Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
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.
Biochem Soc Trans ; 50(3): 1069-1079, 2022 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-35621119

RESUMEN

The ∼30 Mb genomes of the Plasmodium parasites that cause malaria each encode ∼5000 genes, but the functions of the majority remain unknown. This is due to a paucity of functional annotation from sequence homology, which is compounded by low genetic tractability compared with many model organisms. In recent years technical breakthroughs have made forward and reverse genome-scale screens in Plasmodium possible. Furthermore, the adaptation of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and CRISPR-Associated protein 9 (CRISPR/Cas9) technology has dramatically improved gene editing efficiency at the single gene level. Here, we review the arrival of genetic screens in malaria parasites to analyse parasite gene function at a genome-scale and their impact on understanding parasite biology. CRISPR/Cas9 screens, which have revolutionised human and model organism research, have not yet been implemented in malaria parasites due to the need for more complex CRISPR/Cas9 gene targeting vector libraries. We therefore introduce the reader to CRISPR-based screens in the related apicomplexan Toxoplasma gondii and discuss how these approaches could be adapted to develop CRISPR/Cas9 based genome-scale genetic screens in malaria parasites. Moreover, since more than half of Plasmodium genes are required for normal asexual blood-stage reproduction, and cannot be targeted using knockout methods, we discuss how CRISPR/Cas9 could be used to scale up conditional gene knockdown approaches to systematically assign function to essential genes.


Asunto(s)
Parásitos , Plasmodium , Toxoplasma , Animales , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Humanos , Parásitos/genética , Plasmodium/genética , Toxoplasma/genética
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Cell Microbiol ; 17(2): 254-68, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25225164

RESUMEN

The passage through the mosquito is a major bottleneck for malaria parasite populations and a target of interventions aiming to block disease transmission. Here, we used DNA microarrays to profile the developmental transcriptomes of the rodent malaria parasite Plasmodium berghei in vivo, in the midgut of Anopheles gambiae mosquitoes, from parasite stages in the midgut blood bolus to sporulating oocysts on the basal gut wall. Data analysis identified several distinct transcriptional programmes encompassing genes putatively involved in developmental processes or in interactions with the mosquito. At least two of these programmes are associated with the ookinete development that is linked to mosquito midgut invasion and establishment of infection. Targeted disruption by homologous recombination of two of these genes resulted in mutant parasites exhibiting notable infection phenotypes. GAMER encodes a short polypeptide with granular localization in the gametocyte cytoplasm and shows a highly penetrant loss-of-function phenotype manifested as greatly reduced ookinete numbers, linked to impaired male gamete release. HADO encodes a putative magnesium phosphatase with distinctive cortical localization along the concave ookinete periphery. Disruption of HADO compromises ookinete development leading to significant reduction of oocyst numbers. Our data provide important insights into the molecular framework underpinning Plasmodium development in the mosquito and identifies two genes with important functions at initial stages of parasite development in the mosquito midgut.


Asunto(s)
Anopheles/parasitología , Perfilación de la Expresión Génica , Plasmodium berghei/crecimiento & desarrollo , Animales , Tracto Gastrointestinal/parasitología , Malaria/transmisión , Análisis de Secuencia por Matrices de Oligonucleótidos , Plasmodium berghei/genética , Plasmodium berghei/aislamiento & purificación
10.
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
11.
Cell Host Microbe ; 31(2): 305-319.e10, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36634679

RESUMEN

Malaria transmission to mosquitoes requires a developmental switch in asexually dividing blood-stage parasites to sexual reproduction. In Plasmodium berghei, the transcription factor AP2-G is required and sufficient for this switch, but how a particular sex is determined in a haploid parasite remains unknown. Using a global screen of barcoded mutants, we here identify genes essential for the formation of either male or female sexual forms and validate their importance for transmission. High-resolution single-cell transcriptomics of ten mutant parasites portrays the developmental bifurcation and reveals a regulatory cascade of putative gene functions in the determination and subsequent differentiation of each sex. A male-determining gene with a LOTUS/OST-HTH domain as well as the protein interactors of a female-determining zinc-finger protein indicate that germ-granule-like ribonucleoprotein complexes complement transcriptional processes in the regulation of both male and female development of a malaria parasite.


Asunto(s)
Culicidae , Malaria , Parásitos , Animales , Femenino , Masculino , Parásitos/metabolismo , Malaria/parasitología , Plasmodium berghei/genética , Desarrollo Sexual/genética , Culicidae/parasitología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
12.
J Immunol ; 185(9): 5384-91, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20921522

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of respiratory morbidity, resulting in hospitalization for bronchiolitis in some infected infants that is associated with wheeze in later life. Genetic factors are known to affect the severity of the sequelae after RSV infection, but the complexity of the temporal and genetic effects makes it difficult to analyze this response in studies in man. Therefore, we developed a murine genetic model to analyze the sequelae occurring after RSV infection in early life. Haplotype-based genetic analysis of interstrain differences in severity identified the MHC as an important genetic determinant. This was confirmed by analysis of responses in congenic mice with different MHC haplotypes. We also found that susceptible strains had high CD8 levels during secondary infection. Analysis of first filial generation, second filial generation, and back-cross progeny produced by intercrossing resistant (H-2(k), C3H/HeN) and sensitive (H-2(b), BALB/c) strains indicated that susceptibility to sequelae after RSV infection was dominantly inherited but also segregated in a non-MHC-dependent manner. Thus, MHC haplotype and its effect on CD8 cell response is an important determinant of the outcome of neonatal RSV infection.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Complejo Mayor de Histocompatibilidad/genética , Infecciones por Virus Sincitial Respiratorio/genética , Animales , Animales Congénicos , Animales Recién Nacidos/inmunología , Asma/virología , Separación Celular , Mapeo Cromosómico , Modelos Animales de Enfermedad , Citometría de Flujo , Haplotipos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple , Ruidos Respiratorios/etiología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitiales Respiratorios
13.
PLoS Pathog ; 5(8): e1000539, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19662167

RESUMEN

Malaria parasites must undergo sexual and sporogonic development in mosquitoes before they can infect their vertebrate hosts. We report the discovery and characterization of MISFIT, the first protein with paternal effect on the development of the rodent malaria parasite Plasmodium berghei in Anopheles mosquitoes. MISFIT is expressed in male gametocytes and localizes to the nuclei of male gametocytes, zygotes and ookinetes. Gene disruption results in mutant ookinetes with reduced genome content, microneme defects and altered transcriptional profiles of putative cell cycle regulators, which yet successfully invade the mosquito midgut. However, developmental arrest ensues during the ookinete transformation to oocysts leading to malaria transmission blockade. Genetic crosses between misfit mutant parasites and parasites that are either male or female gamete deficient reveal a strict requirement for a male misfit allele. MISFIT belongs to the family of formin-like proteins, which are known regulators of the dynamic remodeling of actin and microtubule networks. Our data identify the ookinete-to-oocyst transition as a critical cell cycle checkpoint in Plasmodium development and lead us to hypothesize that MISFIT may be a regulator of cell cycle progression. This study offers a new perspective for understanding the male contribution to malaria parasite development in the mosquito vector.


Asunto(s)
Culicidae/parasitología , Insectos Vectores/parasitología , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Animales , Southern Blotting , Femenino , Técnica del Anticuerpo Fluorescente , Expresión Génica , Regulación de la Expresión Génica , Genes Protozoarios/genética , Malaria/transmisión , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Filogenia , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Wellcome Open Res ; 5: 71, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32500098

RESUMEN

The rodent parasite Plasmodium chabaudi is an important in vivo model of malaria. The ability to produce chronic infections makes it particularly useful for investigating the development of anti- Plasmodium immunity, as well as features associated with parasite virulence during both the acute and chronic phases of infection. P. chabaudi also undergoes asexual maturation (schizogony) and erythrocyte invasion in culture, so offers an experimentally-amenable in vivo to in vitro model for studying gene function and drug activity during parasite replication. To extend the usefulness of this model, we have further optimised transfection protocols and plasmids for P. chabaudi and generated stable, fluorescent lines that are free from drug-selectable marker genes. These mother-lines show the same infection dynamics as wild-type parasites throughout the lifecycle in mice and mosquitoes; furthermore, their virulence can be increased by serial blood passage and reset by mosquito transmission. We have also adapted the large-insert, linear PlasmoGEM vectors that have revolutionised the scale of experimental genetics in another rodent malaria parasite and used these to generate barcoded P. chabaudi gene-deletion and -tagging vectors for transfection in our fluorescent P. chabaudi mother-lines. This produces a tool-kit of P. chabaudi lines, vectors and transfection approaches that will be of broad utility to the research community.

15.
Mol Microbiol ; 70(1): 209-20, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18761621

RESUMEN

Transmission from the vertebrate host to the mosquito vector represents a major population bottleneck in the malaria life cycle that can successfully be targeted by intervention strategies. However, to date only about 25 parasite proteins expressed during this critical phase have been functionally analysed by gene disruption. We describe the first systematic, larger scale generation and phenotypic analysis of Plasmodium berghei knockout (KO) lines, characterizing 20 genes encoding putatively secreted proteins expressed by the ookinete, the parasite stage responsible for invasion of the mosquito midgut. Of 12 KO lines that were generated, six showed significant reductions in parasite numbers during development in the mosquito, resulting in a block in transmission of five KOs. While expression data, time point of essential function and mutant phenotype correlate well in three KOs defective in midgut invasion, in three KOs that fail at sporulation, maternal inheritance of the mutant phenotype suggests that essential function occurs during ookinete formation and thus precedes morphological abnormalities by several days.


Asunto(s)
Culicidae/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Animales , Expresión Génica , Marcación de Gen , Genes Protozoarios , Malaria/parasitología , Ratones , Fenotipo , Proteómica , Esporozoítos/crecimiento & desarrollo , Transfección
16.
Cell Rep ; 28(6): 1635-1647.e5, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31390575

RESUMEN

Malaria represents a major global health issue, and the identification of new intervention targets remains an urgent priority. This search is hampered by more than one-third of the genes of malaria-causing Plasmodium parasites being uncharacterized. We report a large-scale protein interaction network in Plasmodium schizonts, generated by combining blue native-polyacrylamide electrophoresis with quantitative mass spectrometry and machine learning. This integrative approach, spanning 3 species, identifies >20,000 putative protein interactions, organized into 600 protein clusters. We validate selected interactions, assigning functions in chromatin regulation to previously unannotated proteins and suggesting a role for an EELM2 domain-containing protein and a putative microrchidia protein as mechanistic links between AP2-domain transcription factors and epigenetic regulation. Our interactome represents a high-confidence map of the native organization of core cellular processes in Plasmodium parasites. The network reveals putative functions for uncharacterized proteins, provides mechanistic and structural insight, and uncovers potential alternative therapeutic targets.


Asunto(s)
Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Electroforesis en Gel de Poliacrilamida , Femenino , Ratones , Plasmodium/genética , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Mapas de Interacción de Proteínas , Ratas , Especificidad de la Especie , Espectrometría de Masas en Tándem
17.
Nat Microbiol ; 2: 16276, 2017 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165471

RESUMEN

Malaria is caused by parasites of the genus Plasmodium. All human-infecting Plasmodium species can establish long-lasting chronic infections1-5, creating an infectious reservoir to sustain transmission1,6. It is widely accepted that the maintenance of chronic infection involves evasion of adaptive immunity by antigenic variation7. However, genes involved in this process have been identified in only two of five human-infecting species: Plasmodium falciparum and Plasmodium knowlesi. Furthermore, little is understood about the early events in the establishment of chronic infection in these species. Using a rodent model we demonstrate that from the infecting population, only a minority of parasites, expressing one of several clusters of virulence-associated pir genes, establishes a chronic infection. This process occurs in different species of parasites and in different hosts. Establishment of chronicity is independent of adaptive immunity and therefore different from the mechanism proposed for maintenance of chronic P. falciparum infections7-9. Furthermore, we show that the proportions of parasites expressing different types of pir genes regulate the time taken to establish a chronic infection. Because pir genes are common to most, if not all, species of Plasmodium10, this process may be a common way of regulating the establishment of chronic infections.


Asunto(s)
Malaria/parasitología , Plasmodium/genética , Plasmodium/patogenicidad , Factores de Virulencia/genética , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Enfermedad Crónica , Femenino , Humanos , Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Plasmodium/inmunología , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Plasmodium knowlesi/genética , Plasmodium knowlesi/patogenicidad
18.
Cell Host Microbe ; 17(3): 404-413, 2015 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-25732065

RESUMEN

The genome-wide identification of gene functions in malaria parasites is hampered by a lack of reverse genetic screening methods. We present a large-scale resource of barcoded vectors with long homology arms for effective modification of the Plasmodium berghei genome. Cotransfecting dozens of vectors into the haploid blood stages creates complex pools of barcoded mutants, whose competitive fitness can be measured during infection of a single mouse using barcode sequencing (barseq). To validate the utility of this resource, we rescreen the P. berghei kinome, using published kinome screens for comparison. We find that several protein kinases function redundantly in asexual blood stages and confirm the targetability of kinases cdpk1, gsk3, tkl3, and PBANKA_082960 by genotyping cloned mutants. Thus, parallel phenotyping of barcoded mutants unlocks the power of reverse genetic screening for a malaria parasite and will enable the systematic identification of genes essential for in vivo parasite growth and transmission.


Asunto(s)
Pruebas Genéticas/métodos , Vectores Genéticos , Genética Microbiana/métodos , Plasmodium berghei/genética , Genética Inversa/métodos , Animales , Malaria/parasitología , Malaria/veterinaria , Ratones , Plasmodium berghei/enzimología , Plasmodium berghei/fisiología , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA