Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
mBio ; 12(3): e0098821, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154418

ABSTRACT

Chagas' disease arises as a direct consequence of the lytic cycle of Trypanosoma cruzi in the mammalian host. While invasion is well studied for this pathogen, study of egress has been largely neglected. Here, we provide the first description of T. cruzi egress documenting a coordinated mechanism by which T. cruzi engineers its escape from host cells in which it has proliferated and which is essential for maintenance of infection and pathogenesis. Our results indicate that this parasite egress is a sudden event involving coordinated remodeling of host cell cytoskeleton and subsequent rupture of host cell plasma membrane. We document that host cells maintain plasma membrane integrity until immediately prior to parasite release and report the sequential transformation of the host cell's actin cytoskeleton from normal meshwork in noninfected cells to spheroidal cages-a process initiated shortly after amastigogenesis. Quantification revealed gradual reduction in F-actin over the course of infection, and using cytoskeletal preparations and electron microscopy, we were able to observe disruption of the F-actin proximal to intracellular trypomastigotes. Finally, Western blotting experiments suggest actin degradation driven by parasite proteases, suggesting that degradation of cytoskeleton is a principal component controlling the initiation of egress. Our results provide the first description of the cellular mechanism that regulates the lytic component of the T. cruzi lytic cycle. We show graphically how it is possible to preserve the envelope of host cell plasma membrane during intracellular proliferation of the parasite and how, in cells packed with amastigotes, differentiation into trypomastigotes may trigger sudden egress. IMPORTANCE Understanding how Trypanosoma cruzi interacts with host cells has been transformed by high-quality studies that have examined in detail the mechanisms of T. cruzi host cell invasion. In contrast, little is known about the latter stages of the parasite's lytic cycle: how parasites egress and thereby sustain round after round of infection. Our results show that once in the host cell cytosol and having undergone amastigogenesis, T. cruzi begins to alter the host cell cytoskeleton, remodeling normal F-actin meshworks into encapsulating spheroidal cages. Filamentous actin diminishes over the course of the lytic cycle, and just prior to egress, the filaments comprising the cages are severely degraded where adjacent to the parasites. We conclude that sudden egress follows breach of the containment afforded by the actin cytoskeleton and subsequent plasma membrane rupture-a process that when understood in molecular detail may serve as a target for future novel therapeutic interventions.


Subject(s)
Actin Cytoskeleton/physiology , Cell Membrane/pathology , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Host-Parasite Interactions , Trypanosoma cruzi/physiology , Actins/metabolism , Animals , Cell Membrane/parasitology , Chagas Disease/parasitology , Chlorocebus aethiops , Vero Cells
2.
Commun Biol ; 4(1): 134, 2021 01 29.
Article in English | MEDLINE | ID: mdl-33514854

ABSTRACT

Genome assemblies provide a powerful basis of comparative multi-omics analyses that offer insight into parasite pathogenicity, host-parasite interactions, and invasion biology. As a unique intracellular nematode, Trichinella consists of two clades, encapsulated and non-encapsulated. Genomic correlation of the distinct differences between the two clades is still unclear. Here, we report an annotated draft reference genome of non-encapsulated Trichinella, T. pseudospiralis, and perform comparative multi-omics analyses with encapsulated T. spiralis. Genome and methylome analyses indicate that, during Trichinella evolution, the two clades of Trichinella exhibit differential expansion and methylation of parasitism-related multi-copy gene families, especially for the DNase II members of the phospholipase D superfamily and Glutathione S-transferases. Further, methylome and transcriptome analyses revealed divergent key excretory/secretory (E/S) genes between the two clades. Among these key E/S genes, TP12446 is significantly more expressed across three life stages in T. pseudospiralis. Overexpression of TP12446 in the mouse C2C12 skeletal muscle cell line could induce inhibition of myotube formation and differentiation, further indicating its key role in parasitism of T. pseudospiralis. This multi-omics study provides a foundation for further elucidation of the mechanism of nurse cell formation and immunoevasion, as well as the identification of pharmacological and diagnostic targets of trichinellosis.


Subject(s)
Epigenome , Genes, Helminth , Genome, Protozoan , Helminth Proteins/genetics , Muscle, Skeletal/parasitology , Trichinella/genetics , Trichinellosis/parasitology , Animals , Cell Differentiation , Cell Line , Cytoskeleton/parasitology , Cytoskeleton/pathology , Evolution, Molecular , Genomics , Helminth Proteins/metabolism , Host-Parasite Interactions , Mice , Muscle Fibers, Skeletal/parasitology , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Trichinella/metabolism , Trichinella/pathogenicity , Trichinella spiralis/genetics , Trichinella spiralis/metabolism , Trichinella spiralis/parasitology , Trichinellosis/pathology
3.
Parasit Vectors ; 13(1): 169, 2020 Apr 05.
Article in English | MEDLINE | ID: mdl-32248844

ABSTRACT

BACKGROUND: Trypanosoma brucei exhibits a complex life-cycle alternating between tsetse flies and mammalian hosts. When parasites infect the fly, cells differentiate to adapt to life in various tissues, which is accompanied by drastic morphological and biochemical modifications especially in the proventriculus. This key step represents a bottleneck for salivary gland infection. METHODS: Here, we monitored flagellum assembly in trypanosomes during differentiation from the trypomastigote to the epimastigote stage, i.e. when the nucleus migrates to the posterior end of the cell, by using three-dimensional electron microscopy (focused ion beam scanning electron microscopy, FIB-SEM) and immunofluorescence assays. RESULTS: The combination of light and electron microscopy approaches provided structural and molecular evidence that the new flagellum is assembled while the nucleus migrates towards the posterior region of the body. Two major differences with well-known procyclic cells are reported. First, growth of the new flagellum begins when the associated basal body is found in a posterior position relative to the mature flagellum. Secondly, the new flagellum acquires its own flagellar pocket before rotating on the left side of the anterior-posterior axis. FIB-SEM revealed the presence of a structure connecting the new and mature flagellum and serial sectioning confirmed morphological similarities with the flagella connector of procyclic cells. We discuss the potential function of the flagella connector in trypanosomes from the proventriculus. CONCLUSIONS: These findings show that T. brucei finely modulates its cytoskeletal components to generate highly variable morphologies.


Subject(s)
Flagella/physiology , Trypanosoma brucei brucei/physiology , Tsetse Flies/parasitology , Animals , Cell Differentiation , Cytoskeleton/parasitology , Flagella/genetics , Fluorescent Antibody Technique , Life Cycle Stages , Male , Microscopy, Electron , Protozoan Proteins , Trypanosoma brucei brucei/ultrastructure
4.
Microbiol Mol Biol Rev ; 83(4)2019 11 20.
Article in English | MEDLINE | ID: mdl-31484690

ABSTRACT

The asexual intraerythrocytic development of Plasmodium falciparum, causing the most severe form of human malaria, is marked by extensive host cell remodeling. Throughout the processes of invasion, intracellular development, and egress, the erythrocyte membrane skeleton is remodeled by the parasite as required for each specific developmental stage. The remodeling is facilitated by a plethora of exported parasite proteins, and the erythrocyte membrane skeleton is the interface of most of the observed interactions between the parasite and host cell proteins. Host cell remodeling has been extensively described and there is a vast body of information on protein export or the description of parasite-induced structures such as Maurer's clefts or knobs on the host cell surface. Here we specifically review the molecular level of each host cell-remodeling step at each stage of the intraerythrocytic development of P. falciparum We describe key events, such as invasion, knob formation, and egress, and identify the interactions between exported parasite proteins and the host cell cytoskeleton. We discuss each remodeling step with respect to time and specific requirement of the developing parasite to explain host cell remodeling in a stage-specific manner. Thus, we highlight the interaction with the host membrane skeleton as a key event in parasite survival.


Subject(s)
Cytoskeleton/physiology , Cytoskeleton/parasitology , Erythrocytes/parasitology , Host-Parasite Interactions , Malaria, Falciparum/pathology , Erythrocyte Membrane , Erythrocytes/physiology , Humans , Life Cycle Stages , Plasmodium falciparum , Protein Transport
5.
Mol Biochem Parasitol ; 231: 111189, 2019 07.
Article in English | MEDLINE | ID: mdl-31125575

ABSTRACT

The MESA erythrocyte cytoskeleton binding (MEC) motif is a 13-amino acid sequence found in 14 exported Plasmodium falciparum proteins. First identified in the P. falciparum Mature-parasite-infected Erythrocyte Surface Antigen (MESA), the MEC motif is sufficient to target proteins to the infected red blood cell cytoskeleton. To identify host cell targets, purified MESA MEC motif was incubated with a soluble extract from uninfected erythrocytes, precipitated and subjected to mass spectrometry. The most abundant co-purifying protein was erythrocyte ankyrin (ANK1). A direct interaction between the MEC motif and ANK1 was independently verified using co-purification experiments, the split-luciferase assay, and the yeast two-hybrid assay. A systematic mutational analysis of the core MEC motif demonstrated a critical role for the conserved aspartic acid residue at the C-terminus of the MEC motif for binding to both erythrocyte inside-out vesicles and to ANK1. Using a panel of ANK1 constructs, the MEC motif binding site was localized to the ZU5C domain, which has no known function. The MEC motif had no impact on erythrocyte deformability when introduced into uninfected erythrocyte ghosts, suggesting the MEC motif's primary function is to target exported proteins to the cytoskeleton. Finally, we show that PF3D7_0402100 (PFD0095c) binds to ANK1 and band 4.1, likely through its MEC and PHIST motifs, respectively. In conclusion, we have provided multiple lines of evidence that the MEC motif binds to erythrocyte ANK1.


Subject(s)
Ankyrins/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Motifs , Ankyrins/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Erythrocytes/metabolism , Humans , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Protein Binding , Protozoan Proteins/genetics
6.
Int J Parasitol ; 48(3-4): 275-285, 2018 03.
Article in English | MEDLINE | ID: mdl-29258830

ABSTRACT

Trichinella spiralis infection in skeletal muscle culminates with nurse cell formation. The participation of excretory-secretory products of the muscle larvae has been implicated in this process through different studies performed in infected muscle and the muscle cell line C2C12. In this work, we developed primary myoblast cultures to analyse the changes induced by excretory-secretory products of the muscle larvae in muscle cells. Microarray analyses revealed expression changes in muscle cell differentiation, proliferation, cytoskeleton organisation, cell motion, transcription, cell cycle, apoptosis and signalling pathways such as MAPK, Jak-STAT, Wnt and PI3K-Akt. Some of these changes were further evaluated by other methodologies such as quantitative real-time PCR (qRT-PCR) and western blot, confirming that excretory-secretory products of the muscle larvae treated primary mouse myoblasts undergo increased proliferation, decreased expression of MHC and up-regulation of α-actin. In addition, changes in relevant muscle transcription factors (Pax7, Myf5 and Mef2c) were observed. Taken together, these results provide new information about how T. spiralis could alter the normal process of skeletal muscle repair after ML invasion to accomplish nurse cell formation.


Subject(s)
Helminth Proteins/metabolism , Myoblasts, Skeletal/parasitology , Trichinella spiralis/metabolism , Actins/genetics , Actins/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/parasitology , DNA, Helminth/genetics , DNA, Helminth/metabolism , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique, Indirect , Gene Expression , Hindlimb , Larva/metabolism , Luminescence , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/cytology , Myoblasts, Skeletal/metabolism , RNA, Helminth/genetics , RNA, Helminth/isolation & purification , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Tissue Array Analysis , Trichinella spiralis/genetics
7.
Cell Microbiol ; 19(9)2017 09.
Article in English | MEDLINE | ID: mdl-28409866

ABSTRACT

The successful invasion of Plasmodium is an essential step in their life cycle. The parasite reticulocyte-binding protein homologues (RHs) and erythrocyte-binding like proteins are two families involved in the invasion leading to merozoite-red blood cell (RBC) junction formation. Ca2+ signaling has been shown to play a critical role in the invasion. RHs have been linked to Ca2+ signaling, which triggers the erythrocyte-binding like proteins release ahead of junction formation, consistent with RHs performing an initial sensing function in identifying suitable RBCs. RH5, the only essential RHs, is a highly promising vaccine candidate. RH5-basigin interaction is essential for merozoite invasion and also important in determining host tropism. Here, we show that RH5 has a distinct function from the other RHs. We show that RH5-Basigin interaction on its own triggers a Ca2+ signal in the RBC resulting in changes in RBC cytoskeletal proteins phosphorylation and overall alterations in RBC cytoskeleton architecture. Antibodies targeting RH5 that block the signal prevent invasion before junction formation consistent with the Ca2+ signal in the RBC leading to rearrangement of the cytoskeleton required for invasion. This work provides the first time a functional context for the essential role of RH5 and will now open up new avenues to target merozoite invasion.


Subject(s)
Basigin/metabolism , Calcium Signaling/physiology , Carrier Proteins/metabolism , Erythrocytes/physiology , Merozoites/pathogenicity , Plasmodium falciparum/pathogenicity , Antibodies, Monoclonal/immunology , Antigens, Protozoan/biosynthesis , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Line , Cytoskeleton/parasitology , Cytoskeleton/pathology , Erythrocytes/parasitology , Host-Parasite Interactions/physiology , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Protozoan Proteins/biosynthesis
8.
Blood Cells Mol Dis ; 58: 35-44, 2016 May.
Article in English | MEDLINE | ID: mdl-27067487

ABSTRACT

The phosphorylation status of red blood cell proteins is strongly altered during the infection by the malaria parasite Plasmodium falciparum. We identify the key phosphorylation events that occur in the erythrocyte membrane and cytoskeleton during infection, by a comparative analysis of global phospho-proteome screens between infected (obtained at schizont stage) and uninfected RBCs. The meta-analysis of reported mass spectrometry studies revealed a novel compendium of 495 phosphorylation sites in 182 human proteins with regulatory roles in red cell morphology and stability, with about 25% of these sites specific to infected cells. A phosphorylation motif analysis detected 7 unique motifs that were largely mapped to kinase consensus sequences of casein kinase II and of protein kinase A/protein kinase C. This analysis highlighted prominent roles for PKA/PKC involving 78 phosphorylation sites. We then compared the phosphorylation status of PKA (PKC) specific sites in adducin, dematin, Band 3 and GLUT-1 in uninfected RBC stimulated or not by cAMP to their phosphorylation status in iRBC. We showed cAMP-induced phosphorylation of adducin S59 by immunoblotting and we were able to demonstrate parasite-induced phosphorylation for adducin S726, Band 3 and GLUT-1, corroborating the protein phosphorylation status in our erythrocyte phosphorylation site compendium.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/physiology , Proteome/metabolism , Amino Acid Sequence , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytoskeleton/chemistry , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Erythrocytes/chemistry , Erythrocytes/metabolism , Glucose Transporter Type 1/analysis , Glucose Transporter Type 1/metabolism , Humans , Phosphorylation , Proteome/analysis
9.
Int J Parasitol ; 46(8): 507-17, 2016 07.
Article in English | MEDLINE | ID: mdl-27109557

ABSTRACT

Enteromyxum scophthalmi, an intestinal myxozoan parasite, is the causative agent of a threatening disease for turbot (Scophthalmus maximus, L.) aquaculture. The colonisation of the digestive tract by this parasite leads to a cachectic syndrome associated with high morbidity and mortality rates. This myxosporidiosis has a long pre-patent period and the first detectable clinical and histopathological changes are subtle. The pathogenic mechanisms acting in the early stages of infection are still far from being fully understood. Further information on the host-parasite interaction is needed to assist in finding efficient preventive and therapeutic measures. Here, a RNA-seq-based transcriptome analysis of head kidney, spleen and pyloric caeca from experimentally-infected and control turbot was performed. Only infected fish with early signs of infection, determined by histopathology and immunohistochemical detection of E. scophthalmi, were selected. The RNA-seq analysis revealed, as expected, less intense transcriptomic changes than those previously found during later stages of the disease. Several genes involved in IFN-related pathways were up-regulated in the three organs, suggesting that the IFN-mediated immune response plays a main role in this phase of the disease. Interestingly, an opposite expression pattern had been found in a previous study on severely infected turbot. In addition, possible strategies for immune system evasion were suggested by the down-regulation of different genes encoding complement components and acute phase proteins. At the site of infection (pyloric caeca), modulation of genes related to different structural proteins was detected and the expression profile indicated the inhibition of cell proliferation and differentiation. These transcriptomic changes provide indications regarding the mechanisms of parasite attachment to and invasion of the host. The current results contribute to a better knowledge of the events that characterise the early stages of turbot enteromyxosis and provide valuable information to identify molecular markers for early detection and control of this important parasitosis.


Subject(s)
Fish Diseases/parasitology , Flatfishes/parasitology , Immune Evasion/physiology , Intestinal Diseases, Parasitic/veterinary , Myxozoa/genetics , Parasitic Diseases, Animal/parasitology , Acute-Phase Proteins/genetics , Animals , Cecum/parasitology , Complement System Proteins/genetics , Cytoskeleton/chemistry , Cytoskeleton/genetics , Cytoskeleton/parasitology , Down-Regulation , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/parasitology , Fish Diseases/immunology , Fish Diseases/pathology , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Interferons/genetics , Interferons/immunology , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/parasitology , Intestines/parasitology , Intestines/pathology , Kidney/parasitology , Myxozoa/immunology , Myxozoa/physiology , Parasitic Diseases, Animal/immunology , Parasitic Diseases, Animal/pathology , Sequence Analysis, RNA , Spleen/parasitology , Up-Regulation
10.
FASEB J ; 28(7): 3103-13, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24706359

ABSTRACT

The genomes of malaria parasites (Plasmodium spp.) contain a family of genes encoding proteins with a Plasmodium helical interspersed subtelomeric (PHIST) domain, most of which are predicted to be exported into the parasite-infected human red blood cell (iRBC). Here, using transgenic parasites and a combination of cellular, biochemical, and biophysical assays, we have characterized and determined the function of a novel member of the PHIST protein family in Plasmodium falciparum, termed lysine-rich membrane-associated PHISTb (LyMP). LyMP was shown to associate directly with the cytoskeleton of iRBCs where it plays a role in their abnormal ability to adhere to a protein expressed on vascular endothelial cells, resulting in sequestration. Deletion of LyMP dramatically reduced adhesion of iRBCs to CD36 by 55%, which was completely restored to wild-type levels on complementation. Intriguingly, in the absence of LyMP, formation of RBC membrane knobs and the level of surface exposure of the parasites' major cytoadhesive ligand, PfEMP1, were identical to those for the parental parasite line, demonstrating for the first time an additional mechanism that enhances cytoadherence of iRBCs beyond those already recognized. Our findings identify LyMP as a previously unknown RBC cytoskeletal-binding protein that is likely to be of major significance in the complex pathophysiology of falciparum malaria.-Proellocks, N. I., Herrmann, S., Buckingham, D. W., Hanssen, E., Hodges, E. K., Elsworth, B., Morahan, B. J., Coppel, R. L., Cooke, B. M. A lysine-rich membrane-associated PHISTb protein involved in alteration of the cytoadhesive properties of Plasmodium falciparum infected red blood cells.


Subject(s)
Cell Adhesion/physiology , Cytoskeleton/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Lysine/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Cell Membrane/metabolism , Cell Membrane/parasitology , Cytoskeleton/parasitology , Endothelium, Vascular/metabolism , Endothelium, Vascular/parasitology , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Membrane Proteins/metabolism , Protein Binding/physiology
11.
Int J Med Microbiol ; 302(4-5): 210-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22925990

ABSTRACT

Apicomplexan parasites are obligate intracellular protozoans and are well recognized modulators of the host cell machinery on varying levels such as host cell metabolism, MHC expression, cell cycle, or apoptosis in order to guarantee their intracellular development and survival. One of the most thoroughly examined apicomplexan pathogens demonstrating a potent manipulative capacity with respect to various host cell functions is Toxoplasma gondii, a protozoon exhibiting rapid intracellular development with small meronts in any nucleated cell, almost irrespective of the cell type or host origin. In contrast, Eimeria bovis merogony I is host- and cell type-restricted and occurs exclusively in bovine endothelial host cells. Furthermore, as a peculiarity, intracellular E. bovis meront I development is a long-lasting process (up to 3 weeks), leading to the formation of huge macromeronts of up to 300 µm in size, containing up to 120,000 merozoites I as offspring. In consequence, the necessity for intense host cell modulation to support this particular development appears even more pressing than in other apicomplexan parasite cases. Here we review the data currently available on E. bovis-host cell interactions, indicating the intriguing capacity of this protozoan to exploit and utilize its host cell for its own benefit.


Subject(s)
Eimeria/pathogenicity , Endothelial Cells/parasitology , Host-Parasite Interactions , Sporozoites/growth & development , Animals , Apoptosis , Cattle , Cell Shape , Coccidiosis/immunology , Coccidiosis/parasitology , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Eimeria/genetics , Eimeria/immunology , Eimeria/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation , Species Specificity , Sporozoites/metabolism , Transcription, Genetic
12.
Br J Haematol ; 157(2): 171-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22313394

ABSTRACT

The intra-erythrocyte growth and survival of the malarial parasite Plasmodium falciparum is responsible for both uncomplicated and severe malaria cases and depends on the parasite's ability to remodel its host cell. Host cell remodelling has several functions for the parasite, such as acquiring nutrients from the extracellular milieu because of the loss of membrane transporters upon erythrocyte differentiation, avoiding splenic clearance by conferring cytoadhesive properties to the infected erythrocyte, escaping the host immune response by exporting antigenically variant proteins at the red blood cell surface. In addition, parasite-induced changes at the red blood cell membrane and sub-membrane skeleton are also necessary for the efficient release of the parasite progeny from the host cell. Here we review these cellular and molecular changes, which might not only sustain parasite growth but also prepare, at a very early stage, the last step of egress from the host cell.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Humans
13.
Mem. Inst. Oswaldo Cruz ; 106(8): 1014-1016, Dec. 2011. ilus
Article in English | LILACS | ID: lil-610979

ABSTRACT

It has been recently shown that Trypanosoma cruzi trypomastigotes subvert a constitutive membrane repair mechanism to invade HeLa cells. Using a membrane extraction protocol and high-resolution microscopy, the HeLa cytoskeleton and T. cruzi parasites were imaged during the invasion process after 15 min and 45 min. Parasites were initially found under cells and were later observed in the cytoplasm. At later stages, parasite-driven protrusions with parallel filaments were observed, with trypomastigotes at their tips. We conclude that T. cruzi trypomastigotes induce deformations of the cortical actin cytoskeleton shortly after invasion, leading to the formation of pseudopod-like structures.


Subject(s)
Humans , Cell Membrane/parasitology , Cytoskeleton/parasitology , Trypanosoma cruzi/physiology , Cell Membrane/ultrastructure , Cytoskeleton/ultrastructure , HeLa Cells/parasitology , HeLa Cells/ultrastructure , Time Factors
14.
J Parasitol ; 97(4): 620-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21506833

ABSTRACT

Toxoplasma gondii is an important zoonotic parasite with a worldwide distribution. It infects about one-third of the world's population, causing serious illness in immunosuppressed individuals, fetuses, and infants. Toxoplasma gondii biology within the host cell includes several important phases: (1) active invasion and establishment of a nonfusogenic parasitophorous vacuole in the host cell, (2) extensive modification of the parasitophorous vacuolar membrane for nutrient acquisition, (3) intracellular proliferation by endodyogeny, (4) egress and invasion of new host cells, and (5) stage conversion from tachyzoite to bradyzoite and establishment of chronic infection. During these processes, T. gondii regulates the host cell by modulating morphological, physiological, immunological, genetic, and cellular biological aspects of the host cell. Overall, the infection/development predispositions of T. gondii -host cell interactions overtakes the infection resistance aspects. Upon invasion and development, host cells are modulated to keep a delicate balance between facilitating and eliminating the infection.


Subject(s)
Toxoplasma/physiology , Toxoplasmosis/parasitology , Animals , Apoptosis , Cell Cycle , Cytoskeleton/parasitology , Endoplasmic Reticulum/parasitology , Fibroblasts/cytology , Fibroblasts/parasitology , Humans , Immunocompetence , Immunocompromised Host , Mitochondria/parasitology , Phagocytes/parasitology , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/pathology , Vacuoles/immunology , Vacuoles/parasitology , Vacuoles/physiology
15.
J Biol Chem ; 286(2): 1227-36, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21084299

ABSTRACT

The malaria parasite invades the terminally differentiated erythrocytes, where it grows and multiplies surrounded by a parasitophorous vacuole. Plasmodium blood stages translocate newly synthesized proteins outside the parasitophorous vacuole and direct them to various erythrocyte compartments, including the cytoskeleton and the plasma membrane. Here, we show that the remodeling of the host cell directed by the parasite also includes the recruitment of dematin, an actin-binding protein of the erythrocyte membrane skeleton and its repositioning to the parasite. Internalized dematin was found associated with Plasmodium 14-3-3, which belongs to a family of conserved multitask molecules. We also show that, in vitro, the dematin-14-3-3 interaction is strictly dependent on phosphorylation of dematin at Ser(124) and Ser(333), belonging to two 14-3-3 putative binding motifs. This study is the first report showing that a component of the erythrocyte spectrin-based membrane skeleton is recruited by the malaria parasite following erythrocyte infection.


Subject(s)
14-3-3 Proteins/metabolism , Blood Proteins/metabolism , Erythrocyte Membrane/metabolism , Malaria/metabolism , Phosphoproteins/metabolism , Plasmodium berghei/metabolism , Plasmodium falciparum/metabolism , 14-3-3 Proteins/genetics , Animals , Cell Fractionation , Cyclic AMP/metabolism , Cytoskeletal Proteins , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Erythrocyte Membrane/parasitology , Malaria/parasitology , Mice , Mice, Inbred Strains , Organisms, Genetically Modified , Phosphorylation/physiology , Plasmodium berghei/genetics , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , Protein Transport/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
16.
Mem Inst Oswaldo Cruz ; 106(8): 1014-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241126

ABSTRACT

It has been recently shown that Trypanosoma cruzi trypomastigotes subvert a constitutive membrane repair mechanism to invade HeLa cells. Using a membrane extraction protocol and high-resolution microscopy, the HeLa cytoskeleton and T. cruzi parasites were imaged during the invasion process after 15 min and 45 min. Parasites were initially found under cells and were later observed in the cytoplasm. At later stages, parasite-driven protrusions with parallel filaments were observed, with trypomastigotes at their tips. We conclude that T. cruzi trypomastigotes induce deformations of the cortical actin cytoskeleton shortly after invasion, leading to the formation of pseudopod-like structures.


Subject(s)
Cell Membrane/parasitology , Cytoskeleton/parasitology , Trypanosoma cruzi/physiology , Cell Membrane/ultrastructure , Cytoskeleton/ultrastructure , HeLa Cells/parasitology , HeLa Cells/ultrastructure , Humans , Time Factors
17.
Exp Parasitol ; 126(3): 283-91, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20599990

ABSTRACT

The protozoan parasite Trypanosoma cruzi, the etiologic agent of Chagas disease, is an obligate intracellular protozoan pathogen. Overlapping mechanisms ensure successful infection, yet the relationship between these cellular events and clinical disease remains obscure. This review explores the process of cell invasion from the perspective of cell surface interactions, intracellular signaling, modulation of the host cytoskeleton and endosomal compartment, and the intracellular innate immune response to infection.


Subject(s)
Chagas Disease/parasitology , Trypanosoma cruzi/physiology , Animals , Calcium/metabolism , Cell Membrane/parasitology , Chagas Disease/immunology , Chagas Disease/pathology , Cytoplasm/parasitology , Cytoskeleton/parasitology , Extracellular Matrix/parasitology , Host-Parasite Interactions , Humans , Insect Vectors/parasitology , Membrane Glycoproteins/metabolism , Protozoan Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology
18.
Eukaryot Cell ; 9(11): 1680-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20435700

ABSTRACT

Toxoplasma gondii is an obligate intracellular protozoan parasite that invades and replicates within most nucleated cells of warm-blooded animals. The basis for this wide host cell tropism is unknown but could be because parasites invade host cells using distinct pathways and/or repertoires of host factors. Using synchronized parasite invasion assays, we found that host microtubule disruption significantly reduces parasite invasion into host cells early after stimulating parasite invasion but not at later time points. Host microtubules are specifically associated with the moving junction, which is the site of contact between the host cell and the invading parasite. Host microtubules are specifically associated with the moving junction of those parasites invading early after stimulating invasion but not with those invading later. Disruption of host microtubules has no effect on parasite contact, attachment, motility, or rate of penetration. Rather, host microtubules hasten the time before parasites commence invasion. This effect on parasite invasion is distinct from the role that host microtubules play in bacterial and viral infections, where they function to traffic the pathogen or pathogen-derived material from the host cell's periphery to its interior. These data indicate that the host microtubule cytoskeleton is a structure used by Toxoplasma to rapidly infect its host cell and highlight a novel function for host microtubules in microbial pathogenesis.


Subject(s)
Host-Parasite Interactions/physiology , Microtubules/parasitology , Toxoplasma/pathogenicity , Amino Acid Sequence , Animals , CD59 Antigens/genetics , CD59 Antigens/physiology , Cell Line , Cytoskeleton/parasitology , Cytoskeleton/physiology , Host-Parasite Interactions/drug effects , Humans , Microtubules/drug effects , Microtubules/physiology , Molecular Sequence Data , Nocodazole/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thromboplastin/genetics , Thromboplastin/physiology , Virulence/physiology
19.
APMIS ; 117(5-6): 458-76, 2009 May.
Article in English | MEDLINE | ID: mdl-19400868

ABSTRACT

Toxoplasma gondii is an obligate intracellular protozoan parasite that can infect most warm-blooded animals and cause severe and life-threatening disease in developing fetuses and in immune-compromised patients. Although Toxoplasma was discovered over 100 years ago, we are only now beginning to appreciate the importance of the role that parasite modulation of its host has on parasite growth, bradyzoite development, immune evasion, and virulence. The goal of this review is to highlight these findings, to develop an integrated model for communication between Toxoplasma and its host, and to discuss new questions that arise out of these studies.


Subject(s)
Host-Parasite Interactions , Toxoplasma/physiology , Toxoplasmosis/parasitology , Animals , Cell Adhesion , Cell Cycle , Cytokines/physiology , Cytoskeleton/parasitology , Disease Progression , Gene Expression Regulation , Genotype , Host-Parasite Interactions/immunology , Humans , Mice , Organelles/parasitology , Protozoan Proteins/physiology , Toxoplasma/genetics , Toxoplasma/immunology , Toxoplasma/pathogenicity , Toxoplasmosis/immunology , Toxoplasmosis, Animal/immunology , Transcription, Genetic , Vacuoles/metabolism , Vacuoles/parasitology , Virulence
20.
Vet Res Commun ; 32(7): 521-31, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18668335

ABSTRACT

The first merogony of Eimeria bovis takes place in lymphatic endothelial cells of the ileum, resulting in the formation of macromeronts up to 250 microm. In this study, we investigated the host cell cytoskeleton (actin filaments, microtubules, spectrin, vimentin intermediate filaments) associated with parasitic development in vitro by confocal laser scanning microscopy (CLSM) using primary bovine umbilical vein endothelial cells (BUVEC) and bovine spleen lymphatic endothelial cells (BSLEC) as host cells. No prominent changes in the host cell cytoskeleton were detected 1-3 days after E. bovis sporozoite invasion. With ongoing meront maturation a significant increase in microtubules and actin filaments close to the parasitophorous vacuole (PV) was found. Mature macromeronts within the PV were completely enclosed by these cytoskeletal elements. Our findings suggest, that in order to guarantee the survival of the host cell on the enlargement of macromeronts, E. bovis needs not only to augment but also to rearrange its cytoskeletal system.


Subject(s)
Cytoskeleton/parasitology , Eimeria/physiology , Endothelial Cells/parasitology , Animals , Cattle , Cells, Cultured , Cytoskeleton/metabolism , Spectrin/metabolism , Umbilical Veins/cytology , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...