Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 13: 995327, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36263034

RESUMO

Re-emerging zoonotic pathogen Brucella spp. continues to impact developing countries and persists in expanding populations of wildlife species in the US, constantly threatening infection of our domestic herds. The development of improved animal and human vaccines remains a priority. In this study, immunity to a novel live attenuated B. melitensis strain, termed znBM-mC, was characterized. An oral prime, intranasal (IN) boost strategy conferred exquisite protection against pulmonary challenge, with wild-type (wt) B. melitensis providing nearly complete protection in the lungs and spleens from brucellae colonization. Vaccination with znBM-mC showed an IFN-γ+ CD8+ T-cell bias in the lungs as opposed to Rev 1-vaccinated mice showing IFN-γ+ CD4+ T-cell inclination. Lung CD4+ and CD8+ effector memory T cells (TEMs) increased over 200-fold; and lung CD4+ and CD8+ resident memory T cells (TRMs) increased more than 250- and 150-fold, respectively. These T cells served as the primary producers of IFN-γ in the lungs, which was essential for vaccine clearance and the predominant cytokine generated pre-and post-challenge with wt B. melitensis 16M; znBM-mC growth could not be arrested in IFN-γ-/- mice. Increases in lung TNF-α and IL-17 were also induced, with IL-17 being mostly derived from CD4+ T cells. Vaccination of CD4-/-, CD8-/-, and B6 mice with znBM-mC conferred full protection in the lungs and spleens post-pulmonary challenge with virulent B. melitensis; vaccination of IL-17-/- mice resulted in the protection of the lungs, but not the spleen. These data demonstrate the efficacy of mucosal vaccine administration for the generation of protective memory T cells against wt B. melitensis.


Assuntos
Vacina contra Brucelose , Brucella melitensis , Brucelose , Humanos , Camundongos , Animais , Brucella melitensis/genética , Interleucina-17 , Brucelose/prevenção & controle , Fator de Necrose Tumoral alfa , Vacinação , Linfócitos T CD8-Positivos , Subpopulações de Linfócitos T , Linfócitos T CD4-Positivos
2.
PLoS Pathog ; 17(5): e1009465, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33956909

RESUMO

Salmonella Typhimurium is a causative agent of nontyphoidal salmonellosis, for which there is a lack of a clinically approved vaccine in humans. As an intracellular pathogen, Salmonella impacts many cellular pathways. However, the intercellular communication mechanism facilitated by host-derived small extracellular vesicles (EVs), such as exosomes, is an overlooked aspect of the host responses to this infection. We used a comprehensive proteome-based network analysis of exosomes derived from Salmonella-infected macrophages to identify host molecules that are trafficked via these EVs. This analysis predicted that the host-derived small EVs generated during macrophage infection stimulate macrophages and promote activation of T helper 1 (Th1) cells. We identified that exosomes generated during infection contain Salmonella proteins, including unique antigens previously shown to stimulate protective immune responses against Salmonella in murine studies. Furthermore, we showed that host EVs formed upon infection stimulate a mucosal immune response against Salmonella infection when delivered intranasally to BALB/c mice, a route of antigen administration known to initiate mucosal immunity. Specifically, the administration of these vesicles to animals stimulated the production of anti-Salmonella IgG antibodies, such as anti-OmpA antibodies. Exosomes also stimulated antigen-specific cell-mediated immunity. In particular, splenic mononuclear cells isolated from mice administered with exosomes derived from Salmonella-infected antigen-presenting cells increased CD4+ T cells secreting Th1-type cytokines in response to Salmonella antigens. These results demonstrate that small EVs, formed during infection, contribute to Th1 cell bias in the anti-Salmonella responses. Collectively, this study helps to unravel the role of host-derived small EVs as vehicles transmitting antigens to induce Th1-type immunity against Gram-negative bacteria. Understanding the EV-mediated defense mechanisms will allow the development of future approaches to combat bacterial infections.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Vesículas Extracelulares/imunologia , Imunidade Celular/imunologia , Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Células Th1/imunologia , Animais , Feminino , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia
3.
J Virol ; 93(14)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31068425

RESUMO

The benefits of mucosal vaccines over injected vaccines are difficult to ascertain, since mucosally administered vaccines often induce serum antibody responses of lower magnitude than those induced by injected vaccines. This study aimed to determine if mucosal vaccination using a modified vaccinia virus Ankara expressing human immunodeficiency virus type 1 (HIV-1) gp120 (MVAgp120) prime and a HIV-1 gp120 protein boost could be optimized to induce serum antibody responses similar to those induced by an intramuscularly (i.m.) administered MVAgp120 prime/gp120 boost to allow comparison of an i.m. immunization regimen to a mucosal vaccination regimen for the ability to protect against a low-dose rectal simian-human immunodeficiency virus (SHIV) challenge. A 3-fold higher antigen dose was required for intranasal (i.n.) immunization with gp120 to induce serum anti-gp120 IgG responses not significantly different than those induced by i.m. immunization. gp120 fused to the adenovirus type 2 fiber binding domain (gp120-Ad2F), a mucosal targeting ligand, exhibited enhanced i.n. immunogenicity compared to gp120. MVAgp120 was more immunogenic after i.n. delivery than after gastric or rectal delivery. Using these optimized vaccines, an i.n. MVAgp120 prime/combined i.m. (gp120) and i.n. (gp120-Ad2F) boost regimen (i.n./i.m.-plus-i.n.) induced serum anti-gp120 antibody titers similar to those induced by the intramuscular prime/boost regimen (i.m./i.m.) in rabbits and nonhuman primates. Despite the induction of similar systemic anti-HIV-1 antibody responses, neither the i.m./i.m. nor the i.n./i.m.-plus-i.n. regimen protected against a repeated low-dose rectal SHIV challenge. These results demonstrate that immunization regimens utilizing the i.n. route are able to induce serum antigen-specific antibody responses similar to those induced by systemic immunization.IMPORTANCE Mucosal vaccination is proposed as a method of immunization able to induce protection against mucosal pathogens that is superior to protection provided by parenteral immunization. However, mucosal vaccination often induces serum antigen-specific immune responses of lower magnitude than those induced by parenteral immunization, making the comparison of mucosal and parenteral immunization difficult. We identified vaccine parameters that allowed an immunization regimen consisting of an i.n. prime followed by boosters administered by both i.n. and i.m. routes to induce serum antibody responses similar to those induced by i.m. prime/boost vaccination. Additional studies are needed to determine the potential benefit of mucosal immunization for HIV-1 and other mucosally transmitted pathogens.


Assuntos
Vacinas contra a AIDS/imunologia , Anticorpos Anti-HIV/imunologia , Proteína gp120 do Envelope de HIV/imunologia , HIV-1/imunologia , Imunização Secundária , Vacinação , Vaccinia virus/imunologia , Vacinas contra a AIDS/genética , Administração Intranasal , Animais , Proteína gp120 do Envelope de HIV/genética , HIV-1/genética , Humanos , Imunidade nas Mucosas , Camundongos , Vaccinia virus/genética
4.
Vaccine ; 34(44): 5290-5297, 2016 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-27639282

RESUMO

There remains a need for an improved livestock vaccine for brucellosis since conventional vaccines are only ∼70% efficacious, making some vaccinated animals susceptible to Brucella infections. To address this void, a vaccine capable of evoking protective immunity, while still being sufficiently attenuated to produce minimal disease, is sought. In this pursuit, the ΔnorD ΔznuA B. abortus-lacZ (termed as znBAZ) was developed to be devoid of functional norD and znuA B. abortus genes, and to contain the lacZ as a marker gene. The results show that znBAZ is highly attenuated in mouse and human macrophages, and completely cleared from mouse spleens within eight weeks post-vaccination. Producing less splenic inflammation, znBAZ is significantly more protective than the conventional RB51 vaccine by more than four orders of magnitude. Vaccination with znBAZ elicits elevated numbers of IFN-γ+, TNF-α+, and polyfunctional IFN-γ+ TNF-α+ CD4+ and CD8+ T cells in contrast to RB51-vaccinated mice, which show reduced numbers of proinflammatory cytokine-producing T cells. These results demonstrate that znBAZ is a highly efficacious vaccine candidate capable of eliciting diverse T cell subsets that confer protection against parenteral challenge with virulent, wild-type B. abortus.


Assuntos
Vacina contra Brucelose/genética , Vacina contra Brucelose/imunologia , Brucella abortus/genética , Brucelose/prevenção & controle , Animais , Vacina contra Brucelose/administração & dosagem , Brucella abortus/patogenicidade , Brucella abortus/fisiologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Genes Bacterianos , Imunidade Celular , Interferon gama/biossíntese , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Baço/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Virulência
5.
Immunol Cell Biol ; 94(5): 496-508, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26752510

RESUMO

Brucellosis remains a significant zoonotic threat worldwide. Humans and animals acquire infection via their oropharynx and upper respiratory tract following oral or aerosol exposure. After mucosal infection, brucellosis develops into a systemic disease. Mucosal vaccination could offer a viable alternative to conventional injection practices to deter disease. Using a nasal vaccination approach, the ΔznuA B. melitensis was found to confer potent protection against pulmonary Brucella challenge, and reduce colonization of spleens and lungs by more than 2500-fold, with >50% of vaccinated mice showing no detectable brucellae. Furthermore, 10-fold more brucellae-specific, interferon-γ (IFN-γ)-producing CD8(+) T cells than CD4(+) T cells were induced in the spleen and respiratory lymph nodes. Evaluation of pulmonary and splenic CD8(+) T cells from mice vaccinated with ΔznuA B. melitensis revealed that these expressed an activated effector memory (CD44(hi)CD62L(lo)CCR7(lo)) T cells producing elevated levels of IFN-γ, tumor necrosis factor-α, perforin and granzyme B. To assess the relative importance of these increased numbers of CD8(+) T cells, CD8(-/-) mice were challenged with virulent B. melitensis, and they showed markedly increased bacterial loads in organs in contrast to similarly challenged CD4(-/-) mice. Only ΔznuA B. melitensis- and Rev-1-vaccinated CD4(-/-) and wild-type mice, not CD8(-/-) mice, were completely protected against Brucella challenge. Determination of cytokines responsible for conferring protection showed the relative importance of IFN-γ, but not interleukin-17 (IL-17). Unlike wild-type (wt) mice, IL-17 was greatly induced in IFN-γ(-/-) mice, but IL-17 could not substitute for IFN-γ's protection, although an increase in brucellae dissemination was observed upon in vivo IL-17 neutralization. These results show that nasal ΔznuA B. melitensis vaccination represents an attractive means to stimulate systemic and mucosal immune protection via CD8(+) T-cell engagement.


Assuntos
Brucella melitensis/fisiologia , Brucelose/imunologia , Brucelose/prevenção & controle , Linfócitos T CD8-Positivos/imunologia , Mucosa Nasal/imunologia , Mucosa Nasal/microbiologia , Vacinação , Animais , Vacinas Bacterianas/imunologia , Brucelose/microbiologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Feminino , Memória Imunológica , Mediadores da Inflamação/metabolismo , Interferon gama , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Mucosa Nasal/patologia , Baço/patologia
6.
PLoS One ; 10(1): e0117825, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25629976

RESUMO

Autoimmune diseases arise from the loss of tolerance to self, and because the etiologies of such diseases are largely unknown, symptomatic treatments rely on anti-inflammatory and analgesic agents. Tolerogenic treatments that can reverse disease are preferred, but again, often thwarted by not knowing the responsible auto-antigens (auto-Ags). Hence, a viable alternative to stimulating regulatory T cells (Tregs) is to induce bystander tolerance. Colonization factor antigen I (CFA/I) has been shown to evoke bystander immunity and to hasten Ag-specific Treg development independent of auto-Ag. To translate in treating human autoimmune diseases, the food-based Lactococcus was engineered to express CFA/I fimbriae, and Lactococcus-CFA/I fermented milk fed to arthritic mice proved highly efficacious. Protection occurred via CD39+ Tregs producing TGF-ß and IL-10 to potently suppress TNF-α production and neutrophil influx into the joints. Thus, these data demonstrate the feasibility of oral nutraceuticals for treating arthritis, and potency of protection against arthritis was improved relative to that obtained with Salmonella-CFA/I.


Assuntos
Antígenos CD/metabolismo , Apirase/metabolismo , Artrite Experimental/tratamento farmacológico , Suplementos Nutricionais , Interleucina-10/metabolismo , Leite/imunologia , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Efeito Espectador/imunologia , Tolerância Imunológica/imunologia , Camundongos , Linfócitos T Reguladores/imunologia , Fator de Necrose Tumoral alfa/imunologia
7.
Infect Immun ; 81(9): 3099-105, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23774604

RESUMO

Francisella tularensis is a highly infectious intracellular bacterium that causes the zoonotic infection tularemia. While much literature exists on the host response to F. tularensis infection, the vast majority of work has been conducted using attenuated strains of Francisella that do not cause disease in humans. However, emerging data indicate that the protective immune response against attenuated F. tularensis versus F. tularensis type A differs. Several groups have recently reported that interleukin-17 (IL-17) confers protection against the live vaccine strain (LVS) of Francisella. While we too have found that IL-17Rα(-/-) mice are more susceptible to F. tularensis LVS infection, our studies, using a virulent type A strain of F. tularensis (SchuS4), indicate that IL-17Rα(-/-) mice display organ burdens and pulmonary gamma interferon (IFN-γ) responses similar to those of wild-type mice following infection. In addition, oral LVS vaccination conferred equivalent protection against pulmonary challenge with SchuS4 in both IL-17Rα(-/-) and wild-type mice. While IFN-γ was found to be critically important for survival in a convalescent model of SchuS4 infection, IL-17 neutralization from either wild-type or IFN-γ(-/-) mice had no effect on morbidity or mortality in this model. IL-17 protein levels were also higher in the lungs of mice infected with the LVS rather than F. tularensis type A, while IL-23p19 mRNA expression was found to be caspase-1 dependent in macrophages infected with LVS but not SchuS4. Collectively, these results demonstrate that IL-17 is dispensable for host immunity to type A F. tularensis infection, and that induced and protective immunity differs between attenuated and virulent strains of F. tularensis.


Assuntos
Vacinas Bacterianas/imunologia , Vacinas Bacterianas/farmacologia , Francisella tularensis/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Tularemia/genética , Tularemia/microbiologia , Animais , Caspase 1/genética , Caspase 1/imunologia , Feminino , Francisella tularensis/patogenicidade , Interferon gama/genética , Interferon gama/imunologia , Subunidade p19 da Interleucina-23/genética , Subunidade p19 da Interleucina-23/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Interleucina-17/imunologia , Tularemia/imunologia , Tularemia/prevenção & controle , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/farmacologia
8.
BMC Complement Altern Med ; 13: 149, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23806004

RESUMO

BACKGROUND: Extracts of leaves from Clerodendrum have been used for centuries to treat a variety of medicinal problems in tropical Africa. However, little is known about the high-molecular weight active components conferring therapeutic properties to these extracts. METHODS: Polysaccharides from the leaves of Clerodendrum splendens were extracted and fractionated by ion exchange and size-exclusion chromatography. Molecular weight determination, sugar analysis, degree of methyl esterification, and other chemical characterization of the fractions were performed. Immunomodulatory activity of the fractions was evaluated by determining their ability to induce monocyte/macrophage nitric oxide (NO), cytokine production, and mitogen-activated protein kinase (MAPK) phosphorylation. Experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice, and severity of EAE was monitored in mice treated with intraperitoneal (i.p.) injections of the most active polysaccharide fraction. Lymph nodes (LN) and spleen were harvested, and levels of cytokines in supernatants from LN cells and splenocytes challenged with myelin oligodendrocyte glycoprotein peptide were determined. RESULTS: Fractions containing type II arabinogalactan had potent immunomodulatory activity. Specifically, the high-molecular weight sub-fraction CSP-AU1 (average of 38.5 kDa) induced NO and cytokine [interleukin (IL)-1α, -1ß, -6, -10, tumor necrosis factor (TNF; designated previously as TNF-α), and granulocyte macrophage-colony stimulating factor (GM-CSF)] production by human peripheral blood mononuclear cells (PBMCs) and monocyte/macrophages. CSP-AU1-induced secretion of TNF was prevented by Toll-like receptor 4 (TLR4) antagonist LPS-RS, indicating a role for TLR4 signaling. Treatment with CSP-AU1 also induced phosphorylation of a number of MAPKs in human PBMC and activated AP-1/NF-κB. In vivo treatment of mice with CSP-AU1 and CSP-NU1 resulted in increased serum IL-6, IL-10, TNF, monocyte chemoattractant protein-1 (MCP-1), macrophage inflammatory protein (MIP)-1α/CCL3, and MIP-1ß/CCL4. CSP-AU1 treatment of mice with EAE (50 mg/kg, i.p., daily, 13 days) resulted in significantly reduced disease severity in this experimental model of multiple sclerosis. Levels of IL-13, TNF, interferon (IFN)-γ, IL-17, and GM-CSF were also significantly decreased, whereas transforming growth factor (TGF)-ß was increased in LN cells from CSP-AU1-treated EAE mice. CONCLUSIONS: Polysaccharide CSP-AU1 is a potent natural innate immunomodulator with a broad spectrum of agonist activity in vitro and immunosupressive properties after chronic administration in vivo.


Assuntos
Clerodendrum/química , Encefalomielite Autoimune Experimental/tratamento farmacológico , Fatores Imunológicos/administração & dosagem , Polissacarídeos/administração & dosagem , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/imunologia , Citocinas/genética , Citocinas/imunologia , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Feminino , Humanos , Fatores Imunológicos/isolamento & purificação , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Polissacarídeos/isolamento & purificação , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia
9.
PLoS One ; 8(4): e62500, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23614052

RESUMO

The recognized necessity for new antigen delivery carriers with the capacity to boost, modulate and prolong neutralizing immune responses prompted our approach, in which we describe a multifunctional nanocarrier consisting of an oily nanocontainer protected by a polymeric shell made of chitosan (CS), named CS nanocapsules (CSNC). The CS shell can associate the antigen on its surface, whereas the oily core might provide additional immunostimulating properties. In this first characterization of the system, we intended to study the influence of different antigen organizations on the nanocarrier's surface (using the recombinant hepatitis B surface antigen -rHBsAg- as a model antigen) on their long-term immunopotentiating effect, without any additional immunostimulant. Thus, two prototypes of antigen-loaded CSNC (CSNC+ and CSNC-), exhibiting similar particle size (200 nm) and high antigen association efficiency (>80%), were developed with different surface composition (polymer/antigen ratios) and surface charge (positive/negative, respectively). The biological evaluation of these nanovaccines evidenced the superiority of the CSNC+ as compared to CSNC- and alum-rHBsAg in terms of neutralizing antibody responses, following intramuscular vaccination. Moreover, a single dose of CSNC+ led to similar IgG levels to the positive control. The IgG1/IgG2a ratio suggested a mixed Th1/Th2 response elicited by CSNC+, in contrast to the typical Th2-biased response of alum. Finally, CSNC+ could be freeze-dried without altering its physicochemical properties and adjuvant effect in vivo. In conclusion, the evaluation of CSNC+ confirms its interesting features for enhancing, prolonging and modulating the type of immune response against subunit antigens, such as rHBsAg.


Assuntos
Quitosana/química , Portadores de Fármacos/química , Imunização/métodos , Nanocápsulas/química , Óleos/química , Vacinas/imunologia , Animais , Feminino , Liofilização , Antígenos da Hepatite B/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo , Vacinas/administração & dosagem , Vacinas/química , Água/química
10.
PLoS One ; 7(11): e49491, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23152913

RESUMO

Glycyrrhizin, an abundant bioactive component of the medicinal licorice root is rapidly metabolized by gut commensal bacteria into 18ß-glycyrrhetinic acid (GRA). Either or both of these compounds have been shown to have antiviral, anti-hepatotoxic, anti-ulcerative, anti-tumor, anti-allergenic and anti-inflammatory activity in vitro or in vivo. In this study, the ability of GRA to modulate immune responses at the small intestinal mucosa when delivered orally was investigated. Analysis of cytokine transcription in duodenal and ileal tissue in response to GRA treatment revealed a pattern of chemokine and chemokine receptor gene expression predictive of B cell recruitment to the gut. Consistent with this finding, GRA induced increases in CD19(+) B cells in the lamina propria and B220(+) B cell aggregates framed by CD11c(+) dendritic cells in structures resembling isolated lymphoid follicles (ILF). Using a mouse model of rotavirus infection, GRA reduced the duration of viral antigen shedding, and endpoint serum antibody titers were higher in GRA-treated animals. Together the data suggest GRA delivered orally augments lymphocyte recruitment to the intestinal mucosa and induces maturation of B cell-rich ILF independently of ectopic antigenic stimulus. These results provide further support a role for dietary ligands in modulation of dynamic intestinal lymphoid tissue.


Assuntos
Ácido Glicirretínico/análogos & derivados , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Tecido Linfoide/crescimento & desenvolvimento , Rotavirus/fisiologia , Eliminação de Partículas Virais/efeitos dos fármacos , Administração Oral , Animais , Antígenos CD19/metabolismo , Antígenos Virais/metabolismo , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Complexo CD3 , Agregação Celular/efeitos dos fármacos , Contagem de Células , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glicirretínico/administração & dosagem , Ácido Glicirretínico/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Intestino Delgado/virologia , Antígenos Comuns de Leucócito/metabolismo , Ligantes , Tecido Linfoide/efeitos dos fármacos , Tecido Linfoide/patologia , Tecido Linfoide/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nódulos Linfáticos Agregados/efeitos dos fármacos , Nódulos Linfáticos Agregados/patologia , Nódulos Linfáticos Agregados/virologia , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Rotavirus/efeitos dos fármacos , Rotavirus/imunologia , Infecções por Rotavirus/imunologia , Infecções por Rotavirus/patologia , Sindecana-1/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Transcrição Gênica/efeitos dos fármacos
11.
Mol Pharmacol ; 81(6): 832-45, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22434859

RESUMO

In efforts to identify novel small molecules with anti-inflammatory properties, we discovered a unique series of tetracyclic indenoquinoxaline derivatives that inhibited lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 activation. Compound IQ-1 (11H-indeno[1,2-b]quinoxalin-11-one oxime) was found to be a potent, noncytotoxic inhibitor of pro-inflammatory cytokine [interleukin (IL)-1α, IL-1ß, IL-6, IL-10, tumor necrosis factor (TNF)-α, interferon-γ, and granulocyte-macrophage colony-stimulating factor] and nitric oxide production by human and murine monocyte/macrophages. Three additional potent inhibitors of cytokine production were identified through further screening of IQ-1 analogs. The sodium salt of IQ-1 inhibited LPS-induced TNF-α and IL-6 production in MonoMac-6 cells with IC(50) values of 0.25 and 0.61 µM, respectively. Screening of 131 protein kinases revealed that derivative IQ-3 [11H-indeno[1,2-b]quinoxalin-11-one-O-(2-furoyl)oxime]was a specific inhibitor of the c-Jun N-terminal kinase (JNK) family, with preference for JNK3. This compound, as well as IQ-1 and three additional oxime indenoquinoxalines, were found to be high-affinity JNK inhibitors with nanomolar binding affinity and ability to inhibit c-Jun phosphorylation. Furthermore, docking studies showed that hydrogen bonding interactions of the active indenoquinoxalines with Asn152, Gln155, and Met149 of JNK3 played an important role in enzyme binding activity. Finally, we showed that the sodium salt of IQ-1 had favorable pharmacokinetics and inhibited the ovalbumin-induced CD4(+) T-cell immune response in a murine delayed-type hypersensitivity model in vivo. We conclude that compounds with an indenoquinoxaline nucleus can serve as specific small-molecule modulators for mechanistic studies of JNKs as well as a potential leads for the development of anti-inflammatory drugs.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Oximas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Quinoxalinas/farmacologia , Animais , Western Blotting , Células Cultivadas , Citocinas/antagonistas & inibidores , Ensaio de Imunoadsorção Enzimática , Humanos , Ligação de Hidrogênio , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Modelos Moleculares , Oximas/química , Fosforilação , Inibidores de Proteínas Quinases/química , Quinoxalinas/química
12.
PLoS Pathog ; 8(3): e1002587, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22438809

RESUMO

Pulmonary Francisella tularensis and Burkholderia pseudomallei infections are highly lethal in untreated patients, and current antibiotic regimens are not always effective. Activating the innate immune system provides an alternative means of treating infection and can also complement antibiotic therapies. Several natural agonists were screened for their ability to enhance host resistance to infection, and polysaccharides derived from the Acai berry (Acai PS) were found to have potent abilities as an immunotherapeutic to treat F. tularensis and B. pseudomallei infections. In vitro, Acai PS impaired replication of Francisella in primary human macrophages co-cultured with autologous NK cells via augmentation of NK cell IFN-γ. Furthermore, Acai PS administered nasally before or after infection protected mice against type A F. tularensis aerosol challenge with survival rates up to 80%, and protection was still observed, albeit reduced, when mice were treated two days post-infection. Nasal Acai PS administration augmented intracellular expression of IFN-γ by NK cells in the lungs of F. tularensis-infected mice, and neutralization of IFN-γ ablated the protective effect of Acai PS. Likewise, nasal Acai PS treatment conferred protection against pulmonary infection with B. pseudomallei strain 1026b. Acai PS dramatically reduced the replication of B. pseudomallei in the lung and blocked bacterial dissemination to the spleen and liver. Nasal administration of Acai PS enhanced IFN-γ responses by NK and γδ T cells in the lungs, while neutralization of IFN-γ totally abrogated the protective effect of Acai PS against pulmonary B. pseudomallei infection. Collectively, these results demonstrate Acai PS is a potent innate immune agonist that can resolve F. tularensis and B. pseudomallei infections, suggesting this innate immune agonist has broad-spectrum activity against virulent intracellular pathogens.


Assuntos
Arecaceae/química , Imunidade Inata/efeitos dos fármacos , Melioidose/prevenção & controle , Pneumonia/tratamento farmacológico , Polissacarídeos/farmacologia , Tularemia/prevenção & controle , Administração Intranasal , Animais , Burkholderia pseudomallei/efeitos dos fármacos , Burkholderia pseudomallei/imunologia , Modelos Animais de Doenças , Feminino , Francisella tularensis/efeitos dos fármacos , Francisella tularensis/imunologia , Interferon gama/metabolismo , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Longevidade/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Melioidose/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Extratos Vegetais/administração & dosagem , Extratos Vegetais/farmacologia , Pneumonia/imunologia , Pneumonia/microbiologia , Polissacarídeos/administração & dosagem , Polissacarídeos/isolamento & purificação , Tularemia/imunologia
13.
Int Immunol ; 24(2): 117-28, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22207133

RESUMO

Sublingual (s.l.) vaccination is an efficient way to induce elevated levels of systemic and mucosal immune responses. To mediate mucosal uptake, ovalbumin (OVA) was genetically fused to adenovirus 2 fiber protein (OVA-Ad2F) to assess whether s.l. immunization was as effective as an alternative route of vaccination. Ad2F-delivered vaccines were efficiently taken up by dendritic cells and migrated mostly to submaxillary gland lymph nodes, which could readily stimulate OVA-specific CD4(+) T cells. OVA-Ad2F + cholera toxin (CT)-immunized mice elicited significantly higher OVA-specific serum IgG, IgA and mucosal IgA antibodies among the tested immunization groups. These were supported by elevated OVA-specific IgG and IgA antibody-forming cells. A mixed T(h)-cell response was induced as evident by the enhanced IL-4, IL-10, IFN-γ and TNF-α-specific cytokine-forming cells. To assess whether this approach can stimulate neutralizing antibodies, immunizations were performed with the protein encumbering the ß-trefoil domain of C-terminus heavy chain (Hcßtre) from botulinum neurotoxin A (BoNT/A) as well as when fused to Ad2F. Hcßtre-Ad2F + CT-dosed mice showed the greatest serum IgG, IgA and mucosal IgA titers among the immunization groups. Hcßtre-Ad2F alone also induced elevated antibody production in contrast to Hcßtre alone. Plasma from Hcßtre + CT- and Hcßtre-Ad2F + CT-immunized groups neutralized BoNT/A and protected mice from BoNT/A intoxication. Most importantly, Hcßtre-Ad2F + CT-immunized mice were protected from BoNT/A intoxication relative to Hcßtre + CT-immunized mice, which only showed ∼60% protection. This study shows that s.l. immunization with Ad2F-based vaccines is effective in conferring protective immunity.


Assuntos
Anticorpos Neutralizantes , Toxinas Botulínicas Tipo A/imunologia , Botulismo/imunologia , Proteínas do Capsídeo/imunologia , Clostridium botulinum/imunologia , Administração Sublingual , Animais , Anticorpos Antibacterianos/biossíntese , Anticorpos Neutralizantes/biossíntese , Formação de Anticorpos , Toxinas Botulínicas Tipo A/genética , Toxinas Botulínicas Tipo A/metabolismo , Toxinas Botulínicas Tipo A/toxicidade , Botulismo/complicações , Botulismo/genética , Botulismo/terapia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Clostridium botulinum/patogenicidade , Humanos , Imunoglobulina A/metabolismo , Imunoglobulina G/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas Recombinantes de Fusão/genética , Insuficiência Respiratória/etiologia , Insuficiência Respiratória/prevenção & controle , Vacinação
14.
J Autoimmun ; 37(4): 328-41, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22018711

RESUMO

Natural killer (NK) cells and dendritic cells (DCs) have been shown to link the innate and adaptive immune systems. Likewise, a new innate cell subset, interferon-producing killer DCs (IKDCs), shares phenotypic and functional characteristics with both DCs and NK cells. Here, we show IKDCs play an essential role in the resolution of experimental autoimmune encephalomyelitis (EAE) upon treatment with the tolerizing agent, myelin oligodendrocyte glycoprotein (MOG), genetically fused to reovirus protein σ1 (termed MOG-pσ1). Activated IKDCs were recruited subsequent MOG-pσ1 treatment of EAE, and disease resolution was abated upon NK1.1 cell depletion. These IKDCs were able to kill activated CD4(+) T cells and mature dendritic DCs, thus, contributing to EAE remission. In addition, IKDCs were responsible for MOG-pσ1-mediated MOG-specific regulatory T cell recruitment to the CNS. The IKDCs induced by MOG-pσ1 expressed elevated levels of HVEM for interactions with cognate ligand-positive cells: LIGHT(+) NK and T(eff) cells and BTLA(+) B cells. Further characterization revealed these activated IKDCs being MHC class II(high), and upon their adoptive transfer (CD11c(+)NK1.1(+)MHC class II(high)), IKDCs, but not CD11c(+)NK1.1(+)MHC class II(intermediate/low) (unactivated) cells, conferred protection against EAE. These activated IKDCs showed enhanced CD107a, PD-L1, and granzyme B expression and could present OVA, unlike unactivated IKDCs. Thus, these results demonstrate the interventional potency induced HVEM(+) IKDCs to resolve autoimmune disease.


Assuntos
Células Dendríticas/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/terapia , Proteínas da Mielina/metabolismo , Proteínas Virais/metabolismo , Transferência Adotiva , Animais , Antígenos Ly/metabolismo , Citotoxicidade Imunológica , Células Dendríticas/imunologia , Células Dendríticas/patologia , Progressão da Doença , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/fisiopatologia , Tolerância Imunológica , Interferons/metabolismo , Ativação Linfocitária , Camundongos , Proteínas da Mielina/genética , Proteínas da Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Subfamília B de Receptores Semelhantes a Lectina de Células NK/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Recidiva , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/patologia , Proteínas Virais/genética , Proteínas Virais/imunologia
15.
J Immunol ; 187(9): 4654-66, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21967895

RESUMO

Oral immunization with a Salmonella vaccine vector expressing enterotoxigenic Escherichia coli colonization factor Ag I (CFA/I) can protect against collagen-induced arthritis (CIA) by dampening IL-17 and IFN-γ via enhanced IL-4, IL-10, and TGF-ß. To identify the responsible regulatory CD4(+) T cells making the host refractory to CIA, Salmonella-CFA/I induced CD39(+)CD4(+) T cells with enhanced apyrase activity relative to Salmonella vector-immunized mice. Adoptive transfer of vaccine-induced CD39(+)CD4(+) T cells into CIA mice conferred complete protection, whereas CD39(-)CD4(+) T cells did not. Subsequent analysis of vaccinated Foxp3-GFP mice revealed the CD39(+) T cells were composed of Foxp3-GFP(-) and Foxp3-GFP(+) subpopulations. Although each adoptively transferred Salmonella-CFA/I-induced Foxp3(-) and Foxp3(+)CD39(+)CD4(+) T cells could protect against CIA, each subset was not as efficacious as total CD39(+)CD4(+) T cells, suggesting their interdependence for optimal protection. Cytokine analysis revealed Foxp3(-) CD39(+)CD4(+) T cells produced TGF-ß, and Foxp3(+)CD39(+)CD4(+) T cells produced IL-10, showing a segregation of function. Moreover, donor Foxp3-GFP(-) CD4(+) T cells converted to Foxp3-GFP(+) CD39(+)CD4(+) T cells in the recipients, showing plasticity of these regulatory T cells. TGF-ß was found to be essential for protection because in vivo TGF-ß neutralization reversed activation of CREB and reduced the development of CD39(+)CD4(+) T cells. Thus, CD39 apyrase-expressing CD4(+) T cells stimulated by Salmonella-CFA/I are composed of TGF-ß-producing Foxp3(-) CD39(+)CD4(+) T cells and support the stimulation of IL-10-producing Foxp3(+) CD39(+)CD4(+) T cells.


Assuntos
Antígenos CD/fisiologia , Apirase/fisiologia , Artrite Experimental/imunologia , Linfócitos T CD4-Positivos/imunologia , Colágeno/toxicidade , Fatores de Transcrição Forkhead/fisiologia , Interleucina-10/fisiologia , Fator de Crescimento Transformador beta/biossíntese , Animais , Antígenos CD/biossíntese , Apirase/biossíntese , Artrite Experimental/patologia , Artrite Experimental/prevenção & controle , Linfócitos T CD4-Positivos/metabolismo , Colágeno/administração & dosagem , Proteínas de Fímbrias , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Interleucina-10/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vacinas contra Salmonella/administração & dosagem , Vacinas contra Salmonella/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia
16.
PLoS One ; 6(7): e21978, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21765931

RESUMO

γδ T cells have been postulated to act as a first line of defense against infectious agents, particularly intracellular pathogens, representing an important link between the innate and adaptive immune responses. Human γδ T cells expand in the blood of brucellosis patients and are active against Brucella in vitro. However, the role of γδ T cells in vivo during experimental brucellosis has not been studied. Here we report TCRδ(-/-) mice are more susceptible to B. abortus infection than C57BL/6 mice at one week post-infection as measured by splenic colonization and splenomegaly. An increase in TCRγδ cells was observed in the spleens of B. abortus-infected C57BL/6 mice, which peaked at two weeks post-infection and occurred concomitantly with diminished brucellae. γδ T cells were the major source of IL-17 following infection and also produced IFN-γ. Depletion of γδ T cells from C57BL/6, IL-17Rα(-/-), and GMCSF(-/-) mice enhanced susceptibility to B. abortus infection although this susceptibility was unaltered in the mutant mice; however, when γδ T cells were depleted from IFN-γ(-/-) mice, enhanced susceptibility was observed. Neutralization of γδ T cells in the absence of TNF-α did not further impair immunity. In the absence of TNF-α or γδ T cells, B. abortus-infected mice showed enhanced IFN-γ, suggesting that they augmented production to compensate for the loss of γδ T cells and/or TNF-α. While the protective role of γδ T cells was TNF-α-dependent, γδ T cells were not the major source of TNF-α and activation of γδ T cells following B. abortus infection was TNF-α-independent. Additionally, bovine TCRγδ cells were found to respond rapidly to B. abortus infection upon co-culture with autologous macrophages and could impair the intramacrophage replication of B. abortus via IFN-γ. Collectively, these results demonstrate γδ T cells are important for early protection to B. abortus infections.


Assuntos
Brucella abortus/imunologia , Brucelose Bovina/imunologia , Brucelose/imunologia , Imunidade Inata/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Animais , Brucella abortus/crescimento & desenvolvimento , Brucelose/prevenção & controle , Brucelose Bovina/prevenção & controle , Bovinos , Proliferação de Células , Perfilação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Interferon gama/biossíntese , Interleucina-17/biossíntese , Subpopulações de Linfócitos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Receptores de Antígenos de Linfócitos T gama-delta/deficiência , Baço/imunologia , Baço/microbiologia , Baço/patologia , Fator de Necrose Tumoral alfa/imunologia
17.
J Leukoc Biol ; 90(6): 1043-54, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21693591

RESUMO

Human IBD, including UC and Crohn's disease, is characterized by a chronic, relapsing, and remitting condition that exhibits various features of immunological inflammation and affects at least one/1000 people in Western countries. Polyphenol extracts from a variety of plants have been shown to have immunomodulatory and anti-inflammatory effects. In this study, treatment with APP was investigated to ameliorate chemically induced colitis. Oral but not peritoneal administration of APP during colitis induction significantly protected C57BL/6 mice against disease, as evidenced by the lack of weight loss, colonic inflammation, and shortening of the colon. APP administration dampened the mRNA expression of IL-1ß, TNF-α, IL-6, IL-17, IL-22, CXCL9, CXCL10, CXCL11, and IFN-γ in the colons of mice with colitis. APP-mediated protection requires T cells, as protection was abated in Rag-1(-/-) or TCRα(-/-) mice but not in IL-10(-/-), IRF-1(-/-), µMT, or TCRδ(-/-) mice. Administration of APP during colitis to TCRα(-/-) mice actually enhanced proinflammatory cytokine expression, further demonstrating a requirement for TCRαß cells in APP-mediated protection. APP treatment also inhibited CXCR3 expression by TCRαß cells, but not B or NK cells, in the colons of mice with colitis; however, depletion of CD4(+) or CD8(+) T cells alone did not abolish APP-mediated protection. Collectively, these results show that oral administration of APP protects against experimental colitis and diminishes proinflammatory cytokine expression via T cells.


Assuntos
Colite/imunologia , Citocinas/antagonistas & inibidores , Sulfato de Dextrana/toxicidade , Mediadores da Inflamação/fisiologia , Malus/fisiologia , Polifenóis/fisiologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/patologia , Animais , Células Cultivadas , Colite/induzido quimicamente , Colite/patologia , Citocinas/biossíntese , Citocinas/genética , Sulfato de Dextrana/administração & dosagem , Feminino , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/química , Masculino , Malus/química , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polifenóis/química , Subpopulações de Linfócitos T/imunologia
18.
BMC Immunol ; 11: 36, 2010 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-20609248

RESUMO

BACKGROUND: Immunization with recombinant carboxyl-terminal domain of the heavy chain (Hc domain) of botulinum neurotoxin (BoNT) stimulates protective immunity against native BoNT challenge. Most studies developing a botulism vaccine have focused on the whole Hc; however, since the principal protective epitopes are located within beta-trefoil domain (Hcbetatre), we hypothesize that immunization with the Hcbetatre domain is sufficient to confer protective immunity. In addition, enhancing its uptake subsequent to nasal delivery prompted development of an alternative vaccine strategy, and we hypothesize that the addition of targeting moiety adenovirus 2 fiber protein (Ad2F) may enhance such uptake during vaccination. RESULTS: The Hcbetatre serotype B immunogen was genetically fused to Ad2F (Hcbetatre/B-Ad2F), and its immunogenicity was tested in mice. In combination with the mucosal adjuvant, cholera toxin (CT), enhanced mucosal IgA and serum IgG Ab titers were induced by nasal Hcbetatre-Ad2F relative to Hcbetatre alone; however, similar Ab titers were obtained upon intramuscular immunization. These BoNT/B-specific Abs induced by nasal immunization were generally supported in large part by Th2 cells, as opposed to Hcbetatre-immunized mice that showed more mixed Th1 and Th2 cells. Using a mouse neutralization assay, sera from animals immunized with Hcbetatre and Hcbetatre-Ad2F protected mice against 2.0 LD50. CONCLUSION: These results demonstrate that Hcbetatre-based immunogens are highly immunogenic, especially when genetically fused to Ad2F, and Ad2F can be exploited as a vaccine delivery platform to the mucosa.


Assuntos
Adenoviridae/metabolismo , Toxinas Botulínicas/imunologia , Vacinação/métodos , Proteínas Virais/metabolismo , Administração Intranasal , Animais , Linfócitos B/imunologia , Toxinas Botulínicas/química , Toxinas Botulínicas Tipo A , Botulismo/imunologia , Botulismo/prevenção & controle , Toxina da Cólera/imunologia , Citocinas/imunologia , Imunoensaio , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos C57BL , Testes de Neutralização , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Análise de Sobrevida , Linfócitos T Auxiliares-Indutores/imunologia
19.
J Immunol ; 184(12): 7144-53, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20483737

RESUMO

IL-35 is produced by regulatory T cells, and this novel cytokine can downregulate Th17 cell development and inhibit autoimmune inflammation. In this work, an rIL-35, as a single-chain fusion between murine IL-12p35 and EBV-induced gene 3, was expressed in yeast. This rIL-35 inhibited OVA-specific cellular and Ab responses in OVA-challenged recipients of DO11.10 CD4+ T cells. Likewise, IL-35 inhibited clinical manifestation of collagen-induced arthritis or could cease further disease exacerbation upon initiation of IL-35 treatment. Exogenous IL-35 treatments suppressed Th1 and Th17 cells and promoted CD39 expression by CD4+ T cells. Sorted CD25-CD39+CD4+ T cells from IL-35-treated mice produced IL-10 and, upon adoptive transfer, were sufficiently potent to inhibit subsequent development of inflammation in mice with collagen-induced arthritis, whereas sorted CD25+CD39+CD4+ T cells showed reduced potency. IL-35 treatments of IL-10-/- mice failed to induce protective CD39+CD4+ T cells, demonstrating the effector role of IL-10 by IL-35 immunosuppression.


Assuntos
Artrite Experimental/imunologia , Tolerância Imunológica/imunologia , Interleucina-10/imunologia , Subunidade p35 da Interleucina-12/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Apirase/imunologia , Apirase/metabolismo , Artrite Experimental/metabolismo , Separação Celular , Citometria de Fluxo , Interleucina-10/biossíntese , Subunidade p35 da Interleucina-12/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo
20.
Vaccine ; 28(4): 1069-74, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19914192

RESUMO

To create a new, safe brucellosis live vaccine, a double mutant strain was constructed from Brucella abortus 2308. Using the DeltaznuA B. abortus 2308 mutant, a second mutation was introduced by deleting purE gene. The DeltaznuA DeltapurE B. abortus 2308 strain was less capable of surviving in macrophages. When evaluated in vivo, it was cleared within 8 weeks (wks) from mice, causing significantly less inflammation than spleens obtained from wild-type B. abortus 2308-infected mice. Furthermore, two doses of DeltaznuA DeltapurE B. abortus 2308 conferred 0.79 log protection, similar to S19 as did a single dose of DeltaznuA B. abortus 2308. Thus, this study shows the DeltaznuA DeltapurE B. abortus 2308 strain to be a potential livestock vaccine candidate.


Assuntos
Proteínas de Bactérias/genética , Vacina contra Brucelose/genética , Vacina contra Brucelose/imunologia , Brucella abortus/genética , Brucella abortus/imunologia , Deleção de Genes , Animais , Brucella abortus/patogenicidade , Brucelose/microbiologia , Brucelose/patologia , Feminino , Imunização Secundária/métodos , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Viabilidade Microbiana , Baço/patologia , Vacinação/métodos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA