Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
EMBO J ; 42(23): e113155, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37886905

RESUMEN

Apicomplexan parasites discharge specialized organelles called rhoptries upon host cell contact to mediate invasion. The events that drive rhoptry discharge are poorly understood, yet essential to sustain the apicomplexan parasitic life cycle. Rhoptry discharge appears to depend on proteins secreted from another set of organelles called micronemes, which vary in function from allowing host cell binding to facilitation of gliding motility. Here we examine the function of the microneme protein CLAMP, which we previously found to be necessary for Toxoplasma gondii host cell invasion, and demonstrate its essential role in rhoptry discharge. CLAMP forms a distinct complex with two other microneme proteins, the invasion-associated SPATR, and a previously uncharacterized protein we name CLAMP-linked invasion protein (CLIP). CLAMP deficiency does not impact parasite adhesion or microneme protein secretion; however, knockdown of any member of the CLAMP complex affects rhoptry discharge. Phylogenetic analysis suggests orthologs of the essential complex components, CLAMP and CLIP, are ubiquitous across apicomplexans. SPATR appears to act as an accessory factor in Toxoplasma, but despite incomplete conservation is also essential for invasion during Plasmodium falciparum blood stages. Together, our results reveal a new protein complex that mediates rhoptry discharge following host-cell contact.


Asunto(s)
Toxoplasma , Toxoplasma/metabolismo , Micronema , Proteínas Protozoarias/metabolismo , Filogenia , Orgánulos/metabolismo
3.
Nat Commun ; 11(1): 4813, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32968076

RESUMEN

Artemisinins have revolutionized the treatment of Plasmodium falciparum malaria; however, resistance threatens to undermine global control efforts. To broadly explore artemisinin susceptibility in apicomplexan parasites, we employ genome-scale CRISPR screens recently developed for Toxoplasma gondii to discover sensitizing and desensitizing mutations. Using a sublethal concentration of dihydroartemisinin (DHA), we uncover the putative transporter Tmem14c whose disruption increases DHA susceptibility. Screens performed under high doses of DHA provide evidence that mitochondrial metabolism can modulate resistance. We show that disrupting a top candidate from the screens, the mitochondrial protease DegP2, lowers porphyrin levels and decreases DHA susceptibility, without significantly altering parasite fitness in culture. Deleting the homologous gene in P. falciparum, PfDegP, similarly lowers heme levels and DHA susceptibility. These results expose the vulnerability of heme metabolism to genetic perturbations that can lead to increased survival in the presence of DHA.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Resistencia a Medicamentos/genética , Pruebas Genéticas/métodos , Hemo/genética , Hemo/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Inactivación de Genes , Humanos , Malaria Falciparum/tratamiento farmacológico , Proteínas de Transporte de Membrana/metabolismo , Mutación , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Toxoplasma/efectos de los fármacos , Toxoplasma/genética
4.
ACS Chem Biol ; 15(7): 1801-1807, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32597628

RESUMEN

Apicomplexan parasites include the causative agents of malaria and toxoplasmosis. Cell-based screens in Toxoplasma previously identified a chemical modulator of calcium signaling (ENH1) that blocked parasite egress from host cells and exhibited potent antiparasitic activity. To identify the targets of ENH1, we adapted thermal proteome profiling to Toxoplasma, which revealed calcium-dependent protein kinase 1 (CDPK1) as a target. We confirmed the inhibition of CDPK1 by ENH1 in vitro and in parasites by comparing alleles sensitive or resistant to ENH1. CDPK1 inhibition explained the block in egress; however, the effects of ENH1 on calcium homeostasis and parasite viability were CDPK1-independent, implicating additional targets. Thermal proteome profiling of lysates from parasites expressing the resistant allele of CDPK1 identified additional candidates associated with the mitochondria and the parasite pellicle-compartments that potentially function in calcium release and homeostasis. Our findings illustrate the promise of thermal profiling to identify druggable targets that modulate calcium signaling in apicomplexan parasites.


Asunto(s)
Antiprotozoarios/farmacología , Imidazoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Toxoplasma/efectos de los fármacos , Antiprotozoarios/metabolismo , Señalización del Calcio/efectos de los fármacos , Imidazoles/metabolismo , Pruebas de Sensibilidad Parasitaria , Unión Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Proteoma/metabolismo , Proteómica , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/metabolismo , Piridinas/metabolismo
5.
Cell Host Microbe ; 26(4): 478-492.e8, 2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600500

RESUMEN

Toxoplasma can reach distant organs, especially the brain, leading to a lifelong chronic phase. However, genes involved in related in vivo processes are currently unknown. Here, we use focused CRISPR libraries to identify Toxoplasma genes that affect in vivo fitness. We focus on TgWIP, whose deletion affects Toxoplasma dissemination to distant organs. We show that TgWIP is secreted into the host cell upon invasion and interacts with the host WAVE regulatory complex and SHP2 phosphatase, both of which regulate actin dynamics. TgWIP affects the morphology of dendritic cells and mediates the dissolution of podosomes, which dendritic cells use to adhere to extracellular matrix. TgWIP enhances the motility and transmigration of parasitized dendritic cells, likely explaining its effect on in vivo fitness. Our results provide a framework for systemic identification of Toxoplasma genes with in vivo effects at the site of infection or on dissemination to distant organs, including the brain.


Asunto(s)
Movimiento Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Células Dendríticas/fisiología , Proteínas Protozoarias/genética , Toxoplasma/genética , Animales , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Toxoplasma/patogenicidad , Factores de Virulencia/genética , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo
6.
Nat Protoc ; 13(1): 307-323, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29323662

RESUMEN

Apicomplexan parasites, such as Toxoplasma gondii, cause extensive morbidity and mortality in humans and livestock, highlighting the need for a deeper understanding of their molecular biology. Although techniques for the generation of targeted gene disruptions have long been available for apicomplexans, such methods are not readily scalable to the entire genome. We recently used CRISPR-Cas9 to disrupt all nuclear protein-coding genes in T. gondii using a pooled format. The method relies on transfection of a guide RNA library into parasites constitutively expressing Cas9. Here, we present the complete workflow of such a screen, including preparation of the guide RNA library, growth and testing of the recipient strain, generation of the mutant population, culture conditions for the screen, preparation of genomic DNA libraries, next-generation sequencing of the guide RNA loci, and analysis to detect fitness-conferring genes. This method can be deployed to study how culture conditions affect the repertoire of genes needed by parasites, which will enable studies of their metabolic needs, host specificity, and drug-resistance mechanisms. In addition, by manipulating the background in which the screen is performed, researchers will be able to investigate genetic interactions, which may help uncover redundancy or epistasis in the parasite genome. Using this method, a genome-wide screen and its analysis can be completed in 3 weeks, after ∼1 month of preparation to generate the library and grow the cells needed, making it a powerful tool for uncovering functionally important genes in apicomplexan parasites.


Asunto(s)
Mapeo Cromosómico/métodos , Técnicas de Inactivación de Genes/métodos , Toxoplasma/genética , Sistemas CRISPR-Cas/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Genoma de Protozoos , Biblioteca Genómica , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , ARN Guía de Kinetoplastida , Transfección
7.
Cell ; 166(6): 1423-1435.e12, 2016 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-27594426

RESUMEN

Apicomplexan parasites are leading causes of human and livestock diseases such as malaria and toxoplasmosis, yet most of their genes remain uncharacterized. Here, we present the first genome-wide genetic screen of an apicomplexan. We adapted CRISPR/Cas9 to assess the contribution of each gene from the parasite Toxoplasma gondii during infection of human fibroblasts. Our analysis defines ∼200 previously uncharacterized, fitness-conferring genes unique to the phylum, from which 16 were investigated, revealing essential functions during infection of human cells. Secondary screens identify as an invasion factor the claudin-like apicomplexan microneme protein (CLAMP), which resembles mammalian tight-junction proteins and localizes to secretory organelles, making it critical to the initiation of infection. CLAMP is present throughout sequenced apicomplexan genomes and is essential during the asexual stages of the malaria parasite Plasmodium falciparum. These results provide broad-based functional information on T. gondii genes and will facilitate future approaches to expand the horizon of antiparasitic interventions.


Asunto(s)
Apicomplexa/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Estudio de Asociación del Genoma Completo , Interacciones Huésped-Parásitos , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Células Cultivadas , Claudinas/genética , Claudinas/metabolismo , Fibroblastos/parasitología , Genoma de Protozoos/genética , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/fisiopatología , Plasmodium falciparum/genética , Toxoplasmosis/parasitología , Toxoplasmosis/fisiopatología
8.
Proc Natl Acad Sci U S A ; 113(29): E4133-42, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27382155

RESUMEN

Vaccines have had broad medical impact, but existing vaccine technologies and production methods are limited in their ability to respond rapidly to evolving and emerging pathogens, or sudden outbreaks. Here, we develop a rapid-response, fully synthetic, single-dose, adjuvant-free dendrimer nanoparticle vaccine platform wherein antigens are encoded by encapsulated mRNA replicons. To our knowledge, this system is the first capable of generating protective immunity against a broad spectrum of lethal pathogen challenges, including H1N1 influenza, Toxoplasma gondii, and Ebola virus. The vaccine can be formed with multiple antigen-expressing replicons, and is capable of eliciting both CD8(+) T-cell and antibody responses. The ability to generate viable, contaminant-free vaccines within days, to single or multiple antigens, may have broad utility for a range of diseases.


Asunto(s)
Dendrímeros/uso terapéutico , Nanopartículas/uso terapéutico , ARN/uso terapéutico , Vacunas , Animales , Línea Celular , Ebolavirus/efectos de los fármacos , Femenino , Células HeLa , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Subtipo H1N1 del Virus de la Influenza A/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/prevención & control , Ratas , Linfocitos T/inmunología , Toxoplasma/efectos de los fármacos , Toxoplasmosis/prevención & control
9.
J Biol Chem ; 291(18): 9566-80, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-26933036

RESUMEN

The life cycles of apicomplexan parasites progress in accordance with fluxes in cytosolic Ca(2+) Such fluxes are necessary for events like motility and egress from host cells. We used genetically encoded Ca(2+) indicators (GCaMPs) to develop a cell-based phenotypic screen for compounds that modulate Ca(2+) signaling in the model apicomplexan Toxoplasma gondii In doing so, we took advantage of the phosphodiesterase inhibitor zaprinast, which we show acts in part through cGMP-dependent protein kinase (protein kinase G; PKG) to raise levels of cytosolic Ca(2+) We define the pool of Ca(2+) regulated by PKG to be a neutral store distinct from the endoplasmic reticulum. Screening a library of 823 ATP mimetics, we identify both inhibitors and enhancers of Ca(2+) signaling. Two such compounds constitute novel PKG inhibitors and prevent zaprinast from increasing cytosolic Ca(2+) The enhancers identified are capable of releasing intracellular Ca(2+) stores independently of zaprinast or PKG. One of these enhancers blocks parasite egress and invasion and shows strong antiparasitic activity against T. gondii The same compound inhibits invasion of the most lethal malaria parasite, Plasmodium falciparum Inhibition of Ca(2+)-related phenotypes in these two apicomplexan parasites suggests that depletion of intracellular Ca(2+) stores by the enhancer may be an effective antiparasitic strategy. These results establish a powerful new strategy for identifying compounds that modulate the essential parasite signaling pathways regulated by Ca(2+), underscoring the importance of these pathways and the therapeutic potential of their inhibition.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico , Retículo Endoplásmico , Proteínas Protozoarias , Purinonas/farmacología , Toxoplasma , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Toxoplasma/metabolismo
10.
Cell Host Microbe ; 17(5): 642-52, 2015 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-25974303

RESUMEN

Toxoplasma gondii is a protozoan pathogen in the phylum Apicomplexa that resides within an intracellular parasitophorous vacuole (PV) that is selectively permeable to small molecules through unidentified mechanisms. We have identified GRA17 as a Toxoplasma-secreted protein that localizes to the parasitophorous vacuole membrane (PVM) and mediates passive transport of small molecules across the PVM. GRA17 is related to the putative Plasmodium translocon protein EXP2 and conserved across PV-residing Apicomplexa. The PVs of GRA17-deficient parasites have aberrant morphology, reduced permeability to small molecules, and structural instability. GRA17-deficient parasites proliferate slowly and are avirulent in mice. These GRA17-deficient phenotypes are rescued by complementation with Plasmodium EXP2. GRA17 functions synergistically with a related protein, GRA23. Exogenous expression of GRA17 or GRA23 alters the membrane conductance properties of Xenopus oocytes in a manner consistent with a large non-selective pore. Thus, GRA17 and GRA23 provide a molecular basis for PVM permeability and nutrient access.


Asunto(s)
Antígenos de Protozoos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Toxoplasma/fisiología , Vacuolas/parasitología , Factores de Virulencia/metabolismo , Animales , Antígenos de Protozoos/genética , Transporte Biológico , Eliminación de Gen , Prueba de Complementación Genética , Proteínas de Transporte de Membrana/genética , Ratones , Toxoplasma/crecimiento & desarrollo , Toxoplasma/metabolismo , Toxoplasmosis Animal/parasitología , Toxoplasmosis Animal/patología , Virulencia , Factores de Virulencia/genética , Xenopus
11.
PLoS One ; 10(4): e0122585, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25848798

RESUMEN

Shigellosis is a severe diarrheal disease that affects hundreds of thousands of individuals resulting in significant morbidity and mortality worldwide. Shigellosis is caused by Shigella spp., a gram-negative bacterium that uses a Type 3 Secretion System (T3SS) to deliver effector proteins into the cytosol of infected human cells. Shigella infection triggers multiple signaling programs that result in a robust host transcriptional response that includes the induction of multiple proinflammatory cytokines. PML nuclear bodies (PML-NBs) are dynamic subnuclear structures that coordinate immune signaling programs and have a demonstrated role in controlling viral infection. We show that PML-NB number increases upon Shigella infection. We examined the effects of Shigella infection on SUMOylation and found that upon Shigella infection the localization of SUMOylated proteins is altered and the level of SUMOylated proteins decreases. Although Shigella infection does not alter the abundance of SUMO activating enzymes SAE1 or SAE2, it dramatically decreases the level of the SUMO conjugating enzyme Ubc9. All Shigella-induced alterations to the SUMOylation system are dependent upon a T3SS. Thus, we demonstrate that Shigella uses one or more T3SS effectors to influence both PML-NB number and the SUMOylation machinery in human cells.


Asunto(s)
Estructuras del Núcleo Celular/metabolismo , Estructuras del Núcleo Celular/microbiología , Shigella flexneri/fisiología , Sumoilación , Estructuras del Núcleo Celular/inmunología , Células HeLa , Humanos , Transporte de Proteínas , Proteína SUMO-1/metabolismo , Transducción de Señal
12.
PLoS One ; 9(6): e100450, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24971596

RESUMEN

Toxoplasma gondii is a parasite of humans and animals, and a model for other apicomplexans including Plasmodium spp., the causative agents of malaria. Despite many advances, manipulating the T. gondii genome remains labor intensive, and is often restricted to lab-adapted strains or lines carrying mutations that enable selection. Here, we use the RNA-guided Cas9 nuclease to efficiently generate knockouts without selection, and to introduce point mutations and epitope tags into the T. gondii genome. These methods will streamline the functional analysis of parasite genes and enable high-throughput engineering of their genomes.


Asunto(s)
Proteínas Asociadas a CRISPR , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ingeniería Genética , Toxoplasma/genética , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Secuencia de Bases , Sistemas CRISPR-Cas , Línea Celular , Reparación del ADN por Unión de Extremidades , Orden Génico , Marcación de Gen/métodos , Sitios Genéticos , Genoma de Protozoos , Humanos , Datos de Secuencia Molecular , Plásmidos/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...