Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 11(1): 3532, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32669539

ABSTRACT

Asexual proliferation of the Plasmodium parasites that cause malaria follows a developmental program that alternates non-canonical intraerythrocytic replication with dissemination to new host cells. We carried out a functional analysis of the Plasmodium falciparum homolog of Protein Phosphatase 1 (PfPP1), a universally conserved cell cycle factor in eukaryotes, to investigate regulation of parasite proliferation. PfPP1 is indeed required for efficient replication, but is absolutely essential for egress of parasites from host red blood cells. By phosphoproteomic and chemical-genetic analysis, we isolate two functional targets of PfPP1 for egress: a HECT E3 protein-ubiquitin ligase; and GCα, a fusion protein composed of a guanylyl cyclase and a phospholipid transporter domain. We hypothesize that PfPP1 regulates lipid sensing by GCα and find that phosphatidylcholine stimulates PfPP1-dependent egress. PfPP1 acts as a key regulator that integrates multiple cell-intrinsic pathways with external signals to direct parasite egress from host cells.


Subject(s)
Erythrocytes/parasitology , Plasmodium falciparum/enzymology , Protein Phosphatase 1/metabolism , Protozoan Proteins/metabolism , Animals , Cell Proliferation , Cyclic GMP/metabolism , Gene Expression Regulation, Enzymologic , Humans , Inhibitory Concentration 50 , Mice , Mice, Knockout , Phosphatidylcholines/chemistry , Protein Domains , Proteome , Ubiquitin-Protein Ligases/metabolism
2.
Nano Lett ; 20(6): 4249-4255, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32369369

ABSTRACT

Although near-field imaging techniques reach sub-nanometer resolution on rigid samples, it remains extremely challenging to image soft interfaces, such as biological membranes, due to the deformations induced by the probe. In photonic force microscopy, optical tweezers are used to manipulate and measure the scanning probe, allowing imaging of soft materials without force-induced artifacts. However, the size of the optically trapped probe still limits the maximum resolution. Here, we show a novel and simple nanofabrication protocol to massively produce optically trappable quartz particles which mimic the sharp tips of atomic force microscopy. Imaging rigid nanostructures with our tips, we resolve features smaller than 80 nm. Scanning the membrane of living malaria-infected red blood cells reveals, with no visible artifacts, submicron features termed knobs, related to the parasite activity. The use of nanoengineered particles in photonic force microscopy opens the way to imaging soft samples at high resolution.

3.
Sci Rep ; 8(1): 3543, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29476099

ABSTRACT

Malaria parasites alternate between intracellular and extracellular stages and successful egress from the host cell is crucial for continuation of the life cycle. We investigated egress of Plasmodium berghei gametocytes, an essential process taking place within a few minutes after uptake of a blood meal by the mosquito. Egress entails the rupture of two membranes surrounding the parasite: the parasitophorous vacuole membrane (PVM), and the red blood cell membrane (RBCM). High-speed video microscopy of 56 events revealed that egress in both genders comprises four well-defined phases, although each event is slightly different. The first phase is swelling of the host cell, followed by rupture and immediate vesiculation of the PVM. These vesicles are extruded through a single stabilized pore of the RBCM, and the latter is subsequently vesiculated releasing the free gametes. The time from PVM vesiculation to completion of egress varies between events. These observations were supported by immunofluorescence microscopy using antibodies against proteins of the RBCM and PVM. The combined results reveal dynamic re-organization of the membranes and the cortical cytoskeleton of the erythrocyte during egress.


Subject(s)
Erythrocyte Membrane/ultrastructure , Malaria/parasitology , Plasmodium berghei/genetics , Vacuoles/ultrastructure , Animals , Culicidae/parasitology , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Erythrocyte Membrane/parasitology , Erythrocytes/parasitology , Erythrocytes/ultrastructure , Germ Cells/metabolism , Germ Cells/ultrastructure , Humans , Life Cycle Stages/genetics , Malaria/transmission , Plasmodium berghei/pathogenicity , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Vacuoles/parasitology
4.
PLoS One ; 12(12): e0189556, 2017.
Article in English | MEDLINE | ID: mdl-29244879

ABSTRACT

During the infection process, Apicomplexa discharge their secretory organelles called micronemes, rhoptries and dense granules to sustain host cell invasion, intracellular replication and to modulate host cell pathways and immune responses. Herein, we describe the Toxoplasma gondii Deg-like serine protein (TgDegP), a rhoptry protein homologous to High temperature requirement A (HtrA) or Deg-like family of serine proteases. TgDegP undergoes processing in both types I and II strains as most of the rhoptries proteins. We show that genetic disruption of the degP gene does not impact the parasite lytic cycle in vitro but affects virulence in mice. While in a type I strain DegPI appears dispensable for the establishment of an infection, removal of DegPII in a type II strain dramatically impairs the virulence. Finally, we show that KO-DegPII parasites kill immunodeficient mice as efficiently as the wild-type strain indicating that the protease might be involved in the complex crosstalk that the parasite engaged with the host immune response. Thus, this study unravels a novel rhoptry protein in T. gondii important for the establishment of lethal infection.


Subject(s)
Protozoan Proteins/physiology , Serine Proteases/physiology , Toxoplasma/enzymology , Toxoplasmosis/parasitology , Animals , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Protein Processing, Post-Translational , Proteolysis , Toxoplasma/genetics , Toxoplasma/pathogenicity , Virulence
5.
PLoS One ; 12(10): e0187073, 2017.
Article in English | MEDLINE | ID: mdl-29073264

ABSTRACT

During the erythrocytic cycle of the malaria parasite Plasmodium falciparum, egress and invasion are essential steps finely controlled by reversible phosphorylation. In contrast to the growing number of kinases identified as key regulators, phosphatases have been poorly studied, and calcineurin is the only one identified so far to play a role in invasion. PfShelph2, a bacterial-like phosphatase, is a promising candidate to participate in the invasion process, as it was reported to be expressed late during the asexual blood stage and to reside within an apical compartment, yet distinct from rhoptry bulb, micronemes, or dense granules. It was also proposed to play a role in the control of the red blood cell membrane deformability at the end of the invasion process. However, genetic studies are still lacking to support this hypothesis. Here, we take advantage of the CRISPR-Cas9 technology to tag shelph2 genomic locus while retaining its endogenous regulatory regions. This new strain allows us to follow the endogenous PfShelph2 protein expression and location during asexual blood stages. We show that PfShelph2 apical location is also distinct from the rhoptry neck or exonemes. We further demonstrate PfShelph2 dispensability during the asexual blood stage by generating PfShelph2-KO parasites using CRISPR-Cas9 machinery. Analyses of the mutant during the course of the erythrocytic development indicate that there are no detectable phenotypic consequences of Pfshelph2 genomic deletion. As this lack of phenotype might be due to functional redundancy, we also explore the likelihood of PfShelph1 (PfShelph2 paralog) being a compensatory phosphatase. We conclude that despite its cyclic expression profile, PfShelph2 is a dispensable phosphatase during the Plasmodium falciparum asexual blood stage, whose function is unlikely substituted by PfShelph1.


Subject(s)
Phosphoric Monoester Hydrolases/metabolism , Plasmodium falciparum/enzymology , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Erythrocytes/parasitology , Phosphoric Monoester Hydrolases/genetics , Plasmodium falciparum/growth & development
6.
Trends Parasitol ; 32(2): 90-92, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26652974

ABSTRACT

Das et al. recently reported a role for the major merozoite surface protein MSP1 in malarial parasite egress from the red blood cell (RBC). On the basis of these new data and physical considerations, we propose an updated model for the main steps of this essential process for parasite proliferation.


Subject(s)
Erythrocytes/parasitology , Merozoite Surface Protein 1/metabolism , Merozoites/physiology , Plasmodium falciparum/physiology , Protein Processing, Post-Translational , Protozoan Proteins/metabolism , Spectrin/metabolism , Subtilisins/metabolism , Humans
7.
Traffic ; 16(5): 461-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25615740

ABSTRACT

The export of numerous proteins to the plasma membrane of its host erythrocyte is essential for the virulence and survival of the malaria parasite Plasmodium falciparum. The Maurer's clefts, membrane structures transposed by the parasite in the cytoplasm of its host erythrocyte, play the role of a marshal platform for such exported parasite proteins. We identify here the export pathway of three resident proteins of the Maurer's clefts membrane: the proteins are exported as soluble forms in the red cell cytoplasm to the Maurer's clefts membrane in association with the parasite group II chaperonin (PfTRIC), a chaperone complex known to bind and address a large spectrum of unfolded proteins to their final location. We have also located the domain of interaction with PfTRiC within the amino-terminal domain of one of these Maurer's cleft proteins, PfSBP1. Because several Maurer's cleft membrane proteins with different export motifs seem to follow the same route, we propose a general role for PfTRiC in the trafficking of malarial parasite proteins to the host erythrocyte.


Subject(s)
Erythrocytes/parasitology , Group II Chaperonins/metabolism , Host-Parasite Interactions/physiology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Cells, Cultured , Cytoplasm/metabolism , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Group II Chaperonins/genetics , Humans , Merozoites/metabolism , Merozoites/parasitology , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Protein Transport , Protozoan Proteins/genetics
8.
J Biol Chem ; 288(38): 27002-27018, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23913689

ABSTRACT

In eukaryotic organisms, cysteine palmitoylation is an important reversible modification that impacts protein targeting, folding, stability, and interactions with partners. Evidence suggests that protein palmitoylation contributes to key biological processes in Apicomplexa with the recent palmitome of the malaria parasite Plasmodium falciparum reporting over 400 substrates that are modified with palmitate by a broad range of protein S-acyl transferases. Dynamic palmitoylation cycles require the action of an acyl-protein thioesterase (APT) that cleaves palmitate from substrates and conveys reversibility to this posttranslational modification. In this work, we identified candidates for APT activity in Toxoplasma gondii. Treatment of parasites with low micromolar concentrations of ß-lactone- or triazole urea-based inhibitors that target human APT1 showed varied detrimental effects at multiple steps of the parasite lytic cycle. The use of an activity-based probe in combination with these inhibitors revealed the existence of several serine hydrolases that are targeted by APT1 inhibitors. The active serine hydrolase, TgASH1, identified as the homologue closest to human APT1 and APT2, was characterized further. Biochemical analysis of TgASH1 indicated that this enzyme cleaves substrates with a specificity similar to APTs, and homology modeling points toward an APT-like enzyme. TgASH1 is dispensable for parasite survival, which indicates that the severe effects observed with the ß-lactone inhibitors are caused by the inhibition of non-TgASH1 targets. Other ASH candidates for APT activity were functionally characterized, and one of them was found to be resistant to gene disruption due to the potential essential nature of the protein.


Subject(s)
Enzyme Inhibitors/pharmacology , Lactones/pharmacology , Protozoan Proteins/antagonists & inhibitors , Thiolester Hydrolases/antagonists & inhibitors , Toxoplasma/enzymology , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Humans , Lactones/chemistry , Models, Molecular , Molecular Sequence Data , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Structural Homology, Protein , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Toxoplasma/genetics , Toxoplasmosis/drug therapy , Toxoplasmosis/enzymology , Toxoplasmosis/genetics
9.
Mol Microbiol ; 88(2): 425-42, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23517413

ABSTRACT

The pathogenicity of the most deadly human malaria parasite, Plasmodium falciparum, relies on the export of virulence factors to the surface of infected erythrocytes. A novel membrane compartment, referred to as Maurer's clefts, is transposed to the host erythrocyte, acting as a marshal platform in the red blood cell cytoplasm, for exported parasite proteins addressed to the host cell plasma membrane. We report here the characterization of three new P. falciparum multigene families organized in 9 highly conserved clusters with the Pfmc-2tm genes in the subtelomeric regions of parasite's chromosomes and expressed at early trophozoite stages. Like the PfMC-2TM proteins, the PfEPF1, 3 and 4 proteins encoded by these families are exported to the Maurer's clefts, as peripheral or integral proteins of the Maurer's cleft membrane and largely exposed to the red cell cytosolic face of this membrane. A promoter titration approach was used to question the biological roles of these P. falciparum-specific exported proteins. Using the Pfepf1 family promoter, we observed the specific downregulation of all four families, correlating with the inefficient release of merozoites while the parasite intra-erythrocytic maturation and Maurer's clefts morphology were not impacted.


Subject(s)
Erythrocytes/parasitology , Host-Parasite Interactions , Merozoites/physiology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Animals , Cytoplasm/metabolism , Cytosol/metabolism , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/parasitology , Erythrocytes/cytology , Erythrocytes/metabolism , Humans , Intracellular Membranes , Membrane Proteins/metabolism , Multigene Family , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protein Transport , Protozoan Proteins/genetics , Trophozoites/metabolism , Vacuoles/metabolism , Vacuoles/parasitology
10.
Infect Genet Evol ; 16: 450-4, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23416259

ABSTRACT

For the second consecutive year, teams of the network "Montpellier Infectious Diseases" held their annual meeting. Whereas the 2011 meeting was focused on host-pathogen interaction and pathophysiology, the 2012 meeting was focused on the cooperation between medical and chemical sciences interdisciplinary approaches to fight against virus, bacteria and parasites. Several approaches aimed at designing new bioactive compounds were described during this meeting.


Subject(s)
Communicable Diseases/drug therapy , Animals , Anti-Infective Agents/therapeutic use , Communicable Diseases/microbiology , Communicable Diseases/parasitology , Host-Pathogen Interactions , Humans , Occupational Health , Virulence
11.
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
12.
Blood ; 117(15): 4118-24, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21297002

ABSTRACT

The culminating step of the intraerythrocytic development of Plasmodium falciparum, the causative agent of malaria, is the spectacular release of multiple invasive merozoites on rupture of the infected erythrocyte membrane. This work reports for the first time that the whole process, taking place in time scales as short as 400 milliseconds, is the result of an elastic instability of the infected erythrocyte membrane. Using high-speed differential interference contrast (DIC) video microscopy and epifluorescence, we demonstrate that the release occurs in 3 main steps after osmotic swelling of the infected erythrocyte: a pore opens in ~ 100 milliseconds, ejecting 1-2 merozoites, an outward curling of the erythrocyte membrane is then observed, ending with a fast eversion of the infected erythrocyte membrane, pushing the parasites forward. It is noteworthy that this last step shows slight differences when infected erythrocytes are adhering. We rationalize our observations by considering that during the parasite development, the infected erythrocyte membrane acquires a spontaneous curvature and we present a subsequent model describing the dynamics of the curling rim. Our results show that sequential erythrocyte membrane curling and eversion is necessary for the parasite efficient angular dispersion and might be biologically essential for fast and numerous invasions of new erythrocytes.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Merozoites/growth & development , Plasmodium falciparum/growth & development , Animals , Cell Shape/physiology , Elasticity/physiology , Erythrocyte Membrane/parasitology , Erythrocytes/cytology , Humans , In Vitro Techniques , Merozoites/metabolism , Osmotic Pressure/physiology , Pancreatic Elastase/metabolism , Plasmodium falciparum/metabolism
13.
Int J Parasitol ; 40(11): 1257-68, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20385136

ABSTRACT

Cytidine diphosphate diacylglycerol synthase (CDS) diverts phosphatidic acid towards the biosynthesis of CDP-DAG, an obligatory liponucleotide intermediate in anionic phospholipid biosynthesis. The 78kDa predicted Plasmodium falciparum CDS (PfCDS) is recovered as a 50 kDa conserved C-terminal cytidylyltransferase domain (C-PfCDS) and a 28kDa fragment that corresponds to the unusually long hydrophilic asparagine-rich N-terminal extension (N-PfCDS). Here, we show that the two fragments of PfCDS are the processed forms of the 78 kDa pro-form that is encoded from a single transcript with no alternate translation start site for C-PfCDS. PfCDS, which shares 54% sequence identity with Plasmodium knowlesi CDS (PkCDS), could substitute for PkCDS in P. knowlesi. Experiments to disrupt either the full-length or the N-terminal extension of PkCDS indicate that not only the C-terminal cytidylyltransferase domain but also the N-terminal extension is essential to Plasmodium spp. PkCDS and PfCDS introduced in P. knowlesi were processed in the parasite, suggesting a conserved parasite-dependent mechanism. The N-PfCDS appears to be a peripheral membrane protein and is trafficked outside the parasite to the parasitophorous vacuole. Although the function of this unusual N-PfCDS remains enigmatic, the study here highlights features of this essential gene and its biological importance during the intra-erythrocytic cycle of the parasite.


Subject(s)
Diacylglycerol Cholinephosphotransferase/chemistry , Diacylglycerol Cholinephosphotransferase/metabolism , Plasmodium falciparum/enzymology , Plasmodium knowlesi/enzymology , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Chlorocebus aethiops , Cytidine Diphosphate Diglycerides/biosynthesis , Diacylglycerol Cholinephosphotransferase/genetics , Erythrocytes/parasitology , Humans , Malaria/parasitology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Plasmodium knowlesi/chemistry , Plasmodium knowlesi/genetics , Plasmodium knowlesi/growth & development , Protein Structure, Tertiary , Protozoan Proteins/genetics
14.
Traffic ; 10(2): 137-52, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19054387

ABSTRACT

The human malaria parasite Plasmodium falciparum exports determinants of virulence and pathology to destinations within the host erythrocyte, including the erythrocyte cytoplasm, plasma membrane and membrane profiles of parasite origin termed Maurer's clefts. Most of the exported proteins contain a conserved pentameric motif termed plasmodial export element (PEXEL)/vacuolar transfer signal (VTS) that functions as a cleavable sorting signal permitting export to the host erythrocyte. However, there are some exported proteins, such as the skeleton-binding protein 1 (PfSBP1) that lack the PEXEL/VTS motif and that are not N-terminally processed, suggesting the presence of alternative sorting signals and/or mechanisms. In this study, we have investigated trafficking of PfSBP1 to the Maurer's clefts. Our data show that the transmembrane domain of PfSBP1 functions as an internal signal sequence for entry into the parasite's secretory pathway and for transport to the parasite plasma membrane. Trafficking beyond the parasite's plasma membrane required additional N-terminal domains, which are characterized by a high negative net charge. Biochemical data indicate that these domains affect the solubility and extraction profile, the orientation of the protein within the membrane and the subcellular localization. Our findings suggest new principles of protein export in P. falciparum-infected erythrocytes.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Membrane/metabolism , Gene Deletion , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Protein Binding , Protein Transport , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Trypsin/metabolism
15.
Mol Biochem Parasitol ; 160(2): 81-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18508137

ABSTRACT

The high-molecular mass rhoptry protein complex (PfRhopH), which comprises three distinct gene products, RhopH1, RhopH2, and RhopH3, is known to be secreted and transferred to the parasitophorous vacuole membrane upon invasion of a red blood cell by the malaria parasite Plasmodium falciparum. Here we show that the merozoite-acquired RhopH complex is also transferred to defined domains of the red blood cell cytoplasm, and possibly transiently associated with Maurer's clefts. This is the first report of trafficking in the host cell cytoplasm for P. falciparum rhoptry proteins secreted upon red blood cell invasion. Based on its newly identified sub-cellular location and the phenotype of RhopH1 mutants, we propose that the RhopH complex participate in the assembly of the cytoadherence complex.


Subject(s)
Cytoplasm/chemistry , Erythrocytes/parasitology , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Animals , Blotting, Western , Immunoprecipitation , Microscopy, Fluorescence , Protein Transport
16.
Cell Microbiol ; 8(4): 591-601, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16548885

ABSTRACT

The malarial parasite Plasmodium falciparum transposes a Golgi-like compartment, referred to as Maurer's clefts, into the cytoplasm of its host cell, the erythrocyte, and delivering parasite molecules to the host cell surface. We report here a novel role of the Maurer's clefts implicating a parasite protein phosphatase 1 (PP1) and related to the phosphorylation status of P. falciparum skeleton-binding protein 1 (PfSBP1), a trans-membrane protein of the clefts interacting with the host cell membrane via its carboxy-terminal domain. Based on co-immunoprecipitation and inhibition studies, we show that the parasite PP1 type phosphatase modulates the phosphorylation status of the amino-terminal domain of PfSBP1 in the lumen of Maurer's clefts. Importantly, the addition of a PP1 inhibitor, calyculin A, to late schizonts results in the hyperphosphorylation of PfSBP1 and prevents parasite release from the host cell. We propose that the hyperphosphorylation of PfSBP1 interferes with the release of merozoites, the invasive blood stage of the parasite, by increasing the red cell membrane stability. Moreover, the parasite PP1 phosphatase is the first enzyme essential for the parasite development detected in the Maurer's clefts.


Subject(s)
Erythrocytes/parasitology , Phosphoprotein Phosphatases/metabolism , Plasmodium falciparum/physiology , Amino Acid Sequence , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Membrane/metabolism , Erythrocytes/metabolism , Golgi Apparatus/metabolism , In Vitro Techniques , Marine Toxins , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Molecular Sequence Data , Oxazoles/pharmacology , Phosphorylation , Plasmodium falciparum/enzymology , Protein Phosphatase 1 , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism
17.
Int J Parasitol ; 36(1): 23-36, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16337634

ABSTRACT

Discovered in 1902 by Georg Maurer as a peculiar dotted staining pattern observable by light microscopy in the cytoplasm of erythrocytes infected with the human malarial parasite Plasmodium falciparum, the function of Maurer's clefts have remained obscure for more than a century. The growing interest in protein sorting and trafficking processes in malarial parasites has recently aroused the Maurer's clefts from their deep slumber. Mounting evidence suggests that Maurer's clefts are a secretory organelle, which the parasite establishes within its host erythrocyte, but outside its own confines, to route parasite proteins across the host cell cytoplasm to the erythrocyte surface where they play a role in nutrient uptake and immune evasion processes. Moreover, Maurer's clefts seem to play a role in cell signaling, merozoite egress, phospholipid biosynthesis and, possibly, other biochemical pathways. Here, we review our current knowledge of the ultrastructure of Maurer's clefts, their proteinaceous composition and their function in protein trafficking.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/pathology , Organelles/parasitology , Cytoplasm/metabolism , Cytoplasm/parasitology , Cytoplasm/pathology , Cytoskeleton/metabolism , Cytoskeleton/parasitology , Cytoskeleton/pathology , Erythrocyte Membrane/metabolism , Erythrocyte Membrane/parasitology , Erythrocyte Membrane/pathology , Erythrocytes/metabolism , Erythrocytes/pathology , Host-Parasite Interactions , Humans , Malaria, Falciparum/metabolism , Models, Biological , Organelles/enzymology , Organelles/pathology , Protozoan Proteins/blood , Signal Transduction/physiology , Vesicular Transport Proteins/metabolism
18.
Mol Biochem Parasitol ; 141(1): 39-47, 2005 May.
Article in English | MEDLINE | ID: mdl-15811525

ABSTRACT

As the malarial parasite Plasmodium falciparum develops inside the erythrocyte, parasite-derived membrane structures, referred to as Maurer's clefts, play an important role in parasite development by delivering parasite proteins to the host cell surface, and participating in the assembly of the cytoadherence complex, essential for the pathogenesis of cerebral malaria. PfSBP1 is an integral membrane protein of the clefts, interacting with an erythrocyte cytosolic protein, identified here as the human Lantibiotic synthetase component C-like protein LANCL1. LANCL1 is specifically recruited to the surface of Maurer's clefts in P. falciparum mature blood stages. We propose that the interaction between PfSBP1 and LANCL1 is central for late steps of the parasite development to prevent premature rupture of the red blood cell membrane.


Subject(s)
Carrier Proteins/metabolism , Erythrocytes/metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Erythrocyte Membrane/metabolism , Fluorescent Antibody Technique , Humans , Intracellular Membranes/metabolism , Molecular Sequence Data , Molecular Weight , Peptide Fragments/metabolism , Plasmodium falciparum/growth & development , Protein Binding , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/metabolism
19.
Mol Cell Proteomics ; 4(4): 582-93, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15671043

ABSTRACT

A novel method was validated for the efficient distinction between malaria parasite-derived and host cell proteins in mass spectrometry analyses. This method was applied to a ghost fraction from Plasmodium falciparum-infected erythrocytes containing the red blood cell plasma membrane, the erythrocyte submembrane skeleton, and the Maurer's clefts, a Golgi-like apparatus linked to and addressing parasite proteins to the host cell surface. This method allowed the identification of 78 parasite proteins. Among these we identified seven novel proteins of the Maurer's clefts based on immunofluorescence studies and proteinase K digestion assays. The products of six contiguous genes located on chromosome 5 were identified, and the location within the Maurer's clefts was established for two of them. This suggests a clustering of genes encoding Maurer's cleft proteins. Our study sheds new light on the biological function of the Maurer's clefts, which are central to the pathogenesis and to the intraerythrocytic development of P. falciparum.


Subject(s)
Erythrocyte Membrane/chemistry , Erythrocyte Membrane/parasitology , Membrane Proteins/analysis , Plasmodium falciparum/physiology , Protozoan Proteins/analysis , Animals , Blotting, Western , DNA Primers , DNA, Protozoan/chemistry , Deuterium/metabolism , Endopeptidase K/metabolism , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Glutathione Transferase/metabolism , Host-Parasite Interactions , Humans , Life Cycle Stages , Malaria, Falciparum/parasitology , Mass Spectrometry , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Nucleic Acid Amplification Techniques , Octoxynol/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Plasmodium falciparum/growth & development , Polymerase Chain Reaction , Protozoan Proteins/chemistry , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...