Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 997, 2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38307851

RESUMEN

In the context of continuous emergence of SARS-CoV-2 variants of concern (VOCs), one strategy to prevent the severe outcomes of COVID-19 is developing safe and effective broad-spectrum vaccines. Here, we present preclinical studies of a RBD vaccine derived from the Gamma SARS-CoV-2 variant adjuvanted with Alum. The Gamma-adapted RBD vaccine is more immunogenic than the Ancestral RBD vaccine in terms of inducing broader neutralizing antibodies. The Gamma RBD presents more immunogenic B-cell restricted epitopes and induces a higher proportion of specific-B cells and plasmablasts than the Ancestral RBD version. The Gamma-adapted vaccine induces antigen specific T cell immune responses and confers protection against Ancestral and Omicron BA.5 SARS-CoV-2 challenge in mice. Moreover, the Gamma RBD vaccine induces higher and broader neutralizing antibody activity than homologous booster vaccination in mice previously primed with different SARS-CoV-2 vaccine platforms. Our study indicates that the adjuvanted Gamma RBD vaccine is highly immunogenic and a broad-spectrum vaccine candidate.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , Humanos , Anticuerpos ampliamente neutralizantes , Vacunas contra la COVID-19 , COVID-19/prevención & control , Vacunas de Subunidad , Adyuvantes Inmunológicos , Epítopos de Linfocito B , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Glicoproteína de la Espiga del Coronavirus/genética
2.
Nat Commun ; 14(1): 4551, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507392

RESUMEN

A Gamma Variant RBD-based aluminum hydroxide adjuvanted vaccine called ARVAC CG was selected for a first in human clinical trial. Healthy male and female participants (18-55 years old) with a complete COVID-19-primary vaccine scheme were assigned to receive two intramuscular doses of either a low-dose or a high-dose of ARVAC CG. The primary endpoint was safety. The secondary objective was humoral immunogenicity. Cellular immune responses were studied as an exploratory objective. The trial was prospectively registered in PRIISA.BA (Registration Code 6564) and ANMAT and retrospectively registered in ClinicalTrials.gov (NCT05656508). Samples from participants of a surveillance strategy implemented by the Ministry of Health of the Province of Buenos Aires that were boosted with BNT162b2 were also analyzed to compare with the booster effect of ARVAC CG. ARVAC CG exhibits a satisfactory safety profile, a robust and broad booster response of neutralizing antibodies against the Ancestral strain of SARS-CoV-2 and the Gamma, Delta, Omicron BA.1 and Omicron BA.5 variants of concern and a booster effect on T cell immunity in individuals previously immunized with different COVID-19 vaccine platforms.


Asunto(s)
COVID-19 , Vacunas , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Adyuvantes Inmunológicos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Vacuna BNT162 , COVID-19/prevención & control , Vacunas contra la COVID-19/efectos adversos , SARS-CoV-2
3.
Comput Struct Biotechnol J ; 20: 5098-5114, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187929

RESUMEN

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.

4.
Front Immunol ; 13: 844837, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35296091

RESUMEN

In this work, we evaluated recombinant receptor binding domain (RBD)-based vaccine formulation prototypes with potential for further clinical development. We assessed different formulations containing RBD plus alum, AddaS03, AddaVax, or the combination of alum and U-Omp19: a novel Brucella spp. protease inhibitor vaccine adjuvant. Results show that the vaccine formulation composed of U-Omp19 and alum as adjuvants has a better performance: it significantly increased mucosal and systemic neutralizing antibodies in comparison to antigen plus alum, AddaVax, or AddaS03. Antibodies induced with the formulation containing U-Omp19 and alum not only increased their neutralization capacity against the ancestral virus but also cross-neutralized alpha, lambda, and gamma variants with similar potency. Furthermore, the addition of U-Omp19 to alum vaccine formulation increased the frequency of RBD-specific geminal center B cells and plasmablasts. Additionally, U-Omp19+alum formulation induced RBD-specific Th1 and CD8+ T-cell responses in spleens and lungs. Finally, this vaccine formulation conferred protection against an intranasal severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge of K18-hACE2 mice.


Asunto(s)
Adyuvantes Inmunológicos/metabolismo , Linfocitos B/inmunología , Proteínas de la Membrana Bacteriana Externa/metabolismo , Brucella/metabolismo , Vacunas contra la COVID-19/inmunología , COVID-19/inmunología , Centro Germinal/inmunología , SARS-CoV-2/fisiología , Compuestos de Alumbre/metabolismo , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales , Formación de Anticuerpos , Proteínas de la Membrana Bacteriana Externa/inmunología , Brucella/inmunología , Resistencia a la Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Glicoproteína de la Espiga del Coronavirus/inmunología
5.
Int J Med Microbiol ; 311(6): 151517, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34233227

RESUMEN

Dendritic cells (DCs) are critical in host defense against infection. DC depletion is an early event in the course of sepsis that may impair the host defense mechanisms. Here, we addressed whether DC depletion and dysfunction are pathogen-independent, mediated via pattern recognition receptors, and are due to impaired DC development upon systemic infection with the Gram-negative bacterium Escherichia coli and the Gram-positive pathogen Staphylococcus aureus. Infection with E. coli and S. aureus led to reduced numbers of splenic DC subsets and of DC progenitors in the bone marrow (BM) with this effect persisting significantly longer in mice infected with S. aureus than with E. coli. The reduction of DC subsets and their progenitors was mainly TLR-independent as was the infection-induced monopoiesis. Moreover, de novo DC development was impaired in mice infected with S. aureus, and BM cells from E. coli or S. aureus infected mice favored macrophage differentiation in vitro. As a consequence of reduced DC numbers and their reduced expression of MHC II less CD4+ and CD8+ T cells, especially Th1 and IFN-γ producing CD8+ T cells, could be detected in S. aureus compared to E. coli infected mice. These differences are reflected in the rapid killing of E. coli as opposed to an increase in bacterial load in S. aureus. In summary, our study supports the idea that systemic bacterial infections generally affect the number and development of DCs and thereby the T cell responses, but the magnitude is pathogen-dependent.


Asunto(s)
Sepsis , Infecciones Estafilocócicas , Animales , Linfocitos T CD8-positivos , Diferenciación Celular , Células Dendríticas , Escherichia coli , Ratones , Staphylococcus aureus
6.
J Pharm Sci ; 110(2): 707-718, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33058898

RESUMEN

Unlipidated outer membrane protein 19 (U-Omp19) is a novel mucosal adjuvant in preclinical development to be used in vaccine formulations. U-Omp19 holds two main properties, it is capable of inhibiting gastrointestinal and lysosomal peptidases, increasing the amount of co-administered antigen that reaches the immune inductive sites and its half-life inside cells, and it is able to stimulate antigen presenting cells in vivo. These activities enable U-Omp19 to enhance the adaptive immune response to co-administrated antigens. To characterize the stability of U-Omp19 we have performed an extensive analysis of its physicochemical and biological properties in a 3-year long-term stability study, and under potentially damaging freeze-thawing and lyophilization stress processes. Results revealed that U-Omp19 retains its full protease inhibitor activity, its monomeric state and its secondary structure even when stored in solution for 36 months or after multiple freeze-thawing cycles. Non-enzymatic hydrolysis resulted the major degradation pathway for storage in solution at 4 °C or room temperature which can be abrogated by lyophilization yet increasing protein tendency to form aggregates. This information will play a key role in the development of a stable formulation of U-Omp19, allowing an extended shelf-life during manufacturing, storage, and shipping of a future vaccine containing this pioneering adjuvant.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Vacunas , Adyuvantes Inmunológicos , Animales , Estabilidad de Medicamentos , Lipoproteínas , Ratones , Ratones Endogámicos BALB C
7.
Vaccine ; 38(32): 5027-5035, 2020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32536545

RESUMEN

Acute diarrhea disease caused by bacterial infections is a major global health problem. Enterotoxigenic Escherichia coli (ETEC) is one of the top causes of diarrhea-associated morbidity and mortality in young children and travelers to low-income countries. There are currently no licensed vaccines for ETEC. Induction of immunity at the site of entry of the bacteria is key to prevent infection. Current approaches to ETEC vaccines include a less toxic mutant form of E. coli heat-labile toxin (double-mutant heat-labile enterotoxin -dmLT-) with both antigenic and immunostimulatory properties. U-Omp19 is a protease inhibitor from Brucella spp. with immunostimulatory properties that has been used as oral adjuvant. In this work, we use U-Omp19 as adjuvant in an oral vaccine formulation against ETEC containing dmLT in outbred and inbred mice. To evaluate antigen dose sparing by U-Omp19 three different immunization protocols with three different doses of dmLT were evaluated. We demonstrated that U-Omp19 co-delivery increases anti-LT IgA in feces using a mid-dose of dmLT following a prime-boost protocol (after one or two boosts). Oral immunization with U-Omp19 induced protection against LT challenge when co-formulated with dmLT in CD-1 and BALB/c mice. Indeed, there was a significant increase in anti-LT IgG and IgA avidity after a single oral administration of dmLT plus U-Omp19 in comparison with dmLT delivered alone. Interestingly, sera from dmLT plus U-Omp19 vaccinated mice significantly neutralize LT effect on intestine inflammation in vivo compared with sera from the group immunized with dmLT alone. These results demonstrate the adjuvant capacity of U-Omp19 to increase dmLT immunogenicity by the oral route and support its use in an oral subunit vaccine formulation against ETEC.


Asunto(s)
Escherichia coli Enterotoxigénica , Infecciones por Escherichia coli , Proteínas de Escherichia coli , Vacunas contra Escherichia coli , Animales , Anticuerpos Antibacterianos , Toxinas Bacterianas , Brucella abortus , Enterotoxinas , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/genética , Ratones , Ratones Endogámicos BALB C
8.
Front Immunol ; 10: 1436, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31297115

RESUMEN

Pathogenic microorganisms confront several proteolytic events in the molecular interplay with their host, highlighting that proteolysis and its regulation play an important role during infection. Microbial inhibitors, along with their target endogenous/exogenous enzymes, may directly affect the host's defense mechanisms and promote infection. Omp19 is a Brucella spp. conserved lipoprotein anchored by the lipid portion in the Brucella outer membrane. Previous work demonstrated that purified unlipidated Omp19 (U-Omp19) has protease inhibitor activity against gastrointestinal and lysosomal proteases. In this work, we found that a Brucella omp19 deletion mutant is highly attenuated in mice when infecting by the oral route. This attenuation can be explained by bacterial increased susceptibility to host proteases met by the bacteria during establishment of infection. Omp19 deletion mutant has a cell division defect when exposed to pancreatic proteases that is linked to cell-cycle arrest in G1-phase, Omp25 degradation on the cell envelope and CtrA accumulation. Moreover, Omp19 deletion mutant is more susceptible to killing by macrophage derived microsomes than wt strain. Preincubation with gastrointestinal proteases led to an increased susceptibility of Omp19 deletion mutant to macrophage intracellular killing. Thus, in this work, we describe for the first time a physiological function of B. abortus Omp19. This activity enables Brucella to better thrive in the harsh gastrointestinal tract, where protection from proteolytic degradation can be a matter of life or death, and afterwards invade the host and bypass intracellular proteases to establish the chronic infection.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Brucella abortus/inmunología , Brucelosis/inmunología , Evasión Inmune , Lipoproteínas/inmunología , Inhibidores de Proteasas/inmunología , Animales , Antígenos Bacterianos/genética , Proteínas de la Membrana Bacteriana Externa/genética , Brucella abortus/genética , Brucelosis/genética , Brucelosis/patología , Femenino , Lipoproteínas/genética , Ratones , Ratones Endogámicos BALB C , Péptido Hidrolasas/genética , Péptido Hidrolasas/inmunología
9.
J Control Release ; 293: 158-171, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30496771

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos Bacterianos/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Mucosa Intestinal/metabolismo , Lipoproteínas/administración & dosificación , Inhibidores de Proteasas/administración & dosificación , Vacunas/administración & dosificación , Administración Oral , Animales , Células CACO-2 , Células Epiteliales/metabolismo , Femenino , Células HT29 , Humanos , Ratones Endogámicos BALB C
10.
J Infect Dis ; 217(8): 1257-1266, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29325043

RESUMEN

Brucellaceae are stealthy pathogens with the ability to survive and replicate in the host in the context of a strong immune response. This capacity relies on several virulence factors that are able to modulate the immune system and in their structural components that have low proinflammatory activities. Lipopolysaccharide (LPS), the main component of the outer membrane, is a central virulence factor of Brucella, and it has been well established that it induces a low inflammatory response. We describe here the identification and characterization of a novel periplasmic protein (RomA) conserved in alpha-proteobacteria, which is involved in the homeostasis of the outer membrane. A mutant in this gene showed several phenotypes, such as membrane defects, altered LPS composition, reduced adhesion, and increased virulence and inflammation. We show that RomA is involved in the synthesis of LPS, probably coordinating part of the biosynthetic complex in the periplasm. Its absence alters the normal synthesis of this macromolecule and affects the homeostasis of the outer membrane, resulting in a strain with a hyperinflammatory phenotype. Our results suggest that the proper synthesis of LPS is central to maximize virulence and minimize inflammation.


Asunto(s)
Proteínas Bacterianas/fisiología , Brucella/metabolismo , Brucelosis/microbiología , Lipopolisacáridos/biosíntesis , Animales , Brucella/patogenicidad , Gentamicinas , Inflamación/metabolismo , Ratones , Transporte de Proteínas , Virulencia
11.
Front Immunol ; 8: 171, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28261222

RESUMEN

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.

12.
J Immunol ; 196(10): 4014-29, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27084100

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Brucella/inmunología , Brucelosis/inmunología , Linfocitos T CD8-positivos/fisiología , Vacunas contra el Cáncer/inmunología , Catepsinas/metabolismo , Células Dendríticas/inmunología , Inmunoterapia/métodos , Lipoproteínas/metabolismo , Lisosomas/metabolismo , Melanoma/terapia , Animales , Antígenos de Neoplasias/inmunología , Reactividad Cruzada , Femenino , Activación de Linfocitos , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Vaccine ; 34(4): 430-437, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26707377

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Inmunidad Celular , Lipoproteínas/inmunología , Células TH1/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Antígenos de Protozoos/inmunología , Brucella abortus , Bovinos , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Proteínas Recombinantes/inmunología , Trypanosoma cruzi
14.
Eur J Immunol ; 45(10): 2821-33, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26138432

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Mielopoyesis/inmunología , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Animales , Células Dendríticas/patología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Yersiniosis/patología
15.
PLoS One ; 8(7): e69438, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861971

RESUMEN

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.


Asunto(s)
Adyuvantes Inmunológicos , Proteínas de la Membrana Bacteriana Externa/inmunología , Brucella/inmunología , Hipersensibilidad a la Leche/inmunología , Proteínas de la Leche/inmunología , Células TH1/inmunología , Células Th2/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antígenos/inmunología , Antígenos/metabolismo , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/química , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Bovinos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina E/inmunología , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Hipersensibilidad a la Leche/metabolismo , Mucosa Nasal/inmunología , Mucosa Nasal/metabolismo , Bazo/inmunología , Células TH1/metabolismo , Células Th2/metabolismo
16.
PLoS Pathog ; 8(2): e1002552, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22383883

RESUMEN

Dendritic cells (DCs) as professional antigen-presenting cells play an important role in the initiation and modulation of the adaptive immune response. However, their role in the innate immune response against bacterial infections is not completely defined. Here we have analyzed the role of DCs and their impact on the innate anti-bacterial host defense in an experimental infection model of Yersinia enterocolitica (Ye). We used CD11c-diphtheria toxin (DT) mice to deplete DCs prior to severe infection with Ye. DC depletion significantly increased animal survival after Ye infection. The bacterial load in the spleen of DC-depleted mice was significantly lower than that of control mice throughout the infection. DC depletion was accompanied by an increase in the serum levels of CXCL1, G-CSF, IL-1α, and CCL2 and an increase in the numbers of splenic phagocytes. Functionally, splenocytes from DC-depleted mice exhibited an increased bacterial killing capacity compared to splenocytes from control mice. Cellular studies further showed that this was due to an increased production of reactive oxygen species (ROS) by neutrophils. Adoptive transfer of neutrophils from DC-depleted mice into control mice prior to Ye infection reduced the bacterial load to the level of Ye-infected DC-depleted mice, suggesting that the increased number of phagocytes with additional ROS production account for the decreased bacterial load. Furthermore, after incubation with serum from DC-depleted mice splenocytes from control mice increased their bacterial killing capacity, most likely due to enhanced ROS production by neutrophils, indicating that serum factors from DC-depleted mice account for this effect. In summary, we could show that DC depletion triggers phagocyte accumulation in the spleen and enhances their anti-bacterial killing capacity upon bacterial infection.


Asunto(s)
Células Dendríticas/patología , Inmunidad Innata/fisiología , Fagocitos/fisiología , Yersiniosis/inmunología , Yersinia enterocolitica/inmunología , Traslado Adoptivo , Animales , Bacterias/inmunología , Separación Celular , Células Cultivadas , Femenino , Homeostasis/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/trasplante , Fagocitos/inmunología , Regulación hacia Arriba/inmunología , Yersiniosis/patología , Yersiniosis/terapia
17.
PLoS One ; 6(1): e16203, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21264260

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/uso terapéutico , Proteínas de la Membrana Bacteriana Externa/uso terapéutico , Vacuna contra la Brucelosis/inmunología , Brucella abortus/inmunología , Interleucina-17/inmunología , Lipoproteínas/uso terapéutico , Inmunidad Adaptativa , Animales , Vacuna contra la Brucelosis/administración & dosificación , Brucelosis/prevención & control , Linfocitos T CD4-Positivos/inmunología , Ratones , Células TH1/inmunología , Células Th17/inmunología
18.
J Immunol ; 184(9): 5200-12, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20351187

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Vacuna contra la Brucelosis/inmunología , Brucelosis/prevención & control , Células Dendríticas/inmunología , Interacciones Huésped-Patógeno/inmunología , Células TH1/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Administración Oral , Animales , Antígenos Bacterianos/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/administración & dosificación , Proteínas de la Membrana Bacteriana Externa/biosíntesis , Proteínas de la Membrana Bacteriana Externa/genética , Vacuna contra la Brucelosis/administración & dosificación , Brucelosis/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Adyuvante de Freund/administración & dosificación , Interacciones Huésped-Patógeno/genética , Inmunidad Celular , Inyecciones Intraperitoneales , Lípidos/administración & dosificación , Lipoproteínas/administración & dosificación , Lipoproteínas/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células TH1/microbiología , Nicotiana/genética , Nicotiana/inmunología
19.
Am J Pathol ; 176(3): 1323-38, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20093491

RESUMEN

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.


Asunto(s)
Apoptosis , Astrocitos/microbiología , Astrocitos/patología , Brucella abortus/fisiología , Mediadores de Inflamación/metabolismo , Animales , Antígenos Bacterianos/farmacología , Apoptosis/efectos de los fármacos , Astrocitos/enzimología , Astrocitos/metabolismo , Proteínas de la Membrana Bacteriana Externa/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/microbiología , Encéfalo/patología , Brucella abortus/efectos de los fármacos , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas/metabolismo , Femenino , Calor , Inmunohistoquímica , Lipopolisacáridos/farmacología , Lipoproteínas/farmacología , Ratones , Ratones Endogámicos BALB C , Microglía/efectos de los fármacos , Microglía/microbiología , Microglía/patología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Microbes Infect ; 11(6-7): 689-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19376263

RESUMEN

Human brucellosis is caused by infection with certain species of the genus Brucella and is characterized by bacterial persistence and inflammation of many host tissues. Neutrophils are one of the predominant cell types present in the infiltrate of these inflamed tissues, and due to their potential effect on the inflammatory response and tissue damage, direct activation of neutrophils by Brucella abortus might contribute to the pathology associated with human brucellosis. B. abortus expresses outer membrane lipoproteins (Omp) with inflammatory properties on a variety of cell types. This study examines the effect of B. abortus and its lipoproteins on neutrophil functions. B. abortus induced an increment in CD35 and CD11b expression and a decline in CD62L accompanied by IL-8 secretion, a response compatible with neutrophil activation. B. abortus lipoprotein Omp19 (L-Omp19), but not its unlipidated form, mimicked the changes associated with neutrophil activation induced by B. abortus. L-Omp19 primed neutrophils for oxidative burst as well as promoted neutrophil migration and prolonged neutrophil survival. Thus, Brucella lipoproteins possess pro-inflammatory properties that could contribute to the localize tissue injury and inflammation by direct activation of neutrophils. Data presented here, together with our previous results implicate Brucella lipoproteins in the pathogenesis of human brucellosis.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas de la Membrana Bacteriana Externa/inmunología , Brucella abortus/inmunología , Lipoproteínas/inmunología , Neutrófilos/inmunología , Antígeno CD11b/análisis , Ensayos de Migración de Leucocitos , Movimiento Celular , Supervivencia Celular , Células Cultivadas , Humanos , Interleucina-8/metabolismo , Selectina L/análisis , Neutrófilos/química , Receptores de Complemento 3b/análisis , Estallido Respiratorio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA