Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
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
2.
Nature ; 612(7940): 534-539, 2022 12.
Article in English | MEDLINE | ID: mdl-36477528

ABSTRACT

An effective vaccine is needed for the prevention and elimination of malaria. The only immunogens that have been shown to have a protective efficacy of more than 90% against human malaria are Plasmodium falciparum (Pf) sporozoites (PfSPZ) manufactured in mosquitoes (mPfSPZ)1-7. The ability to produce PfSPZ in vitro (iPfSPZ) without mosquitoes would substantially enhance the production of PfSPZ vaccines and mosquito-stage malaria research, but this ability is lacking. Here we report the production of hundreds of millions of iPfSPZ. iPfSPZ invaded human hepatocytes in culture and developed to mature liver-stage schizonts expressing P. falciparum merozoite surface protein 1 (PfMSP1) in numbers comparable to mPfSPZ. When injected into FRGhuHep mice containing humanized livers, iPfSPZ invaded the human hepatocytes and developed to PfMSP1-expressing late liver stage parasites at 45% the quantity of cryopreserved mPfSPZ. Human blood from FRGhuHep mice infected with iPfSPZ produced asexual and sexual erythrocytic-stage parasites in culture, and gametocytes developed to PfSPZ when fed to mosquitoes, completing the P. falciparum life cycle from infectious gametocyte to infectious gametocyte without mosquitoes or primates.


Subject(s)
Plasmodium falciparum , Sporozoites , Animals , Humans , Mice , Culicidae/parasitology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/biosynthesis , Malaria Vaccines/chemistry , Malaria, Falciparum/parasitology , Plasmodium falciparum/growth & development , Sporozoites/growth & development , Sporozoites/pathogenicity , Hepatocytes/parasitology , Liver/parasitology , Merozoite Surface Protein 1 , Erythrocytes/parasitology , In Vitro Techniques
3.
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
4.
PLoS Pathog ; 17(11): e1010042, 2021 11.
Article in English | MEDLINE | ID: mdl-34748617

ABSTRACT

Rare and potent monoclonal antibodies (mAbs) against the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) on infective sporozoites (SPZ) preferentially bind the PfCSP junctional tetrapeptide NPDP or NVDP minor repeats while cross-reacting with NANP central repeats in vitro. The extent to which each of these epitopes is required for protection in vivo is unknown. Here, we assessed whether junction-, minor repeat- and central repeat-preferring human mAbs (CIS43, L9 and 317 respectively) bound and protected against in vivo challenge with transgenic P. berghei (Pb) SPZ expressing either PfCSP with the junction and minor repeats knocked out (KO), or PbCSP with the junction and minor repeats knocked in (KI). In vivo protection studies showed that the junction and minor repeats are necessary and sufficient for CIS43 and L9 to neutralize KO and KI SPZ, respectively. In contrast, 317 required major repeats for in vivo protection. These data establish that human mAbs can prevent malaria infection by targeting three different protective epitopes (NPDP, NVDP, NANP) in the PfCSP repeat region. This report will inform vaccine development and the use of mAbs to passively prevent malaria.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Epitopes/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Female , Liver/immunology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred C57BL , Sporozoites/growth & development
5.
Parasitol Int ; 85: 102447, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34474178

ABSTRACT

Plasmodium vivax is the most geographically widespread malaria parasite on the planet. This is largely because after mosquito transmission, P. vivax sporozoites can invade hepatocytes and form latent liver stages known as hypnozoites. These persistent liver stages can activate weeks, months or even years after an infected individual suffers a primary clinical infection. Activation then leads to replication and liver stage schizont maturation that ultimately cause relapse of blood stage infection, disease, and onward transmission. Thus, the latent hypnozoite can lie in wait during times when onward transmission is unlikely due to conditions that do not favor the mosquito. For example, in temperate climates where mosquito prevalence is only seasonal. Furthermore, the elimination of hypnozoites is challenging since the hypnozoite reservoir is currently undetectable and not killed by most antimalarial drugs. Here, we review our current knowledge of the pre-erythrocytic stages of the malaria parasite - the sporozoite and liver stages, including the elusive and enigmatic hypnozoite. We focus on our understanding of sporozoite biology, the novel animal models that are available to study the hypnozoite and hypnozoite activation and the ongoing efforts to understand the biological makeup of the hypnozoite that allow for its persistence in the human host.


Subject(s)
Liver/parasitology , Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Sporozoites/physiology , Animals , Disease Models, Animal , Plasmodium vivax/growth & development , Sporozoites/growth & development
6.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34135099

ABSTRACT

Progress towards a protective vaccine against malaria remains slow. To date, only limited protection has been routinely achieved following immunisation with either whole-parasite (sporozoite) or subunit-based vaccines. One major roadblock to vaccine progress, and to pre-erythrocytic parasite biology in general, is the continued reliance on manual salivary gland dissection for sporozoite isolation from infected mosquitoes. Here, we report development of a multi-step method, based on batch processing of homogenised whole mosquitoes, slurry, and density-gradient filtration, which combined with free-flow electrophoresis rapidly produces a pure, infective sporozoite inoculum. Human-infective Plasmodium falciparum and rodent-infective Plasmodium berghei sporozoites produced in this way are two- to threefold more infective than salivary gland dissection sporozoites in in vitro hepatocyte infection assays. In an in vivo rodent malaria model, the same P. berghei sporozoites confer sterile protection from mosquito-bite challenge when immunisation is delivered intravenously or 60-70% protection when delivered intramuscularly. By improving purity, infectivity, and immunogenicity, this method represents a key advancement in capacity to produce research-grade sporozoites, which should impact delivery of a whole-parasite based malaria vaccine at scale in the future.


Subject(s)
Culicidae/parasitology , Malaria/prevention & control , Plasmodium berghei/pathogenicity , Plasmodium falciparum/pathogenicity , Sporozoites/pathogenicity , Animals , Disease Models, Animal , Drosophila , Hep G2 Cells , Humans , Immunization , Male , Rats , Sporozoites/growth & development
7.
Malar J ; 20(1): 221, 2021 May 18.
Article in English | MEDLINE | ID: mdl-34006297

ABSTRACT

BACKGROUND: Efforts to study the biology of Plasmodium vivax liver stages, particularly the latent hypnozoites, have been hampered by the limited availability of P. vivax sporozoites. Anopheles stephensi is a major urban malaria vector in Goa and elsewhere in South Asia. Using P. vivax patient blood samples, a series of standard membrane-feeding experiments were performed with An. stephensi under the US NIH International Center of Excellence for Malaria Research (ICEMR) for Malaria Evolution in South Asia (MESA). The goal was to understand the dynamics of parasite development in mosquitoes as well as the production of P. vivax sporozoites. To obtain a robust supply of P. vivax sporozoites, mosquito-rearing and mosquito membrane-feeding techniques were optimized, which are described here. METHODS: Membrane-feeding experiments were conducted using both wild and laboratory-colonized An. stephensi mosquitoes and patient-derived P. vivax collected at the Goa Medical College and Hospital. Parasite development to midgut oocysts and salivary gland sporozoites was assessed on days 7 and 14 post-feeding, respectively. The optimal conditions for mosquito rearing and feeding were evaluated to produce high-quality mosquitoes and to yield a high sporozoite rate, respectively. RESULTS: Laboratory-colonized mosquitoes could be starved for a shorter time before successful blood feeding compared with wild-caught mosquitoes. Optimizing the mosquito-rearing methods significantly increased mosquito survival. For mosquito feeding, replacing patient plasma with naïve serum increased sporozoite production > two-fold. With these changes, the sporozoite infection rate was high (> 85%) and resulted in an average of ~ 22,000 sporozoites per mosquito. Some mosquitoes reached up to 73,000 sporozoites. Sporozoite production could not be predicted from gametocyte density but could be predicted by measuring oocyst infection and oocyst load. CONCLUSIONS: Optimized conditions for the production of high-quality P. vivax sporozoite-infected An. stephensi were established at a field site in South West India. This report describes techniques for producing a ready resource of P. vivax sporozoites. The improved protocols can help in future research on the biology of P. vivax liver stages, including hypnozoites, in India, as well as the development of anti-relapse interventions for vivax malaria.


Subject(s)
Anopheles/parasitology , Mosquito Vectors/parasitology , Plasmodium vivax/physiology , Animals , Female , India , Plasmodium vivax/growth & development , Sporozoites/growth & development , Sporozoites/physiology
8.
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
9.
PLoS Pathog ; 17(2): e1009353, 2021 02.
Article in English | MEDLINE | ID: mdl-33626094

ABSTRACT

Repeated blood meals provide essential nutrients for mosquito egg development and routes for pathogen transmission. The target of rapamycin, the TOR pathway, is essential for vitellogenesis. However, its influence on pathogen transmission remains to be elucidated. Here, we show that rapamycin, an inhibitor of the TOR pathway, effectively suppresses Plasmodium berghei infection in Anopheles stephensi. An. stephensi injected with rapamycin or feeding on rapamycin-treated mice showed increased resistance to P. berghei infection. Exposing An. stephensi to a rapamycin-coated surface not only decreased the numbers of both oocysts and sporozoites but also impaired mosquito survival and fecundity. Transcriptome analysis revealed that the inhibitory effect of rapamycin on parasite infection was through the enhanced activation of immune responses, especially the NF-κB transcription factor REL2, a regulator of the immune pathway and complement system. Knockdown of REL2 in rapamycin-treated mosquitoes abrogated the induction of the complement-like proteins TEP1 and SPCLIP1 and abolished rapamycin-mediated refractoriness to Plasmodium infection. Together, these findings demonstrate a key role of the TOR pathway in regulating mosquito immune responses, thereby influencing vector competence.


Subject(s)
Anopheles/drug effects , Immunity, Innate/immunology , Malaria/drug therapy , Mosquito Vectors/drug effects , Plasmodium berghei/pathogenicity , Sirolimus/pharmacology , Animals , Anopheles/immunology , Anopheles/parasitology , Female , Gene Expression Profiling , Immunity, Innate/drug effects , Immunosuppressive Agents/pharmacology , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Mice , Mice, Inbred BALB C , Mosquito Vectors/immunology , Mosquito Vectors/parasitology , Oocysts/drug effects , Oocysts/growth & development , Oocysts/immunology , Sporozoites/drug effects , Sporozoites/growth & development , Sporozoites/immunology
10.
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
11.
Mol Microbiol ; 115(5): 870-881, 2021 05.
Article in English | MEDLINE | ID: mdl-33191548

ABSTRACT

Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.


Subject(s)
Hepatocytes/parasitology , Malaria/parasitology , Plasmodium/physiology , Sporozoites/physiology , Animals , Humans , Liver/parasitology , Plasmodium/genetics , Plasmodium/growth & development , Sporozoites/genetics , Sporozoites/growth & development
12.
Parasit Vectors ; 13(1): 373, 2020 Jul 25.
Article in English | MEDLINE | ID: mdl-32711572

ABSTRACT

BACKGROUND: Avian coccidiosis is a widespread, economically significant disease of poultry, caused by several Eimeria species. These parasites have complex and diverse life-cycles that require invasion of their host cells. This is mediated by various proteins secreted from apical secretory organelles. Apical membrane antigen 1 (AMA1), which is released from micronemes and is conserved across all apicomplexans, plays a central role in the host cell invasion. In a previous study, some putative EtAMA1-interacting proteins of E. tenella were screened. In this study, we characterized one putative EtAMA1-interacting protein, E. tenella Eimeria -specific protein (EtEsp). METHODS: Bimolecular fluorescence complementation (BiFC) and glutathione S-transferase (GST) fusion protein pull-down (GST pull-down) were used to confirm the interaction between EtAMA1 and EtEsp in vivo and in vitro. The expression of EtEsp was analyzed in different developmental stages of E. tenella with quantitative PCR and western blotting. The secretion of EtEsp protein was tested with staurosporine when sporozoites were incubated in complete medium at 41 °C. The localization of EtEsp was analyzed with an immunofluorescence assay (IFA). An in vitro invasion inhibition assay was conducted to assess the ability of antibodies against EtEsp to inhibit cell invasion by E. tenella sporozoites. RESULTS: The interaction between EtAMA1 and EtEsp was confirmed with BiFC and by GST pull-down. Our results show that EtEsp is differentially expressed during distinct phases of the parasite life-cycle. IFA showed that the EtEsp protein is mainly distributed on the parasite surface, and that the expression of this protein increases during the development of the parasite in the host cells. Using staurosporine, we showed that EtEsp is a secreted protein, but not from micronemes. In inhibition tests, a polyclonal anti-rEtEsp antibody attenuated the capacity of E. tenella to invade host cells. CONCLUSION: In this study, we show that EtEsp interacts with EtAMA1 and that the protein is secreted protein, but not from micronemes. The protein participates in sporozoite invasion of host cells and is maybe involved in the growth of the parasite. These data have implications for the use of EtAMA1 or EtAMA1-interacting proteins as targets in intervention strategies against avian coccidiosis.


Subject(s)
Chickens/parasitology , Eimeria/metabolism , Host-Parasite Interactions/physiology , Membrane Proteins/metabolism , Animals , Antigens, Protozoan/metabolism , Coccidiosis/parasitology , Coccidiosis/veterinary , Eimeria/growth & development , Eimeria tenella/growth & development , Eimeria tenella/metabolism , Fluorescent Antibody Technique/methods , Life Cycle Stages/physiology , Poultry Diseases/parasitology , Protozoan Proteins/metabolism , Sporozoites/growth & development , Sporozoites/metabolism
13.
Mol Biochem Parasitol ; 237: 111279, 2020 05.
Article in English | MEDLINE | ID: mdl-32360511

ABSTRACT

Studies of Plasmodium sporozoites and liver stages require dissection of Anopheles mosquitoes to obtain sporozoites for experiments. Sporozoites from the rodent parasite P. yoelii are routinely used to infect hepatocytes for liver stage culture, but sometimes these cultures become contaminated. Using standard microbiological techniques, a single colony type of Gram-negative rod-shaped bacteria was isolated from contaminated cultures. Mass spectrometry and sequencing of the bacterial 16S ribosomal RNA gene identified the contaminant as Elizabethkingia spp. Based on sequence comparison and published studies of the Anopheles microbiome, the best match was E. anophelis. Culture contamination was not ameliorated by density gradient purification of sporozoites. However, the addition of vancomycin to the culture media consistently reduced contamination and improved culture outcomes as measured by liver stage parasite size. Thus, mosquito salivary gland-derived E. anophelis is identified a potential contaminant of Plasmodium liver stage cultures that can be mitigated by the addition of antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Flavobacteriaceae/drug effects , Hepatocytes/drug effects , Plasmodium yoelii/growth & development , Sporozoites/growth & development , Vancomycin/pharmacology , Animals , Anopheles/microbiology , Anopheles/parasitology , Bacterial Typing Techniques , Cells, Cultured , Flavobacteriaceae/genetics , Flavobacteriaceae/growth & development , Flavobacteriaceae/pathogenicity , Hepatocytes/microbiology , Hepatocytes/parasitology , Malaria/parasitology , Microbial Sensitivity Tests , Microbiota/drug effects , Microbiota/genetics , Plasmodium yoelii/ultrastructure , RNA, Ribosomal, 16S/genetics , Rodentia/parasitology , Sporozoites/ultrastructure
14.
Parasitol Res ; 119(2): 667-673, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31836921

ABSTRACT

The development cycle of the malaria parasite, Plasmodium sp., in humans takes place after an infected female Anopheles mosquito injects motile infective forms called sporozoites into the bloodstream. Sporozoites migrate via blood vessels to the liver. This pre-erythrocytic tissue stage is widely accepted to occur in humans exclusively in the liver, contrary to avian malaria where this may occur also in other parenchymatous organs. This concept is based on research conducted by English researchers Henry Shortt and P.C.C. Garnham in the late 1940s. Although Italian researchers as, e.g., Giulio Raffaele, additionally claimed the presence of the parasites in the bone marrow, this is not well acknowledged. So, the question remains whether there exists also a tissue life cycle stage in humans.


Subject(s)
Life Cycle Stages/physiology , Liver/parasitology , Plasmodium/growth & development , Sporozoites/growth & development , Animals , Anopheles/parasitology , Female , Humans , Liver/pathology , Malaria, Avian
15.
Malar J ; 18(1): 426, 2019 Dec 18.
Article in English | MEDLINE | ID: mdl-31849326

ABSTRACT

BACKGROUND: The circumsporozoite protein (CSP) of Plasmodium is a key surface antigen that induces antibodies and T-cells, conferring immune protection in animal models and humans. However, much of the work on CSP and immunity has been developed based on studies using rodent or non-human primate CSP antigens, which may not be entirely translatable to CSP expressed by human malaria parasites, especially considering the host specificity of the different species. METHODS: Using a genetically engineered strain of Plasmodium berghei that expresses luciferase, GFP and the Plasmodium falciparum orthologue of CSP, the effect of laboratory preparation, mosquito treatment and mouse factors on sporozoite infectivity was assessed using an in vivo bioluminescence assay on mice. This assay was compared with a PCR-based protection assay using an already described monoclonal antibody that can provide sterile protection against sporozoite challenge. RESULTS: Bioluminescence assay demonstrated similar detection levels of the quantity and kinetics of liver-stage infection, compared to PCR-based detection. This assay was used to evaluate treatment of sporozoite and delivery method on mouse infectivity, as well as the effects of age, sex and strain of mice. Finally, this assay was used to test the protective capacity of monoclonal antibody AB317; results strongly recapitulate the findings of previous work on this antibody. CONCLUSIONS: The PbGFP-Luc line and in vivo bioluminescence imaging provide highly sensitive read-outs of liver-stage infection in mice, and this method can be useful to reliably evaluate potency of pre-erythrocytic interventions.


Subject(s)
Malaria/immunology , Plasmodium berghei/physiology , Animals , Anopheles/parasitology , Female , High-Throughput Screening Assays , Liver/parasitology , Luciferases/metabolism , Male , Mice , Mice, Inbred BALB C , Microorganisms, Genetically-Modified/genetics , Microorganisms, Genetically-Modified/physiology , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Sporozoites/growth & development
16.
Malar J ; 18(1): 330, 2019 Sep 24.
Article in English | MEDLINE | ID: mdl-31551073

ABSTRACT

BACKGROUND: The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS: In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS: The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS: The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.


Subject(s)
Gene Expression Profiling , Hepatocytes/parasitology , Plasmodium berghei/growth & development , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Erythrocytes/parasitology , Gene Expression Regulation , Genome, Protozoan , Humans , Life Cycle Stages , Liver/parasitology , Malaria/parasitology , Merozoites/genetics , Merozoites/growth & development , Promoter Regions, Genetic , RNA-Seq , Sporozoites/genetics , Sporozoites/growth & development
17.
Parasitol Res ; 118(10): 3043-3051, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31471745

ABSTRACT

Using a combination of morphological and molecular data, we describe a new apicomplexan parasite, Isospora svecica sp. n., from the white-spotted bluethroat, Luscinia svecica cyanecula, from the Czech Republic. Oocysts were found in its intestinal tract. Sporulation was exogenous and took 1-3 days. The oocysts were slightly ellipsoidal, of average size 26.17 × 20.33 µm, with a smooth bilayered wall. Micropyle, oocyst residuum, and polar granules were absent. Sporocysts were bottle-shaped, of an average size of 18.82 × 8.82 µm, with a thin, colourless wall. A conspicuous knob-like Stieda body was present. Substieda body was barely visible. Sporocyst residuum was present in the form of granules of various sizes. Sporozoites were banana-shaped and contained large anterior and small posterior refractile bodies. Partial DNA sequences of three genes were obtained from oocysts of Isospora svecica sp. n., being most closely related to other isosporans described from passerines. Little is known about the parasites of the avian family Muscicapidae, including coccidia, a highly prevalent parasitic protist group in all vertebrate classes. Only six species of the genus Isospora have so far been described in Muscicapidae, together with several "Isospora sp." that in fact most likely represent Isospora lacazei. The newly described Isospora svecica sp. n. differs morphologically from other coccidia reported from muscicapid birds, and represents the first coccidian species described from Luscinia svecica.


Subject(s)
Isospora/classification , Isosporiasis/veterinary , Passeriformes/parasitology , Animals , Czech Republic , Genes, Protozoan/genetics , Intestines/parasitology , Isospora/cytology , Isospora/genetics , Isospora/growth & development , Isosporiasis/parasitology , Oocysts/classification , Oocysts/cytology , Oocysts/genetics , Oocysts/growth & development , Sporozoites/classification , Sporozoites/cytology , Sporozoites/genetics , Sporozoites/growth & development
18.
EMBO J ; 38(15): e100984, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31368598

ABSTRACT

Microtubules are cytoskeletal filaments essential for many cellular processes, including establishment and maintenance of polarity, intracellular transport, division and migration. In most metazoan cells, the number and length of microtubules are highly variable, while they can be precisely defined in some protozoan organisms. However, in either case the significance of these two key parameters for cells is not known. Here, we quantitatively studied the impact of modulating microtubule number and length in Plasmodium, the protozoan parasite causing malaria. Using a gene deletion and replacement strategy targeting one out of two α-tubulin genes, we show that chromosome segregation proceeds in the oocysts even in the absence of microtubules. However, fewer and shorter microtubules severely impaired the formation, motility and infectivity of Plasmodium sporozoites, the forms transmitted by the mosquito, which usually contain 16 microtubules. We found that α-tubulin expression levels directly determined the number of microtubules, suggesting a high nucleation barrier as supported by a mathematical model. Infectious sporozoites were only formed in parasite lines featuring at least 10 microtubules, while parasites with 9 or fewer microtubules failed to transmit.


Subject(s)
Malaria/parasitology , Plasmodium/pathogenicity , Tubulin/genetics , Animals , Gene Deletion , Mice , Models, Theoretical , Plasmodium/genetics , Plasmodium/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Sporozoites/genetics , Sporozoites/growth & development , Sporozoites/pathogenicity , Tubulin/metabolism
19.
mBio ; 10(4)2019 08 06.
Article in English | MEDLINE | ID: mdl-31387905

ABSTRACT

Plasmodium sporozoites (SPZs) must traverse the mosquito salivary glands (SGs) to reach a new vertebrate host and continue the malaria disease cycle. Although SGs can harbor thousands of sporozoites, only 10 to 100 are deposited into a host during probing. To determine how the SGs might function as a bottleneck in SPZ transmission, we have characterized Anopheles stephensi SGs infected with the rodent malaria parasite Plasmodium berghei using immunofluorescence confocal microscopy. Our analyses corroborate findings from previous electron microscopy studies and provide new insights into the invasion process. We identified sites of SPZ accumulation within SGs across a range of infection intensities. Although SPZs were most often seen in the distal lateral SG lobes, they were also observed in the medial and proximal lateral lobes. Most parasites were associated with either the basement membrane or secretory cavities. SPZs accumulated at physical barriers, including fused salivary ducts and extensions of the chitinous salivary duct wall into the distal lumen. SPZs were observed only rarely within salivary ducts. SPZs appeared to contact each other in many different quantities, not just in the previously described large bundles. Within parasite bundles, all of the SPZs were oriented in the same direction. We found that moderate levels of infection did not necessarily correlate with major SG disruptions or abundant SG cell death. Altogether, our findings suggest that SG architecture largely acts as a barrier to SPZ transmission.IMPORTANCE Malaria continues to have a devastating impact on human health. With growing resistance to insecticides and antimalarial drugs, as well as climate change predictions indicating expansion of vector territories, the impact of malaria is likely to increase. Additional insights regarding pathogen migration through vector mosquitoes are needed to develop novel methods to prevent transmission to new hosts. Pathogens, including the microbes that cause malaria, must invade the salivary glands (SGs) for transmission. Since SG traversal is required for parasite transmission, SGs are ideal targets for transmission-blocking strategies. The work presented here highlights the role that mosquito SG architecture plays in limiting parasite traversal, revealing how the SG transmission bottleneck is imposed. Further, our data provide unprecedented detail about SG-sporozoite interactions and gland-to-gland variation not provided in previous studies.


Subject(s)
Anopheles/parasitology , Malaria/transmission , Mosquito Vectors/parasitology , Plasmodium berghei/growth & development , Animals , Anopheles/physiology , Female , Humans , Malaria/parasitology , Male , Mice , Mosquito Vectors/physiology , Plasmodium berghei/physiology , Salivary Glands/parasitology , Sporozoites/growth & development , Sporozoites/physiology
20.
Cell Microbiol ; 21(10): e13082, 2019 10.
Article in English | MEDLINE | ID: mdl-31283102

ABSTRACT

The myosin superfamily comprises of actin-dependent eukaryotic molecular motors important in a variety of cellular functions. Although well studied in many systems, knowledge of their functions in Plasmodium, the causative agent of malaria, is restricted. Previously, six myosins were identified in this genus, including three Class XIV myosins found only in Apicomplexa and some Ciliates. The well characterized MyoA is a Class XIV myosin essential for gliding motility and invasion. Here, we characterize all other Plasmodium myosins throughout the parasite life cycle and show that they have very diverse patterns of expression and cellular location. MyoB and MyoE, the other two Class XIV myosins, are expressed in all invasive stages, with apical and basal locations, respectively. Gene deletion revealed that MyoE is involved in sporozoite traversal, MyoF and MyoK are likely essential in the asexual blood stages, and MyoJ and MyoB are not essential. Both MyoB and its essential light chain (MCL-B) are localised at the apical end of ookinetes but expressed at completely different time points. This work provides a better understanding of the role of actomyosin motors in Apicomplexan parasites, particularly in the motile and invasive stages of Plasmodium during sexual and asexual development within the mosquito.


Subject(s)
Myosins/metabolism , Plasmodium/growth & development , Plasmodium/metabolism , Protozoan Proteins/metabolism , Sporozoites/metabolism , Animals , Female , Life Cycle Stages , Mass Spectrometry , Mice , Myosins/chemistry , Myosins/genetics , Phenotype , Plasmodium/genetics , Protein Domains/genetics , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sporozoites/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...