Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 12(8): e0183198, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28817634

RESUMO

Malaria, a disease endemic in many tropical and subtropical regions, is caused by infection of the erythrocyte by the apicomplexan parasite Plasmodium. Host-cell invasion is a complex process but two Plasmodium proteins, Apical Membrane Antigen 1 (AMA1) and the Rhoptry Neck protein complex (RON), play a key role. AMA1, present on the surface of the parasite, binds tightly to the RON2 component of the RON protein complex, which is inserted into the erythrocyte membrane during invasion. Blocking the AMA1-RON2 interaction with antibodies or peptides inhibits invasion, underlining its importance in the Plasmodium life cycle and as a target for therapeutic strategies. We describe the crystal structure of the complex formed between AMA1 from P. vivax (PvAMA1) and a peptide derived from the externally exposed region of P. vivax RON2 (PvRON2sp1), and of the heterocomplex formed between P. falciparum AMA1 (PfAMA1) and PvRON2sp1. Binding studies show that the affinity of PvRON2sp1 for PvAMA1 is weaker than that previously reported for the PfRON2sp1-PfAMA1 association. Moreover, while PvRON2sp1 shows strong cross-reactivity with PfAMA1, PfRON2sp1 displays no detectable interaction with PvAMA1. The structures show that the equivalent residues PvRON2-Thr2055 and PfRON2-Arg2041 largely account for this pattern of reactivity.


Assuntos
Reações Cruzadas , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Animais , Ligantes , Ligação Proteica , Proteínas de Protozoários/metabolismo
2.
Malar J ; 15: 28, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26772184

RESUMO

BACKGROUND: Rosetting, namely the capacity of the Plasmodium falciparum-infected red blood cells to bind uninfected RBCs, is commonly observed in African children with severe malaria. Rosetting results from specific interactions between a subset of variant P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesins encoded by var genes, serum components and RBC receptors. Rosette formation is a redundant phenotype, as there exists more than one var gene encoding a rosette-mediating PfEMP1 in each genome and hence a diverse array of underlying interactions. Moreover, field diversity creates a large panel of rosetting-associated serotypes and studies with human immune sera indicate that surface-reacting antibodies are essentially variant-specific. To gain better insight into the interactions involved in rosetting and map surface epitopes, a panel of monoclonal antibodies (mAbs) was investigated. METHODS: Monoclonal antibodies were isolated from mice immunized with PfEMP1-VarO recombinant domains. They were characterized using ELISA and reactivity with the native PfEMP1-VarO adhesin on immunoblots of reduced and unreduced extracts, as well as SDS-extracts of Palo Alto 89F5 VarO schizonts. Functionality was assessed using inhibition of Palo Alto 89F5 VarO rosette formation and disruption of Palo Alto 89F5 VarO rosettes. Competition ELISAs were performed with biotinylated antibodies against DBL1 to identify reactivity groups. Specificity of mAbs reacting with the DBL1 adhesion domain was explored using recombinant proteins carrying mutations abolishing RBC binding or binding to heparin, a potent inhibitor of rosette formation. RESULTS: Domain-specific, surface-reacting mAbs were obtained for four individual domains (DBL1, CIDR1, DBL2, DBL4). Monoclonal antibodies reacting with DBL1 potently inhibited the formation of rosettes and disrupted Palo Alto 89F5 VarO rosettes. Most surface-reactive mAbs and all mAbs interfering with rosetting reacted on parasite immunoblots with disulfide bond-dependent PfEMP1 epitopes. Based on competition ELISA and binding to mutant DBL1 domains, two distinct binding sites for rosette-disrupting mAbs were identified in close proximity to the RBC-binding site. CONCLUSIONS: Rosette-inhibitory antibodies bind to conformation-dependent epitopes located close to the RBC-binding site and distant from the heparin-binding site. These results provide novel clues for a rational intervention strategy that targets rosetting.


Assuntos
Anticorpos Monoclonais/metabolismo , Moléculas de Adesão Celular/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Ensaio de Imunoadsorção Enzimática , Camundongos , Plasmodium falciparum/efeitos dos fármacos , Ligação Proteica
3.
Sci Rep ; 5: 14868, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26450557

RESUMO

The human malaria parasite, Plasmodium falciparum, is able to evade spleen-mediated clearing from blood stream by sequestering in peripheral organs. This is due to the adhesive properties conferred by the P. falciparum Erythrocyte Membrane Protein 1 (PfEMP1) family exported by the parasite to the surface of infected erythrocytes. Expression of the VAR2CSA variant of PfEMP1 leads to pregnancy-associated malaria, which occurs when infected erythrocytes massively sequester in the placenta by binding to low-sulfated Chondroitin Sulfate A (CSA) present in the intervillous spaces. VAR2CSA is a 350 kDa protein that carries six Duffy-Binding Like (DBL) domains, one Cysteine-rich Inter-Domain Regions (CIDR) and several inter-domain regions. In the present paper, we report for the first time the crystal structure at 2.9 Šof a VAR2CSA double domain, DBL3X-DBL4ε, from the FCR3 strain. DBL3X and DBL4ε share a large contact interface formed by residues that are invariant or highly conserved in VAR2CSA variants, which suggests that these two central DBL domains (DBL3X-DBL4ε) contribute significantly to the structuring of the functional VAR2CSA extracellular region. We have also examined the antigenicity of peptides corresponding to exposed loop regions of the DBL4ε structure.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Placenta/imunologia , Plasmodium falciparum/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Sulfatos de Condroitina/imunologia , Sulfatos de Condroitina/metabolismo , Cristalografia por Raios X , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Interações Hospedeiro-Parasita/imunologia , Humanos , Soros Imunes/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Placenta/metabolismo , Placenta/parasitologia , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Gravidez , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Coelhos , Homologia de Sequência de Aminoácidos
4.
PLoS One ; 10(7): e0134292, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222304

RESUMO

Adhesion of Plasmodium falciparum-infected red blood cells (iRBC) to human erythrocytes (i.e. rosetting) is associated with severe malaria. Rosetting results from interactions between a subset of variant PfEMP1 (Plasmodium falciparum erythrocyte membrane protein 1) adhesins and specific erythrocyte receptors. Interfering with such interactions is considered a promising intervention against severe malaria. To evaluate the feasibility of a vaccine strategy targetting rosetting, we have used here the Palo Alto 89F5 VarO rosetting model. PfEMP1-VarO consists of five Duffy-Binding Like domains (DBL1-5) and one Cysteine-rich Interdomain Region (CIDR1). The binding domain has been mapped to DBL1 and the ABO blood group was identified as the erythrocyte receptor. Here, we study the immunogenicity of all six recombinant PfEMP1-VarO domains and the DBL1- CIDR1 Head domain in BALB/c and outbred OF1 mice. Five readouts of antibody responses are explored: ELISA titres on the recombinant antigen, VarO-iRBC immunoblot reactivity, VarO-iRBC surface-reactivity, capacity to disrupt VarO rosettes and the capacity to prevent VarO rosette formation. For three domains, we explore influence of the expression system on antigenicity and immunogenicity. We show that correctly folded PfEMP1 domains elicit high antibody titres and induce a homogeneous response in outbred and BALB/c mice after three injections. High levels of rosette-disrupting and rosette-preventing antibodies are induced by DBL1 and the Head domain. Reduced-alkylated or denatured proteins fail to induce surface-reacting and rosette-disrupting antibodies, indicating that surface epitopes are conformational. We also report limited cross-reactivity between some PfEMP1 VarO domains. These results highlight the high immunogenicity of the individual domains in outbred animals and provide a strong basis for a rational vaccination strategy targeting rosetting.


Assuntos
Adesinas Bacterianas/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adesinas Bacterianas/química , Adesinas Bacterianas/genética , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Reações Cruzadas , Epitopos/química , Epitopos/genética , Eritrócitos/parasitologia , Feminino , Humanos , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Formação de Roseta
5.
PLoS One ; 10(4): e0123567, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25886591

RESUMO

The malaria parasite Plasmodium knowlesi, previously associated only with infection of macaques, is now known to infect humans as well and has become a significant public health problem in Southeast Asia. This species should therefore be targeted in vaccine and therapeutic strategies against human malaria. Apical Membrane Antigen 1 (AMA1), which plays a role in Plasmodium merozoite invasion of the erythrocyte, is currently being pursued in human vaccine trials against P. falciparum. Recent vaccine trials in macaques using the P. knowlesi orthologue PkAMA1 have shown that it protects against infection by this parasite species and thus should be developed for human vaccination as well. Here, we present the crystal structure of Domains 1 and 2 of the PkAMA1 ectodomain, and of its complex with the invasion-inhibitory monoclonal antibody R31C2. The Domain 2 (D2) loop, which is displaced upon binding the Rhoptry Neck Protein 2 (RON2) receptor, makes significant contacts with the antibody. R31C2 inhibits binding of the Rhoptry Neck Protein 2 (RON2) receptor by steric blocking of the hydrophobic groove and by preventing the displacement of the D2 loop which is essential for exposing the complete binding site on AMA1. R31C2 recognizes a non-polymorphic epitope and should thus be cross-strain reactive. PkAMA1 is much less polymorphic than the P. falciparum and P. vivax orthologues. Unlike these two latter species, there are no polymorphic sites close to the RON2-binding site of PkAMA1, suggesting that P. knowlesi has not developed a mechanism of immune escape from the host's humoral response to AMA1.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Protozoários/química , Proteínas de Membrana/química , Plasmodium knowlesi/imunologia , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/imunologia , Sequência de Bases , Cristalografia por Raios X , Proteínas de Membrana/imunologia , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Proteínas de Protozoários/imunologia
6.
PLoS One ; 10(4): e0124400, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25881166

RESUMO

Infection with Plasmodium knowlesi, a zoonotic primate malaria, is a growing human health problem in Southeast Asia. P. knowlesi is being used in malaria vaccine studies, and a number of proteins are being considered as candidate malaria vaccine antigens, including the Apical Membrane Antigen 1 (AMA1). In order to determine genetic diversity of the ama1 gene and to identify epitopes of AMA1 under strongest immune selection, the ama1 gene of 52 P. knowlesi isolates derived from human infections was sequenced. Sequence analysis of isolates from two geographically isolated regions in Sarawak showed that polymorphism in the protein is low compared to that of AMA1 of the major human malaria parasites, P. falciparum and P. vivax. Although the number of haplotypes was 27, the frequency of mutations at the majority of the polymorphic positions was low, and only six positions had a variance frequency higher than 10%. Only two positions had more than one alternative amino acid. Interestingly, three of the high-frequency polymorphic sites correspond to invariant sites in PfAMA1 or PvAMA1. Statistically significant differences in the quantity of three of the six high frequency mutations were observed between the two regions. These analyses suggest that the pkama1 gene is not under balancing selection, as observed for pfama1 and pvama1, and that the PkAMA1 protein is not a primary target for protective humoral immune responses in their reservoir macaque hosts, unlike PfAMA1 and PvAMA1 in humans. The low level of polymorphism justifies the development of a single allele PkAMA1-based vaccine.


Assuntos
Antígenos de Protozoários/genética , Haplótipos/genética , Malária/genética , Proteínas de Membrana/genética , Mutação/genética , Plasmodium knowlesi/isolamento & purificação , Polimorfismo Genético/genética , Proteínas de Protozoários/genética , Seleção Genética/genética , Sequência de Aminoácidos , Humanos , Malária/parasitologia , Dados de Sequência Molecular , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
7.
J Mol Biol ; 425(10): 1697-711, 2013 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-23429057

RESUMO

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1), a family of adhesins of the falciparum species of the malaria parasite, is exposed on the surface of the infected erythrocyte. In general, only one PfEMP1 variant is expressed at a time but switching between variants occurs, changing both host-cell receptor specificity and serotype. The PfEMP1 variant VAR2CSA causes sequestration of infected erythrocytes in the intervillous spaces of the placenta via the glycosaminoglycan chondroitin sulfate A. This leads to pregnancy-associated malaria, which has severe consequences for the fetus and mother. The extracellular region of VAR2CSA comprises six DBL (Duffy-binding-like) domains and a single CIDR (cysteine-rich inter-domain region) domain. The C-terminal domain DBL6ε, the most polymorphic domain of VAR2CSA, has seven regions of high variability termed variable blocks (VBs). Here we have determined the crystal structure of DBL6ε from the FCR3 parasite line and have compared it with the previously determined structure of that from the 3D7 line. We found significant differences particularly in the N-terminal region, which contains the first VB (VB1). Although DBL6ε is the most variable VAR2CSA domain, DBL6ε-FCR3 and DBL6ε-3D7 react with IgG purified from immune sera of pregnant women. Furthermore, IgG purified on one domain cross-reacts with the other, confirming the presence of cross-reactive epitopes. We also examined reactivity of immune sera to the four least variable VB (VB1, VB2, VB4 and VB5) using peptides with the consensus sequence closest, in turn, to the FCR3 or 3D7 domain. These results provide new molecular insights into immune escape by parasites expressing the VAR2CSA variant.


Assuntos
Antígenos de Protozoários/química , Malária Falciparum/imunologia , Malária Falciparum/metabolismo , Plasmodium falciparum/química , Complicações Parasitárias na Gravidez/imunologia , Proteínas de Protozoários/química , Sequência de Aminoácidos , Antígenos de Protozoários/imunologia , Cristalografia por Raios X , Feminino , Variação Genética/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Malária Falciparum/parasitologia , Dados de Sequência Molecular , Placenta/química , Placenta/imunologia , Placenta/parasitologia , Plasmodium falciparum/classificação , Plasmodium falciparum/imunologia , Gravidez , Complicações Parasitárias na Gravidez/metabolismo , Complicações Parasitárias na Gravidez/parasitologia , Estrutura Terciária de Proteína/genética , Proteínas de Protozoários/imunologia
8.
PLoS Pathog ; 8(7): e1002781, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22807674

RESUMO

The ABO blood group influences susceptibility to severe Plasmodium falciparum malaria. Recent evidence indicates that the protective effect of group O operates by virtue of reduced rosetting of infected red blood cells (iRBCs) with uninfected RBCs. Rosetting is mediated by a subgroup of PfEMP1 adhesins, with RBC binding being assigned to the N-terminal DBL1α1 domain. Here, we identify the ABO blood group as the main receptor for VarO rosetting, with a marked preference for group A over group B, which in turn is preferred to group O RBCs. We show that recombinant NTS-DBL1α1 and NTS-DBL1α1-CIDR1γ reproduce the VarO-iRBC blood group preference and document direct binding to blood group trisaccharides by surface plasmon resonance. More detailed RBC subgroup analysis showed preferred binding to group A1, weaker binding to groups A2 and B, and least binding to groups A(x) and O. The 2.8 Å resolution crystal structure of the PfEMP1-VarO Head region, NTS-DBL1α1-CIDR1γ, reveals extensive contacts between the DBL1α1 and CIDR1γ and shows that the NTS-DBL1α1 hinge region is essential for RBC binding. Computer docking of the blood group trisaccharides and subsequent site-directed mutagenesis localized the RBC-binding site to the face opposite to the heparin-binding site of NTS-DBLα1. RBC binding involves residues that are conserved between rosette-forming PfEMP1 adhesins, opening novel opportunities for intervention against severe malaria. By deciphering the structural basis of blood group preferences in rosetting, we provide a link between ABO blood grouppolymorphisms and rosette-forming adhesins, consistent with the selective role of falciparum malaria on human genetic makeup.


Assuntos
Sistema ABO de Grupos Sanguíneos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Formação de Roseta , Sistema ABO de Grupos Sanguíneos/imunologia , Sequência de Aminoácidos , Anticorpos Antiprotozoários/imunologia , Sítios de Ligação , Cristalografia por Raios X , Eritrócitos/imunologia , Eritrócitos/metabolismo , Humanos , Reação de Imunoaderência , Malária Falciparum/sangue , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmodium falciparum/genética , Plasmodium falciparum/ultraestrutura , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia
9.
PLoS Pathog ; 8(6): e1002755, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737069

RESUMO

Members of the phylum Apicomplexa, which include the malaria parasite Plasmodium, share many features in their invasion mechanism in spite of their diverse host cell specificities and life cycle characteristics. The formation of a moving junction (MJ) between the membranes of the invading apicomplexan parasite and the host cell is common to these intracellular pathogens. The MJ contains two key parasite components: the surface protein Apical Membrane Antigen 1 (AMA1) and its receptor, the Rhoptry Neck Protein (RON) complex, which is targeted to the host cell membrane during invasion. In particular, RON2, a transmembrane component of the RON complex, interacts directly with AMA1. Here, we report the crystal structure of AMA1 from Plasmodium falciparum in complex with a peptide derived from the extracellular region of PfRON2, highlighting clear specificities of the P. falciparum RON2-AMA1 interaction. The receptor-binding site of PfAMA1 comprises the hydrophobic groove and a region that becomes exposed by displacement of the flexible Domain II loop. Mutations of key contact residues of PfRON2 and PfAMA1 abrogate binding between the recombinant proteins. Although PfRON2 contacts some polymorphic residues, binding studies with PfAMA1 from different strains show that these have little effect on affinity. Moreover, we demonstrate that the PfRON2 peptide inhibits erythrocyte invasion by P. falciparum merozoites and that this strong inhibitory potency is not affected by AMA1 polymorphisms. In parallel, we have determined the crystal structure of PfAMA1 in complex with the invasion-inhibitory peptide R1 derived by phage display, revealing an unexpected structural mimicry of the PfRON2 peptide. These results identify the key residues governing the interactions between AMA1 and RON2 in P. falciparum and suggest novel approaches to antimalarial therapeutics.


Assuntos
Antígenos de Protozoários/química , Interações Hospedeiro-Parasita/fisiologia , Proteínas de Membrana/química , Plasmodium falciparum/química , Proteínas de Protozoários/química , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/metabolismo , Membrana Celular/metabolismo , Cristalização , Proteínas de Membrana/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium falciparum/metabolismo , Polimorfismo Genético , Ligação Proteica , Estrutura Quaternária de Proteína , Proteínas de Protozoários/metabolismo , Ressonância de Plasmônio de Superfície
10.
PLoS Negl Trop Dis ; 5(11): e1375, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22069505

RESUMO

Patients with Chronic Chagas' Heart Disease possess high levels of antibodies against the carboxyl-terminal end of the ribosomal P2ß protein of Trypanosoma cruzi (TcP2ß). These antibodies, as well as the murine monoclonal antibody (mAb) 17.2, recognize the last 13 amino acids of TcP2ß (called the R13 epitope: EEEDDDMGFGLFD) and are able to cross-react with, and stimulate, the ß1 adrenergic receptor (ß1-AR). Indeed, the mAb 17.2 was able to specifically detect human ß1-AR, stably transfected into HEK cells, by flow cytometry and to induce repolarisation abnormalities and first degree atrioventricular conduction block after passive transfer to naïve mice. To study the structural basis of this cross-reactivity, we determined the crystal structure of the Fab region of the mAb 17.2 alone at 2.31 Å resolution and in complex with the R13 peptide at 1.89 Å resolution. We identified as key contact residues on R13 peptide Glu3, Asp6 and Phe9 as was previously shown by alanine scanning. Additionally, we generated a model of human ß1-AR to elucidate the interaction with anti-R13 antibodies. These data provide an understanding of the molecular basis of cross-reactive antibodies induced by chronic infection with Trypanosoma cruzi.


Assuntos
Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/química , Anticorpos Antiprotozoários/imunologia , Fosfoproteínas/química , Fosfoproteínas/imunologia , Proteínas Ribossômicas/química , Proteínas Ribossômicas/imunologia , Trypanosoma cruzi/química , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Antiprotozoários/metabolismo , Reações Cruzadas , Cristalografia por Raios X , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Modelos Moleculares , Fosfoproteínas/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Receptores Adrenérgicos beta 1/imunologia , Receptores Adrenérgicos beta 1/metabolismo , Proteínas Ribossômicas/metabolismo
11.
PLoS One ; 6(5): e20270, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21625526

RESUMO

Var2CSA, a key molecule linked with pregnancy-associated malaria (PAM), causes sequestration of Plasmodium falciparum infected erythrocytes (PEs) in the placenta by adhesion to chondroitin sulfate A (CSA). Var2CSA possesses a 300 kDa extracellular region composed of six Duffy-binding like (DBL) domains and a cysteine-rich interdomain region (CIDRpam) module. Although initial studies implicated several individual var2CSA DBL domains as important for adhesion of PEs to CSA, new studies revealed that these individual domains lack both the affinity and specificity displayed by the full-length extracellular region. Indeed, recent evidence suggests the presence of a single CSA-binding site formed by a higher-order domain organization rather than several independent binding sites located on the different domains. Here, we search for the minimal binding region within var2CSA that maintains high affinity and specificity for CSA binding, a characteristic feature of the full-length extracellular region. Accordingly, truncated recombinant var2CSA proteins comprising different domain combinations were expressed and their binding characteristics assessed against different sulfated glycosaminoglycans (GAGs). Our results indicate that the smallest region within var2CSA with similar binding properties to those of the full-length var2CSA is DBL1X-3X. We also demonstrate that inhibitory antibodies raised in rabbit against the full-length DBL1X-6ε target principally DBL3X and, to a lesser extent, DBL5ε. Taken together, our results indicate that efforts should focus on the DBL1X-3X region for developing vaccine and therapeutic strategies aimed at combating PAM.


Assuntos
Antígenos de Protozoários/metabolismo , Sulfatos de Condroitina/metabolismo , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Primers do DNA , Humanos , Reação em Cadeia da Polimerase
12.
Proc Natl Acad Sci U S A ; 108(13): 5243-8, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21402930

RESUMO

The human malaria parasite Plasmodium falciparum can cause infected red blood cells (iRBC) to form rosettes with uninfected RBC, a phenotype associated with severe malaria. Rosetting is mediated by a subset of the Plasmodium falciparum membrane protein 1 (PfEMP1) variant adhesins expressed on the infected host-cell surface. Heparin and other sulfated oligosaccharides, however, can disrupt rosettes, suggesting that therapeutic approaches to this form of severe malaria are feasible. We present a structural and functional study of the N-terminal domain of PfEMP1 from the VarO variant comprising the N-terminal segment (NTS) and the first DBL domain (DBL1α(1)), which is directly implicated in rosetting. We demonstrate that NTS-DBL1α(1)-VarO binds to RBC and that heparin inhibits this interaction in a dose-dependent manner, thus mimicking heparin-mediated rosette disruption. We have determined the crystal structure of NTS-DBL1α(1), showing that NTS, previously thought to be a structurally independent component of PfEMP1, forms an integral part of the DBL1α domain. Using mutagenesis and docking studies, we have located the heparin-binding site, which includes NTS. NTS, unique to the DBL α-class domain, is thus an intrinsic structural and functional component of the N-terminal VarO domain. The specific interaction observed with heparin opens the way for developing antirosetting therapeutic strategies.


Assuntos
Eritrócitos/parasitologia , Heparina/metabolismo , Plasmodium falciparum/metabolismo , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Formação de Roseta , Sequência de Aminoácidos , Cristalografia por Raios X , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
13.
PLoS One ; 6(1): e16544, 2011 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-21298021

RESUMO

The clonally variant Plasmodium falciparum PfEMP1 adhesin is a virulence factor and a prime target of humoral immunity. It is encoded by a repertoire of functionally differentiated var genes, which display architectural diversity and allelic polymorphism. Their serological relationship is key to understanding the evolutionary constraints on this gene family and rational vaccine design. Here, we investigated the Palo Alto/VarO and IT4/R29 and 3D7/PF13_003 parasites lines. VarO and R29 form rosettes with uninfected erythrocytes, a phenotype associated with severe malaria. They express an allelic Cys2/group A NTS-DBL1α(1) PfEMP1 domain implicated in rosetting, whose 3D7 ortholog is encoded by PF13_0003. Using these three recombinant NTS-DBL1α(1) domains, we elicited antibodies in mice that were used to develop monovariant cultures by panning selection. The 3D7/PF13_0003 parasites formed rosettes, revealing a correlation between sequence identity and virulence phenotype. The antibodies cross-reacted with the allelic domains in ELISA but only minimally with the Cys4/group B/C PFL1955w NTS-DBL1α. By contrast, they were variant-specific in surface seroreactivity of the monovariant-infected red cells by FACS analysis and in rosette-disruption assays. Thus, while ELISA can differentiate serogroups, surface reactivity assays define the more restrictive serotypes. Irrespective of cumulated exposure to infection, antibodies acquired by humans living in a malaria-endemic area also displayed a variant-specific surface reactivity. Although seroprevalence exceeded 90% for each rosetting line, the kinetics of acquisition of surface-reactive antibodies differed in the younger age groups. These data indicate that humans acquire an antibody repertoire to non-overlapping serotypes within a serogroup, consistent with an antibody-driven diversification pressure at the population level. In addition, the data provide important information for vaccine design, as production of a vaccine targeting rosetting PfEMP1 adhesins will require engineering to induce variant-transcending responses or combining multiple serotypes to elicit a broad spectrum of immunity.


Assuntos
Epitopos/imunologia , Membrana Eritrocítica/imunologia , Variação Genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Alelos , Animais , Anticorpos Antiprotozoários/biossíntese , Epitopos/genética , Membrana Eritrocítica/parasitologia , Imunidade Humoral/efeitos dos fármacos , Vacinas Antimaláricas , Camundongos , Plasmodium falciparum/imunologia , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/imunologia
14.
PLoS Pathog ; 7(2): e1001276, 2011 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-21347343

RESUMO

Obligate intracellular Apicomplexa parasites share a unique invasion mechanism involving a tight interaction between the host cell and the parasite surfaces called the moving junction (MJ). The MJ, which is the anchoring structure for the invasion process, is formed by secretion of a macromolecular complex (RON2/4/5/8), derived from secretory organelles called rhoptries, into the host cell membrane. AMA1, a protein secreted from micronemes and associated with the parasite surface during invasion, has been shown in vitro to bind the MJ complex through a direct association with RON2. Here we show that RON2 is inserted as an integral membrane protein in the host cell and, using several interaction assays with native or recombinant proteins, we define the region that binds AMA1. Our studies were performed both in Toxoplasma gondii and Plasmodium falciparum and although AMA1 and RON2 proteins have diverged between Apicomplexa species, we show an intra-species conservation of their interaction. More importantly, invasion inhibition assays using recombinant proteins demonstrate that the RON2-AMA1 interaction is crucial for both T. gondii and P. falciparum entry into their host cells. This work provides the first evidence that AMA1 uses the rhoptry neck protein RON2 as a receptor to promote invasion by Apicomplexa parasites.


Assuntos
Antígenos de Protozoários/metabolismo , Apicomplexa/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Proteínas de Protozoários/metabolismo , Internalização do Vírus , Animais , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Apicomplexa/genética , Apicomplexa/metabolismo , Células Cultivadas , Chlorocebus aethiops , Conexinas/metabolismo , Sequência Conservada , Interações Hospedeiro-Parasita/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Modelos Biológicos , Modelos Moleculares , Parasitos/genética , Parasitos/metabolismo , Parasitos/fisiologia , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/fisiologia , Ligação Proteica/genética , Domínios e Motivos de Interação entre Proteínas/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasma/fisiologia , Células Vero
15.
Mol Biochem Parasitol ; 173(2): 115-22, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20562018

RESUMO

Pregnancy-associated malaria (PAM) arises from sequestration of Plasmodium falciparum-parasitized erythrocytes (PE) in the placenta, leading to chronic symptoms in the expectant mother and serious consequences for fetal development. Placental sequestration has been linked to binding of chondroitin sulphate A (CSA) by the var2CSA variant of PfEMP1 expressed on the PE surface, and a substantial body of evidence shows that the immune response to var2CSA gives an effective protection against PAM. We have expressed the var2CSA-DBL5epsilon domain, derived from a placental isolate from Senegal, as soluble product in Escherichia coli and have shown using different criteria that the recombinant protein is obtained with the native conformation. Using surface plasmon resonance techniques, we have examined binding of DBL5epsilon to placental chondroitin sulphate proteoglycan and CSA; however, the recombinant protein also binds to other sulphated oligosaccharides, with higher affinity in some cases, indicating that the single domain lacks the specificity for CSA shown by the complete extra-cellular region of var2CSA and placental parasites. Recombinant DBL5epsilon was specifically recognized by sera from malaria-exposed Senegalese women in a parity-dependent manner but by sera not from children or males from the same endemic region. Conversely, DBL5epsilon induced antibodies in mice that recognized placental isolates from Benin but not isolates from children. The presence of universal epitopes thus supports DBL5epsilon as an interesting component of var2CSA to be considered for vaccine development.


Assuntos
Antígenos de Protozoários/imunologia , Placenta/parasitologia , Plasmodium falciparum/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/sangue , Benin , Criança , Pré-Escolar , Sulfatos de Condroitina/metabolismo , Clonagem Molecular , Escherichia coli/genética , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Plasmodium falciparum/isolamento & purificação , Gravidez , Complicações Infecciosas na Gravidez/parasitologia , Ligação Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Senegal , Alinhamento de Sequência , Ressonância de Plasmônio de Superfície
16.
Proc Natl Acad Sci U S A ; 107(11): 4884-9, 2010 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-20194779

RESUMO

Pregnancy-associated malaria (PAM) is a serious consequence of sequestration of Plasmodium falciparum-parasitized erythrocytes (PE) in the placenta through adhesion to chondroitin sulfate A (CSA) present on placental proteoglycans. Recent work implicates var2CSA, a member of the PfEMP1 family, as the mediator of placental sequestration and as a key target for PAM vaccine development. Var2CSA is a 350 kDa transmembrane protein, whose extracellular region includes six Duffy-binding-like (DBL) domains. Due to its size and high cysteine content, the full-length var2CSA extracellular region has not hitherto been expressed in heterologous systems, thus limiting investigations to individual recombinant domains. Here we report for the first time the expression of the full-length var2CSA extracellular region (domains DBL1X to DBL6epsilon) from the 3D7 parasite strain using the human embryonic kidney 293 cell line. We show that the recombinant extracellular var2CSA region is correctly folded and that, unlike the individual DBL domains, it binds with high affinity and specificity to CSA (K(D) = 61 nM) and efficiently inhibits PE from binding to CSA. Structural characterization by analytical ultracentrifugation and small-angle x-ray scattering reveals a compact organization of the full-length protein, most likely governed by specific interdomain interactions, rather than an extended structure. Collectively, these data suggest that a high-affinity, CSA-specific binding site is formed by the higher-order structure of the var2CSA extracellular region. These results have important consequences for the development of an effective vaccine and therapeutic inhibitors.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Sulfatos de Condroitina/metabolismo , Espaço Extracelular/química , Plasmodium falciparum/metabolismo , Animais , Linhagem Celular , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Dicroísmo Circular , Decorina , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Proteínas da Matriz Extracelular/metabolismo , Feminino , Humanos , Cinética , Modelos Moleculares , Parasitos/metabolismo , Placenta/metabolismo , Gravidez , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteoglicanas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Mol Biochem Parasitol ; 170(2): 84-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20045435

RESUMO

Rosetting of erythrocytes infected with Plasmodium falciparum is frequently observed in children with severe malaria. This adhesion phenomenon has been linked to the DBL1alpha domain of P. falciparum erythrocyte membrane protein 1 (PfEMP1) in three laboratory clones: FCR3S1.2, IT4R29 and Palo Alto varO. Here, we compare the soluble recombinant NTS-DBL1alpha(1)-varO domain (NTS: N-terminal segment) obtained from E. coli, Pichia pastoris and baculovirus/insect cell expression systems. In each case, the presence of NTS was necessary for obtaining a soluble product. Successful expression in E. coli required maltose-binding protein as an N-terminal fusion partner. Each expression system produced an identical, correctly folded protein, as judged by biochemical and biophysical characterisations, and by the capacity to elicit antibodies that react with the surface of VarO-infected erythrocytes and disrupt VarO rosettes. Binding studies using surface plasmon resonance (SPR) techniques showed that NTS-DBL1alpha(1) produced in E. coli binds to heparin with micromolar affinity. IC(50) constants for other sulphated oligosaccharides were determined using SPR by measuring their competitive binding to the soluble protein in the presence of immobilized heparin. The affinity to NTS-DBL1alpha(1) was related to the degree of sulphation of the oligosaccharide, although the position of the sulphate groups on the sugar rings was also important. VarO rosettes could be disrupted by sulphated oligosaccharides with an efficacy that correlated with their binding affinity to recombinant NTS-DBL1alpha(1). Thus high yields of soluble NTS-DBL1alpha(1) with native conformation have been produced, opening novel perspectives for both structure-function studies and vaccine development.


Assuntos
Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Animais , Anticorpos Antiprotozoários/imunologia , Baculoviridae/genética , Adesão Celular , Linhagem Celular , Eritrócitos/parasitologia , Escherichia coli/genética , Feminino , Heparina/metabolismo , Humanos , Insetos , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Formação de Roseta , Ressonância de Plasmônio de Superfície
18.
J Mol Biol ; 388(4): 839-50, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19328810

RESUMO

The use of carbohydrate-mimicking peptides to induce immune responses against surface polysaccharides of pathogenic bacteria offers a novel approach to vaccine development. Factors governing antigenic and immunogenic mimicry, however, are complex and poorly understood. We have addressed this question using the anti-lipopolysaccharide monoclonal antibody F22-4, which was raised against Shigella flexneri serotype 2a and shown to protect against homologous infection in a mouse model. In a previous crystallographic study, we described F22-4 in complex with two synthetic fragments of the O-antigen, the serotype-specific saccharide moiety of lipopolysaccharide. Here, we present a crystallographic and NMR study of the interaction of F22-4 with a dodecapeptide selected by phage display using the monoclonal antibody. Like the synthetic decasaccharide, the peptide binds to F22-4 with micromolar affinity. Although the peptide and decasaccharide use very similar regions of the antigen-binding site, indicating good antigenic mimicry, immunogenic mimicry by the peptide was not observed. The F22-4-antigen interaction is significantly more hydrophobic with the peptide than with oligosaccharides; nonetheless, all hydrogen bonds formed between the peptide and F22-4 have equivalents in the oligosaccharide complex. Two bridging water molecules are also in common, adding to partial structural mimicry. Whereas the bound peptide is entirely helical, its structure in solution, as shown by NMR, is helical in the central region only. Moreover, docking the NMR structure into the antigen-binding site shows that steric hindrance would occur, revealing poor complementarity between the major solution conformation and the antibody that could contribute to the absence of immunogenic mimicry.


Assuntos
Anticorpos , Mimetismo Molecular , Antígenos O , Peptídeos , Shigella flexneri/imunologia , Animais , Anticorpos/química , Anticorpos/imunologia , Sequência de Carboidratos , Cristalografia por Raios X , Camundongos , Conformação Molecular , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Antígenos O/química , Antígenos O/imunologia , Peptídeos/química , Peptídeos/imunologia , Conformação Proteica , Termodinâmica
19.
Infect Immun ; 76(12): 5565-80, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18809668

RESUMO

In the Saimiri sciureus monkey, erythrocytes infected with the varO antigenic variant of the Plasmodium falciparum Palo Alto 89F5 clone bind uninfected red blood cells (rosetting), form autoagglutinates, and have a high multiplication rate, three phenotypic characteristics that are associated with severe malaria in human patients. We report here that varO parasites express a var gene having the characteristics of group A var genes, and we show that the varO Duffy binding-like 1alpha(1) (DBL1alpha(1)) domain is implicated in the rosetting of both S. sciureus and human erythrocytes. The soluble varO N-terminal sequence (NTS)-DBL1alpha(1) recombinant domain, produced in a baculovirus-insect cell system, induced high titers of antibodies that reacted with varO-infected red blood cells and disrupted varO rosettes. varO parasites were culture adapted in vitro using human erythrocytes. They formed rosettes and autoagglutinates, and they had the same surface serotype and expressed the same varO gene as the monkey-propagated parasites. To develop an in vitro model with highly homogeneous varO parasites, rosette purification was combined with positive selection by panning with a varO NTS-DBL1alpha(1)-specific mouse monoclonal antibody. The single-variant, clonal parasites were used to analyze seroprevalence for varO at the village level in a setting where malaria is holoendemic (Dielmo, Senegal). We found 93.6% (95% confidence interval, 89.7 to 96.4%) seroprevalence for varO surface-reacting antibodies and 86.7% (95% confidence interval, 82.8 to 91.6%) seroprevalence for the recombinant NTS-DBL1alpha(1) domain, and virtually all permanent residents had seroconverted by the age of 5 years. These data imply that the varO model is a relevant in vivo and in vitro model for rosetting and autoagglutination that can be used for rational development of vaccine candidates and therapeutic strategies aimed at preventing malaria pathology.


Assuntos
Anticorpos Antiprotozoários , Eritrócitos/metabolismo , Hemaglutinação , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Formação de Roseta/métodos , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Sequência de Bases , Técnicas de Cultura de Células , Células Cultivadas , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Malária Falciparum/sangue , Malária Falciparum/diagnóstico , Malária Falciparum/epidemiologia , Camundongos , Dados de Sequência Molecular , Doenças dos Macacos/diagnóstico , Plasmodium falciparum/imunologia , Plasmodium falciparum/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saimiri , Homologia de Sequência do Ácido Nucleico , Sorotipagem/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...