Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Nat Commun ; 12(1): 1750, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33741942

RESUMO

Malaria elimination requires tools that interrupt parasite transmission. Here, we characterize B cell receptor responses among Malian adults vaccinated against the first domain of the cysteine-rich 230 kDa gamete surface protein Pfs230, a key protein in sexual stage development of P. falciparum parasites. Among nine Pfs230 human monoclonal antibodies (mAbs) that we generated, one potently blocks transmission to mosquitoes in a complement-dependent manner and reacts to the gamete surface; the other eight show only low or no blocking activity. The structure of the transmission-blocking mAb in complex with vaccine antigen reveals a large discontinuous conformational epitope, specific to domain 1 of Pfs230 and comprising six structural elements in the protein. The epitope is conserved, suggesting the transmission-blocking mAb is broadly functional. This study provides a rational basis to improve malaria vaccines and develop therapeutic antibodies for malaria elimination.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/farmacologia , Epitopos/imunologia , Células Germinativas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/efeitos dos fármacos , Adulto , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Sítios de Ligação , Células Cultivadas , Epitopos/química , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/imunologia , Humanos , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Mosquitos Vetores/parasitologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/fisiologia , Conformação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia
2.
EBioMedicine ; 63: 103206, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33429173

RESUMO

BACKGROUND: To deeply understand the role of antibodies in the context of Trypanosoma cruzi infection, we decided to characterize A2R1, a parasite antibody selected from single-chain variable fragment (scFv) phage display libraries constructed from B cells of chronic Chagas heart disease patients. METHODS: Immunoblot, ELISA, cytometry, immunofluorescence and immunohistochemical assays were used to characterize A2R1 reactivity. To identify the antibody target, we performed an immunoprecipitation and two-dimensional electrophoresis coupled to mass spectrometry and confirmed A2R1 specific interaction by producing the antigen in different expression systems. Based on these data, we carried out a comparative in silico analysis of the protein target´s orthologues, focusing mainly on post-translational modifications. FINDINGS: A2R1 recognizes a parasite protein of ~50 kDa present in all life cycle stages of T. cruzi, as well as in other members of the kinetoplastid family, showing a defined immunofluorescence labeling pattern consistent with the cytoskeleton. A2R1 binds to tubulin, but this interaction relies on its post-translational modifications. Interestingly, this antibody also targets mammalian tubulin only present in brain, staining in and around cell bodies of the human peripheral and central nervous system. INTERPRETATION: Our findings demonstrate for the first time the existence of a human antibody against T. cruzi tubulin capable of cross-reacting with a human neural protein. This work re-emphasizes the role of molecular mimicry between host and parasitic antigens in the development of pathological manifestations of T. cruzi infection.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/farmacologia , Doença de Chagas/tratamento farmacológico , Doença de Chagas/parasitologia , Proteínas Recombinantes de Fusão/farmacologia , Trypanosoma cruzi/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/uso terapêutico , Especificidade de Anticorpos/imunologia , Antígenos de Protozoários/imunologia , Linhagem Celular , Clonagem Molecular , Reações Cruzadas/imunologia , Desenvolvimento de Medicamentos , Ensaio de Imunoadsorção Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Imunofluorescência , Expressão Gênica , Humanos , Imunoprecipitação , Espectrometria de Massas , Camundongos , Mimetismo Molecular , Ratos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/uso terapêutico , Análise de Sequência de DNA , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/farmacologia , Anticorpos de Cadeia Única/uso terapêutico
3.
PLoS One ; 14(10): e0223773, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31618282

RESUMO

Trypanosoma cruzi is a flagellate protozoan pathogen that causes Chagas disease. Currently there is no preventive treatment and the efficiency of the two drugs available is limited to the acute phase. Therefore, there is an unmet need for innovative tools to block transmission in endemic areas. In this study, we engineered a novel recombinant molecule able to adhere to the T. cruzi surface, termed scFv-10D8, that consists of a single-chain variable fragment (scFv) derived from mAb-10D8 that targets gp35/50. The synthetic gene encoding scFv-10D8 was cloned and fused to a 6×His tag and expressed in a prokaryotic expression system. Total periplasmic or 6xHis tag affinity-purified fractions of scFv-10D8 retained the capacity to bind to gp35/50, as shown by Western blot analyses. Pre-incubation of metacyclic trypomastigotes with scFv-10D8 showed a remarkable reduction in cell invasion capacity. Our results suggest that scFv-10D8 can be used in a paratransgenic approach to target parasites in insect vectors, avoiding dissemination of infective forms. Such advances in the development of this functional molecule will surely prompt the improvement of alternative strategies to control Chagas disease by targeting mammalian host stages.


Assuntos
Antígenos de Protozoários/imunologia , Engenharia de Proteínas/métodos , Anticorpos de Cadeia Única/genética , Trypanosoma cruzi/imunologia , Anticorpos Antiprotozoários/genética , Anticorpos Antiprotozoários/farmacologia , Linhagem Celular , Doença de Chagas/tratamento farmacológico , Doença de Chagas/parasitologia , Doença de Chagas/prevenção & controle , Células HeLa , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Anticorpos de Cadeia Única/farmacologia , Trypanosoma cruzi/efeitos dos fármacos
4.
Parasitol Int ; 69: 25-29, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30385417

RESUMO

Merozoite surface proteins (MSPs) are considered as promising blood-stage malaria vaccine candidates. MSP3 has long been evaluated for its vaccine candidacy, however, the candidacy of other members of MSP3 family is insufficiently characterized. Here, we investigated Plasmodium falciparum MSP11 (PF3D7_1036000), a member of the MSP3 family, for its potential as a blood-stage vaccine candidate. The full-length protein (MSP11-FL) as well as the N-terminal half-MSP11 (MSP11-N), known to be unique among the MSP3 family members, were expressed by wheat germ cell-free system, and used to raise antibodies in rabbit. Immunoblot analysis of schizont lysates probed with anti-MSP11-N antibodies detected double bands at approximately 40 and 60 kDa, consistent with the previous report thus confirming antibodies specificity. However, inconsistent with previously reported merozoite's surface localization, immunofluorescence assay (IFA) revealed that MSP11 likely localizes to rhoptry neck of merozoites in mature schizonts. After invasion, MSP11 localized to parasitophorous vacuole and thereafter in Maurer's clefts in trophozoites. Anti-MSP11-FL antibody levels were significantly higher in asymptomatic than symptomatic P. falciparum cases in malaria low endemic Thailand. This reconfirmed that anti-MSP11 antibodies play an important role in protection against clinical malaria, as previously reported. Furthermore, in vitro growth inhibition assay revealed that anti-MSP11-FL rabbit antibodies biologically function by inhibiting merozoite invasion of erythrocytes. These findings further support the vaccine candidacy of MSP11.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Antígenos de Protozoários/imunologia , Eritrócitos/parasitologia , Imunoglobulina G/farmacologia , Merozoítos/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Infecções Assintomáticas , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Malária Falciparum/imunologia , Proteínas de Protozoários/genética , Tailândia
5.
Nat Microbiol ; 3(11): 1224-1233, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30349082

RESUMO

The circumsporozoite protein (CSP) is the major surface protein of malaria sporozoites (SPZs), the motile and invasive parasite stage inoculated in the host skin by infected mosquitoes. Antibodies against the central CSP repeats of different plasmodial species are known to block SPZ infectivity1-5, but the precise mechanism by which these effectors operate is not completely understood. Here, using a rodent Plasmodium yoelii malaria model, we show that sterile protection mediated by anti-P. yoelii CSP humoral immunity depends on the parasite inoculation into the host skin, where antibodies inhibit motility and kill P. yoelii SPZs via a characteristic 'dotty death' phenotype. Passive transfer of an anti-repeat monoclonal antibody (mAb) recapitulates the skin inoculation-dependent protection, in a complement- and Fc receptor γ-independent manner. This purified mAb also decreases motility and, notably, induces the dotty death of P. yoelii SPZs in vitro. Cytotoxicity is species-transcendent since cognate anti-CSP repeat mAbs also kill Plasmodium berghei and Plasmodium falciparum SPZs. mAb cytotoxicity requires the actomyosin motor-dependent translocation and stripping of the protective CSP surface coat, rendering the parasite membrane susceptible to the SPZ pore-forming-like protein secreted to wound and traverse the host cell membrane6. The loss of SPZ fitness caused by anti-P. yoelii CSP repeat antibodies is thus a dynamic process initiated in the host skin where SPZs either stop moving7, or migrate and traverse cells to progress through the host tissues7-9 at the eventual expense of their own life.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Malária/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Pele/parasitologia , Animais , Anticorpos Monoclonais/farmacologia , Movimento Celular/efeitos dos fármacos , Culicidae , Feminino , Camundongos , Plasmodium berghei/imunologia , Plasmodium falciparum/imunologia , Plasmodium yoelii/citologia , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Esporozoítos/citologia , Esporozoítos/imunologia
6.
Elife ; 72018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29943728

RESUMO

Antibodies acquired naturally through repeated exposure to Plasmodium falciparum are essential in the control of blood-stage malaria. Antibody-dependent functions may include neutralization of parasite-host interactions, complement activation, and activation of Fc receptor functions. A role of antibody-dependent cellular cytotoxicity (ADCC) by natural killer (NK) cells in protection from malaria has not been established. Here we show that IgG isolated from adults living in a malaria-endemic region activated ADCC by primary human NK cells, which lysed infected red blood cells (RBCs) and inhibited parasite growth in an in vitro assay for ADCC-dependent growth inhibition. RBC lysis by NK cells was highly selective for infected RBCs in a mixed culture with uninfected RBCs. Human antibodies to P. falciparum antigens PfEMP1 and RIFIN were sufficient to promote NK-dependent growth inhibition. As these results implicate acquired immunity through NK-mediated ADCC, antibody-based vaccines that target bloodstream parasites should consider this new mechanism of action.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Imunoglobulina G/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Malária Falciparum/imunologia , Plasmodium falciparum/efeitos dos fármacos , Anticorpos Antiprotozoários/isolamento & purificação , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Técnicas de Cocultura , Eritrócitos/parasitologia , Hemólise , Humanos , Soros Imunes/química , Imunidade Celular/efeitos dos fármacos , Imunoglobulina G/isolamento & purificação , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/parasitologia , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Testes de Sensibilidade Parasitária , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Imagem com Lapso de Tempo
7.
Mol Cell Biochem ; 448(1-2): 9-15, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29435869

RESUMO

The aim of this study was to evaluate the therapeutic efficacy of specific avian polyclonal antibodies (IgY) against Trypanosoma cruzi and their interaction with ecto-enzymes of the purinergic system (NTPDase and adenosine deaminase (ADA) activities) in splenic lymphocytes. For this, mice were divided into six groups: three non-infected (A, B, and C) and three infected (D, E, and F). The groups A and D were composed by negative and positive controls, respectively; while the groups B and E were treated prophylactically with IgY (50 mg/kg), and the groups C and F were treated therapeutically with IgY (50 mg/kg). Treatment with IgY reduced parasitemia on day 6 post-infection (PI) compared to the infected control group, but it was similar on day 8 PI. Moreover, infected and treated animals (the groups E and F) did not show neither amastigotes in the cardiac tissue nor cardiac lesions when compared to the positive control group (the group D). The E-NTPDase (ATP and ADP as substrate) and ADA activities in splenic lymphocytes increased significantly in the positive control group (the group D) compared to the negative control group (the group A). The therapeutic treatment of IgY (the group F) was able to prevent the increase of E-NTPDase and E-ADA activities compared to the positive control group (the group D), but this finding was not observed in animals that received the prophylactic treatment (the group E). The therapeutic treatment of IgY may be considered an interesting approach to improve the immune response of mice experimentally infected by T. cruzi.


Assuntos
Adenosina Desaminase , Anticorpos Antiprotozoários/farmacologia , Proteínas Aviárias/farmacologia , Doença de Chagas , Imunoglobulinas/farmacologia , Proteínas de Protozoários , Baço , Trypanosoma cruzi , Adenosina Desaminase/imunologia , Adenosina Desaminase/metabolismo , Animais , Doença de Chagas/tratamento farmacológico , Doença de Chagas/enzimologia , Doença de Chagas/imunologia , Galinhas , Feminino , Linfócitos/enzimologia , Linfócitos/imunologia , Linfócitos/patologia , Camundongos , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Baço/enzimologia , Baço/imunologia , Baço/parasitologia , Baço/fisiologia , Trypanosoma cruzi/enzimologia , Trypanosoma cruzi/imunologia
8.
Blood ; 131(10): 1111-1121, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29352039

RESUMO

Effective vaccines against malaria caused by Plasmodium falciparum are still lacking, and the molecular mechanism of the host-parasite interaction is not fully understood. Here we demonstrate that the interaction of RAP2, a parasite-secreted rhoptry protein that functions in the parasitophorous vacuole formation stage of the invasion, and CD147 on the host erythrocyte is essential for erythrocyte invasion by P falciparum and is independent from all previously identified interactions involved. Importantly, the blockade of the CD147-RAP2 interaction by HP6H8, a humanized CD147 antibody, completely abolished the parasite invasion with both cure and preventative functions in a humanized mouse model. Together with its long half-life on human red blood cells and its safety profile in cynomolgus monkeys, HP6H8 is the first antibody that offers an advantageous approach by targeting a more conserved late-stage parasite ligand for preventing as well as treating severe malaria.


Assuntos
Basigina/metabolismo , Eritrócitos/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Proteínas rap de Ligação ao GTP/metabolismo , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Antiprotozoários/farmacologia , Eritrócitos/parasitologia , Eritrócitos/patologia , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
9.
Eur J Immunol ; 47(12): 2124-2136, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28833064

RESUMO

Acquired antibodies play an important role in immunity to P. falciparum malaria and are typically directed towards surface antigens expressed by merozoites and infected erythrocytes (IEs). The importance of specific IE surface antigens as immune targets remains unclear. We evaluated antibodies and protective associations in two cohorts of children in Papua New Guinea. We used genetically-modified P. falciparum to evaluate the importance of PfEMP1 and a P. falciparum isolate with a virulent phenotype. Our findings suggested that PfEMP1 was the dominant target of antibodies to the IE surface, including functional antibodies that promoted opsonic phagocytosis by monocytes. Antibodies were associated with increasing age and concurrent parasitemia, and were higher among children exposed to a higher force-of-infection as determined using molecular detection. Antibodies to IE surface antigens were consistently associated with reduced risk of malaria in both younger and older children. However, protective associations for antibodies to merozoite surface antigens were only observed in older children. This suggests that antibodies to IE surface antigens, particularly PfEMP1, play an earlier role in acquired immunity to malaria, whereas greater exposure is required for protective antibodies to merozoite antigens. These findings have implications for vaccine design and serosurveillance of malaria transmission and immunity.


Assuntos
Anticorpos Antiprotozoários/imunologia , Eritrócitos/imunologia , Imunidade/imunologia , Malária Falciparum/imunologia , Merozoítos/imunologia , Plasmodium falciparum/imunologia , Adolescente , Fatores Etários , Anticorpos Antiprotozoários/farmacologia , Linhagem Celular Tumoral , Criança , Pré-Escolar , Estudos de Coortes , Eritrócitos/parasitologia , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Monócitos/imunologia , Monócitos/virologia , Papua Nova Guiné , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Virulência/genética , Virulência/imunologia
10.
Poult Sci ; 95(2): 439-46, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26772659

RESUMO

Eimeria spp. must be controlled in floor-reared poultry to prevent the onset of coccidiosis. Here we use an oral antibody to chicken IL-10 to prevent growth depression due to Eimeria spp. infection. Egg antibody directed against an antigenic peptide of IL-10 was produced in laying hens and measured using an ELISA. In the first experiment, egg yolk powder containing antibody to chicken IL-10 (vlpramqt conjugate) (anti-IL-10 yolk powder) was fed at 3.4 g/kg feed to determine growth response following mixed Eimeria spp. challenge. Chicks were fed either anti-IL-10 antibodies or control antibodies and challenged (d3) with either sterile saline or a 10× attenuated Eimeria spp. vaccine. Control-fed and Eimeria-challenged chicks grew 8.8% slower than those challenged with saline (P < 0.04), whereas anti-IL-10-fed Eimeria challenged chicks were not different from untreated controls. In the second trial a dose response was performed with doses of either 0 (control antibody), 0.34-, or 3.4-g anti-IL-10 yolk powder/kg feed. Control-fed, Eimeria-challenged chicks grew 10.6% slower than control saline-challenged chicks (P < 0.05); however, anti-IL-10-fed chicks fed either dose of anti-IL-10 were not different from saline-challenged chicks. Finally, the effect of anti-IL-10 on acquired immunity was investigated. Chicks were fed control or anti-IL-10 yolk powder and vaccinated with a 1× dose of Eimeria vaccine at d 3. After 14 d, antibody was removed from the diet. Chicks were either saline or 10× Eimeria challenged at d 17. We found that the anti-IL-10-fed chickens did not show a reduction in growth due to challenge; hence anti-IL-10 does not appear to affect adaptive immunity during the primary immunization. Overall, use of an antibody to IL-10 is a novel method in preventing adverse effects of Eimeria spp. infection in poultry.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Proteínas Aviárias/metabolismo , Galinhas , Coccidiose/veterinária , Interleucina-10/metabolismo , Doenças das Aves Domésticas/prevenção & controle , Ração Animal/análise , Animais , Anticorpos Antiprotozoários/administração & dosagem , Galinhas/crescimento & desenvolvimento , Coccidiose/parasitologia , Coccidiose/prevenção & controle , Dieta/veterinária , Eimeria/fisiologia , Feminino , Doenças das Aves Domésticas/parasitologia
11.
Proc Natl Acad Sci U S A ; 112(36): E4975-84, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26305940

RESUMO

Calcium-dependent protein kinases (CDPKs) comprise the major group of Ca2+-regulated kinases in plants and protists. It has long been assumed that CDPKs are activated, like other Ca2+-regulated kinases, by derepression of the kinase domain (KD). However, we found that removal of the autoinhibitory domain from Toxoplasma gondii CDPK1 is not sufficient for kinase activation. From a library of heavy chain-only antibody fragments (VHHs), we isolated an antibody (1B7) that binds TgCDPK1 in a conformation-dependent manner and potently inhibits it. We uncovered the molecular basis for this inhibition by solving the crystal structure of the complex and simulating, through molecular dynamics, the effects of 1B7-kinase interactions. In contrast to other Ca2+-regulated kinases, the regulatory domain of TgCDPK1 plays a dual role, inhibiting or activating the kinase in response to changes in Ca2+ concentrations. We propose that the regulatory domain of TgCDPK1 acts as a molecular splint to stabilize the otherwise inactive KD. This dependence on allosteric stabilization reveals a novel susceptibility in this important class of parasite enzymes.


Assuntos
Proteínas Quinases/química , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Toxoplasma/enzimologia , Regulação Alostérica , Animais , Anticorpos Antiprotozoários/química , Anticorpos Antiprotozoários/metabolismo , Anticorpos Antiprotozoários/farmacologia , Biocatálise/efeitos dos fármacos , Western Blotting , Cálcio/metabolismo , Camelídeos Americanos , Células Cultivadas , Cristalografia por Raios X , Ativação Enzimática/efeitos dos fármacos , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Simulação de Dinâmica Molecular , Mutação , Fosforilação , Ligação Proteica , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Anticorpos de Domínio Único/farmacologia , Toxoplasma/genética
12.
Vet Res ; 45: 25, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24571471

RESUMO

Although IL17A is associated with the immunological control of various infectious diseases, its role in host response to Eimeria infections is not well understood. In an effort to better dissect the role of IL17A in host-pathogen interactions in avian coccidiosis, a neutralizing antibody (Ab) to chicken IL17A was used to counteract IL17A bioactivity in vivo. Chickens infected with Eimeria tenella and treated intravenously with IL17A Ab, exhibited reduced intracellular schizont and merozoite development, diminished lesion score, compared with untreated controls. Immunohistological evaluation of cecal lesions in the parasitized tissues indicated reduced migration and maturation of second-generation schizonts and reduced lesions in lamina propria and submucosa. In contrast, untreated and infected chickens had epithelial cells harboring second-generation schizonts, which extend into the submucosa through muscularis mucosa disruptions, maturing into second generation merozoites. Furthermore, IL17A Ab treatment was associated with increased parameters of Th1 immunity (IL2- and IFNγ- producing cells), reduced levels of reactive oxygen species (ROS), and diminished levels of serum matrix metalloproteinase-9 (MMP-9). Finally, schizonts from untreated and infected chickens expressed S100, Wiskott-Aldrich syndrome protein family member 3 (WASF3), and heat shock protein-70 (HSP70) proteins as merozoites matured, whereas the expression of these proteins was absent in IL17A Ab-treated chickens. These results provide the first evidence that the administration of an IL17A neutralizing Ab to E. tenella-infected chickens inhibits the migration of parasitized epithelial cells, markedly reduces the production of ROS and MMP-9, and decreases cecal lesions, suggesting that IL17A might be a potential therapeutic target for coccidiosis control.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Galinhas , Coccidiose/veterinária , Eimeria tenella/fisiologia , Interleucina-17/administração & dosagem , Doenças das Aves Domésticas/prevenção & controle , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/farmacologia , Anticorpos Antiprotozoários/administração & dosagem , Ceco/efeitos dos fármacos , Ceco/parasitologia , Coccidiose/parasitologia , Coccidiose/prevenção & controle , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/parasitologia , Doenças das Aves Domésticas/parasitologia , Esquizontes/efeitos dos fármacos , Esquizontes/crescimento & desenvolvimento , Esquizontes/fisiologia
13.
PLoS One ; 8(12): e83305, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349483

RESUMO

A gene encoding a Babesia bovis protein that shares significant degree of similarity to other apicomplexan thrombospondin-related anonymous proteins (TRAPs) was found in the genomic database and designated as BbTRAP2. Recombinant protein containing a conserved region of BbTRAP2 was produced in E. coli. A high antigenicity of recombinant BbTRAP2 (rBbTRAP2) was observed with field B. bovis-infected bovine sera collected from geographically different regions of the world. Moreover, antiserum against rBbTRAP2 specifically reacted with the authentic protein by Western blot analysis and an indirect fluorescent antibody test. Three bands corresponding to 104-, 76-, and 44-kDa proteins were identified in the parasite lysates and two bands of 76- and 44-kDa proteins were detected in the supernatant of cultivated parasites, indicating that BbTRAP2 was proteolytically processed and shed into the culture. Apical and surface localizations of BbTRAP2 were observed in the intracellular and extracellular parasites, respectively, by confocal laser microscopic examination. Moreover, native BbTRAP2 was precipitated by bovine erythrocytes, suggesting its role in the attachment to erythrocytes. Furthermore, the specific antibody to rBbTRAP2 inhibited the growth of B. bovis in a concentration-dependent manner. Consistently, pre-incubation of the free merozoites with the antibody to rBbTRAP2 resulted in an inhibition of the parasite invasion into host erythrocytes. Interestingly, the antibody to rBbTRAP2 was the most inhibitive for the parasite's growth as compared to those of a set of antisera produced against different recombinant proteins, including merozoite surface antigen 2c (BbMSA-2c), rhoptry-associated protein 1 C-terminal (BbRAP-1CT), and spherical body protein 1 (BbSBP-1). These results suggest that BbTRAP2 might be a potential candidate for development of a subunit vaccine against B. bovis infection.


Assuntos
Babesia bovis/química , Proteínas de Protozoários/química , Animais , Anticorpos Antiprotozoários/farmacologia , Babesia bovis/genética , Babesia bovis/imunologia , Bovinos , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Vacinas Protozoárias/química , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo
14.
PLoS One ; 8(9): e72504, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24039774

RESUMO

BACKGROUND: A highly effective vaccine against Plasmodium falciparum malaria should induce potent, strain transcending immunity that broadly protects against the diverse population of parasites circulating globally. We aimed to identify vaccine candidates that fulfill the criteria. METHODS: We have measured growth inhibitory activity of antibodies raised to a range of antigens to identify those that can efficiently block merozoite invasion for geographically diverse strains of P. falciparum. RESULTS: This has shown that the conserved Region III-V, of the P. falciparum erythrocyte-binding antigen (EBA)-175 was able to induce antibodies that potently inhibit merozoite invasion across diverse parasite strains, including those reliant on invasion pathways independent of EBA-175 function. Additionally, the conserved RIII-V domain of EBA-140 also induced antibodies with strong in vitro parasite growth inhibitory activity. CONCLUSION: We identify an alternative, highly conserved region (RIV-V) of EBA-175, present in all EBA proteins, that is the target of potent, strain transcending neutralizing antibodies, that represents a strong candidate for development as a component in a malaria vaccine.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinação , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/farmacologia , Anticorpos Antiprotozoários/farmacologia , Antígenos de Protozoários/química , Sequência Conservada , Mapeamento de Epitopos , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunoglobulina G/sangue , Concentração Inibidora 50 , Vacinas Antimaláricas/imunologia , Malária Falciparum/sangue , Malária Falciparum/imunologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/química , Coelhos , Especificidade da Espécie
15.
PLoS Negl Trop Dis ; 6(11): e1902, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166849

RESUMO

BACKGROUND: The discovery of Nanobodies (Nbs) with a direct toxic activity against African trypanosomes is a recent advancement towards a new strategy against these extracellular parasites. The anti-trypanosomal activity relies on perturbing the highly active recycling of the Variant-specific Surface Glycoprotein (VSG) that occurs in the parasite's flagellar pocket. METHODOLOGY/PRINCIPAL FINDINGS: Here we expand the existing panel of Nbs with anti-Trypanosoma brucei potential and identify four categories based on their epitope specificity. We modified the binding properties of previously identified Nanobodies Nb_An05 and Nb_An33 by site-directed mutagenesis in the paratope and found this to strongly affect trypanotoxicity despite retention of antigen-targeting properties. Affinity measurements for all identified anti-trypanosomal Nbs reveal a strong correlation between trypanotoxicity and affinity (K(D)), suggesting that it is a crucial determinant for this activity. Half maximal effective (50%) affinity of 57 nM was calculated from the non-linear dose-response curves. In line with these observations, Nb humanizing mutations only preserved the trypanotoxic activity if the K(D) remained unaffected. CONCLUSIONS/SIGNIFICANCE: This study reveals that the binding properties of Nanobodies need to be compatible with achieving an occupancy of >95% saturation of the parasite surface VSG in order to exert an anti-trypanosomal activity. As such, Nb-based approaches directed against the VSG target would require binding to an accessible, conserved epitope with high affinity.


Assuntos
Anticorpos Antiprotozoários/imunologia , Anticorpos de Domínio Único/imunologia , Trypanosoma brucei brucei/imunologia , Animais , Anticorpos Antiprotozoários/farmacologia , Afinidade de Anticorpos , Camundongos , Camundongos Endogâmicos C57BL , Viabilidade Microbiana/efeitos dos fármacos , Anticorpos de Domínio Único/farmacologia , Trypanosoma brucei brucei/efeitos dos fármacos
16.
PLoS One ; 7(9): e45253, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028883

RESUMO

BACKGROUND: Acquired antibodies are important in human immunity to malaria, but key targets remain largely unknown. Plasmodium falciparum reticulocyte-binding-homologue-4 (PfRh4) is important for invasion of human erythrocytes and may therefore be a target of protective immunity. METHODS: IgG and IgG subclass-specific responses against different regions of PfRh4 were determined in a longitudinal cohort of 206 children in Papua New Guinea (PNG). Human PfRh4 antibodies were tested for functional invasion-inhibitory activity, and expression of PfRh4 by P. falciparum isolates and sequence polymorphisms were determined. RESULTS: Antibodies to PfRh4 were acquired by children exposed to P. falciparum malaria, were predominantly comprised of IgG1 and IgG3 subclasses, and were associated with increasing age and active parasitemia. High levels of antibodies, particularly IgG3, were strongly predictive of protection against clinical malaria and high-density parasitemia. Human affinity-purified antibodies to the binding region of PfRh4 effectively inhibited erythrocyte invasion by P. falciparum merozoites and antibody levels in protected children were at functionally-active concentrations. Although expression of PfRh4 can vary, PfRh4 protein was expressed by most isolates derived from the cohort and showed limited sequence polymorphism. CONCLUSIONS: Evidence suggests that PfRh4 is a target of antibodies that contribute to protective immunity to malaria by inhibiting erythrocyte invasion and preventing high density parasitemia. These findings advance our understanding of the targets and mechanisms of human immunity and evaluating the potential of PfRh4 as a component of candidate malaria vaccines.


Assuntos
Anticorpos Antiprotozoários/imunologia , Imunoglobulina G/imunologia , Malária Falciparum/prevenção & controle , Proteínas de Membrana/imunologia , Parasitemia/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adolescente , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/farmacologia , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Criança , Pré-Escolar , Eritrócitos/efeitos dos fármacos , Eritrócitos/imunologia , Eritrócitos/parasitologia , Feminino , Expressão Gênica , Humanos , Imunidade Humoral/efeitos dos fármacos , Imunoglobulina G/biossíntese , Imunoglobulina G/farmacologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Proteínas de Membrana/genética , Merozoítos/efeitos dos fármacos , Merozoítos/imunologia , Parasitemia/imunologia , Plasmodium falciparum/efeitos dos fármacos , Polimorfismo Genético , Ligação Proteica , Proteínas de Protozoários/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
17.
PLoS Pathog ; 8(8): e1002858, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22912579

RESUMO

Malaria parasites reside inside erythrocytes and the disease manifestations are linked to the growth inside infected erythrocytes (IE). The growth of the parasite is mostly confined to the trophozoite stage during which nuclear division occurs followed by the formation of cell bodies (schizogony). The mechanism and regulation of schizogony are poorly understood. Here we show a novel role for a Plasmodium falciparum 60S stalk ribosomal acidic protein P2 (PfP2) (PFC0400w), which gets exported to the IE surface for 6-8 hrs during early schizogony, starting around 26-28 hrs post-merozoite invasion. The surface exposure is demonstrated using multiple PfP2-specific monoclonal antibodies, and is confirmed through transfection using PfP2-GFP. The IE surface-exposed PfP2-protein occurs mainly as SDS-resistant P2-homo-tetramers. Treatment with anti-PfP2 monoclonals causes arrest of IEs at the first nuclear division. Upon removal of the antibodies, about 80-85% of synchronized parasites can be released even after 24 hrs of antibody treatment. It has been reported that a tubovesicular network (TVN) is set up in early trophozoites which is used for nutrient import. Anti-P2 monoclonal antibodies cause a complete fragmentation of TVN by 36 hrs, and impairs lipid import in IEs. These may be downstream causes for the cell-cycle arrest. Upon antibody removal, the TVN is reconstituted, and the cell division progresses. Each of the above properties is observed in the rodent malaria parasite species P. yoelii and P. berghei. The translocation of the P2 protein to the IE surface is therefore likely to be of fundamental importance in Plasmodium cell division.


Assuntos
Pontos de Checagem do Ciclo Celular/fisiologia , Divisão do Núcleo Celular/fisiologia , Eritrócitos/parasitologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas Ribossômicas/metabolismo , Animais , Anticorpos Monoclonais Murinos/farmacologia , Anticorpos Antiprotozoários/farmacologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Divisão do Núcleo Celular/efeitos dos fármacos , Humanos , Camundongos , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Proteínas de Protozoários/genética , Proteínas Ribossômicas/genética
18.
PLoS One ; 7(5): e36233, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22570698

RESUMO

The ribosomal P proteins are located on the stalk of the ribosomal large subunit and play a critical role during the elongation step of protein synthesis. The single chain recombinant antibody C5 (scFv C5) directed against the C-terminal region of the Trypanosoma cruzi P2ß protein (TcP2ß) recognizes the conserved C-terminal end of all T. cruzi ribosomal P proteins. Although this region is highly conserved among different species, surface plasmon resonance analysis showed that the scFv C5 possesses very low affinity for the corresponding mammalian epitope, despite having only one single amino-acid change. Crystallographic analysis, in silico modelization and NMR assays support the analysis, increasing our understanding on the structural basis of epitope specificity. In vitro protein synthesis experiments showed that scFv C5 was able to specifically block translation by T. cruzi and Crithidia fasciculata ribosomes, but virtually had no effect on Rattus norvegicus ribosomes. Therefore, we used the scFv C5 coding sequence to make inducible intrabodies in Trypanosoma brucei. Transgenic parasites showed a strong decrease in their growth rate after induction. These results strengthen the importance of the P protein C terminal regions for ribosomal translation activity and suggest that trypanosomatid ribosomal P proteins could be a possible target for selective therapeutic agents that could be derived from structural analysis of the scFv C5 antibody paratope.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas de Protozoários/biossíntese , Proteínas Ribossômicas/antagonistas & inibidores , Anticorpos de Cadeia Única/farmacologia , Trypanosoma cruzi/metabolismo , Anticorpos Antiprotozoários/química , Anticorpos Antiprotozoários/genética , Doença de Chagas/tratamento farmacológico , Doença de Chagas/metabolismo , Epitopos/química , Epitopos/imunologia , Expressão Gênica , Humanos , Modelos Moleculares , Filogenia , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Proteínas de Protozoários/classificação , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Proteínas Ribossômicas/biossíntese , Proteínas Ribossômicas/classificação , Proteínas Ribossômicas/imunologia , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/genética , Trypanosoma cruzi/genética , Trypanosoma cruzi/imunologia
19.
PLoS Pathog ; 7(12): e1002448, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22194692

RESUMO

The malarial life cycle involves repeated rounds of intraerythrocytic replication interspersed by host cell rupture which releases merozoites that rapidly invade fresh erythrocytes. Apical membrane antigen-1 (AMA1) is a merozoite protein that plays a critical role in invasion. Antibodies against AMA1 prevent invasion and can protect against malaria in vivo, so AMA1 is of interest as a malaria vaccine candidate. AMA1 is efficiently shed from the invading parasite surface, predominantly through juxtamembrane cleavage by a membrane-bound protease called SUB2, but also by limited intramembrane cleavage. We have investigated the structural requirements for shedding of Plasmodium falciparum AMA1 (PfAMA1), and the consequences of its inhibition. Mutagenesis of the intramembrane cleavage site by targeted homologous recombination abolished intramembrane cleavage with no effect on parasite viability in vitro. Examination of PfSUB2-mediated shedding of episomally-expressed PfAMA1 revealed that the position of cleavage is determined primarily by its distance from the parasite membrane. Certain mutations at the PfSUB2 cleavage site block shedding, and parasites expressing these non-cleavable forms of PfAMA1 on a background of expression of the wild type gene invade and replicate normally in vitro. The non-cleavable PfAMA1 is also functional in invasion. However - in contrast to the intramembrane cleavage site - mutations that block PfSUB2-mediated shedding could not be stably introduced into the genomic pfama1 locus, indicating that some shedding of PfAMA1 by PfSUB2 is essential. Remarkably, parasites expressing shedding-resistant forms of PfAMA1 exhibit enhanced sensitivity to antibody-mediated inhibition of invasion. Drugs that inhibit PfSUB2 activity should block parasite replication and may also enhance the efficacy of vaccines based on AMA1 and other merozoite surface proteins.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Antígenos de Protozoários/imunologia , Proteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/farmacologia , Antígenos de Protozoários/química , Antígenos de Protozoários/metabolismo , Recombinação Homóloga , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Mutação/genética , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo
20.
PLoS Pathog ; 7(6): e1002072, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21698216

RESUMO

The African trypanosome Trypanosoma brucei, which persists within the bloodstream of the mammalian host, has evolved potent mechanisms for immune evasion. Specifically, antigenic variation of the variant-specific surface glycoprotein (VSG) and a highly active endocytosis and recycling of the surface coat efficiently delay killing mediated by anti-VSG antibodies. Consequently, conventional VSG-specific intact immunoglobulins are non-trypanocidal in the absence of complement. In sharp contrast, monovalent antigen-binding fragments, including 15 kDa nanobodies (Nb) derived from camelid heavy-chain antibodies (HCAbs) recognizing variant-specific VSG epitopes, efficiently lyse trypanosomes both in vitro and in vivo. This Nb-mediated lysis is preceded by very rapid immobilisation of the parasites, massive enlargement of the flagellar pocket and major blockade of endocytosis. This is accompanied by severe metabolic perturbations reflected by reduced intracellular ATP-levels and loss of mitochondrial membrane potential, culminating in cell death. Modification of anti-VSG Nbs through site-directed mutagenesis and by reconstitution into HCAbs, combined with unveiling of trypanolytic activity from intact immunoglobulins by papain proteolysis, demonstrates that the trypanolytic activity of Nbs and Fabs requires low molecular weight, monovalency and high affinity. We propose that the generation of low molecular weight VSG-specific trypanolytic nanobodies that impede endocytosis offers a new opportunity for developing novel trypanosomiasis therapeutics. In addition, these data suggest that the antigen-binding domain of an anti-microbial antibody harbours biological functionality that is latent in the intact immunoglobulin and is revealed only upon release of the antigen-binding fragment.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Endocitose/efeitos dos fármacos , Trypanosoma brucei brucei/imunologia , Glicoproteínas Variantes de Superfície de Trypanosoma/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/uso terapêutico , Afinidade de Anticorpos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Nanopartículas , Tripanossomicidas/farmacologia , Tripanossomicidas/uso terapêutico , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/fisiologia , Trypanosoma brucei brucei/ultraestrutura , Tripanossomíase Africana/imunologia , Tripanossomíase Africana/metabolismo , Tripanossomíase Africana/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA