Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
4.
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
5.
PLoS One ; 11(7): e0159449, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27434123

RESUMO

Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery approaches, and improvements are needed to enhance the efficacy of the leading vaccine candidate RTS,S that targets the circumsporozoite protein (CSP) of the infective sporozoite. Here we report a transcriptome-based approach to identify novel pre-erythrocytic vaccine antigens that could potentially be used in combination with CSP. We hypothesized that stage-specific upregulated genes would enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in P. yoelii and P. berghei and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models. In our primary screen using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. Two antigens, when combined with CSP, provided significantly greater protection than CSP alone in both models. Based on the observations reported here, transcriptional patterns of Plasmodium genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/tratamento farmacológico , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Feminino , Humanos , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Plasmodium yoelii/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia , Vacinas de DNA/uso terapêutico
6.
Methods Mol Biol ; 1325: 69-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26450380

RESUMO

Antibodies against the infectious sporozoite stage of malaria have been shown to be effective in preventing infection of the liver and in mitigating the ensuing blood stage. However, only a handful of antibody targets have been vetted and shown to be successful in mediating in vivo protection. Even more limited are the means with which to measure how effectively antibodies can reduce the number of parasites establishing infection in the liver. Traditionally, only qPCR of infected mouse livers could accurately measure liver parasite burden. However, this procedure requires sacrifice of the animal and precludes monitoring of the ensuing blood stage infection. Herein we describe a method of accurately assessing antibody-mediated reduction of parasite liver burden by combining passive or active immunization of mice and mosquito bite challenge with luciferase-expressing transgenic P. yoelii parasites. This method is rapid, reliable and allows for observation of blood stage disease in the same animal. This model will prove integral in screening the efficacy of novel antibody targets as the search for a more effective malaria vaccine continues.


Assuntos
Fígado/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Imagem Molecular/métodos , Animais , Animais Geneticamente Modificados , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/uso terapêutico , Humanos , Fígado/parasitologia , Fígado/patologia , Medições Luminescentes , Malária/parasitologia , Malária/patologia , Camundongos , Plasmodium yoelii/imunologia , Plasmodium yoelii/patogenicidade
7.
Biotechnol J ; 10(10): 1651-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25913888

RESUMO

Multicomponent vaccines targeting different stages of Plasmodium falciparum represent a promising, holistic concept towards better malaria vaccines. Additionally, an effective vaccine candidate should demonstrate cross-strain specificity because many antigens are polymorphic, which can reduce vaccine efficacy. A cocktail of recombinant fusion proteins (VAMAX-Mix) featuring three diversity-covering variants of the blood-stage antigen PfAMA1, each combined with the conserved sexual-stage antigen Pfs25 and one of the pre-erythrocytic-stage antigens PfCSP_TSR or PfCelTOS, or the additional blood-stage antigen PfMSP1_19, was produced in Pichia pastoris and used to immunize rabbits. The immune sera and purified IgG were used to perform various assays determining antigen specific titers and in vitro efficacy against different parasite stages and strains. In functional in vitro assays we observed robust inhibition of blood-stage (up to 90%), and sexual-stage parasites (up to 100%) and biased inhibition of pre-erythrocytic parasites (0-40%). Cross-strain blood-stage efficacy was observed in erythrocyte invasion assays using four different P. falciparum strains. The quantification of antigen-specific IgGs allowed the determination of specific IC50 values. The significant difference in antigen-specific IC50 requirements, the direct correlation between antigen-specific IgG and the relative quantitative representation of antigens within the cocktail, provide valuable implementations for future multi-stage, multi-component vaccine designs.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium falciparum/imunologia , Proteínas Recombinantes de Fusão/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/genética , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/uso terapêutico , Pichia/genética , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Coelhos , Proteínas Recombinantes de Fusão/uso terapêutico
8.
J Immunol Res ; 2015: 738030, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26770994

RESUMO

The last decade has witnessed a steady reduction of the malaria burden worldwide. With various countries targeting disease elimination in the near future, the popular parasite infection or entomological inoculation rates are becoming less and less informative of the underlying malaria burden due to a reduced number of infected individuals or mosquitoes at the time of sampling. To overcome such problem, alternative measures based on antibodies against specific malaria antigens have gained recent interest in malaria epidemiology due to the possibility of estimating past disease exposure in absence of infected individuals. This paper aims then to review current mathematical models and corresponding statistical approaches used in antibody data analysis. The application of these models is illustrated with three data sets from Equatorial Guinea, Brazilian Amazonia region, and western Kenyan highlands. A brief discussion is also carried out on the future challenges of using these models in the context of malaria elimination.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Antimaláricos/uso terapêutico , Imunoterapia , Vacinas Antimaláricas/imunologia , Malária/terapia , Modelos Teóricos , Plasmodium/imunologia , Animais , Brasil , Conjuntos de Dados como Assunto , Guiné Equatorial , Humanos , Quênia , Malária/imunologia
9.
Parasitol Res ; 113(1): 229-37, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24154594

RESUMO

Cryptosporidium parvum is a coccidian protozoan that causes diarrhea in immunocompromised humans and newborn animals. Billions of oocysts of C. parvum can be released from the infected calves and can contaminate the environment. The severity of the disease depends on the immunological status of the individual. Oocysts of Cryptosporidium are extremely resistant to many environmental stresses, and no effective disinfectant and curative agent against this organism is available. In our study, recombinant C. parvum P23 was prepared for application in the isolation and prevention of cryptosporidiosis. P23 is a glycoprotein that belongs to a family of protein of 23-27 kDa and is believed to be expressed in the different life stages of the parasite. Immunostaining analysis using the indirect fluorescent antibody test showed that P23 could be recognized on the surface of the oocysts. The antibody prepared in rabbit against P23 was bound to Sepharose 4B and used for the isolation of oocysts. The results showed that the prepared column was able to bind specifically only the oocysts. The effect of specific recombinant C. parvum IgY antibody against infection with C. parvum was examined in a mouse model. For this aim, purified egg yolk antibody prepared from immunized hens was used to analyze the protective effect of recombinant P23 specific antibody in immunosuppressed adult mice. The results showed more than 70% reduction in oocyst shedding after challenge with 1 × 10(4) oocysts. These results support previous studies of other investigators regarding the protective effect of P23 as an antigen against C. parvum infection and showed that it could be possible to design a passive immunization strategy against C. parvum based on the anti-P23 yolk antibody in animals and immunosuppressed humans.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Criptosporidiose/prevenção & controle , Glicoproteínas/imunologia , Proteínas de Protozoários/imunologia , Animais , Antígenos de Protozoários/imunologia , Bovinos , Galinhas , Criptosporidiose/imunologia , Criptosporidiose/veterinária , Cryptosporidium parvum , Gema de Ovo/imunologia , Feminino , Imunização Passiva , Imunoglobulinas/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Oocistos/imunologia , Coelhos , Proteínas Recombinantes/uso terapêutico
10.
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
11.
PLoS One ; 6(2): e16622, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21326877

RESUMO

The high molecular weight, multidomain VAR2CSA protein mediating adhesion of Plasmodium falciparum-infected erythrocytes in the placenta is the leading candidate for a pregnancy malaria vaccine. However, it has been difficult so far to generate strong and consistent adhesion blocking antibody responses against most single-domain VAR2CSA immunogens. Recent advances in expression of the full-length recombinant protein showed it binds with much greater specificity and affinity to chondroitin sulphate A (CSA) than individual VAR2CSA domains. This raises the possibility that a specific CSA binding pocket(s) is formed in the full length antigen and could be an important target for vaccine development. In this study, we compared the immunogenicity of a full-length VAR2CSA recombinant protein containing all six Duffy binding-like (DBL) domains to that of a three-domain construct (DBL4-6) in mice and rabbits. Animals immunized with either immunogen acquired antibodies reacting with several VAR2CSA individual domains by ELISA, but antibody responses against the highly conserved DBL4 domain were weaker in animals immunized with full-length DBL1-6 recombinant protein compared to DBL4-6 recombinant protein. Both immunogens induced cross-reactive antibodies to several heterologous CSA-binding parasite lines expressing different VAR2CSA orthologues. However, antibodies that inhibited adhesion of parasites to CSA were only elicited in rabbits immunized with full-length immunogen and inhibition was restricted to the homologous CSA-binding parasite. These findings demonstrate that partial and full-length VAR2CSA immunogens induce cross-reactive antibodies, but inhibitory antibody responses to full-length immunogen were highly allele-specific and variable between animal species.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Especificidade de Anticorpos , Antígenos de Protozoários/imunologia , Placenta/parasitologia , Complicações Parasitárias na Gravidez/parasitologia , Sequência de Aminoácidos/fisiologia , Animais , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/uso terapêutico , Especificidade de Anticorpos/imunologia , Especificidade de Anticorpos/fisiologia , Antígenos de Protozoários/química , Antígenos de Protozoários/isolamento & purificação , Células Cultivadas , Reações Cruzadas/imunologia , Feminino , Humanos , Imunização , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/farmacologia , Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/parasitologia , Malária Falciparum/patologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Camundongos , Camundongos Endogâmicos BALB C , Placenta/imunologia , Gravidez , Complicações Parasitárias na Gravidez/patologia , Complicações Parasitárias na Gravidez/terapia , Isoformas de Proteínas/imunologia , Coelhos , Especificidade da Espécie
12.
Niger J Med ; 19(1): 8-13, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20232750

RESUMO

BACKGROUND: Malaria is an important parasitic disease of humans caused by infection with a parasite of the genus Polasmodium and transmitted by female anopheles. Infection caused by P. falciparum is the most serious of all the other species (P. ovale, P. vivax and P. malariae) especially in terms of morbidity and mortality hence the reason why most of the research has been focussed on this species. The disease affects up to about 40 per cent of the world's population with around 300-500 million people currently infected and mainly in the tropics. It has a high morbidity and mortality especially in resource-poor tropical and subtropical regions with an economic fall of about US$ 12 billion annually in Africa alone. METHOD: relevant literatures were reviewed from medical journals, library search and internet source. Other relevant websites like PATH, Malaria Vaccine Initiative and Global Fund were also visited to source for information. The key words employed were: malaria, vaccine, anopheles mosquito, insecticide treated bed-nets, pyrethroids and Plasmodium. RESULTS: several studies have underscored the need to develop an effective human malaria vaccine for the control and possible eradication of malaria across the globe with the view to reduce the morbidity and mortality associated with the disease, improve on the social and economic losses and also protect those at risk. CONCLUSION: It is very obvious that the need for effective human malaria vaccine is not only to serve those living in malaria endemic regions but also the non-immune travellers especially those travelling to malaria endemic areas; this would offer cost effective means of preventing the disease, reducing the morbidity and mortality associated with it in addition to closing the gap left by other control measures. It is very obvious that there is no single control measure known to be effective in the control of malaria, hence the need for combination of more than one method with the aim of achieving synergy in the total control and possible eradication of the disease. It suffices to say that despite the use of combination of more than one method (e.g., drugs treating patients, breaking the life cycle of the vector mosquito using larvicides, clearing swamps and other mosquito breeding sites), no much progress was made towards achieving this goal, hence the renewed interest especially with regards to vaccine development.


Assuntos
Vacinas Antimaláricas/uso terapêutico , Malária/prevenção & controle , Plasmodium/imunologia , Animais , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Humanos , Malária/tratamento farmacológico , Ensaios Clínicos Controlados Aleatórios como Assunto , Viagem
13.
Infect Immun ; 77(1): 549-56, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19001071

RESUMO

Four fully human monoclonal antibodies (MAbs) to Entamoeba histolytica intermediate subunit lectin (Igl) were prepared in XenoMouse mice, which are transgenic mice expressing human immunoglobulin loci. Examination of the reactivities of these MAbs to recombinant Igl1 and Igl2 of E. histolytica showed that XEhI-20 {immunoglobulin G2(kappa) [IgG2(kappa)]} and XEhI-28 [IgG2(kappa)] were specific to Igl1, XEhI-B5 [IgG2(kappa)] was specific to Igl2, and XEhI-H2 [IgM(kappa)] was reactive with both Igls. Gene analyses revealed that the V(H) and V(L) germ lines were VH3-48 and L2 for XEhI-20, VH3-21 and L2 for XEhI-28, VH3-33 and B3 for XEhI-B5, and VH4-4 and A19 for XEhI-H2, respectively. Flow cytometry analyses showed that the epitopes recognized by all of these MAbs were located on the surfaces of living trophozoites. Confocal microscopy demonstrated that most Igl1 and Igl2 proteins were colocalized on the surface and in the cytoplasm, but different localization patterns in intracellular vacuoles were also present. The preincubation of trophozoites with XEhI-20, XEhI-B5, and XEhI-H2 caused significant inhibition of the adherence of trophozoites to Chinese hamster ovary cells, whereas preincubation with XEhI-28 did not do so. XEhI-20, XEhI-B5, and XEhI-H2 were injected intraperitoneally into hamsters 24 h prior to intrahepatic challenge with E. histolytica trophozoites. One week later, the mean abscess size in groups injected with one of the three MAbs was significantly smaller than that in controls injected with polyclonal IgG or IgM isolated from healthy humans. These results demonstrate that human MAbs to Igls may be applicable for immunoprophylaxis of amebiasis.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/isolamento & purificação , Entamoeba histolytica/imunologia , Lectinas/imunologia , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antiprotozoários/uso terapêutico , Células CHO , Adesão Celular , Membrana Celular/química , Cricetinae , Cricetulus , Citoplasma/química , Citometria de Fluxo , Imunização Passiva , Imunoglobulina G/imunologia , Região Variável de Imunoglobulina/genética , Abscesso Hepático/prevenção & controle , Masculino , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA , Vacúolos/química
14.
Peptides ; 30(2): 330-42, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19071172

RESUMO

We have developed monoclonal antibodies directed against the pseudopeptide psi-130, derived from the highly conserved malarial antigen Plasmodium falciparum merozoite surface protein 2 (MSP-2), for obtaining novel molecular tools with potential applications in the control of malaria. Following isotype switching, these antibodies were tested for their ability to suppress blood-stage parasitemia through passive immunization in malaria-infected mice. Some proved totally effective in suppressing a lethal blood-stage challenge infection and others reduced malarial parasitemia. Protection against P. berghei malaria following Ig passive immunization can be associated with specific immunoglobulins induced by a site-directed designed MSP-2 reduced amide pseudopeptide.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Imunização Passiva , Malária/tratamento farmacológico , Parasitemia/tratamento farmacológico , Plasmodium berghei , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Modelos Animais de Doenças , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
PLoS Pathog ; 3(5): e72, 2007 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-17511516

RESUMO

The success of passive immunization suggests that antibody-based therapies will be effective at controlling malaria. We describe the development of fully human antibodies specific for Plasmodium falciparum by antibody repertoire cloning from phage display libraries generated from immune Gambian adults. Although these novel reagents bind with strong affinity to malaria parasites, it remains unclear if in vitro assays are predictive of functional immunity in humans, due to the lack of suitable animal models permissive for P. falciparum. A potentially useful solution described herein allows the antimalarial efficacy of human antibodies to be determined using rodent malaria parasites transgenic for P. falciparum antigens in mice also transgenic for human Fc-receptors. These human IgG1s cured animals of an otherwise lethal malaria infection, and protection was crucially dependent on human FcgammaRI. This important finding documents the capacity of FcgammaRI to mediate potent antimalaria immunity and supports the development of FcgammaRI-directed therapy for human malaria.


Assuntos
Malária/imunologia , Receptores Fc , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/isolamento & purificação , Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários , Antimaláricos , Mapeamento de Epitopos , Humanos , Imunoglobulina G , Malária/terapia , Camundongos , Camundongos Transgênicos , Plasmodium falciparum/imunologia
16.
Exp Parasitol ; 116(1): 53-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17223107

RESUMO

The objective of this study was to test a hypothesis that the predominant variable antigen type (VAT) repertoire of a single stock of Trypanosoma evansi was limited and small. It was further assumed that six rabbits could produce all antibodies against the predominant VAT repertoire of a stock of T. evansi and the antiserum mixture from the six rabbits containing all the antibodies could completely protect mice against any homologous stock infections and partially protect mice against some heterologous stock infections. Mice were each intraperitoneally infected with 100 parasites of clone-derived and non-clone-derived populations of the YNB stock, Kazakhstan strain or Vietnam strain of T. evansi, and treated with the antiserum mixture when trypanosomes had been detected in the blood. All of the 10 mice infected with either non-clone-derived or clone-derived populations of the YNB stock survived, and some (4/10) of mice infected with the heterologous Kazakhstan strain survived, while all those (10/10) infected with the heterologous Vietnam strain died. These results support the hypothesis that the predominant VAT repertoire of a single stock of T. evansi was limited and small, and have important implications in the consideration of treating human trypanosomosis due to drug resistant strains with antiserum mixture.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Soros Imunes/imunologia , Imunização Passiva/veterinária , Trypanosoma/imunologia , Tripanossomíase/veterinária , Testes de Aglutinação/veterinária , Animais , Animais Domésticos , Animais Selvagens , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Variação Antigênica/imunologia , Técnica Indireta de Fluorescência para Anticorpo/veterinária , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos , Camundongos Endogâmicos ICR , Coelhos , Fatores de Tempo , Tripanossomíase/prevenção & controle
17.
Proteins ; 66(3): 513-27, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17173281

RESUMO

Merozoite surface protein 1 (MSP1) of the malaria parasite Plasmodium falciparum is an important vaccine candidate antigen. Antibodies specific for the C-terminal maturation product, MSP1(19), have been shown to inhibit erythrocyte invasion and parasite growth. Specific monoclonal antibodies react with conformational epitopes contained within the two EGF-like domains that constitute the antigen MSP1(19). To gain greater insight into the inhibitory process, the authors selected two strongly inhibitory antibodies (designated 12.8 and 12.10) and modeled their structures by homology. Computational docking was used to generate antigen-antibody complexes and a selection filter based on NMR data was applied to obtain plausible models. Molecular Dynamics simulations of the selected complexes were performed to evaluate the role of specific side chains in the binding. Favorable complexes were obtained that complement the NMR data in defining specific binding sites. These models can provide valuable guidelines for future experimental work that is devoted to the understanding of the action mechanism of invasion-inhibitory antibodies.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Malária/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/química , Complexo Antígeno-Anticorpo , Simulação por Computador , Humanos , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
18.
Clin Sci (Lond) ; 110(1): 11-9, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16336201

RESUMO

Abs (antibodies) are complex glycoproteins that play a crucial role in protective immunity to malaria, but their effectiveness in mediating resistance can be enhanced by genetically engineered modifications that improve on nature. These Abs also aid investigation of immune mechanisms operating to control the disease and are valuable tools in developing neutralization assays for vaccine design. This review explores how this might be achieved.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Imunização Passiva/métodos , Malária/terapia , Receptores Fc/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Humanos , Malária/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium/imunologia , Polimorfismo Genético , Receptores Fc/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico
19.
Vaccine ; 23(2): 232-5, 2004 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-15531042

RESUMO

In this study the effect of chicken egg yolk antibody (IgY) against Cryptosporidium parvum infection was examined. IgY sample was prepared from eggs of chickens immunized with C. parvum oocyst antigens. In vitro, antibody-treated sporozoites showed reduced binding to Caco-2 cells and lost vitality. These phenomena were not observed with a control IgY sample prepared from eggs of non-immunized chickens. Scid mice orally administered with the antibody demonstrated partial reduction in oocyst shedding after challenge with 10(3) oocysts. IgY, however, could not eliminate the infection after 17 days of continuous treatment. The potentials of using specific IgY for treatment and prevention of cryptosporidiosis were discussed.


Assuntos
Anticorpos Antiprotozoários/imunologia , Criptosporidiose/terapia , Cryptosporidium parvum/imunologia , Animais , Anticorpos Antiprotozoários/uso terapêutico , Criptosporidiose/patologia , Cryptosporidium parvum/ultraestrutura , Gema de Ovo , Imunoterapia Adotiva , Camundongos , Camundongos SCID
20.
Immunol Cell Biol ; 82(4): 447-52, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15283856

RESUMO

Sequestration and the attachment of Plasmodium falciparum malaria-infected RBC to venous endothelial cells involves parasite-encoded ligands interacting with up to nine host receptors. Antisequestration immunotherapy as an adjunct to quinine did not alter the dynamics of parasite clearance or prove beneficial for the patient. Estimated concentrations of antibody likely to reverse adherence in patients were based on the concentrations of parasite ligands, host receptors and patient equivalents derived from in vitro observations. Calculations presented here indicate that concentrations in excess of a fivefold increase in antibody concentrations used in the immunotherapy trial and equivalent to doubling normal peripheral blood antibody concentrations are anticipated for the successful reversal of sequestration to occur. It is suggested that immunotherapy aimed at either parasite ligands or host receptors to reverse sequestration in the treatment of severe malaria infections is unlikely to be successful given the complexity and number of receptors and ligands and the calculated concentrations of antibodies required.


Assuntos
Anticorpos/uso terapêutico , Malária Falciparum/terapia , Animais , Anticorpos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/uso terapêutico , Antimaláricos/uso terapêutico , Endotélio Vascular/metabolismo , Eritrócitos/parasitologia , Humanos , Ligantes , Malária Falciparum/diagnóstico , Malária Falciparum/imunologia , Modelos Teóricos , Receptores Imunológicos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA