Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Microbiol ; 23(8): e13332, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33774908

RESUMO

During its intraerythrocytic life cycle, the human malaria parasite Plasmodium falciparum supplements its nutritional requirements by scavenging substrates from the plasma through the new permeability pathways (NPPs) installed in the red blood cell (RBC) membrane. Parasite proteins of the RhopH complex: CLAG3, RhopH2, RhopH3, have been implicated in NPP activity. Here, we studied 13 exported proteins previously hypothesised to interact with RhopH2, to study their potential contribution to the function of NPPs. NPP activity assays revealed that the 13 proteins do not appear to be individually important for NPP function, as conditional knockdown of these proteins had no effect on sorbitol uptake. Intriguingly, reciprocal immunoprecipitation assays showed that five of the 13 proteins interact with all members of the RhopH complex, with PF3D7_1401200 showing the strongest association. Mass spectrometry-based proteomics further identified new protein complexes; a cytoskeletal complex and a Maurer's clefts/J-dot complex, which overall helps clarify protein-protein interactions within the infected RBC (iRBC) and is suggestive of the potential trafficking route of the RhopH complex itself to the RBC membrane.


Assuntos
Parasitos , Plasmodium falciparum , Animais , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Humanos , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Transporte Proteico , Proteínas de Protozoários/metabolismo
2.
Cell Microbiol ; 22(2): e13123, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31652487

RESUMO

A hallmark of the biology of Plasmodium falciparum blood stage parasites is their extensive host cell remodelling, facilitated by parasite proteins that are exported into the erythrocyte. Although this area has received extensive attention, only a few exported parasite proteins have been analysed in detail, and much of this remodelling process remains unknown, particularly for gametocyte development. Recent advances to induce high rates of sexual commitment enable the production of large numbers of gametocytes. We used this approach to study the Plasmodium helical interspersed subtelomeric (PHIST) protein GEXP02, which is expressed during sexual development. We show by immunofluorescence that GEXP02 is exported to the gametocyte-infected host cell periphery. Co-immunoprecipitation revealed potential interactions between GEXP02 and components of the erythrocyte cytoskeleton as well as other exported parasite proteins. This indicates that GEXP02 targets the erythrocyte cytoskeleton and is likely involved in its remodelling. GEXP02 knock-out parasites show no obvious phenotype during gametocyte maturation, transmission through mosquitoes, and hepatocyte infection, suggesting auxiliary or redundant functions for this protein. In summary, we performed a detailed cellular and biochemical analysis of a sexual stage-specific exported parasite protein using a novel experimental approach that is broadly applicable to study the biology of P. falciparum gametocytes.


Assuntos
Membrana Eritrocítica/metabolismo , Células Germinativas/citologia , Malária Falciparum/parasitologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/fisiologia , Interações Hospedeiro-Parasita , Humanos
3.
Biochem Biophys Res Commun ; 456(1): 403-9, 2015 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-25475729

RESUMO

Survival of the malaria parasite Plasmodium falciparum when it infects red blood cells depends upon its ability to export hundreds of its proteins beyond an encasing vacuole. Protein export is mediated by a parasite-derived protein complex, the Plasmodium translocon of exported proteins (PTEX), and requires unfolding of the different cargos prior to their translocation across the vacuolar membrane. Unfolding is performed by the AAA+protein unfoldase HSP101/ClpB2 and the thioredoxin-2 enzyme (TRX2). Protein trafficking is dramatically impaired in parasites with defective HSP101 or lacking TRX2. These two PTEX subunits drive export and are targets for the design of a novel class of antimalarials: protein export inhibitors. To rationalize inhibitor design, we solved the crystal structure of Pfal-TRX2 at 2.2-Å resolution. Within the asymmetric unit, the three different copies of this protein disulfide reductase sample its two redox catalytic states. Size exclusion chromatography and small-angle X-ray scattering (SAXS) analyses demonstrate that Pfal-TRX2 is monomeric in solution. A non-conserved N-terminal extension precedes the canonical thioredoxin-fold; although it is not observed in our structure, our solution analysis suggests it is flexible in contrast to Plasmodium thioredoxin-1. This represents a first step towards the reconstitution of the entire PTEX for mechanistic and structural studies.


Assuntos
Plasmodium falciparum/química , Proteínas de Protozoários/química , Tiorredoxinas/química , Sequência de Aminoácidos , Animais , Antimaláricos/química , Cromatografia em Gel , Cristalografia por Raios X , Escherichia coli/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Transporte Proteico , Homologia de Sequência de Aminoácidos
4.
FASEB J ; 28(7): 3103-13, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24706359

RESUMO

The genomes of malaria parasites (Plasmodium spp.) contain a family of genes encoding proteins with a Plasmodium helical interspersed subtelomeric (PHIST) domain, most of which are predicted to be exported into the parasite-infected human red blood cell (iRBC). Here, using transgenic parasites and a combination of cellular, biochemical, and biophysical assays, we have characterized and determined the function of a novel member of the PHIST protein family in Plasmodium falciparum, termed lysine-rich membrane-associated PHISTb (LyMP). LyMP was shown to associate directly with the cytoskeleton of iRBCs where it plays a role in their abnormal ability to adhere to a protein expressed on vascular endothelial cells, resulting in sequestration. Deletion of LyMP dramatically reduced adhesion of iRBCs to CD36 by 55%, which was completely restored to wild-type levels on complementation. Intriguingly, in the absence of LyMP, formation of RBC membrane knobs and the level of surface exposure of the parasites' major cytoadhesive ligand, PfEMP1, were identical to those for the parental parasite line, demonstrating for the first time an additional mechanism that enhances cytoadherence of iRBCs beyond those already recognized. Our findings identify LyMP as a previously unknown RBC cytoskeletal-binding protein that is likely to be of major significance in the complex pathophysiology of falciparum malaria.-Proellocks, N. I., Herrmann, S., Buckingham, D. W., Hanssen, E., Hodges, E. K., Elsworth, B., Morahan, B. J., Coppel, R. L., Cooke, B. M. A lysine-rich membrane-associated PHISTb protein involved in alteration of the cytoadhesive properties of Plasmodium falciparum infected red blood cells.


Assuntos
Adesão Celular/fisiologia , Citoesqueleto/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Lisina/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Membrana Celular/metabolismo , Membrana Celular/parasitologia , Citoesqueleto/parasitologia , Endotélio Vascular/metabolismo , Endotélio Vascular/parasitologia , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Proteínas de Membrana/metabolismo , Ligação Proteica/fisiologia
5.
FASEB J ; 28(10): 4420-33, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24983468

RESUMO

Uniquely among malaria parasites, Plasmodium falciparum-infected erythrocytes (iRBCs) develop membrane protrusions, known as knobs, where the parasite adhesion receptor P. falciparum erythrocyte membrane protein 1 (PfEMP1) clusters. Knob formation and the associated iRBC adherence to host endothelium are directly linked to the severity of malaria and are functional manifestations of protein export from the parasite to the iRBC. A family of exported proteins featuring Plasmodium helical interspersed subtelomeric (PHIST) domains has attracted attention, with members being implicated in host-parasite protein interactions and differentially regulated in severe disease and among parasite isolates. Here, we show that PHIST member PFE1605w binds the PfEMP1 intracellular segment directly with Kd = 5 ± 0.6 µM, comigrates with PfEMP1 during export, and locates in knobs. PHIST variants that do not locate in knobs (MAL8P1.4) or bind PfEMP1 30 times more weakly (PFI1780w) used as controls did not display the same pattern. We resolved the first crystallographic structure of a PHIST protein and derived a partial model of the PHIST-PfEMP1 interaction from nuclear magnetic resonance. We propose that PFE1605w reinforces the PfEMP1-cytoskeletal connection in knobs and discuss the possible role of PHIST proteins as interaction hubs in the parasite exportome.


Assuntos
Proteínas de Transporte/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/química , Membrana Celular/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Humanos , Dados de Sequência Molecular , Plasmodium falciparum/química , Plasmodium falciparum/patogenicidade , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas de Protozoários/química
6.
Curr Protein Pept Sci ; 25(6): 427-437, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38409726

RESUMO

The apicomplexan pathogenic parasite 'Plasmodium falciparum' (Pf) is responsible for most of the malaria related mortality. It resides in and refurbishes the infected red blood cells (iRBCs) for its own survival and to suffice its metabolic needs. Remodeling of host erythrocytes involves alteration of physical and biochemical properties of the membrane and genesis of new parasite induced structures within the iRBCs. The generated structures include knobs and solute ion channels on the erythrocyte surface and specialized organelles i.e. Maurer's clefts (MCs) in the iRBC cytosol. The above processes are mediated by exporting a large repertoire of proteins to the host cell, most of which are transported via MCs, the sorting stations in parasitized erythrocytes. Information about MC biogenesis and the molecules involved in maintaining MC architecture remains incompletely elucidated. Here, we have compiled a list of experimentally known MC resident proteins, several of which have roles in maintaining its architecture and function. Our short review covers available data on the domain organization, orthologues, topology and specific roles of these proteins. We highlight the current knowledge gaps in our understanding of MCs as crucial organelles involved in parasite biology and disease pathogenesis.


Assuntos
Eritrócitos , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Plasmodium falciparum/metabolismo , Humanos , Eritrócitos/parasitologia , Eritrócitos/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Malária Falciparum/parasitologia , Malária Falciparum/metabolismo , Animais , Interações Hospedeiro-Parasita , Transporte Proteico , Membrana Eritrocítica/parasitologia , Membrana Eritrocítica/metabolismo
7.
Sci Rep ; 14(1): 11242, 2024 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755230

RESUMO

The interaction of Plasmodium falciparum-infected red blood cells (iRBCs) with the vascular endothelium plays a crucial role in malaria pathology and disease. KAHRP is an exported P. falciparum protein involved in iRBC remodelling, which is essential for the formation of protrusions or "knobs" on the iRBC surface. These knobs and the proteins that are concentrated within them allow the parasites to escape the immune response and host spleen clearance by mediating cytoadherence of the iRBC to the endothelial wall, but this also slows down blood circulation, leading in some cases to severe cerebral and placental complications. In this work, we have applied genetic and biochemical tools to identify proteins that interact with P. falciparum KAHRP using enhanced ascorbate peroxidase 2 (APEX2) proximity-dependent biotinylation and label-free shotgun proteomics. A total of 30 potential KAHRP-interacting candidates were identified, based on the assigned fragmented biotinylated ions. Several identified proteins have been previously reported to be part of the Maurer's clefts and knobs, where KAHRP resides. This study may contribute to a broader understanding of P. falciparum protein trafficking and knob architecture and shows for the first time the feasibility of using APEX2-proximity labelling in iRBCs.


Assuntos
Eritrócitos , Plasmodium falciparum , Proteômica , Proteínas de Protozoários , Eritrócitos/parasitologia , Eritrócitos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Humanos , Proteômica/métodos , Malária Falciparum/parasitologia , Malária Falciparum/metabolismo , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Ascorbato Peroxidases/metabolismo , Ligação Proteica , Biotinilação , Endonucleases , Peptídeos , Proteínas , Enzimas Multifuncionais
8.
Vaccines (Basel) ; 8(1)2020 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936739

RESUMO

The need for a malaria vaccine is indisputable. A single vaccine for Plasmodium pre-erythrocytic stages targeting the major sporozoite antigen circumsporozoite protein (CSP) has had partial success. Additionally, CD8+ T cells targeting liver-stage (LS) antigens induced by live attenuated sporozoite vaccines were associated with protection in human challenge experiments. To further evaluate protection mediated by LS antigens, we focused on exported pre-erythrocytic proteins (exported protein 1 (EXP1), profilin (PFN), exported protein 2 (EXP2), inhibitor of cysteine proteases (ICP), transmembrane protein 21 (TMP21), and upregulated in infective sporozoites-3 (UIS3)) expressed in all Plasmodium species and designed optimized, synthetic DNA (synDNA) immunogens. SynDNA antigen cocktails were tested with and without the molecular adjuvant plasmid IL-33. Immunized animals developed robust T cell responses including induction of antigen-specific liver-localized CD8+ T cells, which were enhanced by the co-delivery of plasmid IL-33. In total, 100% of mice in adjuvanted groups and 71%-88% in non-adjuvanted groups were protected from blood-stage disease following Plasmodium yoelii sporozoite challenge. This study supports the potential of synDNA LS antigens as vaccine components for malaria parasite infection.

9.
Trends Parasitol ; 36(2): 85-87, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31883707

RESUMO

A recent report by Jennison et al. reveals an important role for plasmepsin V (PMV), an aspartyl protease, in the development of malaria transmission stages. The authors showed that PMV activity is critical for protein export in these stages and that specific PMV inhibitors block parasite transmission to mosquitoes.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Malária/parasitologia , Malária/transmissão , Plasmodium/enzimologia , Animais , Antimaláricos/farmacologia , Carbamatos/farmacologia , Humanos , Estágios do Ciclo de Vida/efeitos dos fármacos , Estágios do Ciclo de Vida/fisiologia , Malária/prevenção & controle , Oligopeptídeos/farmacologia , Plasmodium/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos
10.
F1000Res ; 9: 1268, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-35600144

RESUMO

Background: The emergence of artemisinin resistance in South East Asia calls for urgent discovery of new drug compounds that have antiplasmodial activity. Unlike the classical compound screening drug discovery methods, the rational approach involving targeted drug discovery is less cumbersome and therefore key for innovation of new antiplasmodial compounds.  Plasmodium falciparum (Pf) utilizes the process of host erythrocyte remodeling using Plasmodium-helical interspersed sub-telomeric domain (PHIST) containing proteins, which are amenable drug targets. The aim of this study is to identify inhibitors of PHIST from sulfated polysaccharides as new antimalarials. Methods: 251 samples from an ongoing study of epidemiology of malaria and drug resistance sensitivity patterns in Kenya were sequenced for PHISTb/RLP1 gene using Sanger sequencing. The sequenced reads were mapped to the reference Pf3D7 protein sequence of PHISTb/RLP1 using CLC Main Workbench. Homology modeling of both reference and mutant protein structures was achieved using the LOMETs tool. The models were refined using ModRefiner for energy minimization. Ramachandran plot was generated by ProCheck to assess the conformation of amino acids in the protein model. Protein binding sites predictions were assessed using FT SITE software. We searched for prospective antimalarials from PubChem. Docking experiments were achieved using AutoDock Vina and analysis results visualized in PyMOL. Results: Sanger sequencing generated 86 complete sequences. Upon mapping of the sequences to the reference, 12 non-synonymous single nucleotide polymorphisms were considered for mutant protein structure analysis. Eleven drug compounds with antiplasmodial activity were identified. Both modelled PHISTb/RLP1 reference and mutant structures had a Ramachandran score of >90% of the amino acids in the favored region. Ten of the drug compounds interacted with amino acid residues in PHISTb and RESA domains, showing potential activity against these proteins. Conclusion: These interactions provide lead compounds for new anti-malarial molecules. Further in vivo testing is recommended.

11.
Genome Biol Evol ; 11(11): 3269-3274, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31697367

RESUMO

The Laverania clade comprises the human malaria parasite Plasmodium falciparum as well as at least seven additional parasite species that infect wild African apes. A recent analysis of Laverania genome sequences (Otto TD, et al. 2018. Genomes of all known members of a Plasmodium subgenus reveal paths to virulent human malaria. Nat Microbiol. 3: 687-697) reported three instances of interspecies gene transfer, one of which had previously been described. Generating gene sequences from additional ape parasites and re-examining sequencing reads generated in the Otto et al. study, we identified one of the newly described gene transfers as an assembly artifact of sequences derived from a sample coinfected by two parasite species. The second gene transfer between ancestors of two divergent chimpanzee parasite lineages was confirmed, but involved a much larger number of genes than originally described, many of which encode exported proteins that remodel, or bind to, erythrocytes. Because successful hybridization between Laverania species is very rare, it will be important to determine to what extent these gene transfers have shaped their host interactions.


Assuntos
Introgressão Genética , Pan troglodytes/parasitologia , Plasmodium/genética , Virulência/genética , Animais , Evolução Molecular , Humanos , Malária/parasitologia , Filogenia , Plasmodium/patogenicidade
12.
Methods Enzymol ; 586: 57-83, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28137577

RESUMO

Protein folding is an intricate and precise process in living cells. Most exported proteins evade cytoplasmic folding, become targeted to the membrane, and then trafficked into/across membranes. Their targeting and translocation-competent states are nonnatively folded. However, once they reach the appropriate cellular compartment, they can fold to their native states. The nonnative states of preproteins remain structurally poorly characterized since increased disorder, protein sizes, aggregation propensity, and the observation timescale are often limiting factors for typical structural approaches such as X-ray crystallography and NMR. Here, we present an alternative approach for the in vitro analysis of nonfolded translocation-competent protein states and their comparison with their native states. We make use of hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS), a method based on differentiated isotope exchange rates in structured vs unstructured protein states/regions, and highly dynamic vs more rigid regions. We present a complete structural characterization pipeline, starting from the preparation of the polypeptides to data analysis and interpretation. Proteolysis and mass spectrometric conditions for the analysis of the labeled proteins are discussed, followed by the analysis and interpretation of HDX-MS data. We highlight the suitability of HDX-MS for identifying short structured regions within otherwise highly flexible protein states, as illustrated by an exported protein example, experimentally tested in our lab. Finally, we discuss statistical analysis in comparative HDX-MS. The protocol is applicable to any protein and protein size, exhibiting slow or fast loss of translocation competence. It could be easily adapted to more complex assemblies, such as the interaction of chaperones with nonnative protein states.


Assuntos
Medição da Troca de Deutério , Proteínas de Escherichia coli/química , Espectrometria de Massas , Proteoma/química , Sequência de Aminoácidos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/isolamento & purificação , Proteínas de Escherichia coli/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteólise , Proteoma/isolamento & purificação , Proteoma/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
13.
Adv Parasitol ; 91: 1-86, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27015947

RESUMO

Malaria, caused by Plasmodium spp., continues to be a major threat to human health and a significant cause of socioeconomic hardship in many countries. Almost half of the world's population live in malaria-endemic regions and many of them suffer one or more, often life-threatening episodes of malaria every year, the symptoms of which are attributable to replication of the parasite within red blood cells (RBCs). In the case of Plasmodium falciparum, the species responsible for most malaria-related deaths, parasite replication within RBCs is accompanied by striking alterations to the morphological, biochemical and biophysical properties of the host cell that are essential for the parasites' survival. To achieve this, the parasite establishes a unique and extensive protein export network in the infected RBC, dedicating at least 6% of its genome to the process. Understanding the full gamut of proteins involved in this process and the mechanisms by which P. falciparum alters the structure and function of RBCs is important both for a more complete understanding of the pathogenesis of malaria and for development of new therapeutic strategies to prevent or treat this devastating disease. This review focuses on what is currently known about exported parasite proteins, their interactions with the RBC and their likely pathophysiological consequences.


Assuntos
Eritrócitos/parasitologia , Plasmodium/fisiologia , Proteínas de Protozoários/fisiologia , Eritrócitos/fisiologia , Eritrócitos/ultraestrutura , Humanos , Chaperonas Moleculares/fisiologia , Plasmodium/química , Transporte Proteico , Proteínas de Protozoários/metabolismo
14.
Mol Biochem Parasitol ; 200(1-2): 25-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25956941

RESUMO

Remodeling of the host red blood cell by Plasmodium falciparum is well established and crucial for infection and parasite virulence. Host cell modifications are not exclusive to human Plasmodium parasites and also occur in hepatocytes and erythrocytes infected with murine Plasmodium parasites. The recently described intra-erythrocytic P. berghei-induced structures (IBIS) share similarities to P. falciparum Maurer's clefts. It is shown here that a potential candidate IBIS1 homologue in P. falciparum, PfHYP12 (PF3D7_1301400), is partially exported into the erythrocyte cytoplasm. To analyze a potential similarity between IBIS and Maurer's clefts we expressed the signature protein of IBIS in P. falciparum parasites. Visualization of the tagged protein revealed that PbIBIS1 can be exported by P. falciparum and localizes to Maurer's clefts in P. falciparum-infected erythrocytes, which indicates that IBIS and Maurer's clefts may be evolutionarily conserved parasite-induced structures in infected erythrocytes.


Assuntos
Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Plasmodium berghei/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Animais , Interações Hospedeiro-Parasita , Humanos , Camundongos , Dados de Sequência Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Transporte Proteico , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Alinhamento de Sequência
15.
Protein Sci ; 24(9): 1508-20, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26130467

RESUMO

Caseinolytic chaperones and proteases (Clp) belong to the AAA+ protein superfamily and are part of the protein quality control machinery in cells. The eukaryotic parasite Plasmodium falciparum, the causative agent of malaria, has evolved an elaborate network of Clp proteins including two distinct ClpB ATPases. ClpB1 and ClpB2 are involved in different aspects of parasitic proteostasis. ClpB1 is present in the apicoplast, a parasite-specific and plastid-like organelle hosting various metabolic pathways necessary for parasite growth. ClpB2 localizes to the parasitophorous vacuole membrane where it drives protein export as core subunit of a parasite-derived protein secretion complex, the Plasmodium Translocon of Exported proteins (PTEX); this process is central to parasite virulence and survival in the human host. The functional associations of these two chaperones with parasite-specific metabolism and protein secretion make them prime drug targets. ClpB proteins function as unfoldases and disaggregases and share a common architecture consisting of four domains-a variable N-terminal domain that binds different protein substrates, followed by two highly conserved catalytic ATPase domains, and a C-terminal domain. Here, we report and compare the first crystal structures of the N terminal domains of ClpB1 and ClpB2 from Plasmodium and analyze their molecular surfaces. Solution scattering analysis of the N domain of ClpB2 shows that the average solution conformation is similar to the crystalline structure. These structures represent the first step towards the characterization of these two malarial chaperones and the reconstitution of the entire PTEX to aid structure-based design of novel anti-malarial drugs.


Assuntos
Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Antimaláricos/química , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/química , Sequência de Aminoácidos , Antimaláricos/farmacologia , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Dados de Sequência Molecular , Terapia de Alvo Molecular , Plasmodium falciparum/genética , Dobramento de Proteína , Estrutura Secundária de Proteína , Transporte Proteico
16.
Int J Parasitol ; 44(5): 319-28, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24530877

RESUMO

Alteration of the adhesive and mechanical properties of red blood cells caused by infection with the malaria parasite Plasmodium falciparum underpin both its survival and extreme pathogenicity. A unique family of parasite putative exported kinases, collectively called FIKK (Phenylalanine (F) - Isoleucine (I) - Lysine (K) - Lysine (K)), has recently been implicated in these pathophysiological processes, however, their precise function in P. falciparum-infected red blood cells or their likely role in malaria pathogenesis remain unknown. Here, for the first time, we demonstrate that one member of the FIKK family, FIKK4.2, can function as an active kinase and is localised in a novel and distinct compartment of the parasite-infected red blood cell which we have called K-dots. Notably, targeted disruption of the gene encoding FIKK4.2 (fikk4.2) dramatically alters the parasite's ability to modify and remodel the red blood cells in which it multiplies. Specifically, red blood cells infected with fikk4.2 knockout parasites were significantly less rigid and less adhesive when compared with red blood cells infected with normal parasites from which the transgenic clones had been derived, despite expressing similar levels of the major cytoadhesion ligand, PfEMP1, on the red blood cell surface. Notably, these changes were accompanied by dramatically altered knob-structures on infected red blood cells that play a key role in cytoadhesion which is responsible for much of the pathogenesis associated with falciparum malaria. Taken together, our data identifies FIKK4.2 as an important kinase in the pathogenesis of P. falciparum malaria and strengthens the attractiveness of FIKK kinases as targets for the development of novel next-generation anti-malaria drugs.


Assuntos
Eritrócitos/parasitologia , Fosfotransferases/metabolismo , Plasmodium falciparum/enzimologia , Fatores de Virulência/metabolismo , Técnicas de Inativação de Genes , Humanos , Fosfotransferases/genética , Plasmodium falciparum/genética , Transporte Proteico , Fatores de Virulência/genética
17.
Mol Biochem Parasitol ; 191(1): 44-52, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24076174

RESUMO

Protein export is considered an essential feature of malaria parasite blood stage development. Here, we examined five components of the candidate Plasmodium translocon of exported proteins (PTEX), a complex thought to mediate protein export across the parasitophorous vacuole membrane into the host cell. Using the murine malaria model parasite Plasmodium berghei, we succeeded in generating parasite lines lacking PTEX88 and thioredoxin 2 (TRX2). Repeated attempts to delete the remaining three translocon components failed, suggesting essential functions for EXP2, PTEX150, and heat shock protein 101 (HSP101) during blood stage development. To analyze blood infections of the null-mutants, we established a flow cytometry-assisted intravital competition assay using three novel high fluorescent lines (Bergreen, Beryellow, and Berred). Although blood stage development of parasites lacking TRX2 was affected, the deficit was much more striking in PTEX88 null-mutants. The multiplication rate of PTEX88-deficient parasites was strongly reduced resulting in out-competition by wild-type parasites. Endogenous tagging revealed that TRX2::tag resides in distinct punctate organelles of unknown identity. PTEX88::tag shows a diffuse intraparasitic pattern in blood stage parasites. In trophozoites, PTEX88::tag also localized to previously unrecognized extensions reaching from the parasite surface into the erythrocyte cytoplasm. Together, our results indicate auxiliary roles for TRX2 and PTEX88 and central roles for EXP2, PTEX150, and HSP101 during P. berghei blood infection.


Assuntos
Sangue/parasitologia , Proteínas de Ligação ao Cálcio/metabolismo , Regulação da Expressão Gênica , Glicoproteínas de Membrana/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Peptídeos/metabolismo , Animais , Proteínas de Ligação ao Cálcio/genética , Deleção de Genes , Genes Essenciais , Genes de Protozoários , Teste de Complementação Genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Transporte Proteico , Proteínas de Protozoários/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Peptídeos/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa