Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Microbiol ; 121(3): 481-496, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38009402

RESUMEN

Plasmodium sporozoite development in and egress from oocysts in the Anopheles mosquito remains largely enigmatic. In a previously performed high-throughput knockout screen, the putative subunit 5 of the prefoldin complex (PbPCS5, PBANKA_0920100) was identified as essential for parasite development during mosquito and liver stage development. Here we generated and analyzed a PbPCS5 knockout parasite line during its development in the mosquito. Interestingly, PbPCS5 deletion does not significantly affect oocyst formation but leads to a growth defect resulting in aberrantly shaped sporozoites. Sporozoites produced in the absence of PbPCS5 were thinner, markedly elongated, and did, in most cases, not contain a nucleus. Sporozoites contained fewer subpellicular microtubules, which reached deep into the sporoblast during sporogony where they contacted and indented nuclei. These aberrantly shaped sporozoites did not reach the salivary glands, and we, therefore, conclude that PbPCS5 is essential for sporogony and the life cycle progression of the parasite during its mosquito stage.


Asunto(s)
Anopheles , Chaperonas Moleculares , Parásitos , Animales , Plasmodium berghei/genética , Oocistos , Esporozoítos , Anopheles/parasitología , Proteínas Protozoarias/genética , Microtúbulos
2.
NPJ Vaccines ; 7(1): 139, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36333336

RESUMEN

Whole-sporozoite (WSp) malaria vaccines induce protective immune responses in animal malaria models and in humans. A recent clinical trial with a WSp vaccine comprising genetically attenuated parasites (GAP) which arrest growth early in the liver (PfSPZ-GA1), showed that GAPs can be safely administered to humans and immunogenicity is comparable to radiation-attenuated PfSPZ Vaccine. GAPs that arrest late in the liver stage (LA-GAP) have potential for increased potency as shown in rodent malaria models. Here we describe the generation of four putative P. falciparum LA-GAPs, generated by CRISPR/Cas9-mediated gene deletion. One out of four gene-deletion mutants produced sporozoites in sufficient numbers for further preclinical evaluation. This mutant, PfΔmei2, lacking the mei2-like RNA gene, showed late liver growth arrest in human liver-chimeric mice with human erythrocytes, absence of unwanted genetic alterations and sensitivity to antimalarial drugs. These features of PfΔmei2 make it a promising vaccine candidate, supporting further clinical evaluation. PfΔmei2 (GA2) has passed regulatory approval for safety and efficacy testing in humans based on the findings reported in this study.

3.
Front Cell Infect Microbiol ; 10: 583812, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33154955

RESUMEN

Kinesin-5 motors play essential roles in spindle apparatus assembly during cell division, by generating forces to establish and maintain the spindle bipolarity essential for proper chromosome segregation. Kinesin-5 is largely conserved structurally and functionally in model eukaryotes, but its role is unknown in the Plasmodium parasite, an evolutionarily divergent organism with several atypical features of both mitotic and meiotic cell division. We have investigated the function and subcellular location of kinesin-5 during cell division throughout the Plasmodium berghei life cycle. Deletion of kinesin-5 had little visible effect at any proliferative stage except sporozoite production in oocysts, resulting in a significant decrease in the number of motile sporozoites in mosquito salivary glands, which were able to infect a new vertebrate host. Live-cell imaging showed kinesin-5-GFP located on the spindle and at spindle poles during both atypical mitosis and meiosis. Fixed-cell immunofluorescence assays revealed kinesin-5 co-localized with α-tubulin and centrin-2 and a partial overlap with kinetochore marker NDC80 during early blood stage schizogony. Dual-color live-cell imaging showed that kinesin-5 is closely associated with NDC80 during male gametogony, but not with kinesin-8B, a marker of the basal body and axonemes of the forming flagella. Treatment of gametocytes with microtubule-specific inhibitors confirmed kinesin-5 association with nuclear spindles and not cytoplasmic axonemal microtubules. Altogether, our results demonstrate that kinesin-5 is associated with the spindle apparatus, expressed in proliferating parasite stages, and important for efficient production of infectious sporozoites.


Asunto(s)
Cinesinas , Esporozoítos , Animales , Segregación Cromosómica , Cinesinas/genética , Masculino , Microtúbulos , Plasmodium berghei , Huso Acromático
4.
Cell Rep ; 30(6): 1883-1897.e6, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049018

RESUMEN

Condensin is a multi-subunit protein complex regulating chromosome condensation and segregation during cell division. In Plasmodium spp., the causative agent of malaria, cell division is atypical and the role of condensin is unclear. Here we examine the role of SMC2 and SMC4, the core subunits of condensin, during endomitosis in schizogony and endoreduplication in male gametogenesis. During early schizogony, SMC2/SMC4 localize to a distinct focus, identified as the centromeres by NDC80 fluorescence and chromatin immunoprecipitation sequencing (ChIP-seq) analyses, but do not form condensin I or II complexes. In mature schizonts and during male gametogenesis, there is a diffuse SMC2/SMC4 distribution on chromosomes and in the nucleus, and both condensin I and condensin II complexes form at these stages. Knockdown of smc2 and smc4 gene expression reveals essential roles in parasite proliferation and transmission. The condensin core subunits (SMC2/SMC4) form different complexes and may have distinct functions at various stages of the parasite life cycle.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Mitosis/fisiología , Complejos Multiproteicos/metabolismo , Parásitos/patogenicidad , Plasmodium/patogenicidad , Animales , Proliferación Celular
5.
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
6.
PLoS Pathog ; 15(10): e1008048, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31600347

RESUMEN

Kinesin-8 proteins are microtubule motors that are often involved in regulation of mitotic spindle length and chromosome alignment. They move towards the plus ends of spindle microtubules and regulate the dynamics of these ends due, at least in some species, to their microtubule depolymerization activity. Plasmodium spp. exhibit an atypical endomitotic cell division in which chromosome condensation and spindle dynamics in the different proliferative stages are not well understood. Genome-wide shared orthology analysis of Plasmodium spp. revealed the presence of two kinesin-8 motor proteins, kinesin-8X and kinesin-8B. Here we studied the biochemical properties of kinesin-8X and its role in parasite proliferation. In vitro, kinesin-8X has motility and depolymerization activities like other kinesin-8 motors. To understand the role of Plasmodium kinesin-8X in cell division, we used fluorescence-tagging and live cell imaging to define its location, and gene targeting to analyse its function, during all proliferative stages of the rodent malaria parasite P. berghei life cycle. The results revealed a spatio-temporal involvement of kinesin-8X in spindle dynamics and an association with both mitotic and meiotic spindles and the putative microtubule organising centre (MTOC). Deletion of the kinesin-8X gene revealed a defect in oocyst development, confirmed by ultrastructural studies, suggesting that this protein is required for oocyst development and sporogony. Transcriptome analysis of Δkinesin-8X gametocytes revealed modulated expression of genes involved mainly in microtubule-based processes, chromosome organisation and the regulation of gene expression, supporting a role for kinesin-8X in cell division. Kinesin-8X is thus required for parasite proliferation within the mosquito and for transmission to the vertebrate host.


Asunto(s)
Cinesinas/metabolismo , Malaria/parasitología , Malaria/transmisión , Oocistos/citología , Plasmodium/fisiología , Proteínas Protozoarias/metabolismo , Huso Acromático/fisiología , Animales , Segregación Cromosómica , Femenino , Cinesinas/genética , Masculino , Ratones Endogámicos BALB C , Microtúbulos/metabolismo , Mitosis , Oocistos/fisiología , Proteínas Protozoarias/genética
7.
Biol Open ; 8(1)2019 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-30541825

RESUMEN

Centrins are calmodulin-like phosphoproteins present in the centrosome and play an active role in the duplication, separation and organization of centrosomal structures such as the microtubule-organizing centre (MTOC) during mitosis. They are also major components of the basal body of flagella and cilia. In Plasmodium spp., the parasite that causes malaria, mitosis is closed during asexual replication and the MTOC is embedded within the intact nuclear membrane. The MTOC has been named the centriolar plaque and is similar to the spindle pole body in yeast. In all phases of asexual replication, repeated rounds of nuclear division precede cell division. However, our knowledge of the location and function of centrins during this process is limited. Previous studies have identified four putative centrins in the human parasite P lasmodium falciparum. We report here the cellular localization of an alveolate-specific centrin (PbCEN-4) during the atypical cell division of asexual replicative stages, using live cell imaging with the rodent malaria parasite P. berghei as a model system. We show that this centrin forms a multi-protein complex with other centrins, but is dispensable for parasite proliferation.

8.
J Biol Chem ; 292(43): 17857-17875, 2017 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-28893907

RESUMEN

Myosin A (MyoA) is a Class XIV myosin implicated in gliding motility and host cell and tissue invasion by malaria parasites. MyoA is part of a membrane-associated protein complex called the glideosome, which is essential for parasite motility and includes the MyoA light chain myosin tail domain-interacting protein (MTIP) and several glideosome-associated proteins (GAPs). However, most studies of MyoA have focused on single stages of the parasite life cycle. We examined MyoA expression throughout the Plasmodium berghei life cycle in both mammalian and insect hosts. In extracellular ookinetes, sporozoites, and merozoites, MyoA was located at the parasite periphery. In the sexual stages, zygote formation and initial ookinete differentiation precede MyoA synthesis and deposition, which occurred only in the developing protuberance. In developing intracellular asexual blood stages, MyoA was synthesized in mature schizonts and was located at the periphery of segmenting merozoites, where it remained throughout maturation, merozoite egress, and host cell invasion. Besides the known GAPs in the malaria parasite, the complex included GAP40, an additional myosin light chain designated essential light chain (ELC), and several other candidate components. This ELC bound the MyoA neck region adjacent to the MTIP-binding site, and both myosin light chains co-located to the glideosome. Co-expression of MyoA with its two light chains revealed that the presence of both light chains enhances MyoA-dependent actin motility. In conclusion, we have established a system to study the interplay and function of the three glideosome components, enabling the assessment of inhibitors that target this motor complex to block host cell invasion.


Asunto(s)
Estadios del Ciclo de Vida/fisiología , Proteínas de la Membrana , Miosinas , Plasmodium berghei , Plasmodium falciparum , Proteínas Protozoarias , Animales , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Miosinas/genética , Miosinas/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
9.
Malar J ; 16(1): 305, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28764716

RESUMEN

BACKGROUND: Malaria research is greatly dependent on and has drastically advanced with the possibility of genetically modifying Plasmodium parasites. The commonly used transfection protocol by Janse and colleagues utilizes blood stage-derived Plasmodium berghei schizonts that have been purified from a blood culture by density gradient centrifugation. Naturally, this transfection protocol depends on the availability of suitably infected mice, constituting a time-based variable. In this study, the potential of transfecting liver stage-derived merozoites was explored. In cell culture, upon merozoite development, infected cells detach from the neighbouring cells and can be easily harvested from the cell culture supernatant. This protocol offers robust experimental timing and temporal flexibility. METHODS: HeLa cells are infected with P. berghei sporozoites to obtain liver stage-derived merozoites, which are harvested from the cell culture supernatant and are transfected using the Amaxa Nucleofector® electroporation technology. RESULTS: Using this protocol, wild type P. berghei ANKA strain and marker-free PbmCherryHsp70-expressing P. berghei parasites were successfully transfected with DNA constructs designed for integration via single- or double-crossover homologous recombination. CONCLUSION: An alternative protocol for Plasmodium transfection is hereby provided, which uses liver stage-derived P. berghei merozoites for transfection. This protocol has the potential to substantially reduce the number of mice used per transfection, as well as to increase the temporal flexibility and robustness of performing transfections, if mosquitoes are routinely present in the laboratory. Transfection of liver stage-derived P. berghei parasites should enable generation of transgenic parasites within 8-18 days.


Asunto(s)
Merozoítos/fisiología , Microorganismos Modificados Genéticamente/fisiología , Plasmodium berghei/fisiología , Animales , Técnicas de Cultivo de Célula , Hígado , Merozoítos/genética , Merozoítos/crecimiento & desarrollo , Ratones , Ratones Endogámicos BALB C , Microorganismos Modificados Genéticamente/genética , Microorganismos Modificados Genéticamente/crecimiento & desarrollo , Plasmodium berghei/genética , Esquizontes/genética , Esquizontes/crecimiento & desarrollo , Esquizontes/fisiología , Transfección
10.
Mol Microbiol ; 102(5): 775-791, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27566438

RESUMEN

To fuel the tremendously fast replication of Plasmodium liver stage parasites, the endoplasmic reticulum (ER) must play a critical role as a major site of protein and lipid biosynthesis. In this study, we analysed the parasite's ER morphology and function. Previous studies exploring the parasite ER have mainly focused on the blood stage. Visualizing the Plasmodium berghei ER during liver stage development, we found that the ER forms an interconnected network throughout the parasite with perinuclear and peripheral localizations. Surprisingly, we observed that the ER additionally generates huge accumulations. Using stimulated emission depletion microscopy and serial block-face scanning electron microscopy, we defined ER accumulations as intricate dense networks of ER tubules. We provide evidence that these accumulations are functional subdivisions of the parasite ER, presumably generated in response to elevated demands of the parasite, potentially consistent with ER stress. Compared to higher eukaryotes, Plasmodium parasites have a fundamentally reduced unfolded protein response machinery for reacting to ER stress. Accordingly, parasite development is greatly impaired when ER stress is applied. As parasites appear to be more sensitive to ER stress than are host cells, induction of ER stress could potentially be used for interference with parasite development.


Asunto(s)
Retículo Endoplásmico/ultraestructura , Plasmodium berghei/ultraestructura , Animales , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , Hígado/parasitología , Malaria/parasitología , Microscopía/métodos , Microscopía Electrónica de Rastreo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Respuesta de Proteína Desplegada
11.
Sci Rep ; 6: 28604, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27339728

RESUMEN

The SAS6-like (SAS6L) protein, a truncated paralogue of the ubiquitous basal body/centriole protein SAS6, has been characterised recently as a flagellum protein in trypanosomatids, but associated with the conoid in apicomplexan Toxoplasma. The conoid has been suggested to derive from flagella parts, but is thought to have been lost from some apicomplexans including the malaria-causing genus Plasmodium. Presence of SAS6L in Plasmodium, therefore, suggested a possible role in flagella assembly in male gametes, the only flagellated stage. Here, we have studied the expression and role of SAS6L throughout the Plasmodium life cycle using the rodent malaria model P. berghei. Contrary to a hypothesised role in flagella, SAS6L was absent during gamete flagellum formation. Instead, SAS6L was restricted to the apical complex in ookinetes and sporozoites, the extracellular invasive stages that develop within the mosquito vector. In these stages SAS6L forms an apical ring, as we show is also the case in Toxoplasma tachyzoites. The SAS6L ring was not apparent in blood-stage invasive merozoites, indicating that the apical complex is differentiated between the different invasive forms. Overall this study indicates that a conoid-associated apical complex protein and ring structure is persistent in Plasmodium in a stage-specific manner.


Asunto(s)
Cuerpos Basales/metabolismo , Mosquitos Vectores/metabolismo , Mosquitos Vectores/parasitología , Plasmodium/metabolismo , Plasmodium/parasitología , Proteínas Protozoarias/metabolismo , Animales , Cuerpos Basales/parasitología , Femenino , Flagelos/metabolismo , Flagelos/parasitología , Estadios del Ciclo de Vida/fisiología , Malaria/metabolismo , Malaria/parasitología , Merozoítos/metabolismo , Ratones , Esporozoítos/metabolismo , Toxoplasma/metabolismo , Toxoplasma/parasitología
12.
Malar J ; 15: 232, 2016 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-27102897

RESUMEN

BACKGROUND: Bioluminescence imaging is widely used for cell-based assays and animal imaging studies, both in biomedical research and drug development. Its main advantages include its high-throughput applicability, affordability, high sensitivity, operational simplicity, and quantitative outputs. In malaria research, bioluminescence has been used for drug discovery in vivo and in vitro, exploring host-pathogen interactions, and studying multiple aspects of Plasmodium biology. While the number of fluorescent proteins available for imaging has undergone a great expansion over the last two decades, enabling simultaneous visualization of multiple molecular and cellular events, expansion of available luciferases has lagged behind. The most widely used bioluminescent probe in malaria research is the Photinus pyralis firefly luciferase, followed by the more recently introduced Click-beetle and Renilla luciferases. Ultra-sensitive imaging of Plasmodium at low parasite densities has not been previously achieved. With the purpose of overcoming these challenges, a Plasmodium berghei line expressing the novel ultra-bright luciferase enzyme NanoLuc, called PbNLuc has been generated, and is presented in this work. RESULTS: NanoLuc shows at least 150 times brighter signal than firefly luciferase in vitro, allowing single parasite detection in mosquito, liver, and sexual and asexual blood stages. As a proof-of-concept, the PbNLuc parasites were used to image parasite development in the mosquito, liver and blood stages of infection, and to specifically explore parasite liver stage egress, and pre-patency period in vivo. CONCLUSIONS: PbNLuc is a suitable parasite line for sensitive imaging of the entire Plasmodium life cycle. Its sensitivity makes it a promising line to be used as a reference for drug candidate testing, as well as the characterization of mutant parasites to explore the function of parasite proteins, host-parasite interactions, and the better understanding of Plasmodium biology. Since the substrate requirements of NanoLuc are different from those of firefly luciferase, dual bioluminescence imaging for the simultaneous characterization of two lines, or two separate biological processes, is possible, as demonstrated in this work.


Asunto(s)
Mediciones Luminiscentes/métodos , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Animales , Culicidae/parasitología , Interacciones Huésped-Parásitos , Humanos , Hígado/parasitología , Luciferasas/genética , Organismos Modificados Genéticamente/genética , Organismos Modificados Genéticamente/metabolismo , Plasmodium berghei/aislamiento & purificación
13.
J Biol Chem ; 290(19): 12147-64, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25802338

RESUMEN

Myosin B (MyoB) is one of the two short class XIV myosins encoded in the Plasmodium genome. Class XIV myosins are characterized by a catalytic "head," a modified "neck," and the absence of a "tail" region. Myosin A (MyoA), the other class XIV myosin in Plasmodium, has been established as a component of the glideosome complex important in motility and cell invasion, but MyoB is not well characterized. We analyzed the properties of MyoB using three parasite species as follows: Plasmodium falciparum, Plasmodium berghei, and Plasmodium knowlesi. MyoB is expressed in all invasive stages (merozoites, ookinetes, and sporozoites) of the life cycle, and the protein is found in a discrete apical location in these polarized cells. In P. falciparum, MyoB is synthesized very late in schizogony/merogony, and its location in merozoites is distinct from, and anterior to, that of a range of known proteins present in the rhoptries, rhoptry neck or micronemes. Unlike MyoA, MyoB is not associated with glideosome complex proteins, including the MyoA light chain, myosin A tail domain-interacting protein (MTIP). A unique MyoB light chain (MLC-B) was identified that contains a calmodulin-like domain at the C terminus and an extended N-terminal region. MLC-B localizes to the same extreme apical pole in the cell as MyoB, and the two proteins form a complex. We propose that MLC-B is a MyoB-specific light chain, and for the short class XIV myosins that lack a tail region, the atypical myosin light chains may fulfill that role.


Asunto(s)
Miosina Tipo IIB no Muscular/química , Plasmodium berghei/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium knowlesi/metabolismo , Proteínas Protozoarias/química , Secuencia de Aminoácidos , Calmodulina/química , Dicroismo Circular , Técnica del Anticuerpo Fluorescente Indirecta , Proteínas Fluorescentes Verdes/química , Datos de Secuencia Molecular , Cadenas Ligeras de Miosina/química , Miosina Tipo IIA no Muscular/química , Péptidos/química , Unión Proteica , Desnaturalización Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
14.
Malar J ; 13: 315, 2014 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25124718

RESUMEN

BACKGROUND: Gametogenesis and fertilization play crucial roles in malaria transmission. While male gametes are thought to be amongst the simplest eukaryotic cells and are proven targets of transmission blocking immunity, little is known about their molecular organization. For example, the pathway of energy metabolism that power motility, a feature that facilitates gamete encounter and fertilization, is unknown. METHODS: Plasmodium berghei microgametes were purified and analysed by whole-cell proteomic analysis for the first time. Data are available via ProteomeXchange with identifier PXD001163. RESULTS: 615 proteins were recovered, they included all male gamete proteins described thus far. Amongst them were the 11 enzymes of the glycolytic pathway. The hexose transporter was localized to the gamete plasma membrane and it was shown that microgamete motility can be suppressed effectively by inhibitors of this transporter and of the glycolytic pathway. CONCLUSIONS: This study describes the first whole-cell proteomic analysis of the malaria male gamete. It identifies glycolysis as the likely exclusive source of energy for flagellar beat, and provides new insights in original features of Plasmodium flagellar organization.


Asunto(s)
Metabolismo Energético , Flagelos/fisiología , Células Germinativas/química , Glucólisis , Plasmodium berghei/química , Plasmodium berghei/fisiología , Proteoma/análisis , Animales , Femenino , Locomoción , Masculino , Ratones
15.
PLoS Pathog ; 10(8): e1004336, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25166051

RESUMEN

Plasmodium parasites express a potent inhibitor of cysteine proteases (ICP) throughout their life cycle. To analyze the role of ICP in different life cycle stages, we generated a stage-specific knockout of the Plasmodium berghei ICP (PbICP). Excision of the pbicb gene occurred in infective sporozoites and resulted in impaired sporozoite invasion of hepatocytes, despite residual PbICP protein being detectable in sporozoites. The vast majority of these parasites invading a cultured hepatocyte cell line did not develop to mature liver stages, but the few that successfully developed hepatic merozoites were able to initiate a blood stage infection in mice. These blood stage parasites, now completely lacking PbICP, exhibited an attenuated phenotype but were able to infect mosquitoes and develop to the oocyst stage. However, PbICP-negative sporozoites liberated from oocysts exhibited defective motility and invaded mosquito salivary glands in low numbers. They were also unable to invade hepatocytes, confirming that control of cysteine protease activity is of critical importance for sporozoites. Importantly, transfection of PbICP-knockout parasites with a pbicp-gfp construct fully reversed these defects. Taken together, in P. berghei this inhibitor of the ICP family is essential for sporozoite motility but also appears to play a role during parasite development in hepatocytes and erythrocytes.


Asunto(s)
Inhibidores de Cisteína Proteinasa/metabolismo , Malaria/parasitología , Plasmodium berghei/crecimiento & desarrollo , Animales , Eritrocitos/parasitología , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Células Hep G2 , Hepatocitos/parasitología , Humanos , Estadios del Ciclo de Vida , Malaria/metabolismo , Ratones , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Transfección
16.
Int J Parasitol ; 43(6): 503-14, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23500072

RESUMEN

Recently it has been shown in rodent malaria models that immunisation with genetically attenuated Plasmodium parasites can confer sterile protection against challenge with virulent parasites. For the mass production of live attenuated Plasmodium parasites for vaccination, safety is a prerequisite. Knockout of a single gene is not sufficient for such a strategy since the parasite can likely compensate for such a genetic modification and a single surviving parasite is sufficient to kill an immunised individual. Parasites must therefore be at least double-attenuated when generating a safe vaccine strain. Genetic double-attenuation can be achieved by knocking out two essential genes or by combining a single gene knockout with the expression of a protein toxic for the parasite. We generated a double-attenuated Plasmodium berghei strain that is deficient in fatty acid synthesis by the knockout of the pdh-e1α gene, introducing a second attenuation by the liver stage-specific expression of the pore-forming bacterial toxin perfringolysin O. With this double genetically attenuated parasite strain, a superior attenuation was indeed achieved compared with single-attenuated strains that were either deficient in pyruvate dehydrogenase (PDH)-E1 or expressed perfringolysin O. In vivo, both single-attenuated strains resulted in breakthrough infections even if low to moderate doses of sporozoites (2,000-5,000) were administered. In contrast, the double genetically attenuated parasite strain, given at moderate doses of 5,000 sporozoites, did not result in blood stage infection and even when administered at 5- to 20-fold higher doses, only single and delayed breakthrough infections were observed. Prime booster immunisation with the double genetically attenuated parasite strain completely protected a susceptible mouse strain from malaria and even a single immunisation conferred protection in some cases and lead to a markedly delayed onset of blood stage infection in others. Importantly, premature rupture of the parasitophorous vacuole membrane by liver stage-specific perfringolysin O expression did not induce host cell death and soluble parasite proteins, which are released into the host cell cytoplasm, have the potential to be processed and presented via MHC class I molecules. This, in turn, might support immunological responses against Plasmodium-infected hepatocytes.


Asunto(s)
Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/inmunología , Plasmodium berghei/inmunología , Plasmodium berghei/patogenicidad , Acidosis Láctica , Animales , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/genética , Sangre/parasitología , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Genes Esenciales , Genes Protozoarios , Malaria/inmunología , Malaria/prevención & control , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Piruvato Deshidrogenasa (Lipoamida)/deficiencia , Análisis de Supervivencia , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
17.
Autophagy ; 9(4): 568-80, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23388496

RESUMEN

Analyzing molecular determinants of Plasmodium parasite cell death is a promising approach for exploring new avenues in the fight against malaria. Three major forms of cell death (apoptosis, necrosis and autophagic cell death) have been described in multicellular organisms but which cell death processes exist in protozoa is still a matter of debate. Here we suggest that all three types of cell death occur in Plasmodium liver-stage parasites. Whereas typical molecular markers for apoptosis and necrosis have not been found in the genome of Plasmodium parasites, we identified genes coding for putative autophagy-marker proteins and thus concentrated on autophagic cell death. We characterized the Plasmodium berghei homolog of the prominent autophagy marker protein Atg8/LC3 and found that it localized to the apicoplast. A relocalization of PbAtg8 to autophagosome-like vesicles or vacuoles that appear in dying parasites was not, however, observed. This strongly suggests that the function of this protein in liver-stage parasites is restricted to apicoplast biology.


Asunto(s)
Autofagia , Estadios del Ciclo de Vida , Hígado/parasitología , Parásitos/citología , Parásitos/crecimiento & desarrollo , Plasmodium berghei/citología , Plasmodium berghei/crecimiento & desarrollo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Bases de Datos de Proteínas , Evolución Molecular , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/metabolismo , Células Hep G2 , Humanos , Metabolismo de los Lípidos , Ratones , Datos de Secuencia Molecular , Parásitos/ultraestructura , Fagosomas/metabolismo , Fagosomas/ultraestructura , Plasmodium berghei/ultraestructura , Transporte de Proteínas , Proteínas Protozoarias/metabolismo , Saccharomyces cerevisiae/metabolismo , Esquizontes/citología , Esquizontes/metabolismo , Esquizontes/ultraestructura , Homología de Secuencia de Aminoácido , Vacuolas/metabolismo
18.
Methods Mol Biol ; 923: 411-27, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22990795

RESUMEN

Exoerythrocytic Plasmodium parasites infect hepatocytes and develop to huge multinucleated schizonts inside a parasitophorous vacuole. Finally, thousands of merozoites are formed and released into the host cell cytoplasm by complete disintegration of the parasitophorous vacuole membrane. This, in turn, results in death and detachment of the infected hepatocyte, followed by the formation of merosomes. The fast growth of the parasite and host cell detachment are hallmarks of liver stage development and can easily be monitored. Here, we describe how to translate these observations into assays for characterizing parasite development. Additionally, other recently introduced techniques and tools to analyze and manipulate liver stage parasites are also discussed.


Asunto(s)
Hepatocitos/metabolismo , Hepatocitos/parasitología , Merozoítos/metabolismo , Plasmodium/crecimiento & desarrollo , Animales , Anopheles/parasitología , Técnicas de Cultivo de Célula , Genes Reporteros , Células Hep G2 , Humanos , Malaria/metabolismo , Malaria/parasitología , Plásmidos/genética , Transfección
19.
FEMS Microbiol Rev ; 36(1): 111-30, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22092244

RESUMEN

Protozoan parasites of the genus Plasmodium are the causative agents of malaria. Despite more than 100 years of research, the complex life cycle of the parasite still bears many surprises and it is safe to say that understanding the biology of the pathogen will keep scientists busy for many years to come. Malaria research has mainly concentrated on the pathological blood stage of Plasmodium parasites, leaving us with many questions concerning parasite development within the mosquito and during the exo-erythrocytic stage in the vertebrate host. After the discovery of the Plasmodium liver stage in the middle of the last century, it remained understudied for many years but the realization that it represents a promising target for vaccination approaches has brought it back into focus. The last decade saw many new and exciting discoveries concerning the exo-erythrocytic stage and in this review we will discuss the highlights of the latest developments in the field.


Asunto(s)
Muerte Celular , Hepatocitos/parasitología , Hígado/parasitología , Plasmodium/patogenicidad , Animales , Humanos , Modelos Biológicos
20.
Cell Microbiol ; 14(3): 416-30, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22128915

RESUMEN

Lipoic acid is an essential cofactor for enzymes that participate in key metabolic pathways in most organisms. While in mammalian cells lipoylated proteins reside exclusively in the mitochondria, apicomplexan parasites of the genus Plasmodium harbour two independent lipoylation pathways in the mitochondrion and the apicoplast, a second organelle of endosymbiotic origin. Protein lipoylation in the apicoplast relies on de novo lipoic acid synthesis while lipoylation of proteins in the mitochondrion depends on scavenging of lipoic acid from the host cell. Here, we analyse the impact of lipoic acid scavenging on the development of Plasmodium berghei liver stage parasites. Treatment of P. berghei-infected HepG2 cells with the lipoic acid analogue 8-bromo-octanoic acid (8-BOA) abolished lipoylation of mitochondrial enzyme complexes in the parasite while lipoylation of apicoplast proteins was not affected. Parasite growth as well as the ability of the parasites to successfully complete liver stage development by merosome formation were severely impaired but not completely blocked by 8-BOA. Liver stage parasites were most sensitive to 8-BOA treatment during schizogony, the phase of development when the parasite grows and undergoes extensive nuclear division to form a multinucleated syncytium. Live cell imaging as well as immunofluorescence analysis and electronmicroscopy studies revealed a close association of both host cell and parasite mitochondria with the parasitophorous vacuole membrane suggesting that host cell mitochondria might be involved in lipoic acid uptake by the parasite from the host cell.


Asunto(s)
Hígado/parasitología , Mitocondrias/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Ácido Tióctico/metabolismo , Caprilatos/farmacología , Acido Graso Sintasa Tipo II/metabolismo , Células Hep G2 , Interacciones Huésped-Parásitos , Humanos , Membranas Intracelulares/metabolismo , Metabolismo de los Lípidos , Lipoilación , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Plasmodium berghei/efectos de los fármacos , Transporte de Proteínas , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética , Vacuolas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA