused the parasite-encoded capsule protein Cap380 with green fluorescent protein in a transgenic P. berghei line, allowing live fluorescence imaging of capsules throughout sporogony and sporozoite excystation. The results show that capsules progressively weaken during sporulation ultimately resulting in sporozoite exit through small holes. Prior to formation of the holes, local thinning of the capsule was observed. Our findings support an excystation model based on local, rather than global, weakening of the capsule likely facilitated by local re-orientation of sporozoites and apical secretion.


Subject(s)
Culicidae , Plasmodium , Animals , Oocysts/metabolism , Sporozoites/metabolism , Plasmodium/metabolism , Animals, Genetically Modified/metabolism , Culicidae/metabolism , Protozoan Proteins/metabolism , Plasmodium berghei/metabolism
13.
Cell Rep ; 42(11): 113419, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37952150

ABSTRACT

Plasmodium parasites contribute to one of the highest global infectious disease burdens. To achieve this success, the parasite has evolved a range of specialized subcellular compartments to extensively remodel the host cell for its survival. The information to fully understand these compartments is likely hidden in the so far poorly characterized Plasmodium species spatial proteome. To address this question, we determined the steady-state subcellular location of more than 12,000 parasite proteins across five different species by extensive subcellular fractionation of erythrocytes infected by Plasmodium falciparum, Plasmodium knowlesi, Plasmodium yoelii, Plasmodium berghei, and Plasmodium chabaudi. This comparison of the pan-species spatial proteomes and their expression patterns indicates increasing species-specific proteins associated with the more external compartments, supporting host adaptations and post-transcriptional regulation. The spatial proteome offers comprehensive insight into the different human, simian, and rodent Plasmodium species, establishing a powerful resource for understanding species-specific host adaptation processes in the parasite.


Subject(s)
Malaria , Proteomics , Humans , Malaria/parasitology , Proteome/metabolism , Plasmodium berghei/metabolism , Erythrocytes/parasitology
14.
Proc Biol Sci ; 290(2011): 20232280, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38018100

ABSTRACT

Vaccination strategies in mice inducing high numbers of memory CD8+ T cells specific to a single epitope are able to provide sterilizing protection against infection with Plasmodium sporozoites. We have recently found that Plasmodium-specific CD8+ T cells cluster around sporozoite-infected hepatocytes but whether such clusters are important in elimination of the parasite remains incompletely understood. Here, we used our previously generated data in which we employed intravital microscopy to longitudinally image 32 green fluorescent protein (GFP)-expressing Plasmodium yoelii parasites in livers of mice that had received activated Plasmodium-specific CD8+ T cells after sporozoite infection. We found significant heterogeneity in the dynamics of the normalized GFP signal from the parasites (termed 'vitality index' or VI) that was weakly correlated with the number of T cells near the parasite. We also found that a simple model assuming mass-action, additive killing by T cells well describes the VI dynamics for most parasites and predicts a highly variable killing efficacy by individual T cells. Given our estimated median per capita kill rate of k = 0.031/h we predict that a single T cell is typically incapable of killing a parasite within the 48 h lifespan of the liver stage in mice. Stochastic simulations of T cell clustering and killing of the liver stage also suggested that: (i) three or more T cells per infected hepatocyte are required to ensure sterilizing protection; (ii) both variability in killing efficacy of individual T cells and resistance to killing by individual parasites may contribute to the observed variability in VI decline, and (iii) the stable VI of some clustered parasites cannot be explained by measurement noise. Taken together, our analysis for the first time provides estimates of efficiency at which individual CD8+ T cells eliminate intracellular parasitic infection in vivo.


Subject(s)
Malaria , Plasmodium yoelii , Mice , Animals , CD8-Positive T-Lymphocytes , Liver/parasitology , Hepatocytes/parasitology , Sporozoites , Plasmodium berghei/metabolism
15.
Proc Natl Acad Sci U S A ; 120(44): e2304339120, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37883438

ABSTRACT

Malaria remains a devastating disease and, with current measures failing to control its transmission, there is a need for novel interventions. A family of proteins that have long been pursued as potential intervention targets are aquaporins, which are channels facilitating the movement of water and other solutes across membranes. We identify an aquaporin in malaria parasites and demonstrate that it is important for completion of Plasmodium development in the mosquito vector. Disruption of AQP2 in the human parasite Plasmodium falciparum and the rodent parasite Plasmodium berghei blocks sporozoite production inside oocysts established on mosquito midguts, greatly limiting parasite infection of salivary glands and transmission to a new host. In vivo epitope tagging of AQP2 in P. berghei, combined with immunofluorescence assays, reveals that the protein is localized in vesicle-like organelles found in the cytoplasm of gametocytes, ookinetes, and sporozoites. The number of these organelles varies between individual parasites and lifecycle stages suggesting that they are likely part of a dynamic endomembrane system. Phylogenetic analysis confirms that AQP2 is unique to malaria and closely related parasites and most closely resembles intracellular aquaporins. Structure prediction analyses identify several unusual features, including a large accessory extracellular loop and an arginine-to-phenylalanine substitution in the selectivity filter principally determining pore function, a unique feature among known aquaporins. This in conjunction with the importance of AQP2 for malaria transmission suggests that AQP2 may be a fruitful target of antimalarial interventions.


Subject(s)
Aquaporin 2 , Mosquito Vectors , Protozoan Proteins , Animals , Malaria , Mosquito Vectors/parasitology , Phylogeny , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Sporozoites/metabolism
16.
Trends Parasitol ; 39(12): 991-995, 2023 12.
Article in English | MEDLINE | ID: mdl-37865610

ABSTRACT

Plasmodium ookinetes and sporozoites were discovered 125 years ago by MacCallum (J. Exp. Med. 1898;3:117-136) and Ross (Ind. Med. Gaz. 1899;34:1-3), respectively. While the migration capacity of ookinetes was noted immediately, the movements of sporozoites remained enigmatic for decades. Today, we know many proteins involved in parasite migration and start to conceptualize a mechanistic understanding of motility.


Subject(s)
Plasmodium , Running , Animals , Sporozoites/metabolism , Plasmodium/metabolism , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism
17.
Antimicrob Agents Chemother ; 67(8): e0035623, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37428074

ABSTRACT

Malaria parasites in the blood stage express a single transmembrane transport protein for the release of the glycolytic end product l-lactate/H+ from the cell. This transporter is a member of the strictly microbial formate-nitrite transporter (FNT) family and a novel putative drug target. Small, drug-like FNT inhibitors potently block lactate transport and kill Plasmodium falciparum parasites in culture. The protein structure of Plasmodium falciparum FNT (PfFNT) in complex with the inhibitor has been resolved and confirms its previously predicted binding site and its mode of action as a substrate analog. Here, we investigated the mutational plasticity and essentiality of the PfFNT target on a genetic level, and established its in vivo druggability using mouse malaria models. We found that, besides a previously identified PfFNT G107S resistance mutation, selection of parasites at 3 × IC50 (50% inhibitory concentration) gave rise to two new point mutations affecting inhibitor binding: G21E and V196L. Conditional knockout and mutation of the PfFNT gene showed essentiality in the blood stage, whereas no phenotypic defects in sexual development were observed. PfFNT inhibitors mainly targeted the trophozoite stage and exhibited high potency in P. berghei- and P. falciparum-infected mice. Their in vivo activity profiles were comparable to that of artesunate, demonstrating strong potential for the further development of PfFNT inhibitors as novel antimalarials.


Subject(s)
Antimalarials , Malaria, Falciparum , Parasites , Animals , Mice , Monocarboxylic Acid Transporters/chemistry , Monocarboxylic Acid Transporters/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Malaria, Falciparum/parasitology , Antimalarials/pharmacology , Antimalarials/chemistry , Parasites/metabolism , Lactates/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism
18.
EMBO Rep ; 24(7): e57064, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37306042

ABSTRACT

Eukaryotic cell adhesion and migration rely on surface adhesins connecting extracellular ligands to the intracellular actin cytoskeleton. Plasmodium sporozoites are transmitted by mosquitoes and rely on adhesion and gliding motility to colonize the salivary glands and to reach the liver after transmission. During gliding, the essential sporozoite adhesin TRAP engages actin filaments in the cytoplasm of the parasite, while binding ligands on the substrate through its inserted (I) domain. Crystal structures of TRAP from different Plasmodium species reveal the I domain in closed and open conformations. Here, we probe the importance of these two conformational states by generating parasites expressing versions of TRAP with the I domain stabilized in either the open or closed state with disulfide bonds. Strikingly, both mutations impact sporozoite gliding, mosquito salivary gland entry, and transmission. Absence of gliding in sporozoites expressing the open TRAP I domain can be partially rescued by adding a reducing agent. This suggests that dynamic conformational change is required for ligand binding, gliding motility, and organ invasion and hence sporozoite transmission from mosquito to mammal.


Subject(s)
Culicidae , Plasmodium , Animals , Sporozoites/metabolism , Ligands , Plasmodium/metabolism , Liver/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Mammals/metabolism
19.
Nat Commun ; 14(1): 1312, 2023 03 10.
Article in English | MEDLINE | ID: mdl-36898988

ABSTRACT

Malaria-causing parasites of the Plasmodium genus undergo multiple developmental phases in the human and the mosquito hosts, regulated by various post-translational modifications. While ubiquitination by multi-component E3 ligases is key to regulate a wide range of cellular processes in eukaryotes, little is known about its role in Plasmodium. Here we show that Plasmodium berghei expresses a conserved SKP1/Cullin1/FBXO1 (SCFFBXO1) complex showing tightly regulated expression and localisation across multiple developmental stages. It is key to cell division for nuclear segregation during schizogony and centrosome partitioning during microgametogenesis. It is additionally required for parasite-specific processes including gamete egress from the host erythrocyte, as well as integrity of the apical and the inner membrane complexes (IMC) in merozoite and ookinete, two structures essential for the dissemination of these motile stages. Ubiquitinomic surveys reveal a large set of proteins ubiquitinated in a FBXO1-dependent manner including proteins important for egress and IMC organisation. We additionally demonstrate an interplay between FBXO1-dependent ubiquitination and phosphorylation via calcium-dependent protein kinase 1. Altogether we show that Plasmodium SCFFBXO1 plays conserved roles in cell division and is also important for parasite-specific processes in the mammalian and mosquito hosts.


Subject(s)
Plasmodium berghei , Humans , Erythrocytes/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Protein Binding , S-Phase Kinase-Associated Proteins/metabolism , Ubiquitination
20.
Cell Rep ; 42(1): 111927, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36640315

ABSTRACT

Plasmodium sporozoites are the motile forms of the malaria parasites that infect hepatocytes. The initial invasion of hepatocytes is thought to be actively driven by sporozoites, but host cell processes might also play a role. Sporozoite invasion triggers a host plasma membrane invagination that forms a vacuole around the intracellular parasite, which is critical for subsequent intracellular parasite replication. Using fast live confocal microscopy, we observed that the initial interactions between sporozoites and hepatocytes induce plasma membrane ruffles and filopodia extensions. Importantly, we find that these host cell processes facilitate invasion and that Rho GTPase signaling, which regulates membrane ruffling and filopodia extension, is critical for productive infection. Interestingly, sporozoite cell traversal stimulates these processes, suggesting that it increases hepatocyte susceptibility to productive infection. Our study identifies host cell signaling events involved in plasma membrane dynamics as a critical host component of successful malaria parasite infection of hepatocytes.


Subject(s)
Malaria , Parasites , Animals , Parasites/metabolism , Protozoan Proteins/metabolism , Hepatocytes/metabolism , Malaria/parasitology , Signal Transduction , Cell Membrane/metabolism , Sporozoites/physiology , Plasmodium berghei/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...