Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters










Publication year range
1.
Parasit Vectors ; 14(1): 373, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34289894

ABSTRACT

BACKGROUND: Plasmodium sp., which causes malaria, must first develop in mosquitoes before being transmitted. Upon ingesting infected blood, gametes form in the mosquito lumen, followed by fertilization and differentiation of the resulting zygotes into motile ookinetes. Within 24 h of blood ingestion, these ookinetes traverse mosquito epithelial cells and lodge below the midgut basal lamina, where they differentiate into sessile oocysts that are protected by a capsule. METHODS: We identified an ookinete surface and oocyst capsule protein (OSCP) that is involved in ookinete motility as well as oocyst capsule formation. RESULTS: We found that knockout of OSCP in parasite decreases ookinete gliding motility and gradually reduces the number of oocysts. On day 15 after blood ingestion, the oocyst wall was significantly thinner. Moreover, adding anti-OSCP antibodies decreased the gliding speed of wild-type ookinetes in vitro. Adding anti-OSCP antibodies to an infected blood meal also resulted in decreased oocyst formation. CONCLUSION: These findings may be useful for the development of a transmission-blocking tool for malaria.


Subject(s)
Antibodies, Protozoan/immunology , Culicidae/parasitology , Malaria/parasitology , Mosquito Vectors/parasitology , Plasmodium berghei/physiology , Protozoan Proteins/metabolism , Animals , Female , Malaria/prevention & control , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Oocysts , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Plasmodium berghei/ultrastructure , Protozoan Proteins/genetics
2.
Zoolog Sci ; 38(2): 187-192, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33812358

ABSTRACT

The ultrastructural features of axoneme organization within the cytoplasm and exflagellation were investigated in detail in microgametes of a malaria parasite, Plasmodium berghei, by electron and fluorescence microscopy. The kinetosomes (basal bodies) of the microgamete were characterized by an electron dense mass in which singlet microtubules (MTs) were embedded. Around the kinetosomes, several singlet and doublet MTs were recognized in transverse sections. Incomplete doublets with growing B-tubule were also observed. As precursors of the axoneme, arrays of over three doublets showed a tendency to encircle the central pair MTs. Some of the doublet MTs were already equipped with inner and outer dynein arms. In the microgamete, which lacks an intraflagellar transport (IFT) system, self-assembly of microtubular and associated components appeared to proceed stepwise from singlet MTs through arrays of one to nine doublet MTs, surrounding the central pair, to form the complete axoneme in a quite short time. At exflagellation, some extra doublets were occasionally included between the axoneme and the flagellar membrane. At high magnification, the outer dynein arm of the Plasmodium microgamete had a pistol-like shape representing a three-headed dynein molecule like that of other Alveolata.


Subject(s)
Axoneme/ultrastructure , Gametogenesis , Germ Cells , Plasmodium berghei , Animals , Axoneme/chemistry , Dyneins/ultrastructure , Female , Germ Cells/chemistry , Germ Cells/ultrastructure , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron , Microscopy, Fluorescence , Plasmodium berghei/physiology , Plasmodium berghei/ultrastructure
3.
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
4.
Mol Microbiol ; 114(3): 454-467, 2020 09.
Article in English | MEDLINE | ID: mdl-32432369

ABSTRACT

Reticulon and REEP family of proteins stabilize the high curvature of endoplasmic reticulum (ER) tubules. Plasmodium berghei Yop1 (PbYop1) is a REEP5 homolog in Plasmodium. Here, we characterize its function using a gene-knockout (Pbyop1∆). Pbyop1∆ asexual stage parasites display abnormal ER architecture and an enlarged digestive vacuole. The erythrocytic cycle of Pbyop1∆ parasites is severely attenuated and the incidence of experimental cerebral malaria is significantly decreased in Pbyop1∆-infected mice. Pbyop1∆ sporozoites have reduced speed, are slower to invade host cells but give rise to equal numbers of infected HepG2 cells, as WT sporozoites. We propose that PbYOP1's disruption may lead to defects in trafficking and secretion of a subset of proteins required for parasite development and invasion of erythrocytes. Furthermore, the maintenance of ER morphology in different parasite stages is likely to depend on different proteins.


Subject(s)
Malaria/pathology , Membrane Transport Proteins/metabolism , Plasmodium berghei/genetics , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Female , Gene Knockout Techniques , Genes, Protozoan , Hep G2 Cells , Humans , Malaria/transmission , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Movement , Plasmodium berghei/ultrastructure , Protein Transport , Protozoan Proteins/genetics , Rats , Rats, Wistar , Sporozoites/metabolism , Virulence
5.
Sci Rep ; 9(1): 7967, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138850

ABSTRACT

Liver stage Plasmodium parasites reside in a parasitophorous vacuole (PV) that associates with lysosomes. It has previously been shown that these organelles can have beneficial as well as harmful effects on the parasite. Yet it is not clear how the association of lysosomes with the parasite is controlled and how interactions with these organelles lead to the antagonistic outcomes. In this study we used advanced imaging techniques to characterize lysosomal interactions with the PV. In host cells harboring successfully developing parasites we observed that these interaction events reach an equilibrium at the PV membrane (PVM). In a population of arrested parasites, this equilibrium appeared to shift towards a strongly increased lysosomal fusion with the PVM witnessed by strong PVM labeling with the lysosomal marker protein LAMP1. This was followed by acidification of the PV and elimination of the parasite. To systematically investigate elimination of arrested parasites, we generated transgenic parasites that express the photosensitizer KillerRed, which leads to parasite killing after activation. Our work provides insights in cellular details of intracellular killing and lysosomal elimination of Plasmodium parasites independent of cells of the immune system.


Subject(s)
Hepatocytes/metabolism , Host-Parasite Interactions/genetics , Lysosomes/metabolism , Malaria/metabolism , Plasmodium berghei/metabolism , Sporozoites/metabolism , Animals , Biomarkers/metabolism , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Hepatocytes/immunology , Hepatocytes/parasitology , Hepatocytes/ultrastructure , Host-Parasite Interactions/immunology , Humans , Hydrogen-Ion Concentration , Light , Liver/immunology , Liver/metabolism , Liver/parasitology , Lysosomal Membrane Proteins/genetics , Lysosomal Membrane Proteins/immunology , Lysosomes/ultrastructure , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Membrane Fusion , Photosensitizing Agents/chemistry , Photosensitizing Agents/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/ultrastructure , Primary Cell Culture , Sporozoites/growth & development , Sporozoites/ultrastructure , Transgenes , Vacuoles/metabolism , Vacuoles/ultrastructure
6.
Nat Rev Microbiol ; 15(1): 37-54, 2017 01.
Article in English | MEDLINE | ID: mdl-27890922

ABSTRACT

Over the past decade, major advances in imaging techniques have enhanced our understanding of Plasmodium spp. parasites and their interplay with mammalian hosts and mosquito vectors. Cryoelectron tomography, cryo-X-ray tomography and super-resolution microscopy have shifted paradigms of sporozoite and gametocyte structure, the process of erythrocyte invasion by merozoites, and the architecture of Maurer's clefts. Intravital time-lapse imaging has been revolutionary for our understanding of pre-erythrocytic stages of rodent Plasmodium parasites. Furthermore, high-speed imaging has revealed the link between sporozoite structure and motility, and improvements in time-lapse microscopy have enabled imaging of the entire Plasmodium falciparum erythrocytic cycle and the complete Plasmodium berghei pre-erythrocytic stages for the first time. In this Review, we discuss the contribution of key imaging tools to these and other discoveries in the malaria field over the past 10 years.


Subject(s)
Erythrocytes/parasitology , Host-Parasite Interactions/physiology , Merozoites/physiology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Sporozoites/physiology , Animals , Cryoelectron Microscopy , Electron Microscope Tomography , Humans , Malaria/parasitology , Malaria/pathology , Merozoites/ultrastructure , Plasmodium berghei/ultrastructure , Plasmodium falciparum/ultrastructure , Sporozoites/ultrastructure , Time-Lapse Imaging
7.
Mol Microbiol ; 102(5): 775-791, 2016 12.
Article in English | MEDLINE | ID: mdl-27566438

ABSTRACT

To fuel the tremendously fast replication of Plasmodium liver stage parasites, the endoplasmic reticulum (ER) must play a critical role as a major site of protein and lipid biosynthesis. In this study, we analysed the parasite's ER morphology and function. Previous studies exploring the parasite ER have mainly focused on the blood stage. Visualizing the Plasmodium berghei ER during liver stage development, we found that the ER forms an interconnected network throughout the parasite with perinuclear and peripheral localizations. Surprisingly, we observed that the ER additionally generates huge accumulations. Using stimulated emission depletion microscopy and serial block-face scanning electron microscopy, we defined ER accumulations as intricate dense networks of ER tubules. We provide evidence that these accumulations are functional subdivisions of the parasite ER, presumably generated in response to elevated demands of the parasite, potentially consistent with ER stress. Compared to higher eukaryotes, Plasmodium parasites have a fundamentally reduced unfolded protein response machinery for reacting to ER stress. Accordingly, parasite development is greatly impaired when ER stress is applied. As parasites appear to be more sensitive to ER stress than are host cells, induction of ER stress could potentially be used for interference with parasite development.


Subject(s)
Endoplasmic Reticulum/ultrastructure , Plasmodium berghei/ultrastructure , Animals , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Liver/parasitology , Malaria/parasitology , Microscopy/methods , Microscopy, Electron, Scanning , Plasmodium berghei/metabolism , Protozoan Proteins/metabolism , Unfolded Protein Response
8.
Cell Host Microbe ; 18(5): 593-603, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26607162

ABSTRACT

Plasmodium sporozoites are deposited in the host skin by Anopheles mosquitoes. The parasites migrate from the dermis to the liver, where they invade hepatocytes through a moving junction (MJ) to form a replicative parasitophorous vacuole (PV). Malaria sporozoites need to traverse cells during progression through host tissues, a process requiring parasite perforin-like protein 1 (PLP1). We find that sporozoites traverse cells inside transient vacuoles that precede PV formation. Sporozoites initially invade cells inside transient vacuoles by an active MJ-independent process that does not require vacuole membrane remodeling or release of parasite secretory organelles typically involved in invasion. Sporozoites use pH sensing and PLP1 to exit these vacuoles and avoid degradation by host lysosomes. Next, parasites enter the MJ-dependent PV, which has a different membrane composition, precluding lysosome fusion. The malaria parasite has thus evolved different strategies to evade host cell defense and establish an intracellular niche for replication.


Subject(s)
Malaria/pathology , Malaria/parasitology , Plasmodium berghei/metabolism , Plasmodium yoelii/metabolism , Sporozoites/pathology , Sporozoites/parasitology , Vacuoles/parasitology , Animals , Anopheles/parasitology , Hep G2 Cells , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Plasmodium berghei/growth & development , Plasmodium berghei/ultrastructure , Plasmodium yoelii/growth & development , Plasmodium yoelii/ultrastructure , Protozoan Proteins/metabolism , Sporozoites/metabolism , Vacuoles/metabolism , Vacuoles/ultrastructure
9.
Autophagy ; 11(9): 1561-79, 2015.
Article in English | MEDLINE | ID: mdl-26208778

ABSTRACT

Plasmodium parasites are transmitted by Anopheles mosquitoes to the mammalian host and actively infect hepatocytes after passive transport in the bloodstream to the liver. In their target host hepatocyte, parasites reside within a parasitophorous vacuole (PV). In the present study it was shown that the parasitophorous vacuole membrane (PVM) can be targeted by autophagy marker proteins LC3, ubiquitin, and SQSTM1/p62 as well as by lysosomes in a process resembling selective autophagy. The dynamics of autophagy marker proteins in individual Plasmodium berghei-infected hepatocytes were followed by live imaging throughout the entire development of the parasite in the liver. Although the host cell very efficiently recognized the invading parasite in its vacuole, the majority of parasites survived this initial attack. Successful parasite development correlated with the gradual loss of all analyzed autophagy marker proteins and associated lysosomes from the PVM. However, other autophagic events like nonselective canonical autophagy in the host cell continued. This was indicated as LC3, although not labeling the PVM anymore, still localized to autophagosomes in the infected host cell. It appears that growing parasites even benefit from this form of nonselective host cell autophagy as an additional source of nutrients, as in host cells deficient for autophagy, parasite growth was retarded and could partly be rescued by the supply of additional amino acid in the medium. Importantly, mouse infections with P. berghei sporozoites confirmed LC3 dynamics, the positive effect of autophagy activation on parasite growth, and negative effects upon autophagy inhibition.


Subject(s)
Cytosol/immunology , Hepatocytes/immunology , Imaging, Three-Dimensional , Immune Evasion , Immunity , Malaria/immunology , Parasites/immunology , Plasmodium berghei/pathogenicity , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy , Biomarkers/metabolism , Galectins/metabolism , Heat-Shock Proteins/metabolism , Hep G2 Cells , Hepatocytes/parasitology , Hepatocytes/ultrastructure , Humans , Intracellular Membranes/metabolism , Intracellular Membranes/ultrastructure , Life Cycle Stages , Liver/parasitology , Lysosomes/metabolism , Lysosomes/ultrastructure , Malaria/parasitology , Mice , Microtubule-Associated Proteins/metabolism , Parasites/growth & development , Parasites/pathogenicity , Parasites/ultrastructure , Plasmodium berghei/growth & development , Plasmodium berghei/ultrastructure , Sequestosome-1 Protein , Sporozoites/physiology , Sporozoites/ultrastructure , Survival Analysis , Time Factors , Ubiquitin/metabolism , Ubiquitination , Vacuoles/metabolism , Vacuoles/ultrastructure , Virulence
10.
Cell Microbiol ; 17(3): 355-68, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25262869

ABSTRACT

Gametogenesis is the earliest event after uptake of malaria parasites by the mosquito vector, with a decisive impact on colonization of the mosquito midgut. This process is triggered by a drop in temperature and contact with mosquito molecules. In a few minutes, male and female gametocytes escape from the host erythrocyte by rupturing the parasitophorous vacuole and the erythrocyte membranes. Electron-dense, oval-shaped organelles, the osmiophilic bodies (OB), have been implicated in the egress of female gametocytes. By comparative electron microscopy and electron tomography analyses combined with immunolocalization experiments, we here define the morphological features distinctive of male secretory organelles, hereafter named MOB (male osmiophilic bodies). These organelles appear as club-shaped, electron-dense vesicles, smaller than female OB. We found that a drop in temperature triggers MOB clustering, independently of exposure to other stimuli. MDV1/PEG3, a protein associated with OB in Plasmodium berghei females, localizes to both non-clustered and clustered MOB, suggesting that clustering precedes vesicle discharge. A P. berghei mutant lacking the OB-resident female-specific protein Pbg377 displays a dramatic reduction in size of the OB, accompanied by a delay in female gamete egress efficiency, while female gamete fertility is not affected. Immunolocalization experiments indicated that MDV1/PEG3 is still recruited to OB-remnant structures.


Subject(s)
Organelles/ultrastructure , Plasmodium berghei/ultrastructure , Animals , Electron Microscope Tomography , Female , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Organelles/chemistry , Plasmodium berghei/chemistry , Protozoan Proteins/analysis
11.
PLoS One ; 9(12): e113923, 2014.
Article in English | MEDLINE | ID: mdl-25438048

ABSTRACT

The circumsporozoite protein (CSP) is the major surface protein of the sporozoite stage of malaria parasites and has multiple functions as the parasite develops and then migrates from the mosquito midgut to the mammalian liver. The overall structure of CSP is conserved among Plasmodium species, consisting of a species-specific central tandem repeat region flanked by two conserved domains: the NH2-terminus and the thrombospondin repeat (TSR) at the COOH-terminus. Although the central repeat region is an immunodominant B-cell epitope and the basis of the only candidate malaria vaccine in Phase III clinical trials, little is known about its functional role(s). We used the rodent malaria model Plasmodium berghei to investigate the role of the CSP tandem repeat region during sporozoite development. Here we describe two mutant parasite lines, one lacking the tandem repeat region (ΔRep) and the other lacking the NH2-terminus as well as the repeat region (ΔNΔRep). We show that in both mutant lines oocyst formation is unaffected but sporozoite development is defective.


Subject(s)
Malaria/parasitology , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , Sporozoites/growth & development , Animals , Plasmodium berghei/chemistry , Plasmodium berghei/genetics , Plasmodium berghei/ultrastructure , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Deletion , Sporozoites/chemistry , Sporozoites/metabolism , Sporozoites/ultrastructure
12.
PLoS One ; 8(4): e61482, 2013.
Article in English | MEDLINE | ID: mdl-23658610

ABSTRACT

Protein export into the host red blood cell is one of the key processes in the pathobiology of the malaria parasite Plasmodiumtrl falciparum, which extensively remodels the red blood cell to ensure its virulence and survival. In this study, we aimed to shed further light on the protein export mechanisms in the rodent malaria parasite P. berghei and provide further proof of the conserved nature of host cell remodeling in Plasmodium spp. Based on the presence of an export motif (R/KxLxE/Q/D) termed PEXEL (Plasmodium export element), we have generated transgenic P. berghei parasite lines expressing GFP chimera of putatively exported proteins and analysed one of the newly identified exported proteins in detail. This essential protein, termed PbCP1 (P. berghei Cleft-like Protein 1), harbours an atypical PEXEL motif (RxLxY) and is further characterised by two predicted transmembrane domains (2TMD) in the C-terminal end of the protein. We have functionally validated the unusual PEXEL motif in PbCP1 and analysed the role of the 2TMD region, which is required to recruit PbCP1 to discrete membranous structures in the red blood cell cytosol that have a convoluted, vesico-tubular morphology by electron microscopy. Importantly, this study reveals that rodent malaria species also induce modifications to their host red blood cell.


Subject(s)
Cytosol/parasitology , Erythrocytes/parasitology , Plasmodium berghei/metabolism , Protozoan Proteins/chemistry , Amino Acid Sequence , Animals , Cytosol/ultrastructure , Erythrocytes/ultrastructure , Gene Expression , Green Fluorescent Proteins , Life Cycle Stages/genetics , Malaria/parasitology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Plasmodium berghei/genetics , Plasmodium berghei/ultrastructure , Protein Structure, Tertiary , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
13.
Autophagy ; 9(4): 568-80, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388496

ABSTRACT

Analyzing molecular determinants of Plasmodium parasite cell death is a promising approach for exploring new avenues in the fight against malaria. Three major forms of cell death (apoptosis, necrosis and autophagic cell death) have been described in multicellular organisms but which cell death processes exist in protozoa is still a matter of debate. Here we suggest that all three types of cell death occur in Plasmodium liver-stage parasites. Whereas typical molecular markers for apoptosis and necrosis have not been found in the genome of Plasmodium parasites, we identified genes coding for putative autophagy-marker proteins and thus concentrated on autophagic cell death. We characterized the Plasmodium berghei homolog of the prominent autophagy marker protein Atg8/LC3 and found that it localized to the apicoplast. A relocalization of PbAtg8 to autophagosome-like vesicles or vacuoles that appear in dying parasites was not, however, observed. This strongly suggests that the function of this protein in liver-stage parasites is restricted to apicoplast biology.


Subject(s)
Autophagy , Life Cycle Stages , Liver/parasitology , Parasites/cytology , Parasites/growth & development , Plasmodium berghei/cytology , Plasmodium berghei/growth & development , Amino Acid Sequence , Animals , Conserved Sequence , Databases, Protein , Evolution, Molecular , Gene Knockout Techniques , Genetic Complementation Test , Green Fluorescent Proteins/metabolism , Hep G2 Cells , Humans , Lipid Metabolism , Mice , Molecular Sequence Data , Parasites/ultrastructure , Phagosomes/metabolism , Phagosomes/ultrastructure , Plasmodium berghei/ultrastructure , Protein Transport , Protozoan Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Schizonts/cytology , Schizonts/metabolism , Schizonts/ultrastructure , Sequence Homology, Amino Acid , Vacuoles/metabolism
14.
Malar J ; 12: 25, 2013 Jan 19.
Article in English | MEDLINE | ID: mdl-23331966

ABSTRACT

BACKGROUND: The apicoplast is a plastid organelle derived from a secondary endosymbiosis, containing biosynthetic pathways essential for the survival of apicomplexan parasites. The Toxoplasma apicoplast clearly possesses four membranes but in related Plasmodium spp. the apicoplast has variably been reported to have either three or four membranes. METHODS: Cryo-electron tomography was employed to image merozoites of Plasmodium falciparum and Plasmodium berghei frozen in their near-native state. Three-dimensional reconstructions revealed the number of apicoplast membranes and the association of the apicoplast with other organelles. Routine transmission electron microscopy of parasites preserved by high-pressure freezing followed by freeze substitution techniques was also used to analyse apicoplast morphology. RESULTS: Cryo-preserved parasites showed clearly four membranes surrounding the apicoplast. A wider gap between the second and third apicoplast membranes was frequently observed. The apicoplast was found in close proximity to the nucleus and to the rhoptries. The apicoplast matrix showed ribosome-sized particles and membranous whorls. CONCLUSIONS: The Plasmodium apicoplast possesses four membranes, as do the apicoplasts of other apicomplexan parasites. This is consistent with a four-membraned secondary endosymbiotic plastid ancestor.


Subject(s)
Intracellular Membranes/ultrastructure , Plasmodium berghei/ultrastructure , Plasmodium falciparum/ultrastructure , Plastids/ultrastructure , Cryoelectron Microscopy , Electron Microscope Tomography , Imaging, Three-Dimensional , Merozoites/ultrastructure , Microscopy, Electron, Transmission
15.
PLoS One ; 7(12): e50772, 2012.
Article in English | MEDLINE | ID: mdl-23227206

ABSTRACT

The proteins P52 and P36 are expressed in the sporozoite stage of the murine malaria parasite Plasmodium berghei. Δp52&p36 sporozoites lacking expression of both proteins are severely compromised in their capability to develop into liver stage parasites and abort development soon after invasion; presumably due to the absence of a parasitophorous vacuole membrane (PVM). However, a small proportion of P. berghei Δp52&p36 parasites is capable to fully mature in hepatocytes causing breakthrough blood stage infections. We have studied the maturation of replicating Δp52&p36 parasites in cultured Huh-7 hepatocytes. Approximately 50% of Δp52&p36 parasites developed inside the nucleus of the hepatocyte but did not complete maturation and failed to produce merosomes. In contrast cytosolic Δp52&p36 parasites were able to fully mature and produced infectious merozoites. These Δp52&p36 parasites developed into mature schizonts in the absence of an apparent parasitophorous vacuole membrane as shown by immunofluorescence and electron microscopy. Merozoites derived from these maturing Δp52&p36 liver stages were infectious for C57BL/6 mice.


Subject(s)
Hepatocytes/parasitology , Intracellular Membranes/parasitology , Mutation/genetics , Parasites/growth & development , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , Vacuoles/parasitology , Animals , Cell Nucleus/parasitology , Cell Nucleus/ultrastructure , Female , Hepatocytes/pathology , Hepatocytes/ultrastructure , Humans , Intracellular Membranes/ultrastructure , Malaria/parasitology , Malaria/pathology , Merozoites/growth & development , Merozoites/ultrastructure , Mice , Mice, Inbred C57BL , Mutant Proteins/metabolism , Parasites/ultrastructure , Plasmodium berghei/ultrastructure , Vacuoles/ultrastructure
16.
Methods Cell Biol ; 111: 59-73, 2012.
Article in English | MEDLINE | ID: mdl-22857923

ABSTRACT

The interaction of a parasite and a host cell is a complex process, which involves several steps: (1) attachment to the plasma membrane, (2) entry inside the host cell, and (3) hijacking of the metabolism of the host. In biochemical experiments, only an event averaged over the whole cell population can be analyzed. The power of microscopy, however, is to investigate individual events in individual cells. Therefore, parasitologists frequently perform experiments with fluorescence microscopy using different dyes to label structures of the parasite or the host cell. Though the resolution of light microscopy has greatly improved, it is not sufficient to reveal interactions at the ultrastructural level. Furthermore, only specifically labeled structures can be seen and related to each other. Here, we want to demonstrate the additional value of electron microscopy in this area of research. Investigation of the different steps of parasite-host cell interaction by electron microscopy, however, is often hampered by the fact that there are only a few cells infected, and therefore it is difficult to find enough cells to study. A solution is to profit from low magnification, hence large overview, and specific location of the players by fluorescence labels in a light microscope with the high power resolution and structural information provided by an electron microscope, in short by correlative light and electron microscopy.


Subject(s)
Host-Parasite Interactions , Macrophages/ultrastructure , Microscopy, Electron, Transmission , Single-Cell Analysis , Animals , Anopheles/parasitology , Cells, Cultured , Contrast Media/chemistry , Cricetinae , Female , Gold/chemistry , Hep G2 Cells , Humans , Image Processing, Computer-Assisted , Insect Vectors/parasitology , Leishmania donovani/physiology , Leishmania donovani/ultrastructure , Macrophages/parasitology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Phase-Contrast , Phagocytosis , Plasmodium berghei/physiology , Plasmodium berghei/ultrastructure , Staining and Labeling
17.
J Struct Biol ; 180(1): 132-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22683346

ABSTRACT

Electron tomography produces highly magnified 3D image volumes useful for investigating the structure and function of cellular components. Image quality is degraded by multiple scattering events and quantum noise, which depend on the angle at which individual tilt projections are collected. We have adapted a biomedical imaging approach to improve image quality by enhancing individual tilt projections prior to volumetric reconstruction. Specifically, we have developed a family of non-linear anisotropic diffusion (NAD) filters parameterized by the tilt angle. We give a quantitative and qualitative evaluation of our pre-processing approach and the NAD filter. We show an improvement in the reconstructed volumes for tomograms generated from both plastic-embedded and cryo-stabilized samples of malaria parasite-infected erythrocytes.


Subject(s)
Electron Microscope Tomography/methods , Imaging, Three-Dimensional/methods , Algorithms , Anisotropy , Electron Microscope Tomography/standards , Erythrocytes/parasitology , Erythrocytes/ultrastructure , Humans , Imaging, Three-Dimensional/standards , Plasmodium berghei/ultrastructure , Plasmodium falciparum/ultrastructure , Quality Improvement , Signal-To-Noise Ratio , Sporozoites/ultrastructure
18.
PLoS One ; 7(12): e52480, 2012.
Article in English | MEDLINE | ID: mdl-23285059

ABSTRACT

BACKGROUND: In Plasmodium, meiosis occurs in diploid zygotes as they develop into haploid motile ookinetes inside the mosquito. Further sporogonic development involves transformation of ookinetes into oocysts and formation of infective sporozoites. METHODOLOGY/PRINCIPAL FINDINGS: Reverse genetics was employed to examine the role of the meiotic specific recombinase Dmc1, a bacterial RecA homolog during sporogony in Plasmodium berghei. PbDmc1 knockout (KO) parasites showed normal asexual growth kinetics compared to WT parasites; however oocyst formation in mosquitoes was reduced by 50 to 80%. Moreover, the majority of oocysts were retarded in their growth and were smaller in size compared to WT parasites. Only a few Dmc1 KO parasites completed maturation resulting in formation of fewer sporozoites which were incapable of infecting naive mice or hepatocytes in vitro. PbDmc1 KO parasites were shown to be approximately 18 times more sensitive to Bizelesin, a DNA alkylating drug compared to WT parasites as reflected by impairment of oocyst formation and sporogonic development in the mosquito vector. CONCLUSIONS/SIGNIFICANCE: Our findings suggest that PbDmc1 plays a critical role in malaria transmission biology.


Subject(s)
Parasites/enzymology , Parasites/growth & development , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Protozoan Proteins/metabolism , Recombinases/deficiency , Sporozoites/growth & development , Alkylating Agents/pharmacology , Animals , Culicidae/parasitology , DNA Damage , Duocarmycins , Female , Gene Knockout Techniques , Gene Targeting , Genes, Protozoan/genetics , Indoles/pharmacology , Kinetics , Life Cycle Stages/drug effects , Male , Mice , Oocysts/cytology , Oocysts/drug effects , Oocysts/growth & development , Oocysts/ultrastructure , Parasites/pathogenicity , Parasites/ultrastructure , Plasmodium berghei/pathogenicity , Plasmodium berghei/ultrastructure , Protozoan Proteins/genetics , Recombinases/genetics , Recombinases/metabolism , Reproduction, Asexual/drug effects , Sporozoites/drug effects , Urea/analogs & derivatives , Urea/pharmacology
19.
Nanomedicine ; 8(1): 17-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22033078

ABSTRACT

Infection of liver cells by Plasmodium, the malaria parasite, is a clinically silent, obligatory step of the parasite's life cycle. The authors studied the progression of Plasmodium infection in hepatic cells by atomic force microscopy, measuring both topographical and nanomechanical changes upon infection. In recent years, several studies have suggested that cellular nanomechanical properties can be correlated with disease progression. The authors' results show that infected cells exhibit considerable topographical changes, which can be correlated with the presence of the parasite, leading to a significant roughening of the cell membrane. The nanomechanical measurements showed that infected cells were significantly stiffer than noninfected cells. Furthermore, the stiffening of the cells appeared to be a cellular reaction to the Plasmodium infection, rather than a result of the stiffness of the invading parasites themselves. This article provides the first evidence of mechanical changes occurring in hepatic cells in response to Plasmodium infection. FROM THE CLINICAL EDITOR: The authors have studied the progression of Plasmodium infection in hepatic cells by atomic force microscopy, measuring topographical and nanomechanical changes upon infection. The nanomechanical measurements demonstrated that infected cells were significantly stiffer than noninfected cells.


Subject(s)
Cell Membrane/ultrastructure , Cell Shape , Hepatocytes/ultrastructure , Liver/ultrastructure , Animals , Cell Line, Tumor , Hepatocytes/parasitology , Humans , Liver/parasitology , Malaria/parasitology , Microscopy, Atomic Force , Plasmodium berghei/ultrastructure
20.
Mol Biochem Parasitol ; 182(1-2): 93-6, 2012.
Article in English | MEDLINE | ID: mdl-22138565

ABSTRACT

Malaria parasite motility relies on an internal parasite actomyosin motor that, when linked to the host cell substrate, propels motile zoites forward. Despite their key role in this process, attempts to visualize actin microfilaments (F-actin) during motility and under native microscopy conditions have not to date been successful. Towards facilitating their visualization we present here a Plasmodium berghei transgenic line in which a green fluorescent protein (GFP)-actin fusion is constitutively expressed through the lifecycle. Focused investigation of the largest motile form, the insect stage ookinete, demonstrates a large cytosolic pool of actin with no obvious F-actin structures. However, following treatment with the actin filament-stabilizing drug Jasplakinolide, we show evidence for concentration of F-actin dynamics in the parasite pellicle and at polar apices. These observations support current models for gliding motility and establish a cellular tool for further exploration of the diverse roles actin is thought to play throughout parasite development.


Subject(s)
Actins/chemistry , Green Fluorescent Proteins/chemistry , Life Cycle Stages , Plasmodium berghei/chemistry , Actins/antagonists & inhibitors , Animals , Animals, Genetically Modified , Carrier Proteins/chemistry , Depsipeptides/pharmacology , Image Processing, Computer-Assisted/methods , Locomotion , Microfilament Proteins/chemistry , Microscopy, Electron , Plasmodium berghei/drug effects , Plasmodium berghei/ultrastructure , Protozoan Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...