Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Microbiol ; 76: 113-134, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35609946

RESUMEN

The malaria parasite life cycle alternates between two hosts: a vertebrate and the female Anopheles mosquito vector. Cell division, proliferation, and invasion are essential for parasite development, transmission, and survival. Most research has focused on Plasmodium development in the vertebrate, which causes disease; however, knowledge of malaria parasite development in the mosquito (the sexual and transmission stages) is now rapidly accumulating, gathered largely through investigation of the rodent malaria model, with Plasmodium berghei. In this review, we discuss the seminal genome-wide screens that have uncovered key regulators of cell proliferation, invasion, and transmission during Plasmodium sexual development. Our focus is on the roles of transcription factors, reversible protein phosphorylation, and molecular motors. We also emphasize the still-unanswered important questions around key pathways in cell division during the vector transmission stages and how they may be targeted in future studies.


Asunto(s)
Anopheles , Malaria , Parásitos , Animales , Anopheles/parasitología , Femenino , Malaria/parasitología , Mosquitos Vectores , Plasmodium berghei/genética
2.
PLoS Biol ; 20(7): e3001704, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35900985

RESUMEN

Kinesins are microtubule (MT)-based motors important in cell division, motility, polarity, and intracellular transport in many eukaryotes. However, they are poorly studied in the divergent eukaryotic pathogens Plasmodium spp., the causative agents of malaria, which manifest atypical aspects of cell division and plasticity of morphology throughout the life cycle in both mammalian and mosquito hosts. Here, we describe a genome-wide screen of Plasmodium kinesins, revealing diverse subcellular locations and functions in spindle assembly, axoneme formation, and cell morphology. Surprisingly, only kinesin-13 is essential for growth in the mammalian host while the other 8 kinesins are required during the proliferative and invasive stages of parasite transmission through the mosquito vector. In-depth analyses of kinesin-13 and kinesin-20 revealed functions in MT dynamics during apical cell polarity formation, spindle assembly, and axoneme biogenesis. These findings help us to understand the importance of MT motors and may be exploited to discover new therapeutic interventions against malaria.


Asunto(s)
Culicidae , Malaria , Parásitos , Plasmodium , Animales , Humanos , Cinesinas/genética , Estadios del Ciclo de Vida/genética , Malaria/metabolismo , Mamíferos , Microtúbulos/metabolismo , Plasmodium/genética
3.
Biochem Soc Trans ; 52(2): 593-602, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38563493

RESUMEN

Malaria, a vector borne disease, is a major global health and socioeconomic problem caused by the apicomplexan protozoan parasite Plasmodium. The parasite alternates between mosquito vector and vertebrate host, with meiosis in the mosquito and proliferative mitotic cell division in both hosts. In the canonical eukaryotic model, cell division is either by open or closed mitosis and karyokinesis is followed by cytokinesis; whereas in Plasmodium closed mitosis is not directly accompanied by concomitant cell division. Key molecular players and regulatory mechanisms of this process have been identified, but the pivotal role of certain protein complexes and the post-translational modifications that modulate their actions are still to be deciphered. Here, we discuss recent evidence for the function of known proteins in Plasmodium cell division and processes that are potential novel targets for therapeutic intervention. We also identify key questions to open new and exciting research to understand divergent Plasmodium cell division.


Asunto(s)
División Celular , Malaria , Plasmodium , Proteínas Protozoarias , Plasmodium/metabolismo , Plasmodium/fisiología , Animales , Humanos , Malaria/parasitología , Malaria/metabolismo , Proteínas Protozoarias/metabolismo , Mitosis , Citocinesis , Meiosis , Procesamiento Proteico-Postraduccional , Interacciones Huésped-Parásitos
4.
PLoS Biol ; 19(3): e3001081, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33705380

RESUMEN

The apical complex is the instrument of invasion used by apicomplexan parasites, and the conoid is a conspicuous feature of this apparatus found throughout this phylum. The conoid, however, is believed to be heavily reduced or missing from Plasmodium species and other members of the class Aconoidasida. Relatively few conoid proteins have previously been identified, making it difficult to address how conserved this feature is throughout the phylum, and whether it is genuinely missing from some major groups. Moreover, parasites such as Plasmodium species cycle through 3 invasive forms, and there is the possibility of differential presence of the conoid between these stages. We have applied spatial proteomics and high-resolution microscopy to develop a more complete molecular inventory and understanding of the organisation of conoid-associated proteins in the model apicomplexan Toxoplasma gondii. These data revealed molecular conservation of all conoid substructures throughout Apicomplexa, including Plasmodium, and even in allied Myzozoa such as Chromera and dinoflagellates. We reporter-tagged and observed the expression and location of several conoid complex proteins in the malaria model P. berghei and revealed equivalent structures in all of its zoite forms, as well as evidence of molecular differentiation between blood-stage merozoites and the ookinetes and sporozoites of the mosquito vector. Collectively, we show that the conoid is a conserved apicomplexan element at the heart of the invasion mechanisms of these highly successful and often devastating parasites.


Asunto(s)
Apicomplexa/metabolismo , Plasmodium/metabolismo , Evolución Biológica , Citoesqueleto/metabolismo , Evolución Molecular , Malaria/parasitología , Mosquitos Vectores/metabolismo , Plasmodium/patogenicidad , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Toxoplasma/patogenicidad
5.
Nature ; 547(7662): 213-216, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28678779

RESUMEN

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


Asunto(s)
Regulación de la Expresión Génica , Malaria/parasitología , Parásitos/metabolismo , Parásitos/patogenicidad , Fosfotransferasas/metabolismo , Plasmodium/metabolismo , Plasmodium/patogenicidad , Animales , Restricción Calórica , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Regulación de la Expresión Génica/efectos de los fármacos , Prueba de Complementación Genética , Glucosa/metabolismo , Glucosa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Parasitemia/sangre , Parasitemia/genética , Parasitemia/metabolismo , Parasitemia/parasitología , Parásitos/genética , Parásitos/crecimiento & desarrollo , Fosfotransferasas/deficiencia , Fosfotransferasas/genética , Plasmodium/genética , Plasmodium/crecimiento & desarrollo , Ratas , Transcriptoma/efectos de los fármacos , Virulencia/efectos de los fármacos
6.
J Biol Chem ; 297(5): 101063, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34375637

RESUMEN

Plasmodium parasites cause malaria and are responsible annually for hundreds of thousands of deaths. Kinesins are a superfamily of microtubule-dependent ATPases that play important roles in the parasite replicative machinery, which is a potential target for antiparasite drugs. Kinesin-5, a molecular motor that cross-links microtubules, is an established antimitotic target in other disease contexts, but its mechanism in Plasmodium falciparum is unclear. Here, we characterized P. falciparum kinesin-5 (PfK5) using cryo-EM to determine the motor's nucleotide-dependent microtubule-bound structure and introduced 3D classification of individual motors into our microtubule image processing pipeline to maximize our structural insights. Despite sequence divergence in PfK5, the motor exhibits classical kinesin mechanochemistry, including ATP-induced subdomain rearrangement and cover neck bundle formation, consistent with its plus-ended directed motility. We also observed that an insertion in loop5 of the PfK5 motor domain creates a different environment in the well-characterized human kinesin-5 drug-binding site. Our data reveal the possibility for selective inhibition of PfK5 and can be used to inform future exploration of Plasmodium kinesins as antiparasite targets.


Asunto(s)
Cinesinas , Plasmodium falciparum , Proteínas Protozoarias , Antimaláricos/química , Microscopía por Crioelectrón , Humanos , Cinesinas/metabolismo , Cinesinas/ultraestructura , Plasmodium falciparum/química , Plasmodium falciparum/metabolismo , Plasmodium falciparum/ultraestructura , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura
7.
J Cell Sci ; 134(5)2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32501284

RESUMEN

Eukaryotic cell proliferation requires chromosome replication and precise segregation to ensure daughter cells have identical genomic copies. Species of the genus Plasmodium, the causative agents of malaria, display remarkable aspects of nuclear division throughout their life cycle to meet some peculiar and unique challenges to DNA replication and chromosome segregation. The parasite undergoes atypical endomitosis and endoreduplication with an intact nuclear membrane and intranuclear mitotic spindle. To understand these diverse modes of Plasmodium cell division, we have studied the behaviour and composition of the outer kinetochore NDC80 complex, a key part of the mitotic apparatus that attaches the centromere of chromosomes to microtubules of the mitotic spindle. Using NDC80-GFP live-cell imaging in Plasmodium berghei, we observe dynamic spatiotemporal changes during proliferation, including highly unusual kinetochore arrangements during sexual stages. We identify a very divergent candidate for the SPC24 subunit of the NDC80 complex, previously thought to be missing in Plasmodium, which completes a canonical, albeit unusual, NDC80 complex structure. Altogether, our studies reveal the kinetochore to be an ideal tool to investigate the non-canonical modes of chromosome segregation and cell division in Plasmodium.


Asunto(s)
Parásitos , Plasmodium , Animales , División Celular , Segregación Cromosómica/genética , Cinetocoros , Microtúbulos , Mitosis/genética , Plasmodium/genética , Huso Acromático/genética
8.
Proc Natl Acad Sci U S A ; 116(8): 3183-3192, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30723152

RESUMEN

The positioning of chromosomes in the nucleus of a eukaryotic cell is highly organized and has a complex and dynamic relationship with gene expression. In the human malaria parasite Plasmodium falciparum, the clustering of a family of virulence genes correlates with their coordinated silencing and has a strong influence on the overall organization of the genome. To identify conserved and species-specific principles of genome organization, we performed Hi-C experiments and generated 3D genome models for five Plasmodium species and two related apicomplexan parasites. Plasmodium species mainly showed clustering of centromeres, telomeres, and virulence genes. In P. falciparum, the heterochromatic virulence gene cluster had a strong repressive effect on the surrounding nuclear space, while this was less pronounced in Plasmodium vivax and Plasmodium berghei, and absent in Plasmodium yoelii In Plasmodium knowlesi, telomeres and virulence genes were more dispersed throughout the nucleus, but its 3D genome showed a strong correlation with gene expression. The Babesia microti genome showed a classical Rabl organization with colocalization of subtelomeric virulence genes, while the Toxoplasma gondii genome was dominated by clustering of the centromeres and lacked virulence gene clustering. Collectively, our results demonstrate that spatial genome organization in most Plasmodium species is constrained by the colocalization of virulence genes. P. falciparum and P. knowlesi, the only two Plasmodium species with gene families involved in antigenic variation, are unique in the effect of these genes on chromosome folding, indicating a potential link between genome organization and gene expression in more virulent pathogens.


Asunto(s)
Genoma de Protozoos/genética , Heterocromatina/genética , Malaria Falciparum/genética , Plasmodium falciparum/genética , Animales , Centrómero/genética , Regulación de la Expresión Génica/genética , Genómica , Humanos , Malaria Falciparum/parasitología , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Plasmodium falciparum/patogenicidad , Plasmodium knowlesi/genética , Plasmodium knowlesi/patogenicidad , Plasmodium vivax/genética , Plasmodium vivax/patogenicidad , Telómero/genética , Toxoplasma/genética , Toxoplasma/patogenicidad
9.
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
10.
Cell Microbiol ; 21(10): e13082, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31283102

RESUMEN

The myosin superfamily comprises of actin-dependent eukaryotic molecular motors important in a variety of cellular functions. Although well studied in many systems, knowledge of their functions in Plasmodium, the causative agent of malaria, is restricted. Previously, six myosins were identified in this genus, including three Class XIV myosins found only in Apicomplexa and some Ciliates. The well characterized MyoA is a Class XIV myosin essential for gliding motility and invasion. Here, we characterize all other Plasmodium myosins throughout the parasite life cycle and show that they have very diverse patterns of expression and cellular location. MyoB and MyoE, the other two Class XIV myosins, are expressed in all invasive stages, with apical and basal locations, respectively. Gene deletion revealed that MyoE is involved in sporozoite traversal, MyoF and MyoK are likely essential in the asexual blood stages, and MyoJ and MyoB are not essential. Both MyoB and its essential light chain (MCL-B) are localised at the apical end of ookinetes but expressed at completely different time points. This work provides a better understanding of the role of actomyosin motors in Apicomplexan parasites, particularly in the motile and invasive stages of Plasmodium during sexual and asexual development within the mosquito.


Asunto(s)
Miosinas/metabolismo , Plasmodium/crecimiento & desarrollo , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Animales , Femenino , Estadios del Ciclo de Vida , Espectrometría de Masas , Ratones , Miosinas/química , Miosinas/genética , Fenotipo , Plasmodium/genética , Dominios Proteicos/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Esporozoítos/crecimiento & desarrollo
11.
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
12.
PLoS Pathog ; 11(11): e1005273, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26565797

RESUMEN

Cell-cycle progression and cell division in eukaryotes are governed in part by the cyclin family and their regulation of cyclin-dependent kinases (CDKs). Cyclins are very well characterised in model systems such as yeast and human cells, but surprisingly little is known about their number and role in Plasmodium, the unicellular protozoan parasite that causes malaria. Malaria parasite cell division and proliferation differs from that of many eukaryotes. During its life cycle it undergoes two types of mitosis: endomitosis in asexual stages and an extremely rapid mitotic process during male gametogenesis. Both schizogony (producing merozoites) in host liver and red blood cells, and sporogony (producing sporozoites) in the mosquito vector, are endomitotic with repeated nuclear replication, without chromosome condensation, before cell division. The role of specific cyclins during Plasmodium cell proliferation was unknown. We show here that the Plasmodium genome contains only three cyclin genes, representing an unusual repertoire of cyclin classes. Expression and reverse genetic analyses of the single Plant (P)-type cyclin, CYC3, in the rodent malaria parasite, Plasmodium berghei, revealed a cytoplasmic and nuclear location of the GFP-tagged protein throughout the lifecycle. Deletion of cyc3 resulted in defects in size, number and growth of oocysts, with abnormalities in budding and sporozoite formation. Furthermore, global transcript analysis of the cyc3-deleted and wild type parasites at gametocyte and ookinete stages identified differentially expressed genes required for signalling, invasion and oocyst development. Collectively these data suggest that cyc3 modulates oocyst endomitotic development in Plasmodium berghei.


Asunto(s)
División Celular/fisiología , Ciclinas/metabolismo , Malaria/parasitología , Plasmodium berghei/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Culicidae , Ciclinas/genética , Femenino , Humanos , Ratones , Oocistos , Proteínas Protozoarias/genética , Esporozoítos/crecimiento & desarrollo
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.
PLoS Pathog ; 9(2): e1003191, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23468629

RESUMEN

Ca(2+) contributes to a myriad of important cellular processes in all organisms, including the apicomplexans, Plasmodium and Toxoplasma. Due to its varied and essential roles, free Ca(2+) is tightly regulated by complex mechanisms. These mechanisms are therefore of interest as putative drug targets. One pathway in Ca(2+) homeostatic control in apicomplexans uses a Ca(2+)/H(+) exchanger (a member of the cation exchanger family, CAX). The P. falciparum CAX (PfCAX) has recently been characterised in asexual blood stage parasites. To determine the physiological importance of apicomplexan CAXs, tagging and knock-out strategies were undertaken in the genetically tractable T. gondii and P. berghei parasites. In addition, a yeast heterologous expression system was used to study the function of apicomplexan CAXs. Tagging of T. gondii and P. berghei CAXs (TgCAX and PbCAX) under control of their endogenous promoters could not demonstrate measureable expression of either CAX in tachyzoites and asexual blood stages, respectively. These results were consistent with the ability of parasites to tolerate knock-outs of the genes for TgCAX and PbCAX at these developmental stages. In contrast, PbCAX expression was detectable during sexual stages of development in female gametocytes/gametes, zygotes and ookinetes, where it was dispersed in membranous networks within the cytosol (with minimal mitochondrial localisation). Furthermore, genetically disrupted parasites failed to develop further from "round" form zygotes, suggesting that PbCAX is essential for ookinete development and differentiation. This impeded phenotype could be rescued by removal of extracellular Ca(2+). Therefore, PbCAX provides a mechanism for free living parasites to multiply within the ionic microenvironment of the mosquito midgut. Ca(2+) homeostasis mediated by PbCAX is critical and suggests plasmodial CAXs may be targeted in approaches designed to block parasite transmission.


Asunto(s)
Antiportadores/metabolismo , Calcio/farmacología , Proteínas de Transporte de Catión/metabolismo , Plasmodium berghei/efectos de los fármacos , Reproducción Asexuada/efectos de los fármacos , Diferenciación Sexual/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Estadios del Ciclo de Vida , Ratones , Datos de Secuencia Molecular , Oogénesis , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/metabolismo , Alineación de Secuencia , Diferenciación Sexual/fisiología , Toxoplasma/efectos de los fármacos , Toxoplasma/crecimiento & desarrollo , Toxoplasma/metabolismo
15.
Cell Microbiol ; 16(5): 657-72, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23937520

RESUMEN

Apicomplexan parasites invade host cells by a conserved mechanism: parasite proteins are secreted from apical organelles, anchored in the host cell plasma membrane, and then interact with integral membrane proteins on the zoite surface to form the moving junction (MJ). The junction moves from the anterior to the posterior of the parasite resulting in parasite internalization into the host cell within a parasitophorous vacuole (PV). Conserved as well as coccidia-unique rhoptry neck proteins (RONs) have been described, some of which associate with the MJ. Here we report a novel RON, which we call RON12. RON12 is found only in Plasmodium and is highly conserved across the genus. RON12 lacks a membrane anchor and is a major soluble component of the nascent PV. The bulk of RON12 secretion happens late during invasion (after parasite internalization) allowing accumulation in the fully formed PV with a small proportion of RON12 also apparent occasionally in structures resembling the MJ. RON12, unlike most other RONs is not essential, but deletion of the gene does affect parasite proliferation. The data suggest that although the overall mechanism of invasion by Apicomplexan parasites is conserved, additional components depending on the parasite-host cell combination are required.


Asunto(s)
Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/metabolismo , Endocitosis , Eliminación de Gen , Plasmodium falciparum/genética , Proteínas Protozoarias/genética
16.
PLoS Pathog ; 8(8): e1002858, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22912579

RESUMEN

Malaria parasites reside inside erythrocytes and the disease manifestations are linked to the growth inside infected erythrocytes (IE). The growth of the parasite is mostly confined to the trophozoite stage during which nuclear division occurs followed by the formation of cell bodies (schizogony). The mechanism and regulation of schizogony are poorly understood. Here we show a novel role for a Plasmodium falciparum 60S stalk ribosomal acidic protein P2 (PfP2) (PFC0400w), which gets exported to the IE surface for 6-8 hrs during early schizogony, starting around 26-28 hrs post-merozoite invasion. The surface exposure is demonstrated using multiple PfP2-specific monoclonal antibodies, and is confirmed through transfection using PfP2-GFP. The IE surface-exposed PfP2-protein occurs mainly as SDS-resistant P2-homo-tetramers. Treatment with anti-PfP2 monoclonals causes arrest of IEs at the first nuclear division. Upon removal of the antibodies, about 80-85% of synchronized parasites can be released even after 24 hrs of antibody treatment. It has been reported that a tubovesicular network (TVN) is set up in early trophozoites which is used for nutrient import. Anti-P2 monoclonal antibodies cause a complete fragmentation of TVN by 36 hrs, and impairs lipid import in IEs. These may be downstream causes for the cell-cycle arrest. Upon antibody removal, the TVN is reconstituted, and the cell division progresses. Each of the above properties is observed in the rodent malaria parasite species P. yoelii and P. berghei. The translocation of the P2 protein to the IE surface is therefore likely to be of fundamental importance in Plasmodium cell division.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , División del Núcleo Celular/fisiología , Eritrocitos/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Ribosómicas/metabolismo , Animales , Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Antiprotozoarios/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , División del Núcleo Celular/efectos de los fármacos , Humanos , Ratones , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Proteínas Protozoarias/genética , Proteínas Ribosómicas/genética
17.
PLoS Pathog ; 8(2): e1002554, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22383885

RESUMEN

Cell-cycle progression is governed by a series of essential regulatory proteins. Two major regulators are cell-division cycle protein 20 (CDC20) and its homologue, CDC20 homologue 1 (CDH1), which activate the anaphase-promoting complex/cyclosome (APC/C) in mitosis, and facilitate degradation of mitotic APC/C substrates. The malaria parasite, Plasmodium, is a haploid organism which, during its life-cycle undergoes two stages of mitosis; one associated with asexual multiplication and the other with male gametogenesis. Cell-cycle regulation and DNA replication in Plasmodium was recently shown to be dependent on the activity of a number of protein kinases. However, the function of cell division cycle proteins that are also involved in this process, such as CDC20 and CDH1 is totally unknown. Here we examine the role of a putative CDC20/CDH1 in the rodent malaria Plasmodium berghei (Pb) using reverse genetics. Phylogenetic analysis identified a single putative Plasmodium CDC20/CDH1 homologue (termed CDC20 for simplicity) suggesting that Plasmodium APC/C has only one regulator. In our genetic approach to delete the endogenous cdc20 gene of P. berghei, we demonstrate that PbCDC20 plays a vital role in male gametogenesis, but is not essential for mitosis in the asexual blood stage. Furthermore, qRT-PCR analysis in parasite lines with deletions of two kinase genes involved in male sexual development (map2 and cdpk4), showed a significant increase in cdc20 transcription in activated gametocytes. DNA replication and ultra structural analyses of cdc20 and map2 mutants showed similar blockage of nuclear division at the nuclear spindle/kinetochore stage. CDC20 was phosphorylated in asexual and sexual stages, but the level of modification was higher in activated gametocytes and ookinetes. Changes in global protein phosphorylation patterns in the Δcdc20 mutant parasites were largely different from those observed in the Δmap2 mutant. This suggests that CDC20 and MAP2 are both likely to play independent but vital roles in male gametogenesis.


Asunto(s)
Proteínas de Ciclo Celular/genética , Gametogénesis/genética , Plasmodium malariae/genética , Proteínas de Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Animales , Proteínas Cdc20 , Proteínas Cdh1 , Genes Protozoarios/fisiología , Células Germinativas/metabolismo , Células Germinativas/fisiología , Cinetocoros/metabolismo , Cinetocoros/fisiología , Malaria/parasitología , Masculino , Ratones , Datos de Secuencia Molecular , Organismos Modificados Genéticamente , Filogenia , Plasmodium malariae/crecimiento & desarrollo , Plasmodium malariae/metabolismo , Plasmodium malariae/fisiología , Homología de Secuencia
18.
PLoS Pathog ; 8(9): e1002948, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23028336

RESUMEN

Protein phosphorylation and dephosphorylation (catalysed by kinases and phosphatases, respectively) are post-translational modifications that play key roles in many eukaryotic signalling pathways, and are often deregulated in a number of pathological conditions in humans. In the malaria parasite Plasmodium, functional insights into its kinome have only recently been achieved, with over half being essential for blood stage development and another 14 kinases being essential for sexual development and mosquito transmission. However, functions for any of the plasmodial protein phosphatases are unknown. Here, we use reverse genetics in the rodent malaria model, Plasmodium berghei, to examine the role of a unique protein phosphatase containing kelch-like domains (termed PPKL) from a family related to Arabidopsis BSU1. Phylogenetic analysis confirmed that the family of BSU1-like proteins including PPKL is encoded in the genomes of land plants, green algae and alveolates, but not in other eukaryotic lineages. Furthermore, PPKL was observed in a distinct family, separate to the most closely-related phosphatase family, PP1. In our genetic approach, C-terminal GFP fusion with PPKL showed an active protein phosphatase preferentially expressed in female gametocytes and ookinetes. Deletion of the endogenous ppkl gene caused abnormal ookinete development and differentiation, and dissociated apical microtubules from the inner-membrane complex, generating an immotile phenotype and failure to invade the mosquito mid-gut epithelium. These observations were substantiated by changes in localisation of cytoskeletal tubulin and actin, and the micronemal protein CTRP in the knockout mutant as assessed by indirect immunofluorescence. Finally, increased mRNA expression of dozi, a RNA helicase vital to zygote development was observed in ppkl(-) mutants, with global phosphorylation studies of ookinete differentiation from 1.5-24 h post-fertilisation indicating major changes in the first hours of zygote development. Our work demonstrates a stage-specific essentiality of the unique PPKL enzyme, which modulates parasite differentiation, motility and transmission.


Asunto(s)
Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/metabolismo , Plasmodium berghei/enzimología , Plasmodium berghei/crecimiento & desarrollo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Alveolados/química , Alveolados/genética , Secuencias de Aminoácidos , Animales , Anopheles/parasitología , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Secuencia de Bases , Diferenciación Celular , Genes Protozoarios , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas Fosfatasas/genética , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteínas Protozoarias/genética , Análisis de Secuencia de ADN , Viridiplantae/química
19.
J Infect Dis ; 208(3): 468-78, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23599312

RESUMEN

BACKGROUND: The mechanism of action of artemisinins against malaria is unclear, despite their widespread use in combination therapies and the emergence of resistance. RESULTS: Here, we report expression of PfATP6 (a SERCA pump) in yeast and demonstrate its inhibition by artemisinins. Mutations in PfATP6 identified in field isolates (such as S769N) and in laboratory clones (such as L263E) decrease susceptibility to artemisinins, whereas they increase susceptibility to unrelated inhibitors such as cyclopiazonic acid. As predicted from the yeast model, Plasmodium falciparum with the L263E mutation is also more susceptible to cyclopiazonic acid. An inability to knockout parasite SERCA pumps provides genetic evidence that they are essential in asexual stages of development. Thaperoxides are a new class of potent antimalarial designed to act by inhibiting PfATP6. Results in yeast confirm this inhibition. CONCLUSIONS: The identification of inhibitors effective against mutated PfATP6 suggests ways in which artemisinin resistance may be overcome.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , ATPasas Transportadoras de Calcio/genética , Resistencia a Medicamentos , Plasmodium falciparum/genética , Polimorfismo Genético , Expresión Génica , Humanos , Pruebas de Sensibilidad Parasitaria/métodos , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética
20.
PLoS Pathog ; 7(2): e1001288, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21379566

RESUMEN

Plasmodium yoelii YM asexual blood stage parasites express multiple members of the py235 gene family, part of the super-family of genes including those coding for Plasmodium vivax reticulocyte binding proteins and Plasmodium falciparum RH proteins. We previously identified a Py235 erythrocyte binding protein (Py235EBP-1, encoded by the PY01365 gene) that is recognized by protective mAb 25.77. Proteins recognized by a second protective mAb 25.37 have been identified by mass spectrometry and are encoded by two genes, PY01185 and PY05995/PY03534. We deleted the PY01365 gene and examined the phenotype. The expression of the members of the py235 family in both the WT and gene deletion parasites was measured by quantitative RT-PCR and RNA-Seq. py235ebp-1 expression was undetectable in the knockout parasite, but transcription of other members of the family was essentially unaffected. The knockout parasites continued to react with mAb 25.77; and the 25.77-binding proteins in these parasites were the PY01185 and PY05995/PY03534 products. The PY01185 product was also identified as erythrocyte binding. There was no clear change in erythrocyte invasion profile suggesting that the PY01185 gene product (designated PY235EBP-2) is able to fulfill the role of EBP-1 by serving as an invasion ligand although the molecular details of its interaction with erythrocytes have not been examined. The PY01365, PY01185, and PY05995/PY03534 genes are part of a distinct subset of the py235 family. In P. falciparum, the RH protein genes are under epigenetic control and expression correlates with binding to distinct erythrocyte receptors and specific invasion pathways, whereas in P. yoelii YM all the genes are expressed and deletion of one does not result in upregulation of another. We propose that simultaneous expression of multiple Py235 ligands enables invasion of a wide range of host erythrocytes even in the presence of antibodies to one or more of the proteins and that this functional redundancy at the protein level gives the parasite phenotypic plasticity in the absence of differences in gene expression.


Asunto(s)
Empalme Alternativo , Antígenos de Protozoos/genética , Eritrocitos/parasitología , Eliminación de Gen , Malaria/genética , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/patogenicidad , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales , Southern Blotting , Western Blotting , Recuento de Eritrocitos , Eritrocitos/inmunología , Eritrocitos/metabolismo , Técnica del Anticuerpo Fluorescente , Genoma de Protozoos , Inmunoprecipitación , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Datos de Secuencia Molecular , Familia de Multigenes , Plasmodium yoelii/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA