Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Chemosphere ; 346: 140630, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37939926

RESUMO

Mercury (Hg) is a globally important pollutant that can negatively impact metabolic, endocrine and immune systems of marine biota. Seabirds are long-lived marine top predators and hence are at risk of bioaccumulating high Hg concentrations from their prey. Here, we measured blood total mercury (THg) concentrations and relationships with physiology and breeding parameters of breeding brown skuas (Stercorarius antarcticus) (n = 49 individuals) at Esperanza/Hope Bay, Antarctic Peninsula. Mean blood THg concentrations were similar in males and females despite the differences in body size and breeding roles, but differed between study years. Immune markers (hematocrit, Immunoglobulin Y [IgY] and albumin) were negatively correlated with blood THg concentrations, which likely indicates a disruptive effect of Hg on immunity. Alanine aminotransferase (GPT) activity, reflecting liver dysfunction, was positively associated with blood THg. Additionally, triacylglycerol and albumin differed between our study years, but did not correlate with Hg levels, and so were more likely to reflect changes in diet and nutritional status rather than Hg contamination. Egg volume correlated negatively with blood THg concentrations. Our study provides new insights into the sublethal effects of Hg contamination on immunity, liver function and breeding parameters in seabirds. In this Antarctic species, exposure to sublethal Hg concentrations reflects the short-term risks which could make individuals more susceptible to environmental stressors, including ongoing climatic changes.


Assuntos
Charadriiformes , Mercúrio , Humanos , Masculino , Animais , Feminino , Aves/metabolismo , Mercúrio/análise , Regiões Antárticas , Monitoramento Ambiental , Charadriiformes/metabolismo , Fígado/metabolismo , Imunocompetência , Albuminas/metabolismo
2.
Environ Pollut ; 297: 118841, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35026328

RESUMO

Antarctic marine ecosystems are often considered to be pristine environments, yet wildlife in the polar regions may still be exposed to high levels of environmental contaminants. Here, we measured total mercury (THg) concentrations in blood samples from adult brown skuas Stercorarius antarcticus lonnbergi (n = 82) from three breeding colonies south of the Antarctic Polar Front in the Southern Ocean (southwest Atlantic region): (i) Bahía Esperanza/Hope Bay, Antarctic Peninsula; (ii) Signy Island, South Orkney Islands; and, (iii) Bird Island, South Georgia. Blood THg concentrations increased from the Antarctic Peninsula towards the Antarctic Polar Front, such that Hg contamination was lowest at Bahía Esperanza/Hope Bay (mean ± SD, 0.95 ± 0.45 µg g-1 dw), intermediate at Signy Island (3.42 ± 2.29 µg g-1 dw) and highest at Bird Island (4.47 ± 1.10 µg g-1 dw). Blood THg concentrations also showed a weak positive correlation with δ15N values, likely reflecting the biomagnification process. Males had higher Hg burdens than females, which may reflect deposition of Hg into eggs by females or potentially differences in their trophic ecology. These data provide important insights into intraspecific variation in contamination and the geographic transfer of Hg to seabirds in the Southern Ocean.


Assuntos
Mercúrio , Animais , Regiões Antárticas , Ecossistema , Monitoramento Ambiental , Feminino , Masculino , Mercúrio/análise , Oceanos e Mares , Caracteres Sexuais
3.
Front Immunol ; 11: 428, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32231667

RESUMO

Gastropod Molluscs rely exclusively on the innate immune system to protect from pathogens, defending their embryos through maternally transferred effectors. In this regard, Pomacea snail eggs, in addition to immune defenses, have evolved the perivitellin-2 or PV2 combining two immune proteins into a neurotoxin: a lectin and a pore-forming protein from the Membrane Attack Complex/Perforin (MACPF) family. This binary structure resembles AB-toxins, a group of toxins otherwise restricted to bacteria and plants. Many of these are enterotoxins, leading us to explore this activity in PV2. Enterotoxins found in bacteria and plants act mainly as pore-forming toxins and toxic lectins, respectively. In animals, although both pore-forming proteins and lectins are ubiquitous, no enterotoxins have been reported. Considering that Pomacea snail eggs ingestion induce morpho-physiological changes in the intestinal mucosa of rodents and is cytotoxic to intestinal cells in culture, we seek for the factor causing these effects and identified PmPV2 from Pomacea maculata eggs. We characterized the enterotoxic activity of PmPV2 through in vitro and in vivo assays. We determined that it withstands the gastrointestinal environment and resisted a wide pH range and enzymatic proteolysis. After binding to Caco-2 cells it promoted changes in surface morphology and an increase in membrane roughness. It was also cytotoxic to both epithelial and immune cells from the digestive system of mammals. It induced enterocyte death by a lytic mechanism and disrupted enterocyte monolayers in a dose-dependent manner. Further, after oral administration to mice PmPV2 attached to enterocytes and induced large dose-dependent morphological changes on their small intestine mucosa, reducing the absorptive surface. Additionally, PmPV2 was detected in the Peyer's patches where it activated lymphoid follicles and triggered apoptosis. We also provide evidence that the toxin can traverse the intestinal barrier and induce oral adaptive immunity with evidence of circulating antibody response. As a whole, these results indicate that PmPV2 is a true enterotoxin, a role that has never been reported to lectins or perforin in animals. This extends by convergent evolution the presence of plant- and bacteria-like enterotoxins to animals, thus expanding the diversity of functions of MACPF proteins in nature.


Assuntos
Enterotoxinas/farmacologia , Imunidade Inata/imunologia , Mucosa Intestinal/efeitos dos fármacos , Venenos de Moluscos/farmacologia , Caramujos/imunologia , Animais , Complexo de Ataque à Membrana do Sistema Complemento , Camundongos , Óvulo/imunologia , Óvulo/metabolismo , Perforina/metabolismo
4.
Heliyon ; 6(12): e05821, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33426331

RESUMO

Zinc oxide nanoparticles were successfully synthesized under precipitation processes, using ZnSO4·7H2O as a Zn2+ precursor and K2CO3 used as a basic source, and hydrozincite was obtained as an intermediary, which was treated under two procedures; first procedure involved multiple stages to get final precipitated with NaOH, and in the second procedure the hydrozincite was straightforwardly dried at 220 °C. By both processes ZnO structures were obtained, which were turned into nanoparticles by a solvothermal treatment, for four hours in ethylene glycol at 200 °C. The final products for the first procedure was conglomerate of spherical nanoparticles with sizes ranged between 5-10 nm and dispersed ellipsoidal nanoparticles for the second procedure. Apart off the two procedures mentioned above, another synthesis was carried out with the same Zn2+ precursor but now using NaOH, and the solvothermal treatment produced ZnO mixed micro-structures which under ultrasonic cavitation disaggregated on mesoporous ZnO nanoplates of hexagonal shapes with nanopore sizes of approximately 0.35 nm. All ZnOs synthesized were structurally characterized with XRD, TEM and FT-IR techniques, and electronically with UV-Vis absorption and diffuse reflectance spectroscopies.

5.
Acta Crystallogr C Struct Chem ; 74(Pt 11): 1547-1552, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30398212

RESUMO

The reaction of 2-aminonicotinaldehyde with 2- or 4-methoxyacetophenone in basic media leads to the new ligands 2-(4-methoxyphenyl)-1,8-naphthyridine and 2-(2-methoxyphenyl)-1,8-naphthyridine, respectively, in high yield. The reaction of these naphthyridine derivatives with [RuCl2(CO)2]n leads to the respective complexes cis-dicarbonyldichloridobis[2-(4-methoxyphenyl)-1,8-naphthyridine-κN8]ruthenium(II) and cis-dicarbonyldichloridobis[2-(2-methoxyphenyl)-1,8-naphthyridine-κN8]ruthenium(II), both [RuCl2(C15H12N2O)2(CO)2], in good yield. Both ruthenium(II) complexes display a slightly distorted octahedron with two cis carbonyl, two cis chloride and two cis naphthyridine ligands, the latter coordinated in a monodentate fashion through the N atom in the 8-position. Both complexes exhibit a moderate catalytic activity in the hydrogen-transfer reaction from propan-2-ol to acetophenone in the presence of a base, with 100% selectivity.

6.
Front Immunol ; 8: 171, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28261222

RESUMO

Most pathogens infect through mucosal surfaces, and parenteral immunization typically fails to induce effective immune responses at these sites. Development of oral-administered vaccines capable of inducing mucosal as well as systemic immunity while bypassing the issues of antigen degradation and immune tolerance could be crucial for the control of enteropathogens. This study demonstrates that U-Omp19, a bacterial protease inhibitor with immunostimulatory features, coadministered with Salmonella antigens by the oral route, enhances mucosal and systemic immune responses in mice. U-Omp19 was able to increase antigen-specific production of IFN-γ and IL-17 and mucosal (IgA) antibody response. Finally, oral vaccination with U-Omp19 plus Salmonella antigens conferred protection against virulent challenge with Salmonella Typhimurium, with a significant reduction in bacterial loads. These findings prove the efficacy of this novel adjuvant in the Salmonella infection model and support the potential of U-Omp19 as a suitable adjuvant in oral vaccine formulations against mucosal pathogens requiring T helper (Th)1-Th17 protective immune responses.

7.
J Immunol ; 196(10): 4014-29, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27084100

RESUMO

In this study, we demonstrate that the unlipidated (U) outer membrane protein (Omp) 19 from Brucella spp. is a competitive inhibitor of human cathepsin L. U-Omp19 inhibits lysosome cathepsins and APC-derived microsome activity in vitro and partially inhibits lysosomal cathepsin L activity within live APCs. Codelivery of U-Omp19 with the Ag can reduce intracellular Ag digestion and increases Ag half-life in dendritic cells (DCs). U-Omp19 retains the Ag in Lamp-2(+) compartments after its internalization and promotes a sustained expression of MHC class I/peptide complexes in the cell surface of DCs. Consequently, U-Omp19 enhances Ag cross-presentation by DCs to CD8(+) T cells. U-Omp19 s.c. delivery induces the recruitment of CD11c(+)CD8α(+) DCs and monocytes to lymph nodes whereas it partially limits in vivo Ag proteolysis inside DCs. Accordingly, this protein is able to induce CD8(+) T cell responses in vivo against codelivered Ag. Antitumor responses were elicited after U-Omp19 coadministration, increasing survival of mice in a murine melanoma challenge model. Collectively, these results indicate that a cysteine protease inhibitor from bacterial origin could be a suitable component of vaccine formulations against tumors.


Assuntos
Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella/imunologia , Brucelose/imunologia , Linfócitos T CD8-Positivos/fisiologia , Vacinas Anticâncer/imunologia , Catepsinas/metabolismo , Células Dendríticas/imunologia , Imunoterapia/métodos , Lipoproteínas/metabolismo , Lisossomos/metabolismo , Melanoma/terapia , Animais , Antígenos de Neoplasias/imunologia , Apresentação Cruzada , Feminino , Ativação Linfocitária , Proteína 2 de Membrana Associada ao Lisossomo/metabolismo , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Vaccine ; 34(4): 430-437, 2016 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-26707377

RESUMO

The discovery of effective adjuvants for many vaccines especially those with limited commercial appeal, such as vaccines to poverty-related diseases, is required. In this work, we demonstrated that subcutaneous co-administration of mice with the outer membrane protein U-Omp19 from Brucella spp. plus OVA as antigen (Ag) increases Ag-specific T cell proliferation and T helper (Th) 1 immune responses in vitro and in vivo. U-Omp19 treated dendritic cells promote IFN-γ production by specific CD4(+) T cells and increases T cell proliferation. U-Omp19 co-administration induces the production of Ag specific effector memory T cell populations (CD4(+) CD44(high) CD62L(low) T cells). Finally, subcutaneous co-administration of U-Omp19 with Trypanosoma cruzi Ags confers protection against virulent parasite challenge, reducing parasitemia and weight loss while increasing mice survival. These results indicate that the bacterial protein U-Omp19 when delivered subcutaneously could be a suitable component of vaccine formulations against infectious diseases requiring Th1 immune responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Imunidade Celular , Lipoproteínas/imunologia , Células Th1/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Protozoários/imunologia , Brucella abortus , Bovinos , Células Cultivadas , Células Dendríticas/imunologia , Feminino , Memória Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Proteínas Recombinantes/imunologia , Trypanosoma cruzi
9.
J Control Release ; 220(Pt A): 18-28, 2015 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-26456256

RESUMO

We report here that a bacterial protease inhibitor from Brucella spp. called U-Omp19 behaves as an ideal constituent for a vaccine formulation against infectious diseases. When co-administered orally with an antigen (Ag), U-Omp19: i) can bypass the harsh environment of the gastrointestinal tract by inhibiting stomach and intestine proteases and consequently increases the half-life of the co-administered Ag at immune inductive sites: Peyer's patches and mesenteric lymph nodes while ii) it induces the recruitment and activation of antigen presenting cells (APCs) and increases the amount of intracellular Ag inside APCs. Therefore, mucosal as well as systemic Ag-specific immune responses, antibodies, Th1, Th17 and CD8(+) T cells are enhanced when U-Omp19 is co-administered with the Ag orally. Finally, this bacterial protease inhibitor in an oral vaccine formulation confers mucosal protection and reduces parasite loads after oral challenge with virulent Toxoplasma gondii.


Assuntos
Antígenos/metabolismo , Proteínas de Bactérias/farmacologia , Brucella/química , Imunidade nas Mucosas , Inibidores de Proteases/farmacologia , Vacinas/imunologia , Administração Oral , Sequência de Aminoácidos , Animais , Feminino , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular
10.
Hum Vaccin Immunother ; 10(7): 2015-23, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25424811

RESUMO

Food allergies are increasingly common disorders and no therapeutic strategies are yet approved. The unlipidated Omp16 (U-Omp16) is the outer membrane protein of 16 kDa from B. abortus and possesses a mucosal adjuvant property. In this study, we aimed to examine the U-Omp16 capacity to abrogate an allergen-specific Th2 immune response when it is administered as an oral adjuvant in a mouse model of food allergy.   Balb/c mice were sensitized with cholera toxin and cow's milk proteins (CMP) by gavage and simultaneously treated with U-Omp16 and CMP. Oral challenge with CMP was performed to evaluate the allergic status of mice. Symptoms, local (small bowel cytokine and transcription factor gene expression) and systemic (specific isotypes and spleen cell-secreted cytokines) parameters, and skin tests were done to evaluate the immune response. We found that the oral administration of U-Omp16 with CMP during sensitization dampened the allergic symptoms, with negativization of immediate skin test and increased skin DTH response. Serum specific IgE and IL-5 were inhibited and a Th1 response was promoted (specific IgG2a antibodies and CMP-induced IFN-γ secretion). We found at the mucosal site an inhibition of the gene expression corresponding to IL-13 and Gata-3, with an induction of IFN-γ and T-bet. These results indicated that the oral administration of U-Omp16 significantly controlled the allergic response in sensitized mice with a shift of the balance of Th1- and Th2-T cells toward Th1 predominance. These findings suggest that U-Omp16 may be useful as a Th1-directing adjuvant in an oral vaccine.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Brucella abortus/imunologia , Hipersensibilidade a Leite/prevenção & controle , Administração Oral , Animais , Imunoglobulina E/sangue , Masculino , Camundongos Endogâmicos BALB C , Proteínas do Leite/imunologia , Proteínas Recombinantes/administração & dosagem , Células Th1/imunologia , Células Th2/imunologia
11.
Clin Vaccine Immunol ; 21(12): 1689-94, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25339409

RESUMO

Canine brucellosis is an infectious disease caused by the Gram-negative bacterium Brucella canis. Unlike conventional control programs for other species of the genus Brucella, currently there is no vaccine available against canine brucellosis, and preventive measures are simply diagnosis and isolation of infected dogs. New approaches are therefore needed to develop an effective and safe immunization strategy against this zoonotic pathogen. In this study, BALB/c mice were subcutaneously immunized with the following: (i) the recombinant Brucella Omp31 antigen formulated in different adjuvants (incomplete Freund adjuvant, aluminum hydroxide, Quil A, and Montanide IMS 3012 VGPR), (ii) plasmid pCIOmp31, or (iii) pCIOmp31 plasmid followed by boosting with recombinant Omp31 (rOmp31). The immune response and the protective efficacy against B. canis infection were characterized. The different strategies induced a strong immunoglobulin G (IgG) response. Furthermore, spleen cells from rOmp31-immunized mice produced gamma interferon and interleukin-4 (IL-4) after in vitro stimulation with rOmp31, indicating the induction of a mixed Th1-Th2 response. Recombinant Omp31 administered with different adjuvants as well as the prime-boost strategy conferred protection against B. canis. In conclusion, our results suggest that Omp31 could be a useful candidate for the development of a subcellular vaccine against B. canis infection.


Assuntos
Anticorpos Antibacterianos/sangue , Proteínas da Membrana Bacteriana Externa/imunologia , Vacina contra Brucelose/imunologia , Brucella canis/imunologia , Brucelose/imunologia , Linfócitos T Citotóxicos/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Brucelose/prevenção & controle , Cães , Camundongos , Camundongos Endogâmicos BALB C , Vacinação
12.
PLoS One ; 9(7): e101279, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24983999

RESUMO

Brucella, the etiological agent of animal and human brucellosis, is a bacterium with the capacity to modulate the inflammatory response. Cyclic ß-1,2-glucan (CßG) is a virulence factor key for the pathogenesis of Brucella as it is involved in the intracellular life cycle of the bacteria. Using comparative studies with different CßG mutants of Brucella, cgs (CßG synthase), cgt (CßG transporter) and cgm (CßG modifier), we have identified different roles for this polysaccharide in Brucella. While anionic CßG is required for bacterial growth in low osmolarity conditions, the sole requirement for a successful Brucella interaction with mammalian host is its transport to periplasmic space. Our results uncover a new role for CßG in promoting splenomegaly in mice. We showed that CßG-dependent spleen inflammation is the consequence of massive cell recruitment (monocytes, dendritics cells and neutrophils) due to the induction of pro-inflammatory cytokines such as IL-12 and TNF-α and also that the reduced splenomegaly response observed with the cgs mutant is not the consequence of changes in expression levels of the characterized Brucella PAMPs LPS, flagellin or OMP16/19. Complementation of cgs mutant with purified CßG increased significantly spleen inflammation response suggesting a direct role for this polysaccharide.


Assuntos
Brucelose/microbiologia , Inflamação/microbiologia , Esplenomegalia/microbiologia , beta-Glucanas/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Brucella abortus/genética , Brucella abortus/metabolismo , Citocinas/metabolismo , Técnicas de Inativação de Genes , Glucosiltransferases/genética , Camundongos
13.
PLoS One ; 8(7): e69438, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861971

RESUMO

The discovery of novel mucosal adjuvants will help to develop new formulations to control infectious and allergic diseases. In this work we demonstrate that U-Omp16 from Brucella spp. delivered by the nasal route (i.n.) induced an inflammatory immune response in bronchoalveolar lavage (BAL) and lung tissues. Nasal co-administration of U-Omp16 with the model antigen (Ag) ovalbumin (OVA) increased the amount of Ag in lung tissues and induced OVA-specific systemic IgG and T helper (Th) 1 immune responses. The usefulness of U-Omp16 was also assessed in a mouse model of food allergy. U-Omp16 i.n. administration during sensitization ameliorated the hypersensitivity responses of sensitized mice upon oral exposure to Cow's Milk Protein (CMP), decreased clinical signs, reduced anti-CMP IgE serum antibodies and modulated the Th2 response in favor of Th1 immunity. Thus, U-Omp16 could be used as a broad Th1 mucosal adjuvant for different Ag formulations.


Assuntos
Adjuvantes Imunológicos , Proteínas da Membrana Bacteriana Externa/imunologia , Brucella/imunologia , Hipersensibilidade a Leite/imunologia , Proteínas do Leite/imunologia , Células Th1/imunologia , Células Th2/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Antígenos/imunologia , Antígenos/metabolismo , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/química , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Bovinos , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Imunoglobulina E/imunologia , Imunoglobulina G/imunologia , Pulmão/imunologia , Pulmão/patologia , Camundongos , Hipersensibilidade a Leite/metabolismo , Mucosa Nasal/imunologia , Mucosa Nasal/metabolismo , Baço/imunologia , Células Th1/metabolismo , Células Th2/metabolismo
14.
J Neuroinflammation ; 10: 47, 2013 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-23587438

RESUMO

BACKGROUND: Central nervous system (CNS) invasion by bacteria of the genus Brucella results in an inflammatory disorder called neurobrucellosis. We have recently demonstrated that B. abortus infects microglia and astrocytes, eliciting the production of a variety of pro-inflammatory cytokines which contribute to CNS damage. Matrix metalloproteinases (MMP) have been implicated in inflammatory tissue destruction in a range of pathological situations in the CNS. Increased MMP secretion is induced by pro-inflammatory cytokines in a variety of CNS diseases characterized by tissue-destructive pathology. METHODS: In this study, the molecular mechanisms that regulate MMP secretion from Brucella-infected astrocytes in vitro were investigated. MMP-9 was evaluated in culture supernatants by ELISA, zymography and gelatinolytic activity. Involvement of mitogen-activated protein kinases (MAPK) signaling pathways was evaluated by Western blot and using specific inhibitors. The role of TNF-α was evaluated by ELISA and by assays with neutralizing antibodies. RESULTS: B. abortus infection induced the secretion of MMP-9 from murine astrocytes in a dose-dependent fashion. The phenomenon was independent of bacterial viability and was recapitulated by L-Omp19, a B. abortus lipoprotein model, but not its LPS. B. abortus and L-Omp19 readily activated p38 and Erk1/2 MAPK, thus enlisting these pathways among the kinase pathways that the bacteria may address as they invade astrocytes. Inhibition of p38 or Erk1/2 significantly diminished MMP-9 secretion, and totally abrogated production of this MMP when both MAPK pathways were inhibited simultaneously. A concomitant abrogation of B. abortus- and L-Omp19-induced TNF-α production was observed when p38 and Erk1/2 pathways were inhibited, indicating that TNF-α could be implicated in MMP-9 secretion. MMP-9 secretion induced by B. abortus or L-Omp19 was completely abrogated when experiments were conducted in the presence of a TNF-α neutralizing antibody. MMP-9 activity was detected in cerebrospinal fluid (CSF) samples from patients suffering from neurobrucellosis. CONCLUSIONS: Our results indicate that the inflammatory response elicited by B. abortus in astrocytes would lead to the production of MMP-9 and that MAPK may play a role in this phenomenon. MAPK inhibition may thus be considered as a strategy to control inflammation and CNS damage in neurobrucellosis.


Assuntos
Brucella abortus , Brucelose/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/fisiologia , Animais , Anticorpos Bloqueadores/farmacologia , Antígenos de Bactérias/fisiologia , Astrócitos/metabolismo , Astrócitos/microbiologia , Astrócitos/fisiologia , Proteínas da Membrana Bacteriana Externa/fisiologia , Citocinas/metabolismo , Gelatinases/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Lipopolissacarídeos/farmacologia , Lipoproteínas/farmacologia , Lipoproteínas/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Cultura Primária de Células , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
15.
Microbes Infect ; 14(7-8): 639-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22387699

RESUMO

Immune evasion is essential for Brucella abortus to survive in the face of robust adaptive CD4+ T cell response. We have previously demonstrated that B. abortus can indirectly inhibit CD4+ T cells by down-regulating MHC-II expression and antigen presentation on macrophages. However, whether B. abortus is able to directly interfere with T lymphocytes is not known. We report here that B. abortus induces apoptosis of human T lymphocytes, even though invasion of T lymphocytes was low and non-replicative. The ability of heat-killed B. abortus to reproduce the same phenomenon suggested that there was a bacterial structural component involved. We demonstrated that a prototypical B. abortus outer membrane lipoprotein (l-Omp19), but not its unlipidated form, induced T lymphocyte apoptosis. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also induced an increase in T lymphocyte cell death, indicating that the structural component implicated in the phenomenon could be any B. abortus lipoprotein. B. abortus-induced T lymphocyte apoptosis was dependent on the secretion of TNF-α since pre-incubation of T lymphocytes with anti-TNF-α mAb inhibited the apoptosis of the cells. Overall, these results represent a new mechanism whereby B. abortus by directly inhibiting T cell-mediated responses may evade adaptive immune responses.


Assuntos
Apoptose , Proteínas da Membrana Bacteriana Externa/imunologia , Brucella abortus/patogenicidade , Lipoproteínas/imunologia , Linfócitos T/imunologia , Linfócitos T/patologia , Fator de Necrose Tumoral alfa/biossíntese , Brucelose/microbiologia , Dinoprostona/biossíntese , Humanos , Linfócitos T/microbiologia
16.
Acta Crystallogr Sect E Struct Rep Online ; 68(Pt 11): m1377-8, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23284355

RESUMO

The mol-ecular structure of the title compound, [Zn(CH(3)COO)(2)(C(12)H(12)N(2))], consists of isolated mol-ecules bis-ected by a twofold rotation axis which goes through the Zn(II) cation and halves the organic base through the central C-C bond. The Zn(II) ion is coordinated by two N atoms from one mol-ecule of the aromatic base and four O atoms from two bidentate, symmetry-related acetate anions, which coordinate asym-metrically [Zn-O distances of 2.058 (2) and 2.362 (3) Å], while the two Zn-N bond distances are equal as imposed by symmetry [2.079 (2) Å]. The crystal structure is supported by a number of weak C-H⋯O inter-actions and C-H⋯π contacts, with no π-π inter-actions present, mainly hindered by the substituent methyl groups and the relative mol-ecular orientation. The result is a three-dimensional structure in which each mol-ecule is linked to eight different neighbors.

17.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 6): m675-6, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21754584

RESUMO

In the title compound, [MoBr(2)(C(12)H(11)N(2))(C(12)H(10)N(2))(C(5)H(7)O(2))], the Mo(VI) atom is six-coordinated in a distorted octa-hedral geometry by two N atoms from the diphenyl-hydrazide(1-) and diphenyl-hydrazide(2-) ligands, two O atoms from a bidentate acetyl-acetonate ligand and two Br(-) ions. The mol-ecules form an inversion dimer via a pair of weak C-H⋯O hydrogen bonds and a π-π stacking inter-action with a centroid-centroid distance of 3.7401 (12) Å. Weak intra-molecular C-H⋯Br inter-actions and an intra-molecular π-π stacking inter-action with a centroid-centroid distance of 3.8118 (15) Šare also observed.

18.
Acta Crystallogr Sect E Struct Rep Online ; 67(Pt 6): o1450-1, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21754825

RESUMO

The mol-ecular structure of the title compound, C(21)H(15)ClN(2)O(2), features one strong intra-molecular N-H⋯O resonance-assisted hydrogen bond (RAHB). In the crystal, mol-ecules form inversion-related dimers via pairs of weak inter-molecular N-H⋯O contacts. These dimers are further stabilized via three weak C-H⋯O contacts, developing the three-dimensional structure.

19.
PLoS One ; 6(1): e16203, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21264260

RESUMO

As Brucella infections occur mainly through mucosal surfaces, the development of mucosal administered vaccines could be radical for the control of brucellosis. In this work we evaluated the potential of Brucella abortus 19 kDa outer membrane protein (U-Omp19) as an edible subunit vaccine against brucellosis. We investigated the protective immune response elicited against oral B. abortus infection after vaccination of mice with leaves from transgenic plants expressing U-Omp19; or with plant-made or E. coli-made purified U-Omp19. All tested U-Omp19 formulations induced protection against Brucella when orally administered without the need of adjuvants. U-Omp19 also induced protection against a systemic challenge when parenterally administered. This built-in adjuvant ability of U-Omp19 was independent of TLR4 and could be explained at least in part by its capability to activate dendritic cells in vivo. While unadjuvanted U-Omp19 intraperitoneally administered induced a specific Th1 response, following U-Omp19 oral delivery a mixed specific Th1-Th17 response was induced. Depletion of CD4(+) T cells in mice orally vaccinated with U-Omp19 resulted in a loss of the elicited protection, indicating that this cell type mediates immune protection. The role of IL-17 against Brucella infection has never been explored. In this study, we determined that if IL-17A was neutralized in vivo during the challenge period, the mucosal U-Omp19 vaccine did not confer mucosal protection. On the contrary, IL-17A neutralization during the infection did not influence at all the subsistence and growth of this bacterium in PBS-immunized mice. All together, our results indicate that an oral unadjuvanted vaccine based on U-Omp19 induces protection against a mucosal challenge with Brucella abortus by inducing an adaptive IL-17 immune response. They also indicate different and important new aspects i) IL-17 does not contribute to reduce the bacterial burden in non vaccinated mice and ii) IL-17 plays a central role in vaccine mediated anti-Brucella mucosal immunity.


Assuntos
Antígenos de Bactérias/uso terapêutico , Proteínas da Membrana Bacteriana Externa/uso terapêutico , Vacina contra Brucelose/imunologia , Brucella abortus/imunologia , Interleucina-17/imunologia , Lipoproteínas/uso terapêutico , Imunidade Adaptativa , Animais , Vacina contra Brucelose/administração & dosagem , Brucelose/prevenção & controle , Linfócitos T CD4-Positivos/imunologia , Camundongos , Células Th1/imunologia , Células Th17/imunologia
20.
Microbes Infect ; 13(3): 239-50, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21070860

RESUMO

The strategies that allow Brucella abortus to persist for years inside macrophages subverting host immune responses are not completely understood. Immunity against this bacterium relies on the capacity of IFN-γ to activate macrophages, endowing them with the ability to destroy intracellular bacteria. We report here that infection with B. abortus down-modulates the expression of the type I receptor for the Fc portion of IgG (FcγRI, CD64) and FcγRI-restricted phagocytosis regulated by IFN-γ in human monocytes/macrophages. Both phenomena were not dependent on bacterial viability, since they were also induced by heat-killed B. abortus (HKBA), suggesting that they were elicited by a structural bacterial component. Accordingly, a prototypical B. abortus lipoprotein (L-Omp19), but not its unlipidated form, inhibited both CD64 expression and FcγRI-restricted phagocytosis regulated by IFN-γ. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also inhibited CD64 expression, indicating that any Brucella lipoprotein could down-modulate CD64 expression and FcγRI-restricted phagocytosis. Pre-incubation of monocytes/macrophages with anti-TLR2 mAb blocked the inhibition of the CD64 expression mediated by HKBA and L-Omp19. These results, together with our previous observations establish that B. abortus utilizes its lipoproteins to inhibit the monocytes/macrophages activation mediated by IFN-γ and to subvert host immunonological responses.


Assuntos
Brucella abortus/fisiologia , Interferon gama/antagonistas & inibidores , Macrófagos/imunologia , Monócitos/imunologia , Receptores de IgG/biossíntese , Receptor 2 Toll-Like/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/química , Brucella abortus/metabolismo , Células Cultivadas , Citometria de Fluxo , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Lipoproteínas/metabolismo , Macrófagos/microbiologia , Monócitos/microbiologia , Fagocitose/fisiologia , Receptores de IgG/antagonistas & inibidores , Receptores de IgG/metabolismo , Estatísticas não Paramétricas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...