Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FEMS Microbiol Rev ; 36(1): 111-30, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22092244

ABSTRACT

Protozoan parasites of the genus Plasmodium are the causative agents of malaria. Despite more than 100 years of research, the complex life cycle of the parasite still bears many surprises and it is safe to say that understanding the biology of the pathogen will keep scientists busy for many years to come. Malaria research has mainly concentrated on the pathological blood stage of Plasmodium parasites, leaving us with many questions concerning parasite development within the mosquito and during the exo-erythrocytic stage in the vertebrate host. After the discovery of the Plasmodium liver stage in the middle of the last century, it remained understudied for many years but the realization that it represents a promising target for vaccination approaches has brought it back into focus. The last decade saw many new and exciting discoveries concerning the exo-erythrocytic stage and in this review we will discuss the highlights of the latest developments in the field.


Subject(s)
Cell Death , Hepatocytes/parasitology , Liver/parasitology , Plasmodium/pathogenicity , Animals , Humans , Models, Biological
2.
PLoS Pathog ; 7(9): e1002224, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21909271

ABSTRACT

The protozoan parasite Plasmodium is transmitted by female Anopheles mosquitoes and undergoes obligatory development within a parasitophorous vacuole in hepatocytes before it is released into the bloodstream. The transition to the blood stage was previously shown to involve the packaging of exoerythrocytic merozoites into membrane-surrounded vesicles, called merosomes, which are delivered directly into liver sinusoids. However, it was unclear whether the membrane of these merosomes was derived from the parasite membrane, the parasitophorous vacuole membrane or the host cell membrane. This knowledge is required to determine how phagocytes will be directed against merosomes. Here, we fluorescently label the candidate membranes and use live cell imaging to show that the merosome membrane derives from the host cell membrane. We also demonstrate that proteins in the host cell membrane are lost during merozoite liberation from the parasitophorous vacuole. Immediately after the breakdown of the parasitophorous vacuole membrane, the host cell mitochondria begin to degenerate and protein biosynthesis arrests. The intact host cell plasma membrane surrounding merosomes allows Plasmodium to mask itself from the host immune system and bypass the numerous Kupffer cells on its way into the bloodstream. This represents an effective strategy for evading host defenses before establishing a blood stage infection.


Subject(s)
Cell Membrane/physiology , Merozoites/ultrastructure , Plasmodium/physiology , Animals , Hep G2 Cells , Hepatocytes/parasitology , Hepatocytes/ultrastructure , Humans , Liver/parasitology , Mice , Mitochondria/pathology , Plasmodium/metabolism , Vacuoles/physiology , Vacuoles/ultrastructure
3.
Cell Microbiol ; 13(11): 1768-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801293

ABSTRACT

The liver stage of the Plasmodium parasite remains one of the most promising targets for intervention against malaria as it is clinically silent, precedes the symptomatic blood stage and represents a bottleneck in the parasite life cycle. However, many aspects of the development of the parasite during this stage are far from understood. During the liver stage, the parasite undergoes extensive replication, forming tens of thousands of infectious merozoites from each invading sporozoite. This implies a very efficient and accurate process of cytokinesis and thus also of organelle development and segregation. We have generated for the first time Plasmodium berghei double-fluorescent parasite lines, allowing visualization of the apicoplast, mitochondria and nuclei in live liver stage parasites. Using these we have seen that in parallel with nuclear division, the apicoplast and mitochondrion become two extensively branched and intertwining structures. The organelles then undergo impressive morphological and positional changes prior to cell division. To form merozoites, the parasite undergoes cytokinesis and the complex process of organelle development and segregation into the forming daughter merozoites could be analysed in detail using the newly generated transgenic parasites.


Subject(s)
Cytokinesis , Liver/parasitology , Merozoites/physiology , Organelles/physiology , Plasmodium berghei/physiology , Genes, Reporter , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Merozoites/growth & development , Microscopy, Fluorescence , Organelles/ultrastructure , Plasmodium berghei/growth & development , Staining and Labeling/methods
4.
Cell Microbiol ; 13(1): 109-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20923452

ABSTRACT

Knowledge of the dynamic features of the processes driven by malaria parasites in the spleen is lacking. To gain insight into the function and structure of the spleen in malaria, we have implemented intravital microscopy and magnetic resonance imaging of the mouse spleen in experimental infections with non-lethal (17X) and lethal (17XL) Plasmodium yoelii strains. Noticeably, there was higher parasite accumulation, reduced motility, loss of directionality, increased residence time and altered magnetic resonance only in the spleens of mice infected with 17X. Moreover, these differences were associated with the formation of a strain-specific induced spleen tissue barrier of fibroblastic origin, with red pulp macrophage-clearance evasion and with adherence of infected red blood cells to this barrier. Our data suggest that in this reticulocyte-prone non-lethal rodent malaria model, passage through the spleen is different from what is known in other Plasmodium species and open new avenues for functional/structural studies of this lymphoid organ in malaria.


Subject(s)
Erythrocytes/parasitology , Macrophages/parasitology , Malaria/pathology , Plasmodium yoelii/pathogenicity , Rodent Diseases/pathology , Spleen/pathology , Spleen/parasitology , Animals , Female , Immune Evasion , Malaria/immunology , Malaria/parasitology , Mice , Mice, Inbred BALB C , Plasmodium yoelii/immunology , Rodent Diseases/immunology , Rodent Diseases/parasitology , Spleen/immunology
5.
Cell Microbiol ; 12(5): 569-79, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20180802

ABSTRACT

Plasmodium parasites, the causative agents of malaria, first invade and develop within hepatocytes before infecting red blood cells and causing symptomatic disease. Because of the low infection rates in vitro and in vivo, the liver stage of Plasmodium infection is not very amenable to biochemical assays, but the large size of the parasite at this stage in comparison with Plasmodium blood stages makes it accessible to microscopic analysis. A variety of imaging techniques has been used to this aim, ranging from electron microscopy to widefield epifluorescence and laser scanning confocal microscopy. High-speed live video microscopy of fluorescent parasites in particular has radically changed our view on key events in Plasmodium liver-stage development. This includes the fate of motile sporozoites inoculated by Anopheles mosquitoes as well as the transport of merozoites within merosomes from the liver tissue into the blood vessel. It is safe to predict that in the near future the application of the latest microscopy techniques in Plasmodium research will bring important insights and allow us spectacular views of parasites during their development in the liver.


Subject(s)
Image Processing, Computer-Assisted/methods , Liver/parasitology , Malaria/parasitology , Microscopy/methods , Plasmodium/cytology , Humans
6.
Nat Protoc ; 4(10): 1433-9, 2009.
Article in English | MEDLINE | ID: mdl-19745825

ABSTRACT

This protocol describes a method for obtaining rodent Plasmodium parasite clones with high efficiency, which takes advantage of the normal course of Plasmodium in vitro exoerythrocytic development. At the completion of development, detached cells/merosomes form, which contain hundreds to thousands of merozoites. As all parasites within a single detached cell/merosome derive from the same sporozoite, we predicted them to be genetically identical. To prove this, hepatoma cells were infected simultaneously with a mixture of Plasmodium berghei sporozoites expressing either GFP or mCherry. Subsequently, individual detached cells/merosomes from this mixed population were selected and injected into mice, resulting in clonal blood stage parasite infections. Importantly, as a large majority of mice become successfully infected using this protocol, significantly less mice are necessary than for the widely used technique of limiting dilution cloning. To produce a clonal P. berghei blood stage infection from a non-clonal infection using this procedure requires between 4 and 5 weeks.


Subject(s)
Malaria/parasitology , Parasitology/methods , Plasmodium berghei/isolation & purification , Animals , Cell Line, Tumor , Humans , Injections , Mice , Plasmodium berghei/growth & development
7.
Biotechnol J ; 4(6): 895-902, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19492329

ABSTRACT

Fluorescent proteins have proven to be important tools for in vitro live imaging of parasites and for imaging of parasites within the living host by intravital microscopy. We observed that a red fluorescent transgenic malaria parasite of rodents, Plasmodium berghei-RedStar, is suitable for in vitro live imaging experiments but bleaches rapidly upon illumination in intravital imaging experiments using mice. We have therefore generated two additional transgenic parasite lines expressing the novel red fluorescent proteins tdTomato and mCherry, which have been reported to be much more photostable than first- and second-generation red fluorescent proteins including RedStar. We have compared all three red fluorescent parasite lines for their use in in vitro live and intravital imaging of P. berghei blood and liver parasite stages, using both confocal and wide-field microscopy. While tdTomato bleached almost as rapidly as RedStar, mCherry showed improved photostability and was bright in all experiments performed.


Subject(s)
Fluorescent Dyes/metabolism , Luminescent Proteins/metabolism , Plasmodium berghei/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Data Interpretation, Statistical , Female , Fluorescent Dyes/chemistry , Humans , Liver/parasitology , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Mice , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Plasmodium berghei/genetics , Red Fluorescent Protein
8.
Protist ; 160(1): 51-63, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19026596

ABSTRACT

The rodent malaria parasite Plasmodium berghei develops in hepatocytes within 48-52h from a single sporozoite into up to 20,000 daughter parasites, so-called merozoites. The cellular and molecular details of this extensive proliferation are still largely unknown. Here we have used a transgenic, RFP-expressing P. berghei parasite line and molecular imaging techniques including intravital microscopy to decipher various aspects of parasite development within the hepatocyte. In late schizont stages, MSP1 is expressed and incorporated into the parasite plasma membrane that finally forms the membrane of developing merozoites by continuous invagination steps. We provide first evidence for activation of a verapamil-sensitive Ca(2+) channel in the plasma membrane of liver stage parasites before invagination occurs. During merozoite formation, the permeability of the parasitophorous vacuole membrane changes considerably before it finally becomes completely disrupted, releasing merozoites into the host cell cytoplasm.


Subject(s)
Cell Membrane/metabolism , Malaria/parasitology , Plasmodium berghei/growth & development , Vacuoles/metabolism , Animals , Calcium Channels/metabolism , Cell Line , Cell Membrane Permeability , Hepatocytes/parasitology , Humans , Liver/parasitology , Merozoites/growth & development , Mice , Microscopy, Electron, Transmission , Microscopy, Fluorescence/methods , Rats , Sporozoites/growth & development , Verapamil
SELECTION OF CITATIONS
SEARCH DETAIL
...