Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Comput Struct Biotechnol J ; 20: 5098-5114, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36187929

RESUMO

U-Omp19 is a bacterial protease inhibitor from Brucella abortus that inhibits gastrointestinal and lysosomal proteases, enhancing the half-life and immunogenicity of co-delivered antigens. U-Omp19 is a novel adjuvant that is in preclinical development with various vaccine candidates. However, the molecular mechanisms by which it exerts these functions and the structural elements responsible for these activities remain unknown. In this work, a structural, biochemical, and functional characterization of U-Omp19 is presented. Dynamic features of U-Omp19 in solution by NMR and the crystal structure of its C-terminal domain are described. The protein consists of a compact C-terminal beta-barrel domain and a flexible N-terminal domain. The latter domain behaves as an intrinsically disordered protein and retains the full protease inhibitor activity against pancreatic elastase, papain and pepsin. This domain also retains the capacity to induce CD8+ T cells in vivo of U-Omp19. This information may lead to future rationale vaccine designs using U-Omp19 as an adjuvant to deliver other proteins or peptides in oral formulations against infectious diseases, as well as to design strategies to incorporate modifications in its structure that may improve its adjuvanticity.

2.
Clin Exp Allergy ; 50(8): 954-963, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32501552

RESUMO

BACKGROUND: IgE-mediated food allergy remains a significant and growing worldwide problem. Sublingual immunotherapy (SLIT) shows an excellent safety profile for food allergy, but the clinical efficacy needs to be improved. This study assessed the effects of the Toll-like receptor 4 agonist outer membrane protein (Omp) 16 from Brucella abortus combined with cow´s milk proteins (CMP) through the sublingual route to modulate cow's milk allergy in an experimental model. METHODS: Mice sensitized with cholera toxin and CMP were orally challenged with the allergen to elicit hypersensitivity reactions. Then, mice were treated with a very low amount of CMP along with Omp16 as a mucosal adjuvant, and finally, animals were re-exposed to CMP. Systemic and mucosal immune parameters were assessed in vivo and in vitro. RESULTS: We found that the sublingual administration of Omp16 + CMP induced a buccal Th1 immune response that modulated the intestinal allergic response with the suppression of symptoms, reduction of IgE and IL-5, and up-regulation of IgG2a and IFN-γ. The adoptive transfer of submandibular IFN-γ-producing α4ß7+ CD4+ and CD8+ cells conferred protection against allergic sensitization. The use of Omp16 + CMP promoted enhanced protection compared to CMP alone. CONCLUSION: In conclusion, Omp16 represents a promising mucosal adjuvant that can be used to improve the clinical and immune efficacy of SLIT for food allergy.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Alérgenos/administração & dosagem , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas de Ciclo Celular/administração & dosagem , Imunidade nas Mucosas/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Hipersensibilidade a Leite/terapia , Proteínas do Leite/administração & dosagem , Imunoterapia Sublingual , Subpopulações de Linfócitos T/efeitos dos fármacos , Administração Sublingual , Transferência Adotiva , Alérgenos/imunologia , Animais , Proteínas da Membrana Bacteriana Externa/imunologia , Proteínas de Ciclo Celular/imunologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Imunoglobulina E/metabolismo , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-5/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos BALB C , Hipersensibilidade a Leite/imunologia , Hipersensibilidade a Leite/metabolismo , Proteínas do Leite/imunologia , Mucosa Bucal/efeitos dos fármacos , Mucosa Bucal/imunologia , Mucosa Bucal/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/transplante , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/metabolismo
3.
J Control Release ; 293: 158-171, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30496771

RESUMO

The study of capture and processing of antigens (Ags) by intestinal epithelial cells is very important for development of new oral administration systems. Efficient oral Ag delivery systems must resist enzymatic degradation by gastric and intestinal proteases and deliver the Ag across biological barriers. The recombinant unlipidated outer membrane protein from Brucella spp. (U-Omp19) is a protease inhibitor with immunostimulatory properties used as adjuvant in oral vaccine formulations. In the present work we further characterized its mechanism of action and studied the interaction and effect of U-Omp19 on the intestinal epithelium. We found that U-Omp19 inhibited protease activity from murine intestinal brush-border membranes and cysteine proteases from human intestinal epithelial cells (IECs) promoting co-administered Ag accumulation within lysosomal compartments of IECs. In addition, we have shown that co-administration of U-Omp19 facilitated the transcellular passage of Ag through epithelial cell monolayers in vitro and in vivo while did not affect epithelial cell barrier permeability. Finally, oral co-delivery of U-Omp19 in mice induced the production of Ag-specific IgA in feces and the increment of CD103+ CD11b- CD8α+ dendritic cells subset at Peyer's patches. Taken together, these data describe a new mechanism of action of a mucosal adjuvant and support the use of this rationale/strategy in new oral delivery systems for vaccines.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Antígenos de Bactérias/administração & dosagem , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Mucosa Intestinal/metabolismo , Lipoproteínas/administração & dosagem , Inibidores de Proteases/administração & dosagem , Vacinas/administração & dosagem , Administração Oral , Animais , Células CACO-2 , Células Epiteliais/metabolismo , Feminino , Células HT29 , Humanos , Camundongos Endogâmicos BALB C
4.
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
5.
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
6.
Infect Immun ; 81(5): 1654-62, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23460520

RESUMO

Brucella abortus is recognized by several Toll-like receptor (TLR)-associated pathways triggering proinflammatory responses that affect both the nature and intensity of the immune response. Previously, we demonstrated that B. abortus-mediated dendritic cell (DC) maturation and control of infection are dependent on the adaptor molecule MyD88. However, the involvement of all TLRs in response to B. abortus infection is not completely understood. Therefore, we decided to evaluate the requirement for TLR6 in host resistance to B. abortus. Here, we demonstrated that TLR6 is an important component for triggering an innate immune response against B. abortus. An in vitro luciferase assay indicated that TLR6 cooperates with TLR2 to sense Brucella and further activates NF-κB signaling. However, in vivo analysis showed that TLR6, not TLR2, is required for the efficient control of B. abortus infection. Additionally, B. abortus-infected dendritic cells require TLR6 to induce tumor necrosis factor alpha (TNF-α) and interleukin-12 (IL-12). Furthermore, our findings demonstrated that the mitogen-activated protein kinase (MAPK) signaling pathway is impaired in TLR2, TLR6, and TLR2/6 knockout (KO) DCs when infected with B. abortus, which may account for the lower proinflammatory cytokine production observed in TLR6 KO mouse dendritic cells. In summary, the results presented here indicate that TLR6 is required to trigger innate immune responses against B. abortus in vivo and is required for the full activation of DCs to induce robust proinflammatory cytokine production.


Assuntos
Brucella abortus/imunologia , Brucelose/imunologia , Imunidade Inata/fisiologia , Receptor 6 Toll-Like/fisiologia , Análise de Variância , Animais , Citocinas/metabolismo , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Baço/microbiologia , Receptor 2 Toll-Like/fisiologia , Receptor 6 Toll-Like/deficiência
7.
Cell Microbiol ; 15(4): 487-502, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23107169

RESUMO

Brucella abortus elicits a vigorous Th1 immune response which activates cytotoxic T lymphocytes. However, B. abortus persists in its hosts in the presence of CD8(+) T cells, establishing a chronic infection. Here, we report that B. abortus infection of human monocytes/macrophages inhibited the IFN-γ-induced MHC-I cell surface expression. This phenomenon was dependent on metabolically active viable bacteria. MHC-I down-modulation correlated with the development of diminished CD8(+) cytotoxic T cell response as evidenced by the reduced expression of the activation marker CD107a on CD8(+) T lymphocytes and a diminished percentage of IFN-γ-producing CD8(+) T cells. Inhibition of MHC-I expression was not due to changes in protein synthesis. Rather, we observed that upon B. abortus infection MHC-I molecules were retained within the Golgi apparatus. Overall, these results describe a novel mechanism based on the intracellular sequestration of MHC-I molecules whereby B. abortus would avoid CD8(+) cytotoxic T cell responses, evading their immunological surveillance.


Assuntos
Brucella abortus/imunologia , Brucella abortus/fisiologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Evasão da Resposta Imune , Macrófagos/imunologia , Macrófagos/microbiologia , Células Cultivadas , Complexo de Golgi/química , Humanos , Interferon gama/metabolismo , Transporte Proteico
8.
J Immunol ; 189(3): 1162-72, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22753933

RESUMO

Aberrant Stat3 activation and signaling contribute to malignant transformation by promoting cell cycle progression, inhibiting apoptosis, and mediating tumor immune evasion. Stat3 inhibition in tumor cells induces the expression of chemokines and proinflammatory cytokines, so we proposed to apply Stat3-inhibited breast cancer cells as a source of immunogens to induce an antitumor immune response. Studies were performed in two murine breast cancer models in which Stat3 is activated: progestin-dependent C4HD cells and 4T1 cells. We immunized BALB/c mice with irradiated cancer cells previously transfected with a dominant-negative Stat3 vector (Stat3Y705F) in either a prophylactic or a therapeutic manner. Prophylactic administration of breast cancer cells transfected with Stat3Y705F (Stat3Y705F-breast cancer cells) inhibited primary tumor growth compared with administration of empty vector-transfected cells in both models. In the 4T1 model, 50% of the challenged mice were tumor free, and the incidence of metastasis decreased by 90%. In vivo assays of C4HD tumors showed that the antitumor immune response involves the participation of CD4(+) T cells and cytotoxic NK cells. Therapeutic immunization with Stat3Y705F-breast cancer cells inhibited tumor growth, promoted tumor cell differentiation, and decreased metastasis. Furthermore, inhibition of Stat3 activation in breast cancer cells induced cellular senescence, contributing to their immunogenic phenotype. In this work, we provide preclinical proof of concept that ablating Stat3 signaling in breast cancer cells results in an effective immunotherapy against breast cancer growth and metastasis. Moreover, our findings showing that Stat3 inactivation results in induction of a cellular senescence program disclose a potential mechanism for immunotherapy research.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Senescência Celular/imunologia , Marcação de Genes , Células Matadoras Naturais/imunologia , Neoplasias Mamárias Animais/imunologia , Neoplasias Mamárias Animais/terapia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Marcação de Genes/métodos , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Cultura Primária de Células , Fator de Transcrição STAT3
9.
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
10.
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
11.
J Immunol ; 184(9): 5200-12, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20351187

RESUMO

Knowing the inherent stimulatory properties of the lipid moiety of bacterial lipoproteins, we first hypothesized that Brucella abortus outer membrane protein (Omp)16 lipoprotein would be able to elicit a protective immune response without the need of external adjuvants. In this study, we demonstrate that Omp16 administered by the i.p. route confers significant protection against B. abortus infection and that the protective response evoked is independent of the protein lipidation. To date, Omp16 is the first Brucella protein that without the requirement of external adjuvants is able to induce similar protection levels to the control live vaccine S19. Moreover, the protein portion of Omp16 (unlipidated Omp16 [U-Omp16]) elicits a protective response when administered by the oral route. Either systemic or oral immunization with U-Omp16 elicits a Th1-specific response. These abilities of U-Omp16 indicate that it is endowed with self-adjuvanting properties. The adjuvanticity of U-Omp16 could be explained, at least in part, by its capacity to activate dendritic cells in vivo. U-Omp16 is also able to stimulate dendritic cells and macrophages in vitro. The latter property and its ability to induce a protective Th1 immune response against B. abortus infection have been found to be TLR4 dependent. The facts that U-Omp16 is an oral protective Ag and possesses a mucosal self-adjuvanting property led us to develop a plant-made vaccine expressing U-Omp16. Our results indicate that plant-expressed recombinant U-Omp16 is able to confer protective immunity, when given orally, indicating that a plant-based oral vaccine expressing U-Omp16 could be a valuable approach to controlling this disease.


Assuntos
Antígenos de Bactérias/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Vacina contra Brucelose/imunologia , Brucelose/prevenção & controle , Células Dendríticas/imunologia , Interações Hospedeiro-Patógeno/imunologia , Células Th1/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/genética , Administração Oral , Animais , Antígenos de Bactérias/administração & dosagem , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/genética , Vacina contra Brucelose/administração & dosagem , Brucelose/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Feminino , Adjuvante de Freund/administração & dosagem , Interações Hospedeiro-Patógeno/genética , Imunidade Celular , Injeções Intraperitoneais , Lipídeos/administração & dosagem , Lipoproteínas/administração & dosagem , Lipoproteínas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Th1/microbiologia , Nicotiana/genética , Nicotiana/imunologia
12.
Am J Pathol ; 176(3): 1323-38, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20093491

RESUMO

Central nervous system (CNS) invasion by bacteria of the genus Brucella results in an inflammatory disorder called neurobrucellosis. In this study we present in vivo and in vitro evidence that B. abortus and its lipoproteins activate the innate immunity of the CNS, eliciting an inflammatory response that leads to astrogliosis, a characteristic feature of neurobrucellosis. Intracranial injection of heat-killed B. abortus (HKBA) or outer membrane protein 19 (Omp19), a B. abortus lipoprotein model, induced astrogliosis in mouse striatum. Moreover, infection of astrocytes and microglia with B. abortus induced the secretion of interleukin (IL)-6, IL-1beta, tumor necrosis factor (TNF)-alpha, macrophage chemoattractant protein-1, and KC (CXCL1). HKBA also induced these inflammatory mediators, suggesting the involvement of a structural component of the bacterium. Accordingly, Omp19 induced the same cytokine and chemokine secretion pattern. B. abortus infection induced astrocyte, but not microglia, apoptosis. Indeed, HKBA and Omp19 elicited not only astrocyte apoptosis but also proliferation, two features observed during astrogliosis. Apoptosis induced by HKBA and L-Omp19 was completely suppressed in cells of TNF receptor p55-/- mice or when the general caspase inhibitor Z-VAD-FMK was added to cultures. Hence, TNF-alpha signaling via TNF receptor (TNFR) 1 through the coupling of caspases determines apoptosis. Our results provide proof of the principle that Brucella lipoproteins could be key virulence factors in neurobrucellosis and that astrogliosis might contribute to neurobrucellosis pathogenesis.


Assuntos
Apoptose , Astrócitos/microbiologia , Astrócitos/patologia , Brucella abortus/fisiologia , Mediadores da Inflamação/metabolismo , Animais , Antígenos de Bactérias/farmacologia , Apoptose/efeitos dos fármacos , Astrócitos/enzimologia , Astrócitos/metabolismo , Proteínas da Membrana Bacteriana Externa/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/microbiologia , Encéfalo/patologia , Brucella abortus/efeitos dos fármacos , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocinas/metabolismo , Feminino , Temperatura Alta , Imuno-Histoquímica , Lipopolissacarídeos/farmacologia , Lipoproteínas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Microglia/efeitos dos fármacos , Microglia/microbiologia , Microglia/patologia , Fator de Necrose Tumoral alfa/metabolismo
13.
Infect Immun ; 76(1): 250-62, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17984211

RESUMO

The strategies that allow Brucella abortus to survive inside macrophages for prolonged periods and to avoid the immunological surveillance of major histocompatibility complex class II (MHC-II)-restricted gamma interferon (IFN-gamma)-producing CD4+ T lymphocytes are poorly understood. We report here that infection of THP-1 cells with B. abortus inhibited expression of MHC-II molecules and antigen (Ag) processing. Heat-killed B. abortus (HKBA) also induced both these phenomena, indicating the independence of bacterial viability and involvement of a structural component of the bacterium. Accordingly, outer membrane protein 19 (Omp19), a prototypical B. abortus lipoprotein, inhibited both MHC-II expression and Ag processing to the same extent as HKBA. Moreover, a synthetic lipohexapeptide that mimics the structure of the protein lipid moiety also inhibited MHC-II expression, indicating that any Brucella lipoprotein could down-modulate MHC-II expression and Ag processing. Inhibition of MHC-II expression and Ag processing by either HKBA or lipidated Omp19 (L-Omp19) depended on Toll-like receptor 2 and was mediated by interleukin-6. HKBA or L-Omp19 also inhibited MHC-II expression and Ag processing of human monocytes. In addition, exposure to the synthetic lipohexapeptide inhibited Ag-specific T-cell proliferation and IFN-gamma production of peripheral blood mononuclear cells from Brucella-infected patients. Together, these results indicate that there is a mechanism by which B. abortus may prevent recognition by T cells to evade host immunity and establish a chronic infection.


Assuntos
Apresentação de Antígeno , Brucella abortus/fisiologia , Regulação da Expressão Gênica , Genes MHC da Classe II/genética , Interleucina-6/metabolismo , Receptor 2 Toll-Like/metabolismo , Antígenos de Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/metabolismo , Brucella abortus/imunologia , Células Cultivadas , Regulação para Baixo , Humanos , Interferon gama , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/microbiologia , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Monócitos/metabolismo , Monócitos/microbiologia
14.
Vaccine ; 25(37-38): 6721-9, 2007 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-17686554

RESUMO

The immunogenicity and protective efficacy of recombinant SurA (rSurA) and rDnaK from Brucella spp. were evaluated in BALB/c mice. Immunization with rSurA in adjuvant induced a vigorous immunoglobulin G (IgG) response, with higher IgG2a than IgG1 titers. In addition, after in vitro stimulation with rSurA, spleen cells from rSurA-immunized mice produced interleukin-2 (IL-2), interferon (IFN)-gamma, IL-4 and IL-5. Immunization with rDnaK plus adjuvant induced a strong humoral response resulting in similar anti-rDnaK IgG titers than immunization with rDnaK alone. IgG2a titers predominated over IgG1 in mice injected with rDnaK alone or rDnaK plus adjuvant. Spleen cells from mice immunized with rDnaK plus adjuvant secreted IFN-gamma and IL-2 upon stimulation with rDnaK and induced a specific cytotoxic response. On the contrary, mice immunized with rDnaK alone did not exhibit a specific T helper or cytotoxic response in vitro. Mice given rSurA or rDnaK with adjuvant exhibited a significant degree of protection whereas immunization with rDnaK alone induced a low but still statistically significant level of protection against B. abortus infection. All studied vaccines were less protected than mice immunized with H38 or B. abortus strain 19 control vaccines. Altogether these results suggest that rSurA or rDnaK induce partial protection against B. abortus infection and could be useful candidates for the development of subunit vaccines against brucellosis.


Assuntos
Adenosina Trifosfatases/imunologia , Vacina contra Brucelose/imunologia , Brucella abortus/imunologia , Brucelose/imunologia , Brucelose/prevenção & controle , Proteínas de Transporte/imunologia , Peptidilprolil Isomerase/imunologia , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Formação de Anticorpos/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Peptidilprolil Isomerase/genética , Peptidilprolil Isomerase/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Linfócitos T Citotóxicos/imunologia , Células Th1/imunologia , Células Th2/imunologia
15.
J Immunol ; 176(6): 3426-37, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16517711

RESUMO

We have demonstrated that in vivo administration of phosphorothioate antisense oligodeoxynucleotides (AS[S]ODNs) to type I insulin-like growth factor receptor (IGF-IR) mRNA resulted in inhibition of C4HD breast cancer growth in BALB/c mice. The present study focused on whether in vivo administration of C4HD tumor cells pretreated with IGF-IR AS[S]ODN and irradiated could provide protection against C4HD wild-type tumor challenge and also on elucidating the mechanism mediating this effect. Our results showed that mice immunized with IGF-IR AS[S]ODN-treated C4HD cells experienced a growth inhibition of 53.4%, 61.6%, and 60.2% when compared with PBS-treated mice, wild-type C4HD cell-injected mice, or phosphorothioate sense oligodeoxynucleotide-treated C4HD cell-injected mice, respectively. The protective effect was C4HD-specific, because no cross-protection was observed against other syngeneic mammary tumor lines. The lack of protection against tumor formation in nude mice indicated that T cells were involved in the antitumoral response. Furthermore, cytotoxicity and splenocyte proliferation assays demonstrated that a cellular CD8(+)-dependent immune response, acting through the Fas/Fas ligand death pathway, could be mediating the antitumor effect induced by immunization with AS[S]ODN-treated cells. Immunization also induced splenocytes to produce Ag-dependent IFN-gamma, indicating the presence of a type 1 response. We demonstrated for the first time that IGF-IR AS[S]ODN treatment of breast cancer cells induced expression of CD86 and heat shock protein 70 molecules, both involved in the induction of the immunogenic phenotype. Immunization with these tumor immunogens imparted protection against parental tumor growth through activation of a specific immune response.


Assuntos
Apoptose , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Glicoproteínas de Membrana/metabolismo , Receptor IGF Tipo 1/deficiência , Linfócitos T Citotóxicos/imunologia , Fatores de Necrose Tumoral/metabolismo , Receptor fas/metabolismo , Animais , Antígeno B7-2/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Vacinas Anticâncer/imunologia , Proliferação de Células , Células Cultivadas , Proteína Ligante Fas , Feminino , Proteínas de Choque Térmico HSP70/metabolismo , Imunização , Interferon gama/biossíntese , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Oligodesoxirribonucleotídeos Antissenso/genética , Fenótipo , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Linfócitos T Citotóxicos/metabolismo
16.
Infect Immun ; 73(12): 8079-88, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16299302

RESUMO

The immunogenicity and protective efficacy of the recombinant 31-kDa outer membrane protein from Brucella melitensis (rOmp31), administered with incomplete Freund's adjuvant, were evaluated in mice. Immunization of BALB/c mice with rOmp31 conferred protection against B. ovis and B. melitensis infection. rOmp31 induced a vigorous immunoglobulin G (IgG) response, with higher IgG1 than IgG2 titers. In addition, spleen cells from rOmp31-immunized mice produced interleukin 2 (IL-2) and gamma interferon, but not IL-10 or IL-4, after in vitro stimulation with rOmp31, suggesting the induction of a T helper 1 (Th1) response. Splenocytes from rOmp31-vaccinated animals also induced a specific cytotoxic-T-lymphocyte activity, which led to the in vitro lysis of Brucella-infected macrophages. In vitro T-cell subset depletion indicated that rOmp31 immunization elicited specific CD4+ T cells that secrete IL-2 and gamma interferon, while CD8+ T cells induced cytotoxic-T-lymphocyte activity. In vivo depletion of T-cell subsets showed that the rOmp31-elicited protection against B. melitensis infection is mediated by CD4+ T cells while the contribution of CD8+ T cells may be limited. We then evaluated the immunogenicity and protective efficacy of a known exposed region from Omp31 on the Brucella membrane, a peptide that contains amino acids 48 to 74 of Omp31. Immunization with the synthetic peptide in adjuvant did not elicit a specific humoral response but elicited a Th1 response mediated by CD4+ T cells. The peptide in adjuvant induced levels of protection similar to those induced by rOmp31 against B. melitensis but less protection than was induced by rOmp31 against B. ovis. Our results indicate that rOmp31 could be a useful candidate for the development of subunit vaccines against B. melitensis and B. ovis.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Vacina contra Brucelose/imunologia , Brucella melitensis/imunologia , Brucelose/prevenção & controle , Fragmentos de Peptídeos/imunologia , Células Th1/imunologia , Sequência de Aminoácidos , Animais , Proteínas da Membrana Bacteriana Externa/administração & dosagem , Vacina contra Brucelose/administração & dosagem , Brucella ovis/imunologia , Brucelose/imunologia , Linfócitos T CD4-Positivos/imunologia , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Feminino , Adjuvante de Freund/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/administração & dosagem , Peptídeos/imunologia , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia
17.
Infect Immun ; 73(10): 6537-46, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16177328

RESUMO

The development of an effective subunit vaccine against brucellosis is a research area of intense interest. The outer membrane proteins (Omps) of Brucella spp. have been extensively characterized as potential immunogenic and protective antigens. This study was conducted to evaluate the immunogenicity and protective efficacy of the B. melitensis Omp31 gene cloned in the pCI plasmid (pCIOmp31). Immunization of BALB/c mice with pCIOmp31 conferred protection against B. ovis and B. melitensis infection. Mice vaccinated with pCIOmp31 developed a very weak humoral response, and in vitro stimulation of their splenocytes with recombinant Omp31 did not induced the secretion of gamma interferon. Splenocytes from Omp31-vaccinated animals induced a specific cytotoxic-T-lymphocyte activity, which leads to the in vitro lysis of Brucella-infected macrophages. pCIOmp31 immunization elicited mainly CD8(+) T cells, which mediate cytotoxicity via perforins, but also CD4(+) T cells, which mediate lysis via the Fas-FasL pathway. In vivo depletion of T-cell subsets showed that the pCIOmp31-induced protection against Brucella infection is mediated predominantly by CD8(+) T cells, although CD4(+)T cells also contribute. Our results demonstrate that the Omp31 DNA vaccine induces cytotoxic responses that have the potential to contribute to protection against Brucella infection. The protective response could be related to the induction of CD8(+) T cells that eliminate Brucella-infected cells via the perforin pathway.


Assuntos
Proteínas da Membrana Bacteriana Externa/imunologia , Vacina contra Brucelose , Brucella melitensis , Brucella ovis , Brucelose/prevenção & controle , Linfócitos T Citotóxicos/imunologia , Animais , Formação de Anticorpos/imunologia , Proteínas da Membrana Bacteriana Externa/genética , Vacina contra Brucelose/genética , Vacina contra Brucelose/imunologia , Brucelose/imunologia , Linfócitos T CD4-Positivos/imunologia , Clonagem Molecular , Citotoxicidade Imunológica , Proteína Ligante Fas , Feminino , Genes Bacterianos , Interferon gama/metabolismo , Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Baço/imunologia , Vacinas de DNA/genética , Vacinas de DNA/imunologia
18.
J Immunol ; 173(7): 4635-42, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15383598

RESUMO

Inflammation is a hallmark of brucellosis. Although Brucella abortus, one of the disease's etiologic agents, possesses cytokine-stimulatory properties, the mechanism by which this bacterium triggers a proinflammatory response is not known. We examined the mechanism whereby heat-killed B. abortus (HKBA), as well as its LPS, induces production of inflammatory cytokines in monocytes/macrophages. Polymyxin B, a specific inhibitor of LPS activity, did not inhibit the production of TNF-alpha- and IL-6-induced HKBA in the human monocytic cell line THP-1. HKBA induced the production of these cytokines in peritoneal macrophages of both C3H/HeJ and C3H/HeN mice, whereas B. abortus LPS only stimulated cells from C3H/HeN mice. Anti-TLR2 Ab, but not anti-TLR4 Ab, blocked HKBA-mediated TNF-alpha and IL-6 production in THP-1 cells. Because bacterial lipoproteins, a TLR2 ligand, have potent inherent stimulatory properties, we investigated the capacity of two B. abortus lipoproteins, outer membrane protein 19 (Omp19) and Omp16, to elicit a proinflammatory response. Lipidated (L)-Omp16 and L-Omp19, but not their unlipidated forms, induced the secretion of TNF-alpha, IL-6, IL-10, and IL-12 in a time- and dose-dependent fashion. Preincubation of THP-1 cells with anti-TLR2 Ab blocked L-Omp19-mediated TNF-alpha and IL-6 production. Together, these results entail a mechanism whereby B. abortus can stimulate cells from the innate immune system and induce cytokine-mediated inflammation in brucellosis. We submit that LPS is not the cause of inflammation in brucellosis; rather, lipoproteins of this organism trigger the production of proinflammatory cytokines, and TLR2 is involved in this process.


Assuntos
Brucella abortus/imunologia , Temperatura Alta , Mediadores da Inflamação/fisiologia , Lipopolissacarídeos/farmacologia , Lipoproteínas/fisiologia , Animais , Antígenos de Bactérias/biossíntese , Antígenos de Bactérias/genética , Antígenos de Bactérias/isolamento & purificação , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Brucella abortus/genética , Linhagem Celular Tumoral , Citocinas/biossíntese , Citocinas/fisiologia , Feminino , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/prevenção & controle , Mediadores da Inflamação/isolamento & purificação , Mediadores da Inflamação/metabolismo , Lipoproteínas/biossíntese , Lipoproteínas/genética , Lipoproteínas/isolamento & purificação , Glicoproteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Receptores de Superfície Celular/fisiologia , Receptor 2 Toll-Like , Receptor 4 Toll-Like , Receptores Toll-Like
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA