Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Rev. bras. parasitol. vet ; 31(3): e009322, 2022. tab
Artículo en Inglés | LILACS, VETINDEX | ID: biblio-1394894

RESUMEN

Abstract The seroprevalence of Sarcocystis spp. and Toxoplasma gondii was researched in swine raised in Santa Maria, RS, Brazil. Serum samples from 84 pigs from 31 farms were tested using indirect immunofluorescence assay (IFA) for both agents. Additionally, 53 samples of pork sausages and tissues destined for human consumption, including: salami, sausage, black pudding, heart, tongue, brain, and rib muscle, were submitted to PCR to detect DNA for each agent. The frequency of anti-Sarcocystis spp. antibodies was 36.9% (31/84), with titers ranging from 32 to 1024, and 25% (21/84) for anti-T. gondii antibodies, with titers ranging from 64 to 2048. Sarcocystis spp. and T. gondii DNA were detected in 67.9% (36/53) and 13.2% (7/53) of samples, respectively. The presence of antibodies and the detection of DNA from Sarcocystis spp., and T. gondii suggests that the pigs were infected and may serve as an important reservoir for both parasites. The infection by these protozoa in the swine population is relevant to public health due to their zoonotic potential.


Resumo A soroprevalência de Sarcocystis spp. e Toxoplasma gondii foi pesquisada em suínos criados em Santa Maria, RS, Brasil. Amostras de soro de 84 suínos de 31 fazendas foram testadas pela reação deimunofluorescência indireta (IFA) para ambos os agentes. Adicionalmente, 53 amostras de embutidos suínos e tecidos cárneos destinados ao consumo humano, incluindo: salame, linguiça, morcela, coração, língua, cérebro e músculo da costela foram submetidas à PCR para detecção de DNA para cada agente. A frequência de anticorpos anti-Sarcocystis spp. foi de 36,9% (31/84), com títulos variando de 32 a 1.024; e 25% (21/84) para anticorpos anti-T. gondii, com títulos variando de 64 a 2048. A presença de DNA de Sarcocystis spp. e T. gondii foi detectada em 67,9% (36/53) e 13,2% (7/53) das amostras avaliadas, respectivamente. A detecção de anticorpos e DNA de Sarcocystis spp. e T. gondii sugere que os suínos foram infectados e podem servir como um importante reservatório de ambos os parasitas. A circulação desses agentes na população suína é relevante para a saúde pública devido ao seu potencial zoonótico.


Asunto(s)
Humanos , Animales , Enfermedades de los Porcinos/diagnóstico , Enfermedades de los Porcinos/parasitología , Toxoplasmosis Animal/diagnóstico , Sarcocistosis/diagnóstico , Sarcocistosis/veterinaria , Porcinos/parasitología , Enfermedades de los Porcinos/epidemiología , Toxoplasma/genética , Toxoplasma/inmunología , Anticuerpos Antiprotozoarios/análisis , Estudios Seroepidemiológicos , Toxoplasmosis Animal/epidemiología , Prevalencia , ADN Protozoario/inmunología , Sarcocystis/genética , Sarcocystis/inmunología , Sarcocistosis/epidemiología , Carne de Cerdo/parasitología
2.
PLoS One ; 16(9): e0256980, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34495988

RESUMEN

BACKGROUND: A DNA-prime/human adenovirus serotype 5 (HuAd5) boost vaccine encoding Plasmodium falciparum (Pf) circumsporozoite protein (PfCSP) and Pf apical membrane antigen-1 (PfAMA1), elicited protection in 4/15 (27%) of subjects against controlled human malaria infection (CHMI) that was statistically associated with CD8+ T cell responses. Subjects with high level pre-existing immunity to HuAd5 were not protected, suggesting an adverse effect on vaccine efficacy (VE). We replaced HuAd5 with chimpanzee adenovirus 63 (ChAd63), and repeated the study, assessing both the two-antigen (CSP, AMA1 = CA) vaccine, and a novel three-antigen (CSP, AMA1, ME-TRAP = CAT) vaccine that included a third pre-erythrocytic stage antigen [malaria multiple epitopes (ME) fused to the Pf thrombospondin-related adhesive protein (TRAP)] to potentially enhance protection. METHODOLOGY: This was an open label, randomized Phase 1 trial, assessing safety, tolerability, and VE against CHMI in healthy, malaria naïve adults. Forty subjects (20 each group) were to receive three monthly CA or CAT DNA priming immunizations, followed by corresponding ChAd63 boost four months later. Four weeks after the boost, immunized subjects and 12 infectivity controls underwent CHMI by mosquito bite using the Pf3D7 strain. VE was assessed by determining the differences in time to parasitemia as detected by thick blood smears up to 28-days post CHMI and utilizing the log rank test, and by calculating the risk ratio of each treatment group and subtracting from 1, with significance calculated by the Cochran-Mantel-Haenszel method. RESULTS: In both groups, systemic adverse events (AEs) were significantly higher after the ChAd63 boost than DNA immunizations. Eleven of 12 infectivity controls developed parasitemia (mean 11.7 days). In the CA group, 15 of 16 (93.8%) immunized subjects developed parasitemia (mean 12.0 days). In the CAT group, 11 of 16 (63.8%) immunized subjects developed parasitemia (mean 13.0 days), indicating significant protection by log rank test compared to infectivity controls (p = 0.0406) and the CA group (p = 0.0229). VE (1 minus the risk ratio) in the CAT group was 25% compared to -2% in the CA group. The CA and CAT vaccines induced robust humoral (ELISA antibodies against CSP, AMA1 and TRAP, and IFA responses against sporozoites and Pf3D7 blood stages), and cellular responses (IFN-γ FluoroSpot responses to CSP, AMA1 and TRAP) that were not associated with protection. CONCLUSIONS: This study demonstrated that the ChAd63 CAT vaccine exhibited significant protective efficacy, and confirmed protection was afforded by adding a third antigen (T) to a two-antigen (CA) formulation to achieve increased VE. Although the ChAd63-CAT vaccine was associated with increased frequencies of systemic AEs compared to the CA vaccine and, historically, compared to the HuAd5 vectored malaria vaccine encoding CSP and AMA1, they were transient and associated with increased vector dosing.


Asunto(s)
Vacunas contra el Adenovirus/inmunología , Adenovirus de los Simios/inmunología , Antígenos de Protozoos/inmunología , ADN Protozoario/inmunología , ADN Recombinante/inmunología , Inmunización Secundaria/métodos , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Vacunas de ADN/inmunología , Vacunas contra el Adenovirus/administración & dosificación , Vacunas contra el Adenovirus/efectos adversos , Adenovirus de los Simios/genética , Adulto , Antígenos de Protozoos/genética , Linfocitos T CD8-positivos/inmunología , ADN Protozoario/genética , Epítopos/genética , Epítopos/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Voluntarios Sanos , Humanos , Inmunogenicidad Vacunal/inmunología , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/efectos adversos , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Masculino , Proteínas de la Membrana/genética , Proteínas Protozoarias/genética , Resultado del Tratamiento , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Adulto Joven
3.
PLoS Pathog ; 16(8): e1008772, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32866214

RESUMEN

The tick-borne apicomplexan parasite, Babesia bovis, a highly persistent bovine pathogen, expresses VESA1 proteins on the infected erythrocyte surface to mediate cytoadhesion. The cytoadhesion ligand, VESA1, which protects the parasite from splenic passage, is itself protected from a host immune response by rapid antigenic variation. B. bovis relies upon segmental gene conversion (SGC) as a major mechanism to vary VESA1 structure. Gene conversion has been considered a form of homologous recombination (HR), a process for which Rad51 proteins are considered pivotal components. This could make BbRad51 a choice target for development of inhibitors that both interfere with parasite genome integrity and disrupt HR-dependent antigenic variation. Previously, we knocked out the Bbrad51 gene from the B. bovis haploid genome, resulting in a phenotype of sensitivity to methylmethane sulfonate (MMS) and apparent loss of HR-dependent integration of exogenous DNA. In a further characterization of BbRad51, we demonstrate here that ΔBbrad51 parasites are not more sensitive than wild-type to DNA damage induced by γ-irradiation, and repair their genome with similar kinetics. To assess the need for BbRad51 in SGC, RT-PCR was used to observe alterations to a highly variant region of ves1α transcripts over time. Mapping of these amplicons to the genome revealed a significant reduction of in situ transcriptional switching (isTS) among ves loci, but not cessation. By combining existing pipelines for analysis of the amplicons, we demonstrate that SGC continues unabated in ΔBbrad51 parasites, albeit at an overall reduced rate, and a reduction in SGC tract lengths was observed. By contrast, no differences were observed in the lengths of homologous sequences at which recombination occurred. These results indicate that, whereas BbRad51 is not essential to babesial antigenic variation, it influences epigenetic control of ves loci, and its absence significantly reduces successful variation. These results necessitate a reconsideration of the likely enzymatic mechanism(s) underlying SGC and suggest the existence of additional targets for development of small molecule inhibitors.


Asunto(s)
Antígenos de Protozoos , Babesia bovis , Conversión Génica/inmunología , Genoma de Protozoos/inmunología , Proteínas Protozoarias , Recombinasa Rad51 , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Babesia bovis/genética , Babesia bovis/inmunología , ADN Protozoario/genética , ADN Protozoario/inmunología , Haploidia , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Recombinasa Rad51/genética , Recombinasa Rad51/inmunología
4.
Trends Parasitol ; 36(9): 773-784, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32736985

RESUMEN

Emerging evidence suggests that the DNA-sensing pathway plays a crucial role in innate immunity against multiple diseases, especially infectious diseases. Cyclic GMP-AMP synthase (cGAS), as a DNA sensor, and stimulator of interferon (IFN) genes (STING), as an adaptor protein, are the central components that link DNA sensing to immunologic functions - including, but not limited to, the type I IFN response. Recently, a series of studies have revealed that genomic DNA from protozoan parasites triggers the cGAS-STING pathway, and these studies identified the positive and negative regulators that modulate the signaling in parasite infection. Here, we summarize current understanding of the critical functions and potential applications of the cGAS-STING axis in parasitic diseases, specifically those caused by malaria parasites.


Asunto(s)
Interacciones Huésped-Parásitos/inmunología , Malaria/inmunología , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Nucleotidiltransferasas/metabolismo , Plasmodium/genética , Plasmodium/inmunología , Animales , ADN Protozoario/genética , ADN Protozoario/inmunología , Humanos , Transducción de Señal/genética
5.
Sci Rep ; 10(1): 7901, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404867

RESUMEN

Schistosomiasis is a human parasitic disease responsible for serious consequences for public health, as well as severe socioeconomic impacts in developing countries. Here, we provide evidence that the adaptor molecule STING plays an important role in Schistosoma mansoni infection. S. mansoni DNA is sensed by cGAS leading to STING activation in murine embryonic fibroblasts (MEFs). Sting-/- and C57BL/6 (WT) mice were infected with schistosome cercariae in order to assess parasite burden and liver pathology. Sting-/- mice showed worm burden reduction but no change in the number of eggs or granuloma numbers and area when compared to WT animals. Immunologically, a significant increase in IFN-γ production by the spleen cells was observed in Sting-/- animals. Surprisingly, Sting-/- mice presented an elevated percentage of neutrophils in lungs, bronchoalveolar lavage, and spleens. Moreover, Sting-/- neutrophils exhibited increased survival rate, but similar ability to kill schistosomula in vitro when stimulated with IFN-γ when compared to WT cells. Finally, microbiota composition was altered in Sting-/- mice, revealing a more inflammatory profile when compared to WT animals. In conclusion, this study demonstrates that STING signaling pathway is important for S. mansoni DNA sensing and the lack of this adaptor molecule leads to enhanced resistance to infection.


Asunto(s)
Interacciones Huésped-Patógeno , Proteínas de la Membrana/metabolismo , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/metabolismo , Esquistosomiasis mansoni/parasitología , Animales , ADN Protozoario/inmunología , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Interacciones Huésped-Patógeno/inmunología , Inmunidad Celular , Inmunidad Humoral , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Noqueados , Nucleotidiltransferasas/deficiencia , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos/inmunología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
6.
Parasitology ; 146(13): 1646-1654, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31452491

RESUMEN

Bovine babesiosis is the most important protozoan disease transmitted by ticks. In Plasmodium falciparum, another Apicomplexa protozoan, the interaction of rhoptry neck protein 2 (RON2) with apical membrane antigen-1 (AMA-1) has been described to have a key role in the invasion process. To date, RON2 has not been described in Babesia bigemina, the causal agent of bovine babesiosis in the Americas. In this work, we found a ron2 gene in the B. bigemina genome. RON2 encodes a protein that is 1351 amino acids long, has an identity of 64% (98% coverage) with RON2 of B. bovis and contains the CLAG domain, a conserved domain in Apicomplexa. B. bigemina ron2 is a single copy gene and it is transcribed and expressed in blood stages as determined by RT-PCR, Western blot, and confocal microscopy. Serum samples from B. bigemina-infected bovines were screened for the presence of RON2-specific antibodies, showing the recognition of conserved B-cell epitopes. Importantly, in vitro neutralization assays showed an inhibitory effect of RON2-specific antibodies on the red blood cell invasion by B. bigemina. Therefore, RON2 is a novel antigen in B. bigemina and contains conserved B-cell epitopes, which induce antibodies that inhibit merozoite invasion.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Babesia/genética , Epítopos de Linfocito B/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Animales , Babesia/inmunología , Babesiosis/parasitología , Bovinos , ADN Protozoario/inmunología , Eritrocitos/parasitología , Genoma de Protozoos , Masculino , Merozoítos/genética , Merozoítos/inmunología , Pruebas de Neutralización
7.
Sci Rep ; 9(1): 9825, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31285443

RESUMEN

Microbial pattern recognition critically contributes to innate response, both at extracellular and intracellular cytosolic surveillance pathway (CSP) interface. However, the role of pattern recognition by host innate receptors in CSP is poorly understood in Leishmania donovani infection. Here, we have demonstrated that cytosolic targeting of L.donovani DNA (Ld-DNA) inhibits macrophage responsiveness to IFNÉ£, through decreased MHC-II expression and lowered pSTAT1 (Y701) levels, involving host three-prime repair exonuclease-1 (TREX-1). The Ld-DNA potently induced type-1 IFNs, i.e. significant over-production of IFNß through activation of the IRF pathway. Interestingly, knockdown of TRIF or MyD88 expression in macrophages had no effect on cytosolic Ld-DNA transfection-mediated IFN-ß production, indicating involvement of a TLR independent pathway. Contrastingly, Ld-DNA failed to induce IFNß in both TBK-1 and IRF3KO knockout macrophages. Although IFNß was not induced by Ld-DNA in STING- knockout macrophages, STING alone was not enough for the induction. Evidently, besides STING, Ld-DNA recognition for induction of IFNß critically required cytosolic cyclic GMP-AMP synthase (cGAS). Furthermore, the cGAS dependent targeting of Ld-DNA induced IFNß over-production that contributed to antimony resistance in L.donovani infection. We provide the first evidence that enhanced cytosolic sensing of Ld-DNA in infection by antimony resistant (SBR-LD), but not antimony sensitive L.donovani strains (SBS-LD), was critically regulated by host MDRs, multi drug resistant associated protein 1 (MRP 1) and permeability glycoprotein (P-gp) in macrophages. Collectively, our results disclose Ld-DNA as a vital pathogen associated molecular pattern (PAMP) driving host Type-I IFN responses and antimony resistance. The findings may help in future development of policies for novel anti-leishmanial therapeutics.


Asunto(s)
ADN Protozoario/inmunología , Interferón gamma/metabolismo , Leishmania donovani/inmunología , Leishmaniasis Visceral/parasitología , Animales , Citosol/inmunología , Citosol/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Leishmania donovani/genética , Leishmania donovani/crecimiento & desarrollo , Leishmaniasis Visceral/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Células RAW 264.7 , Células THP-1
8.
Free Radic Biol Med ; 131: 59-71, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30472364

RESUMEN

Uracil-DNA glycosylase (UNG) initiates the base excision repair pathway by excising uracil from DNA. We have previously shown that Trypanosoma brucei cells defective in UNG exhibit reduced infectivity thus demonstrating the relevance of this glycosylase for survival within the mammalian host. In the early steps of the immune response, nitric oxide (NO) is released by phagocytes, which in combination with oxygen radicals produce reactive nitrogen species (RNS). These species can react with DNA generating strand breaks and base modifications including deaminations. Since deaminated cytosines are the main substrate for UNG, we hypothesized that the glycosylase might confer protection towards nitrosative stress. Our work establishes the occurrence of genotoxic damage in Trypanosoma brucei upon exposure to NO in vitro and shows that deficient base excision repair results in increased levels of damage in DNA and a hypermutator phenotype. We also evaluate the incidence of DNA damage during infection in vivo and show that parasites recovered from mice exhibit higher levels of DNA strand breaks, base deamination and repair foci compared to cells cultured in vitro. Notably, the absence of UNG leads to reduced infectivity and enhanced DNA damage also in animal infections. By analysing mRNA and protein levels, we found that surviving UNG-KO trypanosomes highly express tryparedoxin peroxidase involved in trypanothione/tryparedoxin metabolism. These observations suggest that the immune response developed by the host enhances the activation of genes required to counteract oxidative stress and emphasize the importance of DNA repair pathways in the protection to genotoxic and oxidative stress in trypanosomes.


Asunto(s)
Reparación del ADN , ADN Protozoario/genética , Óxido Nítrico/farmacología , Proteínas Protozoarias/genética , Trypanosoma brucei brucei/genética , Uracil-ADN Glicosidasa/genética , Animales , Daño del ADN , ADN Protozoario/inmunología , Femenino , Expresión Génica , Genotipo , Glutatión/análogos & derivados , Glutatión/metabolismo , Interacciones Huésped-Parásitos , Macrófagos/inmunología , Macrófagos/parasitología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Estrés Nitrosativo/genética , Parasitemia/inmunología , Parasitemia/metabolismo , Parasitemia/parasitología , Peroxidasas/genética , Peroxidasas/metabolismo , Fenotipo , Proteínas Protozoarias/metabolismo , Espermidina/análogos & derivados , Espermidina/metabolismo , Tiorredoxinas/metabolismo , Trypanosoma brucei brucei/efectos de los fármacos , Trypanosoma brucei brucei/metabolismo , Trypanosoma brucei brucei/patogenicidad , Tripanosomiasis/inmunología , Tripanosomiasis/metabolismo , Tripanosomiasis/parasitología , Uracil-ADN Glicosidasa/deficiencia
9.
Front Immunol ; 9: 2888, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30581439

RESUMEN

While half of the world's population is at risk of malaria, the most vulnerable are still children under five, pregnant women and returning travelers. Anopheles mosquitoes transmit malaria parasites to the human host; but how Plasmodium interact with the innate immune system remains largely unexplored. The most recent advances prove that monocytes are a key component to control parasite burden and to protect host from disease. Monocytes' protective roles include phagocytosis, cytokine production and antigen presentation. However, monocytes can be involved in pathogenesis and drive inflammation and sequestration of infected red blood cells in organs such as the brain, placenta or lungs by secreting cytokines that upregulate expression of endothelial adhesion receptors. Plasmodium DNA, hemozoin or extracellular vesicles can impair the function of monocytes. With time, reinfections with Plasmodium change the relative proportion of monocyte subsets and their physical properties. These changes relate to clinical outcomes and might constitute informative biomarkers of immunity. More importantly, at the molecular level, transcriptional, metabolic or epigenetic changes can "prime" monocytes to alter their responses in future encounters with Plasmodium. This mechanism, known as trained immunity, challenges the traditional view of monocytes as a component of the immune system that lacks memory. Overall, this rough guide serves as an update reviewing the advances made during the past 5 years on understanding the role of monocytes in innate immunity to malaria.


Asunto(s)
Malaria/inmunología , Monocitos/inmunología , Plasmodium/inmunología , Anticuerpos Antiprotozoarios/inmunología , Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Citocinas/inmunología , Citocinas/metabolismo , ADN Protozoario/inmunología , Hemoproteínas/inmunología , Humanos , Malaria/parasitología , Monocitos/metabolismo , Fagocitosis/inmunología , Plasmodium/genética , Plasmodium/patogenicidad
10.
Vaccine ; 36(45): 6703-6710, 2018 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-30268732

RESUMEN

Cryptosporidium spp. is a zoonotic intracellular protozoan and a significant cause of diarrhoea in humans and animals worldwide. This parasite can cause high morbidity in immunocompromised people and children in developing countries, livestock being the main reservoir. This study was aimed at performing preliminary tests on Swiss albino weaned mice (ICR) to evaluate the humoral immune response induced against peptides derived from Cryptosporidium parvum CP15 (15 kDa sporozoite surface antigen) and CSL (circumsporozoite-like antigen) proteins. Peptides were identified and characterised using bioinformatics tools and were chemically synthesised. The antibody response was determined and the neutralising effect of antibodies was measured in cell culture. Despite all peptides studied here were capable of stimulating antibody production, neutralising antibodies were detected for just two of the CP15-derived ones. Additional studies aimed at evaluating further the potential of such peptides as vaccine candidates are thus recommended.


Asunto(s)
Antígenos de Protozoos/inmunología , Cryptosporidium parvum/inmunología , Vacunas Antiprotozoos/inmunología , Anticuerpos Antiprotozoarios/inmunología , ADN Protozoario/inmunología , Péptidos/síntesis química , Péptidos/química , Péptidos/inmunología , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología
11.
Korean J Parasitol ; 56(3): 237-245, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29996627

RESUMEN

Toxoplasma gondii can infect all the vertebrates including human, and leads to serious toxoplasmosis and considerable veterinary problems. T. gondii heat shock protein 60 (HSP60) is associated with the activation of antigen presenting cells by inducing initial immune responses and releasing inflammatory cytokines. It might be a potential DNA vaccine candidate for this parasite. A pVAX-HSP60 DNA vaccine was constructed and immune responses was evaluated in Kunming mice in this study. Our data indicated that the innate and adaptive immune responses was elicited by successive immunizations with pVAX-HSP60 DNA, showing apparent increases of CD3e+CD4+ and CD3e+CD8a+ T cells in spleen tissues of the HSP60 DNA-immunized mice (24.70±1.23% and 10.90±0.89%, P<0.05) and higher levels of specific antibodies in sera. Furthermore, the survival period of the immunized mice (10.53±4.78 day) were significantly prolonged during the acute T. gondii infection. Decrease of brain cysts was significant in the experimental group during the chronic infection (P<0.01). Taken together, TgHSP60 DNA can be as a vaccine candidate to prevent the acute and chronic T. gondii infections.


Asunto(s)
Chaperonina 60/inmunología , ADN Protozoario/inmunología , Ratones/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Toxoplasmosis/parasitología , Vacunas de ADN/inmunología , Enfermedad Aguda , Animales , Células Presentadoras de Antígenos/inmunología , Enfermedad Crónica , Citocinas/metabolismo , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Bazo/inmunología , Toxoplasmosis/prevención & control
12.
Recent Pat Biotechnol ; 12(1): 21-32, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28494723

RESUMEN

BACKGROUND: Leishmaniasis is caused by parasites of the genus Leishmania, and represents a group of chronic diseases with an epidemiological and clinical diversity. The disease is endemic in tropical regions, being found in 98 countries, affecting around 12 million people, with an estimated increase of 1.5 million per year. METHODS: The present review aims to analyze recent and most important patents regarding development of vaccines to improve immunization against leishmaniasis. For this purpose, the Web of Science - Derwent Innovations Index was consulted. There is also a short description of the licensed vaccines already on the market for commercialization, and a critical opinion on future developments. RESULTS: The data herein presented comprises national and international filings, thus considering the patent's country of origin, and can be used an indicator of a country's technological development regarding a specific field. Several types of vaccines against Leishmania were studied. The main classes comprise: vaccines using live cells (virulent or attenuated); dead cells; containing recombinant protein; using DNA of the parasite. United States (74 patents) leads the ranking of patent applications for vaccines against Leishmania, followed by Brazil (36 patents), which is an endemic region of leishmaniasis with 20,000 human cases of cutaneous leishmaniasis and over 3,000 cases of visceral form. CONCLUSION: This review showed that there is still a lot of space for development regarding the creation of a feasible, effective vaccine against leishmaniasis. The scientific community appears to be taking steps in the right direction, though.


Asunto(s)
Invenciones/estadística & datos numéricos , Vacunas contra la Leishmaniasis/biosíntesis , Leishmaniasis Cutánea/prevención & control , Leishmaniasis Visceral/prevención & control , Vacunas de ADN/biosíntesis , ADN Protozoario/inmunología , Humanos , Leishmania/inmunología , Leishmania/patogenicidad , Vacunas contra la Leishmaniasis/inmunología , Leishmaniasis Cutánea/inmunología , Leishmaniasis Cutánea/parasitología , Leishmaniasis Visceral/inmunología , Leishmaniasis Visceral/parasitología , Patentes como Asunto , Vacunas de ADN/inmunología , Vacunas Vivas no Atenuadas
13.
Nat Commun ; 8(1): 1985, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29215015

RESUMEN

STING is an innate immune cytosolic adaptor for DNA sensors that engage malaria parasite (Plasmodium falciparum) or other pathogen DNA. As P. falciparum infects red blood cells and not leukocytes, how parasite DNA reaches such host cytosolic DNA sensors in immune cells is unclear. Here we show that malaria parasites inside red blood cells can engage host cytosolic innate immune cell receptors from a distance by secreting extracellular vesicles (EV) containing parasitic small RNA and genomic DNA. Upon internalization of DNA-harboring EVs by human monocytes, P. falciparum DNA is released within the host cell cytosol, leading to STING-dependent DNA sensing. STING subsequently activates the kinase TBK1, which phosphorylates the transcription factor IRF3, causing IRF3 to translocate to the nucleus and induce STING-dependent gene expression. This DNA-sensing pathway may be an important decoy mechanism to promote P. falciparum virulence and thereby may affect future strategies to treat malaria.


Asunto(s)
Citosol/inmunología , ADN Protozoario/inmunología , Vesículas Extracelulares/inmunología , Malaria Falciparum/inmunología , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Línea Celular , Núcleo Celular/metabolismo , Microscopía por Crioelectrón , Citosol/metabolismo , ADN Protozoario/metabolismo , Eritrocitos , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/ultraestructura , Humanos , Inmunidad Innata , Factor 3 Regulador del Interferón/inmunología , Factor 3 Regulador del Interferón/metabolismo , Malaria Falciparum/parasitología , Proteínas de la Membrana/metabolismo , Monocitos , Fosforilación , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Protozoario/inmunología , ARN Protozoario/metabolismo , Transducción de Señal
14.
Nat Commun ; 8(1): 1282, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29101363

RESUMEN

Infectious pathogens contribute to the development of autoimmune disorders, but the mechanisms connecting these processes are incompletely understood. Here we show that Plasmodium DNA induces autoreactive responses against erythrocytes by activating a population of B cells expressing CD11c and the transcription factor T-bet, which become major producers of autoantibodies that promote malarial anaemia. Additionally, we identify parasite DNA-sensing through Toll-like receptor 9 (TLR9) along with inflammatory cytokine receptor IFN-γ receptor (IFN-γR) as essential signals that synergize to promote the development and appearance of these autoreactive T-bet+ B cells. The lack of any of these signals ameliorates malarial anaemia during infection in a mouse model. We also identify both expansion of T-bet+ B cells and production of anti-erythrocyte antibodies in ex vivo cultures of naive human peripheral blood mononuclear cells (PBMC) exposed to P. falciprum infected erythrocyte lysates. We propose that synergistic TLR9/IFN-γR activation of T-bet+ B cells is a mechanism underlying infection-induced autoimmune-like responses.


Asunto(s)
Anemia Hemolítica Autoinmune/etiología , Anemia Hemolítica Autoinmune/inmunología , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/parasitología , ADN Protozoario/inmunología , Malaria Falciparum/complicaciones , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Receptor Toll-Like 9/metabolismo , Anemia Hemolítica Autoinmune/parasitología , Animales , Autoanticuerpos/biosíntesis , Eritrocitos/inmunología , Eritrocitos/parasitología , Femenino , Humanos , Activación de Linfocitos , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium falciparum/patogenicidad , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Receptor Toll-Like 9/deficiencia , Receptor Toll-Like 9/genética , Receptor de Interferón gamma
15.
Korean J Parasitol ; 55(5): 505-512, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29103265

RESUMEN

Toxoplasma gondii cathepsin C proteases (TgCPC1, 2, and 3) are important for the growth and survival of T. gondii. In the present study, B-cell and T-cell epitopes of TgCPC1 were predicted using DNAstar and the Immune Epitope Database. A TgCPC1 DNA vaccine was constructed, and its ability to induce protective immune responses against toxoplasmosis in BALB/c mice was evaluated in the presence or absence of the adjuvant α-GalCer. As results, TgCPC1 DNA vaccine with or without adjuvant α-GalCer showed higher levels of IgG and IgG2a in the serum, as well as IL-2 and IFN-γ in the spleen compared to controls (PBS, pEGFP-C1, and α-Galcer). Upon challenge infection with tachyzoites of T. gondii (RH), pCPC1/α-Galcer immunized mice showed the longest survival among all the groups. Mice vaccinated with DNA vaccine without adjuvant (pCPC1) showed better protective immunity compared to other controls (PBS, pEGFP-C1, and α-Galcer). These results indicate that a DNA vaccine encoding TgCPC1 is a potential vaccine candidate against toxoplasmosis.


Asunto(s)
Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/inmunología , ADN Protozoario/genética , ADN Protozoario/inmunología , Toxoplasma/genética , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología , Toxoplasmosis Animal/prevención & control , Vacunas de ADN/inmunología , Animales , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C
16.
Parasite ; 23: 4, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26842927

RESUMEN

Toxoplasma gondii is an obligate intracellular apicomplexan parasite that affects humans and various vertebrate livestock and causes serious economic losses. To develop an effective vaccine against T. gondii infection, we constructed a DNA vaccine encoding the T. gondii rhoptry protein 17 (TgROP17) and evaluated its immune protective efficacy against acute T. gondii infection in mice. The DNA vaccine (p3×Flag-CMV-14-ROP17) was intramuscularly injected to BALB/c mice and the immune responses of the vaccinated mice were determined. Compared to control mice treated with empty vector or PBS, mice immunized with the ROP17 vaccine showed a relatively high level of specific anti-T. gondii antibodies, and a mixed IgG1/IgG2a response with predominance of IgG2a production. The immunized mice also displayed a specific lymphocyte proliferative response, a Th1-type cellular immune response with production of IFN-γ and interleukin-2, and increased number of CD8(+) T cells. Immunization with the ROP17 DNA significantly prolonged the survival time (15.6 ± 5.4 days, P < 0.05) of mice after challenge infection with the virulent T. gondii RH strain (Type I), compared with the control groups which died within 8 days. Therefore, our data suggest that DNA vaccination with TgROP17 triggers significant humoral and cellular responses and induces effective protection in mice against acute T. gondii infection, indicating that TgROP17 is a promising vaccine candidate against acute toxoplasmosis.


Asunto(s)
ADN Protozoario/genética , Proteínas Protozoarias/inmunología , Toxoplasma/inmunología , Toxoplasmosis Animal/prevención & control , Vacunas de ADN/inmunología , Factores de Virulencia/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , ADN Protozoario/inmunología , ADN Recombinante/genética , ADN Recombinante/inmunología , Femenino , Inmunoglobulina G/sangre , Inyecciones Intramusculares , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/genética , Células TH1/inmunología , Toxoplasmosis Animal/inmunología , Vacunación , Vacunas Sintéticas/inmunología , Factores de Virulencia/genética
17.
mBio ; 6(6): e01605-15, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26578679

RESUMEN

UNLABELLED: High levels of circulating immunocomplexes (ICs) are found in patients with either infectious or sterile inflammation. We report that patients with either Plasmodium falciparum or Plasmodium vivax malaria have increased levels of circulating anti-DNA antibodies and ICs containing parasite DNA. Upon stimulation with malaria-induced ICs, monocytes express an NF-κB transcriptional signature. The main source of IC-induced proinflammatory cytokines (i.e., tumor necrosis factor alpha [TNF-α] and interleukin-1ß [IL-1ß])in peripheral blood mononuclear cells from acute malaria patients was found to be a CD14(+) CD16 (FcγRIIIA)(+) CD64 (FcγRI)(high) CD32 (FcγRIIB)(low) monocyte subset. Monocytes from convalescent patients were predominantly of the classical phenotype (CD14(+) CD16(-)) that produces high levels of IL-10 and lower levels of TNF-α and IL-1ß in response to ICs. Finally, we report a novel role for the proinflammatory activity of ICs by demonstrating their ability to induce inflammasome assembly and caspase-1 activation in human monocytes. These findings illuminate our understanding of the pathogenic role of ICs and monocyte subsets and may be relevant for future development of immunity-based interventions with broad applications to systemic inflammatory diseases. IMPORTANCE: Every year, there are approximately 200 million cases of Plasmodium falciparum and P. vivax malaria, resulting in nearly 1 million deaths, most of which are children. Decades of research on malaria pathogenesis have established that the clinical manifestations are often a consequence of the systemic inflammation elicited by the parasite. Recent studies indicate that parasite DNA is a main proinflammatory component during infection with different Plasmodium species. This finding resembles the mechanism of disease in systemic lupus erythematosus, where host DNA plays a central role in stimulating an inflammatory process and self-damaging reactions. In this study, we disclose the mechanism by which ICs containing Plasmodium DNA activate innate immune cells and consequently stimulate systemic inflammation during acute episodes of malaria. Our results further suggest that Toll-like receptors and inflammasomes have a central role in malaria pathogenesis and provide new insights toward developing novel therapeutic interventions for this devastating disease.


Asunto(s)
Complejo Antígeno-Anticuerpo/metabolismo , Citocinas/metabolismo , ADN Protozoario/inmunología , Inflamasomas/metabolismo , Malaria Falciparum/patología , Malaria Vivax/patología , Monocitos/metabolismo , Complejo Antígeno-Anticuerpo/sangre , Antígenos CD/análisis , Humanos , Inmunofenotipificación , Malaria Falciparum/inmunología , Malaria Vivax/inmunología , Monocitos/química , Multimerización de Proteína
18.
PLoS Pathog ; 11(5): e1004828, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25951312

RESUMEN

In this study, we evaluated the long-term efficacy of a two-component subunit vaccine against Trypanosoma cruzi infection. C57BL/6 mice were immunized with TcG2/TcG4 vaccine delivered by a DNA-prime/Protein-boost (D/P) approach and challenged with T. cruzi at 120 or 180 days post-vaccination (dpv). We examined whether vaccine-primed T cell immunity was capable of rapid expansion and intercepting the infecting T. cruzi. Our data showed that D/P vaccine elicited CD4+ (30-38%) and CD8+ (22-42%) T cells maintained an effector phenotype up to 180 dpv, and were capable of responding to antigenic stimulus or challenge infection by a rapid expansion (CD8>CD4) with type 1 cytokine (IFNγ+ and TFNα+) production and cytolytic T lymphocyte (CTL) activity. Subsequently, challenge infection at 120 or 180 dpv, resulted in 2-3-fold lower parasite burden in vaccinated mice than was noted in unvaccinated/infected mice. Co-delivery of IL-12- and GMCSF-encoding expression plasmids provided no significant benefits in enhancing the anti-parasite efficacy of the vaccine-induced T cell immunity. Booster immunization (bi) with recombinant TcG2/TcG4 proteins 3-months after primary vaccine enhanced the protective efficacy, evidenced by an enhanced expansion (1.2-2.8-fold increase) of parasite-specific, type 1 CD4+ and CD8+ T cells and a potent CTL response capable of providing significantly improved (3-4.5-fold) control of infecting T. cruzi. Further, CD8+T cells in vaccinated/bi mice were predominantly of central memory phenotype, and capable of responding to challenge infection 4-6-months post bi by a rapid expansion to a poly-functional effector phenotype, and providing a 1.5-2.3-fold reduction in tissue parasite replication. We conclude that the TcG2/TcG4 D/P vaccine provided long-term anti-T. cruzi T cell immunity, and bi would be an effective strategy to maintain or enhance the vaccine-induced protective immunity against T. cruzi infection and Chagas disease.


Asunto(s)
Enfermedad de Chagas/inmunología , ADN Protozoario/genética , Inmunización Secundaria/métodos , Linfocitos T/inmunología , Trypanosoma cruzi/inmunología , Animales , Enfermedad de Chagas/patología , Citocinas/inmunología , Citocinas/metabolismo , ADN Protozoario/inmunología , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Plásmidos/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología
19.
J Leukoc Biol ; 97(2): 363-78, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25473100

RESUMEN

Regulation of macrophage PCD plays an important role in pathogenesis of leishmaniasis. However, the precise involvement of any parasite molecule in this process remains uncertain. In the current study, in silico wide analysis demonstrated that genes in the Leishmania donovani genome are highly enriched for CpG motifs, with sequence frequency of 8.7%. Here, we show that unmethylated species-specific CpG motifs in LdDNA significantly (P = 0.01) delay macrophage PCD by endosomal interaction with TLR9 via the adaptor protein MyD88. Importantly, LdDNA triggered high levels of luciferase activity (P = 0.001) under NF-κB-dependent transcription in HEK-TLR9 cells. Furthermore, the activation of caspases in macrophages was inhibited (P = 0.001) in the presence of LdDNA. Notably, the delay of PCD was mediated by modulation of the antiapoptotic proteins, Mcl-1 and Bfl-1, and impairment of loss of Δψm in macrophages through the neutralization of oxidative and nitrosative stress. The inhibition of caspase activation and up-regulation of Mcl-1 by LdDNA were TLR9 dependent. Analysis of the targets of LdDNA identified an early activation of the TLR9-dependent PI3K/Akt and SFK pathways, which were required for the observation of the antiapoptotic effects in macrophages. Moreover, we demonstrate that LdDNA modulates the TLR9-IκB-α pathway by promoting the tyrosine phosphorylation of TLR9 and the TLR9-mediated recruitment of Syk kinase. The results have identified a novel, TLR9-dependent antiapoptotic function of LdDNA, which will provide new opportunities for discovering and evaluating molecular targets for drug and vaccine designing against VL.


Asunto(s)
Apoptosis/inmunología , Islas de CpG/inmunología , Metilación de ADN , ADN Protozoario/inmunología , Leishmania donovani/inmunología , Macrófagos/inmunología , Receptor Toll-Like 9/inmunología , Apoptosis/efectos de los fármacos , Caspasas/inmunología , ADN Protozoario/farmacología , Activación Enzimática/inmunología , Femenino , Células HEK293 , Humanos , Macrófagos/citología , Masculino , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Fosforilación/inmunología , Regulación hacia Arriba/inmunología
20.
PLoS Negl Trop Dis ; 8(11): e3308, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25392997

RESUMEN

DNA sequences purified from distinct organisms, e.g. non vertebrate versus vertebrate ones, were shown to differ in their TLR9 signalling properties especially when either mouse bone marrow-derived- or human dendritic cells (DCs) are probed as target cells. Here we found that the DC-targeting immunostimulatory property of Leishmania major DNA is shared by other Trypanosomatidae DNA, suggesting that this is a general trait of these eukaryotic single-celled parasites. We first documented, in vitro, that the low level of immunostimulatory activity by vertebrate DNA is not due to its limited access to DCs' TLR9. In addition, vertebrate DNA inhibits the activation induced by the parasite DNA. This inhibition could result from the presence of competing elements for TLR9 activation and suggests that DNA from different species can be discriminated by mouse and human DCs. Second, using computational analysis of genomic DNA sequences, it was possible to detect the presence of over-represented inhibitory and under-represented stimulatory sequences in the vertebrate genomes, whereas L. major genome displays the opposite trend. Interestingly, this contrasting features between L. major and vertebrate genomes in the frequency of these motifs are shared by other Trypanosomatidae genomes (Trypanosoma cruzi, brucei and vivax). We also addressed the possibility that proteins expressed in DCs could interact with DNA and promote TLR9 activation. We found that TLR9 is specifically activated with L. major HMGB1-bound DNA and that HMGB1 preferentially binds to L. major compared to mouse DNA. Our results highlight that both DNA sequence and vertebrate DNA-binding proteins, such as the mouse HMGB1, allow the TLR9-signaling to be initiated and achieved by Trypanosomatidae DNA.


Asunto(s)
ADN Protozoario/inmunología , Genoma de Protozoos/inmunología , Motivos de Nucleótidos , Receptor Toll-Like 9/inmunología , Trypanosomatina/genética , Trypanosomatina/inmunología , Animales , Células de la Médula Ósea , ADN/química , ADN/inmunología , ADN/metabolismo , ADN Protozoario/química , ADN Protozoario/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/parasitología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ovinos , Transducción de Señal/inmunología , Porcinos , Receptor Toll-Like 9/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...