Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 2126, 2021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33837203

RESUMO

There is accumulating evidence that the lower airway microbiota impacts lung health. However, the link between microbial community composition and lung homeostasis remains elusive. We combine amplicon sequencing and bacterial culturing to characterize the viable bacterial community in 234 longitudinal bronchoalveolar lavage samples from 64 lung transplant recipients and establish links to viral loads, host gene expression, lung function, and transplant health. We find that the lung microbiota post-transplant can be categorized into four distinct compositional states, 'pneumotypes'. The predominant 'balanced' pneumotype is characterized by a diverse bacterial community with moderate viral loads, and host gene expression profiles suggesting immune tolerance. The other three pneumotypes are characterized by being either microbiota-depleted, or dominated by potential pathogens, and are linked to increased immune activity, lower respiratory function, and increased risks of infection and rejection. Collectively, our findings establish a link between the lung microbial ecosystem, human lung function, and clinical stability post-transplant.


Assuntos
Rejeição de Enxerto/microbiologia , Transplante de Pulmão/efeitos adversos , Pulmão/microbiologia , Microbiota/imunologia , Pneumonia Bacteriana/microbiologia , Adulto , Aloenxertos/imunologia , Aloenxertos/microbiologia , Bactérias/genética , Bactérias/imunologia , Bactérias/isolamento & purificação , Bactérias/patogenicidade , Carga Bacteriana/imunologia , Técnicas Bacteriológicas , Líquido da Lavagem Broncoalveolar/microbiologia , Broncoscopia , DNA Bacteriano/isolamento & purificação , Feminino , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/imunologia , Humanos , Tolerância Imunológica , Estudos Longitudinais , Pulmão/imunologia , Masculino , Metagenômica , Microbiota/genética , Pessoa de Meia-Idade , Pneumonia Bacteriana/diagnóstico , Pneumonia Bacteriana/imunologia , Estudos Prospectivos , RNA Ribossômico 16S/genética
2.
Micron ; 129: 102782, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31775097

RESUMO

Infection of zebrafish with natural pathogen Mycobacterium marinum is a useful surrogate for studying the human granulomatous inflammatory response to infection by Mycobacterium tuberculosis. The adaptive immune system of the adult stage zebrafish offers an advance on the commonly used embryo infection model as adult zebrafish form granulomas with striking similarities to human-M. tuberculosis granulomas. Here, we present workflows to perform high content analyses of granulomas in adult zebrafish infected with M. marinum by cryosectioning to take advantage of strong endogenous transgenic fluorescence adapted from common zebrafish embryo infection tools. Specific guides to classifying granuloma necrosis and organisation, quantifying bacterial burden and leukocyte infiltration of granulomas, visualizing foam cell formation, analysing extracellular matrix remodelling and granuloma fibrosis are also provided. We use these methods to characterize neutrophil recruitment to M. marinum granulomas across time and find an inverse relation to granuloma necrosis suggesting granuloma necrosis is not a marker of immunopathology in the natural infection system of the adult zebrafish-M. marinum pairing. The methods can be easily translated to studying the zebrafish adaptive immune response to other chronic and granuloma-forming pathogens.


Assuntos
Granuloma/imunologia , Infecções por Mycobacterium não Tuberculosas/imunologia , Infecções por Mycobacterium não Tuberculosas/veterinária , Mycobacterium marinum/imunologia , Neutrófilos/imunologia , Imunidade Adaptativa/imunologia , Animais , Carga Bacteriana/imunologia , Modelos Animais de Doenças , Células Espumosas/imunologia , Granuloma/patologia , Inflamação/patologia , Infecções por Mycobacterium não Tuberculosas/patologia , Necrose/imunologia , Necrose/patologia , Infiltração de Neutrófilos/imunologia , Peixe-Zebra
3.
Sci Rep ; 9(1): 6071, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988402

RESUMO

We investigated the anti-inflammatory and antibacterial activities of Hc-cath, a cathelicidin peptide derived from the venom of the sea snake, Hydrophis cyanocyntus, using in vivo models of inflammation and infection. Hc-cath function was evaluated in in vitro, in vivo in the wax moth, Galleria mellonella, and in mouse models of intraperitoneal and respiratory Pseudomonas aeruginosa infection. Hc-Cath downregulated LPS-induced pro-inflammatory responses in macrophages and significantly improved the survival of P. aeruginosa infected G. mellonella over a 5-day period. We also demonstrated, for the first time, that Hc-cath can modulate inflammation in a mouse model of LPS-induced lung inflammation by significantly reducing the release of the pro-inflammatory cytokine and neutrophil chemoattractant, KC, resulting in reduced cellular infiltration into the lungs. Moreover, Hc-cath treatment significantly reduced the bacterial load and inflammation in mouse models of P. aeruginosa intraperitoneal and respiratory infection. The effect of Hc-cath in our studies highlights the potential to develop this peptide as a candidate for therapeutic development.


Assuntos
Anti-Infecciosos/administração & dosagem , Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Produtos Biológicos/administração & dosagem , Hydrophiidae , Pneumonia/tratamento farmacológico , Infecções por Pseudomonas/tratamento farmacológico , Animais , Anti-Infecciosos/síntese química , Peptídeos Catiônicos Antimicrobianos/síntese química , Carga Bacteriana/efeitos dos fármacos , Carga Bacteriana/imunologia , Produtos Biológicos/síntese química , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/metabolismo , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Lipopolissacarídeos/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Mariposas/imunologia , Mariposas/microbiologia , Pneumonia/imunologia , Pneumonia/microbiologia , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Pseudomonas aeruginosa/isolamento & purificação , Células THP-1 , Catelicidinas
4.
J Surg Res ; 233: 323-330, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30502266

RESUMO

BACKGROUND: Previously we have shown that volatile anesthetic isoflurane attenuated neutrophil recruitment and phagocytosis in mouse sepsis and skin inflammation models. The objectives of this study were to test ex vivo function of neutrophils in patients who underwent cardiac catheterization under volatile anesthesia versus intravenous anesthesia (IA), and also to assess the effect of anesthesia on surgical site infections (SSIs) using mouse model to understand the clinical relevance of anesthesia-induced immunomodulation. METHODS: Whole blood from patients who underwent cardiac catheterization procedures either by volatile anesthesia or IA was collected and subjected to phagocytosis assay and a lipopolysaccharide-induced tumor necrosis factor-α assay. Mouse SSI with Staphylococcus aureus USA300 was created, and the effect of isoflurane and propofol exposure (short or long exposure) on bacterial loads was tested. RESULTS: Neutrophil phagocytosis was significantly attenuated after the induction of volatile anesthesia in patients, but not by IA. Monocyte phagocytosis was not affected by the anesthesia regimen. Bacterial loads following SSIs were significantly higher in mice receiving long, but not short, isoflurane exposure. Propofol exposure did not affect bacterial loads. DISCUSSION: Neutrophil phagocytosis can be affected by the type of anesthesia, and preclinical model of SSIs showed potential clinical relevance. The effects of anesthesia regimen on SSIs in patients needs to be studied extensively in the future.


Assuntos
Anestésicos Inalatórios/efeitos adversos , Carga Bacteriana/imunologia , Neutrófilos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Infecção da Ferida Cirúrgica/imunologia , Adolescente , Adulto , Anestésicos Inalatórios/administração & dosagem , Anestésicos Intravenosos/administração & dosagem , Anestésicos Intravenosos/efeitos adversos , Animais , Carga Bacteriana/efeitos dos fármacos , Cateterismo Cardíaco/efeitos adversos , Criança , Pré-Escolar , Modelos Animais de Doenças , Escherichia coli/imunologia , Feminino , Humanos , Isoflurano/administração & dosagem , Isoflurano/efeitos adversos , Masculino , Camundongos , Neutrófilos/imunologia , Propofol/administração & dosagem , Propofol/efeitos adversos , Staphylococcus aureus/imunologia , Staphylococcus aureus/isolamento & purificação , Infecção da Ferida Cirúrgica/microbiologia , Infecção da Ferida Cirúrgica/prevenção & controle , Adulto Jovem
5.
Emerg Microbes Infect ; 7(1): 207, 2018 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-30538219

RESUMO

Tuberculosis (TB) has become the most deadly infectious diseases due to epidemics of HIV/AIDS and multidrug-resistant/extensively drug-resistant TB (MDR-/XDR-TB). Although person-to-person transmission contributes to MDR-TB, it remains unknown whether infection with MDR strains resembles infection with drug-sensitive (DS) TB strains, manipulating limited or broad immune responses. To address these questions, macaques were infected with MDR strain V791 and a drug-sensitive Erdman strain of TB. MDR bacilli burdens in the airway were significantly higher than those of the Erdman control after pulmonary exposure. This productive MDR strain infection upregulated the expression of caspase 3 in macrophages/monocytes and induced appreciable innate-like effector responses of CD3-negative lymphocytes and Ag-specific γδ T-cell subsets. Concurrently, MDR strain infection induced broad immune responses of T-cell subpopulations producing Th1, Th17, Th22, and CTL cytokines. Furthermore, MDR bacilli, like the Erdman strain, were capable of inducing typical TB disease characterized by weight loss, lymphocytopenia, and severe TB lesions. For the first time, our results suggest that MDR-TB infection acts like DS to induce high bacterial burdens in the airway (transmission advantage), innate/adaptive immune responses, and disease processes. Because nonhuman primates are biologically closer to humans than other species, our data may provide useful information for predicting the effects of primary MDR strain infection after person-to-person transmission. The findings also support the hypothesis that a vaccine or host-directed adjunctive modality that is effective for drug-sensitive TB is likely to also impact MDR-TB.


Assuntos
Imunidade Adaptativa , Carga Bacteriana/imunologia , Imunidade Inata , Pulmão/imunologia , Tuberculose Resistente a Múltiplos Medicamentos/imunologia , Animais , Caspase 3 , Citocinas/imunologia , Farmacorresistência Bacteriana Múltipla , Pulmão/microbiologia , Macaca , Macrófagos/imunologia , Macrófagos/microbiologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia , Tuberculose Pulmonar/imunologia
6.
PLoS Pathog ; 14(8): e1007244, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30102746

RESUMO

The early events that shape the innate immune response to restrain pathogens during skin infections remain elusive. Methicillin-resistant Staphylococcus aureus (MRSA) infection engages phagocyte chemotaxis, abscess formation, and microbial clearance. Upon infection, neutrophils and monocytes find a gradient of chemoattractants that influence both phagocyte direction and microbial clearance. The bioactive lipid leukotriene B4 (LTB4) is quickly (seconds to minutes) produced by 5-lipoxygenase (5-LO) and signals through the G protein-coupled receptors LTB4R1 (BLT1) or BLT2 in phagocytes and structural cells. Although it is known that LTB4 enhances antimicrobial effector functions in vitro, whether prompt LTB4 production is required for bacterial clearance and development of an inflammatory milieu necessary for abscess formation to restrain pathogen dissemination is unknown. We found that LTB4 is produced in areas near the abscess and BLT1 deficient mice are unable to form an abscess, elicit neutrophil chemotaxis, generation of neutrophil and monocyte chemokines, as well as reactive oxygen species-dependent bacterial clearance. We also found that an ointment containing LTB4 synergizes with antibiotics to eliminate MRSA potently. Here, we uncovered a heretofore unknown role of macrophage-derived LTB4 in orchestrating the chemoattractant gradient required for abscess formation, while amplifying antimicrobial effector functions.


Assuntos
Abscesso/imunologia , Carga Bacteriana/imunologia , Leucotrieno B4/fisiologia , Macrófagos/metabolismo , Staphylococcus aureus Resistente à Meticilina , Infecções Cutâneas Estafilocócicas/imunologia , Abscesso/genética , Abscesso/microbiologia , Abscesso/patologia , Animais , Araquidonato 5-Lipoxigenase/genética , Carga Bacteriana/genética , Células Cultivadas , Feminino , Leucotrieno B4/metabolismo , Macrófagos/imunologia , Masculino , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Staphylococcus aureus Resistente à Meticilina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Leucotrieno B4/genética , Infecções Cutâneas Estafilocócicas/genética , Infecções Cutâneas Estafilocócicas/patologia
7.
JCI Insight ; 3(3)2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29415887

RESUMO

Neutrophils dominate the early immune response in pathogen-induced acute lung injury, but efforts to harness their responses have not led to therapeutic advancements. Neutrophil extracellular traps (NETs) have been proposed as an innate defense mechanism responsible for pathogen clearance, but there are concerns that NETs may induce collateral damage to host tissues. Here, we detected NETs in abundance in mouse models of severe bacterial pneumonia/acute lung injury and in human subjects with acute respiratory distress syndrome (ARDS) from pneumonia or sepsis. Decreasing NETs reduced lung injury and improved survival after DNase I treatment or with partial protein arginine deiminase 4 deficiency (PAD4+/-). Complete PAD4 deficiency (PAD4-/-) reduced NETs and lung injury but was counterbalanced by increased bacterial load and inflammation. Importantly, we discovered that the lipoxin pathway could be a potent modulator of NET formation, and that mice deficient in the lipoxin receptor (Fpr2-/-) produced excess NETs leading to increased lung injury and mortality. Lastly, we observed in humans that increased plasma NETs were associated with ARDS severity and mortality, and lower plasma DNase I levels were associated with the development of sepsis-induced ARDS. We conclude that a critical balance of NETs is necessary to prevent lung injury and to maintain microbial control, which has important therapeutic implications.


Assuntos
Armadilhas Extracelulares/imunologia , Lesão Pulmonar/imunologia , Pneumonia Bacteriana/imunologia , Síndrome do Desconforto Respiratório/imunologia , Sepse/imunologia , Animais , Bactérias/imunologia , Carga Bacteriana/efeitos dos fármacos , Carga Bacteriana/imunologia , Desoxirribonuclease I/administração & dosagem , Modelos Animais de Doenças , Armadilhas Extracelulares/efeitos dos fármacos , Armadilhas Extracelulares/metabolismo , Humanos , Hidrolases/genética , Hidrolases/metabolismo , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/microbiologia , Lesão Pulmonar/mortalidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Pneumonia Bacteriana/microbiologia , Proteína-Arginina Desiminase do Tipo 4 , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/imunologia , Receptores de Formil Peptídeo/metabolismo , Síndrome do Desconforto Respiratório/microbiologia , Sepse/microbiologia
8.
Nat Commun ; 8(1): 1576, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29146903

RESUMO

Dysregulation of autophagy and inflammasome activity contributes to the development of auto-inflammatory diseases. Emerging evidence highlights the importance of the actin cytoskeleton in modulating inflammatory responses. Here we show that deficiency of Wiskott-Aldrich syndrome protein (WASp), which signals to the actin cytoskeleton, modulates autophagy and inflammasome function. In a model of sterile inflammation utilizing TLR4 ligation followed by ATP or nigericin treatment, inflammasome activation is enhanced in monocytes from WAS patients and in WAS-knockout mouse dendritic cells. In ex vivo models of enteropathogenic Escherichia coli and Shigella flexneri infection, WASp deficiency causes defective bacterial clearance, excessive inflammasome activation and host cell death that are associated with dysregulated septin cage-like formation, impaired autophagic p62/LC3 recruitment and defective formation of canonical autophagosomes. Taken together, we propose that dysregulation of autophagy and inflammasome activities contribute to the autoinflammatory manifestations of WAS, thereby identifying potential targets for therapeutic intervention.


Assuntos
Citoesqueleto de Actina/metabolismo , Autofagia/imunologia , Inflamassomos/imunologia , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/imunologia , Animais , Autofagia/genética , Carga Bacteriana/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Escherichia coli Enteropatogênica/imunologia , Humanos , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Nigericina/farmacologia , Septinas/metabolismo , Shigella flexneri/imunologia , Células THP-1 , Receptor 4 Toll-Like/imunologia , Síndrome de Wiskott-Aldrich/metabolismo
9.
Sci Rep ; 7: 41240, 2017 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-28128217

RESUMO

The protective effects of mycobacterial infections on lung allergy are well documented. However, the inverse relationship between tuberculosis and type 2 immunity is still elusive. Although type 1 immunity is essential to protection against Mycobacterium tuberculosis it might be also detrimental to the host due to the induction of extensive tissue damage. Here, we determined whether lung type 2 immunity induced by allergen sensitization and challenge could affect the outcome of M. tuberculosis infection. We used two different protocols in which sensitization and allergen challenge were performed before or after M. tuberculosis infection. We found an increased resistance to M. tuberculosis only when allergen exposure was given after, but not before infection. Infected mice exposed to allergen exhibited lower bacterial load and cellular infiltrates in the lungs. Enhanced resistance to infection after allergen challenge was associated with increased gene expression of alternatively activated macrophages (M2 macrophages) and IL-33 levels. Accordingly, either adoptive transfer of M2 macrophages or systemic IL-33 treatment was effective in attenuating M. tuberculosis infection. Notably, the enhanced resistance induced by allergen exposure was dependent on IL-33 receptor ST2. Our work indicates that IL-33 might be an alternative therapeutic treatment for severe tuberculosis.


Assuntos
Interleucina-33/imunologia , Pulmão/imunologia , Tuberculose Pulmonar/imunologia , Tuberculose/imunologia , Alérgenos/imunologia , Alérgenos/toxicidade , Animais , Carga Bacteriana/imunologia , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1/imunologia , Interleucina-33/genética , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/patogenicidade , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Tuberculose/genética , Tuberculose/microbiologia , Tuberculose/patologia , Tuberculose Pulmonar/genética , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia
10.
PLoS One ; 11(6): e0158020, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27322540

RESUMO

BACKGROUND: Human Campylobacter jejuni infections are progressively rising worldwide. Information about the molecular mechanisms underlying campylobacteriosis, however, are limited. In the present study we investigated whether cytokines such as IL-23, IL-22 and IL-18, which share pivotal functions in host immunity, were involved in mediating intestinal and systemic immunopathological responses upon C. jejuni infection. METHODOLOGY/PRINCIPAL FINDINGS: To assure stable infection, gnotobiotic (i.e. secondary abiotic) IL-23p19-/-, IL-22-/- and IL-18-/- mice were generated by broad-spectrum antibiotic treatment. Following peroral C. jejuni strain 81-176 infection, mice of all genotypes harbored comparably high pathogenic loads in their intestines. As compared to wildtype controls, however, IL-18-/- mice displayed less distinct C. jejuni induced sequelae as indicated by less pronounced large intestinal shrinkage and lower numbers of apoptotic cells in the colonic epithelial layer at day 8 postinfection (p.i.). Furthermore, lower colonic numbers of adaptive immune cells including regulatory T cells and B lymphocytes were accompanied by less distinct secretion of pro-inflammatory cytokines such as TNF and IFN-γ and lower IL-17A mRNA expression levels in colonic ex vivo biopsies of infected IL-18-/- as compared to wildtype mice. Upon C. jejuni infection, colonic IL-23p19 expression was up-regulated in IL-18-/- mice only, whereas IL-22 mRNA levels were lower in uninfected and infected IL-23p19-/- as well as infected IL-18-/- as compared to respective wildtype control mice. Remarkably, not only intestinal, but also systemic infection-induced immune responses were less pronounced in IL-18-/- mice as indicated by lower TNF, IFN-γ and IL-6 serum levels as compared to wildtype mice. CONCLUSION/SIGNIFICANCE: We here show for the first time that IL-18 is essentially involved in mediating C. jejuni infection in the gnotobiotic mouse model. Future studies need to further unravel the underlying regulatory mechanisms orchestrating pathogen-host interaction.


Assuntos
Infecções por Campylobacter/imunologia , Campylobacter jejuni/imunologia , Vida Livre de Germes/imunologia , Interleucina-18/metabolismo , Animais , Apoptose , Carga Bacteriana/imunologia , Translocação Bacteriana , Biópsia , Infecções por Campylobacter/genética , Infecções por Campylobacter/patologia , Campylobacter jejuni/crescimento & desenvolvimento , Proliferação de Células , Contagem de Colônia Microbiana , Feminino , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Subunidade p19 da Interleucina-23/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/patologia , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Interleucina 22
11.
Pathog Dis ; 74(3)2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26903072

RESUMO

Necrotizing granulomas, exacerbating pathogenesis and neutrophil influx at the site of infection are hallmarks of active pulmonary tuberculosis (TB) in humans. The role of polymorphonuclear neutrophils (PMN) in host defence and TB pathogenesis has recently attracted broader interest. Association of infiltrating PMN, enhanced mycobacterial load and disease exacerbation in both, mice susceptible to experimental TB as well as in TB patients, link PMN to exacerbated pathology. Targeting PMN resulted in smaller lung lesions and reduced mycobacterial burden. Therefore, PMN-associated molecules represent interesting biomarkers to determine TB severity and treatment success. More importantly, PMN are putative targets for host-directed therapies (HDT) in TB. Due to the rise of multi- and extensively drug-resistant Mycobacterium tuberculosis isolates, HDT represent adjunct measures to support antibiotic treatment by ameliorating pathology and local host defences.


Assuntos
Mycobacterium tuberculosis/imunologia , Neutrófilos/imunologia , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/terapia , Animais , Carga Bacteriana/imunologia , Citocinas/imunologia , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Camundongos , Tuberculose Pulmonar/microbiologia
12.
Shock ; 45(1): 40-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26674453

RESUMO

Sepsis following hemorrhagic shock is a common clinical condition, in which innate immune system suffers from severe suppression. B and T lymphocyte attenuator (BTLA) is an immune-regulatory coinhibitory receptor expressed not only on adaptive, but also on innate immune cells. Our previous data showed that BTLA gene deficient mice were protected from septic mortality when compared with wild-type control C57BL/6 mice. Here, we extended our study by treating C57BL/6 mice with an anti-BTLA monoclonal antibody (clone 6A6; reported to have the ability to neutralize or agonize/potentiate BTLA signaling) in a mouse model of hemorrhagic shock (Hem) followed by sepsis induced by cecal ligation and puncture (CLP); positing initially that if BTLA engagement was neutralized, like gene deficiency, an anti-BTLA mAb would have the similar effects on the inflammatory response/morbidity in these mice after such insults. Here, we report that BTLA expression is elevated on innate immune cells after Hem/CLP. However, anti-BTLA antibody treatment increased cytokine (TNF-α, IL-12, IL-10)/chemokine (KC, MIP-2, MCP-1) levels and inflammatory cells (neutrophils, macrophages, dendritic cells) recruitment in the peritoneal cavity, which in turn aggravated organ injury and elevated these animals' mortality in Hem/CLP. When compared with the protective effects of our previous study using BTLA gene deficient mice in a model of lethal septic challenge, we further confirmed BTLA's contribution to enhanced innate cell recruitment, elevated IL-10 levels, and reduced survival, and that engagement of antibody with BTLA potentiates/exacerbates the pathophysiology in Hem/sepsis.


Assuntos
Inflamação/imunologia , Receptores Imunológicos/imunologia , Sepse/imunologia , Choque Hemorrágico/imunologia , Animais , Anticorpos Monoclonais/imunologia , Carga Bacteriana/imunologia , Quimiocinas/biossíntese , Citocinas/biossíntese , Modelos Animais de Doenças , Imunidade Inata , Inflamação/etiologia , Inflamação/microbiologia , Leucócitos/imunologia , Lipopolissacarídeos/imunologia , Macrófagos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Cavidade Peritoneal/citologia , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/biossíntese , Sepse/etiologia , Sepse/microbiologia , Choque Hemorrágico/complicações , Choque Hemorrágico/microbiologia
13.
Immunology ; 146(1): 100-12, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26032199

RESUMO

Streptococcus pneumoniae is a major aetiological agent of pneumonia worldwide, as well as otitis media, sinusitis, meningitis and sepsis. Recent reports have suggested that inflammation of lungs due to S. pneumoniae infection promotes bacterial dissemination and severe disease. However, the contribution of anti-inflammatory molecules to the pathogenesis of S. pneumoniae remains unknown. To elucidate whether the production of the anti-inflammatory cytokine interleukin-10 (IL-10) is beneficial or detrimental for the host during pneumococcal pneumonia, we performed S. pneumoniae infections in mice lacking IL-10 (IL-10(-/-) mice). The IL-10(-/-) mice showed increased mortality, higher expression of pro-inflammatory cytokines, and an exacerbated recruitment of neutrophils into the lungs after S. pneumoniae infection. However, IL-10(-/-) mice showed significantly lower bacterial loads in lungs, spleen, brain and blood, when compared with mice that produced this cytokine. Our results support the notion that production of IL-10 during S. pneumoniae infection modulates the expression of pro-inflammatory cytokines and the infiltration of neutrophils into the lungs. This feature of IL-10 is important to avoid excessive inflammation of tissues and to improve host survival, even though bacterial dissemination is less efficient in the absence of this cytokine.


Assuntos
Interleucina-10/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Pneumonia Pneumocócica/imunologia , Streptococcus pneumoniae/imunologia , Animais , Carga Bacteriana/genética , Carga Bacteriana/imunologia , Encéfalo/microbiologia , Inflamação/imunologia , Interferon gama/biossíntese , Interleucina-10/biossíntese , Interleucina-10/imunologia , Interleucina-1beta/biossíntese , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Pneumocócica/mortalidade , Baço/microbiologia , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
14.
Clin Exp Immunol ; 181(3): 457-67, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25907631

RESUMO

Combined inhibition of complement and CD14 is known to attenuate bacterial-induced inflammation, but the dependency of the bacterial load on this effect is unknown. Thus, we investigated whether the effect of such combined inhibition on Escherichia coli- and Staphylococcus aureus-induced inflammation was preserved during increasing bacterial concentrations. Human whole blood was preincubated with anti-CD14, eculizumab (C5-inhibitor) or compstatin (C3-inhibitor), or combinations thereof. Then heat-inactivated bacteria were added at final concentrations of 5 × 10(4) -1 × 10(8) /ml (E. coli) or 5 × 10(7) -4 × 10(8) /ml (S. aureus). Inflammatory markers were measured using enzyme-linked immunosorbent assay (ELISA), multiplex technology and flow cytometry. Combined inhibition of complement and CD14 significantly (P < 0.05) reduced E. coli-induced interleukin (IL)-6 by 40-92% at all bacterial concentrations. IL-1ß, IL-8 and macrophage inflammatory protein (MIP)-1α were significantly (P < 0.05) inhibited by 53-100%, and the effect was lost only at the highest bacterial concentration. Tumour necrosis factor (TNF) and MIP-1ß were significantly (P < 0.05) reduced by 80-97% at the lowest bacterial concentration. Monocyte and granulocyte CD11b were significantly (P < 0.05) reduced by 63-91% at all bacterial doses. Lactoferrin was significantly (P < 0.05) attenuated to the level of background activity at the lowest bacterial concentration. Similar effects were observed for S. aureus, but the attenuation was, in general, less pronounced. Compared to E. coli, much higher concentrations of S. aureus were required to induce the same cytokine responses. This study demonstrates generally preserved effects of combined complement and CD14 inhibition on Gram-negative and Gram-positive bacterial-induced inflammation during escalating bacterial load. The implications of these findings for future therapy of sepsis are discussed.


Assuntos
Complemento C3/imunologia , Complemento C5/imunologia , Escherichia coli/imunologia , Inflamação/imunologia , Receptores de Lipopolissacarídeos/imunologia , Staphylococcus aureus/imunologia , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Carga Bacteriana/imunologia , Antígeno CD11b/sangue , Antígeno CD11b/imunologia , Complemento C3/antagonistas & inibidores , Complemento C5/antagonistas & inibidores , Citocinas/sangue , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Granulócitos/imunologia , Granulócitos/metabolismo , Temperatura Alta , Humanos , Inflamação/sangue , Inflamação/prevenção & controle , Receptores de Lipopolissacarídeos/sangue , Monócitos/imunologia , Monócitos/metabolismo , Peptídeos Cíclicos/imunologia , Peptídeos Cíclicos/farmacologia
15.
PLoS One ; 10(3): e0122560, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822536

RESUMO

Protein subunit vaccines present a compelling new area of research for control of tuberculosis (TB). Based on the interaction between Mycobacterium tuberculosis and its host, five stage-specific antigens of M. tuberculosis that participate in TB pathogenesis--Rv1813, Rv2660c, Ag85B, Rv2623, and HspX--were selected. These antigens were verified to be recognized by T cells from a total of 42 whole blood samples obtained from active TB patients, patients with latent TB infections (LTBIs), and healthy control donors. The multistage polyprotein A1D4 was developed using the selected five antigens as a potentially more effective novel subunit vaccine. The immunogenicity and protective efficacy of A1D4 emulsified in the adjuvant MTO [monophosphoryl lipid A (MPL), trehalose-6,6'-dibehenate (TDB), components of MF59] was compared with Bacillus Calmette-Guerin (BCG) in C57BL/6 mice. Our results demonstrated that A1D4/MTO could provide more significant protection against M. tuberculosis infection than the PBS control or MTO adjuvant alone judging from the A1D4-specific Th1-type immune response; however, its efficacy was inferior to BCG as demonstrated by the bacterial load in the lung and spleen, and by the pathological changes in the lung. Antigen-specific single IL-2-secreting cells and different combinations with IL-2-secreting CD4+ T cells were beneficial and correlated with BCG vaccine-induced protection against TB. Antigen-specific IFN-γ+ IL-2+ CD4+ T cells were the only effective biomarker significantly induced by A1D4/MTO. Among all groups, A1D4/MTO immunization also conferred the highest number of antigen-specific single IFN-γ+ and IFN-γ+ TNF-α+ CD4+ T cells, which might be related to the antigen load in vivo, and single IFN-γ+ CD8+ T cells by mimicking the immune patterns of LTBIs or curable TB patients. Our strategy seems promising for the development of a TB vaccine based on multistage antigens, and subunit antigen A1D4 suspended in MTO adjuvant warrants preclinical evaluation in animal models of latent infection and may boost BCG vaccination.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/metabolismo , Interleucina-2/metabolismo , Mycobacterium tuberculosis/fisiologia , Vacinas contra a Tuberculose/imunologia , Tuberculose/prevenção & controle , Adjuvantes Imunológicos/farmacologia , Adolescente , Adulto , Idoso , Animais , Anticorpos Antibacterianos/imunologia , Antígenos de Bactérias/sangue , Antígenos de Bactérias/imunologia , Carga Bacteriana/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Feminino , Humanos , Imunização , Pulmão/microbiologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Mycobacterium tuberculosis/imunologia , Poliproteínas/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Adulto Jovem
16.
Infect Immun ; 83(4): 1641-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25667264

RESUMO

Toll-like receptors (TLRs) are evolutionarily conserved host proteins that are essential for effective host defense against pathogens. However, recent studies suggest that some TLRs can negatively regulate immune responses. We observed here that TLR2 and TLR9 played opposite roles in regulating innate immunity against oral infection of Salmonella enterica serovar Typhimurium in mice. While TLR9-/- mice exhibited shortened survival, an increased cytokine storm, and more severe Salmonella hepatitis than wild-type (WT) mice, TLR2-/- mice exhibited the opposite phenomenon. Further studies demonstrated that TLR2 deficiency and TLR9 deficiency in macrophages both disrupted NK cell cytotoxicity against S. Typhimurium-infected macrophages by downregulating NK cell degranulation and gamma interferon (IFN-γ) production through decreased macrophage expression of the RAE-1 NKG2D ligand. But more importantly, we found that S. Typhimurium-infected TLR2-/- macrophages upregulated inducible nitric oxide synthase (iNOS) expression, resulting in a lower bacterial load than that in WT macrophages in vitro and livers in vivo as well as low proinflammatory cytokine levels. In contrast, TLR9-/- macrophages showed decreased reactive oxygen species (ROS) expression concomitant with a high bacterial load in the macrophages and in livers of TLR9-/- mice. TLR9-/- macrophages were also more susceptible than WT macrophages to S. Typhimurium-induced necroptosis in vitro, likely contributing to bacterial spread and transmission in vivo. Collectively, these findings indicate that TLR2 negatively regulates anti-S. Typhimurium immunity, whereas TLR9 is vital to host defense and survival against S. Typhimurium invasion. TLR2 antagonists or TLR9 agonists may thus serve as potential anti-S. Typhimurium therapeutic agents.


Assuntos
Macrófagos/imunologia , Infecções por Salmonella/imunologia , Salmonella typhimurium/imunologia , Receptor 2 Toll-Like/imunologia , Receptor Toll-Like 9/imunologia , Animais , Carga Bacteriana/imunologia , Degranulação Celular/imunologia , Hepatite/imunologia , Hepatite/microbiologia , Imunidade Inata , Inflamação/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/biossíntese , Óxido Nítrico Sintase Tipo II/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Receptor Toll-Like 9/genética
17.
Infect Immun ; 83(2): 560-71, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25404030

RESUMO

IL12RB1 is a human gene that is important for resistance to Mycobacterium tuberculosis infection. IL12RB1 is expressed by multiple leukocyte lineages, and encodes a type I transmembrane protein (IL12Rß1) that associates with IL12p40 and promotes the development of host-protective T(H)1 cells. Recently, we observed that il12rb1­the mouse homolog of IL12RB1­is alternatively spliced by leukocytes to produce a second isoform (IL12Rß1ΔTM) that has biological properties distinct from IL12Rß1. Although the expression of IL12Rß1ΔTM is elicited by M. tuberculosis in vivo, and its overexpression enhances IL12p40 responsiveness in vitro, the contribution of IL12Rß1ΔTM to controlling M. tuberculosis infection has not been tested. Here, we demonstrate that IL12Rß1ΔTM represents a secreted product of il12rb1 that, when absent from mice, compromises their ability to control M. tuberculosis infection in extrapulmonary organs. Furthermore, elevated M. tuberculosis burdens in IL12Rß1ΔTM-deficient animals are associated with decreased lymph node cellularity and a decline in TH1 development. Collectively, these data support a model wherein IL12Rß1ΔTM is a secreted product of il12rb1 that promotes resistance to M. tuberculosis infection by potentiating T(H) cells response to IL-12.


Assuntos
Interleucina-12/imunologia , Mycobacterium tuberculosis/imunologia , Receptores de Interleucina-12/genética , Células Th1/imunologia , Tuberculose/imunologia , Células 3T3 , Processamento Alternativo , Animais , Carga Bacteriana/imunologia , Linhagem Celular , Interferon gama/biossíntese , Interleucina-12/biossíntese , Pulmão/imunologia , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Receptores de Interleucina-12/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
18.
PLoS One ; 9(11): e112848, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25398087

RESUMO

Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb) that is a major public health problem. The vaccine used for TB prevention is Mycobacterium bovis bacillus Calmette-Guérin (BCG), which provides variable efficacy in protecting against pulmonary TB among adults. Consequently, several groups have pursued the development of a new vaccine with a superior protective capacity to that of BCG. Here we constructed a new recombinant BCG (rBCG) vaccine expressing a fusion protein (CMX) composed of immune dominant epitopes from Ag85C, MPT51, and HspX and evaluated its immunogenicity and protection in a murine model of infection. The stability of the vaccine in vivo was maintained for up to 20 days post-vaccination. rBCG-CMX was efficiently phagocytized by peritoneal macrophages and induced nitric oxide (NO) production. Following mouse immunization, this vaccine induced a specific immune response in cells from lungs and spleen to the fusion protein and to each of the component recombinant proteins by themselves. Vaccinated mice presented higher amounts of Th1, Th17, and polyfunctional specific T cells. rBCG-CMX vaccination reduced the extension of lung lesions caused by challenge with Mtb as well as the lung bacterial load. In addition, when this vaccine was used in a prime-boost strategy together with rCMX, the lung bacterial load was lower than the result observed by BCG vaccination. This study describes the creation of a new promising vaccine for TB that we hope will be used in further studies to address its safety before proceeding to clinical trials.


Assuntos
Vacina BCG/imunologia , Células Th1/imunologia , Células Th17/imunologia , Tuberculose/prevenção & controle , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Carga Bacteriana/imunologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Epitopos/imunologia , Feminino , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/imunologia , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Óxido Nítrico/metabolismo , Peritônio/citologia , Fagocitose , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Baço/imunologia , Baço/metabolismo , Células Th1/citologia , Células Th17/citologia , Tuberculose/imunologia
19.
Mol Cell Biol ; 34(23): 4301-14, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25246634

RESUMO

Macrophages regulate cell fate decisions during microbial challenges by carefully titrating signaling events activated by innate receptors such as dectin-1 or Toll-like receptors (TLRs). Here, we demonstrate that dectin-1 activation robustly dampens TLR-induced proinflammatory signature in macrophages. Dectin-1 induced the stabilization of ß-catenin via spleen tyrosine kinase (Syk)-reactive oxygen species (ROS) signals, contributing to the expression of WNT5A. Subsequently, WNT5A-responsive protein inhibitors of activated STAT (PIAS-1) and suppressor of cytokine signaling 1 (SOCS-1) mediate the downregulation of IRAK-1, IRAK-4, and MyD88, resulting in decreased expression of interleukin 12 (IL-12), IL-1ß, and tumor necrosis factor alpha (TNF-α). In vivo activation of dectin-1 with pathogenic fungi or ligand resulted in an increased bacterial burden of Mycobacteria, Klebsiella, Staphylococcus, or Escherichia, with a concomitant decrease in TLR-triggered proinflammatory cytokines. All together, our study establishes a new role for dectin-1-responsive inhibitory mechanisms employed by virulent fungi to limit the proinflammatory environment of the host.


Assuntos
Inflamação/imunologia , Lectinas Tipo C/imunologia , Macrófagos/imunologia , Receptores Toll-Like/imunologia , Via de Sinalização Wnt/imunologia , Animais , Aspergillus flavus/patogenicidade , Aspergillus fumigatus/patogenicidade , Carga Bacteriana/imunologia , Candida albicans/patogenicidade , Regulação para Baixo , Ativação Enzimática , Escherichia/patogenicidade , Quinases Associadas a Receptores de Interleucina-1/biossíntese , Interleucina-12/biossíntese , Interleucina-1beta/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Klebsiella/patogenicidade , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium/patogenicidade , Fator 88 de Diferenciação Mieloide/biossíntese , Proteínas Inibidoras de STAT Ativados/biossíntese , Proteínas Tirosina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus/patogenicidade , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Quinase Syk , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Wnt/biossíntese , Proteína Wnt-5a , beta Catenina/imunologia
20.
Cell Microbiol ; 16(10): 1503-17, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24779433

RESUMO

Uropathogenic Escherichia coli (UPEC) colonizing kidneys is the main cause of acute pyelonephritis. TLR5 that senses flagellin was shown to be highly expressed in the bladder and to participate in host defence against flagellated UPEC, although its role in kidneys still remains elusive. Here we show that TLR5 is expressed in renal medullary collecting duct (MCD) cells, which represent a preferential site of UPEC adhesion. Flagellin, like lipopolysaccharide, stimulated the production of the chemoattractant chemokines CXCL1 and CXCL2, and subsequent migration capacity of neutrophils in cultured wild-type (WT) and Tlr4(-/-) MCDs, but not in Tlr5(-/-) MCDs. UPEC can translocate across intact MCD layers without altering tight junctions. Strikingly, the invasion capacity and transcellular translocation of the UPEC strain HT7 were significantly lower in Tlr5(-/-) than in WT MCDs. The non-motile HT7ΔfliC mutant lacking flagellin also exhibited much lower translocation capacities than the HT7 isolates. Finally, Tlr5(-/-) kidneys exhibited less infiltrating neutrophils than WT kidneys one day after the transurethral inoculation of HT7, and greater delayed renal bacterial loads in the day 4 post-infected Tlr5(-/-) kidneys. Overall, these findings indicate that the epithelial TLR5 participates to renal antibacterial defence, but paradoxically favours the translocation of UPEC across intact MCD cell layers.


Assuntos
Infecções por Escherichia coli/imunologia , Flagelina/imunologia , Túbulos Renais Coletores/imunologia , Receptor 5 Toll-Like/imunologia , Escherichia coli Uropatogênica/patogenicidade , Animais , Aderência Bacteriana/fisiologia , Carga Bacteriana/imunologia , Quimiocina CXCL1/biossíntese , Quimiocina CXCL2/biossíntese , Infecções por Escherichia coli/microbiologia , Feminino , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Pielonefrite/imunologia , Pielonefrite/microbiologia , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Receptor 5 Toll-Like/genética , Bexiga Urinária/imunologia , Bexiga Urinária/metabolismo , Infecções Urinárias/imunologia , Infecções Urinárias/microbiologia , Escherichia coli Uropatogênica/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA