Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 6145, 2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39034325

ABSTRACT

Parasitic diseases, particularly malaria (caused by Plasmodium falciparum) and theileriosis (caused by Theileria spp.), profoundly impact global health and the socioeconomic well-being of lower-income countries. Despite recent advances, identifying host metabolic proteins essential for these auxotrophic pathogens remains challenging. Here, we generate a novel metabolic model of human hepatocytes infected with P. falciparum and integrate it with a genome-wide CRISPR knockout screen targeting Theileria-infected cells to pinpoint shared vulnerabilities. We identify key host metabolic enzymes critical for the intracellular survival of both of these lethal hemoparasites. Remarkably, among the metabolic proteins identified by our synergistic approach, we find that host purine and heme biosynthetic enzymes are essential for the intracellular survival of P. falciparum and Theileria, while other host enzymes are only essential under certain metabolic conditions, highlighting P. falciparum's adaptability and ability to scavenge nutrients selectively. Unexpectedly, host porphyrins emerge as being essential for both parasites. The shared vulnerabilities open new avenues for developing more effective therapies against these debilitating diseases, with the potential for broader applicability in combating apicomplexan infections.


Subject(s)
CRISPR-Cas Systems , Hepatocytes , Malaria, Falciparum , Plasmodium falciparum , Theileria , Plasmodium falciparum/genetics , Humans , Hepatocytes/parasitology , Hepatocytes/metabolism , Malaria, Falciparum/parasitology , Theileria/genetics , Genomics/methods , Heme/metabolism , Host-Parasite Interactions/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Animals , Gene Knockout Techniques
2.
Comput Struct Biotechnol J ; 24: 334-342, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38690550

ABSTRACT

Malaria, a significant global health challenge, is caused by Plasmodium parasites. The Plasmodium liver stage plays a pivotal role in the establishment of the infection. This study focuses on the liver stage development of the model organism Plasmodium berghei, employing fluorescent microscopy imaging and convolutional neural networks (CNNs) for analysis. Convolutional neural networks have been recently proposed as a viable option for tasks such as malaria detection, prediction of host-pathogen interactions, or drug discovery. Our research aimed to predict the transition of Plasmodium-infected liver cells to the merozoite stage, a key development phase, 15 hours in advance. We collected and analyzed hourly imaging data over a span of at least 38 hours from 400 sequences, encompassing 502 parasites. Our method was compared to human annotations to validate its efficacy. Performance metrics, including the area under the receiver operating characteristic curve (AUC), sensitivity, and specificity, were evaluated on an independent test dataset. The outcomes revealed an AUC of 0.873, a sensitivity of 84.6%, and a specificity of 83.3%, underscoring the potential of our CNN-based framework to predict liver stage development of P. berghei. These findings not only demonstrate the feasibility of our methodology but also could potentially contribute to the broader understanding of parasite biology.

3.
Mol Microbiol ; 121(6): 1095-1111, 2024 06.
Article in English | MEDLINE | ID: mdl-38574236

ABSTRACT

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


Subject(s)
Autophagy-Related Proteins , Autophagy , Hepatocytes , Liver , Microtubule-Associated Proteins , Plasmodium berghei , Vacuolar Proton-Translocating ATPases , Vacuoles , Vacuoles/metabolism , Vacuoles/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Plasmodium berghei/enzymology , Animals , Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Liver/parasitology , Mice , Hepatocytes/parasitology , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Malaria/parasitology , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans
4.
Mol Microbiol ; 121(3): 481-496, 2024 03.
Article in English | MEDLINE | ID: mdl-38009402

ABSTRACT

Plasmodium sporozoite development in and egress from oocysts in the Anopheles mosquito remains largely enigmatic. In a previously performed high-throughput knockout screen, the putative subunit 5 of the prefoldin complex (PbPCS5, PBANKA_0920100) was identified as essential for parasite development during mosquito and liver stage development. Here we generated and analyzed a PbPCS5 knockout parasite line during its development in the mosquito. Interestingly, PbPCS5 deletion does not significantly affect oocyst formation but leads to a growth defect resulting in aberrantly shaped sporozoites. Sporozoites produced in the absence of PbPCS5 were thinner, markedly elongated, and did, in most cases, not contain a nucleus. Sporozoites contained fewer subpellicular microtubules, which reached deep into the sporoblast during sporogony where they contacted and indented nuclei. These aberrantly shaped sporozoites did not reach the salivary glands, and we, therefore, conclude that PbPCS5 is essential for sporogony and the life cycle progression of the parasite during its mosquito stage.


Subject(s)
Anopheles , Molecular Chaperones , Parasites , Animals , Plasmodium berghei/genetics , Oocysts , Sporozoites , Anopheles/parasitology , Protozoan Proteins/genetics , Microtubules
5.
J Cell Sci ; 136(22)2023 11 15.
Article in English | MEDLINE | ID: mdl-37942994

ABSTRACT

Fluorescence and light microscopy are important tools in the history of natural science. However, the resolution of microscopes is limited by the diffraction of light. One possible method to circumvent this physical restriction is the recently developed expansion microscopy (ExM). However, the original ultrastructure ExM (U-ExM) protocol is very time-consuming, and some epitopes are lost during the process. In this study, we developed a shortened pre-gelation staining ExM (PS-ExM) protocol and tested it to investigate the Plasmodium liver stage. The protocol presented in this study allows expanding of pre-stained samples, which results in shorter incubation times, better preservation of some epitopes and the advantage that non-expanded controls can be performed alongside using the same staining protocol. The protocol applicability was accessed throughout the Plasmodium liver stage, showing isotropic five-fold expansion. Furthermore, we used PS-ExM to visualise parasite mitochondria as well as the association of lysosomes to the parasitophorous vacuole membrane (PVM) as an example of visualising host-pathogen interaction. We are convinced that this new tool will be helpful for a deeper understanding of the biology of the Plasmodium liver stage.


Subject(s)
Parasites , Plasmodium , Animals , Microscopy , Liver , Epitopes
6.
Autophagy Rep ; 2(1)2023.
Article in English | MEDLINE | ID: mdl-37064813

ABSTRACT

Pathogenic protists are a group of organisms responsible for causing a variety of human diseases including malaria, sleeping sickness, Chagas disease, leishmaniasis, and toxoplasmosis, among others. These diseases, which affect more than one billion people globally, mainly the poorest populations, are characterized by severe chronic stages and the lack of effective antiparasitic treatment. Parasitic protists display complex life-cycles and go through different cellular transformations in order to adapt to the different hosts they live in. Autophagy, a highly conserved cellular degradation process, has emerged as a key mechanism required for these differentiation processes, as well as other functions that are crucial to parasite fitness. In contrast to yeasts and mammals, protist autophagy is characterized by a modest number of conserved autophagy-related proteins (ATGs) that, even though, can drive the autophagosome formation and degradation. In addition, during their intracellular cycle, the interaction of these pathogens with the host autophagy system plays a crucial role resulting in a beneficial or harmful effect that is important for the outcome of the infection. In this review, we summarize the current state of knowledge on autophagy and other related mechanisms in pathogenic protists and their hosts. We sought to emphasize when, how, and why this process takes place, and the effects it may have on the parasitic cycle. A better understanding of the significance of autophagy for the protist life-cycle will potentially be helpful to design novel anti-parasitic strategies.

7.
PLoS Pathog ; 19(3): e1011210, 2023 03.
Article in English | MEDLINE | ID: mdl-36996035

ABSTRACT

Plasmodium parasites have a complex life cycle alternating between a mosquito and a vertebrate host. Following the bite of an Anopheles female mosquito, Plasmodium sporozoites are transmitted from the skin to the liver; their first place of replication within the host. Successfully invaded sporozoites undergo a massive replication and growth involving asynchronous DNA replication and division that results in the generation of tens of thousands or even hundreds of thousands of merozoites depending on the Plasmodium species. The generation of a high number of daughter parasites requires biogenesis and segregation of organelles to finally reach a relatively synchronous cytokinesis event. At the end of liver stage (LS) development, merozoites are packed into merosomes and released into the bloodstream. They are then liberated and infect red blood cells to again produce merozoites by schizogony for the erythrocytic stage of the life cycle. Although parasite LS and asexual blood stage (ABS) differ in many respects, important similarities exist between the two. This review focuses on the cell division of Plasmodium parasite LS in comparison with other life cycle stages especially the parasite blood stage.


Subject(s)
Liver , Plasmodium , Animals , Cytokinesis , Life Cycle Stages , Liver/parasitology , Merozoites , Plasmodium/physiology , Skin , Sporozoites
8.
NPJ Vaccines ; 7(1): 139, 2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36333336

ABSTRACT

Whole-sporozoite (WSp) malaria vaccines induce protective immune responses in animal malaria models and in humans. A recent clinical trial with a WSp vaccine comprising genetically attenuated parasites (GAP) which arrest growth early in the liver (PfSPZ-GA1), showed that GAPs can be safely administered to humans and immunogenicity is comparable to radiation-attenuated PfSPZ Vaccine. GAPs that arrest late in the liver stage (LA-GAP) have potential for increased potency as shown in rodent malaria models. Here we describe the generation of four putative P. falciparum LA-GAPs, generated by CRISPR/Cas9-mediated gene deletion. One out of four gene-deletion mutants produced sporozoites in sufficient numbers for further preclinical evaluation. This mutant, PfΔmei2, lacking the mei2-like RNA gene, showed late liver growth arrest in human liver-chimeric mice with human erythrocytes, absence of unwanted genetic alterations and sensitivity to antimalarial drugs. These features of PfΔmei2 make it a promising vaccine candidate, supporting further clinical evaluation. PfΔmei2 (GA2) has passed regulatory approval for safety and efficacy testing in humans based on the findings reported in this study.

9.
J Cell Sci ; 134(10)2021 05 15.
Article in English | MEDLINE | ID: mdl-34013963

ABSTRACT

The intracellular lifestyle represents a challenge for the rapidly proliferating liver stage Plasmodium parasite. In order to scavenge host resources, Plasmodium has evolved the ability to target and manipulate host cell organelles. Using dynamic fluorescence-based imaging, we here show an interplay between the pre-erythrocytic stages of Plasmodium berghei and the host cell Golgi during liver stage development. Liver stage schizonts fragment the host cell Golgi into miniaturized stacks, which increases surface interactions with the parasitophorous vacuolar membrane of the parasite. Expression of specific dominant-negative Arf1 and Rab GTPases, which interfere with the host cell Golgi-linked vesicular machinery, results in developmental delay and diminished survival of liver stage parasites. Moreover, functional Rab11a is critical for the ability of the parasites to induce Golgi fragmentation. Altogether, we demonstrate that the structural integrity of the host cell Golgi and Golgi-associated vesicular traffic is important for optimal pre-erythrocytic development of P. berghei. The parasite hijacks the Golgi structure of the hepatocyte to optimize its own intracellular development. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Malaria , Parasites , Animals , Hepatocytes , Liver , Plasmodium berghei , Protozoan Proteins
10.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33762334

ABSTRACT

Circulating levels of the adipokine leptin are linked to neuropathology in experimental cerebral malaria (ECM), but its source and regulation mechanism remain unknown. Here, we show that sequestration of infected red blood cells (iRBCs) in white adipose tissue (WAT) microvasculature increased local vascular permeability and leptin production. Mice infected with parasite strains that fail to sequester in WAT displayed reduced leptin production and protection from ECM. WAT sequestration and leptin induction were lost in CD36KO mice; however, ECM susceptibility revealed sexual dimorphism. Adipocyte leptin was regulated by the mechanistic target of rapamycin complex 1 (mTORC1) and blocked by rapamycin. In humans, although Plasmodium falciparum infection did not increase circulating leptin levels, iRBC sequestration, tissue leptin production, and mTORC1 activity were positively correlated with CM in pediatric postmortem WAT. These data identify WAT sequestration as a trigger for leptin production with potential implications for pathogenesis of malaria infection, prognosis, and treatment.


Subject(s)
Malaria, Cerebral , Parasites , Adipose Tissue/pathology , Animals , Child , Humans , Leptin , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice
11.
Cell Microbiol ; 23(1): e13271, 2021 01.
Article in English | MEDLINE | ID: mdl-32979009

ABSTRACT

The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterised by a long-lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3-phosphate (PI3P)-labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P-labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3-labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.


Subject(s)
Autophagy , Hepatocytes/parasitology , Phosphatidylinositol Phosphates/metabolism , Plasmodium berghei/metabolism , Plasmodium berghei/parasitology , Sporozoites/metabolism , Vacuoles/parasitology , Animals , Cell Line , Female , HeLa Cells , Host-Parasite Interactions , Humans , Malaria/parasitology , Mice , Microtubule-Associated Proteins/metabolism , Organisms, Genetically Modified
12.
mSphere ; 5(6)2020 12 23.
Article in English | MEDLINE | ID: mdl-33361125

ABSTRACT

Toxoplasma gondii and members of the genus Plasmodium are obligate intracellular parasites that leave their infected host cell upon a tightly controlled process of egress. Intracellular replication of the parasites occurs within a parasitophorous vacuole, and its membrane as well as the host plasma membrane need to be disrupted during egress, leading to host cell lysis. While several parasite-derived factors governing egress have been identified, much less is known about host cell factors involved in this process. Previously, RNA interference (RNAi)-based knockdown and antibody-mediated depletion identified a host signaling cascade dependent on guanine nucleotide-binding protein subunit alpha q (GNAQ) to be required for the egress of Toxoplasma tachyzoites and Plasmodium blood stage merozoites. Here, we used CRISPR/Cas9 technology to generate HeLa cells deficient in GNAQ and tested their capacity to support the egress of T. gondii tachyzoites and Plasmodium berghei liver stage parasites. While we were able to confirm the importance of GNAQ for the egress of T. gondii, we found that the egress of P. berghei liver stages was unaffected in the absence of GNAQ. These results may reflect differences between the lytic egress process in apicomplexans and the formation of host cell-derived vesicles termed merosomes by P. berghei liver stages.IMPORTANCE The coordinated release of apicomplexan parasites from infected host cells prior to reinvasion is a critical process for parasite survival and the spread of infection. While Toxoplasma tachyzoites and Plasmodium blood stages induce a fast disruption of their surrounding membranes during their egress from host cells, Plasmodium liver stages keep the host cell membrane intact and leave their host cell in host cell-derived vesicles called merosomes. The knockout of GNAQ, a protein involved in G-protein-coupled receptor signaling, demonstrates the importance of this host factor for the lytic egress of T. gondii tachyzoites. Contrastingly, the egress of P. berghei is independent of GNAQ at the liver stage, indicating the existence of a mechanistically distinct strategy to exit the host cell.


Subject(s)
Cell Membrane/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hepatocytes/parasitology , Plasmodium berghei/physiology , Animals , CRISPR-Cas Systems , HeLa Cells , Humans , Malaria/parasitology , Plasmodium berghei/genetics , Toxoplasma/metabolism
13.
Front Immunol ; 11: 585502, 2020.
Article in English | MEDLINE | ID: mdl-33329563

ABSTRACT

Deciphering the mechanisms by which Plasmodium parasites develop inside hepatocytes is an important step toward the understanding of malaria pathogenesis. We propose that the nature and the magnitude of the inflammatory response in the liver are key for the establishment of the infection. Here, we used mice deficient in the multidrug resistance-2 gene (Mdr2-/-)-encoded phospholipid flippase leading to the development of liver inflammation. Infection of Mdr2-/- mice with Plasmodium berghei ANKA (PbANKA) sporozoites (SPZ) resulted in the blockade of hepatic exo-erythrocytic forms (EEFs) with no further development into blood stage parasites. Interestingly, cultured primary hepatocytes from mutant and wild-type mice are equally effective in supporting EEF development. The abortive infection resulted in a long-lasting immunity in Mdr2-/- mice against infectious SPZ where neutrophils and IL-6 appear as key effector components along with CD8+ and CD4+ effector and central memory T cells. Inflammation-induced breakdown of liver tolerance promotes anti-parasite immunity and provides new approaches for the design of effective vaccines against malaria disease.


Subject(s)
Hepatitis/immunology , Hepatocytes/parasitology , Malaria , ATP Binding Cassette Transporter, Subfamily B/deficiency , Animals , Female , Hepatocytes/immunology , Inflammation/immunology , Liver/immunology , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Mice , Plasmodium berghei , Sporozoites , ATP-Binding Cassette Sub-Family B Member 4
14.
PLoS Pathog ; 16(9): e1008891, 2020 09.
Article in English | MEDLINE | ID: mdl-32956401

ABSTRACT

The transitions between developmental stages are critical points in the Plasmodium life cycle. The development of Plasmodium in the livers of their mammalian hosts bridges malaria transmission and the onset of clinical symptoms elicited by red blood cell infection. The egress of Plasmodium parasites from the liver must be a carefully orchestrated process to ensure a successful switch to the blood stage of infection. Cysteine protease activity is known to be required for liver-stage Plasmodium egress, but the crucial cysteine protease(s) remained unidentified. Here, we characterize a member of the papain-like cysteine protease family, Plasmodium berghei serine repeat antigen 4 (PbSERA4), that is required for efficient initiation of blood-stage infection. Through the generation PbSERA4-specific antisera and the creation of transgenic parasites expressing fluorescently tagged protein, we show that PbSERA4 is expressed and proteolytically processed in the liver and blood stages of infection. Targeted disruption of PbSERA4 results in viable and virulent blood-stage parasites. However, upon transmission from mosquitoes to mice, Pbsera4(-) parasites displayed a reduced capacity to initiate a new round of asexual blood-stage replication. Our results from cultured cells indicate that this defect results from an inability of the PbSERA4-deficient parasites to egress efficiently from infected cells at the culmination of liver-stage development. Protection against infection with wildtype P. berghei could be generated in animals in which Pbsera4(-) parasites failed to establish infection. Our findings confirm that liver-stage merozoite release is an active process and demonstrate that this parasite-encoded cysteine protease contributes to parasite escape from the liver.


Subject(s)
Cysteine Proteases/metabolism , Liver/parasitology , Malaria/enzymology , Plasmodium berghei/enzymology , Protozoan Proteins/metabolism , Animals , Cysteine Proteases/genetics , Liver/metabolism , Malaria/genetics , Mice , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Rats , Rats, Sprague-Dawley
15.
Trends Parasitol ; 36(2): 85-87, 2020 02.
Article in English | MEDLINE | ID: mdl-31883707

ABSTRACT

A recent report by Jennison et al. reveals an important role for plasmepsin V (PMV), an aspartyl protease, in the development of malaria transmission stages. The authors showed that PMV activity is critical for protein export in these stages and that specific PMV inhibitors block parasite transmission to mosquitoes.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Malaria/parasitology , Malaria/transmission , Plasmodium/enzymology , Animals , Antimalarials/pharmacology , Carbamates/pharmacology , Humans , Life Cycle Stages/drug effects , Life Cycle Stages/physiology , Malaria/prevention & control , Oligopeptides/pharmacology , Plasmodium/drug effects , Protein Transport/drug effects
16.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31730853

ABSTRACT

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Subject(s)
Genome, Protozoan , Life Cycle Stages/genetics , Liver/metabolism , Liver/parasitology , Plasmodium berghei/growth & development , Plasmodium berghei/genetics , Alleles , Amino Sugars/biosynthesis , Animals , Culicidae/parasitology , Erythrocytes/parasitology , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Gene Knockout Techniques , Genotype , Models, Biological , Mutation/genetics , Parasites/genetics , Parasites/growth & development , Phenotype , Plasmodium berghei/metabolism , Ploidies , Reproduction
17.
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
18.
Cell Microbiol ; 21(9): e13046, 2019 09.
Article in English | MEDLINE | ID: mdl-31099152

ABSTRACT

The virulence strategy of pathogenic Yersinia spp. involves cell-invasive as well as phagocytosis-preventing tactics to enable efficient colonisation of the host organism. Enteropathogenic yersiniae display an invasive phenotype in early infection stages, which facilitates penetration of the intestinal mucosa. Here we show that invasion of epithelial cells by Yersinia enterocolitica is followed by intracellular survival and multiplication of a subset of ingested bacteria. The replicating bacteria were enclosed in vacuoles with autophagy-related characteristics, showing phagophore formation, xenophagy, and recruitment of cytoplasmic autophagosomes to the bacteria-containing compartments. The subsequent fusion of these vacuoles with lysosomes and concomitant vesicle acidification were actively blocked by Yersinia. This resulted in increased intracellular proliferation and detectable egress of yersiniae from infected cells. Notably, deficiency of the core autophagy machinery component FIP200 impaired the development of autophagic features at Yersinia-containing vacuoles as well as intracellular replication and release of bacteria to the extracellular environment. These results suggest that Y. enterocolitica may take advantage of the macroautophagy pathway in epithelial cells to create an autophagosomal niche that supports intracellular bacterial survival, replication, and, eventually, spread of the bacteria from infected cells.


Subject(s)
Autophagosomes/microbiology , Epithelial Cells/microbiology , Yersinia enterocolitica/pathogenicity , Animals , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Cell Death , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , HeLa Cells , Host Microbial Interactions , Humans , Lysosomes/metabolism , Lysosomes/microbiology , Lysosomes/ultrastructure , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Vacuoles/metabolism , Vacuoles/microbiology , Vacuoles/ultrastructure , Yersinia enterocolitica/growth & development , Yersinia enterocolitica/metabolism
19.
Curr Opin Microbiol ; 46: 93-101, 2018 12.
Article in English | MEDLINE | ID: mdl-30317152

ABSTRACT

The use of rodents as model organisms to study human disease is based on the genetic and physiological similarities between the species. Successful molecular methods to generate transgenic reporter or humanized rodents has rendered rodents as powerful tools for understanding biological processes and host-pathogen interactions relevant to humans. In malaria research, rodent models have been pivotal for the study of liver stages, syndromes arising from blood stages of infection, and malaria transmission to and from the mammalian host. Importantly, many in vivo findings are comparable to pathology observed in humans only when adequate combinations of rodent strains and Plasmodium parasites are used.


Subject(s)
Disease Models, Animal , Malaria/parasitology , Rodentia , Animals , Humans , Plasmodium/genetics , Plasmodium/physiology , Rodentia/parasitology
20.
Sci Adv ; 4(5): eaat3775, 2018 05.
Article in English | MEDLINE | ID: mdl-29806032

ABSTRACT

Transmission of Plasmodium parasites to the mosquito requires the formation and development of gametocytes. Studies in infected humans have shown that only the most mature forms of Plasmodium falciparum gametocytes are present in circulation, whereas immature forms accumulate in the hematopoietic environment of the bone marrow. We used the rodent model Plasmodium berghei to study gametocyte behavior through time under physiological conditions. Intravital microscopy demonstrated preferential homing of early gametocyte forms across the intact vascular barrier of the bone marrow and the spleen early during infection and subsequent development in the extravascular environment. During the acute phase of infection, we observed vascular leakage resulting in further parasite accumulation in this environment. Mature gametocytes showed high deformability and were found entering and exiting the intact vascular barrier. We suggest that extravascular gametocyte localization and mobility are essential for gametocytogenesis and transmission of Plasmodium to the mosquito.


Subject(s)
Bone Marrow/parasitology , Malaria/pathology , Malaria/parasitology , Plasmodium/physiology , Transendothelial and Transepithelial Migration , Animals , Disease Models, Animal , Host-Parasite Interactions , Humans , Mice , Molecular Imaging , Mononuclear Phagocyte System/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL