Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(6): e1012360, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38935780

RESUMO

The cGMP-dependent protein kinase (PKG) is the sole cGMP sensor in malaria parasites, acting as an essential signalling hub to govern key developmental processes throughout the parasite life cycle. Despite the importance of PKG in the clinically relevant asexual blood stages, many aspects of malarial PKG regulation, including the importance of phosphorylation, remain poorly understood. Here we use genetic and biochemical approaches to show that reduced cGMP binding to cyclic nucleotide binding domain B does not affect in vitro kinase activity but prevents parasite egress. Similarly, we show that phosphorylation of a key threonine residue (T695) in the activation loop is dispensable for kinase activity in vitro but is essential for in vivo PKG function, with loss of T695 phosphorylation leading to aberrant phosphorylation events across the parasite proteome and changes to the substrate specificity of PKG. Our findings indicate that Plasmodium PKG is uniquely regulated to transduce signals crucial for malaria parasite development.

3.
Dis Model Mech ; 16(2)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36715290

RESUMO

Phenotypic cell-based screens are critical tools for discovering candidate drugs for development, yet identification of the cellular target and mode of action of a candidate drug is often lacking. Using an imaging-based screen, we recently discovered an N-[(4-hydroxychroman-4-yl)methyl]-sulphonamide (N-4HCS) compound, DDD01035881, that blocks male gamete formation in the malaria parasite life cycle and subsequent transmission of the parasite to the mosquito with nanomolar activity. To identify the target(s) of DDD01035881, and of the N-4HCS class of compounds more broadly, we synthesised a photoactivatable derivative, probe 2. Photoaffinity labelling of probe 2 coupled with mass spectrometry identified the 16 kDa Plasmodium falciparum parasitophorous vacuole membrane protein Pfs16 as a potential parasite target. Complementary methods including cellular thermal shift assays confirmed that the parent molecule DDD01035881 stabilised Pfs16 in lysates from activated mature gametocytes. Combined with high-resolution, fluorescence and electron microscopy data, which demonstrated that parasites inhibited with N-4HCS compounds phenocopy the targeted deletion of Pfs16 in gametocytes, these data implicate Pfs16 as a likely target of DDD01035881. This finding establishes N-4HCS compounds as being flexible and effective starting candidates from which transmission-blocking antimalarials can be developed in the future.


Assuntos
Malária , Plasmodium , Animais , Masculino , Proteínas de Membrana/metabolismo , Vacúolos/metabolismo , Sulfonamidas/farmacologia , Sulfonamidas/uso terapêutico , Sulfonamidas/metabolismo
4.
Mol Biochem Parasitol ; 243: 111374, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33974939

RESUMO

Recent studies highlight the emerging role of lipids as important messengers in malaria parasite biology. In an attempt to identify interacting proteins and regulators of these dynamic and versatile molecules, we hypothesised the involvement of phospholipid translocases and their substrates in the infection of the host erythrocyte by the malaria parasite Plasmodium spp. Here, using a data base searching approach of the Plasmodium Genomics Resources (www.plasmodb.org), we have identified a putative phospholipid (PL) scramblase in P. falciparum (PfPLSCR) that is conserved across the genus and in closely related unicellular algae. By reconstituting recombinant PfPLSCR into liposomes, we demonstrate metal ion dependent PL translocase activity and substrate preference, confirming PfPLSCR as a bona fide scramblase. We show that PfPLSCR is expressed during asexual and sexual parasite development, localising to different membranous compartments of the parasite throughout the intra-erythrocytic life cycle. Two different gene knockout approaches, however, suggest that PfPLSCR is not essential for erythrocyte invasion and asexual parasite development, pointing towards a possible role in other stages of the parasite life cycle.


Assuntos
Proteínas de Transferência de Fosfolipídeos/genética , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/genética , Sequência de Aminoácidos , Apicomplexa , Sequência Conservada , Eritrócitos/parasitologia , Regulação Enzimológica da Expressão Gênica , Humanos , Lipossomos/química , Lipossomos/metabolismo , Microrganismos Geneticamente Modificados , Proteínas de Transferência de Fosfolipídeos/isolamento & purificação , Proteínas de Transferência de Fosfolipídeos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
5.
PLoS Pathog ; 16(10): e1009007, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33104759

RESUMO

All symptoms of malaria disease are associated with the asexual blood stages of development, involving cycles of red blood cell (RBC) invasion and egress by the Plasmodium spp. merozoite. Merozoite invasion is rapid and is actively powered by a parasite actomyosin motor. The current accepted model for actomyosin force generation envisages arrays of parasite myosins, pushing against short actin filaments connected to the external milieu that drive the merozoite forwards into the RBC. In Plasmodium falciparum, the most virulent human malaria species, Myosin A (PfMyoA) is critical for parasite replication. However, the precise function of PfMyoA in invasion, its regulation, the role of other myosins and overall energetics of invasion remain unclear. Here, we developed a conditional mutagenesis strategy combined with live video microscopy to probe PfMyoA function and that of the auxiliary motor PfMyoB in invasion. By imaging conditional mutants with increasing defects in force production, based on disruption to a key PfMyoA phospho-regulation site, the absence of the PfMyoA essential light chain, or complete motor absence, we define three distinct stages of incomplete RBC invasion. These three defects reveal three energetic barriers to successful entry: RBC deformation (pre-entry), mid-invasion initiation, and completion of internalisation, each requiring an active parasite motor. In defining distinct energetic barriers to invasion, these data illuminate the mechanical challenges faced in this remarkable process of protozoan parasitism, highlighting distinct myosin functions and identifying potential targets for preventing malaria pathogenesis.


Assuntos
Actomiosina/metabolismo , Eritrócitos/fisiologia , Plasmodium falciparum/metabolismo , Citoesqueleto de Actina/metabolismo , Actomiosina/fisiologia , Animais , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Humanos , Malária/metabolismo , Malária/fisiopatologia , Malária Falciparum/parasitologia , Merozoítos/metabolismo , Miosinas/metabolismo , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIA/fisiologia , Parasitos/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo
6.
Elife ; 92020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33046215

RESUMO

Parasites from the genus Plasmodium are the causative agents of malaria. The mobility, infectivity, and ultimately pathogenesis of Plasmodium falciparum rely on a macromolecular complex, called the glideosome. At the core of the glideosome is an essential and divergent Myosin A motor (PfMyoA), a first order drug target against malaria. Here, we present the full-length structure of PfMyoA in two states of its motor cycle. We report novel interactions that are essential for motor priming and the mode of recognition of its two light chains (PfELC and MTIP) by two degenerate IQ motifs. Kinetic and motility assays using PfMyoA variants, along with molecular dynamics, demonstrate how specific priming and atypical sequence adaptations tune the motor's mechano-chemical properties. Supported by evidence for an essential role of the PfELC in malaria pathogenesis, these structures provide a blueprint for the design of future anti-malarials targeting both the glideosome motor and its regulatory elements.


Assuntos
Antimaláricos/farmacologia , Miosina não Muscular Tipo IIA/química , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/química , Plasmodium falciparum/metabolismo
7.
Sci Rep ; 8(1): 10165, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29976932

RESUMO

Plasmodium knowlesi, a zoonotic parasite causing severe-to-lethal malaria disease in humans, has only recently been adapted to continuous culture with human red blood cells (RBCs). In comparison with the most virulent human malaria, Plasmodium falciparum, there are, however, few cellular tools available to study its biology, in particular direct investigation of RBC invasion by blood-stage P. knowlesi merozoites. This leaves our current understanding of biological differences across pathogenic Plasmodium spp. incomplete. Here, we report a robust method for isolating viable and invasive P. knowlesi merozoites to high purity and yield. Using this approach, we present detailed comparative dissection of merozoite invasion (using a variety of microscopy platforms) and direct assessment of kinetic differences between knowlesi and falciparum merozoites. We go on to assess the inhibitory potential of molecules targeting discrete steps of invasion in either species via a quantitative invasion inhibition assay, identifying a class of polysulfonate polymer able to efficiently inhibit invasion in both, providing a foundation for pan-Plasmodium merozoite inhibitor development. Given the close evolutionary relationship between P. knowlesi and P. vivax, the second leading cause of malaria-related morbidity, this study paves the way for inter-specific dissection of invasion by all three major pathogenic malaria species.


Assuntos
Eritrócitos/patologia , Eritrócitos/parasitologia , Malária/parasitologia , Merozoítos/patogenicidade , Parasitos/patogenicidade , Plasmodium knowlesi/patogenicidade , Animais , Sobrevivência Celular , Eritrócitos/efeitos dos fármacos , Eritrócitos/ultraestrutura , Filtração , Humanos , Cinética , Merozoítos/isolamento & purificação , Merozoítos/ultraestrutura , Parasitos/efeitos dos fármacos , Parasitos/crescimento & desenvolvimento , Parasitos/ultraestrutura , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium knowlesi/efeitos dos fármacos , Plasmodium knowlesi/crescimento & desenvolvimento , Plasmodium knowlesi/ultraestrutura , Polímeros/farmacologia , Sulfonas/farmacologia
8.
Mol Biol Cell ; 26(17): 3001-12, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26157165

RESUMO

Proteins of the actin-depolymerizing factor (ADF)/cofilin family have been shown to be crucial for the motility and survival of apicomplexan parasites. However, the mechanisms by which ADF proteins fulfill their function remain poorly understood. In this study, we investigate the comparative activities of ADF proteins from Toxoplasma gondii and Plasmodium falciparum, the human malaria parasite, using a conditional T. gondii ADF-knockout line complemented with ADF variants from either species. We show that P. falciparum ADF1 can fully restore native TgADF activity, demonstrating functional conservation between parasites. Strikingly, mutation of a key basic residue (Lys-72), previously implicated in disassembly in PfADF1, had no detectable phenotypic effect on parasite growth, motility, or development. In contrast, organelle segregation was severely impaired when complementing with a TgADF mutant lacking the corresponding residue (Lys-68). Biochemical analyses of each ADF protein confirmed the reduced ability of lysine mutants to mediate actin depolymerization via filament disassembly although not severing, in contrast to previous reports. These data suggest that actin filament disassembly is essential for apicomplexan parasite development but not for motility, as well as pointing to genus-specific coevolution between ADF proteins and their native actin.


Assuntos
Destrina/metabolismo , Plasmodium falciparum/metabolismo , Toxoplasma/metabolismo , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Destrina/genética , Técnicas de Inativação de Genes , Estudos de Associação Genética , Lisina/metabolismo
9.
Mol Microbiol ; 89(6): 1167-86, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23869529

RESUMO

Plasmodium parasites remodel their vertebrate host cells by translocating hundreds of proteins across an encasing membrane into the host cell cytosol via a putative export machinery termed PTEX. Previously PTEX150, HSP101 and EXP2 have been shown to be bona fide members of PTEX. Here we validate that PTEX88 and TRX2 are also genuine members of PTEX and provide evidence that expression of PTEX components are also expressed in early gametocytes, mosquito and liver stages, consistent with observations that protein export is not restricted to asexual stages. Although amenable to genetic tagging, HSP101, PTEX150, EXP2 and PTEX88 could not be genetically deleted in Plasmodium berghei, in keeping with the obligatory role this complex is postulated to have in maintaining normal blood-stage growth. In contrast, the putative thioredoxin-like protein TRX2 could be deleted, with knockout parasites displaying reduced grow-rates, both in vivo and in vitro, and reduced capacity to cause severe disease in a cerebral malaria model. Thus, while not essential for parasite survival, TRX2 may help to optimize PTEX activity. Importantly, the generation of TRX2 knockout parasites that display altered phenotypes provides a much-needed tool to dissect PTEX function.


Assuntos
Parasitemia/parasitologia , Plasmodium berghei/enzimologia , Plasmodium berghei/patogenicidade , Tiorredoxinas/metabolismo , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Deleção de Genes , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Análise de Sobrevida , Tiorredoxinas/genética , Virulência , Fatores de Virulência/genética
10.
PLoS One ; 8(4): e61482, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23658610

RESUMO

Protein export into the host red blood cell is one of the key processes in the pathobiology of the malaria parasite Plasmodiumtrl falciparum, which extensively remodels the red blood cell to ensure its virulence and survival. In this study, we aimed to shed further light on the protein export mechanisms in the rodent malaria parasite P. berghei and provide further proof of the conserved nature of host cell remodeling in Plasmodium spp. Based on the presence of an export motif (R/KxLxE/Q/D) termed PEXEL (Plasmodium export element), we have generated transgenic P. berghei parasite lines expressing GFP chimera of putatively exported proteins and analysed one of the newly identified exported proteins in detail. This essential protein, termed PbCP1 (P. berghei Cleft-like Protein 1), harbours an atypical PEXEL motif (RxLxY) and is further characterised by two predicted transmembrane domains (2TMD) in the C-terminal end of the protein. We have functionally validated the unusual PEXEL motif in PbCP1 and analysed the role of the 2TMD region, which is required to recruit PbCP1 to discrete membranous structures in the red blood cell cytosol that have a convoluted, vesico-tubular morphology by electron microscopy. Importantly, this study reveals that rodent malaria species also induce modifications to their host red blood cell.


Assuntos
Citosol/parasitologia , Eritrócitos/parasitologia , Plasmodium berghei/metabolismo , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Citosol/ultraestrutura , Eritrócitos/ultraestrutura , Expressão Gênica , Proteínas de Fluorescência Verde , Estágios do Ciclo de Vida/genética , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium berghei/genética , Plasmodium berghei/ultraestrutura , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
11.
Cell Microbiol ; 12(5): 580-7, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20180801

RESUMO

In order to survive and promote its virulence the malaria parasite must export hundreds of its proteins beyond an encasing vacuole and membrane into the host red blood cell. In the last few years, several major advances have been made that have significantly contributed to our understanding of this export process. These include: (i) the identification of sequences that direct protein export (a signal sequence and a motif termed PEXEL), which have allowed predictions of the exportomes of Plasmodium species that are the cause of malaria, (ii) the recognition that the fate of proteins destined for export is already decided within the parasite's endoplasmic reticulum and involves the PEXEL motif being recognized and cleaved by the aspartic protease plasmepsin V and (iii) the discovery of the Plasmodium translocon of exported proteins (PTEX) that is responsible for the passage of proteins across the vacuolar membrane. We review protein export in Plasmodium and these latest developments in the field that have now provided a new platform from which trafficking of malaria proteins can be dissected.


Assuntos
Plasmodium/metabolismo , Proteínas de Protozoários/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Eritrócitos/parasitologia , Humanos , Sinais Direcionadores de Proteínas , Transporte Proteico
12.
Nat Biotechnol ; 28(1): 91-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20037583

RESUMO

Functions have yet to be defined for the majority of genes of Plasmodium falciparum, the agent responsible for the most serious form of human malaria. Here we report changes in P. falciparum gene expression induced by 20 compounds that inhibit growth of the schizont stage of the intraerythrocytic development cycle. In contrast with previous studies, which reported only minimal changes in response to chemically induced perturbations of P. falciparum growth, we find that approximately 59% of its coding genes display over three-fold changes in expression in response to at least one of the chemicals we tested. We use this compendium for guilt-by-association prediction of protein function using an interaction network constructed from gene co-expression, sequence homology, domain-domain and yeast two-hybrid data. The subcellular localizations of 31 of 42 proteins linked with merozoite invasion is consistent with their role in this process, a key target for malaria control. Our network may facilitate identification of novel antimalarial drugs and vaccines.


Assuntos
Perfilação da Expressão Gênica , Malária Falciparum/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/genética , Transcrição Gênica , Algoritmos , Animais , Antimaláricos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Humanos , Cadeias de Markov , Merozoítos/efeitos dos fármacos , Merozoítos/metabolismo , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Transcrição Gênica/efeitos dos fármacos
13.
PLoS Pathog ; 5(3): e1000322, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19283086

RESUMO

A key process in the lifecycle of the malaria parasite Plasmodium falciparum is the fast invasion of human erythrocytes. Entry into the host cell requires the apical membrane antigen 1 (AMA-1), a type I transmembrane protein located in the micronemes of the merozoite. Although AMA-1 is evolving into the leading blood-stage malaria vaccine candidate, its precise role in invasion is still unclear. We investigate AMA-1 function using live video microscopy in the absence and presence of an AMA-1 inhibitory peptide. This data reveals a crucial function of AMA-1 during the primary contact period upstream of the entry process at around the time of moving junction formation. We generate a Plasmodium falciparum cell line that expresses a functional GFP-tagged AMA-1. This allows the visualization of the dynamics of AMA-1 in live parasites. We functionally validate the ectopically expressed AMA-1 by establishing a complementation assay based on strain-specific inhibition. This method provides the basis for the functional analysis of essential genes that are refractory to any genetic manipulation. Using the complementation assay, we show that the cytoplasmic domain of AMA-1 is not required for correct trafficking and surface translocation but is essential for AMA-1 function. Although this function can be mimicked by the highly conserved cytoplasmic domains of P. vivax and P. berghei, the exchange with the heterologous domain of the microneme protein EBA-175 or the rhoptry protein Rh2b leads to a loss of function. We identify several residues in the cytoplasmic tail that are essential for AMA-1 function. We validate this data using additional transgenic parasite lines expressing AMA-1 mutants with TY1 epitopes. We show that the cytoplasmic domain of AMA-1 is phosphorylated. Mutational analysis suggests an important role for the phosphorylation in the invasion process, which might translate into novel therapeutic strategies.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Vacinas Antimaláricas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/genética , Western Blotting , Citoplasma/metabolismo , Imunofluorescência , Humanos , Immunoblotting , Imunoprecipitação , Vacinas Antimaláricas/genética , Proteínas de Membrana/genética , Microscopia Confocal , Dados de Sequência Molecular , Fosforilação , Reação em Cadeia da Polimerase , Proteínas de Protozoários/genética , Transgenes
14.
Mol Microbiol ; 71(4): 1003-17, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19170882

RESUMO

A short motif termed Plasmodium export element (PEXEL) or vacuolar targeting signal (VTS) characterizes Plasmodium proteins exported into the host cell. These proteins mediate host cell modifications essential for parasite survival and virulence. However, several PEXEL-negative exported proteins indicate that the currently predicted malaria exportome is not complete and it is unknown whether and how these proteins relate to PEXEL-positive export. Here we show that the N-terminal 10 amino acids of the PEXEL-negative exported protein REX2 (ring-exported protein 2) are necessary for its targeting and that a single-point mutation in this region abolishes export. Furthermore we show that the REX2 transmembrane domain is also essential for export and that together with the N-terminal region it is sufficient to promote export of another protein. An N-terminal region and the transmembrane domain of the unrelated PEXEL-negative exported protein SBP1 (skeleton-binding protein 1) can functionally replace the corresponding regions in REX2, suggesting that these sequence features are also present in other PEXEL-negative exported proteins. Similar to PEXEL proteins we find that REX2 is processed, but in contrast, detect no evidence for N-terminal acetylation.


Assuntos
Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Eritrócitos/parasitologia , Proteínas de Membrana/genética , Dados de Sequência Molecular , Plasmodium falciparum/genética , Mutação Puntual , Transporte Proteico , Proteínas de Protozoários/genética , Alinhamento de Sequência , Deleção de Sequência
15.
J Cell Sci ; 121(Pt 13): 2123-9, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18522993

RESUMO

Plasmodium falciparum, the causative agent of malaria, relies on a complex protein-secretion system for protein targeting into numerous subcellular destinations. Recently, a homologue of the Golgi re-assembly stacking protein (GRASP) was identified and used to characterise the Golgi organisation in this parasite. Here, we report on the presence of a splice variant that leads to the expression of a GRASP isoform. Although the first GRASP protein (GRASP1) relies on a well-conserved myristoylation motif, the variant (GRASP2) displays a different N-terminus, similar to GRASPs found in fungi. Phylogenetic analyses between GRASP proteins of numerous taxa point to an independent evolution of the unusual N-terminus that could reflect unique requirements for Golgi-dependent protein sorting and organelle biogenesis in P. falciparum. Golgi association of GRASP2 depends on the hydrophobic N-terminus that resembles a signal anchor, leading to a unique mode of Golgi targeting and membrane attachment.


Assuntos
Evolução Molecular , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Células Eucarióticas , Proteínas da Matriz do Complexo de Golgi , Proteínas de Membrana/genética , Dados de Sequência Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestrutura , Transporte Proteico/genética
16.
Mol Microbiol ; 67(6): 1320-30, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18284574

RESUMO

The Golgi apparatus forms the heart of the secretory pathway in eukaryotic cells where proteins are modified, processed and sorted. The transport of proteins from the endoplasmic reticulum (ER) to the cis-side of the Golgi complex takes place at specialized ER sub-domains known as transitional ER (tER). We used the Plasmodium falciparum orthologue of Sec13p to analyse tER organization. We show that the distribution of PfSec13p is restricted to defined areas of the ER membrane. These foci are juxtaposed to the Golgi apparatus and might represent tER sites. To further analyse cis- to trans-Golgi architecture, we generated a double transfectant parasite line that expresses the Golgi marker Golgi reassembly stacking protein (GRASP) as a green fluorescent protein fusion and the trans-Golgi marker Rab6 as a DsRed fusion protein. Our data demonstrate that Golgi multiplication is closely linked to tER multiplication, and that parasite maturation is accompanied by the spatial separation of the cis- and trans- face of this organelle.


Assuntos
Complexo de Golgi/metabolismo , Plasmodium falciparum/metabolismo , Animais , Western Blotting , Células Cultivadas , Retículo Endoplasmático/metabolismo , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Immunoblotting , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
17.
Infect Immun ; 76(3): 879-87, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18174339

RESUMO

One of the key processes in the pathobiology of the malaria parasite is the invasion and subsequent modification of the human erythrocyte. In this complex process, an unknown number of parasite proteins are involved, some of which are leading vaccine candidates. The majority of the proteins that play pivotal roles in invasion are either stored in the apical secretory organelles or located on the surface of the merozoite, the invasive stage of the parasite. Using transcriptional and structural features of these known proteins, we performed a genomewide search that identified 49 hypothetical proteins with a high probability of being located on the surface of the merozoite or in the secretory organelles. Of these candidates, we characterized a novel leucine zipper-like protein in Plasmodium falciparum that is conserved in Plasmodium spp. This protein is expressed in late blood stages and localizes to the rhoptries of the parasite. We demonstrate that this Plasmodium sp.-specific protein has a high degree of conservation within field isolates and that it is refractory to gene knockout attempts and thus might play an important role in invasion.


Assuntos
Zíper de Leucina , Merozoítos/química , Organelas/química , Plasmodium falciparum/química , Proteínas de Protozoários/análise , Proteínas de Protozoários/genética , Animais , Western Blotting , Deleção de Genes , Genes Essenciais , Humanos , Malária Falciparum/parasitologia , Merozoítos/fisiologia , Microscopia de Fluorescência , Mutagênese Insercional , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/fisiologia
18.
J Biol Chem ; 281(42): 31995-2003, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16935855

RESUMO

The proliferation of the malaria parasite Plasmodium falciparum within the human host is dependent upon invasion of erythrocytes. This process is accomplished by the merozoite, a highly specialized form of the parasite. Secretory organelles including micronemes and rhoptries play a pivotal role in the invasion process by storing and releasing parasite proteins. The mechanism of protein sorting to these compartments is unclear. Using a transgenic approach we show that trafficking of the most abundant micronemal proteins (members of the EBL-family: EBA-175, EBA-140/BAEBL, and EBA-181/JSEBL) is independent of their cytoplasmic and transmembrane domains, respectively. To identify the minimal sequence requirements for microneme trafficking, we generated parasites expressing EBA-GFP chimeric proteins and analyzed their distribution within the infected erythrocyte. This revealed that: (i) a conserved cysteine-rich region in the ectodomain is necessary for protein trafficking to the micronemes and (ii) correct sorting is dependent on accurate timing of expression.


Assuntos
Antígenos de Protozoários/metabolismo , Proteínas de Transporte/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Transporte Biológico , Sequência Conservada , Citoplasma/metabolismo , Complexo de Golgi/metabolismo , Humanos , Proteínas de Membrana , Microscopia de Fluorescência , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...