Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Science ; 359(6371): 48-55, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29302006

RESUMO

Plasmodium vivax shows a strict host tropism for reticulocytes. We identified transferrin receptor 1 (TfR1) as the receptor for P. vivax reticulocyte-binding protein 2b (PvRBP2b). We determined the structure of the N-terminal domain of PvRBP2b involved in red blood cell binding, elucidating the molecular basis for TfR1 recognition. We validated TfR1 as the biological target of PvRBP2b engagement by means of TfR1 expression knockdown analysis. TfR1 mutant cells deficient in PvRBP2b binding were refractory to invasion of P. vivax but not to invasion of P. falciparum Using Brazilian and Thai clinical isolates, we show that PvRBP2b monoclonal antibodies that inhibit reticulocyte binding also block P. vivax entry into reticulocytes. These data show that TfR1-PvRBP2b invasion pathway is critical for the recognition of reticulocytes during P. vivax invasion.


Assuntos
Antígenos CD/metabolismo , Malária Vivax/metabolismo , Malária Vivax/parasitologia , Proteínas de Membrana/química , Plasmodium vivax/patogenicidade , Proteínas de Protozoários/química , Receptores da Transferrina/metabolismo , Reticulócitos/parasitologia , Antígenos CD/genética , Cristalografia por Raios X , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Parasita , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana/ultraestrutura , Plasmodium vivax/metabolismo , Domínios Proteicos , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/ultraestrutura , Receptores da Transferrina/genética
2.
PLoS Negl Trop Dis ; 10(9): e0005014, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27677183

RESUMO

BACKGROUND: Major gaps in our understanding of Plasmodium vivax biology and the acquisition of immunity to this parasite hinder vaccine development. P. vivax merozoites exclusively invade reticulocytes, making parasite proteins that mediate reticulocyte binding and/or invasion potential key vaccine or drug targets. While protein interactions that mediate invasion are still poorly understood, the P. vivax Reticulocyte-Binding Protein family (PvRBP) is thought to be involved in P. vivax restricted host-cell selectivity. METHODOLOGY/PRINCIPAL FINDINGS: We assessed the binding specificity of five members of the PvRBP family (PvRBP1a, PvRBP1b, PvRBP2a, PvRBP2b, PvRBP2-P2 and a non-binding fragment of PvRBP2c) to normocytes or reticulocytes. PvRBP2b was identified as the only reticulocyte-specific binder (P<0.001), whereas the others preferentially bound to normocytes (PvRBP1a/b P≤0.034), or showed comparable binding to both (PvRBP2a/2-P2, P = 0.38). Furthermore, we measured levels of total and IgG subclasses 1, 2, 3 and 4 to the six PvRBPs in a cohort of young Papua New Guinean children, and assessed their relationship with prospective risk of P. vivax malaria. Children had substantial, highly correlated (rho = 0.49-0.82, P<0.001) antibody levels to all six PvRBPs, with dominant IgG1 and IgG3 subclasses. Both total IgG (Incidence Rate Ratio [IRR] 0.63-0.73, P = 0.008-0.041) and IgG1 (IRR 0.56-0.69, P = 0.001-0.035) to PvRBP2b and PvRBP1a were strongly associated with reduced risk of vivax-malaria, independently of age and exposure. CONCLUSION/SIGNIFICANCE: These results demonstrate a diversity of erythrocyte-binding phenotypes of PvRBPs, indicating binding to both reticulocyte-specific and normocyte-specific ligands. Our findings provide further insights into the naturally acquired immunity to P. vivax and highlight the importance of PvRBP proteins as targets of naturally acquired humoral immunity. In-depth studies of the role of PvRBPs in P. vivax invasion and functional validation of the role of anti-PvRBP antibodies in clinical immunity against P. vivax are now required to confirm the potential of the reticulocyte-binding PvRBP2b and PvRBP1a as vaccine candidate antigens.

3.
Cell Host Microbe ; 20(1): 60-71, 2016 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-27374406

RESUMO

Plasmodium falciparum parasites in the merozoite stage invade human erythrocytes and cause malaria. Invasion requires multiple interactions between merozoite ligands and erythrocyte receptors. P. falciparum reticulocyte binding homolog 5 (PfRh5) forms a complex with the PfRh5-interacting protein (PfRipr) and Cysteine-rich protective antigen (CyRPA) and binds erythrocytes via the host receptor basigin. However, the specific role that PfRipr and CyRPA play during invasion is unclear. Using P. falciparum lines conditionally expressing PfRipr and CyRPA, we show that loss of PfRipr or CyRPA function blocks growth due to the inability of merozoites to invade erythrocytes. Super-resolution microscopy revealed that PfRipr, CyRPA, and PfRh5 colocalize at the junction between merozoites and erythrocytes during invasion. PfRipr, CyRPA, and PfRipr/CyRPA/PfRh5-basigin complex is required for triggering the Ca(2+) release and establishing the tight junction. Together, these results establish that the PfRh5/PfRipr/CyRPA complex is essential in the sequential molecular events leading to parasite invasion of human erythrocytes.


Assuntos
Antígenos de Protozoários/metabolismo , Proteínas de Transporte/metabolismo , Endocitose , Eritrócitos/parasitologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Basigina/metabolismo , Cálcio/metabolismo , Cátions Bivalentes/metabolismo , Técnicas de Silenciamento de Genes , Interações Hospedeiro-Patógeno , Humanos , Microscopia , Modelos Biológicos , Ligação Proteica , Multimerização Proteica
4.
Proc Natl Acad Sci U S A ; 113(2): E191-200, 2016 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-26715754

RESUMO

Understanding how malaria parasites gain entry into human red blood cells is essential for developing strategies to stop blood stage infection. Plasmodium vivax preferentially invades reticulocytes, which are immature red blood cells. The organism has two erythrocyte-binding protein families: namely, the Duffy-binding protein (PvDBP) and the reticulocyte-binding protein (PvRBP) families. Several members of the PvRBP family bind reticulocytes, specifically suggesting a role in mediating host cell selectivity of P. vivax. Here, we present, to our knowledge, the first high-resolution crystal structure of an erythrocyte-binding domain from PvRBP2a, solved at 2.12 Å resolution. The monomeric molecule consists of 10 α-helices and one short ß-hairpin, and, although the structural fold is similar to that of PfRh5--the essential invasion ligand in Plasmodium falciparum--its surface properties are distinct and provide a possible mechanism for recognition of alternate receptors. Sequence alignments of the crystallized fragment of PvRBP2a with other PvRBPs highlight the conserved placement of disulfide bonds. PvRBP2a binds mature red blood cells through recognition of an erythrocyte receptor that is neuraminidase- and chymotrypsin-resistant but trypsin-sensitive. By examining the patterns of sequence diversity within field isolates, we have identified and mapped polymorphic residues to the PvRBP2a structure. Using mutagenesis, we have also defined the critical residues required for erythrocyte binding. Characterization of the structural features that govern functional erythrocyte binding for the PvRBP family provides a framework for generating new tools that block P. vivax blood stage infection.


Assuntos
Sequência Conservada , Eritrócitos/metabolismo , Plasmodium vivax/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Área Sob a Curva , Sequência de Bases , Cristalografia por Raios X , Evolução Molecular , Frequência do Gene , Genes de Protozoários , Haplótipos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium vivax/genética , Polimorfismo de Nucleotídeo Único/genética , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Espalhamento a Baixo Ângulo , Alinhamento de Sequência
5.
PLoS Pathog ; 11(12): e1005343, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26694741

RESUMO

The most severe form of malaria in humans is caused by the protozoan parasite Plasmodium falciparum. The invasive form of malaria parasites is termed a merozoite and it employs an array of parasite proteins that bind to the host cell to mediate invasion. In Plasmodium falciparum, the erythrocyte binding-like (EBL) and reticulocyte binding-like (Rh) protein families are responsible for binding to specific erythrocyte receptors for invasion and mediating signalling events that initiate active entry of the malaria parasite. Here we have addressed the role of the cytoplasmic tails of these proteins in activating merozoite invasion after receptor engagement. We show that the cytoplasmic domains of these type 1 membrane proteins are phosphorylated in vitro. Depletion of PfCK2, a kinase implicated to phosphorylate these cytoplasmic tails, blocks P. falciparum invasion of red blood cells. We identify the crucial residues within the PfRh4 cytoplasmic domain that are required for successful parasite invasion. Live cell imaging of merozoites from these transgenic mutants show they attach but do not penetrate erythrocytes implying the PfRh4 cytoplasmic tail conveys signals important for the successful completion of the invasion process.


Assuntos
Eritrócitos/microbiologia , Malária Falciparum/metabolismo , Fosfotransferases/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Humanos , Merozoítos/metabolismo , Dados de Sequência Molecular , Fosforilação , Plasmodium falciparum/patogenicidade
6.
J Biol Chem ; 290(42): 25307-21, 2015 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-26324715

RESUMO

Plasmodium falciparum parasites must invade red blood cells to survive within humans. Entry into red blood cells is governed by interactions between parasite adhesins and red blood cell receptors. Previously we identified that P. falciparum reticulocyte binding protein-like homologue 4 (PfRh4) binds to complement receptor 1 (CR1) to mediate entry of malaria parasites into human red blood cells. In this report we characterize a collection of anti-PfRh4 monoclonal antibodies and CR1 protein fragments that modulate the interaction between PfRh4 and CR1. We identify an anti-PfRh4 monoclonal that blocks PfRh4-CR1 interaction in vitro, inhibits PfRh4 binding to red blood cells, and as a result abolishes the PfRh4-CR1 invasion pathway in P. falciparum. Epitope mapping of anti-PfRh4 monoclonal antibodies identified distinct functional regions within PfRh4 involved in modulating its interaction with CR1. Furthermore, we designed a set of protein fragments based on extensive mutagenesis analyses of the PfRh4 binding site on CR1 and determined their interaction affinities using surface plasmon resonance. These CR1 protein fragments bind tightly to PfRh4 and also function as soluble inhibitors to block PfRh4 binding to red blood cells and to inhibit the PfRh4-CR1 invasion pathway. Our findings can aid future efforts in designing specific single epitope antibodies to block P. falciparum invasion via complement receptor 1.


Assuntos
Anticorpos Monoclonais/imunologia , Eritrócitos/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Protozoários/metabolismo , Receptores de Complemento/metabolismo , Animais , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/imunologia
7.
Trends Parasitol ; 30(2): 75-84, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24393571

RESUMO

Schistosomiasis is one of the world's major neglected tropical diseases. Recent advances in schistosome genomics and transcriptomics have identified components of an intrinsic, B cell lymphoma-2 (Bcl-2)-regulated apoptotic cell death pathway. Molecular characterization of this pathway demonstrates its similarity to that in mammals. Gene expression and functional data indicate that apoptosis is active throughout the lifecycle. Moreover, drugs that activate apoptosis in human cells kill schistosome cells, raising the prospect of developing new treatments against schistosomiasis of humans. The development of new drugs is increasingly important in the face of the potential for resistance to currently available treatments, and the lack of an effective vaccine.


Assuntos
Anti-Helmínticos/uso terapêutico , Apoptose/fisiologia , Esquistossomose/tratamento farmacológico , Animais , Anti-Helmínticos/farmacologia , Apoptose/genética , Genes bcl-2/fisiologia , Genoma Helmíntico/genética , Genômica , Humanos , Schistosoma/efeitos dos fármacos , Schistosoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA