Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Cell Sci ; 132(16)2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31331966

RESUMEN

Here, we show that cells expressing the adherens junction protein nectin-1 capture nectin-4-containing membranes from the surface of adjacent cells in a trans-endocytosis process. We find that internalized nectin-1-nectin-4 complexes follow the endocytic pathway. The nectin-1 cytoplasmic tail controls transfer: its deletion prevents trans-endocytosis, while its exchange with the nectin-4 tail reverses transfer direction. Nectin-1-expressing cells acquire dye-labeled cytoplasmic proteins synchronously with nectin-4, a process most active during cell adhesion. Some cytoplasmic cargo remains functional after transfer, as demonstrated with encapsidated genomes of measles virus (MeV). This virus uses nectin-4, but not nectin-1, as a receptor. Epithelial cells expressing nectin-4, but not those expressing another MeV receptor in its place, can transfer infection to nectin-1-expressing primary neurons. Thus, this newly discovered process can move cytoplasmic cargo, including infectious material, from epithelial cells to neurons. We name the process nectin-elicited cytoplasm transfer (NECT). NECT-related trans-endocytosis processes may be exploited by pathogens to extend tropism. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Endocitosis , Células Epiteliales/metabolismo , Virus del Sarampión/metabolismo , Nectinas/metabolismo , Internalización del Virus , Transporte Biológico Activo/genética , Adhesión Celular/genética , Moléculas de Adhesión Celular/genética , Línea Celular , Humanos , Virus del Sarampión/genética , Nectinas/genética
2.
Immunol Cell Biol ; 94(5): 496-508, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26752510

RESUMEN

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.


Asunto(s)
Brucella melitensis/fisiología , Brucelosis/inmunología , Brucelosis/prevención & control , Linfocitos T CD8-positivos/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/microbiología , Vacunación , Animales , Vacunas Bacterianas/inmunología , Brucelosis/microbiología , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Femenino , Memoria Inmunológica , Mediadores de Inflamación/metabolismo , Interferón gamma , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mucosa Nasal/patología , Bazo/patología
3.
J Immunol ; 192(2): 804-16, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337375

RESUMEN

A Salmonella therapeutic expressing enterotoxigenic Escherichia coli colonization factor Ag I (CFA/I) fimbriae protects against collagen-induced arthritis (CIA) by eliciting two regulatory T cell (Treg) subsets: TGF-ß-producing Foxp3(-)CD39(+)CD4(+) T cells and IL-10-producing Foxp3(+)CD39(+)CD4(+) T cells. However, it is unclear whether CFA/I fimbriae alone are protective and whether other regulatory cytokines are involved, especially in the context for the EBI3-sharing cytokines, Treg-derived IL-35 and APC-derived IL-27, both capable of suppressing Th17 cells and regulating autoimmune diseases. Subsequent evaluation revealed that a single oral dose of purified, soluble CFA/I fimbriae protected against CIA as effectively as did Salmonella-CFA/I and found that Foxp3(+)CD39(+)CD4(+) T cells were the source of secreted IL-35, whereas IL-27 production by CD11c(+) cells was inhibited. Inquiring into their relevance, CFA/I fimbriae-treated IL-27R-deficient (WSX-1(-/-)) mice were equally protected against CIA as were wild-type mice, suggesting a limited role for IL-27. In contrast, CFA/I fimbriae-mediated protection was abated in EBI3(-/-) mice, accompanied by the loss of TGF-ß- and IL-10-producing Tregs. Adoptive transfer of C57BL/6 CD39(+)CD4(+) T cells to EBI3(-/-) mice with concurrent CFA/I plus IL-35 treatment effectively stimulated Tregs suppressing proinflammatory collagen II-specific Th cells. In contrast, recipients cotransferred with C57BL/6 and EBI3(-/-) CD39(+)CD4(+) T cells and treated with CFA/I plus IL-35 were not protected, implicating the importance of endogenous IL-35 for conferring CFA/I-mediated protection. Thus, CFA/I fimbriae stimulate IL-35 required for the coinduction of TGF-ß and IL-10.


Asunto(s)
Artritis Experimental/inmunología , Escherichia coli/inmunología , Proteínas Fimbrias/inmunología , Fimbrias Bacterianas/inmunología , Interleucina-27/inmunología , Interleucinas/inmunología , Animales , Antígenos Bacterianos/inmunología , Antígenos CD/inmunología , Artritis Experimental/inducido químicamente , Factores de Transcripción Forkhead/inmunología , Interleucina-10/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Antígenos de Histocompatibilidad Menor , Receptores de Citocinas/inmunología , Células Th17/inmunología , Factor de Crecimiento Transformador beta/inmunología
4.
J Immunol ; 187(9): 4654-66, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21967895

RESUMEN

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.


Asunto(s)
Antígenos CD/fisiología , Apirasa/fisiología , Artritis Experimental/inmunología , Linfocitos T CD4-Positivos/inmunología , Colágeno/toxicidad , Factores de Transcripción Forkhead/fisiología , Interleucina-10/fisiología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Antígenos CD/biosíntesis , Apirasa/biosíntesis , Artritis Experimental/patología , Artritis Experimental/prevención & control , Linfocitos T CD4-Positivos/metabolismo , Colágeno/administración & dosificación , Proteínas Fimbrias , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Interleucina-10/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vacunas contra la Salmonella/administración & dosificación , Vacunas contra la Salmonella/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/trasplante , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
5.
Infect Immun ; 79(10): 4165-74, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21768283

RESUMEN

Zoonotic transmission of brucellosis often results from exposure to Brucella-infected livestock, feral animals, or wildlife or frequently via consumption of unpasteurized milk products or raw meat. Since natural infection of humans often occurs by the oral route, mucosal vaccination may offer a means to confer protection for both mucosal and systemic tissues. Significant efforts have focused on developing a live brucellosis vaccine, and deletion of the znuA gene involved in zinc transport has been found to attenuate Brucella abortus. A similar mutation has been adapted for Brucella melitensis and tested to determine whether oral administration of ΔznuA B. melitensis can confer protection against nasal B. melitensis challenge. A single oral vaccination with ΔznuA B. melitensis rapidly cleared from mice within 2 weeks and effectively protected mice upon nasal challenge with wild-type B. melitensis 16M. In 83% of the vaccinated mice, no detectable brucellae were found in their spleens, unlike with phosphate-buffered saline (PBS)-dosed mice, and vaccination also enhanced the clearance of brucellae from the lungs. Moreover, vaccinated gamma interferon-deficient (IFN-γ(-/-)) mice also showed protection in both spleens and lungs, albeit protection that was not as effective as in immunocompetent mice. Although IFN-γ, interleukin 17 (IL-17), and IL-22 were stimulated by these live vaccines, only RB51-mediated protection was codependent upon IL-17 in BALB/c mice. These data suggest that oral immunization with the live, attenuated ΔznuA B. melitensis vaccine provides an attractive strategy to protect against inhalational infection with virulent B. melitensis.


Asunto(s)
Vacuna contra la Brucelosis/inmunología , Brucella melitensis/inmunología , Brucelosis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Vacunas Atenuadas/inmunología , Administración Oral , Animales , Vacuna contra la Brucelosis/administración & dosificación , Vacuna contra la Brucelosis/genética , Brucella melitensis/patogenicidad , Brucelosis/prevención & control , Femenino , Interleucina-17/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética
6.
J Biomed Biotechnol ; 2011: 632396, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22007145

RESUMEN

To assess whether anticolonization factor antigen I (CFA/I) fimbriae antibodies (Abs) from enterotoxigenic Escherichia coli (ETEC) can protect against various routes of challenge, BALB/c mice were immunized with a live attenuated Salmonella vaccine vector expressing CFA/I fimbriae. Vaccinated mice elicited elevated systemic IgG and mucosal IgA Abs, unlike mice immunized with the empty Salmonella vector. Mice were challenged with wild-type ETEC by the oral, intranasal (i.n.), and intraperitoneal (i.p.) routes. Naïve mice did not succumb to oral challenge, but did to i.n. challenge, as did immunized mice; however, vaccinated mice were protected against i.p. ETEC challenge. Two intramuscular (i.m.) immunizations with CFA/I fimbriae without adjuvant conferred 100% protection against i.p. ETEC challenge, while a single 30 µg dose conferred 88% protection. Bactericidal assays showed that ETEC is highly sensitive to anti-CFA/I sera. These results suggest that parenteral immunization with purified CFA/I fimbriae can induce protective Abs and may represent an alternative method to elicit protective Abs for passive immunity to ETEC.


Asunto(s)
Anticuerpos/metabolismo , Actividad Bactericida de la Sangre/inmunología , Escherichia coli Enterotoxigénica/inmunología , Proteínas Fimbrias/administración & dosificación , Proteínas Fimbrias/inmunología , Inmunización/métodos , Inyecciones Intraperitoneales , Administración Intranasal , Administración Oral , Animales , Anticuerpos/inmunología , Actividad Bactericida de la Sangre/efectos de los fármacos , Escherichia coli Enterotoxigénica/metabolismo , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Proteínas de Escherichia coli/administración & dosificación , Proteínas de Escherichia coli/inmunología , Femenino , Proteínas Fimbrias/aislamiento & purificación , Inmunidad Mucosa/inmunología , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunas contra la Salmonella/administración & dosificación , Vacunas contra la Salmonella/inmunología , Determinación de Anticuerpos Séricos Bactericidas
7.
Vaccine ; 34(44): 5290-5297, 2016 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-27639282

RESUMEN

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.


Asunto(s)
Vacuna contra la Brucelosis/genética , Vacuna contra la Brucelosis/inmunología , Brucella abortus/genética , Brucelosis/prevención & control , Animales , Vacuna contra la Brucelosis/administración & dosificación , Brucella abortus/patogenicidad , Brucella abortus/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Genes Bacterianos , Inmunidad Celular , Interferón gamma/biosíntesis , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Mutación , Bazo/microbiología , Factor de Necrosis Tumoral alfa/biosíntesis , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Virulencia
8.
PLoS One ; 10(1): e0117825, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25629976

RESUMEN

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.


Asunto(s)
Antígenos CD/metabolismo , Apirasa/metabolismo , Artritis Experimental/tratamiento farmacológico , Suplementos Dietéticos , Interleucina-10/metabolismo , Leche/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Artritis Experimental/inmunología , Artritis Experimental/metabolismo , Efecto Espectador/inmunología , Tolerancia Inmunológica/inmunología , Ratones , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/inmunología
9.
Front Biol (Beijing) ; 8(1): 60-77, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23730309

RESUMEN

Brucella spp. are zoonotic, facultative intracellular pathogens, which cause animal and human disease. Animal disease results in abortion of fetuses; in humans, it manifests flu-like symptoms with an undulant fever, with osteoarthritis as a common complication of infection. Antibiotic regimens for human brucellosis patients may last several months and are not always completely effective. While there are no vaccines for humans, several licensed live Brucella vaccines are available for use in livestock. The performance of these animal vaccines is dependent upon the host species, dose, and route of immunization. Newly engineered live vaccines, lacking well-defined virulence factors, retain low residual virulence, are highly protective, and may someday replace currently used animal vaccines. These also have possible human applications. Moreover, due to their enhanced safety and efficacy in animal models, subunit vaccines for brucellosis show great promise for their application in livestock and humans. This review summarizes the progress of brucellosis vaccine development and presents an overview of candidate vaccines.

10.
PLoS One ; 7(4): e36283, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22558420

RESUMEN

During infection, Yersinia pestis uses its F1 capsule to enhance survival and cause virulence to mammalian host. Since F1 is produced in large quantities and secreted into the host tissues, it also serves as a major immune target. To hold this detrimental effect under proper control, Y. pestis expresses the caf operon (encoding the F1 capsule) in a temperature-dependent manner. However, additional properties of the caf operon limit its expression. By overexpressing the caf operon in wild-type Salmonella enterica serovar Typhimurium under a potent promoter, virulence of Salmonella was greatly attenuated both in vitro and in vivo. In contrast, expression of the caf operon under the regulation of its native promoter exhibited negligible impairment of Salmonellae virulence. In-depth investigation revealed all individual genes in the caf operon attenuated Salmonella when overexpressed. The deleterious effects of caf operon and the caf individual genes were further confirmed when they were overexpressed in Y. pestis KIM6+. This study suggests that by using a weak inducible promoter, the detrimental effects of the caf operon are minimally manifested in Y. pestis. Thus, through tight regulation of the caf operon, Y. pestis precisely balances its capsular anti-phagocytic properties with the detrimental effects of caf during interaction with mammalian host.


Asunto(s)
Cápsulas Bacterianas/genética , Vectores Genéticos/genética , Operón/genética , Salmonella enterica/genética , Yersinia pestis/genética , Animales , Línea Celular , Femenino , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Salmonella enterica/patogenicidad , Salmonella enterica/fisiología
11.
Virulence ; 3(1): 29-42, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22286706

RESUMEN

Generation of a live attenuated vaccine for bacterial pathogens often requires prior knowledge of the pathogen's virulence factors. We hypothesized an alternative approach of heterologous gene expression would make a wild-type (wt) pathogen more susceptible to host cell killing, thus, resulting in immunization. As proof of concept, the heterologous expression of enterotoxigenic E. coli (ETEC) colonization factor antigen I (CFA/I) was tested to attenuate Salmonella. The overexpression of CFA/I resulted in significant attenuation of wt Salmonella. In-depth studies revealed the attenuation depended on the co-expression of chaperone (CfaA) and usher (CfaC) proteins. Remarkably, the CfaAC-attenuated Salmonella conferred protection against wt Salmonella challenge. Mechanistic study indicated CfaAC made Salmonella outer membranes permeable, causing Salmonella to be vulnerable to host destruction. Thus, enhancing bacterial permeability via CfaAC represents an alternative method to attenuate pathogens despite the presence of unknown virulence factors.


Asunto(s)
Proteínas de Escherichia coli/genética , Expresión Génica , Chaperonas Moleculares/genética , Infecciones por Salmonella/microbiología , Salmonella typhimurium/genética , Salmonella typhimurium/inmunología , Factores de Virulencia/genética , Animales , Línea Celular , Proteínas de Escherichia coli/inmunología , Femenino , Proteínas Fimbrias/genética , Proteínas Fimbrias/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Chaperonas Moleculares/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/fisiología , Virulencia , Factores de Virulencia/inmunología
12.
PLoS One ; 7(10): e46828, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23056473

RESUMEN

Flagella are cell surface appendages involved in a number of bacterial behaviors, such as motility, biofilm formation, and chemotaxis. Despite these important functions, flagella can pose a liability to a bacterium when serving as potent immunogens resulting in the stimulation of the innate and adaptive immune systems. Previous work showing appendage overexpression, referred to as attenuating gene expression (AGE), was found to enfeeble wild-type Salmonella. Thus, this approach was adapted to discern whether flagella overexpression could induce similar attenuation. To test its feasibility, flagellar filament subunit FliC and flagellar regulon master regulator FlhDC were overexpressed in Salmonella enterica serovar Typhimurium wild-type strain H71. The results show that the expression of either FliC or FlhDC alone, and co-expression of the two, significantly attenuates Salmonella. The flagellated bacilli were unable to replicate within macrophages and thus were not lethal to mice. In-depth investigation suggests that flagellum-mediated AGE was due to the disruptive effects of flagella on the bacterial membrane, resulting in heightened susceptibilities to hydrogen peroxide and bile. Furthermore, flagellum-attenuated Salmonella elicited elevated immune responses to Salmonella presumably via FliC's adjuvant effect and conferred robust protection against wild-type Salmonella challenge.


Asunto(s)
Flagelos/genética , Salmonella typhimurium/genética , Salmonella typhimurium/patogenicidad , Animales , Proteínas Bacterianas/genética , Femenino , Expresión Génica , Ratones , Ratones Endogámicos C57BL , Viabilidad Microbiana/genética , Salmonella typhimurium/citología , Salmonella typhimurium/crecimiento & desarrollo , Vacunación
13.
J Leukoc Biol ; 92(2): 375-87, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22636321

RESUMEN

Human brucellosis exhibits diverse pathological manifestations that can affect almost any organ. In particular, osteoarticular complications are the most common focal manifestation of brucellosis and occur in 40-80% of patients. In immunocompetent mice, Brucella replication is generally restricted to the spleen, liver, and to a lesser extent, LNs, thereby limiting their use for study of focal inflammation often found in brucellosis. Here, we report that nasal, oral, or peritoneal infection of IFN-γ(-/-) mice with WT Brucella melitensis or Brucella abortus results in joint and periarticular tissue inflammation. Histological analysis of the affected joints revealed inflammatory infiltrates and debris within the joint space colocalizing with Brucella antigen. Osteoarthritis, necrosis, periarticular soft tissue inflammation, and substantial brucellae burdens were observed. Oral rifampicin was effective in clearing infection and halting further progression of focal inflammation from infected IFN-γ(-/-) mice, although some symptoms and swelling remained. Elevated IL-1 ß, but not TNF-α, IL-6, or IL-17, was detected in joint homogenates from infected IFN-γ(-/-) mice. Whereas more susceptible to systemic infection, IL-1R(-/-) mice depleted of IFN-γ were more resistant to focal inflammation than WT mice similarly depleted of IFN-γ. Collectively, these results show IFN-γ(-/-) mice represent a potential model for study of focal inflammation attributed to Brucella infection and will allow evaluation of intervention strategies targeting IL-1, IL-1R, or other inflammatory mediators, with the potential to complement antibiotic-based therapies.


Asunto(s)
Brucelosis/inmunología , Dermatitis/inmunología , Mediadores de Inflamación/fisiología , Interferón gamma/deficiencia , Interleucina-1/fisiología , Miositis/inmunología , Animales , Brucelosis/genética , Brucelosis/microbiología , Células Cultivadas , Dermatitis/genética , Dermatitis/microbiología , Modelos Animales de Enfermedad , Humanos , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Interferón gamma/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Miositis/genética , Miositis/microbiología , Conejos
14.
J Wildl Dis ; 47(3): 501-10, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21719815

RESUMEN

Brucella abortus remains a threat to the health and well-being of livestock in states bordering the Greater Yellowstone Area. During the past several years, cohabitation of infected wildlife with cattle has jeopardized the brucellosis-free status of Idaho, USA; Wyoming, USA; and Montana, USA. Current livestock B. abortus vaccines have not proven to be efficacious in bison (Bison bison) or elk (Cervus elaphus nelsoni). One problem with the lack of vaccine efficacy may stem from the failure to understand wildlife immune responses to vaccines. In an attempt to understand their immune responses, bison were vaccinated with eukaryotic DNA expression vectors encoding the Brucella periplasmic protein, bp26, and the chaperone protein, trigger factor (TF). These DNA vaccines have previously been shown to be protective against Brucella infection in mice. Bison were immunized intramuscularly at weeks 0, 2, and 4 with bp26 and TF DNA vaccines plus CpG adjuvant or empty vector (control) plus CpG. Blood samples were collected before vaccination and at 8, 10, and 12 wk after primary vaccination. The results showed that bison immunized with bp26 and TF DNA vaccines developed enhanced antibody, proliferative T cell, and interferon-gamma (IFN-γ) responses upon in vitro restimulation with purified recombinant bp26 or TF antigens, unlike bison immunized with empty vector. Flow cytometric analysis revealed that the percentages of CD4(+) and CD8(+) T lymphocytes from the DNA-vaccinated groups were significantly greater than they were for those bison given empty vector. These data suggest that DNA vaccination of bison may elicit strong cellular immune responses and serve as an alternative for vaccination of bison for brucellosis.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Bison , Vacuna contra la Brucelosis/administración & dosificación , Brucella abortus/inmunología , Brucelosis/veterinaria , Proteínas de la Membrana/inmunología , Vacunas de ADN/administración & dosificación , Animales , Animales Domésticos , Animales Salvajes , Bison/inmunología , Bison/microbiología , Brucelosis/inmunología , Brucelosis/prevención & control , Brucelosis/transmisión , Femenino , Interferón gamma/sangre , Interferón gamma/inmunología , Linfocitos/inmunología , Masculino , Especificidad de la Especie
15.
PLoS One ; 6(7): e21978, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765931

RESUMEN

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.


Asunto(s)
Brucella abortus/inmunología , Brucelosis Bovina/inmunología , Brucelosis/inmunología , Inmunidad Innata/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Animales , Brucella abortus/crecimiento & desarrollo , Brucelosis/prevención & control , Brucelosis Bovina/prevención & control , Bovinos , Proliferación Celular , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Subgrupos Linfocitarios/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Bazo/inmunología , Bazo/microbiología , Bazo/patología , Factor de Necrosis Tumoral alfa/inmunología
16.
Vaccine ; 28(4): 1069-74, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19914192

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/genética , Vacuna contra la Brucelosis/genética , Vacuna contra la Brucelosis/inmunología , Brucella abortus/genética , Brucella abortus/inmunología , Eliminación de Gen , Animales , Brucella abortus/patogenicidad , Brucelosis/microbiología , Brucelosis/patología , Femenino , Inmunización Secundaria/métodos , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Viabilidad Microbiana , Bazo/patología , Vacunación/métodos , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virulencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA