Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33332286

RESUMEN

CIS43 is a potent neutralizing human mAb that targets a highly conserved "junctional" epitope in the Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP). Enhancing the durability of CIS43 in vivo will be important for clinical translation. Here, 2 approaches were used to improve the durability of CIS43 in vivo while maintaining potent neutralization. First, the Fc domain was modified with the LS mutations (CIS43LS) to increase CIS43 binding affinity for the neonatal Fc receptor (FcRn). CIS43LS and CIS43 showed comparable in vivo protective efficacy. CIS43LS had 9- to 13-fold increased binding affinity for human (6.2 nM versus 54.2 nM) and rhesus (25.1 nM versus 325.8 nM) FcRn at endosomal pH 6.0 compared with CIS43. Importantly, the half-life of CIS43LS in rhesus macaques increased from 22 days to 39 days compared with CIS43. The second approach for sustaining antibody levels of CIS43 in vivo is through adeno-associated virus (AAV) expression. Mice administered once with AAV-expressing CIS43 had sustained antibody levels of approximately 300 µg/mL and mediated protection against sequential malaria challenges up to 36 weeks. Based on these data, CIS43LS has advanced to phase I clinical trials, and AAV delivery provides a potential next-generation approach for malaria prevention.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Sustitución de Aminoácidos , Animales , Anticuerpos Antiidiotipos/biosíntesis , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/genética , Anticuerpos Antiprotozoarios/administración & dosificación , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/genética , Dependovirus/genética , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/genética , Macaca mulatta , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , Proteínas Protozoarias/inmunología
2.
Malar J ; 17(1): 304, 2018 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-30126436

RESUMEN

BACKGROUND: Plasmodium enolase is a target for the growth neutralizing antibodies. Interestingly, the three invasive stages i.e. sporozoites, merozoites, and ookinetes express this protein on their cell surface. Polyclonal anti-Plasmodium falciparum enolase (Pfeno) antibodies disrupt traversal of ookinete through mosquito mid-gut wall as well as have inhibitory effect on parasite growth at erythrocytic stage. In a recent study, it was observed that immunization with a unique epitope of parasite enolase (EWGWS) could confer partial protection against mouse malaria. Further validation is needed for the protective potential of this unique epitope in otherwise highly conserved enolase. METHODS: In order to investigate the efficacy of growth inhibitory potential of the epitope of P falciparum enolase, a monoclonal antibody specific to EWGWS is generated. In vitro parasite growth inhibition assays and passive immunization of Plasmodium yoelii (or Plasmodium berghei) infected mice were used to assess the parasite growth neutralizing activity of the antibody. RESULTS: Screening a panel of monoclonal antibodies raised against recombinant Pfeno that were specific to EWGWS resulted in isolation of H12E1. This antibody recognized only EWGWS epitope containing enolases. H12E1 strongly inhibited parasite growth in culture. This inhibition was strain transcending. Passive infusion of this antibody in P. yoelii or P. berghei infected mice showed significant reduction in parasitemia as compared to controls (p < 0.001). Surface Plasmon Resonance measurements indicated high affinity binding of H12E1 to P. falciparum enolase (KD ~ 7.6 × 10-9M). CONCLUSIONS: A monoclonal antibody directed against EWGWS epitope of Pfeno was shown to inhibit the growth of blood stage malarial parasites. This inhibition was species/strain transcending and is likely to arise due to blockade of enolase on the surface of merozoites, functionally implicating Pfeno in invasion related events. Presence of enolase on the cell surface of merozoites and ookinetes could potentially result in inhibition of host cell invasions at erythrocytic and transmission stages in the parasite life cycle. It is suggested that antibodies against EWGWS epitope have the potential to confer dual stage, species and strain transcending protection against malaria.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Malaria/prevención & control , Fosfopiruvato Hidratasa/inmunología , Plasmodium falciparum/enzimología , Plasmodium falciparum/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Antiprotozoarios/administración & dosificación , Modelos Animales de Enfermedad , Inmunización Pasiva , Malaria/inmunología , Masculino , Ratones , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología
3.
Sci Rep ; 8(1): 10511, 2018 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-30002416

RESUMEN

Plasmodium vivax merozoite invasion is restricted to Duffy positive reticulocytes. Merozoite interaction with the Duffy antigen is mediated by the P. vivax Duffy binding protein (PvDBP). The receptor-binding domain of PvDBP maps to an N-terminal cysteine-rich region referred to as region II (PvDBPII). In addition, a family of P. vivax reticulocyte binding proteins (PvRBPs) mediates interactions with reticulocyte receptors. The receptor binding domain of P. vivax reticulocyte binding protein 1a (PvRBP1a) maps to a 30 kD region (PvRBP1a30). Antibodies raised against recombinant PvRBP1a30 and PvDBPII recognize the native P. vivax antigens and inhibit their binding to host receptors. Rabbit IgG purified from sera raised against PvRBP1a30 and PvDBPII were tested individually and in combination for inhibition of reticulocyte invasion by P. vivax field isolates. While anti-PvDBPII rabbit IgG inhibits invasion, anti-PvRBP1a30 rabbit IgG does not show significant invasion inhibitory activity. Combining antibodies against PvDBPII and PvRBP1a30 also does not increase invasion inhibitory activity. These studies suggest that although PvRBP1a mediates reticulocyte invasion by P. vivax merozoites, it may not be useful to include PvRBP1a30 in a blood stage vaccine for P. vivax malaria. In contrast, these studies validate PvDBPII as a promising blood stage vaccine candidate for P. vivax malaria.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Reticulocitos/parasitología , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Anticuerpos Antiprotozoarios/aislamiento & purificación , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/metabolismo , Bioensayo/métodos , Células COS , Chlorocebus aethiops , Humanos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/inmunología , Inmunoglobulina G/aislamiento & purificación , Vacunas contra la Malaria/administración & dosificación , Malaria Vivax/inmunología , Malaria Vivax/virología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Merozoítos/inmunología , Merozoítos/patogenicidad , Ratones , Plasmodium vivax/genética , Plasmodium vivax/patogenicidad , Dominios y Motivos de Interacción de Proteínas/genética , Dominios y Motivos de Interacción de Proteínas/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Conejos , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Reticulocitos/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
4.
Elife ; 72018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29914622

RESUMEN

Anti-malarial pre-erythrocytic vaccines (PEV) target transmission by inhibiting human infection but are currently partially protective. It has been posited, but never demonstrated, that co-administering transmission-blocking vaccines (TBV) would enhance malaria control. We hypothesized a mechanism that TBV could reduce parasite density in the mosquito salivary glands, thereby enhancing PEV efficacy. This was tested using a multigenerational population assay, passaging Plasmodium berghei to Anopheles stephensi mosquitoes. A combined efficacy of 90.8% (86.7-94.2%) was observed in the PEV +TBV antibody group, higher than the estimated efficacy of 83.3% (95% CrI 79.1-87.0%) if the two antibodies acted independently. Higher PEV efficacy at lower mosquito parasite loads was observed, comprising the first direct evidence that co-administering anti-sporozoite and anti-transmission interventions act synergistically, enhancing PEV efficacy across a range of TBV doses and transmission intensities. Combining partially effective vaccines of differing anti-parasitic classes is a pragmatic, powerful way to accelerate malaria elimination efforts.


Asunto(s)
Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Antiprotozoarios/administración & dosificación , Vacunas contra la Malaria/administración & dosificación , Malaria/prevención & control , Plasmodium berghei/inmunología , Esporozoítos/inmunología , Animales , Anopheles/parasitología , Sinergismo Farmacológico , Femenino , Humanos , Malaria/inmunología , Malaria/parasitología , Ratones , Mosquitos Vectores/parasitología , Carga de Parásitos , Plasmodium berghei/efectos de los fármacos , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Glándulas Salivales/parasitología , Esporozoítos/química , Trofozoítos/química , Trofozoítos/inmunología
5.
PLoS One ; 12(12): e0189878, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29244862

RESUMEN

East Coast Fever (ECF) is the most economically important production disease among traditional beef cattle farmers in Zambia. Despite the disease control efforts by the government, donors, and farmers, ECF cases are increasing. Why does ECF oscillate over time? Can alternative approaches such as systems thinking contribute solutions to the complex ECF problem, avoid unintended consequences, and achieve sustainable results? To answer these research questions and inform the design and implementation of ECF interventions, we qualitatively investigated the influence of dynamic socio-economic, cultural, and ecological factors. We used system dynamics modelling to specify these dynamics qualitatively, and an innovative participatory framework called spatial group model building (SGMB). SGMB uses participatory geographical information system (GIS) concepts and techniques to capture the role of spatial phenomenon in the context of complex systems, allowing stakeholders to identify spatial phenomenon directly on physical maps and integrate such information in model development. Our SGMB process convened focus groups of beef value chain stakeholders in two distinct production systems. The focus groups helped to jointly construct a series of interrelated system dynamics models that described ECF in a broader systems context. Thus, a complementary objective of this study was to demonstrate the applicability of system dynamics modelling and SGMB in animal health. The SGMB process revealed policy leverage points in the beef cattle value chain that could be targeted to improve ECF control. For example, policies that develop sustainable and stable cattle markets and improve household income availability may have positive feedback effects on investment in animal health. The results obtained from a SGMB process also demonstrated that a "one-size-fits-all" approach may not be equally effective in policing ECF in different agro-ecological zones due to the complex interactions of socio-ecological context with important, and often ignored, spatial patterns.


Asunto(s)
Crianza de Animales Domésticos , Theileriosis/virología , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Bovinos , Sistemas de Información Geográfica , Inmunización , Theileriosis/epidemiología , Theileriosis/fisiopatología , Zambia
6.
Poult Sci ; 95(2): 439-46, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26772659

RESUMEN

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.


Asunto(s)
Anticuerpos Antiprotozoarios/farmacología , Proteínas Aviares/metabolismo , Pollos , Coccidiosis/veterinaria , Interleucina-10/metabolismo , Enfermedades de las Aves de Corral/prevención & control , Alimentación Animal/análisis , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Pollos/crecimiento & desarrollo , Coccidiosis/parasitología , Coccidiosis/prevención & control , Dieta/veterinaria , Eimeria/fisiología , Femenino , Enfermedades de las Aves de Corral/parasitología
7.
PLoS Pathog ; 11(6): e1004942, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26110623

RESUMEN

African trypanosomiasis is a deadly neglected disease caused by the extracellular parasite Trypanosoma brucei. Current therapies are characterized by high drug toxicity and increasing drug resistance mainly associated with loss-of-function mutations in the transporters involved in drug import. The introduction of new antiparasitic drugs into therapeutic use is a slow and expensive process. In contrast, specific targeting of existing drugs could represent a more rapid and cost-effective approach for neglected disease treatment, impacting through reduced systemic toxicity and circumventing resistance acquired through impaired compound uptake. We have generated nanoparticles of chitosan loaded with the trypanocidal drug pentamidine and coated by a single domain nanobody that specifically targets the surface of African trypanosomes. Once loaded into this nanocarrier, pentamidine enters trypanosomes through endocytosis instead of via classical cell surface transporters. The curative dose of pentamidine-loaded nanobody-chitosan nanoparticles was 100-fold lower than pentamidine alone in a murine model of acute African trypanosomiasis. Crucially, this new formulation displayed undiminished in vitro and in vivo activity against a trypanosome cell line resistant to pentamidine as a result of mutations in the surface transporter aquaglyceroporin 2. We conclude that this new drug delivery system increases drug efficacy and has the ability to overcome resistance to some anti-protozoal drugs.


Asunto(s)
Resistencia a Medicamentos/efectos de los fármacos , Terapia Molecular Dirigida/métodos , Pentamidina/administración & dosificación , Tripanocidas/administración & dosificación , Tripanosomiasis Africana/tratamiento farmacológico , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Quitosano/administración & dosificación , Quitosano/farmacocinética , Modelos Animales de Enfermedad , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Ensayo de Cambio de Movilidad Electroforética , Femenino , Concentración 50 Inhibidora , Ratones , Ratones Endogámicos C57BL , Nanopartículas/uso terapéutico , Pentamidina/farmacocinética , Reacción en Cadena en Tiempo Real de la Polimerasa , Tripanocidas/farmacocinética
8.
Vet Res ; 45: 25, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24571471

RESUMEN

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.


Asunto(s)
Anticuerpos Antiprotozoarios/farmacología , Pollos , Coccidiosis/veterinaria , Eimeria tenella/fisiología , Interleucina-17/administración & dosificación , Enfermedades de las Aves de Corral/prevención & control , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antiprotozoarios/administración & dosificación , Ciego/efectos de los fármacos , Ciego/parasitología , Coccidiosis/parasitología , Coccidiosis/prevención & control , Células Epiteliales/efectos de los fármacos , Células Epiteliales/parasitología , Enfermedades de las Aves de Corral/parasitología , Esquizontes/efectos de los fármacos , Esquizontes/crecimiento & desarrollo , Esquizontes/fisiología
9.
Br J Pharmacol ; 165(7): 2341-53, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22013955

RESUMEN

BACKGROUND AND PURPOSE: Nanobodies are promising antigen-binding moieties for molecular imaging and therapeutic purposes because of their favourable pharmacological and pharmacokinetic properties. However, the capability of monovalent nanobodies to reach targets in the CNS remains to be demonstrated. EXPERIMENTAL APPROACH: We have assessed the blood-brain barrier permeability of Nb_An33, a nanobody against the Trypanosoma brucei brucei variant-specific surface glycoprotein (VSG). This analysis was performed in healthy rats and in rats that were in the encephalitic stage of African trypanosomiasis using intracerebral microdialysis, single photon emission computed tomography (SPECT) or a combination of both methodologies. This enabled the quantification of unlabelled and (99m) Tc-labelled nanobodies using, respectively, a sensitive VSG-based nanobody-detection elisa, radioactivity measurement in collected microdialysates and SPECT image analysis. KEY RESULTS: The combined read-out methodologies showed that Nb_An33 was detected in the brain of healthy rats following i.v. injection, inflammation-induced damage to the blood-brain barrier, as in the late encephalitic stage of trypanosomiasis, significantly increased the efficiency of passage of the nanobody through this barrier. Complementing SPECT analyses with intracerebral microdialysis improved analysis of brain disposition. There is clear value in assessing penetration of the blood-brain barrier by monovalent nanobodies in models of CNS inflammation. Our data also suggest that rapid clearance from blood might hamper efficient targeting of specific nanobodies to the CNS. CONCLUSIONS AND IMPLICATIONS: Nanobodies can enter the brain parenchyma from the systemic circulation, especially in pathological conditions where the blood-brain barrier integrity is compromised.


Asunto(s)
Anticuerpos Antiprotozoarios/administración & dosificación , Anticuerpos Antiprotozoarios/metabolismo , Barrera Hematoencefálica/inmunología , Nanoestructuras , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Barrera Hematoencefálica/parasitología , Masculino , Microdiálisis/métodos , Ratas , Ratas Wistar , Tecnecio Tc 99m Sestamibi/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único , Trypanosoma brucei brucei/inmunología , Tripanosomiasis Africana/diagnóstico por imagen , Tripanosomiasis Africana/inmunología , Tripanosomiasis Africana/parasitología , Microtomografía por Rayos X
11.
Antimicrob Agents Chemother ; 54(4): 1385-92, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20086143

RESUMEN

At present no completely effective treatments are available for Cryptosporidium parvum infections in humans and livestock. Based on previous data showing the neutralizing potential of a panel of monoclonal antibodies developed against C. parvum, and based on the fact that innate immune peptides and enzymes have anticryptosporidial activity, we engineered several of these antibodies into antibody-biocide fusion proteins. We hypothesized that the combination of high-affinity antibody targeting with innate immune molecule-mediated killing would result in a highly effective new antiprotozoal agent. To test this hypothesis, we expressed antibody-biocide fusion proteins in a mammalian cell culture system and used the resulting products for in vitro and in vivo efficacy experiments. Antibody-biocide fusion proteins efficiently bound to, and destroyed, C. parvum sporozoites in vitro through a membrane-disruptive mechanism. When antibody-biocide fusion proteins were administered orally to neonatal mice in a prophylactic model of cryptosporidiosis, the induction of infection was reduced by as much as 81% in the mucosal epithelium of the gut, as determined on the basis of histopathological scoring of infectious stages. Several versions of antibody fusion proteins that differed in antigen specificity and in the biocide used had strong inhibitory effects on the initiation of infection. The results lay the groundwork for the development of a new class of antimicrobials effective against Cryptosporidium.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Antiprotozoarios/administración & dosificación , Criptosporidiosis/inmunología , Criptosporidiosis/prevención & control , Cryptosporidium parvum , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/genética , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/genética , Anticuerpos Antiprotozoarios/genética , Cryptosporidium parvum/inmunología , Inmunidad Innata , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Esporozoítos/inmunología
12.
J Immunol ; 179(6): 4093-100, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785848

RESUMEN

B and T lymphocyte attenuator (BTLA; CD272) is a coinhibitory receptor that is predominantly expressed on T and B cells and dampens T cell activation. In this study, we analyzed the function of BTLA during infection with Plasmodium berghei ANKA. Infection of C57BL/6 mice with this strain leads to sequestration of leukocytes in brain capillaries that is associated with a pathology resembling cerebral malaria in humans. During the course of infection, we found an induction of BTLA in several organs, which was either due to up-regulation of BTLA expression on T cells in the spleen or due to infiltration of BTLA-expressing T cells into the brain. In the brain, we observed a marked induction of BTLA and its ligand herpesvirus entry mediator during cerebral malaria, which was accompanied by an accumulation of predominantly CD8+ T cells, but also CD4+ T cells. Application of an agonistic anti-BTLA mAb caused a significantly reduced incidence of cerebral malaria compared with control mice. Treatment with this Ab also led to a decreased number of T cells that were sequestered in the brain of P. berghei ANKA-infected mice. Our findings indicate that BTLA-herpesvirus entry mediator interactions are functionally involved in T cell regulation during P. berghei ANKA infection of mice and that BTLA is a potential target for therapeutic interventions in severe malaria.


Asunto(s)
Malaria Cerebral/inmunología , Malaria Cerebral/prevención & control , Receptores Inmunológicos/metabolismo , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Antiprotozoarios/administración & dosificación , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/parasitología , Encéfalo/patología , Movimiento Celular/inmunología , Células Cultivadas , Femenino , Ligandos , Activación de Linfocitos/inmunología , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microcirculación/inmunología , Microcirculación/parasitología , Microcirculación/patología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/inmunología , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/fisiología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/deficiencia , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/fisiología , Linfocitos T/inmunología , Linfocitos T/parasitología , Linfocitos T/patología
13.
J Immunol ; 172(9): 5570-81, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15100300

RESUMEN

Immunizing pregnant women with a malaria vaccine is one approach to protecting the mother and her offspring from malaria infection. However, specific maternal Abs generated in response to vaccination and transferred to the fetus may interfere with the infant's ability to respond to the same vaccine. Using a murine model of malaria, we examined the effect of maternal 19-kDa C-terminal region of merozoite surface protein-1 (MSP1(19)) and Plasmodium yoelii Abs on the pups' ability to respond to immunization with MSP1(19). Maternal MSP1(19)-specific Abs but not P. yoelii-specific Abs inhibited Ab production following MSP1(19) immunization in 2-wk-old pups. This inhibition was correlated with the amount of maternal MSP1(19) Ab present in the pup at the time of immunization and was due to fewer specific B cells. Passively acquired Ab most likely inhibited the development of an Ab response by blocking access to critical B cell epitopes. If a neonate's ability to respond to MSP1(19) vaccination depends on the level of maternal Abs present at the time of vaccination, it may be necessary to delay immunization until Abs specific for the vaccinating Ag have decreased.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/farmacología , Regulación hacia Abajo/inmunología , Intercambio Materno-Fetal/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/fisiología , Plasmodium yoelii/inmunología , Animales , Animales Recién Nacidos , Anticuerpos Antiprotozoarios/administración & dosificación , Especificidad de Anticuerpos , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/metabolismo , División Celular/inmunología , Femenino , Inmunidad Materno-Adquirida/inmunología , Inmunización Pasiva , Inmunofenotipificación , Recuento de Linfocitos , Proteína 1 de Superficie de Merozoito/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Peso Molecular , Embarazo
14.
15.
Infect Immun ; 70(12): 6715-25, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12438346

RESUMEN

Chagas' disease results from infection with Trypanosoma cruzi, a protozoan parasite that establishes systemic intracellular infection after mucosal invasion. We hypothesized that ideal vaccines for mucosally invasive, intracellular pathogens like T. cruzi should induce mucosal type 2 immunity for optimal induction of protective secretory immunoglobulin A (IgA) and systemic type 1 immunity protective against intracellular replication. However, differential mucosal and systemic immune memory could be difficult to induce because of reciprocal inhibitory actions between type 1 and type 2 responses. To test our hypotheses, we investigated the protective effects of type 1 and type 2 biased vaccines against mucosal and systemic T. cruzi challenges. Intranasal vaccinations were given with recombinant interleukin-12 (IL-12)- and IL-4-neutralizing antibody (Ab) for type 1 immune bias, or recombinant IL-4 and gamma interferon-neutralizing Ab for type 2 immune bias. Cytokine RNA and protein studies confirmed that highly polarized memory immune responses were induced by our vaccination protocols. Survival after virulent subcutaneous T. cruzi challenge was used to assess systemic protection. Mucosal protection was assessed by measuring the relative inhibition of parasite replication in mucosal tissues early after oral T. cruzi challenge, using both PCR and quantitative culture techniques. As expected, only type 1 responses protected against systemic challenges (P < 0.01). However, contrary to our original hypothesis, type 1 responses optimally protected against mucosal challenges as well (P < 0.05). Type 1 and type 2 biased vaccines induced similar secretory IgA responses. We conclude that future vaccines for T. cruzi and possibly other mucosally invasive, intracellular pathogens should induce both mucosal and systemic type 1 immunity.


Asunto(s)
Enfermedad de Chagas/prevención & control , Inmunidad Mucosa , Vacunas Antiprotozoos/inmunología , Células TH1/inmunología , Trypanosoma cruzi/inmunología , Administración Intranasal , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/administración & dosificación , Antígenos de Protozoos/inmunología , Enfermedad de Chagas/inmunología , Citocinas/administración & dosificación , Citocinas/genética , Citocinas/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Vacunas Antiprotozoos/administración & dosificación , Células Th2/inmunología , Trypanosoma cruzi/crecimiento & desarrollo , Vacunación
16.
J Immunol ; 166(10): 6236-41, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11342646

RESUMEN

Plasmodium berghei XAT is an irradiation-induced attenuated variant derived from the lethal strain P. berghei NK65, and its blood-stage parasites are spontaneously cleared in immune competent mice. In the present study, we studied the mechanism of host resistance to blood-stage malaria infection using P. berghei XAT. Infection enhanced Ab-dependent phagocytosis of PRBC by splenic macrophages in wild-type C57BL/6 mice. In contrast, FcR gamma-chain knockout (FcRgamma(-/-)) mice, which lack the ability to mediate Ab-dependent phagocytosis and Ab-dependent cell-mediated cytotoxicity through FcgammaRI, FcgammaRII, and FcgammaRIII, could not induce Ab-dependent phagocytic activity. These FcRgamma(-/-) mice showed increased susceptibility to the P. berghei XAT infection, with eventually fatal results, although they produced comparable amounts of IFN-gamma by spleen cells and anti-XAT Abs in serum. In addition, passive transfer of anti-XAT IgG obtained from wild-type mice that had recovered from infection into FcRgamma(-/-) mice could not suppress the increase in parasitemia, and almost all of these mice died after marked parasitemia. In contrast, passive transfer of anti-XAT IgG into control wild-type mice inhibited the increase in parasitemia. IFN-gamma(-/-) mice, which were highly susceptible to the P. berghei XAT infection, failed to induce Ab-dependent phagocytic activity and also showed reduced production of serum anti-XAT IgG2a isotype compared with control wild-type mice. These results suggest that FcR-mediated Ab-dependent phagocytosis, which is located downstream of IFN-gamma production, is important as an effector mechanism to eliminate PRBC in blood-stage P. berghei XAT infection.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Malaria/inmunología , Malaria/parasitología , Fagocitosis/inmunología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/inmunología , Receptores Fc/fisiología , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Transfusión de Eritrocitos , Eritrocitos/parasitología , Femenino , Predisposición Genética a la Enfermedad , Inmunidad Innata/genética , Inmunización Pasiva/métodos , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/sangre , Inyecciones Intravenosas , Interferón gamma/deficiencia , Interferón gamma/genética , Macrófagos/inmunología , Macrófagos/parasitología , Malaria/sangre , Malaria/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Fagocitosis/genética , Bazo/inmunología , Bazo/parasitología
17.
J Exp Med ; 192(11): 1653-60, 2000 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-11104807

RESUMEN

We have recently described that sustained Plasmodium falciparum growth could be obtained in immunodeficient mice. We now report the potential of this new mouse model by assaying the effect of the passive transfer of antibodies (Abs) which in humans have had a well-established effect.Our results show that the total African adult hyperimmune immunoglobulin Gs (HI-IgGs) strongly reduce P. falciparum parasitemia similarly to that reported in humans, but only when mice are concomitantly reconstituted with human monocytes (HuMNs). In contrast, neither HI-IgGs nor HuMNs alone had any direct effect upon parasitemia. We assessed the in vivo effect of epitope-specific human Abs affinity-purified on peptides derived either from the ring erythrocyte surface antigen (RESA) or the merozoite surface protein 3 (MSP3). The inoculation of low concentrations of anti-synthetic peptide from MSP3, but not of anti-RESA Abs, consistently suppressed P. falciparum in the presence of HuMNs. Parasitemia decrease was stronger and faster than that observed using HI-IgGs and as fast as that induced by chloroquine. Our observations demonstrate that this mouse model is of great value to evaluate the protective effect of different Abs with distinct specificity in the same animal, a step hardly accessible and therefore never performed before in humans.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunización Pasiva , Huésped Inmunocomprometido , Malaria Falciparum/sangre , Malaria Falciparum/prevención & control , Masculino , Ratones , Datos de Secuencia Molecular , Monocitos/inmunología , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/inmunología
18.
Clin Immunol ; 97(2): 182-8, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11027459

RESUMEN

Systemic and mucosal and immune responses can be manipulated with immunomodulators. Here we show the modulatory effects of cholera toxin (CT) and beta-1,3-glucan (GLU) on the rat antiamebic serum and fecal antibody responses to one or four intraperitoneal (IP) or intragastric (IG) doses of glutaraldehyde-fixed Entamoeba histolytica trophozoites (GFT). One IP dose of GFT maximized serum IgM and IgG antiamebic antibodies on days 4 and 9, respectively; CT coadministration increased IgM antibodies, whereas IgG titers increased with CT or GLU; coproantibodies were undetectable after GFT alone or coadministered with GLU, whereas CT coadministration maximized fecal IgA antibodies on day 6. One IG dose of GFT alone increased serum IgM and IgG antibodies 2.5 times and no further increases were detected using GLU, whereas CT doubled serum IgG antibodies; GFT did not affect the coproantibody responses, whereas GLU coadministration maximized IgG coproantibody levels on day 6 and CT increased IgG and IgA coproantibody levels on the same day. On the other hand, four IG doses of GFT alone or with GLU induced tolerance, whereas GFT alone via the IP route increased serum antibodies slightly and GLU coadministration increased serum IgG antibody titers 300-fold. CT coadministration by both routes increased IgA coproantibodies, and simultaneous CT+GLU coadministration induced lower responses than either CT or GLU. Different antiamebic immune responses might therefore be attained through the use of different immunization routes and immunomodulators to induce protective immunity against intestinal or extraintestinal amebiasis.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Antígenos de Superficie/inmunología , Toxina del Cólera/farmacología , Entamoeba histolytica/inmunología , Glucanos/farmacología , Mucosa Intestinal/inmunología , beta-Glucanos , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Formación de Anticuerpos/efectos de los fármacos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Infusiones Parenterales , Inyecciones Intraperitoneales , Ratas , Factores de Tiempo
19.
Infect Immun ; 68(7): 4312-8, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10858250

RESUMEN

Antibodies against the Plasmodium falciparum P0 ribosomal phosphoprotein (PfP0) have been detected exclusively but extensively in malaria-immune persons. Polyclonal rabbit and mice sera were raised against two recombinant polypeptides of P. falciparum P0 protein, PfP0N and PfP0C, covering amino acids 17 to 61 and the remaining amino acids 61 to 316, respectively. Sera against both these domains detected a 35-kDa protein from Plasmodium yoelii subsp. yoelii, a rodent malarial parasite, and stained the surface of merozoites in immunofluorescence assays. Total immunoglobulin G (IgG) purified from rabbit and mouse anti-PfP0 sera by ammonium sulfate and DEAE-cellulose chromatography was used for passive transfer experiments in mice. Mice passively immunized with both anti-PfP0N and anti-PfP0C showed distinctly lower levels of parasitemia than control mice. With immunizations on days -1, 0, 1, 3, and 5, about 50% of both sets of mice receiving anti-PfP0N and anti-PfP0C cleared the lethal 17XL strain of P. yoelii and revived by day 25. All the control mice died by day 10. By extending the immunization schedule, the survival period of the mice could be extended for every mouse that received anti-PfP0 IgG. These data demonstrate the cross-protection of the anti-PfP0 IgG and establish parasite P0 protein as a target for invasion-blocking antibodies.


Asunto(s)
Anticuerpos Antiprotozoarios/administración & dosificación , Fosfoproteínas/inmunología , Plasmodium falciparum/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Proteínas Ribosómicas/inmunología , Animales , Humanos , Inmunización Pasiva , Malaria/inmunología , Malaria/prevención & control , Ratones , Ratones Endogámicos BALB C , Parasitemia/inmunología , Parasitemia/prevención & control , Fosfoproteínas/química , Plasmodium yoelii/aislamiento & purificación , Plasmodium yoelii/patogenicidad , Estructura Terciaria de Proteína , Conejos , Proteínas Ribosómicas/química , Factores de Tiempo
20.
J Exp Med ; 191(6): 1063-8, 2000 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-10727468

RESUMEN

We show here that maintenance of Leishmania infections with Leishmania mexicana complex parasites (Leishmania amazonensis and Leishmania pifanoi) is impaired in the absence of circulating antibody. In these studies, we used mice genetically altered to contain no circulating antibody, with and without functional B cells. This experimental design allowed us to rule out a critical role for B cell antigen presentation in Leishmania pathogenesis. In addition, we show that mice lacking the common gamma chain of Fc receptors (FcgammaRI, FcepsilonRI, and FcgammaRIII) are similarly refractory to infection with these parasites. These observations establish a critical role for antibody in the pathogenesis associated with infection by members of the L. mexicana complex.


Asunto(s)
Leishmania mexicana/crecimiento & desarrollo , Leishmania mexicana/metabolismo , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Receptores Fc/fisiología , Animales , Anticuerpos Antiprotozoarios/administración & dosificación , Anticuerpos Antiprotozoarios/biosíntesis , Anticuerpos Antiprotozoarios/sangre , Inmunización Pasiva , Leishmania mexicana/inmunología , Leishmaniasis Cutánea/etiología , Leishmaniasis Cutánea/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Receptores Fc/deficiencia , Receptores Fc/genética , Receptores de IgG/deficiencia , Receptores de IgG/genética , Receptores de IgG/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...