Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Cell Infect Microbiol ; 13: 1090550, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37033482

RESUMO

Introduction: Despite epidemiological associations between community acquired pneumonia (CAP) and myocardial infarction, mechanisms that modify cardiovascular disease during CAP are not well defined. In particular, largely due to a lack of relevant experimental models, the effect of pneumonia on atherosclerotic plaques is unclear. We describe the development of a murine model of the commonest cause of CAP, Streptococcus pneumoniae pneumonia, on a background of established atherosclerosis. We go on to use our model to investigate the effects of pneumococcal pneumonia on atherosclerosis. Methods: C57BL/6J and ApoE-/- mice were fed a high fat diet to promote atherosclerotic plaque formation. Mice were then infected with a range of S. pneumoniae serotypes (1, 4 or 14) with the aim of establishing a model to study atherosclerotic plaque evolution after pneumonia and bacteremia. Laser capture microdissection of plaque macrophages enabled transcriptomic analysis. Results: Intratracheal instillation of S. pneumoniae in mice fed a cholate containing diet resulted in low survival rates following infection, suggestive of increased susceptibility to severe infection. Optimization steps resulted in a final model of male ApoE-/- mice fed a Western diet then infected by intranasal instillation of serotype 4 (TIGR4) S. pneumoniae followed by antibiotic administration. This protocol resulted in high rates of bacteremia (88.9%) and survival (88.5%). Pneumonia resulted in increased aortic sinus plaque macrophage content 2 weeks post pneumonia but not at 8 weeks, and no difference in plaque burden or other plaque vulnerability markers were found at either time point. Microarray and qPCR analysis of plaque macrophages identified downregulation of two E3 ubiquitin ligases, Huwe1 and Itch, following pneumonia. Treatment with atorvastatin failed to alter plaque macrophage content or other plaque features. Discussion: Without antibiotics, ApoE-/- mice fed a high fat diet were highly susceptible to mortality following S. pneumoniae infection. The major infection associated change in plaque morphology was an early increase in plaque macrophages. Our results also hint at a role for the ubiquitin proteasome system in the response to pneumococcal infection in the plaque microenvironment.


Assuntos
Aterosclerose , Bacteriemia , Placa Aterosclerótica , Pneumonia Pneumocócica , Masculino , Camundongos , Animais , Streptococcus pneumoniae , Camundongos Endogâmicos C57BL , Macrófagos , Apolipoproteínas E/genética , Ubiquitinas , Camundongos Knockout , Modelos Animais de Doenças
2.
Front Immunol ; 13: 956991, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35967296

RESUMO

Dysregulated neutrophilic inflammation can be highly destructive in chronic inflammatory diseases due to prolonged neutrophil lifespan and continual release of histotoxic mediators in inflamed tissues. Therapeutic induction of neutrophil apoptosis, an immunologically silent form of cell death, may be beneficial in these diseases, provided that the apoptotic neutrophils are efficiently cleared from the tissue. Previous research in our group identified ErbB inhibitors as able to induce neutrophil apoptosis and reduce neutrophilic inflammation both in vitro and in vivo. Here, we extend that work using a clinical ErbB inhibitor, neratinib, which has the potential to be repurposed in inflammatory diseases. We show that neratinib reduces neutrophilic migration o an inflammatory site in zebrafish larvae. Neratinib upregulates efferocytosis and reduces the number of persisting neutrophil corpses in mouse models of acute, but not chronic, lung injury, suggesting that the drug may have therapeutic benefits in acute inflammatory settings. Phosphoproteomic analysis of human neutrophils shows that neratinib modifies the phosphorylation of proteins regulating apoptosis, migration, and efferocytosis. This work identifies a potential mechanism for neratinib in treating acute lung inflammation by upregulating the clearance of dead neutrophils and, through examination of the neutrophil phosphoproteome, provides important insights into the mechanisms by which this may be occurring.


Assuntos
Neutrófilos , Peixe-Zebra , Animais , Apoptose/fisiologia , Receptores ErbB/metabolismo , Humanos , Inflamação , Macrófagos/metabolismo , Camundongos , Inibidores de Proteínas Quinases , Proteoma/metabolismo , Quinolinas
3.
Artigo em Inglês | MEDLINE | ID: mdl-32984077

RESUMO

Exposure to respiratory pathogens is a leading cause of exacerbations of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD). Pellino-1 is an E3 ubiquitin ligase known to regulate virally-induced inflammation. We wished to determine the role of Pellino-1 in the host response to respiratory viruses in health and disease. Pellino-1 expression was examined in bronchial sections from patients with GOLD stage two COPD and healthy controls. Primary bronchial epithelial cells (PBECs) in which Pellino-1 expression had been knocked down were extracellularly challenged with the TLR3 agonist poly(I:C). C57BL/6 Peli1-/- mice and wild type littermates were subjected to intranasal infection with clinically-relevant respiratory viruses: rhinovirus (RV1B) and influenza A. We found that Pellino-1 is expressed in the airways of normal subjects and those with COPD, and that Pellino-1 regulates TLR3 signaling and responses to airways viruses. In particular we observed that knockout of Pellino-1 in the murine lung resulted in increased production of proinflammatory cytokines IL-6 and TNFα upon viral infection, accompanied by enhanced recruitment of immune cells to the airways, without any change in viral replication. Pellino-1 therefore regulates inflammatory airway responses without altering replication of respiratory viruses.


Assuntos
Infecções por Picornaviridae , Doença Pulmonar Obstrutiva Crônica , Viroses , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares , Rhinovirus , Ubiquitina-Proteína Ligases/genética
4.
ACS Nano ; 14(7): 8287-8298, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32515944

RESUMO

Mononuclear phagocytes such as monocytes, tissue-specific macrophages, and dendritic cells are primary actors in both innate and adaptive immunity. These professional phagocytes can be parasitized by intracellular bacteria, turning them from housekeepers to hiding places and favoring chronic and/or disseminated infection. One of the most infamous is the bacteria that cause tuberculosis (TB), which is the most pandemic and one of the deadliest diseases, with one-third of the world's population infected and an average of 1.8 million deaths/year worldwide. Here we demonstrate the effective targeting and intracellular delivery of antibiotics to infected macrophages both in vitro and in vivo, using pH-sensitive nanoscopic polymersomes made of PMPC-PDPA block copolymer. Polymersomes showed the ability to significantly enhance the efficacy of the antibiotics killing Mycobacterium bovis, Mycobacterium tuberculosis, and another established intracellular pathogen, Staphylococcus aureus. Moreover, they demonstrated to easily access TB-like granuloma tissues-one of the harshest environments to penetrate-in zebrafish models. We thus successfully exploited this targeting for the effective eradication of several intracellular bacteria, including M. tuberculosis, the etiological agent of human TB.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Animais , Humanos , Macrófagos , Monócitos , Tuberculose/tratamento farmacológico , Peixe-Zebra
5.
Front Immunol ; 10: 1721, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31417543

RESUMO

Non-typeable Haemophilus influenzae (NTHi) is a frequent cause of lower respiratory tract infection in people with chronic obstructive pulmonary disease (COPD). Pellino proteins are a family of E3 ubiquitin ligases that are critical regulators of TLR signaling and inflammation. The aim of this study was to identify a role for Pellino-1 in airway defense against NTHi in the context of COPD. Pellino-1 is rapidly upregulated by LPS and NTHi in monocyte-derived macrophages (MDMs) isolated from individuals with COPD and healthy control subjects, in a TLR4 dependent manner. C57BL/6 Peli1-/- and wild-type (WT) mice were subjected to acute (single LPS challenge) or chronic (repeated LPS and elastase challenge) airway inflammation followed by NTHi infection. Both WT and Peli1-/- mice develop airway inflammation in acute and chronic airway inflammation models. Peli1-/- animals recruit significantly more neutrophils to the airway following NTHi infection which is associated with an increase in the neutrophil chemokine, KC, in bronchoalveolar lavage fluid as well as enhanced clearance of NTHi from the lung. These data suggest that therapeutic inhibition of Pellino-1 may augment immune responses in the airway and enhance bacterial clearance in individuals with COPD.


Assuntos
Infecções por Haemophilus/imunologia , Haemophilus influenzae/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Proteínas Nucleares/imunologia , Pneumonia Bacteriana/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Quimiocina CXCL1/genética , Quimiocina CXCL1/imunologia , Infecções por Haemophilus/genética , Infecções por Haemophilus/patologia , Humanos , Macrófagos/patologia , Camundongos , Camundongos Knockout , Monócitos/patologia , Proteínas Nucleares/genética , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/patologia , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia , Ubiquitina-Proteína Ligases/genética
6.
Am J Respir Crit Care Med ; 200(1): 84-97, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30649895

RESUMO

Rationale: Antimicrobial resistance challenges therapy of pneumonia. Enhancing macrophage microbicidal responses would combat this problem but is limited by our understanding of how alveolar macrophages (AMs) kill bacteria. Objectives: To define the role and mechanism of AM apoptosis-associated bacterial killing in the lung. Methods: We generated a unique CD68.hMcl-1 transgenic mouse with macrophage-specific overexpression of the human antiapoptotic Mcl-1 protein, a factor upregulated in AMs from patients at increased risk of community-acquired pneumonia, to address the requirement for apoptosis-associated killing. Measurements and Main Results: Wild-type and transgenic macrophages demonstrated comparable ingestion and initial phagolysosomal killing of bacteria. Continued ingestion (for ≥12 h) overwhelmed initial killing, and a second, late-phase microbicidal response killed viable bacteria in wild-type macrophages, but this response was blunted in CD68.hMcl-1 transgenic macrophages. The late phase of bacterial killing required both caspase-induced generation of mitochondrial reactive oxygen species and nitric oxide, the peak generation of which coincided with the late phase of killing. The CD68.hMcl-1 transgene prevented mitochondrial reactive oxygen species but not nitric oxide generation. Apoptosis-associated killing enhanced pulmonary clearance of Streptococcus pneumoniae and Haemophilus influenzae in wild-type mice but not CD68.hMcl-1 transgenic mice. Bacterial clearance was enhanced in vivo in CD68.hMcl-1 transgenic mice by reconstitution of apoptosis with BH3 mimetics or clodronate-encapsulated liposomes. Apoptosis-associated killing was not activated during Staphylococcus aureus lung infection. Conclusions: Mcl-1 upregulation prevents macrophage apoptosis-associated killing and establishes that apoptosis-associated killing is required to allow AMs to clear ingested bacteria. Engagement of macrophage apoptosis should be investigated as a novel, host-based antimicrobial strategy.


Assuntos
Apoptose/fisiologia , Macrófagos Alveolares/fisiologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Fagocitose/genética , Fagossomos/fisiologia , Pneumonia Bacteriana , Animais , Apoptose/efeitos dos fármacos , Bactérias , Compostos de Bifenilo/farmacologia , Caspases/metabolismo , Ácido Clodrônico/farmacologia , Modelos Animais de Doenças , Haemophilus influenzae , Humanos , Macrófagos Alveolares/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Óxido Nítrico/metabolismo , Nitrofenóis/farmacologia , Piperazinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus , Streptococcus pneumoniae , Sulfonamidas/farmacologia
7.
Front Med (Lausanne) ; 5: 212, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30101145

RESUMO

The tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) is a widely expressed cytokine that can bind five different receptors. TRAIL has been of particular interest for its proposed ability to selectively induce apoptosis in tumour cells. However, it has also been found to regulate a wide variety of non-canonical cellular effects including survival, migration and proliferation via kinase signalling pathways. Lung diseases represent a wide range of conditions affecting multiple tissues. TRAIL has been implicated in several biological processes underlying lung diseases, including angiogenesis, inflammation, and immune regulation. For example, TRAIL is detrimental in pulmonary arterial hypertension-it is upregulated in patient serum and lungs, and drives the underlying proliferative pulmonary vascular remodelling in rodent models. However, TRAIL protects against pulmonary fibrosis in mice models-by inducing apoptosis of neutrophils-and reduced serum TRAIL is found in patients. Conversely, in the airways TRAIL positively regulates inflammation and immune response. In COPD patients and asthmatic patients challenged with antigen, TRAIL and its death receptors are upregulated in serum and airways. Furthermore, TRAIL-deleted mouse models have reduced airway inflammation and remodelling. In the context of respiratory infections, TRAIL assists in immune response, e.g., via T-cell toxicity in influenza infection, and neutrophil killing in S. pneumoniae infection. In this mini-review, we examine the functions of TRAIL and highlight the diverse roles TRAIL has in diseases affecting the lung. Disentangling the facets of TRAIL signalling in lung diseases could help in understanding their pathogenic processes and targeting novel treatments.

8.
Am J Respir Crit Care Med ; 197(12): 1604-1615, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29365279

RESUMO

RATIONALE: People living with HIV are at significantly increased risk of invasive pneumococcal disease, despite long-term antiretroviral therapy (ART). The mechanism explaining this observation remains undefined. OBJECTIVES: To determine if apoptosis-associated microbicidal mechanisms, required to clear intracellular pneumococci that survive initial phagolysosomal killing, are perturbed. METHODS: Alveolar macrophages (AM) were obtained by BAL from healthy donors or HIV-1-seropositive donors on long-term ART with undetectable plasma viral load. Monocyte-derived macrophages (MDM) were obtained from healthy donors and infected with HIV-1BaL or treated with gp120. Macrophages were challenged with opsonized serotype 2 Streptococcus pneumoniae and assessed for apoptosis, bactericidal activity, protein expression, and mitochondrial reactive oxygen species (mROS). AM phenotyping, ultrasensitive HIV-1 RNA quantification, and gp120 measurement were also performed in BAL. MEASUREMENTS AND MAIN RESULTS: HIV-1BaL infection impaired apoptosis, induction of mROS, and pneumococcal killing by MDM. Apoptosis-associated pneumococcal killing was also reduced in AM from ART-treated HIV-1-seropositive donors. BAL fluid from these individuals demonstrated persistent lung CD8+ T lymphocytosis, and gp120 or HIV-1 RNA was also detected. Despite this, transcriptional activity in AM freshly isolated from people living with HIV was broadly similar to healthy volunteers. Instead, gp120 phenocopied the defect in pneumococcal killing in healthy MDM through post-translational modification of Mcl-1, preventing apoptosis induction, caspase activation, and increased mROS generation. Moreover, gp120 also inhibited mROS-dependent pneumococcal killing in MDM. CONCLUSIONS: Despite ART, HIV-1, via gp120, drives persisting innate immune defects in AM microbicidal mechanisms, enhancing susceptibility to pneumococcal disease.


Assuntos
Antirretrovirais/efeitos adversos , Antirretrovirais/uso terapêutico , Resistência à Doença/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Pneumopatias/imunologia , Macrófagos Alveolares/imunologia , Infecções Pneumocócicas/imunologia , Adulto , Feminino , Proteína gp120 do Envelope de HIV/sangue , Humanos , Pneumopatias/microbiologia , Pneumopatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Infecções Pneumocócicas/fisiopatologia
9.
J Clin Invest ; 127(9): 3407-3420, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28805660

RESUMO

Fully activated innate immune cells are required for effective responses to infection, but their prompt deactivation and removal are essential for limiting tissue damage. Here, we have identified a critical role for the prolyl hydroxylase enzyme Phd2 in maintaining the balance between appropriate, predominantly neutrophil-mediated pathogen clearance and resolution of the innate immune response. We demonstrate that myeloid-specific loss of Phd2 resulted in an exaggerated inflammatory response to Streptococcus pneumonia, with increases in neutrophil motility, functional capacity, and survival. These enhanced neutrophil responses were dependent upon increases in glycolytic flux and glycogen stores. Systemic administration of a HIF-prolyl hydroxylase inhibitor replicated the Phd2-deficient phenotype of delayed inflammation resolution. Together, these data identify Phd2 as the dominant HIF-hydroxylase in neutrophils under normoxic conditions and link intrinsic regulation of glycolysis and glycogen stores to the resolution of neutrophil-mediated inflammatory responses. These results demonstrate the therapeutic potential of targeting metabolic pathways in the treatment of inflammatory disease.


Assuntos
Glicogênio/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Neutrófilos/citologia , Infecções Pneumocócicas/imunologia , Doença Aguda , Animais , Lavagem Broncoalveolar , Colite/metabolismo , Glicólise , Humanos , Imunidade Inata , Inflamação , Leucócitos/citologia , Lesão Pulmonar/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Transdução de Sinais
10.
Am J Respir Crit Care Med ; 196(7): 845-855, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28557543

RESUMO

RATIONALE: Chronic obstructive pulmonary disease (COPD) is characterized by impaired clearance of pulmonary bacteria. OBJECTIVES: The effect of COPD on alveolar macrophage (AM) microbicidal responses was investigated. METHODS: AMs were obtained from bronchoalveolar lavage from healthy donors or patients with COPD and challenged with opsonized serotype 14 Streptococcus pneumoniae. Cells were assessed for apoptosis, bactericidal activity, and mitochondrial reactive oxygen species (mROS) production. A transgenic mouse line in which the CD68 promoter ensures macrophage-specific expression of human induced myeloid leukemia cell differentiation protein Mcl-1 (CD68.hMcl-1) was used to model the molecular aspects of COPD. MEASUREMENTS AND MAIN RESULTS: COPD AMs had elevated levels of Mcl-1, an antiapoptotic B-cell lymphoma 2 family member, with selective reduction of delayed intracellular bacterial killing. CD68.hMcl-1 AMs phenocopied the microbicidal defect because transgenic mice demonstrated impaired clearance of pulmonary bacteria and increased neutrophilic inflammation. Murine bone marrow-derived macrophages and human monocyte-derived macrophages generated mROS in response to pneumococci, which colocalized with bacteria and phagolysosomes to enhance bacterial killing. The Mcl-1 transgene increased oxygen consumption rates and mROS expression in mock-infected bone marrow-derived macrophages but reduced caspase-dependent mROS production after pneumococcal challenge. COPD AMs also increased basal mROS expression, but they failed to increase production after pneumococcal challenge, in keeping with reduced intracellular bacterial killing. The defect in COPD AM intracellular killing was associated with a reduced ratio of mROS/superoxide dismutase 2. CONCLUSIONS: Up-regulation of Mcl-1 and chronic adaption to oxidative stress alter mitochondrial metabolism and microbicidal function, reducing the delayed phase of intracellular bacterial clearance in COPD.


Assuntos
Anti-Infecciosos/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Animais , Western Blotting , Lavagem Broncoalveolar , Modelos Animais de Doenças , Citometria de Fluxo , Humanos , Camundongos , Camundongos Transgênicos , Estresse Oxidativo/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/fisiopatologia
11.
Eur J Pharmacol ; 793: 49-55, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27832943

RESUMO

The principal mechanism by which bronchodilator ß-adrenoceptor agonists act is to relax airways smooth muscle although they may also be anti-inflammatory. However, the extent of anti-inflammatory activity and the cell types affected by these agonists are uncertain. The purpose of this study was to evaluate whether ß-adrenoceptor agonists prevent pro-inflammatory cytokine generation from activated human lung macrophages. Macrophages were isolated and purified from human lung. The cells were pre-treated with both short-acting (isoprenaline, salbutamol, terbutaline) and long-acting (formoterol, salmeterol, indacaterol) ß-agonists before activation with lipopolysaccharide (LPS) to induce cytokine (TNFα, IL-6, IL-8 and IL-10) generation. The experiments showed that short-acting ß-agonists were poor inhibitors of cytokine generation. Of the long-acting ß-agonists studied, formoterol was also a weak inhibitor of cytokine generation whereas only indacaterol and salmeterol showed moderate inhibitory activity. Further experiments using the ß2-adrenoceptor antagonist ICI-118,551 suggested that the effects of indacaterol were likely to be mediated by ß2-adrenoceptors whereas those of salmeterol were not. These findings were corroborated by functional desensitization studies in which the inhibitory effects of indacaterol appeared to be receptor-mediated whereas those of salmeterol were not. Taken together, the data indicate that the anti-inflammatory effects of ß-adrenoceptor agonists on human lung macrophages are modest.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Anti-Inflamatórios/farmacologia , Pulmão/citologia , Macrófagos/efeitos dos fármacos , Adulto , Idoso , Idoso de 80 Anos ou mais , Citocinas/biossíntese , Citocinas/metabolismo , Feminino , Humanos , Indanos/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Monócitos/citologia , Quinolonas/farmacologia , Fatores de Tempo
12.
Br J Pharmacol ; 173(21): 3099-3109, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27460634

RESUMO

BACKGROUND AND PURPOSE: PGE2 inhibits cytokine generation from human lung macrophages. However, the EP receptor that mediates this beneficial anti-inflammatory effect of PGE2 has not been defined. The aim of this study was to identify the EP receptor by which PGE2 inhibits cytokine generation from human lung macrophages. This was determined by using recently developed EP receptor ligands. EXPERIMENTAL APPROACH: The effects of PGE2 and EP-selective agonists on LPS-induced generation of TNF-α and IL-6 from macrophages were evaluated. The effects of EP2 -selective (PF-04852946, PF-04418948) and EP4 -selective (L-161,982, CJ-042794) receptor antagonists on PGE2 responses were studied. The expression of EP receptor subtypes by human lung macrophages was determined by RT-PCR. KEY RESULTS: PGE2 inhibited LPS-induced and Streptococcus pneumoniae-induced cytokine generation from human lung macrophages. Analysis of mRNA levels indicated that macrophages expressed EP2 and EP4 receptors. L-902,688 (EP4 receptor-selective agonist) was considerably more potent than butaprost (EP2 receptor-selective agonist) as an inhibitor of TNF-α generation from macrophages. EP2 receptor-selective antagonists had marginal effects on the PGE2 inhibition of TNF-α generation, whereas EP4 receptor-selective antagonists caused rightward shifts in the PGE2 concentration-response curves. CONCLUSIONS AND IMPLICATIONS: These studies demonstrate that the EP4 receptor is the principal receptor that mediates the anti-inflammatory effects of PGE2 on human lung macrophages. This suggests that EP4 receptor agonists could be effective anti-inflammatory agents in human lung disease.


Assuntos
Antibacterianos/farmacologia , Anti-Inflamatórios não Esteroides/farmacologia , Dinoprostona/farmacologia , Macrófagos Alveolares/efeitos dos fármacos , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Antibacterianos/química , Anti-Inflamatórios não Esteroides/química , Dinoprostona/química , Humanos , Lipopolissacarídeos/antagonistas & inibidores , Lipopolissacarídeos/farmacologia , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/microbiologia , Testes de Sensibilidade Microbiana , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/genética , Streptococcus pneumoniae/efeitos dos fármacos
13.
Cell Microbiol ; 18(1): 80-96, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26248337

RESUMO

Macrophages are critical effectors of the early innate response to bacteria in tissues. Phagocytosis and killing of bacteria are interrelated functions essential for bacterial clearance but the rate-limiting step when macrophages are challenged with large numbers of the major medical pathogen Staphylococcus aureus is unknown. We show that macrophages have a finite capacity for intracellular killing and fail to match sustained phagocytosis with sustained microbial killing when exposed to large inocula of S. aureus (Newman, SH1000 and USA300 strains). S. aureus ingestion by macrophages is associated with a rapid decline in bacterial viability immediately after phagocytosis. However, not all bacteria are killed in the phagolysosome, and we demonstrate reduced acidification of the phagolysosome, associated with failure of phagolysosomal maturation and reduced activation of cathepsin D. This results in accumulation of viable intracellular bacteria in macrophages. We show macrophages fail to engage apoptosis-associated bacterial killing. Ultittop mately macrophages with viable bacteria undergo cell lysis, and viable bacteria are released and can be internalized by other macrophages. We show that cycles of lysis and reuptake maintain a pool of viable intracellular bacteria over time when killing is overwhelmed and demonstrate intracellular persistence in alveolar macrophages in the lungs in a murine model.


Assuntos
Macrófagos/imunologia , Macrófagos/microbiologia , Viabilidade Microbiana , Fagocitose , Pneumonia Estafilocócica/patologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/fisiologia , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Camundongos
14.
Elife ; 42015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-25951517

RESUMO

Lack of reproducibility is an ongoing problem in some areas of the biomedical sciences. Poor experimental design and a failure to engage with experienced statisticians at key stages in the design and analysis of experiments are two factors that contribute to this problem. The RIPOSTE (Reducing IrreProducibility in labOratory STudiEs) framework has been developed to support early and regular discussions between scientists and statisticians in order to improve the design, conduct and analysis of laboratory studies and, therefore, to reduce irreproducibility. This framework is intended for use during the early stages of a research project, when specific questions or hypotheses are proposed. The essential points within the framework are explained and illustrated using three examples (a medical equipment test, a macrophage study and a gene expression study). Sound study design minimises the possibility of bias being introduced into experiments and leads to higher quality research with more reproducible results.


Assuntos
Pesquisa Biomédica/métodos , Projetos de Pesquisa/normas , Pesquisa Biomédica/tendências , Interpretação Estatística de Dados , Reprodutibilidade dos Testes
15.
mBio ; 5(5): e01710-14, 2014 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-25293758

RESUMO

Intracellular killing of Streptococcus pneumoniae is complemented by induction of macrophage apoptosis. Here, we show that the toxin pneumolysin (PLY) contributes both to lysosomal/phagolysosomal membrane permeabilization (LMP), an upstream event programing susceptibility to apoptosis, and to apoptosis execution via a mitochondrial pathway, through distinct mechanisms. PLY is necessary but not sufficient for the maximal induction of LMP and apoptosis. PLY's ability to induce both LMP and apoptosis is independent of its ability to form cytolytic pores and requires only the first three domains of PLY. LMP involves TLR (Toll-like receptor) but not NLRP3/ASC (nucleotide-binding oligomerization domain [Nod]-like receptor family, pyrin domain-containing protein 3/apoptosis-associated speck-like protein containing a caspase recruitment domain) signaling and is part of a PLY-dependent but phagocytosis-independent host response that includes the production of cytokines, including interleukin-1 beta (IL-1ß). LMP involves progressive and selective permeability to 40-kDa but not to 250-kDa fluorescein isothiocyanate (FITC)-labeled dextran, as PLY accumulates in the cytoplasm. In contrast, the PLY-dependent execution of apoptosis requires phagocytosis and is part of a host response to intracellular bacteria that also includes NO generation. In cells challenged with PLY-deficient bacteria, reconstitution of LMP using the lysomotrophic detergent LeuLeuOMe favored cell necrosis whereas PLY reconstituted apoptosis. The results suggest that PLY contributes to macrophage activation and cytokine production but also engages LMP. Following bacterial phagocytosis, PLY triggers apoptosis and prevents macrophage necrosis as a component of a broad-based antimicrobial strategy. This illustrates how a key virulence factor can become the focus of a multilayered and coordinated innate response by macrophages, optimizing pathogen clearance and limiting inflammation. Importance: Streptococcus pneumoniae, the commonest cause of bacterial pneumonia, expresses the toxin pneumolysin, which can make holes in cell surfaces, causing tissue damage. Macrophages, resident immune cells essential for responses to bacteria in tissues, activate a program of cell suicide called apoptosis, maximizing bacterial clearance and limiting harmful inflammation. We examined pneumolysin's role in activating this response. We demonstrate that pneumolysin did not directly form holes in cells to trigger apoptosis and show that pneumolysin has two distinct roles which require only part of the molecule. Pneumolysin and other bacterial factors released by bacteria that have not been eaten by macrophages activate macrophages to release inflammatory factors but also make the cell compartment containing ingested bacteria leaky. Once inside the cell, pneumolysin ensures that the bacteria activate macrophage apoptosis, rather than necrosis, enhancing bacterial killing and limiting inflammation. This dual response to pneumolysin is critical for an effective immune response to S. pneumoniae.


Assuntos
Apoptose , Membranas Intracelulares/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos , Streptococcus pneumoniae/imunologia , Estreptolisinas/toxicidade , Animais , Proteínas de Bactérias/toxicidade , Células Cultivadas , Humanos , Camundongos , Streptococcus pneumoniae/metabolismo
16.
Blood ; 123(3): 366-76, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24196071

RESUMO

Neutrophil lifespan and function are regulated by hypoxia via components of the hypoxia inducible factor (HIF)/von Hippel Lindau/hydroxylase pathway, including specific roles for HIF-1α and prolyl hydroxylase-3. HIF-2α has both distinct and overlapping biological roles with HIF-1α and has not previously been studied in the context of neutrophil biology. We investigated the role of HIF-2α in regulating key neutrophil functions. Human and murine peripheral blood neutrophils expressed HIF-2α, with expression up-regulated by acute and chronic inflammatory stimuli and in disease-associated inflammatory neutrophil. HIF2A gain-of-function mutations resulted in a reduction in neutrophil apoptosis both ex vivo, through the study of patient cells, and in vivo in a zebrafish tail injury model. In contrast, HIF-2α-deficient murine inflammatory neutrophils displayed increased sensitivity to nitrosative stress induced apoptosis ex vivo and increased neutrophil apoptosis in vivo, resulting in a reduction in neutrophilic inflammation and reduced tissue injury. Expression of HIF-2α was temporally dissociated from HIF-1α in vivo and predominated in the resolution phase of inflammation. These data support a critical and selective role for HIF-2α in persistence of neutrophilic inflammation and provide a platform to dissect the therapeutic utility of targeting HIF-2α in chronic inflammatory diseases.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica , Inflamação , Neutrófilos/metabolismo , Animais , Apoptose , Hipóxia Celular , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Muramidase , Neutrófilos/citologia , Fagocitose , Fenótipo , RNA/metabolismo , Explosão Respiratória , Peixe-Zebra
17.
J Infect ; 66(1): 1-17, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23046969

RESUMO

Cardiovascular disease has emerged as the leading cause of mortality worldwide. Acute coronary syndromes occur as a result of thrombotic complications at the site of atherosclerotic plaques, often following rupture of the fibrous cap of vulnerable plaques. A growing body of evidence from clinical and experimental studies suggests that acute respiratory tract infections can act as a trigger for acute coronary syndromes. The mechanism underlying this association has yet to be established. We explore the mechanistic links between acute respiratory tract infection and acute coronary syndromes, with a particular focus on the host response to infection and its potential interaction with pathogenic processes involved in atherosclerosis and atherosclerotic plaque rupture. The prothrombotic and haemodynamic effects of acute respiratory infection are also discussed. We review mechanistic studies as well as clinical trial data to investigate potential links between acute coronary syndromes and acute respiratory infection. Understanding the link between acute respiratory infections and acute coronary syndromes should help improve the outcome of acute coronary syndromes.


Assuntos
Arritmias Cardíacas/epidemiologia , Infecções Comunitárias Adquiridas/epidemiologia , Insuficiência Cardíaca/epidemiologia , Pneumonia/epidemiologia , Feminino , Humanos , Masculino
18.
Cell Tissue Res ; 350(3): 455-64, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22986921

RESUMO

Despite being initially identified in mice, little is known about the sites of production of members of the BPI fold (BPIF) containing (PLUNC) family of putative innate defence proteins in this species. These proteins have largely been considered to be specificaly expressed in the respiratory tract, and we have recently shown that they exhibit differential expression in the epithelium of the proximal airways. In this study, we have used species-specific antibodies to systematically localize two members of this protein family; BPIFA1 (PLUNC/SPLUNC1) and BPIFB1 (LPLUNC1) in adult mice. In general, these proteins exhibit distinct and only partially overlapping localization. BPIFA1 is highly expressed in the respiratory epithelium and Bowman's glands of the nasal passages, whereas BPIFB1 is present in small subset of goblet cells in the nasal passage and pharynx. BPIFB1 is also present in the serous glands in the proximal tongue where is co-localised with the salivary gland specific family member, BPIFA2E (parotid secretory protein) and also in glands of the soft palate. Both proteins exhibit limited expression outside of these regions. These results are consistent with the localization of the proteins seen in man. Knowledge of the complex expression patterns of BPIF proteins in these regions will allow the use of tractable mouse models of disease to dissect their function.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas/metabolismo , Boca/citologia , Boca/metabolismo , Cavidade Nasal/citologia , Cavidade Nasal/metabolismo , Fosfoproteínas/metabolismo , Sequência de Aminoácidos , Animais , Proteínas de Transporte/genética , Diferenciação Celular/fisiologia , Glicoproteínas/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fosfoproteínas/genética
19.
PLoS Pathog ; 8(7): e1002814, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22829769

RESUMO

Monocytes and T-cells are critical to the host response to acute bacterial infection but monocytes are primarily viewed as amplifying the inflammatory signal. The mechanisms of cell death regulating T-cell numbers at sites of infection are incompletely characterized. T-cell death in cultures of peripheral blood mononuclear cells (PBMC) showed 'classic' features of apoptosis following exposure to pneumococci. Conversely, purified CD3(+) T-cells cultured with pneumococci demonstrated necrosis with membrane permeabilization. The death of purified CD3(+) T-cells was not inhibited by necrostatin, but required the bacterial toxin pneumolysin. Apoptosis of CD3(+) T-cells in PBMC cultures required 'classical' CD14(+) monocytes, which enhanced T-cell activation. CD3(+) T-cell death was enhanced in HIV-seropositive individuals. Monocyte-mediated CD3(+) T-cell apoptotic death was Fas-dependent both in vitro and in vivo. In the early stages of the T-cell dependent host response to pneumococci reduced Fas ligand mediated T-cell apoptosis was associated with decreased bacterial clearance in the lung and increased bacteremia. In summary monocytes converted pathogen-associated necrosis into Fas-dependent apoptosis and regulated levels of activated T-cells at sites of acute bacterial infection. These changes were associated with enhanced bacterial clearance in the lung and reduced levels of invasive pneumococcal disease.


Assuntos
Apoptose , Proteína Ligante Fas/metabolismo , Monócitos/imunologia , Infecções Pneumocócicas/imunologia , Linfócitos T/fisiologia , Animais , Bacteriemia , Proteínas de Bactérias , Complexo CD3/biossíntese , Células Cultivadas , Infecções por HIV/imunologia , HIV-1/imunologia , Humanos , Imidazóis/farmacologia , Indóis/farmacologia , Receptores de Lipopolissacarídeos/biossíntese , Pulmão/microbiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/microbiologia , Necrose , Streptococcus pneumoniae/imunologia , Streptococcus pneumoniae/patogenicidade , Estreptolisinas , Linfócitos T/imunologia , Linfócitos T/microbiologia
20.
Thorax ; 67(11): 985-92, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22735687

RESUMO

BACKGROUND: Therapeutic strategies to modulate the host response to bacterial pneumonia are needed to improve outcomes during community-acquired pneumonia. This study used mice with impaired Fas signalling to examine susceptibility to pneumococcal pneumonia and decoy receptor 3 analogue (DcR3-a) to correct factors associated with increased susceptibility. METHODS: Wild-type mice and those with varying degrees of impairment of Fas (lpr) or Fas ligand signalling (gld) were challenged with Streptococcus pneumoniae and microbiological and immunological outcomes measured in the presence or absence of DcR3-a. RESULTS: During established pneumonia, neutrophils became the predominant cell in the airway and gld mice were less able to clear bacteria from the lungs, demonstrating localised impairment of pulmonary neutrophil function in comparison to lpr or wild-type mice. T-cells from gld mice had enhanced activation and reduced apoptosis in comparison to wild-type and lpr mice during established pneumonia. Treatment with DcR3-a reduced T-cell activation and corrected the defect in pulmonary bacterial clearance in gld mice. CONCLUSIONS: The results suggest that imbalance in tumour necrosis factor superfamily signalling and excessive T-cell activation can impair bacterial clearance in the lung but that DcR3-a treatment can reduce T-cell activation, restore optimal pulmonary neutrophil function and enhance bacterial clearance during S pneumoniae infection.


Assuntos
Proteína Ligante Fas/metabolismo , Neutrófilos/imunologia , Fagócitos/imunologia , Pneumonia Pneumocócica/imunologia , Membro 6b de Receptores do Fator de Necrose Tumoral/farmacologia , Animais , Modelos Animais de Doenças , Proteína Ligante Fas/farmacologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neutrófilos/efeitos dos fármacos , Fagócitos/efeitos dos fármacos , Pneumonia Pneumocócica/metabolismo , Pneumonia Pneumocócica/microbiologia , Pneumonia Pneumocócica/terapia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Streptococcus pneumoniae/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA