Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Immunol ; 45(6): 1716-26, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25789453

RESUMO

Type 2 helper cell (Th2) dominated chronic lung diseases such as asthma are characterized by an increased risk for bacterial lung infections. However, the underlying mechanisms are poorly defined. Arginase 1 (Arg1) has been suggested to play an important role in the pathophysiology of asthma, and is rapidly induced in lung macrophages by Th2 cytokines, thereby limiting macrophage-derived antimicrobial nitric oxide (NO) production. Here we examined the effect of Th2 cytokine induced upregulation or lung myeloid cell specific conditional knockdown of Arg1 on lung resistance against Streptococcus pneumoniae (Spn) in mice. Lung macrophages responded with a profound induction of Arg1 mRNA and protein to treatment with IL-13 both in vitro and in vivo. IL-13-induced Arg1 activity in the lungs of mice led to significantly attenuated lung-protective immunity against Spn, while conditional Arg1 knockdown had no effect on lung-protective immunity against Spn. Collectively, the data show that Th2 cytokine induced increased Arg1 activity worsens lung-protective immunity against Spn, and interventions to block Th2 cytokine induced lung Arg1 activity may thus be a novel immunomodulatory strategy to lower the risk of bacterial infections in asthmatic patients.


Assuntos
Arginase/metabolismo , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/metabolismo , Streptococcus pneumoniae/imunologia , Animais , Arginase/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Ativação Enzimática , Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/metabolismo , Pneumonia Pneumocócica/genética , Pneumonia Pneumocócica/patologia , Células Th2/imunologia , Células Th2/metabolismo
2.
J Infect Dis ; 210(1): 14-24, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24470504

RESUMO

BACKGROUND: Protein-based vaccination using pneumococcal proteins is a promising approach for efficient vaccines against Streptococcus pneumoniae. Basophils play an important role in enhancing memory immune responses to intact proteins. We examined the impact of increased basophil pool sizes on humoral memory responses to pneumococcal surface protein A (PspA). METHODS: Basophil pool sizes in blood, spleen, and bone marrow were increased by either interleukin 3 (IL-3) treatment or by adoptive basophil transfer before secondary PspA immunization. Subsequently, PspA-specific antibody titers and resistance of mice against invasive pneumococcal disease (IPD) was determined. RESULTS: Mice treated with IL-3, which increased basophil pool sizes, and mice receiving a single basophil transfusion responded with significantly higher PspA-specific antibody titers after immunization with PspA. Importantly, however, just a single transfusion of flow-sorted basophils into mice before secondary immunization with PspA significantly protected mice from lethal IPD. Moreover, concomitant blockade of inhibitory FcγRIIB on transfused basophils further substantially increased basophil-mediated protection against IPD in mice. CONCLUSIONS: This is the first study to find that a single transfusion of basophils is sufficient to boost protein-based memory responses against pneumococcal protein antigens, thereby providing significant protection against IPD in mice.


Assuntos
Proteínas de Bactérias/imunologia , Basófilos/imunologia , Infecções Pneumocócicas/imunologia , Streptococcus pneumoniae/imunologia , Animais , Anticorpos Antibacterianos/sangue , Antígenos de Bactérias/imunologia , Sangue/imunologia , Medula Óssea/imunologia , Modelos Animais de Doenças , Resistência à Doença , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia
3.
Innate Immun ; 19(5): 451-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23283733

RESUMO

Mycobacterium avium ssp. paratuberculosis (MAP) causes Johne's disease, a chronic, granulomatous enteritis of ruminants. Dendritic cells (DC) of the gut are ideally placed to combat invading mycobacteria; however, little is known about their interaction with MAP. Here, we investigated the interaction of MAP and the closely related M. avium ssp. avium (MAA) with murine DC and the effect of infected macrophages on DC maturation. The infection of DC with MAP or MAA induced DC maturation, which differed to that of LPS as maturation was accompanied by higher production of IL-10 and lower production of IL-12. Treatment of maturing DC with supernatants from mycobacteria-infected macrophages resulted in impaired DC maturation, leading to a semi-mature, tolerogenic DC phenotype expressing low levels of MHCII, CD86 and TNF-α after LPS stimulation. Though the cells were not completely differentiated they responded with an increased IL-10 and a decreased IL-12 production. Using recombinant cytokines we provide evidence that the semi-mature DC phenotype results from a combination of secreted cytokines and released antigenic mycobacterial components of the infected macrophage. Our results indicate that MAP and MAA are able to subvert DC function directly by infecting and indirectly via the milieu created by infected macrophages.


Assuntos
Células Dendríticas/imunologia , Macrófagos Peritoneais/imunologia , Mycobacterium avium/imunologia , Paratuberculose/imunologia , Tuberculose/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem Celular , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Equilíbrio Th1-Th2
4.
J Exp Med ; 209(11): 1937-52, 2012 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-23071253

RESUMO

Apoptotic death of alveolar macrophages observed during lung infection with Streptococcus pneumoniae is thought to limit overwhelming lung inflammation in response to bacterial challenge. However, the underlying apoptotic death mechanism has not been defined. Here, we examined the role of the TNF superfamily member TNF-related apoptosis-inducing ligand (TRAIL) in S. pneumoniae-induced macrophage apoptosis, and investigated the potential benefit of TRAIL-based therapy during pneumococcal pneumonia in mice. Compared with WT mice, Trail(-/-) mice demonstrated significantly decreased lung bacterial clearance and survival in response to S. pneumoniae, which was accompanied by significantly reduced apoptosis and caspase 3 cleavage but rather increased necrosis in alveolar macrophages. In WT mice, neutrophils were identified as a major source of intraalveolar released TRAIL, and their depletion led to a shift from apoptosis toward necrosis as the dominant mechanism of alveolar macrophage cell death in pneumococcal pneumonia. Therapeutic application of TRAIL or agonistic anti-DR5 mAb (MD5-1) dramatically improved survival of S. pneumoniae-infected WT mice. Most importantly, neutropenic mice lacking neutrophil-derived TRAIL were protected from lethal pneumonia by MD5-1 therapy. We have identified a previously unrecognized mechanism by which neutrophil-derived TRAIL induces apoptosis of DR5-expressing macrophages, thus promoting early bacterial killing in pneumococcal pneumonia. TRAIL-based therapy in neutropenic hosts may represent a novel antibacterial treatment option.


Assuntos
Anticorpos Monoclonais/farmacologia , Pneumonia Pneumocócica/tratamento farmacológico , Streptococcus pneumoniae/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Anticorpos Monoclonais/imunologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Citocinas/metabolismo , Feminino , Citometria de Fluxo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Neutrófilos/patologia , Pneumonia Pneumocócica/genética , Pneumonia Pneumocócica/microbiologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Streptococcus pneumoniae/fisiologia , Análise de Sobrevida , Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Resultado do Tratamento
5.
Infect Immun ; 80(12): 4281-90, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23006850

RESUMO

FMS-like tyrosine kinase-3 ligand (Flt3L) is a dendritic cell (DC) growth and differentiation factor with potential in antitumor therapies and antibacterial immunization strategies. However, the effect of systemic Flt3L treatment on lung-protective immunity against bacterial infection is incompletely defined. Here, we examined the impact of deficient (in Flt3L knockout [KO] mice), normal (in wild-type [WT] mice), or increased Flt3L availability (in WT mice pretreated with Flt3L for 3, 5, or 7 days) on lung DC subset profiles and lung-protective immunity against the major lung-tropic pathogen, Streptococcus pneumoniae. Although in Flt3L-deficient mice the numbers of DCs positive for CD11b (CD11b(pos) DCs) and for CD103 (CD103(pos) DCs) were diminished, lung permeability, a marker of injury, was unaltered in response to S. pneumoniae. In contrast, WT mice pretreated with Flt3L particularly responded with increased numbers of CD11b(pos) DCs and with less pronounced numbers of CD103(pos) DCs and impaired bacterial clearance and with increased lung permeability following S. pneumoniae challenge. Notably, infection of Flt3L-pretreated mice with S. pneumoniae lacking the pore-forming toxin, pneumolysin (PLY), resulted in substantially less lung CD11b(pos) DCs activation and reduced lung permeability. Collectively, this study establishes that Flt3L treatment enhances the accumulation of proinflammatory activated lung CD11b(pos) DCs which contribute to acute lung injury in response to PLY released by S. pneumoniae.


Assuntos
Lesão Pulmonar Aguda/imunologia , Células Dendríticas/imunologia , Proteínas de Membrana/uso terapêutico , Streptococcus pneumoniae/patogenicidade , Estreptolisinas/metabolismo , Lesão Pulmonar Aguda/terapia , Animais , Proteínas de Bactérias/metabolismo , Antígeno CD11b/metabolismo , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Humanos , Inflamação/imunologia , Ligantes , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/patologia
6.
Infect Immun ; 79(12): 4893-901, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21911460

RESUMO

Neutrophil serine proteases cathepsin G (CG), neutrophil elastase (NE), and proteinase 3 (PR3) have recently been shown to contribute to killing of Streptococcus pneumoniae in vitro. However, their relevance in lung-protective immunity against different serotypes of S. pneumoniae in vivo has not been determined so far. Here, we examined the effect of CG and CG/NE deficiency on the lung host defense against S. pneumoniae in mice. Despite similar neutrophil recruitment, both CG knockout (KO) mice and CG/NE double-KO mice infected with focal pneumonia-inducing serotype 19 S. pneumoniae demonstrated a severely impaired bacterial clearance, which was accompanied by lack of CG and NE but not PR3 proteolytic activity in recruited neutrophils, as determined using fluorescence resonance energy transfer (FRET) substrates. Moreover, both CG and CG/NE KO mice but not wild-type mice responded with increased lung permeability to infection with S. pneumoniae, resulting in severe respiratory distress and progressive mortality. Both neutrophil depletion and ablation of hematopoietic CG/NE in bone marrow chimeras abolished intra-alveolar CG and NE immunoreactivity and led to bacterial outgrowth in the lungs of mice, thereby identifying recruited neutrophils as the primary cellular source of intra-alveolar CG and NE. This is the first study showing a contribution of neutrophil-derived neutral serine proteases CG and NE to lung-protective immunity against focal pneumonia-inducing serotype 19 S. pneumoniae in mice. These data may be important for the development of novel intervention strategies to improve lung-protective immune mechanisms in critically ill patients suffering from severe pneumococcal pneumonia.


Assuntos
Catepsina G/metabolismo , Elastase de Leucócito/metabolismo , Pulmão/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/fisiologia , Animais , Líquido da Lavagem Broncoalveolar , Catepsina G/genética , Elastase de Leucócito/genética , Pulmão/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/fisiologia , Oxigênio/sangue , Peptídeo Hidrolases/metabolismo , Permeabilidade , Streptococcus pneumoniae/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...