Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
FASEB J ; 36(3): e22197, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35147989

RESUMEN

Neonatal meningitis-associated Escherichia coli (NMEC) is among the leading causes of bacterial meningitis and sepsis in newborn infants. Several virulence factors have been identified as common among NMEC, and have been shown to play an important role in the development of bacteremia and/or meningitis. However, there is significant variability in virulence factor expression between NMEC isolates, and relatively little research has been done to assess the impact of variable virulence factor expression on immune cell activation and the outcome of infection. Here, we investigated the role of NMEC strain-dependent P2X receptor (P2XR) signaling on the outcome of infection in neonatal mice. We found that alpha-hemolysin (HlyA)-expressing NMEC (HlyA+ ) induced robust P2XR-dependent macrophage cell death in vitro, while HlyA- NMEC did not. P2XR-dependent cell death was inflammasome independent, suggesting an uncoupling of P2XR and inflammasome activation in the context of NMEC infection. In vivo inhibition of P2XRs was associated with increased mortality in neonatal mice infected with HlyA+ NMEC, but had no effect on the survival of neonatal mice infected with HlyA- NMEC. Furthermore, we found that P2XR-dependent protection against HlyA+ NMEC in vivo required macrophages, but not neutrophils or NLRP3. Taken together, these data suggest that HlyA+ NMEC activates P2XRs which in turn confers macrophage-dependent protection against infection in neonates. In addition, our findings indicate that strain-dependent virulence factor expression should be taken into account when studying the immune response to NMEC.


Asunto(s)
Proteínas de Escherichia coli/toxicidad , Proteínas Hemolisinas/toxicidad , Inflamasomas/metabolismo , Meningitis por Escherichia coli/metabolismo , Sepsis Neonatal/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animales , Células Cultivadas , Escherichia coli K12 , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/metabolismo , Macrófagos/metabolismo , Meningitis por Escherichia coli/microbiología , Ratones , Ratones Endogámicos C57BL , Sepsis Neonatal/microbiología , Receptores Purinérgicos P2X/genética
2.
Infect Immun ; 89(10): e0015621, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34125603

RESUMEN

Brucellosis is one of the most common global zoonoses and is caused by facultative intracellular bacteria of the genus Brucella. Numerous studies have found that MyD88 signaling contributes to protection against Brucella; however, the underlying mechanism has not been entirely defined. Here, we show that MyD88 signaling in hematopoietic cells contributes both to inflammation and to control of Brucella melitensis infection in vivo. While the protective role of MyD88 in Brucella infection has often been attributed to promotion of gamma interferon (IFN-γ) production, we found that MyD88 signaling restricts host colonization by B. melitensis even in the absence of IFN-γ. In vitro, we show that MyD88 promotes macrophage glycolysis in response to B. melitensis. Interestingly, a B. melitensis mutant lacking the glucose transporter, GluP, was more highly attenuated in MyD88-/- than in wild-type mice, suggesting MyD88 deficiency results in an increased availability of glucose in vivo, which Brucella can exploit via GluP. Metabolite profiling of macrophages identified several metabolites regulated by MyD88 in response to B. melitensis, including itaconate. Subsequently, we found that itaconate has antibacterial effects against Brucella and also regulates the production of proinflammatory cytokines in B. melitensis-infected macrophages. Mice lacking the ability to produce itaconate were also more susceptible to B. melitensis in vivo. Collectively, our findings indicate that MyD88-dependent changes in host metabolism contribute to control of Brucella infection.


Asunto(s)
Brucelosis/metabolismo , Glucosa/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Succinatos/metabolismo , Animales , Brucella melitensis/patogenicidad , Brucelosis/microbiología , Citocinas/metabolismo , Glucólisis/fisiología , Interferón gamma/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología
3.
Infect Immun ; 86(9)2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29941463

RESUMEN

Brucellosis, caused by the intracellular bacterial pathogen Brucella, is a zoonotic disease for which arthritis is the most common focal complication in humans. Here we investigated the role of inflammasomes and their effectors, including interleukin-1 (IL-1), IL-18, and pyroptosis, on inflammation and control of infection during Brucella-induced arthritis. Early in infection, both caspase-1 and caspase-11 were found to initiate joint inflammation and proinflammatory cytokine production. However, by 1 week postinfection, caspase-1 and caspase-11 also contributed to control of Brucella joint infection. Inflammasome-dependent restriction of Brucella joint burdens did not require AIM2 (absent in melanoma 2) or NLRP3 (NLR family, pyrin domain containing 3). IL-1R had a modest effect on Brucella-induced joint swelling, but mice lacking IL-1R were not impaired in their ability to control infection of the joint by Brucella In contrast, IL-18 contributed to the initiation of joint swelling and control of joint Brucella infection. Caspase1/11-dependent cell death was observed in vivo, and in vitro studies demonstrated that both caspase-1 and caspase-11 induce pyroptosis, which limited Brucella infection in macrophages. Brucella lipopolysaccharide alone was also able to induce caspase-11-dependent pyroptosis. Collectively, these data demonstrate that inflammasomes induce inflammation in an IL-18-dependent manner and that inflammasome-dependent IL-18 and pyroptosis restrict Brucella infection.


Asunto(s)
Brucelosis/inmunología , Caspasa 1/fisiología , Caspasas/fisiología , Inflamasomas/fisiología , Inflamación/inmunología , Artropatías/inmunología , Piroptosis/fisiología , Animales , Caspasas Iniciadoras , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos
4.
J Leukoc Biol ; 102(6): 1441-1450, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28951422

RESUMEN

Francisella tularensis is a highly infectious intracellular bacterium that causes the potentially fatal disease tularemia. We used mice with conditional MyD88 deficiencies to investigate cellular and molecular mechanisms by which MyD88 restricts type A F. tularensis infection. F. tularensis-induced weight loss was predominately dependent on MyD88 signaling in nonhematopoietic cells. In contrast, MyD88 signaling in hematopoietic cells, but not in myeloid and dendritic cells, was essential for control of F. tularensis infection in tissue. Myeloid and dendritic cell MyD88 deficiency also did not markedly impair cytokine production during infection. Although the production of IL-12 or -18 was not significantly reduced in hematopoietic MyD88-deficient mice, IFN-γ production was abolished in these animals. In addition, neutralization studies revealed that control of F. tularensis infection mediated by hematopoietic MyD88 was entirely dependent on IFN-γ. Although IL-18 production was not significantly affected by MyD88 deficiency, IL-18 was essential for IFN-γ production and restricted bacterial replication in an IFN-γ-dependent manner. Caspase-1 was also found to be partially necessary for the production of IL-18 and IFN-γ and for control of F. tularensis replication. Our collective data show that the response of leukocytes to caspase-1-dependent IL-18 via MyD88 is critical, whereas MyD88 signaling in myeloid and dendritic cells is dispensable for IFN-γ-dependent control of type A F. tularensis infection.


Asunto(s)
Francisella tularensis/fisiología , Hematopoyesis , Interferón gamma/metabolismo , Interleucina-18/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Tularemia/metabolismo , Tularemia/microbiología , Animales , Caspasa 1/metabolismo , Células Dendríticas/metabolismo , Francisella tularensis/patogenicidad , Mediadores de Inflamación/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/metabolismo , Transducción de Señal , Tularemia/patología , Virulencia
5.
Infect Immun ; 81(9): 3099-105, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23774604

RESUMEN

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


Asunto(s)
Vacunas Bacterianas/inmunología , Vacunas Bacterianas/farmacología , Francisella tularensis/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Tularemia/genética , Tularemia/microbiología , Animales , Caspasa 1/genética , Caspasa 1/inmunología , Femenino , Francisella tularensis/patogenicidad , Interferón gamma/genética , Interferón gamma/inmunología , Subunidad p19 de la Interleucina-23/genética , Subunidad p19 de la Interleucina-23/inmunología , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores de Interleucina-17/inmunología , Tularemia/inmunología , Tularemia/prevención & control , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/farmacología
6.
PLoS Pathog ; 8(3): e1002587, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438809

RESUMEN

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


Asunto(s)
Arecaceae/química , Inmunidad Innata/efectos de los fármacos , Melioidosis/prevención & control , Neumonía/tratamiento farmacológico , Polisacáridos/farmacología , Tularemia/prevención & control , Administración Intranasal , Animales , Burkholderia pseudomallei/efectos de los fármacos , Burkholderia pseudomallei/inmunología , Modelos Animales de Enfermedad , Femenino , Francisella tularensis/efectos de los fármacos , Francisella tularensis/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Longevidad/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Melioidosis/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Extractos Vegetales/administración & dosificación , Extractos Vegetales/farmacología , Neumonía/inmunología , Neumonía/microbiología , Polisacáridos/administración & dosificación , Polisacáridos/aislamiento & purificación , Tularemia/inmunología
7.
J Autoimmun ; 37(4): 328-41, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22018711

RESUMEN

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


Asunto(s)
Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Proteínas de la Mielina/metabolismo , Proteínas Virales/metabolismo , Traslado Adoptivo , Animales , Antígenos Ly/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/fisiopatología , Tolerancia Inmunológica , Interferones/metabolismo , Activación de Linfocitos , Ratones , Proteínas de la Mielina/genética , Proteínas de la Mielina/inmunología , Glicoproteína Mielina-Oligodendrócito , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Recurrencia , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Proteínas Virales/genética , Proteínas Virales/inmunología
8.
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
9.
J Leukoc Biol ; 90(6): 1043-54, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21693591

RESUMEN

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


Asunto(s)
Colitis/inmunología , Citocinas/antagonistas & inhibidores , Sulfato de Dextran/toxicidad , Mediadores de Inflamación/fisiología , Malus/fisiología , Polifenoles/fisiología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/patología , Animales , Células Cultivadas , Colitis/inducido químicamente , Colitis/patología , Citocinas/biosíntesis , Citocinas/genética , Sulfato de Dextran/administración & dosificación , Femenino , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/química , Masculino , Malus/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Polifenoles/química , Subgrupos de Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA