Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 135
Filtrar
1.
PLoS Negl Trop Dis ; 15(8): e0009627, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34403413

RESUMO

Visceral leishmaniasis is a protozoan disease associated with high fatality rate in developing countries. Although the drug pipeline is constantly improving, available treatments are costly and live-threatening side effects are not uncommon. Moreover, an approved vaccine against human leishmaniasis does not exist yet. Using whole antigens from Leishmania donovani promastigotes (LdAg), we investigated the protective potential of a novel adjuvant-free vaccine strategy. Immunization of mice with LdAg via the intradermal or the intranasal route prior to infection decreases the parasitic burden in primary affected internal organs, including the liver, spleen, and bone marrow. Interestingly, the intranasal route is more efficient than the intradermal route, leading to better parasite clearance and remarkable induction of adaptive immune cells, notably the helper and cytotoxic T cells. In vitro restimulation experiments with Leishmania antigens led to significant IFN-γ secretion by splenocytes; therefore, exemplifying specificity of the adaptive immune response. To improve mucosal delivery and the immunogenic aspects of our vaccine strategy, we used polysaccharide-based nanoparticles (NP) that carry the antigens. The NP-LdAg formulation is remarkably taken up by dendritic cells and induces their maturation in vitro, as revealed by the increased expression of CD80, CD86 and MHC II. Intranasal immunization with NP-LdAg does not improve the parasite clearance in our experimental timeline; however, it does increase the percentage of effector and memory T helper cells in the spleen, suggesting a potential induction of long-term memory. Altogether, this study provides a simple and cost-effective vaccine strategy against visceral leishmaniasis based on LdAg administration via the intranasal route, which could be applicable to other parasitic diseases.


Assuntos
Antígenos de Protozoários/imunologia , Medula Óssea/parasitologia , Vacinas contra Leishmaniose/imunologia , Leishmaniose Visceral/imunologia , Fígado/parasitologia , Baço/parasitologia , Imunidade Adaptativa , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/sangue , Medula Óssea/metabolismo , Feminino , Imunização , Interferon gama/metabolismo , Leishmania donovani/imunologia , Vacinas contra Leishmaniose/administração & dosagem , Leishmaniose Visceral/parasitologia , Leishmaniose Visceral/prevenção & controle , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Baço/metabolismo
2.
Parasitol Res ; 119(8): 2609-2622, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32535734

RESUMO

The treatment against visceral leishmaniasis (VL) presents problems, mainly related to the toxicity and/or high cost of the drugs. In this context, a prophylactic vaccination is urgently required. In the present study, a Leishmania protein called LiHyE, which was suggested recently as an antigenic marker for canine and human VL, was evaluated regarding its immunogenicity and protective efficacy in BALB/c mice against Leishmania infantum infection. In addition, the protein was used to stimulate peripheral blood mononuclear cells (PBMCs) from VL patients before and after treatment, as well as from healthy subjects. Vaccination results showed that the recombinant (rLiHyE) protein associated with liposome or saponin induced effective protection in the mice, since significant reductions in the parasite load in spleen, liver, draining lymph nodes, and bone marrow were found. The parasitological protection was associated with Th1-type cell response, since high IFN-γ, IL-12, and GM-CSF levels, in addition to low IL-4 and IL-10 production, were found. Liposome induced a better parasitological and immunological protection than did saponin. Experiments using PBMCs showed rLiHyE-stimulated lymphoproliferation in treated patients' and healthy subjects' cells, as well as high IFN-γ levels in the cell supernatant. In conclusion, rLiHyE could be considered for future studies as a vaccine candidate against VL.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Protozoários/administração & dosagem , Leishmania infantum/imunologia , Leishmaniose Visceral/prevenção & controle , Animais , Antígenos de Protozoários/imunologia , Feminino , Humanos , Imunogenicidade da Vacina , Leishmaniose Visceral/imunologia , Leishmaniose Visceral/parasitologia , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Carga Parasitária , Células Th1/imunologia , Vacinação
3.
PLoS One ; 15(2): e0229121, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32101539

RESUMO

Since dogs play a central role in the contamination of humans and livestock with Echinococcus granulosus, the development of an effective vaccine for dogs is essential to control the disease caused by this parasite. For this purpose, a formulation based on biodegradable polymeric nanoparticles (NPs) as delivery system of recombinant Echinococcus granulosus antigen (tropomyosin EgTrp) adjuved with monophosphoryl lipid A (MPLA) has been developed. The obtained nanoparticles had a size of approximately 200 nm in diameter into which the antigen was correctly preserved and encapsulated. The efficiency of this system to deliver the antigen was evaluated in vitro on canine monocyte-derived dendritic cells (cMoDCs) generated from peripheral blood monocytes. After 48 h of contact between the formulations and cMoDCs, we observed no toxic effect on the cells but a strong internalization of the NPs, probably through different pathways depending on the presence or not of MPLA. An evaluation of cMoDCs activation by flow cytometry showed a stronger expression of CD80, CD86, CD40 and MHCII by cells treated with any of the tested formulations or with LPS (positive control) in comparison to cells treated with PBS (negative control). A higher activation was observed for cells challenged with EgTrp-NPs-MPLA compared to EgTrp alone. Formulations with MPLA, even at low ratio of MPLA, give better results than formulations without MPLA, proving the importance of the adjuvant in the nanoparticles structure. Moreover, autologous T CD4+ cell proliferation observed in presence of cMoDCs challenged with EgTrp-NPs-MPLA was higher than those observed after challenged with EgTrp alone (p<0.05). These first results suggest that our formulation could be used as an antigen delivery system to targeting canine dendritic cells in the course of Echinococcus granulosus vaccine development.


Assuntos
Antígenos de Protozoários/administração & dosagem , Células Dendríticas/imunologia , Cães/parasitologia , Equinococose/prevenção & controle , Echinococcus granulosus/imunologia , Vacinas Protozoárias/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Cães/sangue , Cães/imunologia , Portadores de Fármacos/química , Portadores de Fármacos/toxicidade , Equinococose/imunologia , Equinococose/parasitologia , Equinococose/veterinária , Echinococcus granulosus/genética , Imunogenicidade da Vacina , Lipídeo A/análogos & derivados , Lipídeo A/química , Lipídeo A/toxicidade , Ativação Linfocitária/imunologia , Monócitos/fisiologia , Nanopartículas/química , Nanopartículas/toxicidade , Poliésteres/química , Poliésteres/toxicidade , Cultura Primária de Células , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Testes de Toxicidade Aguda , Tropomiosina/administração & dosagem , Tropomiosina/genética , Tropomiosina/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
4.
Parasite ; 26: 58, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31535970

RESUMO

Toxoplasma gondii infection is prevalent in humans and animals worldwide. In this study, recombinant eukaryotic expression plasmids (pVAX-GRA24, pVAX-GRA25 and pVAX-MIC6) were constructed, and then injected into Kunming mice intramuscularly, as cocktailed plasmids or as single-gene plasmids. We evaluated immune protective responses by detecting the titer of antibodies and cytokine production of IFN-γ, IL-2, IL-4, IL-10, IL-12 and IL-23, the percentages of the subclasses of T lymphocytes, as well as the records of the survival time and cyst decrement in the brain of the mouse model after challenge with the T. gondii RH and Pru strains, respectively. Compared with the control groups, antibody and cytokine production were significantly increased, while the survival times of mice in all immunized groups were also prolonged, and the number of T. gondii cysts in their brains were decreased significantly (29.03% for pVAX-GRA24; 40.88% for pVAX-GRA25; 37.70% for pVAX-MIC6; 48.06% for pVAX-GRA24 + pVAX-GRA25; and 55.37% for pVAX-GRA24 + pVAX-GRA25 + pVAX-MIC6). The mouse group immunized with the three-gene cocktail (TgGRA24 + TgGRA25 + TgMIC6) had better performance in each detection index than the mouse groups immunized with the two-gene cocktail of TgGRA24 + TgGRA25, which was better than that in the group immunized with the single gene vaccine of TgGRA24, TgMIC6 or TgGRA25. In conclusion, TgGRA24 or TgGRA25 may be good vaccine candidates against T. gondii infection, but the three-gene cocktail of TgGRA24, TgMIC6 and TgGRA25 may induce the strongest protective immunity. Further studies of multi-antigenic DNA vaccines or cocktailed vaccines against T. gondii infection are necessary.


TITLE: Évaluation de la protection immunitaire contre l'infection par Toxoplasma gondii chez la souris, induite par un vaccin à ADN multi-antigénique contenant TgGRA24, TgGRA25 et TgMIC6. ABSTRACT: L'infection à Toxoplasma gondii est répandue chez les humains et les animaux dans le monde entier. Dans cette étude, des plasmides d'expression eucaryotes recombinants (pVAX-GRA24, pVAX-GRA25 et pVAX-MIC6) ont été construits, puis injectés à des souris Kunming par voie intramusculaire, comme cocktails de plasmides ou comme plasmides à un seul gène. Nous avons évalué les réponses de protection immunitaire en détectant le titre des anticorps et la production de cytokines IFN-γ, IL-2, IL-4, IL-10, IL-12 et IL-23, les pourcentages des sous-classes des lymphocytes T, ainsi qu'en mesurant les temps de survie et de décrément des kystes dans le cerveau du modèle souris après challenge par les souches RH et Pru de T. gondii, respectivement. Comparativement aux groupes témoins, la production d'anticorps et de cytokines était significativement accrue, tandis que le temps de survie des souris de tous les groupes immunisés était également prolongé et que le nombre de kystes de T. gondii dans leur cerveau diminuait de manière significative (29,03 % pour pVAX-GRA24 ; 40,88 % pour pVAX-GRA25 ; 37,70 % pour pVAX-MIC6 ; 48,06 % pour pVAX-GRA24 + pVAX-GRA25 ; 55,37 % pour pVAX-GRA24 + pVAX-GRA25 + pVAX-MIC6). Le groupe de souris immunisées avec les cocktails à trois gènes (TgGRA24 + TgGRA25 + TgMIC6) présentait la meilleure performance dans chaque indice de détection par rapport aux groupes de souris immunisées avec des cocktails à deux gènes de TgGRA24 + TgGRA25, qui était supérieur à ceux immunisés avec les vaccins monogéniques TgGRA24, TgMIC6 ou TgGRA25. En conclusion, TgGRA24 ou TgGRA25 peuvent être de bons candidats au vaccin contre l'infection à T. gondii, mais un cocktail à trois gènes de TgGRA24, TgMIC6 et TgGRA25 peut induire la plus forte immunité protectrice. Des études supplémentaires sur les vaccins à ADN multi-antigéniques ou les vaccins en cocktail contre l'infection à T. gondii sont nécessaires.


Assuntos
Antígenos de Protozoários/imunologia , Moléculas de Adesão Celular/genética , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/imunologia , Toxoplasmose Animal/prevenção & controle , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/administração & dosagem , Moléculas de Adesão Celular/imunologia , Citocinas/imunologia , Feminino , Imunidade Celular , Imunogenicidade da Vacina , Camundongos , Proteínas de Protozoários/genética , Vacinas Protozoárias/administração & dosagem , Organismos Livres de Patógenos Específicos , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
5.
Parasit Vectors ; 11(1): 575, 2018 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-30390674

RESUMO

BACKGROUND: Babesia bovis belongs to the phylum Apicomplexa and is the major causal agent of bovine babesiosis, the most important veterinary disease transmitted by arthropods. In apicomplexan parasites, the interaction between AMA1 and RON2 is necessary for the invasion process, and it is a target for vaccine development. In B. bovis, the existence of AMA1 has already been reported; however, the presence of a homolog of RON2 is unknown. The aim of this study was to characterize RON2 in B. bovis. RESULTS: The B. bovis ron2 gene has a similar synteny with the orthologous gene in the B. bigemina genome. The entire ron2 gene was sequenced from different B. bovis strains showing > 99% similarity at the amino acid and nucleotide level among all the sequences obtained, including the characteristic CLAG domain for cytoadherence in the amino acid sequence, as is described in other Apicomplexa. The in silico transcription analysis showed similar levels of transcription between attenuated and virulent B. bovis strains, and expression of RON2 was confirmed by western blot in the B. bovis T3Bo virulent strain. Four conserved peptides, containing predicted B-cell epitopes in hydrophilic regions of the protein, were designed and chemically synthesized. The humoral immune response generated by the synthetic peptides was characterized in bovines, showing that anti-RON2 antibodies against peptides recognized intraerythrocytic merozoites of B. bovis. Only peptides P2 and P3 generated partially neutralizing antibodies that had an inhibitory effect of 28.10% and 21.42%, respectively, on the invasion process of B. bovis in bovine erythrocytes. Consistently, this effect is additive since inhibition increased to 42.09% when the antibodies were evaluated together. Finally, P2 and P3 peptides were also recognized by 83.33% and 87.77%, respectively, of naturally infected cattle from endemic areas. CONCLUSIONS: The data support RON2 as a novel B. bovis vaccine candidate antigen that contains conserved B-cell epitopes that elicit partially neutralizing antibodies.


Assuntos
Antígenos de Protozoários/imunologia , Babesia bovis/imunologia , Babesiose/imunologia , Epitopos de Linfócito B/imunologia , Imunidade Humoral , Peptídeos/imunologia , Proteínas de Protozoários/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Babesia bovis/patogenicidade , Bovinos , Doenças dos Bovinos/imunologia , Doenças dos Bovinos/parasitologia , Simulação por Computador , Epitopos de Linfócito B/genética , Eritrócitos/parasitologia , Imunização , Peptídeos/administração & dosagem , Peptídeos/síntese química , Peptídeos/genética , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética
6.
Nat Nanotechnol ; 13(12): 1174-1181, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30297818

RESUMO

Pfs25 is a malaria transmission-blocking vaccine antigen candidate, but its apparently limited immunogenicity in humans has hindered clinical development. Here, we show that recombinant, polyhistidine-tagged (his-tagged) Pfs25 can be mixed at the time of immunization with pre-formed liposomes containing cobalt porphyrin-phospholipid, resulting in spontaneous nanoliposome antigen particleization (SNAP). Antigens are stably presented in uniformly orientated display via his-tag insertion in the cobalt porphyrin-phospholipid bilayer, without covalent modification or disruption of antigen conformation. SNAP immunization of mice and rabbits is well tolerated with minimal local reactogenicity, and results in orders-of-magnitude higher functional antibody generation compared with other 'mix-and-inject' adjuvants. Serum-stable antigen binding during transit to draining lymph nodes leads to enhanced antigen uptake by phagocytic antigen-presenting cells, with subsequent generation of long-lived, antigen-specific plasma cells. Seamless multiplexing with four additional his-tagged Plasmodium falciparum polypeptides induces strong and balanced antibody production, illustrating the simplicity of developing multistage particulate vaccines with SNAP immunization.


Assuntos
Antígenos de Protozoários/imunologia , Lipossomos/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Animais , Formação de Anticorpos , Antígenos de Protozoários/administração & dosagem , Feminino , Humanos , Imunização , Lipossomos/administração & dosagem , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/imunologia , Camundongos , Proteínas de Protozoários/administração & dosagem , Células RAW 264.7 , Coelhos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/imunologia
7.
Vet Parasitol ; 260: 12-21, 2018 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-30197008

RESUMO

Toxoplasma gondii rhoptry proteins (TgROPs) have been considered the main targets and indicator molecules for immune diagnosis and prophylaxis because they present during the initial process of invasion. The aim of this study was to evaluate the protective effect of a TgROP35 DNA vaccine on experimental mice subjected to T. gondii challenge. The effect of intramuscularly injecting the genetic vaccine pVAX-ROP35 into BALB/c mice was evaluated, by first inserting the TgROP35 sequence into the pVAX I eukaryotic expression vector. The levels of IgG, IgG1 and IgG2a in pVAX-ROP35-vaccinated animals were integrally increased. Cytokine profile analyses revealed that IFN-γ, IL-2 and IL-10 levels were significantly increased, while there were no significant differences in IL-4 expression between the pVAX-ROP35 immunized and control groups. Additionally, we found that immunization with pVAX-ROP35 significantly prolonged the survival time (13.90 ±â€¯1.97 days) of mice after challenge infection with the virulent T. gondii RH strain compared with that in non-vaccinated control animals (mice died within 10 days). Moreover, the number of brain cysts (1450 ±â€¯287) in the animals subjected to pVAX-TgROP35 vaccination decreased remarkably (P <  0.05) compared to that in the blank control mice (2333 ± 473). Furthermore, the size of brain cysts in the pVAX-TgROP35 group was significantly smaller than that in the blank control, PBS and pVAXI groups. The findings indicated that intense cell-mediated and humoural immunity was triggered and that defence mechanisms against T. gondii were partially induced after vaccination by TgROP35.


Assuntos
Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/prevenção & controle , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Encéfalo/parasitologia , Citocinas/sangue , Feminino , Imunidade Celular , Imunidade Humoral , Imunoglobulina G/sangue , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/administração & dosagem , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Toxoplasma/genética , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
8.
Vet Parasitol ; 258: 108-113, 2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-30105970

RESUMO

Refractile body protein, SO7, is a highly immunogenic protein which is essentially involved in the early development of Eimeria species infecting the domestic chicken. In the present study, the immune response and protective efficacy of recombinant Eimeria tenella SO7 (rEtSO7) protein was assessed in broiler chickens following homologous oocyst challenge. Broiler chicks were subcutaneously immunized with rEtSO7 antigen adjuvanted with Montanide ISA 71 VG on 7 and 21 days of age and protective efficacy of vaccination was evaluated in terms of body weight gain, lesion score and reduction in oocyst output. The peripheral blood lymphocyte proliferation, serum IgY response, and levels of interferon gamma (IFN-γ), interleukin 2 (IL-2), interleukin 4 (IL-4), tumor growth factor beta (TGF-ß) and nitric oxide (NO) were assessed. The results revealed significant reduction (p < 0.05) in the oocyst output and increased weight gain in immunized birds as compared to unimmunized birds. Significantly increased levels of serum IgY, IFN-γ and proliferation of lymphocytes were evident in rEtSO7 immunized chickens. The results demonstrated that the recombinant protein could effectively elicit the cellular and humoral immune responses in immunized chickens, and provided significant protection against caecal coccidiosis in chickens.


Assuntos
Antígenos de Protozoários/imunologia , Coccidiose/veterinária , Eimeria tenella/química , Eimeria tenella/imunologia , Imunização/veterinária , Doenças das Aves Domésticas/prevenção & controle , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Ceco/parasitologia , Galinhas/imunologia , Coccidiose/imunologia , Coccidiose/prevenção & controle , Eimeria tenella/genética , Fezes/parasitologia , Imunidade Celular , Imunidade Humoral , Imunoglobulinas/sangue , Interferon gama/imunologia , Interleucina-2/imunologia , Oocistos/isolamento & purificação , Oocistos/fisiologia , Doenças das Aves Domésticas/imunologia , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Proteínas Recombinantes/imunologia , Células Th1/imunologia , Aumento de Peso
9.
Biomed Pharmacother ; 106: 599-604, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29990848

RESUMO

Gamma radiation induces protein changes that enhance immunogenicity for venoms, used in antivenin production. Coccidian parasites exposed to gamma radiation elicit immune response with protection in mice and man, but without studies on the effect of gamma radiation in soluble acellular extracts or isolated proteins. Toxoplasmosis is a highly prevalent coccidian disease with only one vaccine for veterinary use but with remaining tissue cysts. Total parasite extracts or recombinant proteins used as immunogen induce usually low protection. Here, we study gamma radiation effect on T. gondii extracts proteins (STAG) and its induced immunity in experimental mice models. By SDS-PAGE, protein degradation is seen at high radiation doses, but at ideal dose (1500 Gy), there are preservation of the antigenicity and immunogenicity, detected by specific antibody recognition or production after mice immunization. Immunization with STAG irradiated at 1500 Gy induced significant protection in mice immunized and challenged with distinct T. gondii strains. In their blood, higher levels of specific CD19+, CD3+CD4+ and CD3+CD8+ activated cells were found when compared to mice immunized with STAG. Irradiated T. gondii tachyzoites extracts induce immune response and protection in mice in addition, could be a feasible alternative for Toxoplasma vaccine.


Assuntos
Antígenos de Protozoários/efeitos da radiação , Raios gama , Imunogenicidade da Vacina , Vacinas Protozoárias/efeitos da radiação , Toxoplasma/efeitos da radiação , Toxoplasmose/prevenção & controle , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/imunologia , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Imunidade Celular , Imunidade Humoral , Imunização , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Desnaturação Proteica , Estabilidade Proteica , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/imunologia , Fatores de Tempo , Toxoplasma/imunologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia
10.
Acta Parasitol ; 63(2): 368-374, 2018 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-29654677

RESUMO

In the present study, we have investigated the protective effect of a heterologous prime-boost strategy with priming plasmid DNA followed by recombinant adenovirus, both expressing BmAMA1, against Babesia microti infection. Four groups consisting of 3 hamsters per group were immunized with pBmAMA1/Ad5BmAMA1, pNull/Ad5BmAMA1, pBmAMA1/Ad5Null and pNull/Ad5Null, followed by challenge infection with B. microti. Our results showed that hamsters immunized with plasmid and adenovirus expressing BmAMA1 developed a robust IgG and IgG2a antibody response against BmAMA1, suggesting the DNA vaccine or viral vector vaccine tend to induce a Th1-biased response. Compared to the control hamsters, the hamsters vaccinated either with the prime-boost strategy or one of the two "vaccines" exhibited no significant protection against B. microti challenge. Although a slight difference in terms of parasitemia and hematocrit values at days 14-16 post challenge infection was observed, no other statistical difference was detected. Our results indicate that the prime-boost vaccination strategy of injection of plasmid and adenovirus expressing BmAMA1 is not efficient in protecting against B. microti infection.


Assuntos
Adenoviridae/genética , Antígenos de Protozoários/imunologia , Babesiose/prevenção & controle , Imunização Secundária/métodos , Proteínas de Protozoários/genética , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Babesia microti/genética , Babesia microti/imunologia , Babesiose/parasitologia , Cricetinae , Imunoglobulina G/sangue , Parasitemia/prevenção & controle , Plasmídeos/administração & dosagem , Plasmídeos/genética , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/imunologia , Vacinação/métodos , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
11.
Parasitology ; 145(4): 508-526, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28691653

RESUMO

Despite immense efforts, vaccine against visceral leishmaniasis has yet not been developed. Earlier our proteomic study revealed a novel protein, cofactor-independent phoshoglycerate mutase (LdiPGAM), an important enzyme in glucose metabolism, in T helper cells type 1 (Th1) stimulatory region of soluble Leishmania donovani antigen. In this study, LdiPGAM was biochemically and molecularly characterized and evaluated for its immunogenicity and prophylactic efficacy against L. donovani. Immunogenicity of recombinant LdiPGAM (rLdiPGAM) was initially assessed in naïve hamsters immunized with it by analysing mRNA expression of inducible nitric oxide (NO) synthase (iNOS) and other Th1/T helper cells type 2 cytokines, which revealed an upregulation of Th1 cytokines along with iNOS. Immunogenicity of rLdiPGAM was further evaluated in lymphocytes of treated Leishmania-infected hamsters and peripheral blood mononuclear cells of Leishmania patients in clinical remission by various parameters, viz. lymphoproliferation assay and NO production (hamsters and patients) and levels of various cytokines (patients). rLdiPGAM induced remarkable Lymphoproliferative response and NO production in treated Leishmania-infected hamsters as well as in patients and increase in interferon gamma (IFN-γ), interleukin-12 (IL-12p40) responses in Leishmania patients in clinical remission. Vaccination with rLdiPGAM exerted considerable prophylactic efficacy (73%) supported by increase in mRNA expression of iNOS, IFN-γ and IL-12p40 with decrease in transforming growth factor beta and interleukin-10. Above results indicate the importance of rLdiPGAM protein as a potential vaccine candidate against visceral leishmaniasis.


Assuntos
Antígenos de Protozoários/imunologia , Leishmania donovani/imunologia , Vacinas contra Leishmaniose/imunologia , Leishmaniose Visceral/prevenção & controle , Fosfoglicerato Mutase/genética , Fosfoglicerato Mutase/imunologia , Adolescente , Adulto , Animais , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Criança , Pré-Escolar , Cricetinae , Feminino , Humanos , Imunogenicidade da Vacina , Interferon gama/genética , Leishmania donovani/enzimologia , Vacinas contra Leishmaniose/administração & dosagem , Vacinas contra Leishmaniose/genética , Leishmaniose Visceral/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Óxido Nítrico , Fosfoglicerato Mutase/administração & dosagem , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Células Th1 , Células Th2 , Vacinação , Adulto Jovem
12.
Vaccine ; 35(30): 3726-3732, 2017 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-28578824

RESUMO

Malaria is a devastating disease caused by Plasmodium parasites, resulting in almost 0.5 million deaths per year. The Pfs48/45 protein exposed on the P. falciparum sexual stages is one of the most advanced antigen candidates for a transmission-blocking (TB) vaccine in the clinical pipeline. However, it remains essential to identify an optimal vaccine formulation that can facilitate induction of a long-lasting TB anti-Pfs48/45 response. Here we report on the development and evaluation of two Pfs48/45-based virus-like particle (VLP) vaccines generated using the AP205 SpyTag/Catcher VLP system. Two different recombinant proteins (SpyCatcher-R0.6C and SpyCatcher-6C), comprising the Pfs48/45-6C region, were covalently attached to the surface of Spy-tagged Acinetobacter phage AP205 VLPs. Resulting Pfs48/45-VLP complexes appeared as non-aggregated particles of ∼30nm, each displaying an average of 216 (R0.6C) or 291 (6C) copies of the antigens. Both R0.6C and 6C VLP conjugates were strongly reactive with a monoclonal antibody (mAb45.1) targeting a conformational TB Pfs48/45 epitope, suggesting that the TB epitope is accessible for immune recognition on the particles. To select the most suitable vaccine formulation for downstream clinical studies the two VLP vaccines were tested in CD1 mice using different adjuvant formulations. The study demonstrates that VLP-display of R0.6C and 6C significantly increases antigen immunogenicity when using Montanide ISA 720 VG as extrinsic adjuvant.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Peptídeos/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Adjuvantes Imunológicos , Animais , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Feminino , Imunogenicidade da Vacina , Malária Falciparum/transmissão , Camundongos , Peptídeos/química , Peptídeos/genética , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Proteínas Recombinantes/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Vacinas de Partículas Semelhantes a Vírus/química
13.
Vaccine ; 35(24): 3171-3177, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28483194

RESUMO

A CD1d-binding, invariant (i) natural killer T (NKT)-cell stimulatory glycolipid, α-Galactosylceramide (αGalCer), has been shown to act as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying a higher binding affinity for CD1d molecule and more potent adjuvant activity than αGalCer. In the present study, 7DW8-5 co-administered intramuscularly (i.m.) with a recombinant adenovirus expressing a Plasmodium yoelii circumsporozoite protein (PyCSP), AdPyCS, has led to a co-localization of 7DW8-5 and a PyCSP in draining lymph nodes (dLNs), particularly in dendritic cells (DCs). This occurrence initiates a cascade of events, such as the recruitment of DCs to dLNs and their activation and maturation, and the enhancement of the ability of DCs to prime CD8+ T cells induced by AdPyCS and ultimately leading to a potent adjuvant effect and protection against malaria.


Assuntos
Adenoviridae/genética , Adjuvantes Imunológicos , Antígenos CD1d/imunologia , Galactosilceramidas/imunologia , Vacinas Antimaláricas/imunologia , Animais , Antígenos CD1d/metabolismo , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Galactosilceramidas/química , Galactosilceramidas/metabolismo , Imunogenicidade da Vacina , Injeções Intramusculares , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Malária/imunologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Camundongos , Células T Matadoras Naturais/imunologia , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Vacinas Sintéticas/imunologia
14.
Clin Vaccine Immunol ; 24(4)2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28179403

RESUMO

Four different vaccine platforms, each targeting the human malaria parasite Plasmodium vivax cell-traversal protein for ookinetes and sporozoites (PvCelTOS), were generated and assessed for protective efficacy. These platforms consisted of a recombinant chimpanzee adenoviral vector 63 (ChAd63) expressing PvCelTOS (Ad), a recombinant modified vaccinia virus Ankara expressing PvCelTOS (MVA), PvCelTOS conjugated to bacteriophage Qß virus-like particles (VLPs), and a recombinant PvCelTOS protein expressed in eukaryotic HEK293T cells (protein). Inbred BALB/c mice and outbred CD-1 mice were immunized using the following prime-boost regimens: Ad-MVA, Ad-VLPs, and Ad-protein. Protective efficacy against sporozoite challenge was assessed after immunization using a novel chimeric rodent Plasmodium berghei parasite (Pb-PvCelTOS). This chimeric parasite expresses P. vivax CelTOS in place of the endogenous P. berghei CelTOS and produces fully infectious sporozoites. A single Ad immunization in BALB/c and CD-1 mice induced anti-PvCelTOS antibodies which were boosted efficiently using MVA, VLP, or protein immunization. PvCelTOS-specific gamma interferon- and tumor necrosis factor alpha-producing CD8+ T cells were induced at high frequencies by all prime-boost regimens in BALB/c mice but not in CD-1 mice; in CD-1 mice, they were only marginally increased after boosting with MVA. Despite the induction of anti-PvCelTOS antibodies and PvCelTOS-specific CD8+ T-cell responses, only low levels of protective efficacy against challenge with Pb-PvCelTOS sporozoites were obtained using any immunization strategy. In BALB/c mice, no immunization regimens provided significant protection against a Pb-PvCelTOS chimeric sporozoite challenge. In CD-1 mice, modest protective efficacy against challenge with chimeric P. berghei sporozoites expressing either PvCelTOS or P. falciparum CelTOS was observed using the Ad-protein vaccination regimen.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Portadores de Fármacos , Feminino , Interferon gama/metabolismo , Vacinas Antimaláricas/administração & dosagem , Camundongos , Fator de Necrose Tumoral alfa/metabolismo , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
15.
Parasitol Int ; 66(3): 324-330, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28110081

RESUMO

Humoral and cellular immune responses play an important role during Giardia lamblia infection. Several Giardia proteins have been identified as immunogenic antigens based on their elicited humoral immune response. Poorly is known about Giardia antigens that stimulate a cellular immune response. The main purpose of this study was to isolate and partial characterize an immunogenic antigen (5G8) of G. lamblia. The 5G8 protein was isolated from G. lamblia trophozoite lysates by affinity chromatography using moAb 5G8-coupled CNBr-Sepharose. The isolated protein was analysed by electrospray tandem mass spectrometry (ESI-MS/MS), and by diverse bioinformatics tools (GiardiaDB, BLASTn, BLASTp and ExPASy). Additionally, several biochemical and immunological characteristics of the isolated protein were analysed. By ESI-MS/MS the amino acidic 5G8 sequence was deduced. The 5G8 antigen belongs to the VSP family proteins of G. lamblia. This protein is composed by one polypeptide chain (±71kDa). Using the algorithm SYFPHEITI, we identified candidate CD4+ T-cell epitopes from the 5G8 antigen, which can elicit cell-mediated immune responses. In this study, we have identified a G. lamblia protein that induces a strong immune response in infected mice. The biochemical and immunological characterization of the immunogenic 5G8 antigen may contribute to the rational design of a Giardia vaccine.


Assuntos
Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Giardia lamblia/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Antígenos de Protozoários/isolamento & purificação , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/isolamento & purificação , Epitopos de Linfócito T/química , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/isolamento & purificação , Imunidade Celular , Imunidade Humoral , Proteínas de Membrana/genética , Camundongos , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/isolamento & purificação , Proteínas de Protozoários/metabolismo , Espectrometria de Massas em Tandem , Trofozoítos/imunologia
16.
Malar J ; 15: 161, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26979066

RESUMO

BACKGROUND: The pathogenesis of malaria is primarily associated with blood-stage infection and there is strong evidence that antibodies specific for parasite blood-stage antigens can control parasitaemia. This provides a strong rationale for incorporation of asexual blood-stage antigen components into an effective multivalent malaria subunit vaccine. On the basis of available genome-wide transcriptomic and proteomic data, previously uncharacterized Plasmodium falciparum open reading frames were screened for new blood stage vaccine candidates. This has led to the identification of the cysteine-rich protective antigen (PfCyRPA), which forms together with PfRH5 and PfRipr a multiprotein complex that is crucial for erythrocyte invasion. METHODS: Glycosylated and non-glycosylated variants of recombinant PfCyRPA were expressed and produced as secreted protein in mammalian cells. Adjuvanted formulations of purified PfCyRPA were tested to assess whether they can effectively elicit parasite inhibitory antibodies, and to investigate whether or not the glycosylation status affects antibody binding. For this purpose, two sets of PfCyRPA-specific mouse monoclonal antibodies (mAbs) have been raised and evaluated for functional activity. RESULTS: Generated PfCyRPA-specific mAbs, irrespective of the immunogen's glycosylation status, showed substantial parasite in vitro growth-inhibitory activity due to inhibition of erythrocyte invasion by merozoites. Furthermore, passive immunization experiments in P. falciparum infected NOD-scid IL2Rγ (null) mice engrafted with human erythrocytes demonstrated potent in vivo growth-inhibitory activity of generated mAbs. CONCLUSIONS: Recombinantly expressed PfCyRPA tested as adjuvanted vaccine formulations in mice elicited antibodies that significantly inhibit P. falciparum asexual blood stage parasite growth both in vitro and in vivo. These findings render PfCyRPA a promising blood-stage candidate antigen for inclusion into a multicomponent malaria subunit vaccine.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Monoclonais/isolamento & purificação , Anticorpos Antiprotozoários/isolamento & purificação , Antígenos de Protozoários/administração & dosagem , Vacinas Antimaláricas/administração & dosagem , Camundongos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
17.
Exp Parasitol ; 163: 46-56, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26836446

RESUMO

Toxoplasma gondii infects animals habiting terrestrial and aquatic environments. Its oocysts and tissue cysts are important for the horizontal transmission of this parasite. The oocyst and tissue cyst walls are crucial for the ability of the parasite to persist in the environment or in animal tissues, respectively. However, the composition of these walls is not well understood. We report the generation of monoclonal antibodies directed against wall components using mice immunized with oocyst antigens of T. gondii. One monoclonal antibody (mAb) G1/19 reacted solely with T. gondii sporozoites. The respective antigen had a relative molecular weight (Mr) of 30 kDa. MAb G1/19 failed to react with sporozoites of any other coccidian parasite species tested (Hammondia hammondi, Hammondia heydorni, Cystoisospora felis, Eimeria bovis, Sarcocystis sp.). Another mAb, designated K8/15-15, recognized antigens in sporocyst walls of the parasite and in the walls of in vivo or in vitro produced tissue cysts, as demonstrated by immunofluorescence and immunoblot assays. Antigens of 80 to a high molecular weight protein of about 350 kDa Mr were recognized by this antibody using antigen extracts from sporocysts, and from in vitro or in vivo generated tissue cysts of the parasite. Tissue cyst and sporocyst walls of H. hammondi and H. heydorni, and tissue cysts of Neospora caninum were also recognized by mAb K8/15-15. Sporocyst walls of C. felis also reacted to this mAb. The cyst walls of Sarcocystis sp. and Besnoitia besnoiti were not recognized by mAb K8/15-15. Reactivity by a single mAb against T. gondii antigens in tissue cysts and sporocysts had not been reported previously. MAb K8/15-15 may be a practical tool for the identification of both cysts and sporocysts of the parasite, and may also be potentially employed in proteomic studies on the identification of new components of the cyst and sporocyst walls of T. gondii.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos de Protozoários/imunologia , Imunoglobulina G/imunologia , Toxoplasma/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Antígenos de Protozoários/administração & dosagem , Gatos , Bovinos , Coccídios/classificação , Coccídios/imunologia , Cães , Imunofluorescência , Hibridomas , Imunização Secundária , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Oocistos/imunologia , Ovinos
18.
Parasit Vectors ; 8: 498, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26420606

RESUMO

BACKGROUND: Toxoplasma gondii is a widely prevalent intracellular parasite which infects almost all warm-blooded animals including humans and causes serious zoonotic toxoplasmosis. DNA vaccines have proved effective in the protection against parasites. However, the problems of weak immunity and inefficient delivery of DNA vaccine remain major issues. Therefore, comprehensive antigens derived from all stages of the parasite, effective adjuvants and delivery systems should be considered in the vaccine construction. METHODS: SAG3101-144,ROP18347-396, MIC6288-347, GRA7182-224, MAG158-125, BAG1156-211 and SPA142-200, derived from antigens in tachyzoite, bradyzoite and sporozoite stages of T. gondii were screened based on CD8(+) T cell epitope binding affinity to HLA and H-2. We constructed a recombinant DNA vaccine and an adenovirus vaccine encoding multi-stage antigen of T. gondii linked to ubiquitin molecules and vaccinated BALB/c mice with different strategies. Antibodies, cytokines, splenocytes proliferation, as well as the percentage of CD4(+) and CD8(+) T cells in immunized mouse were analyzed by the Enzyme-Linked Immunosorbent Assays (ELISA), Flow Cytometry (FCM). Protective efficacy was evaluated by challenging immunized mice with type I and type II parasite. RESULTS: Our results indicated that the DNA vaccine had the advantage of inducing a stronger humoral response, whereas the adenovirus-vectored vaccine effectively improved the cellular immune response. Priming with DNA vaccine and boosting with adenovirus-vectored vaccine induced Th1-type immune responses with highest levels of IgG2a and secretion of cytokines IL-2 and IFN-γ. Effective protection against type I and type II parasite with an increase in survival rate and a decrease in brain cyst burden was achieved in immunized mice. CONCLUSIONS: Priming vaccination with DNA vaccine and boosting with the recombinant adenovirus vaccine encoding ubiquitin conjugated multi-stage antigens of T. gondii was proved to be a potential strategy against the infection of type I and type II parasite.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Protozoárias/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Ubiquitina/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Linfócitos T CD8-Positivos/imunologia , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Vacinas Protozoárias/administração & dosagem , Vacinas Protozoárias/genética , Toxoplasma/genética , Toxoplasmose/parasitologia , Toxoplasmose/prevenção & controle , Ubiquitina/administração & dosagem , Ubiquitina/genética , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética , Vacinas de DNA/imunologia
19.
Infect Immun ; 83(12): 4476-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26351281

RESUMO

CD11b(+) Gr1(+) myeloid-derived suppressor cells (MDSCs), a heterogeneous population of precursor cells, modulate protective immunity against visceral leishmaniasis by suppressing T cell functions. We observed that CD11b(+) Gr1(+) MDSCs, which initially expanded in soluble leishmanial antigen (SLA)-immunized mice and later diminished, suppressed proliferation of T cells isolated from SLA-immunized mice, but to a lesser extent than the case in naive mice. This lesser suppression of MDSCs accompanied the expression of F4/80 and the production of Cox-2, arginase I, nitric oxide, and PGE2. However, with SLA immunization, there was no difference in the expression of interleukin-2 (IL-2) or gamma interferon (IFN-γ) by T cells, in contrast to the case in nonimmunized mice, in which there is an influence. Glycyrrhizic acid (a triterpenoid compound)-mediated inhibition of Cox-2 in myeloid-derived suppressor cells influenced the capacity of T cells to proliferate and the expression of IL-2 and IFN-γ in Leishmania donovani-infected BALB/c mice. Further characterization confirmed that administration of glycyrrhizic acid to L. donovani-infected BALB/c mice results in an impairment of the generation of MDSCs and a reciprocal organ-specific proliferation of IFN-γ- and IL-10-expressing CD4(+) and CD8(+) T cells. Comprehensive knowledge on the Cox-2-mediated regulation of myeloid-derived suppressor cells might be involved in unlocking a new avenue for therapeutic interventions during visceral leishmaniasis.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Ácido Glicirrízico/farmacologia , Fatores Imunológicos/farmacologia , Leishmania donovani/efeitos dos fármacos , Leishmaniose Visceral/tratamento farmacológico , Células Mieloides/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/imunologia , Antígenos de Protozoários/administração & dosagem , Arginase/genética , Arginase/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/patologia , Proliferação de Células , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/antagonistas & inibidores , Dinoprostona/biossíntese , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Imunização , Interferon gama/genética , Interferon gama/imunologia , Interleucina-2/genética , Interleucina-2/imunologia , Leishmania donovani/crescimento & desenvolvimento , Leishmania donovani/imunologia , Leishmania donovani/patogenicidade , Leishmaniose Visceral/imunologia , Leishmaniose Visceral/microbiologia , Leishmaniose Visceral/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Células Mieloides/microbiologia , Células Mieloides/patologia , Óxido Nítrico/antagonistas & inibidores , Óxido Nítrico/biossíntese , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia , Linfócitos T Reguladores/patologia
20.
Parasitol Res ; 114(11): 4195-203, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26243574

RESUMO

Toxoplasma gondii infection in humans and animals is a worldwide zoonosis. Prevention and control of toxoplasmosis based on vaccination is one of the promising strategies. In the present study, recombinant T. gondii rhoptry proteins 38 and 18 (TgROP38 and TgROP18) were encapsulated into poly (lactide-co-glycolide) (PLG) (1:1), respectively, to obtain the stable water-in-oil-in-water double emulsion. Female Kunming mice were then immunized with the protein vaccines twice at a 2-week interval. Eight weeks after the second immunization, 10 mice from each group were challenged with T. gondii PRU strain (genotype II). The entrapment rates of PLG-rROP38 and PLG-rROP18 ranged from 65.5 to 77.7% and 58.1 to 72.3%, respectively. Immunization of mice with rROP38 and rROP18 proteins encapsulated into PLG microparticles elicited strongly humoral and cell-mediated responses against T. gondii, associated with relatively high levels of total IgG, IgG2a isotype, and IFN-γ, as well as the mixed Th1/Th2 immunity responses. Immunization with various protein vaccines induced significant reduction of the brain cysts after chronic infection with the T. gondii PRU strain, and the most effective protection was achieved in the PLG-rROP38-rROP18-immunized mice, with a cyst reduction of 81.3%. The findings of the present study indicated that recombinant rhoptry antigens encapsulated in PLG could maintain the protein immunogenicity in an extended period and elicit effective protection against chronic T. gondii infection, which has implications for the development of long-lasting vaccines against chronic toxoplasmosis in animals.


Assuntos
Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/imunologia , Proteínas de Protozoários/administração & dosagem , Vacinas Protozoárias/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Portadores de Fármacos/administração & dosagem , Feminino , Humanos , Imunização , Camundongos , Modelos Animais , Poliésteres/administração & dosagem , Proteínas de Protozoários/imunologia , Proteínas Recombinantes , Toxoplasmose Animal/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA