Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
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.
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
3.
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
4.
Eur J Immunol ; 45(10): 2821-33, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26138432

RESUMO

DCs are professional APCs playing a crucial role in the initiation of T-cell responses to combat infection. However, systemic bacterial infection with various pathogens leads to DC-depletion in humans and mice. The mechanisms of pathogen-induced DC-depletion remain poorly understood. Previously, we showed that mice infected with Yersinia enterocolitica (Ye) had impaired de novo DC-development, one reason for DC-depletion. Here, we extend these studies to gain insight into the molecular mechanisms of DC-depletion and the impact of different bacteria on DC-development. We show that the number of bone marrow (BM) hematopoietic progenitors committed to the DC lineage is reduced following systemic infection with different Gram-positive and Gram-negative bacteria. This is associated with a TLR4- and IFN-γ-signaling dependent increase of committed monocyte progenitors in the BM and mature monocytes in the spleen upon Ye-infection. Adoptive transfer experiments revealed that infection-induced monopoiesis occurs at the expense of DC-development. Our data provide evidence for a general response of hematopoietic progenitors upon systemic bacterial infections to enhance monocyte production, thereby increasing the availability of innate immune cells for pathogen control, whereas impaired DC-development leads to DC-depletion, possibly driving transient immunosuppression in bacterial sepsis.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Imunidade Inata , Mielopoese/imunologia , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Células Dendríticas/patologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Interferon gama/imunologia , Camundongos , Camundongos Knockout , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Yersiniose/patologia
5.
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
6.
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
7.
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
8.
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
9.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA