Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 19(8): e1011566, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37651449

RESUMEN

As an obligate intracellular parasite, Toxoplasma gondii must import essential nutrients from the host cell into the parasitophorous vacuole. We previously reported that the parasite scavenges cholesterol from host endocytic organelles for incorporation into membranes and storage as cholesteryl esters in lipid droplets. In this study, we have investigated whether Toxoplasma utilizes cholesterol as a precursor for the synthesis of metabolites, such as steroids. In mammalian cells, steroidogenesis occurs in mitochondria and involves membrane-bound type I cytochrome P450 oxidases that are activated through interaction with heme-binding proteins containing a cytochrome b5 domain, such as members of the membrane-associated progesterone receptor (MAPR) family. Our LC-MS targeted lipidomics detect selective classes of hormone steroids in Toxoplasma, with a predominance for anti-inflammatory hydroxypregnenolone species, deoxycorticosterone and dehydroepiandrosterone. The genome of Toxoplasma contains homologs encoding a single type I CYP450 enzyme (we named TgCYP450mt) and a single MAPR (we named TgMAPR). We showed that TgMAPR is a hemoprotein with conserved residues in a heme-binding cytochrome b5 domain. Both TgCYP450 and TgMAPR localize to the mitochondrion and show interactions in in situ proximity ligation assays. Genetic ablation of cyp450mt is not tolerated by Toxoplasma; we therefore engineered a conditional knockout strain and showed that iΔTgCYP450mt parasites exhibit growth impairment in cultured cells. Parasite strains deficient for mapr could be generated; however, ΔTgMAPR parasites suffer from poor global fitness, loss of plasma membrane integrity, aberrant mitochondrial cristae, and an abnormally long S-phase in their cell cycle. Compared to wild-type parasites, iΔTgCYP450mt and ΔTgMAPR lost virulence in mice and metabolomics studies reveal that both mutants have reduced levels of steroids. These observations point to a steroidogenic pathway operational in the mitochondrion of a protozoan that involves an evolutionary conserved TgCYP450mt enzyme and its binding partner TgMAPR.


Asunto(s)
Toxoplasma , Animales , Ratones , Toxoplasma/genética , Citocromos b5/genética , Mitocondrias , Sistema Enzimático del Citocromo P-450 , Membranas Mitocondriales , Progesterona , Mamíferos
2.
J Cell Sci ; 136(4)2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36718630

RESUMEN

Intracellular pathogens exploit cellular resources through host cell manipulation. Within its nonfusogenic parasitophorous vacuole (PV), Toxoplasma gondii targets host nutrient-filled organelles and sequesters them into the PV through deep invaginations of the PV membrane (PVM) that ultimately detach from this membrane. Some of these invaginations are generated by an intravacuolar network (IVN) of parasite-derived tubules attached to the PVM. Here, we examined the usurpation of host ESCRT-III and Vps4A by the parasite to create PVM buds and vesicles. CHMP4B associated with the PVM/IVN, and dominant-negative (DN) CHMP4B formed many long PVM invaginations containing CHMP4B filaments. These invaginations were shorter in IVN-deficient parasites, suggesting cooperation between the IVN and ESCRT. In infected cells expressing Vps4A-DN, enlarged intra-PV structures containing host endolysosomes accumulated, reflecting defects in PVM scission. Parasite mutants lacking T. gondii (Tg)GRA14 or TgGRA64, which interact with ESCRT, reduced CHMP4B-DN-induced PVM invaginations and intra-PV host organelles, with greater defects in a double knockout, revealing the exploitation of ESCRT to scavenge host organelles by Toxoplasma.


Asunto(s)
Toxoplasma , Animales , Toxoplasma/metabolismo , Vacuolas/metabolismo , Interacciones Huésped-Parásitos , Lisosomas/metabolismo , Proteínas Protozoarias/metabolismo , Mamíferos/metabolismo
3.
mBio ; 13(4): e0144222, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35730903

RESUMEN

The intracellular parasite Toxoplasma gondii adapts to diverse host cell environments within a replicative compartment that is heavily decorated by secreted proteins. In an attempt to identify novel parasite secreted proteins that influence host cell activity, we identified and characterized a transmembrane dense granule protein dubbed GRA64 (TGME49_202620). We found that GRA64 is on the parasitophorous vacuolar membrane (PVM) and is partially exposed to the host cell cytoplasm in both tachyzoite and bradyzoite parasitophorous vacuoles. Using co-immunoprecipitation and proximity-based biotinylation approaches, we demonstrated that GRA64 appears to interact with components of the host endosomal sorting complexes required for transport (ESCRT). Genetic disruption of GRA64 does not affect acute Toxoplasma virulence or encystation in mice, as observed via tissue cyst burdens in mice during chronic infection. However, ultrastructural analysis of Δgra64 tissue cysts using electron tomography revealed enlarged vesicular structures underneath the cyst membrane, suggesting a role for GRA64 in organizing the recruitment of ESCRT proteins and subsequent intracystic vesicle formation. This study uncovers a novel host-parasite interaction that contributes to an emerging paradigm in which specific host ESCRT proteins are recruited to the limiting membranes (PVMs) of tachyzoite and bradyzoite vacuoles formed during acute and chronic Toxoplasma infection. IMPORTANCE Toxoplasma gondii is a widespread foodborne parasite that causes congenital disease and life-threatening complications in immunocompromised individuals. Part of this parasite's success lies in its ability to infect diverse organisms and host cells and to persist as a latent infection within parasite-constructed structures called tissue cysts. In this study, we characterized a protein that is secreted by T. gondii into its parasitophorous vacuole during intracellular infection, which we dub GRA64. On the vacuolar membrane, this protein is exposed to the host cell cytosol and interacts with specific host ESCRT proteins. Parasites lacking the GRA64 protein exhibit ultrastructural changes in tissue cysts during chronic infection. This study lays the foundation for future studies on the mechanics and consequences of host ESCRT-parasite protein interactions.


Asunto(s)
Toxoplasma , Toxoplasmosis , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Ratones , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Toxoplasmosis/parasitología , Vacuolas/metabolismo
4.
Front Cell Infect Microbiol ; 12: 882166, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35573773

RESUMEN

The Apicomplexa are famously named for their apical complex, a constellation of organelles at their apical end dedicated to invasion of their host cells. In contrast, at the other end of the cell, the basal complex (BC) has been overshadowed since it is much less prominent and specific functions were not immediately obvious. However, in the past decade a staggering array of functions have been associated with the BC and strides have been made in understanding its structure. Here, these collective insights are supplemented with new data to provide an overview of the understanding of the BC in Toxoplasma gondii. The emerging picture is that the BC is a dynamic and multifunctional complex, with a series of (putative) functions. The BC has multiple roles in cell division: it is the site where building blocks are added to the cytoskeleton scaffold; it exerts a two-step stretch and constriction mechanism as contractile ring; and it is key in organelle division. Furthermore, the BC has numerous putative roles in 'import', such as the recycling of mother cell remnants, the acquisition of host-derived vesicles, possibly the uptake of lipids derived from the extracellular medium, and the endocytosis of micronemal proteins. The latter process ties the BC to motility, whereas an additional role in motility is conferred by Myosin C. Furthermore, the BC acts on the assembly and/or function of the intravacuolar network, which may directly or indirectly contribute to the establishment of chronic tissue cysts. Here we provide experimental support for molecules acting in several of these processes and identify several new BC proteins critical to maintaining the cytoplasmic bridge between divided parasites. However, the dispensable nature of many BC components leaves many questions unanswered regarding its function. In conclusion, the BC in T. gondii is a dynamic and multifunctional structure at the posterior end of the parasite.


Asunto(s)
Toxoplasma , División Celular , Citoesqueleto/metabolismo , Orgánulos/metabolismo , Proteínas Protozoarias/genética , Toxoplasma/metabolismo
5.
Mol Biol Cell ; 33(5): ar34, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35274991

RESUMEN

After mammalian cell invasion, the parasite Toxoplasma multiplies in a self-made membrane-bound compartment, the parasitophorous vacuole (PV). We previously showed that Toxoplasma interacts with many host cell organelles, especially from recycling pathways, and sequestrates Rab11A and Rab11B vesicles into the PV. Here, we examine the specificity of host Rab11 vesicle interaction with the PV by focusing on the recruitment of subpopulations of Rab11 vesicles characterized by different effectors, for example, Rab11-family interacting roteins (FIPs) or Arf6. Our quantitative microscopic analysis illustrates the presence of intra-PV vesicles with FIPs from class I (FIP1C, FIP2, FIP5) and class II (FIP3, FIP4) but to various degrees. The intra-PV delivery of vesicles with class I, but not class II, FIPs is dependent on Rab11 binding. Cell depletion of Rab11A results in a significant decrease in intra-PV FIP5, but not FIP3 vesicles. Class II FIPs also bind to Arf6, and we observe vesicles associated with FIP3-Rab11A or FIP3-Arf6 complexes concomitantly within the PV. Abolishing FIP3 binding to both Rab11 and Arf6 reduces the number of intra-PV FIP3 vesicles. These data point to a selective process of mammalian Rab11 vesicle recognition and scavenging mediated by Toxoplasma, suggesting that specific parasite PV proteins may be involved in these processes.


Asunto(s)
Toxoplasma , Factor 6 de Ribosilación del ADP , Animales , Endosomas/metabolismo , Células HeLa , Humanos , Mamíferos/metabolismo , Unión Proteica , Toxoplasma/metabolismo , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo
6.
Methods Mol Biol ; 2293: 295-305, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34453726

RESUMEN

Fluorescence microscopy and image analysis are powerful techniques to examine the distribution and interactions of different cellular compartments, including mammalian organelles with intravacuolar pathogens. Toxoplasma gondii is an obligate intracellular protozoan parasite that forms a membrane-bound compartment, the parasitophorous vacuole (PV), upon invasion of mammalian cells. From within the PV, the parasite interacts with many host organelles (without fusion), redirects host vesicles decorated with Rab GTPases to the PV, and internalizes many of these nutrient-filled Rab vesicles into the PV. Here, we report a method to distinguish the host Rab vesicles that are exclusively trapped in the Toxoplasma PV from those localized along the edge of the vacuole. Such a discrimination between the two Rab vesicle populations (inside versus outside of the PV) allows the selective characterization of the intra-PV Rab vesicles, for example, number per PV, volume, and distance from the PV centroid, as well as comparisons between wild-type and mutant Toxoplasma.


Asunto(s)
Toxoplasma , Animales , Interacciones Huésped-Parásitos , Humanos , Microscopía Fluorescente , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo
7.
NPJ Vaccines ; 6(1): 98, 2021 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376691

RESUMEN

Genetically attenuated sporozoite vaccines can elicit long-lasting protection against malaria but pose risks of breakthrough infection. Chemoprophylaxis vaccination (CVac) has proven to be the most effective vaccine strategy against malaria. Here, we demonstrate that a liver stage-specific autophagy mutant of Plasmodium berghei (ATG8 overexpressor), when used as a live vaccine under a CVac regimen, provides superior long-lasting protection, in both inbred and outbred mice, as compared to WT-CVac. Uniquely, the protection elicited by this mutant is predominantly dependent on a CD8+ T-cell response through an IFN-γ-independent mechanism and is associated with a stable population of antigen-experienced CD8+ T cells. Jointly, our findings support the exploitation of liver-stage mutants as vaccines under a CVac protocol. This vaccination strategy is also a powerful model to study the mechanisms of protective immunity and discover new protective antigens.

8.
PLoS Pathog ; 16(12): e1009067, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33383579

RESUMEN

Inorganic ions such as phosphate, are essential nutrients required for a broad spectrum of cellular functions and regulation. During infection, pathogens must obtain inorganic phosphate (Pi) from the host. Despite the essentiality of phosphate for all forms of life, how the intracellular parasite Toxoplasma gondii acquires Pi from the host cell is still unknown. In this study, we demonstrated that Toxoplasma actively internalizes exogenous Pi by exploiting a gradient of Na+ ions to drive Pi uptake across the plasma membrane. The Na+-dependent phosphate transport mechanism is electrogenic and functionally coupled to a cipargarmin sensitive Na+-H+-ATPase. Toxoplasma expresses one transmembrane Pi transporter harboring PHO4 binding domains that typify the PiT Family. This transporter named TgPiT, localizes to the plasma membrane, the inward buds of the endosomal organelles termed VAC, and many cytoplasmic vesicles. Upon Pi limitation in the medium, TgPiT is more abundant at the plasma membrane. We genetically ablated the PiT gene, and ΔTgPiT parasites are impaired in importing Pi and synthesizing polyphosphates. Interestingly, ΔTgPiT parasites accumulate 4-times more acidocalcisomes, storage organelles for phosphate molecules, as compared to parental parasites. In addition, these mutants have a reduced cell volume, enlarged VAC organelles, defects in calcium storage and a slightly alkaline pH. Overall, these mutants exhibit severe growth defects and have reduced acute virulence in mice. In survival mode, ΔTgPiT parasites upregulate several genes, including those encoding enzymes that cleave or transfer phosphate groups from phosphometabolites, transporters and ions exchangers localized to VAC or acidocalcisomes. Taken together, these findings point to a critical role of TgPiT for Pi supply for Toxoplasma and also for protection against osmotic stresses.


Asunto(s)
Osmorregulación/genética , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/fisiología , Toxoplasma , Animales , Animales Modificados Genéticamente , Transporte Biológico/genética , Células Cultivadas , Humanos , Ratones , Proteínas Cotransportadoras de Sodio-Fosfato/genética , Toxoplasma/genética , Toxoplasma/metabolismo
9.
Biol Cell ; 112(7): 187-195, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32180234

RESUMEN

Many intracellular microbial pathogens subvert, disrupt or otherwise modulate host membrane trafficking pathways to establish a successful infection. Among them, bacteria that are trapped in a phagosome during mammalian cell invasion, disengage the programmed degradation process by altering the identity of their replicative niche through the exclusion or recruitment of specific Rab GTPases to their vacuole. Many viruses co-opt essential cellular trafficking pathways to perform key steps in their lifecycles. Among protozoan parasites, Apicomplexa are obligate intracellular microbes that invade mammalian cells by creating a unique, nonfusogenic membrane-bound compartment that protects the parasites straightaway from lysosomal degradation. Recent compelling evidence demonstrates that apicomplexan parasites are master manipulators of mammalian Rab GTPase proteins, and benefit or antagonise Rab functions for development within host cells. This review covers the exploitation of mammalian Rab proteins and vesicles by Apicomplexa, focusing on Toxoplasma, Neospora, Plasmodium and Theileria parasites.


Asunto(s)
Apicomplexa/enzimología , Proteínas de Unión al GTP rab/metabolismo , Humanos
10.
PLoS One ; 13(12): e0208558, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30521607

RESUMEN

Three-dimensional (3D) cell culture models bridge the gap between two-dimensional (2D) monolayer cultures and animal models. Physiologically relevant, 3D culture models have significantly advanced basic cell science and provide unique insights into host-pathogen interactions intrinsically linked to cell morphology. Toxoplasma gondii is an obligate intravacuolar parasite that chronically infects a large portion of the global human population. Our current understanding of Toxoplasma infection is largely based on 2D cell cultures, in which mammalian cells are grown on flat surfaces. However, 2D cell cultures may not recapitulate key conditions of in vivo infections as they introduce artificial pressures and tensions, which may subsequently alter infectious processes that are dependent on spatiality, e.g., invasion, replication and egress. In this study, we adapted a collagen-based 3D cell culture system to reproduce the 3D environment of T. gondii natural infections for investigation of the replication and egress of the parasite from the parasitophorous vacuole. Suspended in the 3D matrix, Toxoplasma-infected VERO cells have round morphology, as opposed to infected VERO cells in 2D monolayers. The doubling time of Toxoplasma in VERO cells within the matrix is comparable to that of parasites cultivated in VERO cell monolayers. In the absence of the pressure of flattened host cells grown in 2D cultures, the parasitophorous vacuole of T. gondii has a globular shape, with intravacuolar parasites distributed radially, forming 3D spherical 'rosette' structures. Parasites egress radially away from the ruptured host cell in 3D matrices, in contrast to Toxoplasma cultivated in 2D monolayer cultures, where the parasites escape perpendicularly from the flat surface below the host cells. These observations demonstrate the utility of collagen matrices for studying parasite modes of infection as these 3D assays more closely mimic in vivo conditions.


Asunto(s)
Técnicas de Cultivo de Célula , Toxoplasma/fisiología , Animales , Microambiente Celular , Chlorocebus aethiops , Modelos Biológicos , Toxoplasmosis/parasitología , Células Vero
11.
Artículo en Inglés | MEDLINE | ID: mdl-30061287

RESUMEN

Toxoplasma gondii, an obligate intracellular parasite replicating in mammalian cells within a parasitophorous vacuole (PV), is an avid scavenger of lipids retrieved from the host cell. Following lipid uptake, this parasite stores excess lipids in lipid droplets (LD). Here, we examined the lipid storage capacities of Toxoplasma upon supplementation of the culture medium with various fatty acids at physiological concentrations. Supplemental unsaturated fatty acids (oleate [OA], palmitoleate, linoleate) accumulate in large LD and impair parasite replication, whereas saturated fatty acids (palmitate, stearate) neither stimulate LD formation nor impact growth. Examination of parasite growth defects with 0.4 mM OA revealed massive lipid deposits outside LD, indicating enzymatic inadequacies for storing neutral lipids in LD in response to the copious salvage of OA. Toxoplasma exposure to 0.5 mM OA led to irreversible growth arrest and lipid-induced damage, confirming a major disconnect between fatty acid uptake and the parasite's cellular lipid requirements. The importance of neutral lipid synthesis and storage to avoid lipotoxicity was further highlighted by the selective vulnerability of Toxoplasma, both the proliferative and the encysted forms, to subtoxic concentrations of the acyl coenzyme A:diacylglycerol acyltransferase 1 (DGAT1) pharmacological inhibitor T863. T863-treated parasites did not form LD but instead built up large membranous structures within the cytoplasm, which suggests improper channeling and management of the excess lipid. Dual addition of OA and T863 to infected cells intensified the deterioration of the parasite. Overall, our data pinpoint Toxoplasma DGAT as a promising drug target for the treatment of toxoplasmosis that would not incur the risk of toxicity for mammalian cells.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Gotas Lipídicas/metabolismo , Toxoplasma/metabolismo , Animales , Ácidos Grasos Monoinsaturados/metabolismo , Ácido Linoleico/metabolismo , Ácido Oléico/metabolismo , Ácido Palmítico/metabolismo
12.
PLoS Pathog ; 14(4): e1007018, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29689101

RESUMEN

[This corrects the article DOI: 10.1371/journal.ppat.1006893.].

14.
J Cell Biol ; 216(12): 4235-4254, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29070609

RESUMEN

Many intracellular pathogens subvert host membrane trafficking pathways to promote their replication. Toxoplasma multiplies in a membrane-bound parasitophorous vacuole (PV) that interacts with mammalian host organelles and intercepts Golgi Rab vesicles to acquire sphingolipids. The mechanisms of host vesicle internalization and processing within the PV remain undefined. We demonstrate that Toxoplasma sequesters a broad range of Rab vesicles into the PV. Correlative light and electron microscopy analysis of infected cells illustrates that intravacuolar Rab1A vesicles are surrounded by the PV membrane, suggesting a phagocytic-like process for vesicle engulfment. Rab11A vesicles concentrate to an intravacuolar network (IVN), but this is reduced in Δgra2 and Δgra2Δgra6 parasites, suggesting that tubules stabilized by the TgGRA2 and TgGRA6 proteins secreted by the parasite within the PV contribute to host vesicle sequestration. Overexpression of a phospholipase TgLCAT, which is localized to the IVN, results in a decrease in the number of intravacuolar GFP-Rab11A vesicles, suggesting that TgLCAT controls lipolytic degradation of Rab vesicles for cargo release.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Interacciones Huésped-Parásitos , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Toxoplasma/metabolismo , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Células CHO , Chlorocebus aethiops , Cricetulus , Vesículas Citoplasmáticas/ultraestructura , Fibroblastos/metabolismo , Fibroblastos/parasitología , Fibroblastos/ultraestructura , Regulación de la Expresión Génica , Genes Reporteros , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Fagocitosis , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esfingolípidos/metabolismo , Toxoplasma/ultraestructura , Vacuolas/ultraestructura , Células Vero , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab1/genética , Proteínas de Unión al GTP rab1/metabolismo
15.
PLoS Pathog ; 13(6): e1006362, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28570716

RESUMEN

Toxoplasma is an obligate intracellular parasite that replicates in mammalian cells within a parasitophorous vacuole (PV) that does not fuse with any host organelles. One mechanism developed by the parasite for nutrient acquisition is the attraction of host organelles to the PV. Here, we examined the exploitation of host lipid droplets (LD), ubiquitous fat storage organelles, by Toxoplasma. We show that Toxoplasma replication is reduced in host cells that are depleted of LD, or impaired in TAG lipolysis or fatty acid catabolism. In infected cells, the number of host LD and the expression of host LD-associated genes (ADRP, DGAT2), progressively increase until the onset of parasite replication. Throughout infection, the PV are surrounded by host LD. Toxoplasma is capable of accessing lipids stored in host LD and incorporates these lipids into its own membranes and LD. Exogenous addition of oleic acid stimulates LD biogenesis in the host cell and results in the overaccumulation of neutral lipids in very large LD inside the parasite. To access LD-derived lipids, Toxoplasma intercepts and internalizes within the PV host LD, some of which remaining associated with Rab7, which become wrapped by an intravacuolar network of membranes (IVN). Mutant parasites impaired in IVN formation display diminished capacity of lipid uptake from host LD. Moreover, parasites lacking an IVN-localized phospholipase A2 are less proficient in salvaging lipids from host LD in the PV, suggesting a major contribution of the IVN for host LD processing in the PV and, thus lipid content release. Interestingly, gavage of parasites with lipids unveils, for the first time, the presence in Toxoplasma of endocytic-like structures containing lipidic material originating from the PV lumen. This study highlights the reliance of Toxoplasma on host LD for its intracellular development and the parasite's capability in scavenging neutral lipids from host LD.


Asunto(s)
Gotas Lipídicas/parasitología , Toxoplasma/fisiología , Toxoplasmosis/parasitología , Línea Celular , Interacciones Huésped-Parásitos , Humanos , Toxoplasma/genética , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis/metabolismo , Toxoplasmosis/fisiopatología
16.
J Biol Chem ; 291(8): 3725-46, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26694607

RESUMEN

The protozoan parasite Toxoplasma gondii develops within a parasitophorous vacuole (PV) in mammalian cells, where it scavenges cholesterol. When cholesterol is present in excess in its environment, the parasite expulses this lipid into the PV or esterifies it for storage in lipid bodies. Here, we characterized a unique T. gondii homologue of mammalian lecithin:cholesterol acyltransferase (LCAT), a key enzyme that produces cholesteryl esters via transfer of acyl groups from phospholipids to the 3-OH of free cholesterol, leading to the removal of excess cholesterol from tissues. TgLCAT contains a motif characteristic of serine lipases "AHSLG" and the catalytic triad consisting of serine, aspartate, and histidine (SDH) from LCAT enzymes. TgLCAT is secreted by the parasite, but unlike other LCAT enzymes it is cleaved into two proteolytic fragments that share the residues of the catalytic triad and need to be reassembled to reconstitute enzymatic activity. TgLCAT uses phosphatidylcholine as substrate to form lysophosphatidylcholine that has the potential to disrupt membranes. The released fatty acid is transferred to cholesterol, but with a lower transesterification activity than mammalian LCAT. TgLCAT is stored in a subpopulation of dense granule secretory organelles, and following secretion, it localizes to the PV and parasite plasma membrane. LCAT-null parasites have impaired growth in vitro, reduced virulence in animals, and exhibit delays in egress from host cells. Parasites overexpressing LCAT show increased virulence and faster egress. These observations demonstrate that TgLCAT influences the outcome of an infection, presumably by facilitating replication and egress depending on the developmental stage of the parasite.


Asunto(s)
Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/enzimología , Toxoplasma/patogenicidad , Toxoplasmosis/enzimología , Dominio Catalítico , Línea Celular , Humanos , Fosfatidilcolina-Esterol O-Aciltransferasa/química , Fosfatidilcolina-Esterol O-Aciltransferasa/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Toxoplasma/genética , Toxoplasmosis/genética , Toxoplasmosis/patología
17.
Eukaryot Cell ; 14(5): 454-73, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25750213

RESUMEN

Toxoplasma gondii and Neospora caninum, which cause the diseases toxoplasmosis and neosporosis, respectively, are two closely related apicomplexan parasites. They have similar heteroxenous life cycles and conserved genomes and share many metabolic features. Despite these similarities, T. gondii and N. caninum differ in their transmission strategies and zoonotic potential. Comparative analyses of the two parasites are important to identify the unique biological features that underlie the basis of host preference and pathogenicity. T. gondii and N. caninum are obligate intravacuolar parasites; in contrast to T. gondii, events that occur during N. caninum infection remain largely uncharacterized. We examined the capability of N. caninum (Liverpool isolate) to interact with host organelles and scavenge nutrients in comparison to that of T. gondii (RH strain). N. caninum reorganizes the host microtubular cytoskeleton and attracts endoplasmic reticulum (ER), mitochondria, lysosomes, multivesicular bodies, and Golgi vesicles to its vacuole though with some notable differences from T. gondii. For example, the host ER gathers around the N. caninum parasitophorous vacuole (PV) but does not physically associate with the vacuolar membrane; the host Golgi apparatus surrounds the N. caninum PV but does not fragment into ministacks. N. caninum relies on plasma lipoproteins and scavenges cholesterol from NPC1-containing endocytic organelles. This parasite salvages sphingolipids from host Golgi Rab14 vesicles that it sequesters into its vacuole. Our data highlight a remarkable degree of conservation in the intracellular infection program of N. caninum and T. gondii. The minor differences between the two parasites related to the recruitment and rearrangement of host organelles around their vacuoles likely reflect divergent evolutionary paths.


Asunto(s)
Neospora/metabolismo , Toxoplasma/metabolismo , Vacuolas/metabolismo , Animales , Línea Celular , Retículo Endoplásmico/metabolismo , Interacciones Huésped-Parásitos , Humanos , Metabolismo de los Lípidos , Lípidos
18.
Autophagy ; 10(2): 269-84, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24342964

RESUMEN

Plasmodium parasites successfully colonize different habitats within mammals and mosquitoes, and adaptation to various environments is accompanied by changes in their organelle composition and size. Previously, we observed that during hepatocyte infection, Plasmodium discards organelles involved in invasion and expands those implicated in biosynthetic pathways. We hypothesized that this process is regulated by autophagy. Plasmodium spp. possess a rudimentary set of known autophagy-related proteins that includes the ortholog of yeast Atg8. In this study, we analyzed the activity of the ATG8-conjugation pathway over the course of the lifecycle of Plasmodium falciparum and during the liver stage of Plasmodium berghei. We engineered a transgenic P. falciparum strain expressing mCherry-PfATG8. These transgenic parasites expressed mCherry-PfATG8 in human hepatocytes and erythrocytes, and in the midgut and salivary glands of Anopheles mosquitoes. In all observed stages, mCherry-PfATG8 was localized to tubular structures. Our EM and colocalization studies done in P. berghei showed the association of PbATG8 on the limiting membranes of the endosymbiont-derived plastid-like organelle known as the apicoplast. Interestingly, during parasite replication in hepatocytes, the association of PbATG8 with the apicoplast increases as this organelle expands in size. PbATG3, PbATG7 and PbATG8 are cotranscribed in all parasitic stages. Molecular analysis of PbATG8 and PbATG3 revealed a novel mechanism of interaction compared with that observed for other orthologs. This is further supported by the inability of Plasmodium ATG8 to functionally complement atg8Δ yeast or localize to autophagosomes in starved mammalian cells. Altogether, these data suggests a unique role for the ATG8-conjugation system in Plasmodium parasites.


Asunto(s)
Apicoplastos/inmunología , Autofagia/inmunología , Hígado/microbiología , Parásitos/inmunología , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Animales , Antígenos de Protozoos/inmunología , Familia de las Proteínas 8 Relacionadas con la Autofagia , Femenino , Hepatocitos/metabolismo , Hígado/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/inmunología , Parásitos/metabolismo , Fagosomas/inmunología , Saccharomyces cerevisiae/inmunología , Proteínas de Saccharomyces cerevisiae/inmunología
19.
Pathog Dis ; 69(2): 72-86, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23821471

RESUMEN

The bacterium Chlamydia trachomatis and the protozoan parasite Toxoplasma gondii are the causative agents of chlamydiosis and toxoplasmosis in humans, respectively. Both microorganisms are obligate intracellular pathogens and notorious for extensively modifying the cytoskeletal architecture and the endomembrane system of their host cells to establish productive infections. This review highlights the similar tactics developed by these two pathogens to manipulate their host cell despite their genetic unrelatedness. Using an in vitro cell culture model whereby single fibroblasts are infected by C. trachomatis and T. gondii simultaneously, thus setting up an intracellular competition, we demonstrate that the solutions to the problem of intracellular survival deployed by the parasite and the bacterium may represent an example of convergent evolution, driven by the necessity to acquire nutrients in a hostile environment.


Asunto(s)
Infecciones por Chlamydia/complicaciones , Coinfección/microbiología , Coinfección/parasitología , Orgánulos/microbiología , Orgánulos/parasitología , Toxoplasmosis/complicaciones , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/fisiología , Fibroblastos/microbiología , Fibroblastos/parasitología , Humanos , Modelos Teóricos , Toxoplasma/fisiología , Toxoplasmosis/parasitología
20.
Mol Biol Cell ; 24(12): 1974-95, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23615442

RESUMEN

The obligate intracellular protozoan Toxoplasma gondii actively invades mammalian cells and, upon entry, forms its own membrane-bound compartment, named the parasitophorous vacuole (PV). Within the PV, the parasite replicates and scavenges nutrients, including lipids, from host organelles. Although T. gondii can synthesize sphingolipids de novo, it also scavenges these lipids from the host Golgi. How the parasite obtains sphingolipids from the Golgi remains unclear, as the PV avoids fusion with host organelles. In this study, we explore the host Golgi-PV interaction and evaluate the importance of host-derived sphingolipids for parasite growth. We demonstrate that the PV preferentially localizes near the host Golgi early during infection and remains closely associated with this organelle throughout infection. The parasite subverts the structure of the host Golgi, resulting in its fragmentation into numerous ministacks, which surround the PV, and hijacks host Golgi-derived vesicles within the PV. These vesicles, marked with Rab14, Rab30, or Rab43, colocalize with host-derived sphingolipids in the vacuolar space. Scavenged sphingolipids contribute to parasite replication since alterations in host sphingolipid metabolism are detrimental for the parasite's growth. Thus our results reveal that T. gondii relies on host-derived sphingolipids for its development and scavenges these lipids via Golgi-derived vesicles.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Aparato de Golgi/metabolismo , Esfingolípidos/metabolismo , Toxoplasma/metabolismo , Vacuolas/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Células CHO , Línea Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Vesículas Citoplasmáticas/parasitología , Vesículas Citoplasmáticas/ultraestructura , Aparato de Golgi/parasitología , Aparato de Golgi/ultraestructura , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Interacciones Huésped-Parásitos , Humanos , Immunoblotting , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Mutación , Toxoplasma/crecimiento & desarrollo , Toxoplasma/fisiología , Vacuolas/parasitología , Vacuolas/ultraestructura , Células Vero , Proteínas de Unión al GTP rab/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...