Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730853

RESUMEN

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Asunto(s)
Genoma de Protozoos , Estadios del Ciclo de Vida/genética , Hígado/metabolismo , Hígado/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Alelos , Amino Azúcares/biosíntesis , Animales , Culicidae/parasitología , Eritrocitos/parasitología , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Técnicas de Inactivación de Genes , Genotipo , Modelos Biológicos , Mutación/genética , Parásitos/genética , Parásitos/crecimiento & desarrollo , Fenotipo , Plasmodium berghei/metabolismo , Ploidias , Reproducción
2.
Cell ; 170(2): 260-272.e8, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28708996

RESUMEN

The genomes of malaria parasites contain many genes of unknown function. To assist drug development through the identification of essential genes and pathways, we have measured competitive growth rates in mice of 2,578 barcoded Plasmodium berghei knockout mutants, representing >50% of the genome, and created a phenotype database. At a single stage of its complex life cycle, P. berghei requires two-thirds of genes for optimal growth, the highest proportion reported from any organism and a probable consequence of functional optimization necessitated by genomic reductions during the evolution of parasitism. In contrast, extreme functional redundancy has evolved among expanded gene families operating at the parasite-host interface. The level of genetic redundancy in a single-celled organism may thus reflect the degree of environmental variation it experiences. In the case of Plasmodium parasites, this helps rationalize both the relative successes of drugs and the greater difficulty of making an effective vaccine.


Asunto(s)
Genoma de Protozoos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Animales , Evolución Biológica , Femenino , Técnicas de Inactivación de Genes , Genes Esenciales , Interacciones Huésped-Parásitos , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos BALB C , Plasmodium berghei/metabolismo , Saccharomyces cerevisiae/genética , Toxoplasma/genética , Trypanosoma brucei brucei/genética
3.
Nature ; 547(7662): 213-216, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28678779

RESUMEN

The lifestyle of intracellular pathogens, such as malaria parasites, is intimately connected to that of their host, primarily for nutrient supply. Nutrients act not only as primary sources of energy but also as regulators of gene expression, metabolism and growth, through various signalling networks that enable cells to sense and adapt to varying environmental conditions. Canonical nutrient-sensing pathways are presumed to be absent from the causative agent of malaria, Plasmodium, thus raising the question of whether these parasites can sense and cope with fluctuations in host nutrient levels. Here we show that Plasmodium blood-stage parasites actively respond to host dietary calorie alterations through rearrangement of their transcriptome accompanied by substantial adjustment of their multiplication rate. A kinome analysis combined with chemical and genetic approaches identified KIN as a critical regulator that mediates sensing of nutrients and controls a transcriptional response to the host nutritional status. KIN shares homology with SNF1/AMPKα, and yeast complementation studies suggest that it is part of a functionally conserved cellular energy-sensing pathway. Overall, these findings reveal a key parasite nutrient-sensing mechanism that is critical for modulating parasite replication and virulence.


Asunto(s)
Regulación de la Expresión Génica , Malaria/parasitología , Parásitos/metabolismo , Parásitos/patogenicidad , Fosfotransferasas/metabolismo , Plasmodium/metabolismo , Plasmodium/patogenicidad , Animales , Restricción Calórica , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Complementación Genética , Glucosa/metabolismo , Glucosa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Parasitemia/sangre , Parasitemia/genética , Parasitemia/metabolismo , Parasitemia/parasitología , Parásitos/genética , Parásitos/crecimiento & desarrollo , Fosfotransferasas/deficiencia , Fosfotransferasas/genética , Plasmodium/genética , Plasmodium/crecimiento & desarrollo , Ratas , Transcriptoma/efectos de los fármacos , Virulencia/efectos de los fármacos
4.
Mol Cell Proteomics ; 20: 100038, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33515807

RESUMEN

Sporozoites are a motile form of malaria-causing Plasmodium falciparum parasites that migrate from the site of transmission in the dermis through the bloodstream to invade hepatocytes. Sporozoites interact with many cells within the host, but the molecular identity of these interactions and their role in the pathology of malaria is poorly understood. Parasite proteins that are secreted and embedded within membranes are known to be important for these interactions, but our understanding of how they interact with each other to form functional complexes is largely unknown. Here, we compile a library of recombinant proteins representing the repertoire of cell surface and secreted proteins from the P. falciparum sporozoite and use an assay designed to detect extracellular interactions to systematically identify complexes. We identify three protein complexes including an interaction between two components of the p24 complex that is involved in the trafficking of glycosylphosphatidylinositol-anchored proteins through the secretory pathway. Plasmodium parasites lacking either gene are strongly inhibited in the establishment of liver-stage infections. These findings reveal an important role for the p24 complex in malaria pathogenesis and show that the library of recombinant proteins represents a valuable resource to investigate P. falciparum sporozoite biology.


Asunto(s)
Interacciones Huésped-Parásitos , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Animales , Femenino , Malaria/parasitología , Ratones Endogámicos BALB C , Organismos Modificados Genéticamente , Fenotipo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/fisiología , Mapas de Interacción de Proteínas , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Esporozoítos/fisiología
5.
Genome Res ; 28(4): 547-560, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29500236

RESUMEN

Avian malaria parasites are prevalent around the world and infect a wide diversity of bird species. Here, we report the sequencing and analysis of high-quality draft genome sequences for two avian malaria species, Plasmodium relictum and Plasmodium gallinaceum We identify 50 genes that are specific to avian malaria, located in an otherwise conserved core of the genome that shares gene synteny with all other sequenced malaria genomes. Phylogenetic analysis suggests that the avian malaria species form an outgroup to the mammalian Plasmodium species, and using amino acid divergence between species, we estimate the avian- and mammalian-infective lineages diverged in the order of 10 million years ago. Consistent with their phylogenetic position, we identify orthologs of genes that had previously appeared to be restricted to the clades of parasites containing Plasmodium falciparum and Plasmodium vivax, the species with the greatest impact on human health. From these orthologs, we explore differential diversifying selection across the genus and show that the avian lineage is remarkable in the extent to which invasion-related genes are evolving. The subtelomeres of the P. relictum and P. gallinaceum genomes contain several novel gene families, including an expanded surf multigene family. We also identify an expansion of reticulocyte binding protein homologs in P. relictum, and within these proteins, we detect distinct regions that are specific to nonhuman primate, humans, rodent, and avian hosts. For the first time in the Plasmodium lineage, we find evidence of transposable elements, including several hundred fragments of LTR-retrotransposons in both species and an apparently complete LTR-retrotransposon in the genome of P. gallinaceum.


Asunto(s)
Malaria Aviar/genética , Plasmodium falciparum/genética , Plasmodium vivax/genética , Plasmodium/genética , Animales , Aves/parasitología , Evolución Molecular , Humanos , Malaria Aviar/parasitología , Mamíferos/parasitología , Filogenia , Plasmodium/patogenicidad , Plasmodium falciparum/patogenicidad , Plasmodium vivax/patogenicidad
6.
Proc Natl Acad Sci U S A ; 115(17): 4477-4482, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29632205

RESUMEN

Malaria-causing Plasmodium sporozoites are deposited in the dermis by the bite of an infected mosquito and move by gliding motility to the liver where they invade and develop within host hepatocytes. Although extracellular interactions between Plasmodium sporozoite ligands and host receptors provide important guidance cues for productive infection and are good vaccine targets, these interactions remain largely uncharacterized. Thrombospondin-related anonymous protein (TRAP) is a parasite cell surface ligand that is essential for both gliding motility and invasion because it couples the extracellular binding of host receptors to the parasite cytoplasmic actinomyosin motor; however, the molecular nature of the host TRAP receptors is poorly defined. Here, we use a systematic extracellular protein interaction screening approach to identify the integrin αvß3 as a directly interacting host receptor for Plasmodium falciparum TRAP. Biochemical characterization of the interaction suggests a two-site binding model, requiring contributions from both the von Willebrand factor A domain and the RGD motif of TRAP for integrin binding. We show that TRAP binding to cells is promoted in the presence of integrin-activating proadhesive Mn2+ ions, and that cells genetically targeted so that they lack cell surface expression of the integrin αv-subunit are no longer able to bind TRAP. P. falciparum sporozoites moved with greater speed in the dermis of Itgb3-deficient mice, suggesting that the interaction has a role in sporozoite migration. The identification of the integrin αvß3 as the host receptor for TRAP provides an important demonstration of a sporozoite surface ligand that directly interacts with host receptors.


Asunto(s)
Integrina alfaVbeta3/metabolismo , Modelos Biológicos , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Animales , Células HEK293 , Humanos , Integrina alfaVbeta3/genética , Ratones , Ratones Noqueados , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/genética , Esporozoítos/genética
7.
Genes Dev ; 27(10): 1198-215, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23699412

RESUMEN

Fertilization is a crucial yet poorly characterized event in eukaryotes. Our previous discovery that the broadly conserved protein HAP2 (GCS1) functioned in gamete membrane fusion in the unicellular green alga Chlamydomonas and the malaria pathogen Plasmodium led us to exploit the rare biological phenomenon of isogamy in Chlamydomonas in a comparative transcriptomics strategy to uncover additional conserved sexual reproduction genes. All previously identified Chlamydomonas fertilization-essential genes fell into related clusters based on their expression patterns. Out of several conserved genes in a minus gamete cluster, we focused on Cre06.g280600, an ortholog of the fertilization-related Arabidopsis GEX1. Gene disruption, cell biological, and immunolocalization studies show that CrGEX1 functions in nuclear fusion in Chlamydomonas. Moreover, CrGEX1 and its Plasmodium ortholog, PBANKA_113980, are essential for production of viable meiotic progeny in both organisms and thus for mosquito transmission of malaria. Remarkably, we discovered that the genes are members of a large, previously unrecognized family whose first-characterized member, KAR5, is essential for nuclear fusion during yeast sexual reproduction. Our comparative transcriptomics approach provides a new resource for studying sexual development and demonstrates that exploiting the data can lead to the discovery of novel biology that is conserved across distant taxa.


Asunto(s)
Chlamydomonas/genética , Hongos/genética , Genes Esenciales , Membrana Nuclear/metabolismo , Proteínas Nucleares/clasificación , Plasmodium/genética , Vertebrados/genética , Animales , Proteínas de Arabidopsis/clasificación , Proteínas de Arabidopsis/metabolismo , Fertilización/genética , Hongos/crecimiento & desarrollo , Perfilación de la Expresión Génica , Meiosis , Proteínas de la Membrana/clasificación , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plantas/genética , Reproducción/genética , Proteínas de Saccharomyces cerevisiae/clasificación , Proteínas de Saccharomyces cerevisiae/metabolismo , Transcriptoma/genética
8.
Nature ; 507(7491): 253-257, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24572359

RESUMEN

Commitment to and completion of sexual development are essential for malaria parasites (protists of the genus Plasmodium) to be transmitted through mosquitoes. The molecular mechanism(s) responsible for commitment have been hitherto unknown. Here we show that PbAP2-G, a conserved member of the apicomplexan AP2 (ApiAP2) family of DNA-binding proteins, is essential for the commitment of asexually replicating forms to sexual development in Plasmodium berghei, a malaria parasite of rodents. PbAP2-G was identified from mutations in its encoding gene, PBANKA_143750, which account for the loss of sexual development frequently observed in parasites transmitted artificially by blood passage. Systematic gene deletion of conserved ApiAP2 genes in Plasmodium confirmed the role of PbAP2-G and revealed a second ApiAP2 member (PBANKA_103430, here termed PbAP2-G2) that significantly modulates but does not abolish gametocytogenesis, indicating that a cascade of ApiAP2 proteins are involved in commitment to the production and maturation of gametocytes. The data suggest a mechanism of commitment to gametocytogenesis in Plasmodium consistent with a positive feedback loop involving PbAP2-G that could be exploited to prevent the transmission of this pernicious parasite.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Germinativas/crecimiento & desarrollo , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Desarrollo Sexual/genética , Animales , Culicidae/parasitología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Retroalimentación Fisiológica , Femenino , Regulación de la Expresión Génica , Células Germinativas/citología , Células Germinativas/metabolismo , Masculino , Mutación/genética , Plasmodium berghei/citología , Transporte de Proteínas , Proteínas Protozoarias/genética , Reproducción Asexuada , Transcripción Genética
9.
Mol Microbiol ; 100(3): 397-408, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26748879

RESUMEN

Ca(2+) is a ubiquitous intracellular messenger in malaria parasites with important functions in asexual blood stages responsible for malaria symptoms, the preceding liver-stage infection and transmission through the mosquito. Intracellular messengers amplify signals by binding to effector molecules that trigger physiological changes. The characterisation of some Ca(2+) effector proteins has begun to provide insights into the vast range of biological processes controlled by Ca(2+) signalling in malaria parasites, including host cell egress and invasion, protein secretion, motility and cell cycle regulation. Despite the importance of Ca(2+) signalling during the life cycle of malaria parasites, little is known about Ca(2+) homeostasis. Recent findings highlighted that upstream of stage-specific Ca(2+) effectors is a conserved interplay between second messengers to control critical intracellular Ca(2+) signals throughout the life cycle. The identification of the molecular mechanisms integrating stage-transcending mechanisms of Ca(2+) homeostasis in a network of stage-specific regulator and effector pathways now represents a major challenge for a meaningful understanding of Ca(2+) signalling in malaria parasites.


Asunto(s)
Señalización del Calcio/fisiología , Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Malaria Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Movimiento Celular/fisiología , Eritrocitos/parasitología , Estadios del Ciclo de Vida/fisiología , Malaria Falciparum/parasitología
10.
PLoS Pathog ; 11(9): e1005119, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26407009

RESUMEN

Inhibition of nitric oxide (NO) signaling may contribute to pathological activation of the vascular endothelium during severe malaria infection. Dimethylarginine dimethylaminohydrolase (DDAH) regulates endothelial NO synthesis by maintaining homeostasis between asymmetric dimethylarginine (ADMA), an endogenous NO synthase (NOS) inhibitor, and arginine, the NOS substrate. We carried out a community-based case-control study of Gambian children to determine whether ADMA and arginine homeostasis is disrupted during severe or uncomplicated malaria infections. Circulating plasma levels of ADMA and arginine were determined at initial presentation and 28 days later. Plasma ADMA/arginine ratios were elevated in children with acute severe malaria compared to 28-day follow-up values and compared to children with uncomplicated malaria or healthy children (p<0.0001 for each comparison). To test the hypothesis that DDAH1 is inactivated during Plasmodium infection, we examined DDAH1 in a mouse model of severe malaria. Plasmodium berghei ANKA infection inactivated hepatic DDAH1 via a post-transcriptional mechanism as evidenced by stable mRNA transcript number, decreased DDAH1 protein concentration, decreased enzyme activity, elevated tissue ADMA, elevated ADMA/arginine ratio in plasma, and decreased whole blood nitrite concentration. Loss of hepatic DDAH1 activity and disruption of ADMA/arginine homeostasis may contribute to severe malaria pathogenesis by inhibiting NO synthesis.


Asunto(s)
Amidohidrolasas/sangre , Arginina/sangre , Malaria/metabolismo , Óxido Nítrico/metabolismo , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Gambia , Homeostasis/fisiología , Humanos , Hígado/enzimología , Ratones
11.
Cell Microbiol ; 18(11): 1625-1641, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27084458

RESUMEN

As the Plasmodium parasite transitions between mammalian and mosquito host, it has to adjust quickly to new environments. Palmitoylation, a reversible and dynamic lipid post-translational modification, plays a central role in regulating this process and has been implicated with functions for parasite morphology, motility and host cell invasion. While proteins associated with the gliding motility machinery have been described to be palmitoylated, no palmitoyl transferase responsible for regulating gliding motility has previously been identified. Here, we characterize two palmityol transferases with gene tagging and gene deletion approaches. We identify DHHC3, a palmitoyl transferase, as a mediator of ookinete development, with a crucial role for gliding motility in ookinetes and sporozoites, and we co-localize the protein with a marker for the inner membrane complex in the ookinete stage. Ookinetes and sporozoites lacking DHHC3 are impaired in gliding motility and exhibit a strong phenotype in vivo; with ookinetes being significantly less infectious to their mosquito host and sporozoites being non-infectious to mice. Importantly, genetic complementation of the DHHC3-ko parasite completely restored virulence. We generated parasites lacking both DHHC3, as well as the palmitoyl transferase DHHC9, and found an enhanced phenotype for these double knockout parasites, allowing insights into the functional overlap and compensational nature of the large family of PbDHHCs. These findings contribute to our understanding of the organization and mechanism of the gliding motility machinery, which as is becoming increasingly clear, is mediated by palmitoylation.


Asunto(s)
Aciltransferasas/fisiología , Anopheles/parasitología , Hígado/parasitología , Plasmodium berghei/enzimología , Proteínas Protozoarias/fisiología , Animales , Células Hep G2 , Interacciones Huésped-Parásitos , Humanos , Lipoilación , Ratones , Oocistos/enzimología , Oocistos/crecimiento & desarrollo , Plasmodium berghei/fisiología , Procesamiento Proteico-Postraduccional , Glándulas Salivales/parasitología , Esporozoítos/enzimología , Esporozoítos/crecimiento & desarrollo
12.
PLoS Biol ; 12(3): e1001806, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24594931

RESUMEN

Many critical events in the Plasmodium life cycle rely on the controlled release of Ca²âº from intracellular stores to activate stage-specific Ca²âº-dependent protein kinases. Using the motility of Plasmodium berghei ookinetes as a signalling paradigm, we show that the cyclic guanosine monophosphate (cGMP)-dependent protein kinase, PKG, maintains the elevated level of cytosolic Ca²âº required for gliding motility. We find that the same PKG-dependent pathway operates upstream of the Ca²âº signals that mediate activation of P. berghei gametocytes in the mosquito and egress of Plasmodium falciparum merozoites from infected human erythrocytes. Perturbations of PKG signalling in gliding ookinetes have a marked impact on the phosphoproteome, with a significant enrichment of in vivo regulated sites in multiple pathways including vesicular trafficking and phosphoinositide metabolism. A global analysis of cellular phospholipids demonstrates that in gliding ookinetes PKG controls phosphoinositide biosynthesis, possibly through the subcellular localisation or activity of lipid kinases. Similarly, phosphoinositide metabolism links PKG to egress of P. falciparum merozoites, where inhibition of PKG blocks hydrolysis of phosphatidylinostitol (4,5)-bisphosphate. In the face of an increasing complexity of signalling through multiple Ca²âº effectors, PKG emerges as a unifying factor to control multiple cellular Ca²âº signals essential for malaria parasite development and transmission.


Asunto(s)
Señalización del Calcio , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , Fosfatidilinositoles/metabolismo , Plasmodium falciparum/fisiología , Animales , Culicidae/parasitología , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Interacciones Huésped-Parásitos , Humanos , Estadios del Ciclo de Vida , Malaria/parasitología , Modelos Biológicos , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo
13.
Nucleic Acids Res ; 43(Database issue): D1176-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25593348

RESUMEN

The Plasmodium Genetic Modification (PlasmoGEM) database (http://plasmogem.sanger.ac.uk) provides access to a resource of modular, versatile and adaptable vectors for genome modification of Plasmodium spp. parasites. PlasmoGEM currently consists of >2000 plasmids designed to modify the genome of Plasmodium berghei, a malaria parasite of rodents, which can be requested by non-profit research organisations free of charge. PlasmoGEM vectors are designed with long homology arms for efficient genome integration and carry gene specific barcodes to identify individual mutants. They can be used for a wide array of applications, including protein localisation, gene interaction studies and high-throughput genetic screens. The vector production pipeline is supported by a custom software suite that automates both the vector design process and quality control by full-length sequencing of the finished vectors. The PlasmoGEM web interface allows users to search a database of finished knock-out and gene tagging vectors, view details of their designs, download vector sequence in different formats and view available quality control data as well as suggested genotyping strategies. We also make gDNA library clones and intermediate vectors available for researchers to produce vectors for themselves.


Asunto(s)
Bases de Datos Genéticas , Plasmodium berghei/genética , Vectores Genéticos , Genoma de Protozoos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Internet , Mutación , Plásmidos , Plasmodium/genética , Programas Informáticos
14.
J Infect Dis ; 214(12): 1840-1849, 2016 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-27923948

RESUMEN

BACKGROUND: Plasmodium infection depletes arginine, the substrate for nitric oxide synthesis, and impairs endothelium-dependent vasodilation. Increased conversion of arginine to ornithine by parasites or host arginase is a proposed mechanism of arginine depletion. METHODS: We used high-performance liquid chromatography to measure plasma arginine, ornithine, and citrulline levels in Malawian children with cerebral malaria and in mice infected with Plasmodium berghei ANKA with or without the arginase gene. Heavy isotope-labeled tracers measured by quadrupole time-of-flight liquid chromatography-mass spectrometry were used to quantify the in vivo rate of appearance and interconversion of plasma arginine, ornithine, and citrulline in infected mice. RESULTS: Children with cerebral malaria and P. berghei-infected mice demonstrated depletion of plasma arginine, ornithine, and citrulline. Knock out of Plasmodium arginase did not alter arginine depletion in infected mice. Metabolic tracer analysis demonstrated that plasma arginase flux was unchanged by P. berghei infection. Instead, infected mice exhibited decreased rates of plasma arginine, ornithine, and citrulline appearance and decreased conversion of plasma citrulline to arginine. Notably, plasma arginine use by nitric oxide synthase was decreased in infected mice. CONCLUSIONS: Simultaneous arginine and ornithine depletion in malaria parasite-infected children cannot be fully explained by plasma arginase activity. Our mouse model studies suggest that plasma arginine depletion is driven primarily by a decreased rate of appearance.


Asunto(s)
Arginina/sangre , Malaria Cerebral/patología , Malaria/patología , Plasma/química , Plasmodium berghei/crecimiento & desarrollo , Animales , Arginasa/genética , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Citrulina/sangre , Femenino , Humanos , Lactante , Malaui , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ornitina/sangre , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
PLoS Pathog ; 10(7): e1004263, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25032958

RESUMEN

While the apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii are thought to primarily depend on glycolysis for ATP synthesis, recent studies have shown that they can fully catabolize glucose in a canonical TCA cycle. However, these parasites lack a mitochondrial isoform of pyruvate dehydrogenase and the identity of the enzyme that catalyses the conversion of pyruvate to acetyl-CoA remains enigmatic. Here we demonstrate that the mitochondrial branched chain ketoacid dehydrogenase (BCKDH) complex is the missing link, functionally replacing mitochondrial PDH in both T. gondii and P. berghei. Deletion of the E1a subunit of T. gondii and P. berghei BCKDH significantly impacted on intracellular growth and virulence of both parasites. Interestingly, disruption of the P. berghei E1a restricted parasite development to reticulocytes only and completely prevented maturation of oocysts during mosquito transmission. Overall this study highlights the importance of the molecular adaptation of BCKDH in this important class of pathogens.


Asunto(s)
Mitocondrias , Proteínas Mitocondriales/genética , Oxidorreductasas/genética , Plasmodium berghei , Proteínas Protozoarias/genética , Toxoplasma , Mitocondrias/enzimología , Mitocondrias/genética , Plasmodium berghei/enzimología , Plasmodium berghei/genética , Toxoplasma/enzimología , Toxoplasma/genética
16.
Traffic ; 14(8): 895-911, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23638681

RESUMEN

The advent of techniques to study palmitoylation on a whole proteome scale has revealed that it is an important reversible modification that plays a role in regulating multiple biological processes. Palmitoylation can control the affinity of a protein for lipid membranes, which allows it to impact protein trafficking, stability, folding, signalling and interactions. The publication of the palmitome of the schizont stage of Plasmodium falciparum implicated a role for palmitoylation in host cell invasion, protein export and organelle biogenesis. However, nothing is known so far about the repertoire of protein S-acyl transferases (PATs) that catalyse this modification in Apicomplexa. We undertook a comprehensive analysis of the repertoire of Asp-His-His-Cys cysteine-rich domain (DHHC-CRD) PAT family in Toxoplasma gondii and Plasmodium berghei by assessing their localization and essentiality. Unlike functional redundancies reported in other eukaryotes, some apicomplexan-specific DHHCs are essential for parasite growth, and several are targeted to organelles unique to this phylum. Of particular interest is DHHC7, which localizes to rhoptry organelles in all parasites tested, including the major human pathogen P. falciparum. TgDHHC7 interferes with the localization of the rhoptry palmitoylated protein TgARO and affects the apical positioning of the rhoptry organelles. This PAT has a major impact on T. gondii host cell invasion, but not on the parasite's ability to egress.


Asunto(s)
Acetiltransferasas/metabolismo , Plasmodium berghei/enzimología , Proteínas Protozoarias/metabolismo , Toxoplasma/enzimología , Acetiltransferasas/química , Acetiltransferasas/genética , Secuencias de Aminoácidos , Técnicas de Cultivo de Célula , Eliminación de Gen , Genoma de Protozoos , Humanos , Filogenia , Plasmodium berghei/patogenicidad , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Toxoplasma/patogenicidad
17.
PLoS Pathog ; 9(12): e1003811, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24348254

RESUMEN

Transmission of the malaria parasite to its vertebrate host involves an obligatory exoerythrocytic stage in which extensive asexual replication of the parasite takes place in infected hepatocytes. The resulting liver schizont undergoes segmentation to produce thousands of daughter merozoites. These are released to initiate the blood stage life cycle, which causes all the pathology associated with the disease. Whilst elements of liver stage merozoite biology are similar to those in the much better-studied blood stage merozoites, little is known of the molecular players involved in liver stage merozoite production. To facilitate the study of liver stage biology we developed a strategy for the rapid production of complex conditional alleles by recombinase mediated engineering in Escherichia coli, which we used in combination with existing Plasmodium berghei deleter lines expressing Flp recombinase to study subtilisin-like protease 1 (SUB1), a conserved Plasmodium serine protease previously implicated in blood stage merozoite maturation and egress. We demonstrate that SUB1 is not required for the early stages of intrahepatic growth, but is essential for complete development of the liver stage schizont and for production of hepatic merozoites. Our results indicate that inhibitors of SUB1 could be used in prophylactic approaches to control or block the clinically silent pre-erythrocytic stage of the malaria parasite life cycle.


Asunto(s)
Estadios del Ciclo de Vida/genética , Hígado/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Subtilisinas/fisiología , Animales , Anopheles/parasitología , Femenino , Células Hep G2 , Humanos , Merozoítos/crecimiento & desarrollo , Merozoítos/metabolismo , Ratones , Ratones Endogámicos C57BL , Organismos Modificados Genéticamente , Esquizontes/crecimiento & desarrollo , Esquizontes/metabolismo
18.
BMC Biol ; 12: 86, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25359557

RESUMEN

BACKGROUND: Rodent malaria parasites (RMP) are used extensively as models of human malaria. Draft RMP genomes have been published for Plasmodium yoelii, P. berghei ANKA (PbA) and P. chabaudi AS (PcAS). Although availability of these genomes made a significant impact on recent malaria research, these genomes were highly fragmented and were annotated with little manual curation. The fragmented nature of the genomes has hampered genome wide analysis of Plasmodium gene regulation and function. RESULTS: We have greatly improved the genome assemblies of PbA and PcAS, newly sequenced the virulent parasite P. yoelii YM genome, sequenced additional RMP isolates/lines and have characterized genotypic diversity within RMP species. We have produced RNA-seq data and utilised it to improve gene-model prediction and to provide quantitative, genome-wide, data on gene expression. Comparison of the RMP genomes with the genome of the human malaria parasite P. falciparum and RNA-seq mapping permitted gene annotation at base-pair resolution. Full-length chromosomal annotation permitted a comprehensive classification of all subtelomeric multigene families including the 'Plasmodium interspersed repeat genes' (pir). Phylogenetic classification of the pir family, combined with pir expression patterns, indicates functional diversification within this family. CONCLUSIONS: Complete RMP genomes, RNA-seq and genotypic diversity data are excellent and important resources for gene-function and post-genomic analyses and to better interrogate Plasmodium biology. Genotypic diversity between P. chabaudi isolates makes this species an excellent parasite to study genotype-phenotype relationships. The improved classification of multigene families will enhance studies on the role of (variant) exported proteins in virulence and immune evasion/modulation.


Asunto(s)
Expresión Génica , Genoma de Protozoos , Plasmodium falciparum/genética , Plasmodium/clasificación , Secuencia de Bases , Mapeo Cromosómico , Regulación de la Expresión Génica , Genotipo , Datos de Secuencia Molecular , Familia de Multigenes , Plasmodium/genética , Plasmodium falciparum/clasificación , ARN Protozoario/genética , Análisis de Secuencia de ARN , Transcriptoma/genética
19.
Infect Immun ; 82(3): 1277-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24379295

RESUMEN

Plasmodium vivax is the world's most widely distributed malaria parasite and a potential cause of morbidity and mortality for approximately 2.85 billion people living mainly in Southeast Asia and Latin America. Despite this dramatic burden, very few vaccines have been assessed in humans. The clinically relevant vectors modified vaccinia virus Ankara (MVA) and the chimpanzee adenovirus ChAd63 are promising delivery systems for malaria vaccines due to their safety profiles and proven ability to induce protective immune responses against Plasmodium falciparum thrombospondin-related anonymous protein (TRAP) in clinical trials. Here, we describe the development of new recombinant ChAd63 and MVA vectors expressing P. vivax TRAP (PvTRAP) and show their ability to induce high antibody titers and T cell responses in mice. In addition, we report a novel way of assessing the efficacy of new candidate vaccines against P. vivax using a fully infectious transgenic Plasmodium berghei parasite expressing P. vivax TRAP to allow studies of vaccine efficacy and protective mechanisms in rodents. Using this model, we found that both CD8+ T cells and antibodies mediated protection against malaria using virus-vectored vaccines. Our data indicate that ChAd63 and MVA expressing PvTRAP are good preerythrocytic-stage vaccine candidates with potential for future clinical application.


Asunto(s)
Adenoviridae/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/inmunología , Plasmodium berghei/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/genética , Virus Vaccinia/inmunología , Adenoviridae/genética , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Culicidae/inmunología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vacunas contra la Malaria/genética , Malaria Vivax/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Pan troglodytes/inmunología , Pan troglodytes/virología , Plasmodium berghei/genética , Proteínas Protozoarias/inmunología , Vaccinia/genética , Vaccinia/inmunología , Virus Vaccinia/genética
20.
Nat Methods ; 8(12): 1078-82, 2011 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-22020067

RESUMEN

In malaria parasites, the systematic experimental validation of drug and vaccine targets by reverse genetics is constrained by the inefficiency of homologous recombination and by the difficulty of manipulating adenine and thymine (A+T)-rich DNA of most Plasmodium species in Escherichia coli. We overcame these roadblocks by creating a high-integrity library of Plasmodium berghei genomic DNA (>77% A+T content) in a bacteriophage N15-based vector that can be modified efficiently using the lambda Red method of recombineering. We built a pipeline for generating P. berghei genetic modification vectors at genome scale in serial liquid cultures on 96-well plates. Vectors have long homology arms, which increase recombination frequency up to tenfold over conventional designs. The feasibility of efficient genetic modification at scale will stimulate collaborative, genome-wide knockout and tagging programs for P. berghei.


Asunto(s)
ADN Protozoario/genética , ADN Recombinante/genética , Ingeniería Genética , Malaria/parasitología , Plasmodium berghei/genética , Escherichia coli/genética , Biblioteca de Genes , Vectores Genéticos/genética , Genoma de Protozoos/genética , Recombinación Homóloga
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA