Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 20(6): e1011162, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38885280

RESUMEN

Very little is known about the process of meiosis in the apicomplexan parasite Cryptosporidium despite the essentiality of sex in its life cycle. Most cell lines only support asexual growth of Cryptosporidium parvum (C. parvum), but stem cell derived intestinal epithelial cells grown under air-liquid interface (ALI) conditions support the sexual cycle. To examine chromosomal dynamics during meiosis in C. parvum, we generated two transgenic lines of parasites that were fluorescently tagged with mCherry or GFP on chromosomes 1 or 5, respectively. Infection of ALI cultures or Ifngr1-/- mice with mCherry and GFP parasites resulted in cross-fertilization and the formation of "yellow" oocysts, which contain 4 haploid sporozoites that are the product of meiosis. Recombinant oocysts from the F1 generation were purified and used to infect HCT-8 cultures, and phenotypes of the progeny were observed by microscopy. All possible phenotypes predicted by independent segregation were represented equally (~25%) in the population, indicating that C. parvum chromosomes exhibit a Mendelian inheritance pattern. The most common pattern observed from the outgrowth of single oocysts included all possible parental and recombinant phenotypes derived from a single meiotic event, suggesting a high rate of crossover. To estimate the frequency of crossover, additional loci on chromosomes 1 and 5 were tagged and used to monitor intrachromosomal crosses in Ifngr1-/- mice. Both chromosomes showed a high frequency of crossover compared to other apicomplexans with map distances (i.e., 1% recombination) of 3-12 kb. Overall, a high recombination rate may explain many unique characteristics observed in Cryptosporidium spp. such as high rates of speciation, wide variation in host range, and rapid evolution of host-specific virulence factors.


Asunto(s)
Criptosporidiosis , Cryptosporidium parvum , Meiosis , Oocistos , Recombinación Genética , Animales , Cryptosporidium parvum/genética , Ratones , Criptosporidiosis/parasitología , Criptosporidiosis/genética , Meiosis/genética , Humanos , Receptores de Interferón/genética , Receptor de Interferón gamma , Segregación Cromosómica/genética , Esporozoítos/genética , Ratones Noqueados , Fenotipo
2.
Genomics ; 116(2): 110792, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38215860

RESUMEN

Eimeria tenella is the main pathogen responsible for coccidiosis in chickens. The life cycle of E. tenella is, arguably, the least complex of all Coccidia, with only one host. However, it presents different developmental stages, either in the environment or in the host and either intracellular or extracellular. Its signaling and metabolic pathways change with its different developmental stages. Until now, little is known about the developmental regulation and transformation mechanisms of its life cycle. In this study, protein profiles from the five developmental stages, including unsporulated oocysts (USO), partially sporulated (7 h) oocysts (SO7h), sporulated oocysts (SO), sporozoites (S) and second-generation merozoites (M2), were harvested using the label-free quantitative proteomics approach. Then the differentially expressed proteins (DEPs) for these stages were identified. A total of 314, 432, 689, and 665 DEPs were identified from the comparison of SO7h vs USO, SO vs SO7h, S vs SO, and M2 vs S, respectively. By conducting weighted gene coexpression network analysis (WGCNA), six modules were dissected. Proteins in blue and brown modules were calculated to be significantly positively correlated with the E. tenella developmental stages of sporozoites (S) and second-generation merozoites (M2), respectively. In addition, hub proteins with high intra-module degree were identified. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway enrichment analyses revealed that hub proteins in blue modules were involved in electron transport chain and oxidative phosphorylation. Hub proteins in the brown module were involved in RNA splicing. These findings provide new clues and ideas to enhance our fundamental understanding of the molecular mechanisms underlying parasite development.


Asunto(s)
Eimeria tenella , Animales , Eimeria tenella/genética , Proteómica , Pollos/parasitología , Oocistos/fisiología , Esporozoítos/genética , Esporozoítos/metabolismo , Estadios del Ciclo de Vida
3.
J Biol Chem ; 298(9): 102241, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35809642

RESUMEN

Malaria and other apicomplexan-caused diseases affect millions of humans, agricultural animals, and pets. Cell traversal is a common feature used by multiple apicomplexan parasites to migrate through host cells and can be exploited to develop therapeutics against these deadly parasites. Here, we provide insights into the mechanism of the Cell-traversal protein for ookinetes and sporozoites (CelTOS), a conserved cell-traversal protein in apicomplexan parasites and malaria vaccine candidate. CelTOS has previously been shown to form pores in cell membranes to enable traversal of parasites through cells. We establish roles for the distinct protein regions of Plasmodium vivax CelTOS and examine the mechanism of pore formation. We further demonstrate that CelTOS dimer dissociation is required for pore formation, as disulfide bridging between monomers inhibits pore formation, and this inhibition is rescued by disulfide-bridge reduction. We also show that a helix-destabilizing amino acid, Pro127, allows CelTOS to undergo significant conformational changes to assemble into pores. The flexible C terminus of CelTOS is a negative regulator that limits pore formation. Finally, we highlight that lipid binding is a prerequisite for pore assembly as mutation of a phospholipids-binding site in CelTOS resulted in loss of lipid binding and abrogated pore formation. These findings identify critical regions in CelTOS and will aid in understanding the egress mechanism of malaria and other apicomplexan parasites as well as have implications for studying the function of other essential pore-forming proteins.


Asunto(s)
Vacunas contra la Malaria , Malaria Vivax , Plasmodium vivax , Proteínas Protozoarias , Sitios de Unión , Disulfuros/química , Humanos , Vacunas contra la Malaria/química , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Fosfolípidos/inmunología , Plasmodium vivax/genética , Plasmodium vivax/inmunología , Prolina/química , Prolina/genética , Conformación Proteica en Hélice alfa , Multimerización de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Esporozoítos/genética , Esporozoítos/inmunología
4.
EMBO J ; 38(15): e100984, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31368598

RESUMEN

Microtubules are cytoskeletal filaments essential for many cellular processes, including establishment and maintenance of polarity, intracellular transport, division and migration. In most metazoan cells, the number and length of microtubules are highly variable, while they can be precisely defined in some protozoan organisms. However, in either case the significance of these two key parameters for cells is not known. Here, we quantitatively studied the impact of modulating microtubule number and length in Plasmodium, the protozoan parasite causing malaria. Using a gene deletion and replacement strategy targeting one out of two α-tubulin genes, we show that chromosome segregation proceeds in the oocysts even in the absence of microtubules. However, fewer and shorter microtubules severely impaired the formation, motility and infectivity of Plasmodium sporozoites, the forms transmitted by the mosquito, which usually contain 16 microtubules. We found that α-tubulin expression levels directly determined the number of microtubules, suggesting a high nucleation barrier as supported by a mathematical model. Infectious sporozoites were only formed in parasite lines featuring at least 10 microtubules, while parasites with 9 or fewer microtubules failed to transmit.


Asunto(s)
Malaria/parasitología , Plasmodium/patogenicidad , Tubulina (Proteína)/genética , Animales , Eliminación de Gen , Ratones , Modelos Teóricos , Plasmodium/genética , Plasmodium/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo , Esporozoítos/patogenicidad , Tubulina (Proteína)/metabolismo
5.
Mol Microbiol ; 115(5): 870-881, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33191548

RESUMEN

Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.


Asunto(s)
Hepatocitos/parasitología , Malaria/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Humanos , Hígado/parasitología , Plasmodium/genética , Plasmodium/crecimiento & desarrollo , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo
6.
Nucleic Acids Res ; 48(5): 2303-2311, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32034421

RESUMEN

Chromatin conformation assays such as Hi-C cannot directly measure differences in 3D architecture between cell types or cell states. For this purpose, two or more Hi-C experiments must be carried out, but direct comparison of the resulting Hi-C matrices is confounded by several features of Hi-C data. Most notably, the genomic distance effect, whereby contacts between pairs of genomic loci that are proximal along the chromosome exhibit many more Hi-C contacts that distal pairs of loci, dominates every Hi-C matrix. Furthermore, the form that this distance effect takes often varies between different Hi-C experiments, even between replicate experiments. Thus, a statistical confidence measure designed to identify differential Hi-C contacts must accurately account for the genomic distance effect or risk being misled by large-scale but artifactual differences. ACCOST (Altered Chromatin COnformation STatistics) accomplishes this goal by extending the statistical model employed by DEseq, re-purposing the 'size factors,' which were originally developed to account for differences in read depth between samples, to instead model the genomic distance effect. We show via analysis of simulated and real data that ACCOST provides unbiased statistical confidence estimates that compare favorably with competing methods such as diffHiC, FIND and HiCcompare. ACCOST is freely available with an Apache license at https://bitbucket.org/noblelab/accost.


Asunto(s)
Cromatina/química , ADN/química , Sitios Genéticos , Genoma , Programas Informáticos , Animales , Línea Celular , Cromatina/metabolismo , ADN/metabolismo , Epistasis Genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Ratones , Conformación Molecular , Plasmodium falciparum/genética , Esporozoítos/genética , Trofozoítos/genética
7.
Malar J ; 20(1): 430, 2021 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-34717635

RESUMEN

BACKGROUND: Plasmodium sporozoites are the highly motile forms of malaria-causing parasites that are transmitted by the mosquito to the vertebrate host. Sporozoites need to enter and cross several cellular and tissue barriers for which they employ a set of surface proteins. Three of these proteins are members of the thrombospondin related anonymous protein (TRAP) family. Here, potential additive, synergistic or antagonistic roles of these adhesion proteins were investigated. METHODS: Four transgenic Plasmodium berghei parasite lines that lacked two or all three of the TRAP family adhesins TRAP, TLP and TREP were generated using positive-negative selection. The parasite lines were investigated for their capacity to attach to and move on glass, their ability to egress from oocysts and their capacity to enter mosquito salivary glands. One strain was in addition interrogated for its capacity to infect mice. RESULTS: The major phenotype of the TRAP single gene deletion dominates additional gene deletion phenotypes. All parasite lines including the one lacking all three proteins were able to conduct some form of active, if unproductive movement. CONCLUSIONS: The individual TRAP-family adhesins appear to play functionally distinct roles during motility and infection. Other proteins must contribute to substrate adhesion and gliding motility.


Asunto(s)
Plasmodium berghei/fisiología , Proteínas Protozoarias/genética , Esporozoítos/fisiología , Microorganismos Modificados Genéticamente/genética , Microorganismos Modificados Genéticamente/fisiología , Plasmodium berghei/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/genética
8.
Malar J ; 20(1): 308, 2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34243763

RESUMEN

BACKGROUND: Vaccination with radiation-attenuated Plasmodium falciparum sporozoites is known to induce protective immunity. However, the mechanisms underlying this protection remain unclear. In this work, two recent radiation-attenuated sporozoite vaccination studies were used to identify potential transcriptional correlates of vaccination-induced protection. METHODS: Longitudinal whole blood RNAseq transcriptome responses to immunization with radiation-attenuated P. falciparum sporozoites were analysed and compared across malaria-naïve adult participants (IMRAS) and malaria-experienced adult participants (BSPZV1). Parasite dose and method of delivery differed between trials, and immunization regimens were designed to achieve incomplete protective efficacy. Observed protective efficacy was 55% in IMRAS and 20% in BSPZV1. Study vaccine dosings were chosen to elicit both protected and non-protected subjects, so that protection-associated responses could be identified. RESULTS: Analysis of comparable time points up to 1 week after the first vaccination revealed a shared cross-study transcriptional response programme, despite large differences in number and magnitude of differentially expressed genes between trials. A time-dependent regulatory programme of coherent blood transcriptional modular responses was observed, involving induction of inflammatory responses 1-3 days post-vaccination, with cell cycle responses apparent by day 7 in protected individuals from both trials. Additionally, strongly increased induction of inflammation and interferon-associated responses was seen in non-protected IMRAS participants. All individuals, except for non-protected BSPZV1 participants, showed robust upregulation of cell-cycle associated transcriptional responses post vaccination. CONCLUSIONS: In summary, despite stark differences between the two studies, including route of vaccination and status of malaria exposure, responses were identified that were associated with protection after PfRAS vaccination. These comprised a moderate early interferon response peaking 2 days post vaccination, followed by a later proliferative cell cycle response steadily increasing over the first 7 days post vaccination. Non-protection is associated with deviations from this model, observed in this study with over-induction of early interferon responses in IMRAS and failure to mount a cell cycle response in BSPZV1.


Asunto(s)
Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/prevención & control , Anticuerpos Antiprotozoarios/sangre , Ensayos Clínicos como Asunto , Humanos , Vacunas contra la Malaria/administración & dosificación , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Proteínas Protozoarias/genética , Esporozoítos/genética , Esporozoítos/inmunología , Transcripción Genética , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/uso terapéutico
9.
Exp Parasitol ; 220: 108034, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33188795

RESUMEN

Eimeria ninakohlyakimovae represents a highly pathogenic coccidian parasite causing severe haemorrhagic typhlocolitis in goat kids worldwide. NETosis was recently described as an efficient defense mechanism of polymorphonuclear neutrophils (PMN) acting against different parasites in vitro and in vivo. In vitro interactions of caprine PMN with parasitic stages of E. ninakohlyakimovae (i. e. oocysts and sporozoites) as well as soluble oocyst antigens (SOA) were analyzed at different ratios, concentrations and time spans. Extracellular DNA staining was used to illustrate classical molecules induced during caprine NETosis [i. e. histones (H3) and neutrophil elastase (NE)] via antibody-based immunofluorescence analyses. Functional inhibitor treatments with DPI and DNase I were applied to unveil role of NADPH oxidase (NOX) and characterize DNA-backbone composition of E. ninakohlyakimovae-triggered caprine NETosis. Scanning electron microscopy (SEM)- and immunofluorescence-analyses demonstrated that caprine PMN underwent NETosis upon contact with sporozoites and oocysts of E. ninakohlyakimovae, ensnaring filaments which firmly entrapped parasites. Detailed co-localization studies of E. ninakohlyakimovae-induced caprine NETosis revealed presence of PMN-derived DNA being adorned with nuclear H3 and NE corroborating molecular characteristics of NETosis. E. ninakohlyakoimovae-induced caprine NETosis was found to be NOX-independent since DPI inhibition led to a slight decrease of NETosis. Exposure of caprine PMN to vital E. ninakohlyakimovae sporozoites as well as SOA resulted in up-regulation of IL-12, TNF-α, IL-6, CCL2 and iNOS gene transcription in stimulated PMN. Since vital E. ninakohlyakimovae-sporozoites induced caprine NETosis, this effective entrapment mechanism might reduce initial sporozoite epithelial host cell invasion during goat coccidiosis ultimately resulting in less macromeront formation and reduced merozoites I production.


Asunto(s)
Coccidiosis/veterinaria , Citocinas/genética , Eimeria/inmunología , Enfermedades de las Cabras/parasitología , Neutrófilos/parasitología , Análisis de Varianza , Animales , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Coccidiosis/inmunología , Coccidiosis/parasitología , Colitis/parasitología , Colitis/veterinaria , Citocinas/metabolismo , Eimeria/genética , Eimeria/ultraestructura , Hemorragia Gastrointestinal/parasitología , Hemorragia Gastrointestinal/veterinaria , Enfermedades de las Cabras/inmunología , Cabras , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Microscopía Electrónica de Rastreo/veterinaria , NADPH Oxidasas/metabolismo , Neutrófilos/inmunología , Neutrófilos/ultraestructura , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oocistos/genética , Oocistos/inmunología , Reacción en Cadena de la Polimerasa/veterinaria , Esporozoítos/genética , Esporozoítos/inmunología , Transcripción Genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Tiflitis/parasitología , Tiflitis/veterinaria , Regulación hacia Arriba
10.
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
11.
Parasitol Res ; 120(7): 2689-2693, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34146126

RESUMEN

Poultry coccidiosis causes considerable economical losses to the livestock industry. Eimeria parasites are responsible for this disease. On a global scale, E. acervulina and E. tenella are amongst the most common Eimeria spp. infecting broilers. E. tenella is commonly used as infection model in in vivo and in vitro studies. On the other hand, E. acervulina has barely been studied under in vitro conditions. A well established and widely used in vitro model for E. tenella infection is the Madin-Darby bovine kidney cell line (MDBK); however, little is known regarding suitability of MDBK cells as host cells for E. acervulina. We infected MDBK monolayers with two different doses, 5 × 104 and 2 × 105, of E. acervulina sporozoites and evaluated cultures at 24 and 96 h post infection (hpi). For comparison, we ran an identical infection assay using E. tenella sporozoites. To assess parasite reproduction, the number of DNA copies of E. acervulina SCAR marker and E. tenella ITS-1 gene was quantified using real-time quantitative PCR. We found that the number of E. acervulina copies increased significantly at 24 hpi in comparison to E. tenella (p < 0.05). After 96 hpi, E. acervulina gene copies were considerably reduced while E. tenella continued to multiply (p < 0.05). Our results show that MDBK monolayers could be used for in vitro research aimed to study E. acervulina sporozoite cell invasion. Nevertheless, modifications of in vitro cultivation appear necessary to allow qualitative and quantitative studies over longer periods of parasite reproduction.


Asunto(s)
Eimeria/fisiología , Riñón/parasitología , Animales , Bovinos , Línea Celular , Pollos/parasitología , Coccidiosis/parasitología , Coccidiosis/veterinaria , Eimeria/clasificación , Eimeria/genética , Eimeria tenella/genética , Eimeria tenella/fisiología , Células Epiteliales , Riñón/citología , Enfermedades de las Aves de Corral/parasitología , Reacción en Cadena en Tiempo Real de la Polimerasa , Esporozoítos/clasificación , Esporozoítos/genética , Esporozoítos/fisiología
12.
Cell Microbiol ; 21(7): e13027, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30941872

RESUMEN

Coccidia are obligate intracellular protozoan parasites responsible for human and veterinary diseases. Eimeria tenella, the aetiologic agent of caecal coccidiosis, is a major pathogen of chickens. In Toxoplasma gondii, some kinases from the rhoptry compartment (ROP) are key virulence factors. ROP kinases hijack and modulate many cellular functions and pathways, allowing T. gondii survival and development. E. tenella's kinome comprises 28 putative members of the ROP kinase family; most of them are predicted, as pseudokinases and their functions have never been characterised. One of the predicted kinase, EtROP1, was identified in the rhoptry proteome of E. tenella sporozoites. Here, we demonstrated that EtROP1 is active, and the N-terminal extension is necessary for its catalytic kinase activity. Ectopic expression of EtROP1 followed by co-immunoprecipitation identified cellular p53 as EtROP1 partner. Further characterisation confirmed the interaction and the phosphorylation of p53 by EtROP1. E. tenella infection or overexpression of EtROP1 resulted both in inhibition of host cell apoptosis and G0/G1 cell cycle arrest. This work functionally described the first ROP kinase from E. tenella and its noncanonical structure. Our study provides the first mechanistic insight into host cell apoptosis inhibition by E. tenella. EtROP1 appears as a new candidate for coccidiosis control.


Asunto(s)
Coccidiosis/genética , Eimeria tenella/genética , Proteínas de la Membrana/genética , Proteínas Protozoarias/genética , Animales , Apoptosis/genética , Pollos/parasitología , Coccidiosis/parasitología , Eimeria tenella/patogenicidad , Puntos de Control de la Fase G1 del Ciclo Celular , Fosfotransferasas/genética , Proteoma/genética , Esporozoítos/genética , Esporozoítos/patogenicidad , Toxoplasma/genética , Toxoplasma/patogenicidad , Factores de Virulencia/genética
13.
Parasitol Res ; 119(11): 3817-3828, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33009946

RESUMEN

Buffalo-derived Theileria parva can 'break through' the immunity induced by the infection and treatment vaccination method (ITM) in cattle. However, no such 'breakthroughs' have been reported in northern Tanzania where there has been long and widespread ITM use in pastoralist cattle, and the Cape buffalo (Syncerus caffer) is also present. We studied the exposure of vaccinated and unvaccinated cattle in northern Tanzania to buffalo-derived T. parva using p67 gene polymorphisms and compared this to its distribution in vaccinated cattle exposed to buffalo-derived T. parva in central Kenya, where vaccine 'breakthroughs' have been reported. Additionally, we analysed the CD8+ T cell target antigen Tp2 for positive selection. Our results showed that 10% of the p67 sequences from Tanzanian cattle (n = 39) had a buffalo type p67 (allele 4), an allele that is rare among East African isolates studied so far. The percentage of buffalo-derived p67 alleles observed in Kenyan cattle comprised 19% of the parasites (n = 36), with two different p67 alleles (2 and 3) of presumptive buffalo origin. The Tp2 protein was generally conserved with only three Tp2 variants from Tanzania (n = 33) and five from Kenya (n = 40). Two Tanzanian Tp2 variants and two Kenyan Tp2 variants were identical to variants present in the trivalent Muguga vaccine. Tp2 evolutionary analysis did not show evidence for positive selection within previously mapped epitope coding sites. The p67 data indicates that some ITM-vaccinated cattle are protected against disease induced by a buffalo-derived T. parva challenge in northern Tanzania and suggests that the parasite genotype may represent one factor explaining this.


Asunto(s)
Antígenos de Superficie/genética , Búfalos/parasitología , Theileria parva/genética , Theileriosis/parasitología , Alelos , Animales , Animales Salvajes/parasitología , Bovinos , Genotipo , Especificidad del Huésped , Kenia , Ganado/parasitología , Polimorfismo Genético/genética , Esporozoítos/genética , Tanzanía , Theileria parva/clasificación , Theileriosis/transmisión , Vacunación/veterinaria
14.
Malar J ; 18(1): 330, 2019 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-31551073

RESUMEN

BACKGROUND: The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS: In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS: The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS: The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.


Asunto(s)
Perfilación de la Expresión Génica , Hepatocitos/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Eritrocitos/parasitología , Regulación de la Expresión Génica , Genoma de Protozoos , Humanos , Estadios del Ciclo de Vida , Hígado/parasitología , Malaria/parasitología , Merozoítos/genética , Merozoítos/crecimiento & desarrollo , Regiones Promotoras Genéticas , RNA-Seq , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo
15.
Parasitology ; 146(13): 1690-1698, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31391126

RESUMEN

Apicomplexan haemoparasites of the genera Schellackia Reichenow, 1919, and Karyolysus Labbé, 1894, seem to be common in lizards and widespread across the world. For decades, their identification has been based on morphological descriptions and life cycle patterns, with molecular characterizations, applied only recently. We used molecular characterization to confirm the identification of haemoparasites detected by microscopy in blood smears of Lacerta schreiberi Bedriaga, 1878, a lizard of the Iberian Peninsula. Since blood samples other than blood smears were not available from the studied lizards, 264 engorged ticks Ixodes ricinus (Linneaus, 1758) collected from them were used as an alternative non-invasive source of haemoparasite DNA for molecular genetic analyses. Of the 48 blood smears microscopically examined, 31 were positive for blood parasites (64.6% prevalence). We identified trophozoites and gamonts similar to Karyolysus lacazei (Labbé, 1894) (24/48; 50%) and Schellackia-like sporozoites (20/48; 41.7%). Mixed infections with both species occurred in 13 blood smears (27.1%). Sequence data were obtained for both parasites from engorged ticks. Phylogenetic analyses placed our unique haemogregarine sequence within the Karyolysus clade, nevertheless, within substantial polytomy. Thus, according to its morphology and effect on the host cell, we refer to this haemogregarine as Karyolysus cf. lacazei. Besides the Schellackia sequences being identical to a previously identified haplotype, we also obtained sequences of three new closely related haplotypes.


Asunto(s)
Coccidiosis/veterinaria , Eucoccidiida/clasificación , Variación Genética , Ixodes/parasitología , Lagartos/parasitología , Animales , Coccidiosis/sangre , Coccidiosis/parasitología , ADN Protozoario/genética , Haplotipos , Filogenia , Análisis de Secuencia de ADN , Esporozoítos/genética , Trofozoítos/genética
16.
Parasitol Res ; 118(10): 3043-3051, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31471745

RESUMEN

Using a combination of morphological and molecular data, we describe a new apicomplexan parasite, Isospora svecica sp. n., from the white-spotted bluethroat, Luscinia svecica cyanecula, from the Czech Republic. Oocysts were found in its intestinal tract. Sporulation was exogenous and took 1-3 days. The oocysts were slightly ellipsoidal, of average size 26.17 × 20.33 µm, with a smooth bilayered wall. Micropyle, oocyst residuum, and polar granules were absent. Sporocysts were bottle-shaped, of an average size of 18.82 × 8.82 µm, with a thin, colourless wall. A conspicuous knob-like Stieda body was present. Substieda body was barely visible. Sporocyst residuum was present in the form of granules of various sizes. Sporozoites were banana-shaped and contained large anterior and small posterior refractile bodies. Partial DNA sequences of three genes were obtained from oocysts of Isospora svecica sp. n., being most closely related to other isosporans described from passerines. Little is known about the parasites of the avian family Muscicapidae, including coccidia, a highly prevalent parasitic protist group in all vertebrate classes. Only six species of the genus Isospora have so far been described in Muscicapidae, together with several "Isospora sp." that in fact most likely represent Isospora lacazei. The newly described Isospora svecica sp. n. differs morphologically from other coccidia reported from muscicapid birds, and represents the first coccidian species described from Luscinia svecica.


Asunto(s)
Isospora/clasificación , Isosporiasis/veterinaria , Passeriformes/parasitología , Animales , República Checa , Genes Protozoarios/genética , Intestinos/parasitología , Isospora/citología , Isospora/genética , Isospora/crecimiento & desarrollo , Isosporiasis/parasitología , Oocistos/clasificación , Oocistos/citología , Oocistos/genética , Oocistos/crecimiento & desarrollo , Esporozoítos/clasificación , Esporozoítos/citología , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo
17.
Parasitol Res ; 118(10): 2989-2999, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31473858

RESUMEN

Chicken coccidiosis is caused by the apicomplexan parasite Eimeria spp. At present, drug resistance of Eimeria is common because of the indiscriminate use of anticoccidial drugs. The gene encoding surface antigen 10 of Eimeria tenella (EtSAG10) is differentially expressed between drug-resistant and drug-sensitive strains. RNA-seq analysis indicated that this gene was downregulated in strains resistant to maduramicin and diclazuril compared to susceptible strains. EtSAG10 DNA sequence alignment revealed that they contained one and ten mutations in MRR and DZR, compared with DS, respectively. A full-length EtSAG10 cDNA was successfully cloned and expressed, and the polyclonal antibody was prepared. The transcription and translation levels of EtSAG10 were analyzed by quantitative real-time PCR (qPCR) and Western blotting. The localization of EtSAG10 in Spz, Mrz, and parasites in the first asexual stage was determined by indirect immunofluorescence. The potential association of EtSAG10 with sporozoite invasion of host cells was assessed by invasion inhibition assays. The results showed that EtSAG10 had a predicted transmembrane domain at the C-terminal end and a predicted signal peptide at the N-terminal end. EtSAG10 was downregulated in drug-resistant strains, which is consistent with the RNA-seq results. The EtSAG10 protein was localized to the parasite surface and parasitophorous vacuole membrane. This protein was shown to play a role in the infection of chicken intestine by sporozoites.


Asunto(s)
Antígenos de Protozoos/genética , Antígenos de Superficie/genética , Pollos/parasitología , Coccidiosis/veterinaria , Eimeria tenella/inmunología , Enfermedades de las Aves de Corral/parasitología , Animales , Antígenos de Protozoos/metabolismo , Antígenos de Superficie/metabolismo , Coccidiosis/parasitología , Coccidiostáticos/farmacología , Resistencia a Medicamentos/genética , Eimeria tenella/efectos de los fármacos , Eimeria tenella/genética , Eimeria tenella/crecimiento & desarrollo , Regulación de la Expresión Génica , Mutación , Esporozoítos/genética , Esporozoítos/inmunología
18.
Infect Immun ; 86(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29440367

RESUMEN

Genetically attenuated malaria parasites (GAP) that arrest during liver stage development are powerful immunogens and afford complete and durable protection against sporozoite infection. Late liver stage-arresting GAP provide superior protection against sporozoite challenge in mice compared to early live stage-arresting attenuated parasites. However, very few late liver stage-arresting GAP have been generated to date. Therefore, identification of additional loci that are critical for late liver stage development and can be used to generate novel late liver stage-arresting GAPs is of importance. We further explored genetic attenuation in Plasmodium yoelii by combining two gene deletions, PlasMei2 and liver-specific protein 2 (LISP2), that each cause late liver stage arrest with various degrees of infrequent breakthrough to blood stage infection. The dual gene deletion resulted in a synthetic lethal phenotype that caused complete attenuation in a highly susceptible mouse strain. P. yoeliiplasmei2-lisp2- arrested late in liver stage development and did not persist in livers beyond 3 days after infection. Immunization with this GAP elicited robust protective antibody responses in outbred and inbred mice against sporozoites, liver stages, and blood stages as well as eliciting protective liver-resident T cells. The immunization afforded protection against both sporozoite challenge and blood stage challenge. These findings provide evidence that completely attenuated late liver stage-arresting GAP are achievable via the synthetic lethal approach and might enable a path forward for the creation of a completely attenuated late liver stage-arresting P. falciparum GAP.


Asunto(s)
Eritrocitos/inmunología , Hígado/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Animales , Inmunización/métodos , Malaria/prevención & control , Ratones , Ratones Endogámicos BALB C , Plasmodium yoelii/genética , Proteínas Protozoarias/genética , Esporozoítos/genética
19.
Parasitol Res ; 117(8): 2487-2497, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29797085

RESUMEN

Malaria is transmitted through the injection of Plasmodium sporozoites into the skin by Anopheles mosquitoes. The parasites first replicate within the liver before infecting red blood cells, which leads to the symptoms of the disease. Experimental immunization with attenuated sporozoites that arrest their development in the liver has been extensively investigated in rodent models and humans. Recent technological advances have included the capacity to cryopreserve sporozoites for injection, which has enabled a series of controlled studies on human infection with sporozoites. Here, we used a cryopreservation protocol to test the efficiency of genetically attenuated cryopreserved sporozoites for immunization of mice in comparison with freshly isolated controls. This showed that cryopreserved sporozoites are highly viable as judged by their capacity to migrate in vitro but show only 20% efficiency in liver infection, which impacts their capacity to generate protection of animals in immunization experiments.


Asunto(s)
Malaria/prevención & control , Plasmodium berghei/inmunología , Esporozoítos/inmunología , Vacunación , Vacunas Atenuadas/inmunología , Animales , Anopheles/parasitología , Línea Celular Tumoral , Movimiento Celular/fisiología , Criopreservación , Células Hep G2 , Humanos , Hígado/parasitología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Esporozoítos/genética , Esporozoítos/metabolismo
20.
BMC Genomics ; 18(1): 686, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28870168

RESUMEN

BACKGROUND: Parasites can either respond to differences in immune defenses that exist between individual hosts plastically or, alternatively, follow a genetically canalized ("hard wired") program of infection. Assuming that large-scale functional plasticity would be discernible in the parasite transcriptome we have performed a dual RNA-seq study of the lifecycle of Eimeria falciformis using infected mice with different immune status as models for coccidian infections. RESULTS: We compared parasite and host transcriptomes (dual transcriptome) between naïve and challenge infected mice, as well as between immune competent and immune deficient ones. Mice with different immune competence show transcriptional differences as well as differences in parasite reproduction (oocyst shedding). Broad gene categories represented by differently abundant host genes indicate enrichments for immune reaction and tissue repair functions. More specifically, TGF-beta, EGF, TNF and IL-1 and IL-6 are examples of functional annotations represented differently depending on host immune status. Much in contrast, parasite transcriptomes were neither different between Coccidia isolated from immune competent and immune deficient mice, nor between those harvested from naïve and challenge infected mice. Instead, parasite transcriptomes have distinct profiles early and late in infection, characterized largely by biosynthesis or motility associated functional gene groups, respectively. Extracellular sporozoite and oocyst stages showed distinct transcriptional profiles and sporozoite transcriptomes were found enriched for species specific genes and likely pathogenicity factors. CONCLUSION: We propose that the niche and host-specific parasite E. falciformis uses a genetically canalized program of infection. This program is likely fixed in an evolutionary process rather than employing phenotypic plasticity to interact with its host. This in turn might limit the potential of the parasite to adapt to new host species or niches, forcing it to coevolve with its host.


Asunto(s)
Coccidios/inmunología , Coccidios/parasitología , Eimeria/genética , Eimeria/fisiología , Interacciones Huésped-Parásitos , Análisis de Secuencia de ARN , Eimeria/crecimiento & desarrollo , Evolución Molecular , Perfilación de la Expresión Génica , Reproducción Asexuada/genética , Esporozoítos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA