Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
EMBO Rep ; 24(7): e57064, 2023 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-37306042

RESUMEN

Eukaryotic cell adhesion and migration rely on surface adhesins connecting extracellular ligands to the intracellular actin cytoskeleton. Plasmodium sporozoites are transmitted by mosquitoes and rely on adhesion and gliding motility to colonize the salivary glands and to reach the liver after transmission. During gliding, the essential sporozoite adhesin TRAP engages actin filaments in the cytoplasm of the parasite, while binding ligands on the substrate through its inserted (I) domain. Crystal structures of TRAP from different Plasmodium species reveal the I domain in closed and open conformations. Here, we probe the importance of these two conformational states by generating parasites expressing versions of TRAP with the I domain stabilized in either the open or closed state with disulfide bonds. Strikingly, both mutations impact sporozoite gliding, mosquito salivary gland entry, and transmission. Absence of gliding in sporozoites expressing the open TRAP I domain can be partially rescued by adding a reducing agent. This suggests that dynamic conformational change is required for ligand binding, gliding motility, and organ invasion and hence sporozoite transmission from mosquito to mammal.


Asunto(s)
Culicidae , Plasmodium , Animales , Esporozoítos/metabolismo , Ligandos , Plasmodium/metabolismo , Hígado/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Mamíferos/metabolismo
2.
J Cell Sci ; 136(1)2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36511329

RESUMEN

Malaria is a devastating mosquito-borne parasitic disease that manifests when Plasmodium parasites replicate within red blood cells. During the development within the red blood cell, the parasite digests hemoglobin and crystalizes the otherwise toxic heme. The resulting hemozoin crystals limit imaging by STED nanoscopy owing to their high light-absorbing capacity, which leads to immediate cell destruction upon contact with the laser. Here, we establish CUBIC-P-based clearing of hemozoin crystals, enabling whole-cell STED nanoscopy of parasites within red blood cells. Hemozoin-cleared infected red blood cells could reliably be stained with antibodies, and hence proteins in the hemozoin-containing digestive vacuole membrane, as well as in secretory vesicles of gametocytes, could be imaged at high resolution. Thus, this process is a valuable tool to study and understand parasite biology and the potential molecular mechanisms mediating drug resistance. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Antimaláricos , Malaria , Parásitos , Plasmodium , Humanos , Animales , Microscopía , Malaria/parasitología , Plasmodium/metabolismo , Eritrocitos , Plasmodium falciparum , Antimaláricos/metabolismo , Antimaláricos/uso terapéutico
3.
Redox Biol ; 58: 102536, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36401887

RESUMEN

The redox state of the host-parasite unit has been hypothesized to play a central role for the fitness of the intraerythrocytic blood stages of the human malaria parasite Plasmodium falciparum. In particular, hemoglobinopathies have been suggested to cause a more oxidizing environment, thereby protecting from severe malaria. Here we determined the redox potential of infected wild-type (hemoglobin AA) or sickle trait (hemoglobin AS) erythrocytes using parasite-encoded variants of the redox-sensitive green-fluorescent protein 2 (roGFP2). Our non-invasive roGFP2 single-cell measurements revealed a reducing steady-state redox potential of -304 ± 11 mV for the erythrocyte cytosol during ring-stage development and a rather sudden oxidation to -278 ± 12 mV during trophozoite-stage development around 28 h post invasion. There was no significant difference between wild-type or sickle trait erythrocytes regarding the stage dependence and the detected increase of the redox potential during the intraerythrocytic life cycle. The steady-state redox potential of the parasite cytosol, between -304 and -313 mV, was highly reducing throughout the life cycle. The redox potential in the parasitophorous vacuole at the interface between the secretory pathway and the erythrocyte was -284 ± 10 mV and remained stable during trophozoite-stage development with implications for the export of disulfide-containing proteins. In summary, P. falciparum blood stage development from the late ring to the early trophozoite stage causes a physiological jump in erythrocyte redox potential irrespective of the presence or absence of hemoglobin S.


Asunto(s)
Malaria Falciparum , Rasgo Drepanocítico , Humanos , Plasmodium falciparum/metabolismo , Rasgo Drepanocítico/genética , Rasgo Drepanocítico/metabolismo , Eritrocitos/metabolismo , Malaria Falciparum/parasitología , Proteínas Fluorescentes Verdes/metabolismo , Oxidación-Reducción
4.
EMBO Rep ; 23(7): e54719, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35403820

RESUMEN

During transmission of malaria-causing parasites from mosquitoes to mammals, Plasmodium sporozoites migrate rapidly in the skin to search for a blood vessel. The high migratory speed and narrow passages taken by the parasites suggest considerable strain on the sporozoites to maintain their shape. Here, we show that the membrane-associated protein, concavin, is important for the maintenance of the Plasmodium sporozoite shape inside salivary glands of mosquitoes and during migration in the skin. Concavin-GFP localizes at the cytoplasmic periphery and concavin(-) sporozoites progressively round up upon entry of salivary glands. Rounded concavin(-) sporozoites fail to pass through the narrow salivary ducts and are rarely ejected by mosquitoes, while normally shaped concavin(-) sporozoites are transmitted. Strikingly, motile concavin(-) sporozoites disintegrate while migrating through the skin leading to parasite arrest or death and decreased transmission efficiency. Collectively, we suggest that concavin contributes to cell shape maintenance by riveting the plasma membrane to the subtending inner membrane complex. Interfering with cell shape maintenance pathways might hence provide a new strategy to prevent a malaria infection.


Asunto(s)
Anopheles , Malaria , Parásitos , Plasmodium , Animales , Anopheles/parasitología , Mamíferos , Esporozoítos/metabolismo
5.
Mol Microbiol ; 117(3): 553-568, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34587292

RESUMEN

In the last 10 years, proximity-dependent biotinylation (PDB) techniques greatly expanded the ability to study protein environments in the living cell that range from specific protein complexes to entire compartments. This is achieved by using enzymes such as BirA* and APEX that are fused to proteins of interest and biotinylate proteins in their proximity. PDB techniques are now also increasingly used in apicomplexan parasites. In this review, we first give an overview of the main PDB approaches and how they compare with other techniques that address similar questions. PDB is particularly valuable to detect weak or transient protein associations under physiological conditions and to study cellular structures that are difficult to purify or have a poorly understood protein composition. We also highlight new developments such as novel smaller or faster-acting enzyme variants and conditional PDB approaches, providing improvements in both temporal and spatial resolution which may offer broader application possibilities useful in apicomplexan research. In the second part, we review work using PDB techniques in apicomplexan parasites and how this expanded our knowledge about these medically important parasites.


Asunto(s)
Biología , Proteínas , Biotinilación , Proteínas/metabolismo
6.
Malar J ; 20(1): 430, 2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717635

RESUMEN

BACKGROUND: Plasmodium sporozoites are the highly motile forms of malaria-causing parasites that are transmitted by the mosquito to the vertebrate host. Sporozoites need to enter and cross several cellular and tissue barriers for which they employ a set of surface proteins. Three of these proteins are members of the thrombospondin related anonymous protein (TRAP) family. Here, potential additive, synergistic or antagonistic roles of these adhesion proteins were investigated. METHODS: Four transgenic Plasmodium berghei parasite lines that lacked two or all three of the TRAP family adhesins TRAP, TLP and TREP were generated using positive-negative selection. The parasite lines were investigated for their capacity to attach to and move on glass, their ability to egress from oocysts and their capacity to enter mosquito salivary glands. One strain was in addition interrogated for its capacity to infect mice. RESULTS: The major phenotype of the TRAP single gene deletion dominates additional gene deletion phenotypes. All parasite lines including the one lacking all three proteins were able to conduct some form of active, if unproductive movement. CONCLUSIONS: The individual TRAP-family adhesins appear to play functionally distinct roles during motility and infection. Other proteins must contribute to substrate adhesion and gliding motility.


Asunto(s)
Plasmodium berghei/fisiología , Proteínas Protozoarias/genética , Esporozoítos/fisiología , Microorganismos Modificados Genéticamente/genética , Microorganismos Modificados Genéticamente/fisiología , Plasmodium berghei/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/genética
7.
EMBO Mol Med ; 13(4): e13933, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33666362

RESUMEN

Transmission of malaria-causing parasites to and by the mosquito relies on active parasite migration and constitutes bottlenecks in the Plasmodium life cycle. Parasite adaption to the biochemically and physically different environments must hence be a key evolutionary driver for transmission efficiency. To probe how subtle but physiologically relevant changes in environmental elasticity impact parasite migration, we introduce 2D and 3D polyacrylamide gels to study ookinetes, the parasite forms emigrating from the mosquito blood meal and sporozoites, the forms transmitted to the vertebrate host. We show that ookinetes adapt their migratory path but not their speed to environmental elasticity and are motile for over 24 h on soft substrates. In contrast, sporozoites evolved more short-lived rapid gliding motility for rapidly crossing the skin. Strikingly, sporozoites are highly sensitive to substrate elasticity possibly to avoid adhesion to soft endothelial cells on their long way to the liver. Hence, the two migratory stages of Plasmodium evolved different strategies to overcome the physical challenges posed by the respective environments and barriers they encounter.


Asunto(s)
Malaria , Parásitos , Plasmodium , Animales , Elasticidad , Células Endoteliales , Esporozoítos
8.
Mol Biochem Parasitol ; 241: 111347, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33347893

RESUMEN

Substrate-dependent gliding motility is key to malaria transmission. It mediates host cell traversal, invasion and infection by Plasmodium and related apicomplexan parasites. The 110 amino acid-long cell surface protein LIMP is essential for P. berghei sporozoites where it is required for the invasion of the mosquito's salivary glands and the liver cells of the rodent host. Here we define an additional role for LIMP during mosquito invasion by the ookinete. limp mRNA is provided as a translationally repressed mRNP (messenger ribonucleoprotein) by the female gametocyte and the protein translated in the ookinete. Parasites depleted of limp (Δlimp) develop ookinetes with apparent normal morphology and no defect during in vitro gliding motility, and yet display a pronounced reduction in oocyst numbers; compared to wildtype 82 % more Δlimp ookinetes remain within the mosquito blood meal explaining the decrease in oocysts. As in the sporozoite, LIMP exerts a profound role on ookinete infection of the mosquito.


Asunto(s)
Culicidae/metabolismo , Culicidae/parasitología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/parasitología , Proteínas de Membrana de los Lisosomas/genética , Plasmodium berghei , Proteínas Protozoarias/genética , Animales , Expresión Génica , Genes Reporteros , Proteínas de Membrana de los Lisosomas/metabolismo , Malaria/parasitología , Malaria/transmisión , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo
9.
PLoS One ; 15(9): e0238134, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936796

RESUMEN

Malaria is a life-threatening disease, caused by Apicomplexan parasites of the Plasmodium genus. The Anopheles mosquito is necessary for the sexual replication of these parasites and for their transmission to vertebrate hosts, including humans. Imaging of the parasite within the insect vector has been attempted using multiple microscopy methods, most of which are hampered by the presence of the light scattering opaque cuticle of the mosquito. So far, most imaging of the Plasmodium mosquito stages depended on either sectioning or surgical dissection of important anatomical sites, such as the midgut and the salivary glands. Optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) enable imaging fields of view in the centimeter scale whilst providing micrometer resolution. In this paper, we compare different optical clearing protocols and present reconstructions of the whole body of Plasmodium-infected, optically cleared Anopheles stephensi mosquitoes and their midguts. The 3D-reconstructions from OPT imaging show detailed features of the mosquito anatomy and enable overall localization of parasites in midguts. Additionally, LSFM imaging of mosquito midguts shows detailed distribution of oocysts in extracted midguts. This work was submitted as a pre-print to bioRxiv, available at https://www.biorxiv.org/content/10.1101/682054v2.


Asunto(s)
Anopheles/fisiología , Imagenología Tridimensional , Insectos Vectores/fisiología , Microscopía Fluorescente , Plasmodium/fisiología , Tomografía Óptica , Animales , Anopheles/parasitología
10.
Elife ; 92020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32648541

RESUMEN

Inserted (I) domains function as ligand-binding domains in adhesins that support cell adhesion and migration in many eukaryotic phyla. These adhesins include integrin αß heterodimers in metazoans and single subunit transmembrane proteins in apicomplexans such as TRAP in Plasmodium and MIC2 in Toxoplasma. Here we show that the I domain of TRAP is essential for sporozoite gliding motility, mosquito salivary gland invasion and mouse infection. Its replacement with the I domain from Toxoplasma MIC2 fully restores tissue invasion and parasite transmission, while replacement with the aX I domain from human integrins still partially restores liver infection. Mutations around the ligand binding site allowed salivary gland invasion but led to inefficient transmission to the rodent host. These results suggest that apicomplexan parasites appropriated polyspecific I domains in part for their ability to engage with multiple ligands and to provide traction for emigration into diverse organs in distant phyla.


Asunto(s)
Anopheles/parasitología , Malaria/parasitología , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Femenino , Ligandos , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Glándulas Salivales/parasitología , Alineación de Secuencia , Esporozoítos/fisiología
11.
PLoS One ; 14(9): e0222226, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31553751

RESUMEN

Ookinetes, one of the motile and invasive forms of the malaria parasite, rely on gliding motility in order to establish an infection in the mosquito host. Here we characterize the protein PBANKA_0407300 which is conserved in the Plasmodium genus but lacks significant similarity to proteins of other eukaryotes. It is expressed in gametocytes and throughout the invasive mosquito stages of P. berghei, but is absent from asexual blood stages. Mutants lacking the protein developed morphologically normal ookinetes that were devoid of productive motility although some stretching movement could be detected. We therefore named the protein Ookinete Motility Deficient (OMD). Several key factors known to be involved in motility however were normally expressed and localized in the mutant. Importantly, the mutant failed to establish an infection in the mosquito which resulted in a total malaria transmission blockade.


Asunto(s)
Anopheles/parasitología , Malaria/transmisión , Plasmodium berghei/fisiología , Proteínas Protozoarias/fisiología , Animales , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Técnicas de Silenciamiento del Gen , Malaria/parasitología , Ratones , Microscopía Electrónica de Rastreo , Proteínas Protozoarias/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Expert Opin Ther Targets ; 23(3): 251-261, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30700216

RESUMEN

BACKGROUND: Cellular metabolism generates reactive oxygen species. The oxidation and deamination of the deoxynucleoside triphosphate (dNTP) pool results in the formation of non-canonical, toxic dNTPs that can cause mutations, genome instability, and cell death. House-cleaning or sanitation enzymes that break down and detoxify non-canonical nucleotides play major protective roles in nucleotide metabolism and constitute key drug targets for cancer and various pathogens. We hypothesized that owing to their protective roles in nucleotide metabolism, these house-cleaning enzymes are key drug targets in the malaria parasite. METHODS: Using the rodent malaria parasite Plasmodium berghei we evaluate here, by gene targeting, a group of conserved proteins with a putative function in the detoxification of non-canonical nucleotides as potential antimalarial drug targets: they are inosine triphosphate pyrophosphatase (ITPase), deoxyuridine triphosphate pyrophosphatase (dUTPase) and two NuDiX hydroxylases, the diadenosine tetraphosphate (Ap4A) hydrolase and the nucleoside triphosphate hydrolase (NDH). RESULTS: While all four proteins are expressed constitutively across the intraerythrocytic developmental cycle, neither ITPase nor NDH are required for parasite viability. dutpase and ap4ah null mutants, on the other hand, are not viable suggesting an essential function for these proteins for the malaria parasite. CONCLUSIONS: Plasmodium dUTPase and Ap4A could be drug targets in the malaria parasite.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Malaria/parasitología , Plasmodium berghei/enzimología , Pirofosfatasas/genética , Ácido Anhídrido Hidrolasas/metabolismo , Animales , Antimaláricos/farmacología , Humanos , Ratones , Ratones Endogámicos C57BL , Nucleósido-Trifosfatasa/genética , Nucleósido-Trifosfatasa/metabolismo , Plasmodium berghei/genética , Pirofosfatasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Inosina Trifosfatasa
13.
J Cell Sci ; 131(20)2018 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-30237220

RESUMEN

Gene expression of malaria parasites is mediated by the apicomplexan Apetala2 (ApiAP2) transcription factor family. Different ApiAP2s control gene expression at distinct stages in the complex life cycle of the parasite, ensuring timely expression of stage-specific genes. ApiAP2s recognize short cis-regulatory elements that are enriched in the upstream/promoter region of their target genes. This should, in principle, allow the generation of 'synthetic' promoters that drive gene expression at desired stages of the Plasmodium life cycle. Here we test this concept by combining cis-regulatory elements of two genes expressed successively within the mosquito part of the life cycle. Our tailored 'synthetic' promoters, named Spooki 1.0 and Spooki 2.0, activate gene expression in early and late mosquito stages, as shown by the expression of a fluorescent reporter. We used these promoters to address the specific functionality of two related adhesins that are exclusively expressed either during the early or late mosquito stage. By modifying the expression profile of both adhesins in absence of their counterpart we were able to test for complementary functions in gliding and invasion. We discuss the possible advantages and drawbacks of our approach.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Culicidae/metabolismo , Estadios del Ciclo de Vida/genética , Malaria/genética , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/metabolismo , Animales , Humanos , Plasmodium
14.
Sci Rep ; 8(1): 11249, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-30050042

RESUMEN

The nuclear pore complex (NPC) is a large macromolecular assembly of around 30 different proteins, so-called nucleoporins (Nups). Embedded in the nuclear envelope the NPC mediates bi-directional exchange between the cytoplasm and the nucleus and plays a role in transcriptional regulation that is poorly understood. NPCs display modular arrangements with an overall structure that is generally conserved among many eukaryotic phyla. However, Nups of yeast or human origin show little primary sequence conservation with those from early-branching protozoans leaving those of the malaria parasite unrecognized. Here we have combined bioinformatic and genetic methods to identify and spatially characterize Nup components in the rodent infecting parasite Plasmodium berghei and identified orthologs from the human malaria parasite P. falciparum, as well as the related apicomplexan parasite Toxoplasma gondii. For the first time we show the localization of selected Nups throughout the P. berghei life cycle. Largely restricted to apicomplexans we identify an extended C-terminal poly-proline extension in SEC13 that is essential for parasite survival and provide high-resolution images of Plasmodium NPCs obtained by cryo electron tomography. Our data provide the basis for full characterization of NPCs in malaria parasites, early branching unicellular eukaryotes with significant impact on human health.


Asunto(s)
Proteínas de Complejo Poro Nuclear/análisis , Proteínas de Complejo Poro Nuclear/genética , Plasmodium berghei/enzimología , Biología Computacional , Genes Esenciales , Biología Molecular , Plasmodium berghei/genética , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Toxoplasma/enzimología , Toxoplasma/genética
15.
Cell Microbiol ; 20(8): e12843, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29579782

RESUMEN

Compared with other eukaryotic cell types, malaria parasites appear to possess a more rudimentary Golgi apparatus being composed of dispersed, unstacked cis and trans-cisternae. Despite playing a central role in the secretory pathway of the parasite, few Plasmodium Golgi resident proteins have been characterised. We had previously identified a new Golgi resident protein of unknown function, which we had named Golgi Protein 1, and now show that it forms a complex with a previously uncharacterised transmembrane protein (Golgi Protein 2, GP2). The Golgi Protein complex localises to the cis-Golgi throughout the erythrocytic cycle and potentially also during the mosquito stages. Analysis of parasite strains where GP1 expression is conditionally repressed and/or the GP2 gene is inactivated reveals that though the Golgi protein complex is not essential at any stage of the parasite life cycle, it is important for optimal asexual development in the blood stages.


Asunto(s)
Eritrocitos/parasitología , Aparato de Golgi/metabolismo , Complejos Multiproteicos/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/metabolismo , Humanos
16.
PLoS Pathog ; 13(5): e1006412, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28552953

RESUMEN

Profilin is an actin monomer binding protein that provides ATP-actin for incorporation into actin filaments. In contrast to higher eukaryotic cells with their large filamentous actin structures, apicomplexan parasites typically contain only short and highly dynamic microfilaments. In apicomplexans, profilin appears to be the main monomer-sequestering protein. Compared to classical profilins, apicomplexan profilins contain an additional arm-like ß-hairpin motif, which we show here to be critically involved in actin binding. Through comparative analysis using two profilin mutants, we reveal this motif to be implicated in gliding motility of Plasmodium berghei sporozoites, the rapidly migrating forms of a rodent malaria parasite transmitted by mosquitoes. Force measurements on migrating sporozoites and molecular dynamics simulations indicate that the interaction between actin and profilin fine-tunes gliding motility. Our data suggest that evolutionary pressure to achieve efficient high-speed gliding has resulted in a unique profilin-actin interface in these parasites.


Asunto(s)
Actinas/metabolismo , Malaria/parasitología , Plasmodium berghei/citología , Plasmodium berghei/metabolismo , Profilinas/metabolismo , Proteínas Protozoarias/metabolismo , Actinas/genética , Animales , Movimiento Celular , Femenino , Humanos , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Profilinas/genética , Unión Proteica , Proteínas Protozoarias/genética , Esporozoítos/citología , Esporozoítos/crecimiento & desarrollo , Esporozoítos/metabolismo
17.
Elife ; 62017 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-28525314

RESUMEN

Gliding motility allows malaria parasites to migrate and invade tissues and cells in different hosts. It requires parasite surface proteins to provide attachment to host cells and extracellular matrices. Here, we identify the Plasmodium protein LIMP (the name refers to a gliding phenotype in the sporozoite arising from epitope tagging of the endogenous protein) as a key regulator for adhesion during gliding motility in the rodent malaria model P. berghei. Transcribed in gametocytes, LIMP is translated in the ookinete from maternal mRNA, and later in the sporozoite. The absence of LIMP reduces initial mosquito infection by 50%, impedes salivary gland invasion 10-fold, and causes a complete absence of liver invasion as mutants fail to attach to host cells. GFP tagging of LIMP caused a limping defect during movement with reduced speed and transient curvature changes of the parasite. LIMP is an essential motility and invasion factor necessary for malaria transmission.


Asunto(s)
Culicidae/parasitología , Locomoción , Proteínas de Membrana de los Lisosomas/metabolismo , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Esporozoítos/fisiología , Factores de Virulencia/metabolismo , Animales , Modelos Animales de Enfermedad , Hígado/parasitología , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Ratones
18.
PLoS Pathog ; 12(7): e1005710, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27409081

RESUMEN

Parasites causing malaria need to migrate in order to penetrate tissue barriers and enter host cells. Here we show that the actin filament-binding protein coronin regulates gliding motility in Plasmodium berghei sporozoites, the highly motile forms of a rodent malaria-causing parasite transmitted by mosquitoes. Parasites lacking coronin show motility defects that impair colonization of the mosquito salivary glands but not migration in the skin, yet result in decreased transmission efficiency. In non-motile sporozoites low calcium concentrations mediate actin-independent coronin localization to the periphery. Engagement of extracellular ligands triggers an intracellular calcium release followed by the actin-dependent relocalization of coronin to the rear and initiation of motility. Mutational analysis and imaging suggest that coronin organizes actin filaments for productive motility. Using coronin-mCherry as a marker for the presence of actin filaments we found that protein kinase A contributes to actin filament disassembly. We finally speculate that calcium and cAMP-mediated signaling regulate a switch from rapid parasite motility to host cell invasion by differentially influencing actin dynamics.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Malaria/parasitología , Proteínas de Microfilamentos/metabolismo , Plasmodium berghei/metabolismo , Esporozoítos/metabolismo , Animales , Western Blotting , Culicidae/microbiología , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Células Hep G2 , Humanos , Insectos Vectores/microbiología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/metabolismo , Transfección
19.
PLoS Pathog ; 12(7): e1005734, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27427910

RESUMEN

Regulated protein secretion is required for malaria parasite life cycle progression and transmission between the mammalian host and mosquito vector. During transmission from the host to the vector, exocytosis of highly specialised secretory vesicles, such as osmiophilic bodies, is key to the dissolution of the red blood cell and parasitophorous vacuole membranes enabling gamete egress. The positioning of adhesins from the TRAP family, from micronemes to the sporozoite surface, is essential for gliding motility of the parasite and transmission from mosquito to mammalian host. Here we identify a conserved role for the putative pantothenate transporter PAT in Plasmodium berghei in vesicle fusion of two distinct classes of vesicles in gametocytes and sporozoites. PAT is a membrane component of osmiophilic bodies in gametocytes and micronemes in sporozoites. Despite normal formation and trafficking of osmiophilic bodies to the cell surface upon activation, PAT-deficient gametes fail to discharge their contents, remain intraerythrocytic and unavailable for fertilisation and further development in the mosquito. Sporozoites lacking PAT fail to secrete TRAP, are immotile and thus unable to infect the subsequent rodent host. Thus, P. berghei PAT appears to regulate exocytosis in two distinct populations of vesicles in two different life cycle forms rather than acting as pantothenic transporter during parasite transmission.


Asunto(s)
Anopheles/parasitología , Malaria/transmisión , Perilipinas/metabolismo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Modelos Animales de Enfermedad , Exocitosis/fisiología , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Insectos Vectores/parasitología , Ratones , Microscopía Electrónica , Vesículas Secretoras/metabolismo , Esporozoítos/metabolismo , Transfección
20.
Mol Cell Proteomics ; 15(9): 2852-62, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27371728

RESUMEN

Malaria transmission from an infected host to the mosquito vector requires the uptake of intraerythrocytic sexual precursor cells into the mosquito midgut. For the release of mature extracellular gametes two membrane barriers-the parasite parasitophorous vacuole membrane and the host red blood cell membrane-need to be dissolved. Membrane lysis occurs after the release of proteins from specialized secretory vesicles including osmiophilic bodies. In this study we conducted proteomic analyses of the P. berghei gametocyte egressome and developed a vesicular bioID approach to identify hitherto unknown proteins with a potential function in gametocyte egress. This first Plasmodium gametocyte egressome includes the proteins released by the parasite during the lysis of the parasitophorous vacuole membrane and red blood cell membrane. BioID of the osmiophilic body protein MDV1/PEG3 revealed a vesicular proteome of these gametocyte-specific secretory vesicles. Fluorescent protein tagging and gene deletion approaches were employed to validate and identify a set of novel factors essential for this lysis and egress process. Our study provides the first in vivo bioID for a rodent malaria parasite and together with the first Plasmodium gametocyte egressome identifies MTRAP as a novel factor essential for mosquito transmission. Our data provide an important resource for proteins potentially involved in a key step of gametogenesis.


Asunto(s)
Malaria/transmisión , Plasmodium berghei/fisiología , Proteómica/métodos , Proteínas Protozoarias/metabolismo , Animales , Membrana Eritrocítica/parasitología , Estadios del Ciclo de Vida , Malaria/veterinaria , Espectrometría de Masas , Ratones , Plasmodium berghei/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA