Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-38405843

RESUMEN

Plasmodium parasites, which are the causative agents of malaria, undergo closed mitosis without breakdown of the nuclear envelope. Unlike the closed mitosis in yeast, P. berghei parasites undergo multiple rounds of asynchronous nuclear divisions in a shared cytoplasm result in a multinucleated (8-24) organism prior to formation of daughter cells within an infected red blood cell. During this replication process, intact nuclear pore complexes (NPCs) and their component nucleoporins are likely to play critical roles in parasite growth, facilitating selective bi-directional nucleocytoplasmic transport and genome organization. Here we utilize ultrastructure expansion microscopy (U-ExM) to investigate P. berghei Nup138, Nup221, and Nup313 at the single nucleus level throughout the 24 hour blood-stage replication cycle. Our findings reveal that these Nups are evenly distributed around the nuclei and organized in a rosette structure previously undescribed around the centriolar plaque, which is responsible for intranuclear microtubule nucleation during mitosis. We also detect an increased number of NPCs compared with previously reported, highlighting the power of U-ExM. By adapting the recombination-induced tag exchange (RITE) system to P. berghei, we provide evidence of NPC maintenance, demonstrating Nup221 turnover during parasite asexual replication. Our data shed light on the distribution of NPCs and their homeostasis during the blood-stage replication of P. berghei parasites. Further studies into the nuclear surface of these parasites will allow for a better understanding of parasites nuclear mechanics and organization.

2.
mBio ; 13(6): e0309622, 2022 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-36445080

RESUMEN

During vertebrate infection, obligate intracellular malaria parasites develop within a parasitophorous vacuole, which constitutes the interface between the parasite and its hepatocyte or erythrocyte host cells. To traverse this barrier, Plasmodium spp. utilize a dual-function pore formed by EXP2 for nutrient transport and, in the context of the PTEX translocon, effector protein export across the vacuole membrane. While critical to blood-stage survival, less is known about EXP2/PTEX function in the liver stage, although major differences in the export mechanism are suggested by absence of the PTEX unfoldase HSP101 in the intrahepatic vacuole. Here, we employed the glucosamine-activated glmS ribozyme to study the role of EXP2 during Plasmodium berghei liver-stage development in hepatoma cells. Insertion of the glmS sequence into the exp2 3' untranslated region (UTR) enabled glucosamine-dependent depletion of EXP2 after hepatocyte invasion, allowing separation of EXP2 function during intrahepatic development from a recently reported role in hepatocyte invasion. Postinvasion EXP2 knockdown reduced parasite size and largely abolished expression of the mid- to late-liver-stage marker LISP2. As an orthogonal approach to monitor development, EXP2-glmS parasites and controls were engineered to express nanoluciferase. Activation of glmS after invasion substantially decreased luminescence in hepatoma monolayers and in culture supernatants at later time points corresponding to merosome detachment, which marks the culmination of liver-stage development. Collectively, our findings extend the utility of the glmS ribozyme to study protein function in the liver stage and reveal that EXP2 is important for intrahepatic parasite development, indicating that PTEX components also function at the hepatocyte-parasite interface. IMPORTANCE After the mosquito bite that initiates a Plasmodium infection, parasites first travel to the liver and develop in hepatocytes. This liver stage is asymptomatic but necessary for the parasite to transition to the merozoite form, which infects red blood cells and causes malaria. To take over their host cells, avoid immune defenses, and fuel their growth, these obligately intracellular parasites must import nutrients and export effector proteins across a vacuole membrane in which they reside. In the blood stage, these processes depend on a translocon called PTEX, but it is unclear if PTEX also functions during the liver stage. Here, we adapted the glmS ribozyme to control expression of EXP2, the membrane pore component of PTEX, during the liver stage of the rodent malaria parasite Plasmodium berghei. Our results show that EXP2 is important for intracellular development in the hepatocyte, revealing that PTEX components are also functionally important during liver-stage infection.


Asunto(s)
Eritrocitos , Hepatocitos , Malaria , Plasmodium berghei , Proteínas Protozoarias , Carcinoma Hepatocelular , Eritrocitos/metabolismo , Eritrocitos/parasitología , Neoplasias Hepáticas , Malaria/genética , Malaria/metabolismo , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/genética , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN Catalítico/metabolismo , Animales , Ratones , Hepatocitos/metabolismo , Hepatocitos/parasitología
3.
mBio ; 13(5): e0181522, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36040030

RESUMEN

Twenty years since the publication of the Plasmodium falciparum and P. berghei genomes one-third of their protein-coding genes still lack functional annotation. In the absence of sequence and structural homology, protein-protein interactions can facilitate functional prediction of such orphan genes by mapping protein complexes in their natural cellular environment. The Plasmodium nuclear pore complex (NPC) is a case in point: it remains poorly defined; its constituents lack conservation with the 30+ proteins described in the NPC of many opisthokonts, a clade of eukaryotes that includes fungi and animals, but not Plasmodium. Here, we developed a labeling methodology based on TurboID fusion proteins, which allows visualization of the P. berghei NPC and facilitates the identification of its components. Following affinity purification and mass spectrometry, we identified 4 known nucleoporins (Nups) (138, 205, 221, and the bait 313), and verify interaction with the putative phenylalanine-glycine (FG) Nup637; we assigned 5 proteins lacking annotation (and therefore meaningful homology with proteins outside the genus) to the NPC, which is confirmed by green fluorescent protein (GFP) tagging. Based on gene deletion attempts, all new Nups - Nup176, 269, 335, 390, and 434 - are essential to parasite survival. They lack primary sequence homology with proteins outside the Plasmodium genus; albeit 2 incorporate short domains with structural homology to human Nup155 and yeast Nup157, and the condensin SMC (Structural Maintenance Of Chromosomes 4). The protocols developed here showcase the power of proximity labeling for elucidating protein complex composition and annotation of taxonomically restricted genes in Plasmodium. It opens the door to exploring the function of the Plasmodium NPC and understanding its evolutionary position. IMPORTANCE The nuclear pore complex (NPC) is a platform for constant evolution and has been used to study the evolutionary patterns of early-branching eukaryotes. The Plasmodium NPC is poorly defined due to its evolutionary divergent nature making it impossible to characterize it via homology searches. Although 2 decades have passed since the publication of the Plasmodium genome, 30% of the genes still lack functional annotation. Our study demonstrates the ability of proximity labeling using TurboID to assign function to orphan proteins in the malaria parasite. We have identified a total of 10 Nups that will allow further study of NPC dynamics, structural elements, involvement in nucleocytoplasmic transport, and unique non-transport functions of nucleoporins that provide adaptability to this malaria parasite.


Asunto(s)
Malaria , Poro Nuclear , Humanos , Transporte Activo de Núcleo Celular/genética , Glicina/metabolismo , Proteínas Fluorescentes Verdes/análisis , Malaria/metabolismo , Poro Nuclear/química , Poro Nuclear/genética , Poro Nuclear/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Fenilalanina/química , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Saccharomyces cerevisiae/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(34): e2111932119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969762

RESUMEN

Glutamate-gated chloride channels (GluCls) are unique to invertebrates and are targeted by macrocyclic lactones. In this study, we cloned an AVR-14B GluCl subunit from adult Brugia malayi, a causative agent of lymphatic filariasis in humans. To elucidate this channel's pharmacological properties, we used Xenopus laevis oocytes for expression and performed two-electrode voltage-clamp electrophysiology. The receptor was gated by the natural ligand L-glutamate (effective concentration, 50% [EC50] = 0.4 mM) and ivermectin (IVM; EC50 = 1.8 nM). We also characterized the effects of nodulisporic acid (NA) on Bma-AVR-14B and NA-produced dual effects on the receptor as an agonist and a type II positive allosteric modulator. Here we report characterization of the complex activity of NA on a nematode GluCl. Bma-AVR-14B demonstrated some unique pharmacological characteristics. IVM did not produce potentiation of L-glutamate-mediated responses but instead, reduced the channel's sensitivity for the ligand. Further electrophysiological exploration showed that IVM (at a moderate concentration of 0.1 nM) functioned as an inhibitor of both agonist and positive allosteric modulatory effects of NA. This suggests that IVM and NA share a complex interaction. The pharmacological properties of Bma-AVR-14B indicate that the channel is an important target of IVM and NA. In addition, the unique electrophysiological characteristics of Bma-AVR-14B could explain the observed variation in drug sensitivities of various nematode parasites. We have also shown the inhibitory effects of IVM and NA on adult worm motility using Worminator. RNA interference (RNAi) knockdown suggests that AVR-14 plays a role in influencing locomotion in B. malayi.


Asunto(s)
Brugia Malayi , Canales de Cloruro , Indoles , Animales , Brugia Malayi/efectos de los fármacos , Brugia Malayi/genética , Brugia Malayi/metabolismo , Canales de Cloruro/efectos de los fármacos , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Ácido Glutámico/metabolismo , Indoles/farmacología , Ivermectina/farmacología , Ligandos
5.
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
6.
Invert Neurosci ; 20(4): 17, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32978688

RESUMEN

Cestodes are common gastrointestinal parasites of humans and livestock. They attach to the host gut and, without a mouth or intestinal system, absorb nutrients through their epidermis. Here we show that despite this simplified anatomy and sessile lifestyle, they maintain a complex neuromuscular system. We used fluorescently labelled phalloidin as a specific probe for filamentous actin to define the overall organisation of several distinct muscle systems in the cyclophyllidean Moniezia expansa. Like all flatworms, the body wall musculature below the neodermis of this intestinal parasite of sheep is characterised by outer circular and inner longitudinal muscle fibres. Diagonal fibres, typically found in free-living and trematode platyhelminths, on the other hand, are notably absent. Prominent longitudinal sheaths dominate the parenchyma and provide retractor muscles to the four acetabula in the scolex; they attach at the bottom of each cup-shaped holdfast. Within sexually mature proglottids, circular fibres dominate the duct walls of the male and female reproductive systems. Nerve cells and fibres that express serotonin or neuropeptide F supply well-developed innervation to several of the described muscle systems: emanating from the central nervous system, fibres in the periphery develop pervasive nerve nets that anastomose within body wall musculature as well as the parenchymal longitudinal and oblique muscle fibres, and innervate the sexual organs and gonopore in mature proglottids. Using homology searches, we provide evidence for 20 neuropeptide precursors together with four prepropeptide processing enzymes as well as several 5-HT signalling components to be represented in the Moniezia transcriptome.


Asunto(s)
Cestodos/fisiología , Músculos/fisiología , Sistema Nervioso , Actinas , Animales , Neuropéptidos , Faloidina , Ovinos
7.
PLoS Pathog ; 16(4): e1008396, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32243475

RESUMEN

Nematode parasites infect approximately 1.5 billion people globally and are a significant public health concern. There is an accepted need for new, more effective anthelmintic drugs. Nicotinic acetylcholine receptors on parasite nerve and somatic muscle are targets of the cholinomimetic anthelmintics, while glutamate-gated chloride channels in the pharynx of the nematode are affected by the avermectins. Here we describe a novel nicotinic acetylcholine receptor on the nematode pharynx that is a potential new drug target. This homomeric receptor is comprised of five non-α EAT-2 subunits and is not sensitive to existing cholinomimetic anthelmintics. We found that EAT-18, a novel auxiliary subunit protein, is essential for functional expression of the receptor. EAT-18 directly interacts with the mature receptor, and different homologs alter the pharmacological properties. Thus we have described not only a novel potential drug target but also a new type of obligate auxiliary protein for nAChRs.


Asunto(s)
Antinematodos/farmacología , Ascaris suum/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Helminto/metabolismo , Faringe/metabolismo , Receptores Nicotínicos/metabolismo , Acetilcolina/farmacología , Animales , Ascaris suum/efectos de los fármacos , Ascaris suum/genética , Caenorhabditis elegans/efectos de los fármacos , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas del Helminto/genética , Faringe/efectos de los fármacos , Receptores Nicotínicos/genética
8.
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
9.
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
10.
Cell Microbiol ; 21(5): e12999, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30597708

RESUMEN

Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.


Asunto(s)
Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Células Germinativas/metabolismo , Malaria/transmisión , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/metabolismo , Animales , Culicidae/parasitología , Detergentes/farmacología , Membrana Eritrocítica/efectos de los fármacos , Membrana Eritrocítica/genética , Membrana Eritrocítica/parasitología , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Exocitosis/genética , Femenino , Células Germinativas/citología , Células Germinativas/crecimiento & desarrollo , Células Germinativas/ultraestructura , Interacciones Huésped-Patógeno , Estadios del Ciclo de Vida/genética , Malaria/genética , Malaria/metabolismo , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Mosquitos Vectores/genética , Mosquitos Vectores/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Dominios Proteicos/genética , Proteínas Protozoarias/genética
11.
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
12.
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
13.
Nat Microbiol ; 3(1): 17-25, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29109477

RESUMEN

The causative agent of malaria, Plasmodium, replicates inside a membrane-bound parasitophorous vacuole (PV), which shields this intracellular parasite from the cytosol of the host cell 1 . One common threat for intracellular pathogens is the homeostatic process of autophagy, through which cells capture unwanted intracellular material for lysosomal degradation 2 . During the liver stage of a malaria infection, Plasmodium parasites are targeted by the autophagy machinery of the host cell, and the PV membrane (PVM) becomes decorated with several autophagy markers, including LC3 (microtubule-associated protein 1 light chain 3) 3,4 . Here we show that Plasmodium berghei parasites infecting hepatic cells rely on the PVM transmembrane protein UIS3 to avoid elimination by host-cell-mediated autophagy. We found that UIS3 binds host LC3 through a non-canonical interaction with a specialized surface on LC3 where host proteins with essential functions during autophagy also bind. UIS3 acts as a bona fide autophagy inhibitor by competing with host LC3-interacting proteins for LC3 binding. Our work identifies UIS3, one of the most promising candidates for a genetically attenuated vaccine against malaria 5 , as a unique and potent mediator of autophagy evasion in Plasmodium. We propose that the protein-protein interaction between UIS3 and host LC3 represents a target for antimalarial drug development.


Asunto(s)
Autofagia/fisiología , Hepatocitos/patología , Malaria/patología , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Plasmodium berghei/genética , Animales , Autofagosomas/metabolismo , Línea Celular , Células HEK293 , Células Hep G2 , Hepatocitos/parasitología , Hepatocitos/ultraestructura , Interacciones Huésped-Patógeno , Humanos , Malaria/fisiopatología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Plasmodium berghei/metabolismo , Plasmodium berghei/patogenicidad , Unión Proteica , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Vacuolas/metabolismo
14.
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
15.
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
16.
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
17.
Parasitol Int ; 65(5 Pt A): 463-71, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27312996

RESUMEN

The technical challenges of working with the sexual stages of the malaria parasite Plasmodium have hindered the characterization of sexual stage antigens in the quest for a successful malaria transmission-blocking vaccine. One such predicted and largely uncharacterized group of sexual stage candidate antigens is the CPW-WPC family of proteins. CPW-WPC proteins are named for a characteristic domain that contains two conserved motifs, CPxxW and WPC. Conserved across Apicomplexa, this family is also present earlier in the Alveolata in the free-living, non-parasitophorous, photosynthetic chromerids, Chromera and Vitrella. In Plasmodium falciparum and Plasmodium berghei blood stage parasites, the transcripts of all nine cpw-wpc genes have been detected in gametocytes. RNA immunoprecipitation followed by reverse transcriptase-PCR reveals all P. berghei cpw-wpc transcripts to be bound by the translational repressors DOZI and CITH, and thus are likely under translational control prior to transmission from the rodent host to the mosquito vector in P. berghei. The GFP tagging of two endogenous P. berghei genes confirmed translational silencing in the gametocyte and translation in ookinetes. By establishing a luciferase transgene assay, we show that the 3' untranslated region of PF3D7_1331400 controls protein expression of this reporter in P. falciparum gametocytes. Our analyses suggest that cpw-wpc genes are translationally silenced in gametocytes across Plasmodium spp. and activated during ookinete formation and thus may have a role in transmission to the mosquito.


Asunto(s)
Anopheles/parasitología , Genes Protozoarios/genética , Malaria Falciparum/transmisión , Plasmodium berghei/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Regiones no Traducidas 3'/genética , Secuencia de Aminoácidos , Animales , Evolución Biológica , Femenino , Humanos , Masculino , Ratones , Familia de Multigenes/genética , Biosíntesis de Proteínas/genética
18.
Proc Natl Acad Sci U S A ; 113(26): 7183-8, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27303037

RESUMEN

Transmission of the malaria parasite from the mammalian host to the mosquito vector requires the formation of adequately adapted parasite forms and stage-specific organelles. Here we show that formation of the crystalloid-a unique and short-lived organelle of the Plasmodium ookinete and oocyst stage required for sporogony-is dependent on the precisely timed expression of the S-acyl-transferase DHHC10. DHHC10, translationally repressed in female Plasmodium berghei gametocytes, is activated translationally during ookinete formation, where the protein is essential for the formation of the crystalloid, the correct targeting of crystalloid-resident protein LAP2, and malaria parasite transmission.


Asunto(s)
Aciltransferasas/fisiología , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/fisiología , Animales , Femenino , Malaria/transmisión , Ratones Endogámicos BALB C , Oocistos/fisiología , Orgánulos/fisiología , Plasmodium berghei/enzimología , Plasmodium berghei/fisiología
19.
Nucleic Acids Res ; 44(13): 6087-101, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27298255

RESUMEN

Sexual differentiation of malaria parasites into gametocytes in the vertebrate host and subsequent gamete fertilization in mosquitoes is essential for the spreading of the disease. The molecular processes orchestrating these transitions are far from fully understood. Here, we report the first transcriptome analysis of male and female Plasmodium falciparum gametocytes coupled with a comprehensive proteome analysis. In male gametocytes there is an enrichment of proteins involved in the formation of flagellated gametes; proteins involved in DNA replication, chromatin organization and axoneme formation. On the other hand, female gametocytes are enriched in proteins required for zygote formation and functions after fertilization; protein-, lipid- and energy-metabolism. Integration of transcriptome and proteome data revealed 512 highly expressed maternal transcripts without corresponding protein expression indicating large scale translational repression in P. falciparum female gametocytes for the first time. Despite a high degree of conservation between Plasmodium species, 260 of these 'repressed transcripts' have not been previously described. Moreover, for some of these genes, protein expression is only reported in oocysts and sporozoites indicating that repressed transcripts can be partitioned into short- and long-term storage. Finally, these data sets provide an essential resource for identification of vaccine/drug targets and for further mechanistic studies.


Asunto(s)
Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteoma/genética , Transcriptoma/genética , Cromatina/genética , Replicación del ADN/genética , Femenino , Gametogénesis/genética , Regulación de la Expresión Génica/genética , Humanos , Malaria Falciparum/parasitología , Masculino , Redes y Vías Metabólicas/genética , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Biosíntesis de Proteínas , Caracteres Sexuales
20.
Open Biol ; 6(4): 160034, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27053680

RESUMEN

Myzozoans (which include dinoflagellates, chromerids and apicomplexans) display notable divergence from their ciliate sister group, including a reduced mitochondrial genome and divergent metabolic processes. The factors contributing to these divergent processes are still poorly understood and could serve as potential drug targets in disease-causing protists. Here, we report the identification and characterization of a small mitochondrial protein from the rodent-infecting apicomplexan parasite Plasmodium berghei that is essential for development in its mosquito host. Parasites lacking the gene mitochondrial protein ookinete developmental defect (mpodd) showed malformed parasites that were unable to transmit to mosquitoes. Knockout parasites displayed reduced mitochondrial mass without affecting organelle integrity, indicating no role of the protein in mitochondrial biogenesis or morphology maintenance but a likely role in mitochondrial import or metabolism. Using genetic complementation experiments, we identified a previously unrecognized Plasmodium falciparum homologue that can rescue the mpodd(-) phenotype, thereby showing that the gene is functionally conserved. As far as can be detected, mpodd is found in myzozoans, has homologues in the phylum Apicomplexa and appears to have arisen in free-living dinoflagellates. This suggests that the MPODD protein has a conserved mitochondrial role that is important for myzozoans. While previous studies identified a number of essential proteins which are generally highly conserved evolutionarily, our study identifies, for the first time, a non-canonical protein fulfilling a crucial function in the mitochondrion during parasite transmission.


Asunto(s)
Malaria/transmisión , Proteínas Mitocondriales/metabolismo , Plasmodium berghei/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Culicidae/parasitología , Evolución Molecular , Humanos , Estadios del Ciclo de Vida , Malaria/parasitología , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/química , Parásitos/crecimiento & desarrollo , Parásitos/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...