Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pathogens ; 9(5)2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32422907

RESUMEN

The targeting of immunogens/vaccines to specific immune cells is a promising approach for amplifying immune responses in the absence of exogenous adjuvants. However, the targeting approaches reported thus far require novel, labor-intensive reagents for each vaccine and have primarily been shown as proof-of-concept with isolated proteins and/or inactivated bacteria. We have engineered a plasmid-based, complement receptor-targeting platform that is readily applicable to live forms of multiple gram-negative bacteria, including, but not limited to, Escherichia coli, Klebsiella pneumoniae, and Francisella tularensis. Using F. tularensis as a model, we find that targeted bacteria show increased binding and uptake by macrophages, which coincides with increased p38 and p65 phosphorylation. Mice vaccinated with targeted bacteria produce higher titers of specific antibody that recognizes a greater diversity of bacterial antigens. Following challenge with homologous or heterologous isolates, these mice exhibited less weight loss and/or accelerated weight recovery as compared to counterparts vaccinated with non-targeted immunogens. Collectively, these findings provide proof-of-concept for plasmid-based, complement receptor-targeting of live gram-negative bacteria.

2.
Vaccines (Basel) ; 8(2)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340134

RESUMEN

Lack of safe and effective mucosal adjuvants has severely hampered the development of mucosal subunit vaccines. In this regard, we have previously shown that immunogenicity of vaccine antigens can be improved by targeting the antigens to the antigen-presenting cells. Specifically, groups of mice immunized intranasally with a fusion protein (Bivalent-FP) containing a fragment of pneumococcal-surface-protein-A (PspA) as antigen and a single-chain bivalent antibody raised against the anti-human Fc-gamma-receptor-I (hFcγRI) elicited protective immunity to pulmonary Streptococcus pneumoniae infection. In order to further enhance the immunogenicity, an additional hFcγRI-binding moiety of the single chain antibody was incorporated. The modified vaccine (Trivalent-FP) induced significantly improved protection against lethal pulmonary S. pneumoniae challenge compared to Bivalent-FP. In addition, the modified vaccine exhibited over 85% protection with only two immunizations. Trivalent-FP also induced S. pneumoniae-specific systemic and mucosal antibodies. Moreover, Trivalent-FP also induced IL-17- and IL-22-producing CD4+ T cells. Furthermore, it was found that the hFcγRI facilitated uptake and presentation of Trivalent-FP. In addition, Trivalent-FP also induced IL-1α, MIP-1α, and TNF-α; modulated recruitment of dendritic cells and macrophages; and induced CD80/86 and MHC-II expression on antigen presenting cells.

3.
Cell Immunol ; 345: 103962, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31582169

RESUMEN

Previous in vivo studies established that inactivated Francisella tularensis immune complexes (mAb-iFt) are a more protective vaccine against lethal tularemia than iFt alone. Subsequent in vitro studies revealed enhanced DC maturation marker expression with mAb-iFt stimulation. The goal of this study was to determine the mechanism of enhanced DC maturation. Multiparameter analysis of surface marker expression and cytokine secretion demonstrates a requirement for FcγR signaling in enhanced DC maturation. MyD88 was also found to be essential for heightened DC maturation, implicating MyD88-dependent TLRs in DC maturation. Upon further study, we discovered that TLRs 2 & 4 drive cytokine secretion, but surprisingly TLR9 is required for DC maturation marker upregulation. These studies reveal a separation of DC cytokine and maturation marker induction pathways and demonstrate that FcγR-TLR/MyD88 synergy underlies the enhanced dendritic cell maturation in response to the mAb-iFt vaccine.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Receptores de IgG/inmunología , Receptor Toll-Like 9/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Vacunas Bacterianas/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Células Dendríticas/metabolismo , Francisella tularensis/inmunología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/metabolismo , Receptores de IgG/genética , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Tularemia/inmunología , Tularemia/microbiología
4.
Front Immunol ; 10: 1144, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191528

RESUMEN

Adjuvants have been used in vaccines for over a century, however, the search for safe and effective vaccine adjuvants continues. In recent decades toll-like-receptor (TLR) agonists have been investigated as potential vaccine adjuvants. In this regard, the majority of the currently investigated TLR agonists are non-protein microbial components such as lipopolysaccharides, oligonucleotides, and lipopeptides. On the other hand, a growing number of studies reveal that TLR signaling and immune responses can be activated by numerous bacterial proteins. However, their potential roles as adjuvants have been somewhat overlooked. Herein, we discuss several such bacterial proteins which exhibit adjuvant properties, including the activation of TLR signaling, antigen presenting cell maturation, pro-inflammatory cytokine production and adaptive immune response. The protein nature of these TLR agonists presents several unique features not shared by non-protein TLR agonists. These properties include the amenability for modifying the structure and function as necessary for optimal immunogenicity and minimal toxicity. Protein adjuvants can be genetically fused to protein antigens which ensure the co-delivery of adjuvant-antigen not only into the same cell but also in the same endocytic cargo, leading to more effective activation of innate and adaptive immune response.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas Bacterianas/inmunología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 5/agonistas , Vacunas/inmunología , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Antígenos Heterófilos/inmunología , Autoantígenos/inmunología , Proteínas Bacterianas/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Dimerización , Endocitosis , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/farmacología , Ligandos , Macrófagos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Ingeniería de Proteínas , Receptores de Reconocimiento de Patrones/fisiología , Autotolerancia/inmunología , Relación Estructura-Actividad
5.
PLoS One ; 13(12): e0207587, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30533047

RESUMEN

Francisella tularensis (Ft) is a biothreat agent for which there is no FDA-approved human vaccine. Currently, there are substantial efforts underway to develop both vaccines and the tools to assess these vaccines. Tularemia laboratory research has historically relied primarily upon a small number of inbred mouse strains, but the utility of such findings to outbred animals may be limited. Specifically, C57BL/6 mice are more susceptible than BALB/c mice to Ft infection and less easily protected against challenge with highly virulent type A Ft. Thus, depending on the inbred mouse strain used, one could be misled as to which immunogen(s)/vaccine will ultimately be effective in an outbred human population. Accordingly, we evaluated an outbred Swiss Webster (SW) mouse model in direct comparison to a well-established, inbred C57BL/6 mouse model. Mucosal vaccination with the live, attenuated Ft LVS superoxide dismutase (sodB) mutant demonstrated significantly higher protection in outbred SW mice compared to inbred C57BL/6 mice against Ft SchuS4 respiratory challenge. The protection observed in vaccinated outbred mice correlated with lower bacterial density, reduced tissue inflammation, and reduced levels of pro-inflammatory cytokine production. This protection was CD4+ and CD8+ T cell-dependent and characterized by lower titers of serum antibody (Ab) that qualitatively differed from vaccinated inbred mice. Enhanced protection of vaccinated outbred mice correlated with early and robust production of IFN-γ and IL-17A. Neutralizing Ab administered at the time of challenge revealed that IFN-γ was central to this protection, while IL-17A neutralization did not alter bacterial burden or survival. The present study demonstrates the utility of the outbred mouse as an alternative vaccination model for testing tularemia vaccines. Given the limited MHC repertoire in inbred mice, this outbred model is more analogous to the human in terms of immunological diversity.


Asunto(s)
Vacunas Bacterianas/inmunología , Francisella tularensis/inmunología , Animales , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , Femenino , Francisella tularensis/genética , Francisella tularensis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Superóxido Dismutasa/genética , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Vacunación
6.
Front Immunol ; 9: 1594, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30042767

RESUMEN

Francisella tularensis (Ft) is a biothreat agent for which there is no FDA-approved human vaccine. Currently, there are substantial efforts underway to develop both vaccines and improved tools to assess these vaccines. Ft expresses distinct sets of antigens (Ags) in vivo as compared to those expressed in vitro. Importantly, Ft grown in brain-heart infusion medium (BHIM) more closely mimics the antigenic profile of macrophage-grown Ft when compared to Mueller-Hinton medium (MHM)-grown Ft. Thus, we predicted that when used as a live vaccine BHIM-grown Ft (BHIM-Ft) would provide better protection, as compared to MHM-Ft. We first determined if there was a difference in growth kinetics between BHIM and MHM-Ft. We found that BHIM-Ft exhibited an initial growth advantage ex vivo that manifests as slightly hastened intracellular replication as compared to MHM-Ft. We also observed that BHIM-Ft exhibited an initial growth advantage in vivo represented by rapid bacterial expansion and systemic dissemination associated with a slightly shorter mean survival time of naive animals. Next, using two distinct strains of Ft LVS (WT and sodB), we observed that mice vaccinated with live BHIM-Ft LVS exhibited significantly better protection against Ft SchuS4 respiratory challenge compared to MHM-Ft-immunized mice. This enhanced protection correlated with lower bacterial burden, reduced tissue inflammation, and reduced pro-inflammatory cytokine production late in infection. Splenocytes from BHIM-Ft sodB-immunized mice contained more CD4+, effector, memory T-cells, and were more effective at limiting intracellular replication of Ft LVS in vitro. Concurrent with enhanced killing of Ft LVS, BHIM-Ft sodB-immune splenocytes produced significantly higher levels of IFN-γ and IL-17A cytokines than their MHM-Ft sodB-immunized counterparts indicating development of a more effective T cell memory response when immunizing mice with BHIM-Ft.

7.
Front Microbiol ; 8: 1158, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28690600

RESUMEN

The gram-negative bacterium Francisella tularensis (Ft) is both a potential biological weapon and a naturally occurring microbe that survives in arthropods, fresh water amoeba, and mammals with distinct phenotypes in various environments. Previously, we used a number of measurements to characterize Ft grown in Brain-Heart Infusion (BHI) broth as (1) more similar to infection-derived bacteria, and (2) slightly more virulent in naïve animals, compared to Ft grown in Mueller Hinton Broth (MHB). In these studies we observed that the free amino acids in MHB repress expression of select Ft virulence factors by an unknown mechanism. Here, we tested the hypotheses that Ft grown in BHI (BHI-Ft) accurately displays a full protein composition more similar to that reported for infection-derived Ft and that this similarity would make BHI-Ft more susceptible to pre-existing, vaccine-induced immunity than MHB-Ft. We performed comprehensive proteomic analysis of Ft grown in MHB, BHI, and BHI supplemented with casamino acids (BCA) and compared our findings to published "omics" data derived from Ft grown in vivo. Based on the abundance of ~1,000 proteins, the fingerprint of BHI-Ft is one of nutrient-deprived bacteria that-through induction of a stringent-starvation-like response-have induced the FevR regulon for expression of the bacterium's virulence factors, immuno-dominant antigens, and surface-carbohydrate synthases. To test the notion that increased abundance of dominant antigens expressed by BHI-Ft would render these bacteria more susceptible to pre-existing, vaccine-induced immunity, we employed a battery of LVS-vaccination and S4-challenge protocols using MHB- and BHI-grown Ft S4. Contrary to our hypothesis, these experiments reveal that LVS-immunization provides a barrier to infection that is significantly more effective against an MHB-S4 challenge than a BHI-S4 challenge. The differences in apparent virulence to immunized mice are profoundly greater than those observed with primary infection of naïve mice. Our findings suggest that tularemia vaccination studies should be critically evaluated in regard to the growth conditions of the challenge agent.

8.
Vaccine ; 35(7): 1080-1086, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069362

RESUMEN

Numerous studies have demonstrated that administration of antigen (Ag)-pulsed dendritic cells (DCs) is an effective strategy for enhancing immunity to tumors and infectious disease organisms. However, the generation and/or isolation of DCs can require substantial time and expense. Therefore, using inactivated F. tularensis (iFt) Ag as a model immunogen, we first sought to determine if DCs could be replaced with peripheral blood mononuclear cells (PBMCs) during the ex-vivo pulse phase and still provide protection against Ft infection. Follow up studies were then conducted using the S. pneumoniae (Sp) vaccine Prevnar ®13 as the Ag in the pulse phase followed by immunization and Sp challenge. In both cases, we demonstrate that PBMCs can be used in place of DCs when pulsing with iFt and/or Prevnar ®13 ex vivo and re-administering the Ag-pulsed PBMCs as a vaccine. In addition, utilization of the i.n. route for Ag-pulsed PBMC administration is superior to use of the i.v. route in the case of Sp immunization, as well as when compared to direct injection of Prevnar ®13 vaccine i.m. or i.n. Furthermore, this PBMC-based vaccine strategy provides a more marked and enduring protective immune response and is also capable of serving as a multi-organism vaccine platform. The potential for this ex-vivo vaccine strategy to provide a simpler, less time consuming, and less expensive approach to DC-based vaccines and vaccination in general is also discussed.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Antígenos Bacterianos/farmacología , Vacunas Bacterianas/administración & dosificación , Leucocitos Mononucleares/efectos de los fármacos , Infecciones Neumocócicas/prevención & control , Tularemia/prevención & control , Administración Intranasal , Traslado Adoptivo , Animales , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Francisella tularensis/química , Francisella tularensis/inmunología , Inyecciones Intramusculares , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos C57BL , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/mortalidad , Cultivo Primario de Células , Streptococcus pneumoniae/química , Streptococcus pneumoniae/inmunología , Análisis de Supervivencia , Tularemia/inmunología , Tularemia/microbiología , Tularemia/mortalidad
9.
Vaccine ; 34(29): 3396-404, 2016 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-27182819

RESUMEN

Francisella tularensis (Ft) is a Category A biothreat agent for which there currently is no FDA-approved vaccine. Thus, there is a substantial effort underway to develop an effective tularemia vaccine. While it is well established that gender can significantly impact susceptibility to primary infection, the impact of gender on vaccine efficacy is not well established. Thus, development of a successful vaccine against tularemia will require an understanding of the impact gender has on vaccine-induced protection against this organism. In this study, a role for gender in vaccine-induced protection following Ft challenge is identified for the first time. In the present study, mucosal vaccination with inactivated Ft (iFt) LVS elicited gender-based protection in C57BL/6Tac mice against respiratory challenge with Ft LVS. Specifically, vaccinated male mice were more susceptible to subsequent Ft LVS challenge. This increased susceptibility in male mice correlated with increased bacterial burden, increased tissue inflammation, and increased proinflammatory cytokine production late in post-challenge infection. In contrast, improved survival of iFt-vaccinated female mice correlated with reduced bacterial burden and enhanced levels of Ft-specific Abs in serum and broncho-alveolar lavage (BAL) fluid post-challenge. Furthermore, vaccination with a live attenuated vaccine consisting of an Ft LVS superoxide dismutase (SodB) mutant, which has proven efficacious against the highly virulent Ft SchuS4 strain, demonstrated similar gender bias in protection post-Ft SchuS4 challenge. Of particular significance is the fact that these are the first studies to demonstrate that gender differences impact disease outcome in the case of lethal respiratory tularemia following mucosal vaccination. In addition, these studies further emphasize the fact that gender differences must be a serious consideration in any future tularemia vaccine development studies.


Asunto(s)
Vacunas Bacterianas/inmunología , Factores Sexuales , Administración a través de la Mucosa , Animales , Anticuerpos Antibacterianos/sangre , Citocinas/inmunología , Femenino , Francisella tularensis , Inmunidad Celular , Inmunidad Humoral , Masculino , Ratones Endogámicos C57BL , Tularemia/prevención & control , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/inmunología
10.
Vaccine (Auckl) ; 6: 9-23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27200274

RESUMEN

Francisella tularensis (Ft) is a gram-negative intercellular pathogen and category A biothreat agent. However, despite 15 years of strong government investment and intense research focused on the development of a US Food and Drug Administration-approved vaccine against Ft, the primary goal remains elusive. This article reviews research efforts focused on developing an Ft vaccine, as well as a number of important factors, some only recently recognized as such, which can significantly impact the development and evaluation of Ft vaccine efficacy. Finally, an assessment is provided as to whether a US Food and Drug Administration-approved Ft vaccine is likely to be forthcoming and the potential means by which this might be achieved.

11.
Front Immunol ; 7: 677, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28119692

RESUMEN

Francisella tularensis (Ft) is a category A biothreat agent for which there is no Food and Drug Administration-approved vaccine. Ft can survive in a variety of habitats with a remarkable ability to adapt to changing environmental conditions. Furthermore, Ft expresses distinct sets of antigens (Ags) when inside of macrophages (its in vivo host) as compared to those grown in vitro with Mueller Hinton Broth (MHB). However, in contrast to MHB-grown Ft, Ft grown in Brain-Heart Infusion (BHI) more closely mimics the antigenic profile of macrophage-grown Ft. Thus, we anticipated that when used as a vaccine, BHI-grown Ft would provide better protection compared to MHB-grown Ft, primarily due to its greater antigenic similarity to Ft circulating inside the host (macrophages) during natural infection. Our investigation, however, revealed that inactivated Ft (iFt) grown in MHB (iFt-MHB) exhibited superior protective activity when used as a vaccine, as compared to iFt grown in BHI (iFt-BHI). The superior protection afforded by iFt-MHB compared to that of iFt-BHI was associated with significantly lower bacterial burden and inflammation in the lungs and spleens of vaccinated mice. Moreover, iFt-MHB also induced increased levels of Ft-specific IgG. Further evaluation of early immunological cues also revealed that iFt-MHB exhibits increased engagement of Ag-presenting cells including increased iFt binding to dendritic cells, increased expression of costimulatory markers, and increased secretion of pro-inflammatory cytokines. Importantly, these studies directly demonstrate that Ft growth conditions strongly impact Ft vaccine efficacy and that the growth medium used to produce whole cell vaccines to Ft must be a key consideration in the development of a tularemia vaccine.

12.
PLoS One ; 10(6): e0129981, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26114641

RESUMEN

Production of pro-inflammatory cytokines by innate immune cells at the early stages of bacterial infection is important for host protection against the pathogen. Many intracellular bacteria, including Francisella tularensis, the agent of tularemia, utilize the anti-inflammatory cytokine IL-10, to evade the host immune response. It is well established that IL-10 has the ability to inhibit robust antigen presentation by dendritic cells and macrophages, thus suppressing the generation of protective immunity. The pathogenesis of F. tularensis is not fully understood, and research has failed to develop an effective vaccine to this date. In the current study, we hypothesized that F. tularensis polarizes antigen presenting cells during the early stages of infection towards an anti-inflammatory status characterized by increased synthesis of IL-10 and decreased production of IL-12p70 and TNF-α in an IFN-É£-dependent fashion. In addition, F. tularensis drives an alternative activation of alveolar macrophages within the first 48 hours post-infection, thus allowing the bacterium to avoid protective immunity. Furthermore, we demonstrate that targeting inactivated F. tularensis (iFt) to Fcγ receptors (FcÉ£Rs) via intranasal immunization with mAb-iFt complexes, a proven vaccine strategy in our laboratories, reverses the anti-inflammatory effects of the bacterium on macrophages by down-regulating production of IL-10. More specifically, we observed that targeting of iFt to FcγRs enhances the classical activation of macrophages not only within the respiratory mucosa, but also systemically, at the early stages of infection. These results provide important insight for further understanding the protective immune mechanisms generated when targeting immunogens to Fc receptors.


Asunto(s)
Antígenos Bacterianos/inmunología , Francisella tularensis/inmunología , Receptores Fc/metabolismo , Tularemia/inmunología , Tularemia/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Activación de Macrófagos/inmunología , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Noqueados , Receptores de IgG/metabolismo , Tularemia/microbiología
13.
J Immunol Res ; 2015: 840842, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25961064

RESUMEN

Fc gamma receptor IIB (FcγRIIB) is the only Fc gamma receptor (FcγR) which negatively regulates the immune response, when engaged by antigen- (Ag-) antibody (Ab) complexes. Thus, the generation of Ag-specific IgG in response to infection or immunization has the potential to downmodulate immune protection against infection. Therefore, we sought to determine the impact of FcγRIIB on immune protection against Francisella tularensis (Ft), a Category A biothreat agent. We utilized inactivated Ft (iFt) as an immunogen. Naïve and iFt-immunized FcγRIIB knockout (KO) or wildtype (WT) mice were challenged with Ft-live vaccine strain (LVS). While no significant difference in survival between naïve FcγRIIB KO versus WT mice was observed, iFt-immunized FcγRIIB KO mice were significantly better protected than iFt-immunized WT mice. Ft-specific IgA in serum and bronchial alveolar lavage, as well as IFN-γ, IL-10, and TNF-α production by splenocytes harvested from iFt-immunized FcγRIIB KO, were also significantly elevated. In addition, iFt-immunized FcγRIIB KO mice exhibited a reduction in proinflammatory cytokine levels in vivo at 5 days after challenge, which correlates with increased survival following Ft-LVS challenge in published studies. Thus, these studies demonstrate for the first time the ability of FcγRIIB to regulate vaccine-induced IgA production and downmodulate immunity and protection. The immune mechanisms behind the above observations and their potential impact on vaccine development are discussed.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Francisella tularensis/inmunología , Inmunoglobulina A/sangre , Receptores de IgG/genética , Receptores de IgG/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interferón gamma/inmunología , Interleucina-10/inmunología , Interleucina-17/inmunología , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/citología , Tularemia/inmunología , Tularemia/microbiología , Factor de Necrosis Tumoral alfa/inmunología , Vacunación , Vacunas Atenuadas/inmunología
14.
PLoS One ; 10(4): e0124326, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25897786

RESUMEN

Tularemia is caused by a gram-negative, intracellular bacterial pathogen, Francisella tularensis (Ft). The history weaponization of Ft in the past has elevated concerns that it could be used as a bioweapon or an agent of bioterrorism. Since the discovery of Ft, three broad approaches adopted for tularemia vaccine development have included inactivated, live attenuated, or subunit vaccines. Shortcomings in each of these approaches have hampered the development of a suitable vaccine for prevention of tularemia. Recently, we reported an oxidant sensitive mutant of Ft LVS in putative EmrA1 (FTL_0687) secretion protein. The emrA1 mutant is highly sensitive to oxidants, attenuated for intramacrophage growth and virulence in mice. We reported that EmrA1 contributes to oxidant resistance by affecting the secretion of antioxidant enzymes SodB and KatG. This study investigated the vaccine potential of the emrA1 mutant in prevention of respiratory tularemia caused by Ft LVS and the virulent SchuS4 strain in C57BL/6 mice. We report that emrA1 mutant is safe and can be used at an intranasal (i. n.) immunization dose as high as 1x106 CFU without causing any adverse effects in immunized mice. The emrA1 mutant is cleared by vaccinated mice by day 14-21 post-immunization, induces minimal histopathological lesions in lungs, liver and spleen and a strong humoral immune response. The emrA1 mutant vaccinated mice are protected against 1000-10,000LD100 doses of i.n. Ft LVS challenge. Such a high degree of protection has not been reported earlier against respiratory challenge with Ft LVS using a single immunization dose with an attenuated mutant generated on Ft LVS background. The emrA1 mutant also provides partial protection against i.n. challenge with virulent Ft SchuS4 strain in vaccinated C57BL/6 mice. Collectively, our results further support the notion that antioxidants of Ft may serve as potential targets for development of effective vaccines for prevention of tularemia.


Asunto(s)
Vacunas Bacterianas/inmunología , Francisella tularensis/inmunología , Tularemia/prevención & control , Vacunación , Animales , Anticuerpos Antibacterianos/sangre , Proteínas Bacterianas/genética , Citocinas/sangre , Femenino , Francisella tularensis/genética , Hígado/microbiología , Hígado/patología , Pulmón/microbiología , Pulmón/patología , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Bazo/microbiología , Bazo/patología
15.
Infect Immun ; 83(1): 77-89, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312957

RESUMEN

Targeting antigens (Ag) to Fcγ receptors (FcγR) intranasally (i.n.) enhances immunogenicity and protection against intracellular and extracellular pathogens. Specifically, we have demonstrated that targeting fixed (inactivated) Francisella tularensis (iFT) organisms to FcR in mice i.n., with MAb-iFT immune complexes, enhances F. tularensis-specific immune responses and protection against F. tularensis challenge. Furthermore, traditional adjuvant is not required. In addition, we have demonstrated that the increased immunogenicity following the targeting of iFT to FcR is due, in part, to enhanced dendritic cell (DC) maturation, enhanced internalization, and processing and presentation of iFT by DCs, as well as neonatal FcR (FcRn)-enhanced trafficking of iFT from the nasal passage to the nasal mucosa-associated lymphoid tissue (NALT). Using this immunization and challenge model, we expanded on these studies to identify specific in vivo immune responses impacted and enhanced by FcR targeting of iFT i.n. Specifically, the results of this study demonstrate for the first time that targeting iFT to FcR increases the frequency of activated DCs within the lungs of MAb-iFT-immunized mice subsequent to F. tularensis LVS challenge. In addition, the frequency and number of gamma interferon (IFN-γ)-secreting effector memory (EM) CD4(+) T cells elicited by F. tularensis infection (postimmunization) is increased in an interleukin 12 (IL-12)-dependent manner. In summary, these studies build significantly upon previously published work utilizing this vaccine platform. We have identified a number of additional mechanisms by which this novel, adjuvant-independent, FcR-targeted mucosal vaccine approach enhances immunity and protection against infection, while further validating its potential as a universal vaccine platform against mucosal pathogens.


Asunto(s)
Anticuerpos Antibacterianos/metabolismo , Vacunas Bacterianas/inmunología , Francisella tularensis/inmunología , Receptores Fc/metabolismo , Tularemia/prevención & control , Administración Intranasal , Animales , Anticuerpos Monoclonales/metabolismo , Presentación de Antígeno , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Ratones Endogámicos C57BL , Tularemia/inmunología
16.
Vaccine ; 32(40): 5212-20, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25068496

RESUMEN

Dendritic cells (DCs) play a critical role in the generation of adaptive immunity via the efficient capture, processing, and presentation of antigen (Ag) to naïve T cells. Administration of Ag-pulsed DCs is also an effective strategy for enhancing immunity to tumors and infectious disease organisms. Studies have also demonstrated that targeting Ags to Fcγ receptors (FcγR) on Ag presenting cells can enhance humoral and cellular immunity in vitro and in vivo. Specifically, our studies using a Francisella tularensis (Ft) infectious disease vaccine model have demonstrated that targeting immunogens to FcγR via intranasal (i.n.) administration of monoclonal antibody (mAb)-inactivated Ft (iFt) immune complexes (ICs) enhances protection against Ft challenge. Ft is the causative agent of tularemia, a debilitating disease of humans and other mammals and a category A biothreat agent for which there is no approved vaccine. Therefore, using iFt Ag as a model immunogen, we sought to determine if ex vivo targeting of iFt to FcγR on DCs would enhance the potency of i.n. administered iFt-pulsed DCs. In this study, bone marrow-derived DCs (BMDCs) were pulsed ex vivo with iFt or mAb-iFt ICs. Intranasal administration of mAb-iFt-pulsed BMDCs enhanced humoral and cellular immune responses, as well as protection against Ft live vaccine strain (LVS) challenge. Increased protection correlated with increased iFt loading on the BMDC surface as a consequence of FcγR-targeting. However, the inhibitory FcγRIIB had no impact on this enhancement. In conclusion, targeting Ag ex vivo to FcγR on DCs provides a method for enhanced Ag loading of DCs ex vivo, thereby reducing the amount of Ag required, while also avoiding the inhibitory impact of FcγRIIB. Thus, this represents a simple and less invasive strategy for increasing the potency of ex vivo-pulsed DC vaccines against chronic infectious diseases and cancer.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , Receptores de IgG/inmunología , Vacunación/métodos , Administración Intranasal , Animales , Antígenos Bacterianos/inmunología , Vacunas Bacterianas/inmunología , Citocinas/metabolismo , Francisella tularensis , Inmunidad Celular , Inmunidad Humoral , Ratones Endogámicos C57BL , Ratones Noqueados , Tularemia/prevención & control
18.
Immunol Cell Biol ; 91(2): 139-48, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23247654

RESUMEN

We have previously demonstrated that immunization with the inactivated Francisella tularensis, a Category A intracellular mucosal pathogen, combined with IgG2a anti-F. tularensis monoclonal antibody (Ab), enhances protection against subsequent F. tularensis challenge. To understand the mechanism(s) involved, we examined the binding, internalization, presentation, and in vivo trafficking of inactivated F. tularensis in the presence and absence of opsonizing monoclonal Ab. We found that when inactivated F. tularensis is combined with anti-F. tularensis monoclonal Ab, presentation to F. tularensis-specific T cells is enhanced. This enhancement is Fc receptor (FcR)-dependent, and requires a physical linkage between the monoclonal Ab and the inactivated F. tularensis immunogen. This enhanced presentation is due, in part, to enhanced binding and internalization of inactivated F. tularensis by antigen(Ag)-presenting cells, and involves interactions with multiple FcR types. Furthermore, targeting inactivated F. tularensis to FcRs enhances dendritic cell maturation and extends the time period over which Ag-presenting cells stimulate T cells. In vivo trafficking studies reveal enhanced transport of inactivated F. tularensis immunogen to the nasal-associated lymphoid tissue in the presence of monoclonal Ab, which is FcRn-dependent. In summary, these are the first comprehensive studies using a single-vaccine protection model/immunogen to establish the array of mechanisms involved in enhanced immunity/protection mediated by an FcR-targeted mucosal immunogen. These results demonstrate that multiple cellular/immune mechanisms contribute to FcR-enhanced immunity.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Antígenos Bacterianos/inmunología , Francisella tularensis/inmunología , Inmunidad/inmunología , Viabilidad Microbiana/inmunología , Administración Intranasal , Animales , Especificidad de Anticuerpos/inmunología , Presentación de Antígeno/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Endocitosis/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Unión Proteica , Receptores Fc/metabolismo , Especificidad de la Especie , Linfocitos T/inmunología
19.
Retina ; 33(4): 807-11, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23222459

RESUMEN

PURPOSE: Previous investigations have explored molecular differences between proliferative vitreoretinopathy and primary retinal detachment. An exploration of a greater number of molecules might provide novel insight into the biology of this disorder and identify potential therapeutic targets. METHODS: Vitreous specimens were obtained from patients with epiretinal membranes or macular puckers (n = 15), patients with a primary retinal detachment without proliferative vitreoretinopathy (n = 15), and patients with retinal detachments and proliferative vitreoretinopathy (n = 15). A multiplex assay was performed to calculate the concentrations of 48 different cytokines and chemokines, and statistical analyses were performed to identify differences between the groups. RESULTS: Of the 48 molecules that were studied, we identified 10 that were statistically significantly different in cases of proliferative vitreoretinopathy, including interleukins 4, 5, 6, and 15; granulocyte-macrophage colony-stimulating factors; stem cell factor; stem cell growth factor; macrophage inflammatory protein 1α; and interferon γ-induced protein 10. CONCLUSION: Proliferative vitreoretinopathy represents a highly ordered molecular process that involves discrete changes in the concentrations of specific cytokines and chemokines. These molecules may represent novel therapeutic targets.


Asunto(s)
Citocinas/metabolismo , Vitreorretinopatía Proliferativa/metabolismo , Cuerpo Vítreo/metabolismo , Anciano , Membrana Epirretinal/metabolismo , Membrana Epirretinal/cirugía , Humanos , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa Multiplex , Desprendimiento de Retina/metabolismo , Desprendimiento de Retina/cirugía , Vitrectomía , Vitreorretinopatía Proliferativa/cirugía
20.
Ophthalmic Plast Reconstr Surg ; 28(4): 273-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22689135

RESUMEN

PURPOSE: A prior investigation has demonstrated that innate immune-specific cytokines are enriched in idiopathic orbital inflammation (IOI). To further document the role of innate immunity in IOI, the authors sought to determine whether toll-like receptors (TLRs) are present in biopsy specimens of this disorder. METHODS: Immunohistochemical staining for TLR2, TLR3, and TLR4 was performed on biopsy specimens taken from patients with IOI, and the number of TLR-positive cells was counted across five 40× light microscopic fields. These results were compared with an isotype control and with orbital adipose tissue taken from patients without evidence of inflammation. RESULTS: All IOI specimens demonstrated positivity for all 3 TLRs, and sections stained for isotype controls did not demonstrate any positivity. Furthermore, orbital adipose tissue did not demonstrate any significant signal. The mean number of positive cells was 24.4 cells/high power field (hpf; standard deviation = 11.6 cells/hpf), 7.23 cells/hpf (standard deviation = 5.59 cells/hpf), and 11.7 cells/hpf for TLR2, TLR3, and TLR4, respectively. CONCLUSIONS: This study provides the first documentation of TLRs in orbital disease. Toll-like receptors are present in IOI, and IOI may represent an aberrant innate immune response. Interference with TLRs may represent an additional potential therapeutic mechanism in the management of IOI.


Asunto(s)
Seudotumor Orbitario/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Tejido Adiposo/metabolismo , Adulto , Biopsia , Blefaroplastia , Humanos , Inmunidad Innata , Técnicas para Inmunoenzimas , Persona de Mediana Edad , Seudotumor Orbitario/inmunología , Seudotumor Orbitario/patología , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...