Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Immunol Cell Biol ; 95(5): 454-460, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27990018

RESUMEN

We have comprehensively demonstrated using the mouse model that intranasal immunization with recombinant chlamydial protease-like activity factor (rCPAF) leads to a significant reduction in bacterial burden, genital tract pathology and preserves fertility following intravaginal genital chlamydial challenge. In the present report, we evaluated the protective efficacy of rCPAF immunization in guinea pigs, a second animal model for genital chlamydial infection. Using a vaccination strategy similar to the mouse model, we intranasally immunized female guinea pigs with rCPAF plus CpG deoxynucleotides (CpG; as an adjuvant), and challenged intravaginally with C. trachomatis serovar D (CT-D). Immunization with rCPAF/CpG significantly reduced vaginal CT-D shedding and induced resolution of infection by day 24, compared with day 33 in CpG alone treated and challenged animals. Immunization induced robust anti-rCPAF serum IgG 2 weeks following the last immunization, and was sustained at a high-level 4 weeks post challenge. Upregulation of antigen-specific IFN-γ gene expression was observed in rCPAF/CpG-vaccinated splenocytes. Importantly, a significant reduction in inflammation in the genital tissue in rCPAF/CpG-immunized guinea pigs compared with CpG-immunized animals was observed. Taken together, this study provides evidence of the protective efficacy of rCPAF as a vaccine candidate in a second animal model of genital chlamydial infection.


Asunto(s)
Infecciones por Chlamydia/inmunología , Infecciones por Chlamydia/microbiología , Chlamydia trachomatis/fisiología , Endopeptidasas/inmunología , Animales , Infecciones por Chlamydia/genética , Regulación de la Expresión Génica , Genitales/microbiología , Genitales/patología , Cobayas , Inmunización , Inmunoglobulina G/inmunología , Interferón gamma/genética , Interferón gamma/metabolismo , Oligodesoxirribonucleótidos/inmunología
2.
Cell Immunol ; 295(2): 83-91, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25863744

RESUMEN

The human pathogen Chlamydia pneumoniae has been implicated in chronic inflammatory diseases including type 2 diabetes. Therefore, we designed a study to evaluate pancreatic beta cells and mast cells during chlamydial infection. Our study revealed that C. pneumoniae infected mast cells significantly (p<0.005) decreased beta cell ATP and insulin production, in contrast to uninfected mast cells co-cultured with beta cells. Infected mast cells exhibited pyknotic nuclei and active caspase-3 and caspase-1 expression. Additionally, ex vivo analyses of tissues collected from C. pneumoniae infected mice showed increased interleukin-1ß production in splenocytes and pancreatic tissues as was observed with in vitro mast cell-beta cell co-cultures during C. pneumoniae infection. Notably, infected mast cells promoted beta cell destruction. Our findings reveal the negative effect of C. pneumoniae on mast cells, and the consequential impact on pancreatic beta cell function and viability.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydophila pneumoniae/inmunología , Diabetes Mellitus Tipo 2/microbiología , Células Secretoras de Insulina/microbiología , Mastocitos/microbiología , Animales , Caspasa 1/análisis , Caspasa 3/análisis , Supervivencia Celular/inmunología , Infecciones por Chlamydia/microbiología , Técnicas de Cocultivo , Diabetes Mellitus Tipo 2/inmunología , Citometría de Flujo , Células Secretoras de Insulina/inmunología , Interleucina-1beta/análisis , Hígado/citología , Mastocitos/inmunología , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía Confocal , Microscopía Electrónica de Rastreo , Organismos Libres de Patógenos Específicos , Bazo/citología
3.
Infect Immun ; 82(9): 3910-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25001601

RESUMEN

Multidrug-resistant Acinetobacter baumannii is among the most prevalent bacterial pathogens associated with trauma-related wound and bloodstream infections. Although septic shock and disseminated intravascular coagulation have been reported following fulminant A. baumannii sepsis, little is known about the protective host immune response to this pathogen. In this study, we examined the role of PTX3, a soluble pattern recognition receptor with reported antimicrobial properties and stored within neutrophil granules. PTX3 production by murine J774a.1 macrophages was assessed following challenge with A. baumannii strains ATCC 19606 and clinical isolates (CI) 77, 78, 79, 80, and 86. Interestingly, only CI strains 79, 80, and 86 induced PTX3 synthesis in murine J774a.1 macrophages, with greatest production observed following CI 79 and 86 challenge. Subsequently, C57BL/6 mice were challenged intraperitoneally with CI 77 and 79 to assess the role of PTX3 in vivo. A. baumannii strain CI 79 exhibited significantly (P < 0.0005) increased mortality, with an approximate 50% lethal dose (LD50) of 10(5) CFU, while an equivalent dose of CI 77 exhibited no mortality. Plasma leukocyte chemokines (KC, MCP-1, and RANTES) and myeloperoxidase activity were also significantly elevated following challenge with CI 79, indicating neutrophil recruitment/activation associated with significant elevation in serum PTX3 levels. Furthermore, 10-fold-greater PTX3 levels were observed in mouse serum 12 h postchallenge, comparing CI 79 to CI 77 (1,561 ng/ml versus 145 ng/ml), with concomitant severe pathology (liver and spleen) and coagulopathy. Together, these results suggest that elevation of PTX3 is associated with fulminant disease during A. baumannii sepsis.


Asunto(s)
Acinetobacter baumannii/inmunología , Proteína C-Reactiva/inmunología , Proteínas del Tejido Nervioso/inmunología , Sepsis/inmunología , Choque Séptico/inmunología , Infecciones por Acinetobacter/sangre , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/microbiología , Animales , Línea Celular , Quimiocinas/sangre , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/microbiología , Proteínas del Tejido Nervioso/sangre , Neutrófilos/inmunología , Neutrófilos/microbiología , Peroxidasa/sangre , Sepsis/sangre , Sepsis/microbiología , Choque Séptico/sangre , Choque Séptico/mortalidad
4.
J Immunol ; 188(11): 5604-11, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22529298

RESUMEN

TLR signaling is critical for early host defense against pathogens, but the contributions of mast cell TLR-mediated mechanisms and subsequent effector functions during pulmonary infection are largely unknown. We have previously demonstrated that mast cells, through the production of IL-4, effectively control Francisella tularensis replication. In this study, the highly human virulent strain of F. tularensis SCHU S4 and the live vaccine strain were used to investigate the contribution of mast cell/TLR regulation of Francisella. Mast cells required TLR2 for effective bacterial killing, regulation of the hydrolytic enzyme cathepsin L, and for coordination and trafficking of MHC class II and lysosomal-associated membrane protein 2. Infected TLR2(-/-) mast cells, in contrast to wild-type and TLR4(-/-) cells, lacked detectable IL-4 and displayed increased cell death with a 2-3 log increase of F. tularensis replication, but could be rescued with rIL-4 treatment. Importantly, MHC class II and lysosomal-associated membrane protein 2 localization with labeled F. tularensis in the lungs was greater in wild-type than in TLR2(-/-) mice. These results provide evidence for the important effector contribution of mast cells and TLR2-mediated signaling on early innate processes in the lung following pulmonary F. tularensis infection and provide additional insight into possible mechanisms by which intracellular pathogens modulate respiratory immune defenses.


Asunto(s)
Francisella tularensis/crecimiento & desarrollo , Francisella tularensis/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Transducción de Señal/inmunología , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/fisiología , Animales , Muerte Celular/genética , Muerte Celular/inmunología , Interleucina-4/deficiencia , Mastocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Transducción de Señal/genética , Receptor Toll-Like 4/fisiología , Tularemia/inmunología , Tularemia/microbiología , Tularemia/prevención & control
5.
Vaccines (Basel) ; 12(4)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38675740

RESUMEN

Multi-drug-resistant (MDR) Acinetobacter baumannii is an opportunistic pathogen associated with hospital-acquired infections. Due to its environmental persistence, virulence, and limited treatment options, this organism causes both increased patient mortality and incurred healthcare costs. Thus, prophylactic vaccination could be ideal for intervention against MDR Acinetobacter infection in susceptible populations. In this study, we employed immunoinformatics to identify peptides containing both putative B- and T-cell epitopes from proteins associated with A. baumannii pathogenesis. A novel Acinetobacter Multi-Epitope Vaccine (AMEV2) was constructed using an A. baumannii thioredoxin A (TrxA) leading protein sequence followed by five identified peptide antigens. Antisera from A. baumannii infected mice demonstrated reactivity to rAMEV2, and subcutaneous immunization of mice with rAMEV2 produced high antibody titer against the construct as well as peptide components. Immunization results in increased frequency of IL-4-secreting splenocytes indicative of a Th2 response. AMEV2-immunized mice were protected against intranasal challenge with a hypervirulent strain of A. baumannii and demonstrated reduced bacterial burden at 48 h. In contrast, all mock vaccinated mice succumbed to infection within 3 days. Results presented here provide insight into the effectiveness of immunoinformatic-based vaccine design and its potential as an effective strategy to combat the rise of MDR pathogens.

6.
Infect Immun ; 80(6): 2177-85, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22493083

RESUMEN

A licensed vaccine against Francisella tularensis is currently not available. Two Francisella tularensis subsp. novicida (herein referred to by its earlier name, Francisella novicida) attenuated strains, the ΔiglB and ΔfopC strains, have previously been evaluated as potential vaccine candidates against pneumonic tularemia in experimental animals. F. novicida ΔiglB, a Francisella pathogenicity island (FPI) mutant, is deficient in phagosomal escape and intracellular growth, whereas F. novicida ΔfopC, lacking the outer membrane lipoprotein FopC, which is required for evasion of gamma interferon (IFN-γ)-mediated signaling, is able to escape and replicate in the cytosol. To dissect the difference in protective immune mechanisms conferred by these two vaccine strains, we examined the efficacy of the F. novicida ΔiglB and ΔfopC mutants against pulmonary live-vaccine-strain (LVS) challenge and found that both strains provided comparable protection in wild-type, major histocompatibility complex class I (MHC I) knockout, and MHC II knockout mice. However, F. novicida ΔfopC-vaccinated but not F. novicida ΔiglB-vaccinated perforin-deficient mice were more susceptible and exhibited greater bacterial burdens than similarly vaccinated wild-type mice. Moreover, perforin produced by natural killer (NK) cells and release of granzyme contributed to inhibition of LVS replication within macrophages. This NK cell-mediated LVS inhibition was enhanced with anti-F. novicida ΔfopC immune serum, suggesting antibody-dependent cell-mediated cytotoxicity (ADCC) in F. novicida ΔfopC-mediated protection. Overall, this study provides additional immunological insight into the basis for protection conferred by live attenuated F. novicida strains with different phenotypes and supports further investigation of this organism as a vaccine platform for tularemia.


Asunto(s)
Vacunas Bacterianas , Francisella tularensis/inmunología , Granzimas/metabolismo , Perforina/metabolismo , Tularemia/prevención & control , Animales , Proteínas Bacterianas/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/fisiología , Células Cultivadas , Técnicas de Cocultivo , Regulación de la Expresión Génica , Genes MHC Clase I/genética , Genes MHC Clase I/fisiología , Genes MHC Clase II/genética , Genes MHC Clase II/fisiología , Granzimas/genética , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perforina/genética , Tularemia/inmunología , Vacunación , Vacunas Atenuadas
7.
Exp Biol Med (Maywood) ; 247(3): 282-288, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34713732

RESUMEN

Acinetobacter baumannii is a Gram-negative bacterium responsible for many hospital-acquired infections including ventilator-associated pneumonia and sepsis. We have previously identified A. baumannii thioredoxin A protein (TrxA) as a virulence factor with a multitude of functions including reduction of protein disulfides. TrxA plays an important role in resistance to oxidative stress facilitating host immune evasion in part by alteration of type IV pili and cell surface hydrophobicity. Other virulence factors such as outer membrane vesicles (OMV) shed by bacteria have been shown to mediate bacterial intercellular communication and modulate host immune response. To investigate whether OMVs can be modulated by TrxA, we isolated OMVs from wild type (WT) and TrxA-deficient (ΔtrxA) A. baumannii clinical isolate Ci79 and carried out a functional and proteomic comparison. Despite attenuation of ΔtrxA in a mouse challenge model, pulmonary inoculation of ΔtrxA OMVs resulted in increased lung permeability compared to WT OMVs. Furthermore, ΔtrxA OMVs induced more J774 macrophage-like cell death than WT OMVs. This ΔtrxA OMV-mediated cell death was abrogated when cells were incubated with protease-K-treated OMVs suggesting OMV proteins were responsible for cytotoxicity. We therefore compared WT and mutant OMV proteins using proteomic analysis. We observed that up-regulated and unique ΔtrxA OMV proteins consisted of many membrane bound proteins involved in small molecule transport as well as proteolytic activity. Bacterial OmpA, metalloprotease, and fimbrial protein have been shown to enhance mammalian cell apoptosis through various mechanisms. Differential packaging of these proteins in ΔtrxA OMVs may contribute to the increased cytotoxicity observed in this study.


Asunto(s)
Acinetobacter baumannii/patogenicidad , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/patología , Tiorredoxinas/metabolismo , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/aislamiento & purificación , Animales , Membrana Externa Bacteriana/metabolismo , Vesículas Extracelulares/patología , Interacciones Huésped-Patógeno/fisiología , Humanos , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos C57BL , Tiorredoxinas/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
8.
Infect Immun ; 79(7): 2928-35, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21536799

RESUMEN

The immunopathogenesis of Chlamydia trachomatis-induced oviduct pathological sequelae is not well understood. Mice genetically deficient in perforin (perforin(-/-) mice) or tumor necrosis factor alpha (TNF-α) production (TNF-α(-/-) mice) displayed comparable vaginal chlamydial clearance rates but significantly reduced oviduct pathology (hydrosalpinx) compared to that of wild-type mice. Since both perforin and TNF-α are effector mechanisms of CD8(+) T cells, we evaluated the role of CD8(+) T cells during genital Chlamydia muridarum infection and oviduct sequelae. Following vaginal chlamydial challenge, (i) mice deficient in TAP I (and therefore the major histocompatibility complex [MHC] I pathway and CD8(+) T cells), (ii) wild-type mice depleted of CD8(+) T cells, and (iii) mice genetically deficient in CD8 (CD8(-/-) mice) all displayed similar levels of vaginal chlamydial clearance but significantly reduced hydrosalpinx, compared to those of wild-type C57BL/6 mice, suggesting a role for CD8(+) T cells in chlamydial pathogenesis. Repletion of CD8(-/-) mice with wild-type or perforin(-/-), but not TNF-α(-/-), CD8(+) T cells at the time of challenge restored hydrosalpinx to levels observed in wild-type C57BL/6 mice, suggesting that TNF-α production from CD8(+) T cells is important for pathogenesis. Additionally, repletion of TNF-α(-/-) mice with TNF-α(+/+) CD8(+) T cells significantly enhanced the incidence of hydrosalpinx and oviduct dilatation compared to those of TNF-α(-/-) mice but not to the levels found in wild-type mice, suggesting that TNF-α production from CD8(+) T cells and non-CD8(+) cells cooperates to induce optimal oviduct pathology following genital chlamydial infection. These results provide compelling new evidence supporting the contribution of CD8(+) T cells and TNF-α production to Chlamydia-induced reproductive tract sequelae.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Infecciones por Chlamydia/inmunología , Chlamydia muridarum , Trompas Uterinas/patología , Enfermedades de los Genitales Femeninos/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Linfocitos T CD8-positivos/inmunología , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Trompas Uterinas/microbiología , Femenino , Enfermedades de los Genitales Femeninos/microbiología , Enfermedades de los Genitales Femeninos/patología , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Perforina/biosíntesis , Perforina/genética , Factor de Necrosis Tumoral alfa/genética , Vagina/microbiología
9.
Lab Invest ; 91(10): 1530-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21769086

RESUMEN

Respiratory dysfunction in adults has been correlated with neonatal Chlamydia trachomatis pneumonia in several studies, but a causal association has not been clearly demonstrated. In this study, we examined radial alveolar counts (RACs) by microscopy, and airway and parenchymal lung function using a small animal ventilator in juvenile (5 weeks age) and adult (8 weeks age) BALB/c mice challenged as neonates with Chlamydia muridarum (C. mur) on day 1 or day 7 after birth, representing saccular (human pre-term neonates) and alveolar (human term neonates) stages of lung development, respectively. Pups challenged with C. mur on either day 1 or 7 after birth demonstrated significantly enhanced airway hyperreactivity and lung compliance, both as juveniles (5 weeks age) and adults (8 weeks age), compared with mock-challenged mice. Moreover, mice challenged neonatally with Chlamydia displayed significantly reduced RACs, suggesting emphysematous changes. Antimicrobial treatment during the neonatal infection induced early bacterial clearance and partially ameliorated the Chlamydia-induced lung dysfunction as adults. These results suggest that neonatal chlamydial pneumonia, especially in pre-term neonates, is a cause of respiratory dysfunction continuing into adulthood, and that antimicrobial administration may be partially effective in preventing the adverse respiratory sequelae in adulthood. The results of our studies also emphasize the importance of prenatal screening and treatment of pregnant women for C. trachomatis in order to prevent the infection of neonates.


Asunto(s)
Envejecimiento , Animales Recién Nacidos , Infecciones por Chlamydia/patología , Infecciones por Chlamydia/fisiopatología , Neumonía Bacteriana/patología , Neumonía Bacteriana/fisiopatología , Sistema Respiratorio/patología , Sistema Respiratorio/fisiopatología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Antibacterianos/administración & dosificación , Hiperreactividad Bronquial/etiología , Infecciones por Chlamydia/complicaciones , Infecciones por Chlamydia/tratamiento farmacológico , Esquema de Medicación , Eritromicina/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Rendimiento Pulmonar , Ratones , Ratones Endogámicos BALB C , Neumonía Bacteriana/complicaciones , Neumonía Bacteriana/tratamiento farmacológico , Alveolos Pulmonares/crecimiento & desarrollo
10.
Appl Environ Microbiol ; 77(12): 4119-25, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21531837

RESUMEN

Proteolytic cleavage activation of influenza virus hemagglutinin (HA0) is required for cell entry via receptor-mediated endocytosis. Despite numerous studies describing bacterial protease-mediated influenza A viral activation in mammals, very little is known about the role of intestinal bacterial flora of birds in hemagglutinin cleavage/activation. Therefore, the cloaca of wild waterfowl was examined for (i) representative bacterial types and (ii) their ability to cleave in a "trypsin-like" manner the precursor viral hemagglutinin molecule (HA0). Using radiolabeled HA0, bacterial secretion-mediated trypsin-like conversion of HA0 to HA1 and HA2 peptide products was observed to various degrees in 42 of 44 bacterial isolates suggestive of influenza virus activation in the cloaca of wild waterfowl. However, treatment of uncleaved virus with all bacterial isolates gave rise to substantially reduced emergent virus progeny compared with what was expected. Examination of two isolates exhibiting pronounced trypsin-like conversion of HA0 to HA1 and HA2 peptide products and low infectivity revealed lipase activity to be present. Because influenza virus possesses a complex lipid envelope, the presence of lipid hydrolase activity could in part account for the observed less-than-expected level of viable progeny. A thorough characterization of respective isolate protease HA0 hydrolysis products as well as other resident activities (i.e., lipase) is ongoing such that the role of these respective contributors in virus activation/inactivation can be firmly established.


Asunto(s)
Bacterias/metabolismo , Cloaca/microbiología , Patos/microbiología , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Orthomyxoviridae/patogenicidad , Factores de Virulencia/metabolismo , Animales , Bacterias/enzimología , Proteínas Bacterianas/metabolismo , ADN Bacteriano/química , ADN Bacteriano/genética , Lipasa/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia de ADN
11.
Cytokine ; 55(2): 211-20, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21565523

RESUMEN

Mast cells have classically been implicated in the triggering of allergic and anaphylactic reactions. However, recent findings have elucidated the ability of these cells to selectively release a variety of cytokines leading to bacterial clearance through neutrophil and dendritic cell mobilization, and suggest an important role in innate host defenses. Our laboratory has established a primary bone marrow derived mast cell-macrophage co-culture system and found that mast cells mediated a significant inhibition of Francisella tularensis live vaccine strain (LVS) uptake and replication within macrophages through contact and the secreted product interleukin-4 (IL-4). In this study, we utilized P815 mast cells and J774 macrophages to further investigate whether mast cell activation by non-FcεR driven signals could produce IL-4 and control intramacrophage LVS replication. P815 supernatants collected upon activation by the mast cell activating peptide MP7, as well as P815 cells co-cultured with J774 macrophages, exhibited marked inhibition of bacterial uptake and replication, which correlated with the production of IL-4. The inhibition noted in vitro was titratable and preserved at ratios relevant to cellular infiltration events following pulmonary challenge. Collectively, our data suggest that both primary mast cell and P815 mast cell (lacking FcεR) secreted IL-4 can control intramacrophage Francisella replication.


Asunto(s)
Francisella tularensis/fisiología , Interleucina-4/metabolismo , Macrófagos/microbiología , Mastocitos/metabolismo , Receptores de IgE/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/fisiología , Células Cultivadas , Técnicas de Cocultivo , Francisella tularensis/inmunología , Macrófagos/citología , Macrófagos/inmunología , Mastocitos/citología , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Receptores de IgE/genética , Tularemia/inmunología , Factor de Necrosis Tumoral alfa/inmunología
12.
Proc Natl Acad Sci U S A ; 105(27): 9313-8, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18591675

RESUMEN

Francisella tularensis is an intracellular, Gram-negative bacterium that is the causative agent of pulmonary tularemia. The pathogenesis and mechanisms related to innate resistance against F. tularensis are not completely understood. Mast cells are strategically positioned within mucosal tissues, the major interface with the external environment, to initiate innate responses at the site of infection. Mast cell numbers in the cervical lymph nodes and the lungs progressively increased as early as 48 h after intranasal F. tularensis live vaccine strain (LVS) challenge. We established a primary bone marrow-derived mast cell-macrophage coculture system and found that mast cells significantly inhibit F. tularensis LVS uptake and growth within macrophages. Importantly, mice deficient in either mast cells or IL-4 receptor displayed greater susceptibility to the infection when compared with corresponding wild-type animals. Contact-dependent events and secreted products including IL-4 from mast cells, and IL-4 production from other cellular sources, appear to mediate the observed protective effects. These results demonstrate a previously unrecognized role for mast cells and IL-4 and provide a new dimension to our understanding of the innate immune mechanisms involved in controlling intramacrophage Francisella replication.


Asunto(s)
Inhibición de Contacto , Replicación del ADN , Francisella tularensis/inmunología , Interleucina-4/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Mastocitos/citología , Administración Intranasal , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/inmunología , Adhesión Celular , Inmunidad Innata/inmunología , Espacio Intracelular/microbiología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Mastocitos/inmunología , Mastocitos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Tularemia/inmunología , Tularemia/microbiología
13.
Infect Immun ; 78(9): 3942-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20605976

RESUMEN

We have previously demonstrated the efficacy of recombinant chlamydial protease-like activity factor (rCPAF; a secreted chlamydial protein) in inducing antigen-specific CD4+ T cell/gamma interferon (IFN-gamma)-mediated but not antibody-mediated chlamydial clearance and reduction of upper genital tract (UGT) pathological sequelae. Since chlamydial integral antigens may induce neutralizing antibody protection, we further evaluated induction of protective immunity using a combination of rCPAF and UV-inactivated chlamydial elementary bodies (UV-EB) against vaginal chlamydial challenge in comparison to immunization with the individual components or live EB. The rCPAF-UV-EB immunization induced a significantly enhanced anti-UV-EB cellular and antibody response and a reduced anti-CPAF cellular and antibody response, compared to immunization with the respective individual components. Moreover, vaccination with UV-EB and rCPAF-UV-EB induced serum antibodies that neutralized chlamydial infectivity. The rCPAF-UV-EB immunization resulted in a significant reduction of vaginal chlamydial shedding and induced earlier bacterial clearance than vaccination of mice with the individual components. Importantly, the UGT sequelae were significantly reduced in mice immunized with rCPAF or rCPAF-UV-EB, but not in those immunized with UV-EB alone, and approached the levels of protection induced by live EB. These results collectively suggest that a combination of neutralizing antibodies induced by integral chlamydial antigens and cell-mediated responses induced by secreted proteins such as CPAF induces optimal protective immunity against genital chlamydial infections.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Infecciones por Chlamydia/prevención & control , Chlamydia muridarum/inmunología , Enfermedades de los Genitales Femeninos/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Cricetinae , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/inmunología , Vacunas Sintéticas/inmunología , Vagina/microbiología
14.
PLoS One ; 14(7): e0218505, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31265467

RESUMEN

The Gram-negative pathogen, Acinetobacter baumannii has emerged as a global nosocomial health threat affecting the majority of hospitals in the U.S. and abroad. The redox protein thioredoxin has been shown to play several roles in modulation of cellular functions affecting various virulence factors in Gram-negative pathogens. This study aims to explore the role of thioredoxin-A protein (TrxA) in A. baumannii virulence. We determined that deletion of the TrxA gene did not significantly affect resistance to environmental stressors such as temperature, salt, and pH. However, TrxA was critical for survival in the presence of elevated levels of hydrogen peroxide. Lack of TrxA was associated with decreased expression of type IV pili related genes and an inability to undergo normal twitching motility. Interestingly, the TrxA-null mutant was able to form biofilms better than the wildtype (WT) and was observed to be significantly less virulent than the WT in a pulmonary infection model. These results are supportive of thioredoxin playing a key role in A. baumannii virulence.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Proteínas Bacterianas , Fimbrias Bacterianas , Tiorredoxinas , Factores de Virulencia , Infecciones por Acinetobacter/genética , Infecciones por Acinetobacter/metabolismo , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/patogenicidad , Animales , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Femenino , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Ratones , Tiorredoxinas/biosíntesis , Tiorredoxinas/genética , Factores de Virulencia/biosíntesis , Factores de Virulencia/genética
15.
Front Microbiol ; 10: 2849, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921031

RESUMEN

Acinetobacter baumannii, a Gram-negative coccobacillus, has become a prevalent nosocomial health threat affecting the majority of hospitals both in the U.S. and around the globe. Microbial cell surface hydrophobicity (CSH) has previously been correlated with virulence, uptake by immune cells, and attachment to epithelial cells. A mutant strain of A. baumannii (ΔtrxA) lacking the redox protein thioredoxin A was found to be more hydrophobic than its wild type (WT) and complemented counterparts, as measured by both Microbial Adhesion to Hydrocarbon (MATH) and salt aggregation. The hydrophobicity of the mutant could be abrogated through treatment with sodium cyanoborohydride (SCBH). This modulation correlated with reduction of disulfide bonds, as SCBH was able to reduce 5,5'-dithio-bis-[2-nitrobenzoic acid] and treatment with the known disulfide reducer, ß-mercaptoethanol, also decreased ΔtrxA CSH. Additionally, the ΔtrxA mutant was more readily taken up than WT by J774 macrophages and this differential uptake could be abrogated though SCBH treatment. When partitioned into aqueous and hydrophobic phases, ΔtrxA recovered from the hydrophobic partition was phagocytosed more readily than from the aqueous phase further supporting the contribution of CSH to A. baumannii uptake by phagocytes. A second Gram-negative bacterium, Francisella novicida, also showed the association of TrxA deficiency (FnΔtrxA) with increased hydrophobicity and uptake by J774 cells. We previously have demonstrated that modification of the type IV pilus system (T4P) was associated with the A. baumannii ΔtrxA phenotype, and the Francisella FnΔtrxA mutant also was found to have a marked T4P deficiency. Interestingly, a F. novicida mutant lacking pilT also showed increased hydrophobicity over FnWT. Collective evidence presented in this study suggests that Gram-negative bacterial thioredoxin mediates CSH through multiple mechanisms including disulfide-bond reduction and T4P modulation.

16.
mBio ; 9(4)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29991584

RESUMEN

Multidrug-resistant Acinetobacter baumannii is among the most common causes of infectious complications associated with combat-related trauma in military personnel serving overseas. However, little is currently known about its pathogenesis. While the gastrointestinal (GI) tract has been found to be a major reservoir for A. baumannii, as well as to potentially contribute to development of multidrug resistance, no studies have addressed the mechanisms involved in gut colonization. In this study, we address this critical gap in knowledge by first assessing the interaction between secretory IgA (SIgA), the principal humoral immune defense on mucosal surfaces, and the A. baumannii clinical isolate Ci79. Surprisingly, SIgA appeared to enhance A. baumannii GI tract colonization, in a process mediated by bacterial thioredoxin A (TrxA), as evidenced by reduction of bacterial attachment in the presence of TrxA inhibitors. Additionally, a trxA targeted deletion mutant (ΔtrxA) showed reduced bacterial burdens within the GI tract 24 h after oral challenge by in vivo live imaging, along with loss of thiol-reductase activity. Surprisingly, not only was GI tract colonization greatly reduced but the associated 50% lethal dose (LD50) of the ΔtrxA mutant was increased nearly 100-fold in an intraperitoneal sepsis model. These data suggest that TrxA not only mediates A. baumannii GI tract colonization but also may contribute to pathogenesis in A. baumannii sepsis following escape from the GI tract under conditions when the intestinal barrier is compromised, as occurs with cases of severe shock and trauma.IMPORTANCEAcinetobacter baumannii is an emerging bacterial pathogen recently classified as a serious threat to U.S. and global health by both the Centers for Disease Control and Prevention and the World Health Organization. It also is one of the leading causes of combat-related infections associated with injured military personnel serving overseas. Little is known regarding mechanisms of gastrointestinal tract colonization despite this site being shown to serve as a reservoir for multidrug-resistant (MDR) A. baumannii isolates. Here, we establish that secretory IgA, the major immunoglobulin of mucosal surfaces, promotes A. baumannii GI tract colonization via bacterial thioredoxin A as evidenced through significant reduction in colonization in IgA-deficient animals. Additionally, bacterial colonization and mortality were significantly reduced in animals challenged with a thioredoxin A-deficient A. baumannii mutant. Combined, these data suggest that thioredoxin A is a novel virulence factor, for which antithioredoxin therapies could be developed, for this important multidrug-resistant pathogen.


Asunto(s)
Acinetobacter baumannii/fisiología , Adhesión Bacteriana , Tracto Gastrointestinal/microbiología , Inmunoglobulina A Secretora/metabolismo , Factores Inmunológicos/metabolismo , Tiorredoxinas/metabolismo , Factores de Virulencia/metabolismo , Acinetobacter baumannii/genética , Acinetobacter baumannii/patogenicidad , Animales , Modelos Animales de Enfermedad , Eliminación de Gen , Ratones Endogámicos C57BL , Oxidación-Reducción , Sepsis/microbiología , Sepsis/patología , Análisis de Supervivencia , Tiorredoxinas/antagonistas & inhibidores , Tiorredoxinas/genética , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/genética
17.
Innate Immun ; 24(3): 152-162, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29482417

RESUMEN

Macrophages are important innate immune cells that respond to microbial insults. In response to multi-bacterial infection, the macrophage activation state may change upon exposure to nascent mediators, which results in different bacterial killing mechanism(s). In this study, we utilized two respiratory bacterial pathogens, Mycobacterium bovis (Bacillus Calmette Guerin, BCG) and Francisella tularensis live vaccine strain (LVS) with different phagocyte evasion mechanisms, as model microbes to assess the influence of initial bacterial infection on the macrophage response to secondary infection. Non-activated (M0) macrophages or activated M2-polarized cells (J774 cells transfected with the mouse IL-4 gene) were first infected with BCG for 24-48 h, subsequently challenged with LVS, and the results of inhibition of LVS replication in the macrophages was assessed. BCG infection in M0 macrophages activated TLR2-MyD88 and Mincle-CARD9 signaling pathways, stimulating nitric oxide (NO) production and enhanced killing of LVS. BCG infection had little effect on LVS escape from phagosomes into the cytosol in M0 macrophages. In contrast, M2-polarized macrophages exhibited enhanced endosomal acidification, as well as inhibiting LVS replication. Pre-infection with BCG did not induce NO production and thus did not further reduce LVS replication. This study provides a model for studies of the complexity of macrophage activation in response to multi-bacterial infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Coinfección/inmunología , Macrófagos/inmunología , Fagosomas/inmunología , Animales , Polaridad Celular , Endosomas/inmunología , Humanos , Evasión Inmune , Inmunidad Innata/inmunología , Interleucina-4/biosíntesis , Ratones , Infecciones por Mycobacterium/inmunología , Mycobacterium bovis/inmunología , Óxido Nítrico/biosíntesis , Transducción de Señal/inmunología , Transfección , Tularemia/inmunología , Vacunas Vivas no Atenuadas
18.
Vaccine ; 35(26): 3387-3394, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28522011

RESUMEN

Multi-drug resistant Acinetobacter baumannii (MDR-Ab), an opportunistic pathogen associated with nosocomial and combat related infections, has a high mortality due to its virulence and limited treatment options. Deletion of the thioredoxin gene (TrxA) from a clinical isolate of MDR-Ab resulted in a 100-fold increase in 50% lethal dose (LD50) in a systemic challenge murine model. Thus, we investigated the potential use of this attenuated strain as a live vaccine against MDR-Ab. Mice were vaccinated by subcutaneous (s.c.) injection of 2×105 CFU of the ΔtrxA mutant, boosted 14days later with an equivalent inoculum, and then challenged 30days post-vaccination by i.p. injection with 10 LD50 of the wild type (WT) Ci79 strain. Efficacy of vaccination was evaluated by monitoring MDR-Ab specific antibody titers and cytokine production, observing pathology and organ burdens after WT challenge, and measuring levels of serum pentraxin-3, a molecular correlate of A. baumannii infection severity, before and after challenge. Mice vaccinated with ΔtrxA were fully protected against the lethal challenge of WT. However, minimal immunoglobulin class switching was observed with IgM predominating. Spleens harvested from vaccinated mice exhibited negligible levels of IL-4, IFN-γ and IL-17 production when stimulated with UV-inactivated WT Ci79. Importantly, tissues obtained from vaccinated mice displayed reduced pathology and organ burden compared to challenged non-vaccinated mice. Additionally, serum pentraxin-3 concentrations were not increased 24h after challenge in vaccinated mice, correlating with reduction of WT MDR-Ab infection in ΔtrxA immunized mice. Furthermore, passive immunization with ΔtrxA-immune sera provided protection against lethal systemic Ci79 challenge. Collectively, the defined live attenuated ΔtrxA strain is a vaccine candidate against emerging MDR Acinetobacter infection.


Asunto(s)
Infecciones por Acinetobacter/prevención & control , Acinetobacter baumannii/inmunología , Vacunas Bacterianas/inmunología , Tiorredoxinas/genética , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/genética , Animales , Anticuerpos Antibacterianos/sangre , Carga Bacteriana , Proteína C-Reactiva/análisis , Citocinas/inmunología , Eliminación de Gen , Inmunidad Mucosa , Inmunización Pasiva , Cambio de Clase de Inmunoglobulina , Inmunoglobulina M/sangre , Ratones , Ratones Endogámicos C57BL , Sepsis/prevención & control , Componente Amiloide P Sérico/análisis , Bazo/inmunología , Vacunas Atenuadas/inmunología , Factores de Virulencia/genética
19.
Innate Immun ; 22(7): 567-74, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27554051

RESUMEN

Understanding innate immune intercellular communication following microbial infection remains a key biological issue. Using live cell imaging, we demonstrate that mast cells actively extend cellular projections to sample the macrophage periphery during Francisella tularensis LVS infection. Mast cell MHCII(hi) expression was elevated from less than 1% to 13% during LVS infection. Direct contact during co-culture with macrophages further increased mast cell MHCII(hi) expression to approximately 87%. Confocal analyses of the cellular perimeter revealed mast cell caspase-1 was localized in close proximity with FcɛRI in uninfected mast cells, and repositioned to clustered regions upon LVS infection. Importantly, mast cell FcɛRI-encompassed vesicles are transferred to macrophages by trogocytosis, and macrophage caspase-1 expression is further up-regulated upon direct contact with mast cells. Our study reveals direct cellular interactions between innate cells that may impact the function of caspase-1, a known sensor of microbial danger and requirement for innate defense against many pathogenic microbes including F. tularensis.


Asunto(s)
Caspasa 1/metabolismo , Vesículas Citoplasmáticas/metabolismo , Francisella tularensis/inmunología , Macrófagos/inmunología , Mastocitos/inmunología , Receptores de IgE/metabolismo , Tularemia/inmunología , Animales , Comunicación Celular , Extensiones de la Superficie Celular/metabolismo , Extensiones de la Superficie Celular/patología , Células Cultivadas , Técnicas de Cocultivo , Inmunidad Innata , Macrófagos/microbiología , Mastocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Transporte de Proteínas
20.
Metabolomics ; 12(4)2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27642272

RESUMEN

INTRODUCTION: Chlamydia trachomatis (Ct), is the leading cause of sexually transmitted infections worldwide. Host transcriptomic- or proteomic profiling studies have identified key molecules involved in establishment of Ct infection or the generation of anti Ct-immunity. However, the contribution of the host metabolome is not known. OBJECTIVES: The objective of this study was to determine the contribution of host metabolites in genital Ct infection. METHODS: We used high-performance liquid chromatography-mass spectrometry, and mapped lipid profiles in genital swabs obtained from female guinea pigs at days 3, 9, 15, 30 and 65 post Ct serovar D intravaginal infection. RESULTS: Across all time points assessed, 13 distinct lipid species including choline, ethanolamine and glycerol were detected. Amongst these metabolites, phosphatidylcholine (PC) was the predominant phospholipid detected from animals actively shedding bacteria i.e., at 3, 9, and 15 days post infection. However, at days 30 and 65 when the animals had cleared the infection, PC was observed to be decreased compared to previous time points. Mass spectrometry analyses of PC produced in guinea pigs (in vivo) and 104C1 guinea pig cell line (in vitro) revealed distinct PC species following Ct D infection. Amongst these, PC 16:0/18:1 was significantly upregulated following Ct D infection (p < 0.05, >twofold change) in vivo and in vitro infection models investigated in this report. Exogenous addition of PC 16:0/18:1 resulted in significant increase in Ct D in Hela 229 cells. CONCLUSION: This study demonstrates a role for host metabolite, PC 16:0/18:1 in regulating genital Ct infection in vivo and in vitro.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA