Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 270
Filter
Add more filters











Publication year range
1.
Commun Biol ; 7(1): 1175, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39294220

ABSTRACT

Biological studies of the determinants of Cryptosporidium infectivity are lacking despite the fact that cryptosporidiosis is a major public health problem. Recently, the 60-kDa glycoprotein (GP60) has received attention because of its high sequence polymorphism and association with host infectivity of isolates and protection against reinfection. However, studies of GP60 function have been hampered by its heavy O-linked glycosylation. Here, we used advanced genetic tools to investigate the processing, fate, and function of GP60. Endogenous gene tagging showed that the GP60 cleavage products, GP40 and GP15, are both highly expressed on the surface of sporozoites, merozoites and male gametes. During invasion, GP40 translocates to the apical end of the zoites and remains detectable at the parasite-host interface. Deletion of the signal peptide, GPI anchor, and GP15 sequences affects the membrane localization of GP40. Deletion of the GP60 gene significantly reduces parasite growth and severity of infection, and replacement of the GP60 gene with sequence from an avirulent isolate reduces the pathogenicity of a highly infective isolate. These results have revealed dynamic changes in GP60 expression during parasite development. They further suggest that GP60 is a key protein mediating host infectivity and pathogenicity.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Protozoan Proteins , Cryptosporidium parvum/genetics , Cryptosporidium parvum/pathogenicity , Cryptosporidium parvum/metabolism , Animals , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Cryptosporidiosis/parasitology , Host-Parasite Interactions , Mice , Humans , Sporozoites/metabolism , Sporozoites/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism
2.
Front Cell Infect Microbiol ; 14: 1438019, 2024.
Article in English | MEDLINE | ID: mdl-39149419

ABSTRACT

The malaria-causing parasites have to complete a complex infection cycle in the mosquito vector that also involves attack by the insect's innate immune system, especially at the early stages of midgut infection. However, Anopheles immunity to the late Plasmodium sporogonic stages, such as oocysts, has received little attention as they are considered to be concealed from immune factors due to their location under the midgut basal lamina and for harboring an elaborate cell wall comprising an external layer derived from the basal lamina that confers self-properties to an otherwise foreign structure. Here, we investigated whether Plasmodium berghei oocysts and sporozoites are susceptible to melanization-based immunity in Anopheles gambiae. Silencing of the negative regulator of melanization response, CLIPA14, increased melanization prevalence without significantly increasing the numbers of melanized oocysts, while co-silencing CLIPA14 with CLIPA2, a second negative regulator of melanization, resulted in a significant increase in melanized oocysts and melanization prevalence. Only late-stage oocysts were found to be melanized, suggesting that oocyst rupture was a prerequisite for melanization-based immune attack, presumably due to the loss of the immune-evasive features of their wall. We also found melanized sporozoites inside oocysts and in the hemocoel, suggesting that sporozoites at different maturation stages are susceptible to melanization. Silencing the melanization promoting factors TEP1 and CLIPA28 rescued oocyst melanization in CLIPA2/CLIPA14 co-silenced mosquitoes. Interestingly, silencing of CTL4, that protects early stage ookinetes from melanization, had no effect on oocysts and sporozoites, indicating differential regulation of immunity to early and late sporogonic stages. Similar to previous studies addressing ookinete stage melanization, the melanization of Plasmodium falciparum oocysts was significantly lower than that observed for P. berghei. In summary, our results provide conclusive evidence that late sporogonic malaria parasite stages are susceptible to melanization, and we reveal distinct regulatory mechanisms for ookinete and oocyst melanization.


Subject(s)
Anopheles , Melanins , Oocysts , Plasmodium berghei , Sporozoites , Animals , Anopheles/parasitology , Anopheles/immunology , Plasmodium berghei/immunology , Oocysts/metabolism , Melanins/metabolism , Sporozoites/immunology , Sporozoites/metabolism , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Insect Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/immunology , Malaria/immunology , Malaria/parasitology , Gene Silencing , Immunity, Innate , Female
3.
Parasite ; 31: 51, 2024.
Article in English | MEDLINE | ID: mdl-39212528

ABSTRACT

Cryptosporidium is a globally distributed zoonotic protozoan parasite that can cause severe diarrhea in humans and animals. L-type lectins are carbohydrate-binding proteins involved in multiple pathways in animals and plants, including protein transportation, secretion, innate immunity, and the unfolded protein response signaling pathway. However, the biological function of the L-type lectins remains unknown in Cryptosporidium parvum. Here, we preliminarily characterized an L-type lectin in C. parvum (CpLTL) that contains a lectin-leg-like domain. Immunofluorescence assay confirmed that CpLTL is located on the wall of oocysts, the surface of the mid-anterior region of the sporozoite and the cytoplasm of merozoites. The involvement of CpLTL in parasite invasion is partly supported by experiments showing that an anti-CpLTL antibody could partially block the invasion of C. parvum sporozoites into host cells. Moreover, the recombinant CpLTL showed binding ability with mannose and the surface of host cells, and competitively inhibited the invasion of C. parvum. Two host cell proteins were identified by proteomics which should be prioritized for future validation of CpLTL-binding. Our data indicated that CpLTL is potentially involved in the adhesion and invasion of C. parvum.


Title: Une protéine mono-transmembranaire, lectine de type L spécifique du mannose, potentiellement impliquée dans l'adhésion et l'invasion de Cryptosporidium parvum. Abstract: Cryptosporidium est un parasite protozoaire zoonotique répandu dans le monde entier qui peut provoquer de graves diarrhées chez les humains et les animaux. Les lectines de type L sont des protéines liant les glucides impliquées dans de multiples voies chez les animaux et les plantes, notamment le transport des protéines, la sécrétion, l'immunité innée et la voie de signalisation de la réponse protéique dépliée. Cependant, la fonction biologique des lectines de type L reste inconnue chez Cryptosporidium parvum. Ici, nous avons caractérisé de manière préliminaire une lectine de type L chez C. parvum (CpLTL) qui contient un domaine de type jambe de lectine. Le test d'immunofluorescence a confirmé que CpLTL est localisée sur la paroi des oocystes, la surface de la région médio-antérieure du sporozoïte et le cytoplasme des mérozoïtes. L'implication de CpLTL dans l'invasion parasitaire est en partie étayée par des expériences montrant qu'un anticorps anti-CpLTL peut bloquer partiellement l'invasion des sporozoïtes de C. parvum dans les cellules hôtes. De plus, la CpLTL recombinante a montré une capacité de liaison avec le mannose et la surface des cellules hôtes et a inhibé de manière compétitive l'invasion de C. parvum. Deux protéines de cellules hôtes ont été identifiées par protéomique et devraient être prioritaires pour la validation future de la liaison avec CpLTL. Nos données indiquent que CpLTL est potentiellement impliquée dans l'adhésion et l'invasion de C. parvum.


Subject(s)
Cryptosporidium parvum , Mannose , Protozoan Proteins , Sporozoites , Cryptosporidium parvum/physiology , Cryptosporidium parvum/metabolism , Cryptosporidium parvum/genetics , Sporozoites/physiology , Sporozoites/metabolism , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans , Mannose/metabolism , Oocysts/physiology , Cryptosporidiosis/parasitology , Merozoites/physiology , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Adhesion , Proteomics
4.
J Biol Chem ; 300(8): 107557, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39002668

ABSTRACT

Glycosylphosphatidylinositol (GPI) anchor protein modification in Plasmodium species is well known and represents the principal form of glycosylation in these organisms. The structure and biosynthesis of GPI anchors of Plasmodium spp. has been primarily studied in the asexual blood stage of Plasmodium falciparum and is known to contain the typical conserved GPI structure of EtN-P-Man3GlcN-PI. Here, we have investigated the circumsporozoite protein (CSP) for the presence of a GPI anchor. CSP is the major surface protein of Plasmodium sporozoites, the infective stage of the malaria parasite. While it is widely assumed that CSP is a GPI-anchored cell surface protein, compelling biochemical evidence for this supposition is absent. Here, we employed metabolic labeling and mass-spectrometry-based approaches to confirm the presence of a GPI anchor in CSP. Biosynthetic radiolabeling of CSP with [3H]-palmitic acid and [3H]-ethanolamine, with the former being base-labile and therefore ester-linked, provided strong evidence for the presence of a GPI anchor on CSP, but these data alone were not definitive. To provide further evidence, immunoprecipitated CSP was analyzed for the presence of myo-inositol (a characteristic component of GPI anchor) using strong acid hydrolysis and GC-MS for highly sensitive and quantitative detection. The single ion monitoring (SIM) method for GC-MS analysis confirmed the presence of the myo-inositol component in CSP. Taken together, these data provide confidence that the long-assumed presence of a GPI anchor on this important parasite protein is correct.


Subject(s)
Cell Membrane , Glycosylphosphatidylinositols , Plasmodium falciparum , Protozoan Proteins , Sporozoites , Protozoan Proteins/metabolism , Glycosylphosphatidylinositols/metabolism , Glycosylphosphatidylinositols/chemistry , Cell Membrane/metabolism , Sporozoites/metabolism , Plasmodium falciparum/metabolism , Animals , Membrane Proteins/metabolism , Humans
5.
Proc Natl Acad Sci U S A ; 121(28): e2403442121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968107

ABSTRACT

Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.


Subject(s)
Hepatocytes , Liver , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Sporozoites , Plasmodium falciparum/growth & development , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Sporozoites/metabolism , Sporozoites/growth & development , Mice , Liver/parasitology , Liver/metabolism , Humans , Hepatocytes/parasitology , Hepatocytes/metabolism , Malaria, Falciparum/parasitology
6.
J Cell Sci ; 137(11)2024 06 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
7.
Malar J ; 23(1): 151, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755636

ABSTRACT

BACKGROUND: Sporozoite invasion of hepatocytes is an essential step in the Plasmodium life-cycle and has similarities, at the cellular level, to merozoite invasion of erythrocytes. In the case of the Plasmodium blood-stage, efforts to identify host-pathogen protein-protein interactions have yielded important insights including vaccine candidates. In the case of sporozoite-hepatocyte invasion, the host-pathogen protein-protein interactions involved are poorly understood. METHODS: To gain a better understanding of the protein-protein interaction between the sporozoite ligands and host receptors, a systematic screen was performed. The previous Plasmodium falciparum and human surface protein ectodomain libraries were substantially extended, resulting in the creation of new libraries comprising 88 P. falciparum sporozoite protein coding sequences and 182 sequences encoding human hepatocyte surface proteins. Having expressed recombinant proteins from these sequences, a plate-based assay was used, capable of detecting low affinity interactions between recombinant proteins, modified for enhanced throughput, to screen the proteins for interactions. The novel interactions identified in the screen were characterized biochemically, and their essential role in parasite invasion was further elucidated using antibodies and genetically manipulated Plasmodium parasites. RESULTS: A total of 7540 sporozoite-hepatocyte protein pairs were tested under conditions capable of detecting interactions of at least 1.2 µM KD. An interaction between the human fibroblast growth factor receptor 4 (FGFR4) and the P. falciparum protein Pf34 is identified and reported here, characterizing its affinity and demonstrating the blockade of the interaction by reagents, including a monoclonal antibody. Furthermore, further interactions between Pf34 and a second P. falciparum rhoptry neck protein, PfRON6, and between human low-density lipoprotein receptor (LDLR) and the P. falciparum protein PIESP15 are identified. Conditional genetic deletion confirmed the essentiality of PfRON6 in the blood-stage, consistent with the important role of this protein in parasite lifecycle. Pf34 was refractory to attempted genetic modification. Antibodies to Pf34 abrogated the interaction and had a modest effect upon sporozoite invasion into primary human hepatocytes. CONCLUSION: Pf34 and PfRON6 may be members of a functionally important invasion complex which could be a target for future interventions. The modified interaction screening assay, protein expression libraries and P. falciparum mutant parasites reported here may be a useful tool for protein interaction discovery and antigen candidate screening which could be of wider value to the scientific community.


Subject(s)
Hepatocytes , Plasmodium falciparum , Protozoan Proteins , Sporozoites , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Hepatocytes/parasitology , Humans , Sporozoites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Host-Pathogen Interactions , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Host-Parasite Interactions , Protein Binding
8.
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
9.
Front Cell Infect Microbiol ; 14: 1367359, 2024.
Article in English | MEDLINE | ID: mdl-38660488

ABSTRACT

Cryptosporidium parvum is a common cause of a zoonotic disease and a main cause of diarrhea in newborns. Effective drugs or vaccines are still lacking. Oocyst is the infective form of the parasite; after its ingestion, the oocyst excysts and releases four sporozoites into the host intestine that rapidly attack the enterocytes. The membrane protein CpRom1 is a large rhomboid protease that is expressed by sporozoites and recognized as antigen by the host immune system. In this study, we observed the release of CpRom1 with extracellular vesicles (EVs) that was not previously described. To investigate this phenomenon, we isolated and resolved EVs from the excystation medium by differential ultracentrifugation. Fluorescence flow cytometry and transmission electron microscopy (TEM) experiments identified two types of sporozoite-derived vesicles: large extracellular vesicles (LEVs) and small extracellular vesicles (SEVs). Nanoparticle tracking analysis (NTA) revealed mode diameter of 181 nm for LEVs and 105 nm for SEVs, respectively. Immunodetection experiments proved the presence of CpRom1 and the Golgi protein CpGRASP in LEVs, while immune-electron microscopy trials demonstrated the localization of CpRom1 on the LEVs surface. TEM and scanning electron microscopy (SEM) showed that LEVs were generated by means of the budding of the outer membrane of sporozoites; conversely, the origin of SEVs remained uncertain. Distinct protein compositions were observed between LEVs and SEVs as evidenced by their corresponding electrophoretic profiles. Indeed, a dedicated proteomic analysis identified 5 and 16 proteins unique for LEVs and SEVs, respectively. Overall, 60 proteins were identified in the proteome of both types of vesicles and most of these proteins (48 in number) were already identified in the molecular cargo of extracellular vesicles from other organisms. Noteworthy, we identified 12 proteins unique to Cryptosporidium spp. and this last group included the immunodominant parasite antigen glycoprotein GP60, which is one of the most abundant proteins in both LEVs and SEVs.


Subject(s)
Cryptosporidium parvum , Extracellular Vesicles , Protozoan Proteins , Sporozoites , Extracellular Vesicles/metabolism , Cryptosporidium parvum/metabolism , Sporozoites/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/analysis , Microscopy, Electron, Transmission , Animals , Cryptosporidiosis/parasitology , Humans , Proteome/analysis , Proteomics , Flow Cytometry
10.
Parasit Vectors ; 17(1): 146, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38504274

ABSTRACT

BACKGROUND: Cryptosporidium parvum is an apicomplexan zoonotic parasite causing the diarrheal illness cryptosporidiosis in humans and animals. To invade the host intestinal epithelial cells, parasitic proteins expressed on the surface of sporozoites interact with host cells to facilitate the formation of parasitophorous vacuole for the parasite to reside and develop. The gp40 of C. parvum, named Cpgp40 and located on the surface of sporozoites, was proven to participate in the process of host cell invasion. METHODS: We utilized the purified Cpgp40 as a bait to obtain host cell proteins interacting with Cpgp40 through the glutathione S-transferase (GST) pull-down method. In vitro analysis, through bimolecular fluorescence complementation assay (BiFC) and coimmunoprecipitation (Co-IP), confirmed the solid interaction between Cpgp40 and ENO1. In addition, by using protein mutation and parasite infection rate analysis, it was demonstrated that ENO1 plays an important role in the C. parvum invasion of HCT-8 cells. RESULTS: To illustrate the functional activity of Cpgp40 interacting with host cells, we identified the alpha-enolase protein (ENO1) from HCT-8 cells, which showed direct interaction with Cpgp40. The mRNA level of ENO1 gene was significantly decreased at 3 and 24 h after C. parvum infection. Antibodies and siRNA specific to ENO1 showed the ability to neutralize C. parvum infection in vitro, which indicated the participation of ENO1 during the parasite invasion of HCT-8 cells. In addition, we further demonstrated that ENO1 protein was involved in the regulation of cytoplasmic matrix of HCT-8 cells during C. parvum invasion. Functional study of the protein mutation illustrated that ENO1 was also required for the endogenous development of C. parvum. CONCLUSIONS: In this study, we utilized the purified Cpgp40 as a bait to obtain host cell proteins ENO1 interacting with Cpgp40. Functional studies illustrated that the host cell protein ENO1 was involved in the regulation of tight junction and adherent junction proteins during C. parvum invasion and was required for endogenous development of C. parvum.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Humans , Animals , Cryptosporidium parvum/genetics , Cryptosporidiosis/parasitology , Sporozoites/metabolism , Protozoan Proteins/metabolism , Membrane Proteins/metabolism , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , DNA-Binding Proteins/metabolism , Biomarkers, Tumor/metabolism , Tumor Suppressor Proteins/metabolism
11.
Sci Rep ; 14(1): 4851, 2024 02 28.
Article in English | MEDLINE | ID: mdl-38418588

ABSTRACT

Eimeria species serve as promising eukaryotic vaccine vectors. And that the location of heterologous antigens in the subcellular components of genetically modified Eimeria may determine the magnitude and type of immune responses. Therefore, our study aimed to target a heterologous fluorescent protein to the cell surface or microneme, two locations where are more effective in inducing protective immunity, of Eimeria tenella and E. acervulina sporozoites. We used an enhanced yellow fluorescent protein (EYFP) as a tagging biomarker, fusing variously with some localization or whole sequences of compartmental proteins for targeting. After acquiring stable transgenic Eimeria populations, we observed EYFP expressing in expected locations with certain strategies. That is, EYFP successfully localized to the surface when it was fused between signal peptides and mature products of surface antigen 1 (SAG1). Furthermore, EYFP was efficiently targeted to the apical end, an optimal location for secretory organelle known as the microneme, when fused to the C terminus of microneme protein 2. Unexpectedly, EYFP exhibited dominantly in the apical end with only weak expression on the surface of the transgenic sporozoites when the parasites were transfected with plasmid with EYFP fused between signal peptides and mature products of E. tenella SAG 13. These strategies worked in both E. tenella and E. acervulina, laying a solid foundation for studying E. tenella and E. acervulina-based live vaccines that can be further tailored to the inclusion of cargo immunogens from other pathogens.


Subject(s)
Coccidiosis , Eimeria , Parasites , Poultry Diseases , Animals , Coccidiosis/parasitology , Animals, Genetically Modified , Protein Sorting Signals , Sporozoites/metabolism , Chickens/parasitology
12.
Mol Cell Proteomics ; 23(3): 100736, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38342407

ABSTRACT

The oocyst is a sporogonic stage of Plasmodium development that takes place in the mosquito midgut in about 2 weeks. The cyst is protected by a capsule of unknown composition, and little is known about oocyst biology. We carried out a proteomic analysis of oocyst samples isolated at early, mid, and late time points of development. Four biological replicates for each time point were analyzed, and almost 600 oocyst-specific candidates were identified. The analysis revealed that, in young oocysts, there is a strong activity of protein and DNA synthesis, whereas in mature oocysts, proteins involved in oocyst and sporozoite development, gliding motility, and invasion are mostly abundant. Among the proteins identified at early stages, 17 candidates are specific to young oocysts. Thirty-four candidates are common to oocyst and the merosome stages (sporozoite proteins excluded), sharing common features as replication and egress. Western blot and immunofluorescence analyses of selected candidates confirm the expression profile obtained by proteomic analysis.


Subject(s)
Anopheles , Plasmodium , Animals , Oocysts/metabolism , Proteomics , Sporozoites/metabolism , Protozoan Proteins/metabolism
13.
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
14.
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
15.
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
16.
Sci Rep ; 14(1): 1260, 2024 01 13.
Article in English | MEDLINE | ID: mdl-38218737

ABSTRACT

In Plasmodium vivax, the most studied vaccine antigens are aimed at blocking merozoite invasion of erythrocytes and disease development. Very few studies have evaluated pre-erythrocytic (PE) stage antigens. The P. vivax circumsporozoite protein (CSP), is considered the leading PE vaccine candidate, but immunity to CSP is short-lived and variant specific. Thus, there is a need to identify other potential candidates to partner with CSP in a multivalent vaccine to protect against infection and disease. We hypothesize that sporozoite antigens important for host cell infection are considered potential targets. In this study, we evaluated the magnitude and quality of naturally acquired antibody responses to four P. vivax PE antigens: sporozoite surface protein 3 (SSP3), sporozoite protein essential for traversal 1 (SPECT1), cell traversal protein of ookinetes and sporozoites (CelTOS) and CSP in plasma of P. vivax infected patients from Thailand. Naturally acquired antibodies to these antigens were prevalent in the study subjects, but with significant differences in magnitude of IgG antibody responses. About 80% of study participants had antibodies to all four antigens and only 2% did not have antibodies to any of the antigens. Most importantly, these antibodies inhibited sporozoite infection of hepatocytes in vitro. Significant variations in magnitude of antigen-specific inhibitory antibody responses were observed with individual samples. The highest inhibitory responses were observed with anti-CelTOS antibodies, followed by anti-SPECT1, SSP3 and CSP antibodies respectively. These data highlight the vaccine potential of these antigens in protecting against hepatocyte infection and the need for a multi-valent pre-erythrocytic vaccine to prevent liver stage development of P. vivax sporozoites.


Subject(s)
Malaria, Vivax , Vaccines , Animals , Humans , Plasmodium vivax , Sporozoites/metabolism , Protozoan Proteins/metabolism , Antigens, Protozoan , Membrane Proteins/metabolism , Erythrocytes/metabolism , Hepatocytes/metabolism , Antibodies, Protozoan , Plasmodium falciparum/metabolism
17.
mBio ; 15(2): e0315823, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38265238

ABSTRACT

The zoonotic Cryptosporidium parvum is a global contributor to infantile diarrheal diseases and opportunistic infections in immunocompromised or weakened individuals. Like other apicomplexans, it possesses several specialized secretory organelles, including micronemes, rhoptry, and dense granules. However, the understanding of cryptosporidial micronemal composition and secretory pathway remains limited. Here, we report a new micronemal protein in C. parvum, namely, thrombospondin (TSP)-repeat domain-containing protein-4 (CpTSP4), providing insights into these ambiguities. Immunostaining and enzyme-linked assays show that CpTSP4 is prestored in the micronemes of unexcysted sporozoites but secreted during sporozoite excystation, gliding, and invasion. In excysted sporozoites, CpTSP4 is also distributed on the two central microtubules unique to Cryptosporidium. The secretion and microtubular distribution could be completely blocked by the selective kinesin-5 inhibitors SB-743921 and SB-715992, resulting in the accumulation of CpTSP4 in micronemes. These support the kinesin-dependent microtubular trafficking of CpTSP4 for secretion. We also localize γ-tubulin, consistent with kinesin-dependent anterograde trafficking. Additionally, recombinant CpTSP4 displays nanomolar binding affinity to the host cell surface, for which heparin acts as one of the host ligands. A novel heparin-binding motif is identified and validated biochemically for its contribution to the adhesive property of CpTSP4 by peptide competition assays and site-directed mutagenesis. These findings shed light on the mechanisms of intracellular trafficking and secretion of a cryptosporidial micronemal protein and the interaction of a TSP-family protein with host cells.IMPORTANCECryptosporidium parvum is a globally distributed apicomplexan parasite infecting humans and/or animals. Like other apicomplexans, it possesses specialized secretory organelles in the zoites, in which micronemes discharge molecules to facilitate the movement and invasion of zoites. Although past and recent studies have identified several proteins in cryptosporidial micronemes, our understanding of the composition, secretory pathways, and domain-ligand interactions of micronemal proteins remains limited. This study identifies a new micronemal protein, namely, CpTSP4, that is discharged during excystation, gliding, and invasion of C. parvum sporozoites. The CpTSP4 secretion depends on the intracellular trafficking on the two Cryptosporidium-unique microtubes that could be blocked by kinesin-5/Eg5 inhibitors. Additionally, a novel heparin-binding motif is identified and biochemically validated, which contributes to the nanomolar binding affinity of CpTSP4 to host cells. These findings indicate that kinesin-dependent microtubular trafficking is critical to CpTSP4 secretion, and heparin/heparan sulfate is one of the ligands for this micronemal protein.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Humans , Animals , Cryptosporidium parvum/metabolism , Cryptosporidiosis/metabolism , Kinesins/metabolism , Microtubules/metabolism , Sporozoites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Heparin/metabolism
18.
Genomics ; 116(2): 110792, 2024 03.
Article in English | MEDLINE | ID: mdl-38215860

ABSTRACT

Eimeria tenella is the main pathogen responsible for coccidiosis in chickens. The life cycle of E. tenella is, arguably, the least complex of all Coccidia, with only one host. However, it presents different developmental stages, either in the environment or in the host and either intracellular or extracellular. Its signaling and metabolic pathways change with its different developmental stages. Until now, little is known about the developmental regulation and transformation mechanisms of its life cycle. In this study, protein profiles from the five developmental stages, including unsporulated oocysts (USO), partially sporulated (7 h) oocysts (SO7h), sporulated oocysts (SO), sporozoites (S) and second-generation merozoites (M2), were harvested using the label-free quantitative proteomics approach. Then the differentially expressed proteins (DEPs) for these stages were identified. A total of 314, 432, 689, and 665 DEPs were identified from the comparison of SO7h vs USO, SO vs SO7h, S vs SO, and M2 vs S, respectively. By conducting weighted gene coexpression network analysis (WGCNA), six modules were dissected. Proteins in blue and brown modules were calculated to be significantly positively correlated with the E. tenella developmental stages of sporozoites (S) and second-generation merozoites (M2), respectively. In addition, hub proteins with high intra-module degree were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway enrichment analyses revealed that hub proteins in blue modules were involved in electron transport chain and oxidative phosphorylation. Hub proteins in the brown module were involved in RNA splicing. These findings provide new clues and ideas to enhance our fundamental understanding of the molecular mechanisms underlying parasite development.


Subject(s)
Eimeria tenella , Animals , Eimeria tenella/genetics , Proteomics , Chickens/parasitology , Oocysts/physiology , Sporozoites/genetics , Sporozoites/metabolism , Life Cycle Stages
19.
Microb Pathog ; 188: 106549, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38281605

ABSTRACT

The five epidermal growth factor-like domains (EGF) of Eimeria tenella microneme protein 8 (EtMIC8) (EtMIC8-EGF) plays a vital role in host cell attachment and invasion. These processes require interactions between parasite proteins and receptors on the surface of host cells. In this study, five chicken membrane proteins potentially interacting with EtMIC8-EGF were identified using the GST pull-down assay and mass spectrometry analysis, and only chicken (Gallus gallus) epithelial cell adhesion molecule (EPCAM) could bind to EtMIC8-EGF. EPCAM-specific antibody and recombinant EPCAM protein (rEPCAM) inhibited the EtMIC8-EGF binding to host cells in a concentration-dependent manner. Furthermore, the rEPCAM protein showed a binding activity to sporozoites in vitro, and a significant reduction of E. tenella invasion in DF-1 cells was further observed after pre-incubation of sporozoites with rEPCAM. The specific anti-EPCAM antibody further significantly decreased weight loss, lesion score and oocyst output during E. tenella infection, displaying partial inhibition of E. tenella infection. These results indicate that chicken EPCAM is an important EtMIC8-interacting host protein involved in E. tenella-host cell adhesion and invasion. The findings will contribute to a better understanding of the role of adhesion-associated microneme proteins in E. tenella.


Subject(s)
Coccidiosis , Eimeria tenella , Poultry Diseases , Animals , Eimeria tenella/chemistry , Eimeria tenella/metabolism , Epithelial Cell Adhesion Molecule/metabolism , Chickens , Protozoan Proteins , Epidermal Growth Factor/metabolism , Recombinant Proteins , Sporozoites/metabolism , Coccidiosis/veterinary , Coccidiosis/parasitology , Poultry Diseases/parasitology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL