Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 68(5): e0169023, 2024 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-38501806

RESUMO

Malaria tropica, caused by the parasite Plasmodium falciparum (P. falciparum), remains one of the greatest public health burdens for humankind. Due to its pivotal role in parasite survival, the energy metabolism of P. falciparum is an interesting target for drug design. To this end, analysis of the central metabolite adenosine triphosphate (ATP) is of great interest. So far, only cell-disruptive or intensiometric ATP assays have been available in this system, with various drawbacks for mechanistic interpretation and partly inconsistent results. To address this, we have established fluorescent probes, based on Förster resonance energy transfer (FRET) and known as ATeam, for use in blood-stage parasites. ATeams are capable of measuring MgATP2- levels in a ratiometric manner, thereby facilitating in cellulo measurements of ATP dynamics in real-time using fluorescence microscopy and plate reader detection and overcoming many of the obstacles of established ATP analysis methods. Additionally, we established a superfolder variant of the ratiometric pH sensor pHluorin (sfpHluorin) in P. falciparum to monitor pH homeostasis and control for pH fluctuations, which may affect ATeam measurements. We characterized recombinant ATeam and sfpHluorin protein in vitro and stably integrated the sensors into the genome of the P. falciparum NF54attB cell line. Using these new tools, we found distinct sensor response patterns caused by several different drug classes. Arylamino alcohols increased and redox cyclers decreased ATP; doxycycline caused first-cycle cytosol alkalization; and 4-aminoquinolines caused aberrant proteolysis. Our results open up a completely new perspective on drugs' mode of action, with possible implications for target identification and drug development.


Assuntos
Trifosfato de Adenosina , Antimaláricos , Transferência Ressonante de Energia de Fluorescência , Plasmodium falciparum , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Trifosfato de Adenosina/metabolismo , Antimaláricos/farmacologia , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/química , Humanos , Quinina/farmacologia , Doxiciclina/farmacologia , Artemisininas/farmacologia , Cloroquina/farmacologia , Concentração de Íons de Hidrogênio
2.
Antioxid Redox Signal ; 37(1-3): 1-18, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35072524

RESUMO

Aims: Genetically encoded green fluorescent protein (GFP)-based redox biosensors are widely used to monitor specific and dynamic redox processes in living cells. Over the last few years, various biosensors for a variety of applications were engineered and enhanced to match the organism and cellular environments, which should be investigated. In this context, the unicellular intraerythrocytic parasite Plasmodium, the causative agent of malaria, represents a challenge, as the small size of the organism results in weak fluorescence signals that complicate precise measurements, especially for cell compartment-specific observations. To address this, we have functionally and structurally characterized an enhanced redox biosensor superfolder roGFP2 (sfroGFP2). Results: SfroGFP2 retains roGFP2-like behavior, yet with improved fluorescence intensity (FI) in cellulo. SfroGFP2-based redox biosensors are pH insensitive in a physiological pH range and show midpoint potentials comparable with roGFP2-based redox biosensors. Using crystallography and rigidity theory, we identified the superfolding mutations as being responsible for improved structural stability of the biosensor in a redox-sensitive environment, thus explaining the improved FI in cellulo. Innovation: This work provides insight into the structure and function of GFP-based redox biosensors. It describes an improved redox biosensor (sfroGFP2) suitable for measuring oxidizing effects within small cells where applicability of other redox sensor variants is limited. Conclusion: Improved structural stability of sfroGFP2 gives rise to increased FI in cellulo. Fusion to hGrx1 (human glutaredoxin-1) provides the hitherto most suitable biosensor for measuring oxidizing effects in Plasmodium. This sensor is of major interest for studying glutathione redox changes in small cells, as well as subcellular compartments in general. Antioxid. Redox Signal. 37, 1-18.


Assuntos
Técnicas Biossensoriais , Glutationa , Plasmodium , Técnicas Biossensoriais/métodos , Glutationa/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Oxirredução , Plasmodium/isolamento & purificação
3.
Parasitol Int ; 87: 102513, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34785370

RESUMO

Plasmodium falciparum macrophage migration inhibitory factor (PfMIF) is a homologue of the multifunctional human host cytokine MIF (HsMIF). Upon schizont rupture it is released into the human blood stream where it acts as a virulence factor, modulating the host immune system. Whereas for HsMIF a tautomerase, an oxidoreductase, and a nuclease activity have been identified, the latter has not yet been studied for PfMIF. Furthermore, previous studies identified PfMIF as a target for several redox post-translational modifications. Therefore, we analysed the impact of S-glutathionylation and S-nitrosation on the protein's functions. To determine the impact of the four cysteines of PfMIF we produced His-tagged cysteine to alanine mutants of PfMIF via site-directed mutagenesis. Recombinant proteins were analysed via mass spectrometry, and enzymatic assays. Here we show for the first time that PfMIF acts as a DNase of human genomic DNA and that this activity is greater than that shown by HsMIF. Moreover, we observed a significant decrease in the maximum velocity of the DCME tautomerase activity of PfMIF upon alanine replacement of Cys3, and Cys3/Cys4 double mutant. Lastly, using a yeast reporter system, we were able to verify binding of PfMIF to the human chemokine receptors CXCR4, and demonstrate a so-far overlooked binding to CXCR2, both of which function as non-cognate receptors for HsMIF. While S-glutathionylation and S-nitrosation of PfMIF did not impair the tautomerase activity of PfMIF, activation of these receptors was significantly decreased.


Assuntos
Cisteína/deficiência , Fatores Inibidores da Migração de Macrófagos/química , Fatores Inibidores da Migração de Macrófagos/genética , Plasmodium falciparum/química , Alanina/química , Cisteína/genética , Desoxirribonucleases/metabolismo , Humanos , Plasmodium falciparum/genética , Proteínas Recombinantes/genética
4.
Mol Biochem Parasitol ; 242: 111362, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33513391

RESUMO

Plasmodium falciparum causes the deadliest form of malaria. Adequate redox control is crucial for this protozoan parasite to overcome oxidative and nitrosative challenges, thus enabling its survival. Sulfenylation is an oxidative post-translational modification, which acts as a molecular on/off switch, regulating protein activity. To obtain a better understanding of which proteins are redox regulated in malaria parasites, we established an optimized affinity capture protocol coupled with mass spectrometry analysis for identification of in vivo sulfenylated proteins. The non-dimedone based probe BCN-Bio1 shows reaction rates over 100-times that of commonly used dimedone-based probes, allowing for a rapid trapping of sulfenylated proteins. Mass spectrometry analysis of BCN-Bio1 labeled proteins revealed the first insight into the Plasmodium falciparum trophozoite sulfenylome, identifying 102 proteins containing 152 sulfenylation sites. Comparison with Plasmodium proteins modified by S-glutathionylation and S-nitrosation showed a high overlap, suggesting a common core of proteins undergoing redox regulation by multiple mechanisms. Furthermore, parasite proteins which were identified as targets for sulfenylation were also identified as being sulfenylated in other organisms, especially proteins of the glycolytic cycle. This study suggests that a number of Plasmodium proteins are subject to redox regulation and it provides a basis for further investigations into the exact structural and biochemical basis of regulation, and a deeper understanding of cross-talk between post-translational modifications.


Assuntos
Compostos Bicíclicos com Pontes/química , Sondas Moleculares/química , Plasmodium falciparum/metabolismo , Processamento de Proteína Pós-Traducional , Proteínas de Protozoários/metabolismo , Ácidos Sulfênicos/metabolismo , Trofozoítos/metabolismo , Células Cultivadas , Cisteína/metabolismo , Eritrócitos/parasitologia , Ontologia Genética , Glutationa/metabolismo , Humanos , Espectrometria de Massas , Anotação de Sequência Molecular , Compostos Nitrosos/metabolismo , Oxirredução , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Coloração e Rotulagem/métodos , Trofozoítos/genética
5.
Sci Rep ; 10(1): 4193, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144363

RESUMO

The regulation of human Arf1 GTPase activity by ArfGEFs that stimulate GDP/GTP exchange and ArfGAPs that mediate GTP hydrolysis has attracted attention for the discovery of Arf1 inhibitors as potential anti-cancer agents. The malaria parasite Plasmodium falciparum encodes a Sec7 domain-containing protein - presumably an ArfGEF - and two putative ArfGAPs, as well as an Arf1 homologue (PfArf1) that is essential for blood-stage parasite viability. However, ArfGEF and ArfGAP-mediated activation/deactivation of PfArf1 has not been demonstrated. In this study, we established an in vitro colorimetric microtiter plate-based assay to detect the activation status of truncated human and P. falciparum Arf1 and used it to demonstrate the activation of both proteins by the Sec7 domain of ARNO, their deactivation by the GAP domain of human ArfGAP1 and the inhibition of the respective reactions by the compounds SecinH3 and QS11. In addition, we found that the GAP domains of both P. falciparum ArfGAPs have activities equivalent to that of human ArfGAP1, but are insensitive to QS11. Library screening identified a novel inhibitor which selectively inhibits one of the P. falciparum GAP domains (IC50 4.7 µM), suggesting that the assay format is suitable for screening compound collections for inhibitors of Arf1 regulatory proteins.


Assuntos
Proteínas de Bactérias/metabolismo , Bioensaio/métodos , Colorimetria/métodos , Proteínas Ativadoras de GTPase/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Bactérias/química , Proteínas Ativadoras de GTPase/química , Guanosina Trifosfato/metabolismo , Humanos , Hidrólise
6.
PLoS One ; 13(4): e0195823, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29672587

RESUMO

An increasing number of monoclonal antibodies and derivatives such as antibody-drug conjugates (ADC) are of the IgG1 and IgG4 isotype with distinct structural and functional properties. In cases where antibody-mediated cytotoxicity is not desired, IgG4 is often used, as its Fc region is relatively poor at inducing antibody-dependent cell-mediated or complement-dependent cytotoxicity. IgG4 ADCs with highly cytotoxic drugs against proliferating target cells but which lack or have diminished antibody effector functions against quiescent cells may have a favorable safety profile compared to IgG1. Another unique property of the IgG4 subclass is the capability to exchange half antibodies in vivo creating randomly bispecific antibodies. To investigate the functional properties of process-derived antibody species, and determine the influence of shuffling on the therapeutic efficacy, several model antibodies on the basis of the anti-CD138 antibody-drug conjugate BT062 (Indatuximab ravtansine) were generated: (I) A wild type nBT062, (II) a stable nBT062 comprising mutations to prevent half-antibody exchange, (III) a half nBT062 lacking covalent binding between two heavy chains and (IV) a stabilized, bispecific nBT062-natalizumab antibody with a second, monovalent specificity against CD49d. All nBT062 model variants were capable of CD138-specific binding and antigen-mediated internalization into cells. Furthermore, all nBT062 models inhibited tumor growth in vitro after conjugation with the maytansinoid DM4. The in vivo effects of the different molecular variants were assessed in the MAXF1322 xenograft model. The bispecific nBT062-natalizumab-DM4 demonstrated the least efficacy and was only moderately active even without the co-administration of a human IgG preparation. Wild type, stable and half nBT062-DM4 models demonstrated great anti-tumor activities. The efficacy of wild type and half nBT062-DM4 was reduced in the presence of IgG, while stable nBT062-DM4 was only marginally influenced. These pre-clinical data demonstrate the advantage of introducing half-antibody exchange-preventing mutations into therapeutic IgG4-based antibody drug-conjugates.


Assuntos
Anticorpos Biespecíficos/farmacologia , Anticorpos Monoclonais/farmacologia , Imunoconjugados/farmacologia , Imunoglobulina G/farmacologia , Animais , Anticorpos Biespecíficos/genética , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos/imunologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Variação Genética , Humanos , Imunoconjugados/imunologia , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Camundongos , Mutação , Ligação Proteica , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Free Radic Biol Med ; 104: 104-117, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28062360

RESUMO

The malaria parasite Plasmodium falciparum is exposed to multiple sources of oxidative challenge during its complex life cycle in the Anopheles vector and its human host. In order to further elucidate redox-based parasite host cell interactions and mechanisms of drug action, we targeted the genetically encoded glutathione redox sensor roGFP2 coupled to human glutaredoxin 1 (roGFP2-hGrx1) as well as the ratiometric pH sensor pHluorin to the apicoplast and the mitochondrion of P. falciparum. Using live cell imaging, this allowed for the first time the determination of the pH values of the apicoplast (7.12±0.40) and mitochondrion (7.37±0.09) in the intraerythrocytic asexual stages of the parasite. Based on the roGFP2-hGrx1 signals, glutathione-dependent redox potentials of -267mV and -328mV, respectively, were obtained. Employing these novel tools, initial studies on the effects of redox-active agents and clinically employed antimalarial drugs were carried out on both organelles.


Assuntos
Glutarredoxinas/metabolismo , Interações Hospedeiro-Parasita/genética , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Animais , Anopheles/parasitologia , Antimaláricos/metabolismo , Antimaláricos/uso terapêutico , Apicoplastos/metabolismo , Glutarredoxinas/genética , Glutationa/genética , Glutationa/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Mitocôndrias/metabolismo , Oxirredução , Estresse Oxidativo/genética , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade
8.
Nat Commun ; 7: 10470, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26832821

RESUMO

Plasmodium falciparum exports proteins into erythrocytes using the Plasmodium export element (PEXEL) motif, which is cleaved in the endoplasmic reticulum (ER) by plasmepsin V (PMV). A recent study reported that phosphatidylinositol-3-phosphate (PI(3)P) concentrated in the ER binds to PEXEL motifs and is required for export independent of PMV, and that PEXEL motifs are functionally interchangeable with RxLR motifs of oomycete effectors. Here we show that the PEXEL does not bind PI(3)P, and that this lipid is not concentrated in the ER. We find that RxLR motifs cannot mediate export in P. falciparum. Parasites expressing a mutated version of KAHRP, with the PEXEL motif repositioned near the signal sequence, prevented PMV cleavage. This mutant possessed the putative PI(3)P-binding residues but is not exported. Reinstatement of PEXEL to its original location restores processing by PMV and export. These results challenge the PI(3)P hypothesis and provide evidence that PEXEL position is conserved for co-translational processing and export.


Assuntos
Fosfatos de Fosfatidilinositol/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Motivos de Aminoácidos , Membrana Celular , Escherichia coli , Lopinavir/farmacologia , Plasmodium falciparum/genética , Ligação Proteica , Proteínas de Protozoários/genética
9.
PLoS One ; 10(4): e0125191, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25909331

RESUMO

Plasmodium falciparum invades human red blood cells, residing in a parasitophorous vacuole (PV), with a parasitophorous vacuole membrane (PVM) separating the PV from the host cell cytoplasm. Here we have investigated the role of N-myristoylation and two other N-terminal motifs, a cysteine potential S-palmitoylation site and a stretch of basic residues, as the driving force for protein targeting to the parasite plasma membrane (PPM) and subsequent translocation across this membrane. Plasmodium falciparum adenylate kinase 2 (Pf AK2) contains these three motifs, and was previously proposed to be targeted beyond the parasite to the PVM, despite the absence of a signal peptide for entry into the classical secretory pathway. Biochemical and microscopy analyses of PfAK2 variants tagged with green fluorescent protein (GFP) showed that these three motifs are involved in targeting the protein to the PPM and translocation across the PPM to the PV. It was shown that the N-terminal 37 amino acids of PfAK2 alone are sufficient to target and translocate GFP across the PPM. As a control we examined the N-myristoylated P. falciparum ADP-ribosylation factor 1 (PfARF1). PfARF1 was found to co-localise with a Golgi marker. To determine whether or not the putative palmitoylation and the cluster of lysine residues from the N-terminus of PfAK2 would modulate the subcellular localization of PfARF1, a chimeric fusion protein containing the N-terminus of PfARF1 and the two additional PfAK2 motifs was analysed. This chimeric protein was targeted to the PPM, but not translocated across the membrane into the PV, indicating that other features of the N-terminus of PfAK2 also play a role in the secretion process.


Assuntos
Malária Falciparum/parasitologia , Parasitos/fisiologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Via Secretória/fisiologia , Fator 1 de Ribosilação do ADP/metabolismo , Adenilato Quinase/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Membrana Celular/metabolismo , Cisteína/metabolismo , Eritrócitos/parasitologia , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Lisina/metabolismo , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Sinais Direcionadores de Proteínas/fisiologia , Transporte Proteico/fisiologia , Vacúolos/metabolismo
10.
Int J Biochem Cell Biol ; 62: 47-53, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25701168

RESUMO

Heat shock proteins, many of which function as molecular chaperones, play important roles in the lifecycle and pathogenesis of the malaria parasite, Plasmodium falciparum. The P. falciparum heat shock protein 70 (PfHsp70) family of chaperones is potentially regulated by a large complement of J proteins that localize to various intracellular compartments including the infected erythrocyte cytosol. While PfHsp70-1 has been shown to be an abundant cytosolic chaperone, its regulation by J proteins is poorly understood. In this study, we characterized the J protein PFB0595w, a homologue of the well-studied yeast cytosolic J protein, Sis1. PFB0595w, similarly to PfHsp70-1, was localized to the parasite cytosol and its expression was upregulated by heat shock. Additionally, recombinant PFB0595w was shown to be dimeric and to stimulate the in vitro ATPase activity of PfHsp70-1. Overall, the expression, localization and biochemical data for PFB0595w suggest that it may function as a cochaperone of PfHsp70-1, and advances current knowledge on the chaperone machinery of the parasite.


Assuntos
Trifosfato de Adenosina/metabolismo , Proteínas de Choque Térmico HSP72/metabolismo , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Adenosina Trifosfatases/metabolismo , Citosol/metabolismo , Eritrócitos/parasitologia , Humanos , Hidrólise , Técnicas In Vitro , Plasmodium falciparum/citologia , Ligação Proteica , Multimerização Proteica , Distribuição Tecidual
11.
Cell Host Microbe ; 12(5): 717-29, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23159060

RESUMO

For proliferation, the malaria parasite Plasmodium falciparum needs to modify the infected host cell extensively. To achieve this, the parasite exports proteins containing a Plasmodium export element (PEXEL) into the host cell. Phosphatidylinositol-3-phosphate binding and cleavage of the PEXEL are thought to mediate protein export. We show that these requirements can be bypassed, exposing a second level of export control in the N terminus generated after PEXEL cleavage that is sufficient to distinguish exported from nonexported proteins. Furthermore, this region also corresponds to the export domain of a second group of exported proteins lacking PEXELs (PNEPs), indicating shared export properties among different exported parasite proteins. Concordantly, export of both PNEPs and PEXEL proteins depends on unfolding, revealing translocation as a common step in export. However, translocation of transmembrane proteins occurs at the parasite plasma membrane, one step before translocation of soluble proteins, indicating unexpectedly complex translocation events at the parasite periphery.


Assuntos
Proteínas de Transporte/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Humanos , Fosfatos de Fosfatidilinositol/metabolismo , Transporte Proteico , Desdobramento de Proteína , Proteínas de Protozoários/química
12.
Eukaryot Cell ; 11(12): 1472-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23042132

RESUMO

Protein import into complex plastids of red algal origin is a multistep process including translocons of different evolutionary origins. The symbiont-derived ERAD-like machinery (SELMA), shown to be of red algal origin, is proposed to be the transport system for preprotein import across the periplastidal membrane of heterokontophytes, haptophytes, cryptophytes, and apicomplexans. In contrast to the canonical endoplasmic reticulum-associated degradation (ERAD) system, SELMA translocation is suggested to be uncoupled from proteasomal degradation. We investigated the distribution of known and newly identified SELMA components in organisms with complex plastids of red algal origin by intensive data mining, thereby defining a set of core components present in all examined organisms. These include putative pore-forming components, a ubiquitylation machinery, as well as a Cdc48 complex. Furthermore, the set of known 20S proteasomal components in the periplastidal compartment (PPC) of diatoms was expanded. These newly identified putative SELMA components, as well as proteasomal subunits, were in vivo localized as PPC proteins in the diatom Phaeodactylum tricornutum. The presented data allow us to speculate about the specific features of SELMA translocation in contrast to the canonical ERAD system, especially the uncoupling of translocation from degradation.


Assuntos
Diatomáceas/enzimologia , Proteínas de Plantas/metabolismo , Plastídeos/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Rodófitas/enzimologia , Ubiquitina/metabolismo , Adenosina Trifosfatases/metabolismo , Sequência de Aminoácidos , Proteínas de Ciclo Celular/metabolismo , Diatomáceas/genética , Diatomáceas/metabolismo , Degradação Associada com o Retículo Endoplasmático , Dados de Sequência Molecular , Proteínas de Plantas/química , Plastídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/química , Estrutura Terciária de Proteína , Transporte Proteico , Proteólise , Rodófitas/genética , Rodófitas/metabolismo , Proteína com Valosina
13.
Antioxid Redox Signal ; 17(4): 534-43, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22229886

RESUMO

AIMS: The malaria parasite Plasmodium falciparum possesses four unique selenoproteins (PfSel1-PfSel4) which are likely to represent important components of the redox-regulatory network of this infectious agent. So far these proteins have only been characterized in silico. The aim of the present study was to gain further insight into the structural, biochemical, and functional properties of P. falciparum selenoproteins. RESULTS: Using (75)Se labeling in P. falciparum cell culture, the presence of selenoproteins in the parasite could be verified for the first time. Bioinformatic analyses indicated distant relatedness between the Plasmodium proteins and selenoproteins described in other organisms, namely between PfSel1 and SelK, PfSel2 and SelT, and between PfSel4 and SelS. For PfSel3 no remarkable similarities with proteins from other organisms were identified. All four proteins were recombinantly produced in Escherichia coli as UGA→UGU (selenocysteine→cysteine) mutants. Using green fluorescent protein (GFP)-fusion proteins and immunofluorescence, the subcellular localization of the four selenoprotein mutants was studied. PfSel1, PfSel2, and PfSel4 localized to the endoplasmic reticulum whereas PfSel3 was visualized in the nucleus and/or the apicoplast. Functional assays support the roles of PfSel1 and PfSel4 in cellular redox reactions. Transcriptional profiles of the four selenoproteins, and proteins involved in selenoprotein biosynthesis, indicate that their expression is regulated via the availability of selenium and via oxidative and nitrosative stress. INNOVATION: In this study the presence of selenoproteins in Plasmodium has been proven for the first time; the subcellular localization of the proteins and their relatedness to known selenoproteins have been systematically studied, and recombinant proteins as well as information on regulation of transcript levels have been obtained. CONCLUSION: Taken together, our data enhance our understanding of the functional role of selenoproteins in Plasmodium.


Assuntos
Plasmodium falciparum/metabolismo , Proteoma , Proteínas de Protozoários/metabolismo , Selenoproteínas/metabolismo , Animais , Autorradiografia , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Nitrosação , Estresse Oxidativo , Plasmodium falciparum/crescimento & desenvolvimento , Frações Subcelulares/metabolismo
14.
Cell Microbiol ; 13(6): 897-912, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21501358

RESUMO

Malaria parasites reside in human erythrocytes within a parasitophorous vacuole. The parasites are transmitted from the human to the mosquito by the uptake of intraerythrocytic gametocytes during a blood meal, which in the midgut become activated by external stimuli and subsequently egress from the enveloping erythrocyte. Gametocyte egress is a crucial step for the parasite to prepare for fertilization, but the molecular mechanisms of egress are not well understood. Via electron microscopy, we show that Plasmodium falciparum gametocytes exit the erythrocyte by an inside-out type of egress. The parasitophorous vacuole membrane (PVM) ruptures at multiple sites within less than a minute following activation, a process that requires a temperature drop and parasite contact with xanthurenic acid. PVM rupture can also be triggered by the ionophore nigericin and is sensitive to the cysteine protease inhibitor E-64d. Following PVM rupture the subpellicular membrane begins to disintegrate. This membrane is specific to malaria gametocytes, and disintegration is impaired by the aspartic protease inhibitor EPNP and the cysteine/serine protease inhibitor TLCK. Approximately 15 min post activation, the erythrocyte membrane ruptures at a single breaking point, which can be inhibited by inhibitors TLCK and TPCK. In all cases inhibitor treatment results in interrupted gametogenesis.


Assuntos
Culicidae/parasitologia , Eritrócitos/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Animais , Compostos de Epóxi/metabolismo , Eritrócitos/ultraestrutura , Humanos , Membranas Intracelulares/ultraestrutura , Leucina/análogos & derivados , Leucina/metabolismo , Microscopia Eletrônica , Nigericina/metabolismo , Nitrofenóis/metabolismo , Plasmodium falciparum/ultraestrutura , Temperatura , Tosilina Clorometil Cetona/metabolismo , Vacúolos/parasitologia , Vacúolos/ultraestrutura , Xanturenatos/metabolismo
15.
Cell Stress Chaperones ; 16(4): 389-401, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21191678

RESUMO

Heat shock protein 70 (Hsp70) and heat shock protein 40 (Hsp40) function as molecular chaperones during the folding and trafficking of proteins within most cell types. However, the Hsp70-Hsp40 chaperone partnerships within the malaria parasite, Plasmodium falciparum, have not been elucidated. Only one of the 43 P. falciparum Hsp40s is predicted to be a cytosolic, canonical Hsp40 (termed PfHsp40) capable of interacting with the major cytosolic P. falciparum-encoded Hsp70, PfHsp70. Consistent with this hypothesis, we found that PfHsp40 is upregulated under heat shock conditions in a similar pattern to PfHsp70. In addition, PfHsp70 and PfHsp40 reside mainly in the parasite cytosol, as assessed using indirect immunofluorescence microscopy. Recombinant PfHsp40 stimulated the ATP hydrolytic rates of both PfHsp70 and human Hsp70 similar to other canonical Hsp40s of yeast (Ydj1) and human (Hdj2) origin. In contrast, the Hsp40-stimulated plasmodial and human Hsp70 ATPase activities were differentially inhibited in the presence of pyrimidinone-based small molecule modulators. To further probe the chaperone properties of PfHsp40, protein aggregation suppression assays were conducted. PfHsp40 alone suppressed protein aggregation, and cooperated with PfHsp70 to suppress aggregation. Together, these data represent the first cellular and biochemical evidence for a PfHsp70-PfHsp40 partnership in the malaria parasite, and furthermore that the plasmodial and human Hsp70-Hsp40 chaperones possess unique attributes that are differentially modulated by small molecules.


Assuntos
Proteínas de Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Resposta ao Choque Térmico , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/metabolismo , Adenosina Trifosfatases/metabolismo , Citosol/metabolismo , Expressão Gênica , Hidrólise , Plasmodium falciparum/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA