Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 39(2): 110691, 2022 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-35417716

RESUMEN

The innate immune recognition of the malaria-causing pathogen Plasmodium falciparum (P. falciparum) is not fully explored. Here, we identify the nucleoside 5'-methylthioinosine (MTI), a Plasmodium-specific intermediate of the purine salvage pathway, as a pathogen-derived Toll-like receptor 8 (TLR8) agonist. Co-incubation of MTI with the TLR8 enhancer poly(dT) as well as synthetic or P. falciparum-derived RNA strongly increase its stimulatory activity. Of note, MTI generated from methylthioadenosine (MTA) by P. falciparum lysates activates TLR8 when MTI metabolism is inhibited by immucillin targeting the purine nucleoside phosphorylase (PfPNP). Importantly, P. falciparum-infected red blood cells incubated with MTI or cultivated with MTA and immucillin lead to TLR8-dependent interleukin-6 (IL-6) production in human monocytes. Our data demonstrate that the nucleoside MTI is a natural human TLR8 ligand with possible in vivo relevance for innate sensing of P. falciparum.


Asunto(s)
Malaria Falciparum , Metiltioinosina , Receptor Toll-Like 8 , Humanos , Metiltioinosina/análogos & derivados , Nucleósidos , Plasmodium falciparum/metabolismo , Purina-Nucleósido Fosforilasa/metabolismo , Purinas , Receptor Toll-Like 8/metabolismo
2.
Int J Med Microbiol ; 308(1): 148-154, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29089241

RESUMEN

Human red blood cells infected with the malaria parasite Plasmodium falciparum show an increased permeability to a number of solutes. We have previously demonstrated that such infected cells take up glutamate via a member of the excitatory amino acid transporter protein family (EAAT), namely EAAT3. Babesia divergens is a parasite that also infects human erythrocytes, and also induces increased solute permeability, including for glutamate. Here we have investigated whether glutamate uptake in B. divergens infected human red blood cells is also dependent on EAAT3 activity. We find that, although B. divergens infected cells do take up glutamate, this uptake is independent on EAAT3. Thus, though infecting the same host cell, two related parasites have developed distinct pathways to obtain access to nutrients from the extracellular milieu.


Asunto(s)
Babesia/fisiología , Eritrocitos/parasitología , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Colina/farmacología , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/fisiología , Eritrocitos/efectos de los fármacos , Eritrocitos/fisiología , Transportador 3 de Aminoácidos Excitadores/antagonistas & inhibidores , Glutamatos/farmacología , Nitrobenzoatos/farmacología
3.
Eur J Cell Biol ; 94(7-9): 332-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26105829

RESUMEN

The mammalian red blood cell is a terminally differentiated cell that lacks a genetic programme and that has only a very limited metabolic capacity. Nonetheless, it serves as habitat for two parasites belonging to the monophyletic group of Apicomplexa, namely Plasmodium and Babesia. Studies of the parasitized red blood cell have revealed several properties that are unknown in the non-infected cell and that are difficult to conceptualize based on our view of red blood cell function. Here we review the current knowledge on host cell invasion and nutrient acquisition by these parasites. We attempt to dissect the factors that are directly contributed by the parasites from those that exist but have remained undetected in the non-infected cell.


Asunto(s)
Babesia/metabolismo , Fenómenos Fisiológicos Celulares/fisiología , Eritrocitos/parasitología , Plasmodium/metabolismo , Animales , Babesia/genética , Babesia/patogenicidad , Compartimento Celular/fisiología , Eritrocitos/fisiología , Humanos , Plasmodium/genética , Plasmodium/patogenicidad
4.
PLoS One ; 10(4): e0125191, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25909331

RESUMEN

Plasmodium falciparum invades human red blood cells, residing in a parasitophorous vacuole (PV), with a parasitophorous vacuole membrane (PVM) separating the PV from the host cell cytoplasm. Here we have investigated the role of N-myristoylation and two other N-terminal motifs, a cysteine potential S-palmitoylation site and a stretch of basic residues, as the driving force for protein targeting to the parasite plasma membrane (PPM) and subsequent translocation across this membrane. Plasmodium falciparum adenylate kinase 2 (Pf AK2) contains these three motifs, and was previously proposed to be targeted beyond the parasite to the PVM, despite the absence of a signal peptide for entry into the classical secretory pathway. Biochemical and microscopy analyses of PfAK2 variants tagged with green fluorescent protein (GFP) showed that these three motifs are involved in targeting the protein to the PPM and translocation across the PPM to the PV. It was shown that the N-terminal 37 amino acids of PfAK2 alone are sufficient to target and translocate GFP across the PPM. As a control we examined the N-myristoylated P. falciparum ADP-ribosylation factor 1 (PfARF1). PfARF1 was found to co-localise with a Golgi marker. To determine whether or not the putative palmitoylation and the cluster of lysine residues from the N-terminus of PfAK2 would modulate the subcellular localization of PfARF1, a chimeric fusion protein containing the N-terminus of PfARF1 and the two additional PfAK2 motifs was analysed. This chimeric protein was targeted to the PPM, but not translocated across the membrane into the PV, indicating that other features of the N-terminus of PfAK2 also play a role in the secretion process.


Asunto(s)
Malaria Falciparum/parasitología , Parásitos/fisiología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Vías Secretoras/fisiología , Factor 1 de Ribosilacion-ADP/metabolismo , Adenilato Quinasa/metabolismo , Secuencias de Aminoácidos/fisiología , Animales , Membrana Celular/metabolismo , Cisteína/metabolismo , Eritrocitos/parasitología , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lisina/metabolismo , Parásitos/metabolismo , Plasmodium falciparum/metabolismo , Señales de Clasificación de Proteína/fisiología , Transporte de Proteínas/fisiología , Vacuolas/metabolismo
5.
Cell Microbiol ; 17(7): 1052-68, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25628009

RESUMEN

Plasmodium falciparum invades human red blood cells (RBC), while Babesia divergens infects bovine and, occasionally, human RBC. The mammalian RBC is normally unable to endocytose or phagocytose and the events leading to invasion are incompletely understood. Initially, both parasites are surrounded by the RBC plasma membrane-derived parasitophorous vacuolar membrane (PVM) that is formed during invasion. In P. falciparum-infected RBC, the PVM persists at least until parasite replication is completed whereas it has been proposed that the B. divergens PVM is disintegrated soon upon invasion. Here, we have used a B. divergens strain adapted to human RBC to investigate the formation and fate of the PVM. Using ultrastructural analysis and whole-mount or on-section immunofluorescence and immunogold labelling, we demonstrate that the initial vacuolar membrane is formed from protein and lipid components of the RBC plasma membrane. Integral membrane proteins band 3 and glycophorin A and the cytoskeletal protein spectrin are associated with the PVM of the B. divergens, but are absent from the PVM of P. falciparum at the ring or the trophozoite stage. Our results provide evidence that the biophysical properties of the RBC cytoskeleton per se do not preclude the internalization of cytoskeletal proteins by invading parasites.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Babesia/fisiología , Endocitosis , Eritrocitos/parasitología , Glicoforinas/metabolismo , Interacciones Huésped-Patógeno , Espectrina/metabolismo , Técnicas Citológicas , Humanos , Vacuolas/química , Vacuolas/parasitología
6.
Med Microbiol Immunol ; 203(6): 383-93, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24985035

RESUMEN

The digestive vacuole (DV) of Plasmodium falciparum, which is released into the bloodstream upon rupture of each parasitized red blood cell (RBC), was recently discovered to activate the alternative complement pathway. In the present work, we show that C3- and C5-convertases assembling on the parasitic organelle are able to provoke deposition of activated C3 and C5b-9 on non-infected bystander erythrocytes. Direct contact of DVs with cells is mandatory for the effect, and bystander complement deposition occurs focally, possibly at the sites of contact. Complement opsonization promotes protracted erythrophagocytosis by human macrophages, an effect that is magnified when ring-stage infected RBCs with reduced CD55 and CD59, or paroxysmal nocturnal hemoglobinuria (PNH)-RBCs lacking these complement inhibitors are employed as targets. Bystander attack can also directly induce lysis of PNH-RBCs. Direct evidence for complement activation and bystander attack mediated by DVs was obtained through immunohistochemical analyses of brain paraffin sections from autopsies of patients who had died of cerebral malaria. C3d and the assembled C5b-9 complex could be detected in all sections, colocalizing with and often extending locally beyond massive accumulations of DVs that were identified under polarized light. This is the first demonstration that a complement-activating particle can mediate opsonization of bystander cells to promote their antibody-independent phagocytosis. The phenomenon may act in concert with other pathomechanisms to promote the development of anemia in patients with severe malaria.


Asunto(s)
Efecto Espectador , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Eritrocitos/inmunología , Fagocitosis , Plasmodium falciparum/inmunología , Vacuolas/inmunología , Encéfalo/patología , Eritrocitos/patología , Humanos , Inmunohistoquímica
7.
Nucleic Acids Res ; 42(11): 7113-31, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24799432

RESUMEN

Babesia spp. are tick-borne, intraerythrocytic hemoparasites that use antigenic variation to resist host immunity, through sequential modification of the parasite-derived variant erythrocyte surface antigen (VESA) expressed on the infected red blood cell surface. We identified the genomic processes driving antigenic diversity in genes encoding VESA (ves1) through comparative analysis within and between three Babesia species, (B. bigemina, B. divergens and B. bovis). Ves1 structure diverges rapidly after speciation, notably through the evolution of shortened forms (ves2) from 5' ends of canonical ves1 genes. Phylogenetic analyses show that ves1 genes are transposed between loci routinely, whereas ves2 genes are not. Similarly, analysis of sequence mosaicism shows that recombination drives variation in ves1 sequences, but less so for ves2, indicating the adoption of different mechanisms for variation of the two families. Proteomic analysis of the B. bigemina PR isolate shows that two dominant VESA1 proteins are expressed in the population, whereas numerous VESA2 proteins are co-expressed, consistent with differential transcriptional regulation of each family. Hence, VESA2 proteins are abundant and previously unrecognized elements of Babesia biology, with evolutionary dynamics consistently different to those of VESA1, suggesting that their functions are distinct.


Asunto(s)
Variación Antigénica , Babesia/genética , Evolución Molecular , Genes Protozoarios , Interacciones Huésped-Parásitos/genética , Puntos de Rotura del Cromosoma , Genoma de Protozoos , Proteínas Protozoarias/genética , Recombinación Genética
8.
Blood ; 119(18): 4301-10, 2012 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-22403252

RESUMEN

Severe Plasmodium falciparum malaria evolves through the interplay among capillary sequestration of parasitized erythrocytes, deregulated inflammatory responses, and hemostasis dysfunction. After rupture, each parasitized erythrocyte releases not only infective merozoites, but also the digestive vacuole (DV), a membrane-bounded organelle containing the malaria pigment hemozoin. In the present study, we report that the intact organelle, but not isolated hemozoin, dually activates the alternative complement and the intrinsic clotting pathway. Procoagulant activity is destroyed by phospholipase C treatment, indicating a critical role of phospholipid head groups exposed at the DV surface. Intravenous injection of DVs caused alternative pathway complement consumption and provoked apathy and reduced nociceptive responses in rats. Ultrasonication destroyed complement-activating and procoagulant properties in vitro and rendered the DVs biologically inactive in vivo. Low-molecular-weight dextran sulfate blocked activation of both complement and coagulation and protected animals from the harmful effects of DV infusion. We surmise that in chronic malaria, complement activation by and opsonization of the DV may serve a useful function in directing hemozoin to phagocytic cells for safe disposal. However, when the waste disposal system of the host is overburdened, DVs may transform into a trigger of pathology and therefore represent a potential therapeutic target in severe malaria.


Asunto(s)
Coagulación Sanguínea/fisiología , Vía Alternativa del Complemento/fisiología , Eritrocitos/parasitología , Plasmodium falciparum/fisiología , Vacuolas/fisiología , Animales , Coagulación Sanguínea/efectos de los fármacos , Vía Alternativa del Complemento/efectos de los fármacos , Sulfato de Dextran/farmacología , Hemoproteínas/fisiología , Hemólisis , Humanos , Hipoestesia/etiología , Membranas Intracelulares/fisiología , Pulmón/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/complicaciones , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Masculino , Monocitos/parasitología , Umbral del Dolor , Fagocitosis , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/ultraestructura , Ratas , Ratas Sprague-Dawley , Bazo/parasitología
9.
Blood ; 119(15): 3604-12, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22389252

RESUMEN

Human erythrocytes have a low basal permeability to L-glutamate and are not known to have a functional glutamate transporter. Here, treatment of human erythrocytes with arsenite was shown to induce the uptake of L-glutamate and D-aspartate, but not that of D-glutamate or L-alanine. The majority of the arsenite-induced L-glutamate influx was via a high-affinity, Na(+)-dependent system showing characteristics of members of the "excitatory amino acid transporter" (EAAT) family. Western blots and immunofluorescence assays revealed the presence of a member of this family, EAAT3, on the erythrocyte membrane. Erythrocytes infected with the malaria parasite Plasmodium falciparum take up glutamate from the extracellular environment. Although the majority of uptake is via a low-affinity Na(+)-independent pathway there is, in addition, a high-affinity uptake component, raising the possibility that the parasite activates the host cell glutamate transporter.


Asunto(s)
Eritrocitos/metabolismo , Transportador 3 de Aminoácidos Excitadores/agonistas , Ácido Glutámico/farmacocinética , Malaria Falciparum/metabolismo , Plasmodium falciparum/fisiología , Anestésicos/farmacología , Arsenitos/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Células Cultivadas , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Transportador 3 de Aminoácidos Excitadores/metabolismo , Ácido Glutámico/metabolismo , Ácido Glutámico/farmacología , Interacciones Huésped-Parásitos/fisiología , Humanos , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Pregnanodionas/farmacología , Estimulación Química , Teratógenos/farmacología
10.
Blood ; 118(18): 4946-56, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21911835

RESUMEN

Sequestration of parasitized erythrocytes and dysregulation of the coagulation and complement system are hallmarks of severe Plasmodium falciparum malaria. A link between these events emerged through the discovery that the parasite digestive vacuole (DV), which is released together with infective merozoites into the bloodstream, dually activates the intrinsic clotting and alternative complement pathway. Complement attack occurs exclusively on the membrane of the DVs, and the question followed whether DVs might be marked for uptake by polymorphonuclear granulocytes (PMNs). We report that DVs are indeed rapidly phagocytosed by PMNs after schizont rupture in active human serum. Uptake of malaria pigment requires an intact DV membrane and does not occur when the pigment is extracted from the organelle. Merozoites are not opsonized and escape phagocytosis in nonimmune serum. Antimalarial Abs mediate some uptake of the parasites, but to an extent that is not sufficient to markedly reduce reinvasion rates. Phagocytosis of DVs induces a vigorous respiratory burst that drives the cells into a state of functional exhaustion, blunting the production of reactive oxygen species (ROS) and microbicidal activity upon challenge with bacterial pathogens. Systemic overloading of PMNs with DVs may contribute to the enhanced susceptibility of patients with severe malaria toward invasive bacterial infections.


Asunto(s)
Neutrófilos/parasitología , Fagocitosis/fisiología , Plasmodium falciparum/patogenicidad , Vacuolas/fisiología , Animales , Recuento de Células Sanguíneas , Muerte Celular/inmunología , Eritrocitos/parasitología , Eritrocitos/patología , Humanos , Malaria Falciparum/sangre , Malaria Falciparum/inmunología , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Merozoítos/inmunología , Merozoítos/metabolismo , Merozoítos/patología , Merozoítos/fisiología , Modelos Biológicos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/fisiología , Fagocitosis/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/metabolismo , Plasmodium falciparum/ultraestructura , Especies Reactivas de Oxígeno/metabolismo , Especificidad por Sustrato , Factores de Tiempo , Vacuolas/metabolismo , Vacuolas/parasitología
11.
PLoS One ; 6(5): e19334, 2011 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-21573242

RESUMEN

BACKGROUND: Highly charged compounds typically suffer from low membrane permeability and thus are generally regarded as sub-optimal drug candidates. Nonetheless, the highly charged drug fosmidomycin and its more active methyl-derivative FR900098 have proven parasiticidal activity against erythrocytic stages of the malaria parasite Plasmodium falciparum. Both compounds target the isoprenoid biosynthesis pathway present in bacteria and plastid-bearing organisms, like apicomplexan parasites. Surprisingly, the compounds are inactive against a range of apicomplexans replicating in nucleated cells, including Toxoplasma gondii. METHODOLOGY/PRINCIPAL FINDINGS: Since non-infected erythrocytes are impermeable for FR90098, we hypothesized that these drugs are taken up only by erythrocytes infected with Plasmodium. We provide evidence that radiolabeled FR900098 accumulates in theses cells as a consequence of parasite-induced new properties of the host cell, which coincide with an increased permeability of the erythrocyte membrane. Babesia divergens, a related parasite that also infects human erythrocytes and is also known to induce an increase in membrane permeability, displays a similar susceptibility and uptake behavior with regard to the drug. In contrast, Toxoplasma gondii-infected cells do apparently not take up the compounds, and the drugs are inactive against the liver stages of Plasmodium berghei, a mouse malaria parasite. CONCLUSIONS/SIGNIFICANCE: Our findings provide an explanation for the observed differences in activity of fosmidomycin and FR900098 against different Apicomplexa. These results have important implications for future screens aimed at finding new and safe molecular entities active against P. falciparum and related parasites. Our data provide further evidence that parasite-induced new permeability pathways may be exploited as routes for drug delivery.


Asunto(s)
Antimaláricos/metabolismo , Antimaláricos/farmacología , Babesia/patogenicidad , Eritrocitos/metabolismo , Eritrocitos/parasitología , Fosfomicina/análogos & derivados , Plasmodium falciparum/patogenicidad , Animales , Babesia/efectos de los fármacos , Western Blotting , Células Cultivadas , Eritrocitos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Fosfomicina/metabolismo , Fosfomicina/farmacología , Humanos , Ratones , Plasmodium falciparum/efectos de los fármacos , Toxoplasma/efectos de los fármacos , Toxoplasma/patogenicidad
12.
PLoS One ; 6(3): e18396, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21483790

RESUMEN

Upon invading the host erythrocyte, the human malaria parasite P. falciparum lives and replicates within a membrane bound compartment referred to as the parasitophorous vacuole. Recently, interest in this compartment and its protein content has grown, due to the important roles these play in parasite egress and protein traffic to the host cell. Surprisingly, the function of many proteins within this compartment has not been experimentally addressed. Here, we study the importance of one of these proteins, termed PfPV1, for intra-erythrocytic parasite survival. Despite numerous attempts to inactivate the gene encoding PfPV1, we were unable to recover deletion mutants. Control experiments verified that the pv1 gene locus was per se open for gene targeting experiments, allowing us to exclude technical limitations in our experimental strategy. Our data provide strong genetic evidence that PfPV1 is essential for survival of blood stage P. falciparum, and further highlight the importance of parasitophorous vacuole proteins in this part of the parasite's life cycle.


Asunto(s)
Eritrocitos/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Southern Blotting , Western Blotting , Humanos , Estadios del Ciclo de Vida/genética , Estadios del Ciclo de Vida/fisiología , Plasmodium falciparum/genética , Reacción en Cadena de la Polimerasa , Proteínas Protozoarias/genética
13.
Cell Stress Chaperones ; 16(4): 389-401, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21191678

RESUMEN

Heat shock protein 70 (Hsp70) and heat shock protein 40 (Hsp40) function as molecular chaperones during the folding and trafficking of proteins within most cell types. However, the Hsp70-Hsp40 chaperone partnerships within the malaria parasite, Plasmodium falciparum, have not been elucidated. Only one of the 43 P. falciparum Hsp40s is predicted to be a cytosolic, canonical Hsp40 (termed PfHsp40) capable of interacting with the major cytosolic P. falciparum-encoded Hsp70, PfHsp70. Consistent with this hypothesis, we found that PfHsp40 is upregulated under heat shock conditions in a similar pattern to PfHsp70. In addition, PfHsp70 and PfHsp40 reside mainly in the parasite cytosol, as assessed using indirect immunofluorescence microscopy. Recombinant PfHsp40 stimulated the ATP hydrolytic rates of both PfHsp70 and human Hsp70 similar to other canonical Hsp40s of yeast (Ydj1) and human (Hdj2) origin. In contrast, the Hsp40-stimulated plasmodial and human Hsp70 ATPase activities were differentially inhibited in the presence of pyrimidinone-based small molecule modulators. To further probe the chaperone properties of PfHsp40, protein aggregation suppression assays were conducted. PfHsp40 alone suppressed protein aggregation, and cooperated with PfHsp70 to suppress aggregation. Together, these data represent the first cellular and biochemical evidence for a PfHsp70-PfHsp40 partnership in the malaria parasite, and furthermore that the plasmodial and human Hsp70-Hsp40 chaperones possess unique attributes that are differentially modulated by small molecules.


Asunto(s)
Proteínas del Choque Térmico HSP40/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Respuesta al Choque Térmico , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/metabolismo , Adenosina Trifosfatasas/metabolismo , Citosol/metabolismo , Expresión Génica , Hidrólisis , Plasmodium falciparum/genética , Regulación hacia Arriba
14.
Cell Microbiol ; 12(10): 1398-420, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20482550

RESUMEN

Plasmodium falciparum is predicted to transport over 300 proteins to the cytosol of its chosen host cell, the mature human erythrocyte, including 19 members of the Hsp40 family. Here, we have generated transfectant lines expressing GFP- or HA-Strep-tagged versions of these proteins, and used these to investigate both localization and other properties of these Hsp40 co-chaperones. These fusion proteins labelled punctate structures within the infected erythrocyte, initially suggestive of a Maurer's clefts localization. Further experiments demonstrated that these structures were distinct from the Maurer's clefts in protein composition. Transmission electron microscopy verifies a non-cleft localization for HA-Strep-tagged versions of these proteins. We were not able to label these structures with BODIPY-ceramide, suggesting a lower size and/or different lipid composition compared with the Maurer's clefts. Solubility studies revealed that the Hsp40-GFP fusion proteins appear to be tightly associated with membranes, but could be released from the bilayer under conditions affecting membrane cholesterol content or organization, suggesting interaction with a binding partner localized to cholesterol-rich domains. These novel structures are highly mobile in the infected erythrocyte, but based on velocity calculations, can be distinguished from the 'highly mobile vesicles' previously described. Our study identifies a further extra-parasitic structure in the P. falciparum-infected erythrocyte, which we name 'J-dots' (as their defining characteristic so far is the content of J-proteins). We suggest that these J-dots are involved in trafficking of parasite-encoded proteins through the cytosol of the infected erythrocyte.


Asunto(s)
Citosol/parasitología , Eritrocitos/parasitología , Proteínas del Choque Térmico HSP40/metabolismo , Interacciones Huésped-Parásitos , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas del Choque Térmico HSP40/genética , Hemaglutininas/genética , Hemaglutininas/metabolismo , Humanos , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Proteínas Protozoarias/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Coloración y Etiquetado/métodos , Estreptavidina/genética , Estreptavidina/metabolismo
15.
Protoplasma ; 240(1-4): 3-12, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19949823

RESUMEN

Apicomplexan parasites obligatorily invade and multiply within eukaryotic cells. Phylogenetically, they are related to a group of algae which, during their evolution, have acquired a secondary endosymbiont. This organelle, which in the parasite is called the apicoplast, is highly reduced compared to the endosymbionts of algae, but still contains many plant-specific biosynthetic pathways. The malaria parasite Plasmodium falciparum infects mammalian erythrocytes which are devoid of intracellular compartments and which largely lack biosynthetic pathways. Despite the limited resources of nutrition, the parasite grows and generates up to 32 merozoites which are the infectious stages of the complex life cycle. A large part of the intra-erythrocytic development takes place in the so-called parasitophorous vacuole, a compartment which forms an interface between the parasite and the cytoplasm of the host cell. In the course of parasite growth, the host cell undergoes dramatic alterations which on one hand contribute directly to the symptoms of severe malaria and which, on the other hand, are also required for parasite survival. Some of these alterations facilitate the acquisition of nutrients from the extracellular environment which are not provided by the host cell. Here, we describe the cell biologically unique interactions between an intracellular eukaryotic pathogen and its metabolically highly reduced host cell. We further discuss current models to explain the appearance of pathogen-induced novel physiological properties in a host cell which has lost its genetic programme.


Asunto(s)
Plasmodium falciparum/fisiología , Plasmodium falciparum/patogenicidad , Animales , Compartimento Celular , Permeabilidad de la Membrana Celular , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Eritrocitos/ultraestructura , Interacciones Huésped-Parásitos/fisiología , Humanos , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Modelos Biológicos , Orgánulos/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Vacuolas/parasitología
16.
Mol Biochem Parasitol ; 167(1): 48-53, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19393693

RESUMEN

The molecular mechanisms underlying the formation of the parasitophorous vacuolar membrane in Plasmodium falciparum infected erythrocytes are incompletely understood, and the protein composition of this membrane is still enigmatic. Although the differentiated mammalian erythrocyte lacks the machinery required for endocytosis, some reports have described a localisation of host cell membrane proteins at the parasitophorous vacuolar membrane. Aquaporin 3 is an abundant plasma membrane protein of various cells, including mammalian erythrocytes where it is found in distinct oligomeric states. Here we show that human aquaporin 3 is internalized into infected erythrocytes, presumably during or soon after invasion. It is integrated into the PVM where it is organized in novel oligomeric states which are not found in non-infected cells.


Asunto(s)
Acuaporina 3/análisis , Membrana Celular/química , Eritrocitos/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Vacuolas/parasitología , Animales , Humanos
17.
Mol Microbiol ; 71(3): 613-28, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19040635

RESUMEN

Plasmodium falciparum traffics a large number of proteins to its host cell, the mature human erythrocyte. How exactly these proteins gain access to the red blood cell is poorly understood. Here we have investigated the effect of protein folding on the transport of model substrate proteins to the host cell. We find that proteins must pass into the erythrocyte cytoplasm in an unfolded state. Our data strongly support the presence of a protein-conducting channel in the parasitophorous vacuolar membrane, and additionally imply an important role for molecular chaperones in keeping parasite proteins in a 'translocation competent' state prior to membrane passage.


Asunto(s)
Eritrocitos/metabolismo , Plasmodium falciparum/metabolismo , Pliegue de Proteína , Proteínas Protozoarias/metabolismo , Vacuolas/metabolismo , Animales , Permeabilidad de la Membrana Celular , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Humanos , Transporte de Proteínas , Vacuolas/parasitología
18.
Cell Physiol Biochem ; 22(5-6): 395-404, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19088421

RESUMEN

The intraerythrocytic development of P. falciparum induces New Permeability Pathways (NPP) in the membrane of the parasitized erythrocyte which provide the parasite with nutrients, adjust the erythrocyte electrolyte composition to the needs of the parasite, and dispose of metabolic waste products and osmolytes. Patch-clamp recordings identified inwardly and outwardly rectifying (OR) anion conductances in the host erythrocyte membrane as electrophysiological correlate of the NPP. The OR conductance is regulated by serum. Here we show that serum albumin (SA) stimulated OR-generated Cl(-) and lactate outward currents with an EC(50) of approximately 100 nM while other proteins such as ovalbumin or casein did not. The stimulatory efficacy did not differ between fatty acid free bovine SA and recombinant human SA and disruption of the SA tertiary structure abolished the effect suggesting that intact SA protein and not other bound factors interact with the erythrocyte membrane. Taken together, the data indicate a high affinity and specificity interaction of native SA with the parasitized erythrocytes which might underlie the observed dependence of P. falciparum growth in vitro on SA.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/parasitología , Activación del Canal Iónico , Malaria/metabolismo , Plasmodium falciparum/metabolismo , Albúmina Sérica/metabolismo , Animales , Caseínas/farmacología , Canales de Cloruro/metabolismo , Eritrocitos/efectos de los fármacos , Humanos , Activación del Canal Iónico/efectos de los fármacos , Ácido Láctico/metabolismo , Ovalbúmina/farmacología , Plasmodium falciparum/efectos de los fármacos , Proteínas Recombinantes/metabolismo , Albúmina Sérica Bovina/farmacología
19.
PLoS One ; 3(2): e1560, 2008 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-18253504

RESUMEN

We performed a bioinformatical analysis of protein export elements (PEXEL) in the putative proteome of the malaria parasite Plasmodium falciparum. A protein family-specific conservation of physicochemical residue profiles was found for PEXEL-flanking sequence regions. We demonstrate that the family members can be clustered based on the flanking regions only and display characteristic hydrophobicity patterns. This raises the possibility that the flanking regions may contain additional information for a family-specific role of PEXEL. We further show that signal peptide cleavage results in a positional alignment of PEXEL from both proteins with, and without, a signal peptide.


Asunto(s)
Biología Computacional , Plasmodium falciparum/química , Señales de Clasificación de Proteína , Proteoma , Proteínas Protozoarias/química , Animales , Análisis por Conglomerados , Interacciones Hidrofóbicas e Hidrofílicas
20.
J Biol Chem ; 282(30): 21609-17, 2007 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-17545162

RESUMEN

Sulfosuccinimidyl-6-(biotinamido) hexanoate and derivatives thereof covalently bind to the epsilon-amino group of lysine residues. Our observation that access of the biotin derivative to specific lysine residues depends on conformational properties of the entire polypeptide chain prompted us to investigate whether differential biotinylation patterns of a protein can be used as indicators for conformational changes. Bovine serum albumin is a soluble protein with characteristic unfolding kinetics upon exposure to high temperature. First, we show that biotinylation patterns of proteins are highly reproducible. Second, we demonstrate by mass spectrometry and tandem mass spectrometry that unfolding of the protein correlates with the accessibility of the biotin derivative to specific lysine residues. We have applied this experimental strategy to the analysis of a cell-surface protein, viz. the human band 3 anion exchanger of erythrocytes infected with the malaria parasite Plasmodium falciparum. We found that Lys(826) in a highly flexible loop can be biotinylated in non-infected (but not infected) erythrocytes, confirming earlier observations (Winograd, E., and Sherman, I. W. (2004) Mol. Biochem. Parasitol. 138, 83-87) based on epitope-specific monoclonal antibodies suggesting that this region undergoes a conformational change upon infection.


Asunto(s)
Biotina/análogos & derivados , Biotina/química , Eritrocitos/parasitología , Lisina , Proteínas de la Membrana/química , Conformación Proteica , Proteínas/química , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteínas Sanguíneas/química , Proteínas Sanguíneas/aislamiento & purificación , Anhidrasas Carbónicas/química , Membrana Eritrocítica/química , Humanos , Espectrometría de Masas , Proteínas de la Membrana/sangre , Proteínas de la Membrana/aislamiento & purificación , Plasmodium falciparum , Albúmina Sérica Bovina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...