Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell ; 187(7): 1666-1684.e26, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38490194

RESUMO

Diminished hepatocyte regeneration is a key feature of acute and chronic liver diseases and after extended liver resections, resulting in the inability to maintain or restore a sufficient functional liver mass. Therapies to restore hepatocyte regeneration are lacking, making liver transplantation the only curative option for end-stage liver disease. Here, we report on the structure-based development and characterization (nuclear magnetic resonance [NMR] spectroscopy) of first-in-class small molecule inhibitors of the dual-specificity kinase MKK4 (MKK4i). MKK4i increased liver regeneration upon hepatectomy in murine and porcine models, allowed for survival of pigs in a lethal 85% hepatectomy model, and showed antisteatotic and antifibrotic effects in liver disease mouse models. A first-in-human phase I trial (European Union Drug Regulating Authorities Clinical Trials [EudraCT] 2021-000193-28) with the clinical candidate HRX215 was conducted and revealed excellent safety and pharmacokinetics. Clinical trials to probe HRX215 for prevention/treatment of liver failure after extensive oncological liver resections or after transplantation of small grafts are warranted.


Assuntos
Inibidores Enzimáticos , Falência Hepática , MAP Quinase Quinase 4 , Animais , Humanos , Camundongos , Hepatectomia/métodos , Hepatócitos , Fígado , Hepatopatias/tratamento farmacológico , Falência Hepática/tratamento farmacológico , Falência Hepática/prevenção & controle , Regeneração Hepática , Suínos , MAP Quinase Quinase 4/antagonistas & inibidores , Inibidores Enzimáticos/uso terapêutico
2.
EMBO J ; 42(12): e111272, 2023 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-37143403

RESUMO

Patients with chronic obstructive pulmonary disease (COPD) are still waiting for curative treatments. Considering its environmental cause, we hypothesized that COPD will be associated with altered epigenetic signaling in lung cells. We generated genome-wide DNA methylation maps at single CpG resolution of primary human lung fibroblasts (HLFs) across COPD stages. We show that the epigenetic landscape is changed early in COPD, with DNA methylation changes occurring predominantly in regulatory regions. RNA sequencing of matched fibroblasts demonstrated dysregulation of genes involved in proliferation, DNA repair, and extracellular matrix organization. Data integration identified 110 candidate regulators of disease phenotypes that were linked to fibroblast repair processes using phenotypic screens. Our study provides high-resolution multi-omic maps of HLFs across COPD stages. We reveal novel transcriptomic and epigenetic signatures associated with COPD onset and progression and identify new candidate regulators involved in the pathogenesis of chronic lung diseases. The presence of various epigenetic factors among the candidates demonstrates that epigenetic regulation in COPD is an exciting research field that holds promise for novel therapeutic avenues for patients.


Assuntos
Doença Pulmonar Obstrutiva Crônica , Transcriptoma , Humanos , Epigênese Genética , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/patologia , Pulmão/patologia , Perfilação da Expressão Gênica , Metilação de DNA
3.
ERJ Open Res ; 6(4)2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33313305

RESUMO

BACKGROUND: Epithelial sodium channel (ENaC) is an important regulator of airway surface liquid volume; ENaC is hyperactivated in cystic fibrosis (CF). ENaC inhibition is a potential therapeutic target for CF. Here, we report in vitro and in vivo results for BI 1265162, an inhaled ENaC inhibitor currently in Phase II clinical development, administered via the Respimat® Soft Mist™ inhaler. METHODS: In vitro inhibition of sodium ion (Na+) transport by BI 1265162 was tested in mouse renal collecting duct cells (M1) and human bronchial epithelial cells (NCI-H441); inhibition of water transport was measured using M1 cells. In vivo inhibition of liquid absorption from rat airway epithelium and acceleration of mucociliary clearance (MCC) in sheep lungs were assessed. Fully differentiated normal and CF human epithelium was used to measure the effect of BI 1265162 with or without ivacaftor and lumacaftor on water transport and MCC. RESULTS: BI 1265162 dose-dependently inhibited Na+ transport and decreased water resorption in cell line models. BI 1265162 reduced liquid absorption in rat lungs and increased MCC in sheep. No effects on renal function were seen in the animal models. BI 1265162 alone and in combination with CF transmembrane conductance regulator (CFTR) modulators decreased water transport and increased MCC in both normal and CF airway human epithelial models and also increased the effects of CFTR modulators in CF epithelium to reach the effect size seen in healthy epithelium with ivacaftor/lumacaftor alone. CONCLUSION: These results demonstrate the potential of BI 1265162 as a mutation agnostic, ENaC-inhibitor-based therapy for CF.

4.
Shock ; 51(1): 68-77, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29424792

RESUMO

INTRODUCTION: Hemorrhagic shock accounts for a large amount of trauma-related mortality. The severity of trauma can be further aggravated by an additional blunt chest trauma (TxT), which independently contributes to mortality upon the development of an acute lung injury (ALI). Besides, cigarette smoke (CS) exposure before TxT enhanced posttraumatic inflammation, thereby aggravating ALI. We therefore aimed to characterize the impact of an acute and/or chronic lung injury on organ dysfunction in a murine model of traumatic hemorrhagic shock (HS). METHODS: After 3 weeks of CS exposure, anesthetized mice underwent HS with/without TxT. Hemorrhagic shock was implemented for 1 h followed by retransfusion of shed blood and intensive care therapy for 4 h including lung-protective mechanical ventilation, fluid resuscitation, and noradrenaline titrated to maintain mean arterial pressure ≥50 mmHg. Lung mechanics and gas exchange were assessed together with systemic hemodynamics, metabolism, and acid-base status. Postmortem blood and tissue samples were analyzed for cytokine and chemokine levels, protein expression, mitochondrial respiration, and histological changes. RESULTS: CS exposure and HS alone coincided with increased inflammation, decreased whole blood sulfide concentrations, and decreased diaphragmatic mitochondrial respiration. CS-exposed mice, which were subjected to TxT and subsequent HS, showed hemodynamic instability, acute kidney injury, and high mortality. CONCLUSIONS: Chronic CS exposure per se had the strongest impact on inflammatory responses. The degree of inflammation was similar upon an additional TxT, however, mice presented with organ dysfunction and increased mortality rates. Hence, in mice the degree of inflammation may be dissociated from the severity of organ dysfunction or injury.


Assuntos
Injúria Renal Aguda/sangue , Lesão Pulmonar Aguda/sangue , Fumar Cigarros , Choque Hemorrágico/sangue , Sulfetos/sangue , Ferimentos não Penetrantes/sangue , Doença Aguda , Animais , Fumar Cigarros/efeitos adversos , Fumar Cigarros/sangue , Modelos Animais de Doenças , Inflamação/sangue , Masculino , Camundongos
5.
Transl Res ; 203: 15-30, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30142307

RESUMO

The present study examined the effects of simultaneous inhibition of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) receptor signaling with BIBF1000, a novel triple tyrosine kinase inhibitor on preventing and reversing the progression of severe pulmonary arterial hypertension (PAH) in an experimental model in rats. Left pneumonectomized male Wistar rats were injected with monocrotaline to induce PAH. Treatment with BIBF1000 from day 1 to day 21 after monocrotaline injection attenuated PAH development, as evidenced by lower values for pulmonary artery pressure (mPAP), right ventricular pressure (RVSP), pulmonary arterial neointimal formation, and the ratio of right ventricular weight to left ventricular and septum weight [RV/(LV+S)] on day 21 compared to control rats. Treatment with BIBF1000 from day 21 to day 42 after monocrotaline injection reversed established PAH as shown by normalized values for mPAP and RVSP, RV/(LV+S) ratio, pulmonary arterial occlusion scores, levels of heart and lung fibrosis, as well as improved survival. Treatment with BIBF1000 reduced inflammatory cell recruitment in bronchoalveolar lavage and lung tissues, reduced CD-68 positive macrophages and expression of proliferating cell nuclear antigen in the perivascular areas, and reduced TNF-α and growth factor productions, and inhibited the phosphorylation of AKT and GSK3ß in lungs. In addition, BIBF1000 inhibited pulmonary artery smooth muscle cells migration and proliferation from rat pulmonary artery explant cultures. Simultaneous inhibition of VEGF, PDGF, and FGF receptor signaling by BIBF1000 prevents and reverses the progression of severe pulmonary arterial hypertension and vascular remodeling in this experimental model.


Assuntos
Hipertensão Pulmonar/tratamento farmacológico , Indóis/farmacologia , Neointima , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Glicogênio Sintase Quinase 3 beta/metabolismo , Inflamação , Masculino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular
6.
Am J Respir Crit Care Med ; 199(5): 622-630, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30141961

RESUMO

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a fatal disease with a variable and unpredictable course. OBJECTIVES: To determine whether BAL cell gene expression is predictive of survival in IPF. METHODS: This retrospective study analyzed the BAL transcriptome of three independent IPF cohorts: Freiburg (Germany), Siena (Italy), and Leuven (Belgium) including 212 patients. BAL cells from 20 healthy volunteers, 26 patients with sarcoidosis stage III and IV, and 29 patients with chronic obstructive pulmonary disease were used as control subjects. Survival analysis was performed by Cox models and component-wise boosting. Presence of airway basal cells was tested by immunohistochemistry and flow cytometry. MEASUREMENTS AND MAIN RESULTS: A total of 1,582 genes were predictive of mortality in the IPF derivation cohort in univariate analyses adjusted for age and sex at false discovery rate less than 0.05. A nine-gene signature, derived from the discovery cohort (Freiburg), performed well in both replication cohorts, Siena (P < 0.0032) and Leuven (P = 0.0033). nCounter expression analysis confirmed the array results (P < 0.0001). The genes associated with mortality in BAL cells were significantly enriched for genes expressed in airway basal cells. Further analyses by gene expression, flow cytometry, and immunohistochemistry showed an increase in airway basal cells in BAL and tissues of IPF compared with control subjects, but not in chronic obstructive pulmonary disease or sarcoidosis. CONCLUSIONS: Our results identify and validate a BAL signature that predicts mortality in IPF and improves the accuracy of outcome prediction based on clinical parameters. The BAL signature associated with mortality unmasks a potential role for airway basal cells in IPF.


Assuntos
Líquido da Lavagem Broncoalveolar/citologia , Fibrose Pulmonar Idiopática/metabolismo , Mucosa Respiratória/metabolismo , Idoso , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Humanos , Fibrose Pulmonar Idiopática/mortalidade , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Valor Preditivo dos Testes , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Análise de Sobrevida
7.
Pulm Pharmacol Ther ; 44: 96-105, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28315490

RESUMO

Smoking is an important risk factor for the development of chronic obstructive pulmonary disease (COPD) and viral infections are believed to be major triggers of exacerbations, which periodically lead to a worsening of symptoms. The pro-inflammatory IL-1 family members IL-1α and IL-1ß are increased in COPD patients and might contribute to disease pathology. We investigated whether individual or combined inhibition of these cytokines reduced lung inflammation in cigarette smoke (CS)-exposed and H1N1-infected BALB/c mice. Animals were treated with individual or combined antibodies (Abs) directed against IL-1α, IL-1ß or IL-1R1. Cells in BAL fluid and cytokines/chemokines in lung homogenate were determined. The viral load was investigated. Blocking IL-1α had significant suppressive effects on total cells, neutrophils, and macrophages. Furthermore, it reduced KC levels significantly. Blocking of IL-1ß did not provide significant activity. In primary human bronchial epithelial air-liquid-interface cell cultures infected with H1N1, IL-1α Abs but not IL-1ß Abs reduced levels of TNF-α and IL-6. Concomitant usage of Abs against IL-1α/IL-1ß revealed strong effects in vivo and reduced total cells, neutrophils and macrophages. Additionally, levels of KC, IL-6, TNF-α, MCP-1, MIP-1α and MIP-1ß were significantly reduced and ICAM-1 and MUC5 A/C mRNA expression was attenuated. The viral load decreased significantly upon combined IL-1α/IL-1ß Ab treatment. Blocking the IL-1R1 provided significant effects on total cells, neutrophils and macrophages but was inferior compared to inhibiting both its soluble ligands IL-1α/IL-1ß. Our results suggest that combined inhibition of IL-1α/IL-1ß might be beneficial to reduce CS/H1N1-induced airway inflammation. Moreover, combined targeting of both IL-1α/IL-1ß might be more efficient compared to individual neutralization IL-1α or IL-1ß or inhibition of the IL-1R1.


Assuntos
Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Pneumonia/prevenção & controle , Fumar/efeitos adversos , Animais , Anticorpos , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/etiologia , Inflamação/patologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Influenza Humana/complicações , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo , Infecções por Orthomyxoviridae/complicações , Pneumonia/etiologia , Fatores de Risco , Fumaça/efeitos adversos , Nicotiana , Fator de Necrose Tumoral alfa/metabolismo
8.
Shock ; 47(2): 193-199, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27559703

RESUMO

Both acute and chronic lung injury are associated with up-regulation of the pulmonary expression of the purinergic receptors P2XR4 and P2XR7. Genetic deletion or blockade of P2XR7 attenuated pulmonary hyperinflammation, but simultaneous P2XR4 up-regulation compensated for P2XR7 deletion. Therefore, we tested the hypothesis whether genetic P2XR4 deletion would attenuate the pulmonary inflammatory response and thereby improve organ function after blunt chest trauma in mice with and without pretraumatic cigarette smoke (CS) exposure.After 3 weeks to 4 weeks of exposure to CS, anesthetized wildtype or P2XR4 mice (n = 32) underwent a blast wave-induced blunt chest trauma followed by 4 h of lung-protective mechanical ventilation, fluid resuscitation, and noradrenaline support to maintain mean arterial pressure >55 mm Hg. Hemodynamics, lung mechanics, gas exchange, and acid-base status were measured together with blood and tissue cytokine and chemokine concentrations, heme oxygenase-1, B-cell lymphoma-extra large (Bcl-xL), endogenous nuclear factor-κB inhibitor (IκBα) expression, nitrotyrosine formation, purinergic receptor expression, and histological scoring.Despite a significant increase in the histopathology score in both CS-exposed groups, neither CS exposure nor P2XR4 deletion had any significant effect on post-traumatic pulmonary function and inflammatory response. However, P2XR4 deletion was associated with attenuated impairment of glucose homeostasis and acid-base-status after CS exposure and chest trauma.In conclusion, genetic P2XR4 deletion failed to attenuate the acute post-traumatic pulmonary inflammatory response. The improved glucose homeostasis and acid-base-status after CS exposure in the P2XR4 group was possibly due to less alveolar hypoxia-induced right ventricular remodeling resulting in preserved liver metabolic capacity.


Assuntos
Receptores Purinérgicos P2Y2/metabolismo , Receptores Purinérgicos/metabolismo , Fumar/efeitos adversos , Traumatismos Torácicos/metabolismo , Ferimentos não Penetrantes/metabolismo , Animais , Immunoblotting , Imuno-Histoquímica , Camundongos , Receptores Purinérgicos/genética
9.
Shock ; 47(4): 491-499, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27685807

RESUMO

Pretraumatic cigarette smoke (CS) exposure aggravates posttraumatic acute lung injury (ALI). Cystathionine-γ-lyase (CSE) protects against ALI and CS exposure-induced chronic obstructive lung disease (COPD). Therefore, we tested the hypothesis whether genetic CSE knockout (CSE) would aggravate posttraumatic ALI after CS exposure. After 3 to 4 weeks of CS exposure, anesthetized wild-type (WT) and CSE mice underwent blunt chest trauma, surgical instrumentation and 4 h of lung-protective mechanical ventilation. We measured hemodynamics, lung mechanics, gas exchange, metabolism, and acid-base status together with blood and tissue cytokine and chemokine levels, tissue expression of mediator proteins, parameters of oxidative and nitrosative stress, and histology. CSE mice without CS exposure showed higher cytokine and chemokine levels, and this was further enhanced by CS exposure, particularly in males. CS exposure in WT mice aggravated posttraumatic alveolar membrane thickening, dystelectasis, and inflammatory cell accumulation, which was associated with higher thoracopulmonary compliance. Pretraumatic CS exposure in CSE mice produced a similar response, except for less alveolar membrane thickening, most likely due to lung hyperinflation. CS-exposed WT mice showed the most pronounced metabolic acidosis, while CS exposure in CSE mice resulted in the lowest blood glucose levels. Urinary output and anesthesia rate were highest in male CS-exposed CSE animals. In conclusion, in murine acute-on-chronic pulmonary disease, CSE knockout aggravated posttraumatic inflammation, which was further worsened upon pretraumatic CS exposure, and this effect was particularly pronounced in males. Hence, maintaining CSE expression is critically important for stress adaptation during ALI and CS-induced COPD, most likely in a gender-dependent manner.


Assuntos
Fumar Cigarros/efeitos adversos , Cistationina gama-Liase/metabolismo , Traumatismos Torácicos/enzimologia , Traumatismos Torácicos/metabolismo , Ferimentos não Penetrantes/enzimologia , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/metabolismo , Animais , Quimiocinas/metabolismo , Ensaio Cometa , Cistationina gama-Liase/deficiência , Cistationina gama-Liase/genética , Citocinas/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Immunoblotting , Pulmão/imunologia , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença Pulmonar Obstrutiva Crônica/enzimologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Traumatismos Torácicos/genética , Ferimentos não Penetrantes/genética , Ferimentos não Penetrantes/metabolismo
10.
J Pharmacol Exp Ther ; 357(3): 606-18, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27016458

RESUMO

Viral infections trigger exacerbations in chronic obstructive pulmonary disease (COPD), and tiotropium, a M3 receptor antagonist, reduces exacerbations in patients by unknown mechanisms. In this report, we investigated whether tiotropium has anti-inflammatory effects in mice exposed to cigarette smoke (CS) and infected with influenza virus A/PR/8/34 (H1N1) or respiratory syncytial virus (RSV) and compared these effects with those of steroid fluticasone and PDE4-inhibitor roflumilast. Mice were exposed to CS; infected with H1N1 or RSV; and treated with tiotropium, fluticasone, or roflumilast. The amount of cells and cytokine levels in the airways, lung function, and viral load was determined. NCI-H292 cells were infected with H1N1 or RSV and treated with the drugs. In CS/H1N1-exposed mice, tiotropium reduced neutrophil and macrophage numbers and levels of interleukin-6 (IL-6) and interferon-γ (IFN-γ) in the airways and improved lung function. In contrast, fluticasone increased the loss of body weight; failed to reduce neutrophil or macrophage numbers; increased IL-6, KC, and tumor necrosis factor-α (TNF-α) in the lungs; and worsened lung function. Treatment with roflumilast reduced macrophage numbers, IL-6, and KC in the lungs but had no effect on neutrophil numbers or lung function. In CS/RSV-exposed mice, treatment with tiotropium, but not fluticasone or roflumilast, reduced neutrophil numbers and IL-6 and TNF-α levels in the lungs. Viral load of H1N1 and RSV was significantly elevated in CS/virus-exposed mice and NCI-H292 cells after fluticasone treatment, whereas tiotropium and roflumilast had no effect. In conclusion, tiotropium has anti-inflammatory effects on CS/virus-induced inflammation in mice that are superior to the effects of roflumilast and fluticasone. This finding might help to explain the observed reduction of exacerbation rates in COPD patients.


Assuntos
Anti-Inflamatórios/farmacologia , Nicotiana/química , Pneumonia/induzido quimicamente , Pneumonia/virologia , Fumaça/efeitos adversos , Brometo de Tiotrópio/farmacologia , Animais , Anti-Inflamatórios/uso terapêutico , Peso Corporal/efeitos dos fármacos , Contagem de Células , Colina O-Acetiltransferase/genética , Citocinas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Vírus da Influenza A Subtipo H1N1/fisiologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Pulmão/fisiopatologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos C57BL , Oxo-Ácido-Liases/genética , Pneumonia/tratamento farmacológico , Pneumonia/patologia , Vírus Sinciciais Respiratórios/fisiologia , Brometo de Tiotrópio/uso terapêutico
11.
Pharmacol Res ; 104: 132-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26747401

RESUMO

Kinin B1 receptors are implicated in asthmatic airway inflammation. Here we tested this hypothesis by examining the anti-inflammatory effects of BI113823, a novel non-peptide orally active kinin B1 receptor antagonist in mice sensitized to ovalbumin (OVA). Male Balb-c mice were randomly assigned to four study groups: (1) control, (2) OVA+vehicle, (3) OVA+BI113823, (4) OVA+dexamethasone. Mice were sensitized intraperitoneally with 75µg ovalbumin on days 1 and 8. On days 15-17, mice were challenged intranasally with 50µg of ovalbumin. Mice received vehicle, BI113823, or dexamethasone (positive control) on days 16-18. On day 19, bronchoalveolar lavage (BAL) and lung tissue were collected for biochemical and immuno-histological analysis. Compared to controls treatment with BI113823 significantly reduced the numbers of BAL eosinophils, macrophages, neutrophils and lymphocytes by 58.3%, 61.1%, 66.4% and 56.0%, respectively. Mice treated with dexamethasone showed similar reductions in BAL cells. Treatment with BI113823 and dexamethasone also significantly reduced total protein content, IgE, TNF-α and IL-1ß in lavage fluid, reduced myeloperoxidase activity, mucus secretion in lung tissues, and reduced the expression of B1 receptors, matrix metalloproteinase (MMP)-2 and cyclooxygenase (COX)-2 compared to vehicle-treated mice. Only BI113823 reduced MMP-9 and inducible nitric oxide synthase (iNOS). BI113823 effectively reduced OVA-induced inflammatory cell, mediator and signaling pathways equal to or greater than that seen with steroids in a mouse asthma model. BI113823 might be useful in modulating inflammation in asthma.


Assuntos
Anti-Inflamatórios/uso terapêutico , Asma/tratamento farmacológico , Antagonistas de Receptor B1 da Bradicinina/uso terapêutico , Alérgenos , Animais , Anti-Inflamatórios/farmacologia , Asma/imunologia , Asma/metabolismo , Asma/patologia , Antagonistas de Receptor B1 da Bradicinina/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Contagem de Células , Ciclo-Oxigenase 2/imunologia , Dexametasona/farmacologia , Imunoglobulina E/imunologia , Interleucina-1beta/imunologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Metaloproteinase 2 da Matriz/imunologia , Camundongos Endogâmicos BALB C , Muco/metabolismo , Ovalbumina , Fator de Necrose Tumoral alfa/imunologia
12.
J Infect Dis ; 213(4): 532-40, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26310310

RESUMO

BACKGROUND: This study examined the therapeutic effects of an orally active nonpeptide kinin B1 receptor antagonist, BI113823, in a clinically relevant experimental model of polymicrobial sepsis in rats. METHODS: Sepsis was induced by cecal ligation and puncture (CLP). Animals received treatment with either vehicle or BI113823. The experiment was terminated in the first set of animals 15 hours after CLP. Seven-day survival following CLP was determined in the second set of animals. RESULTS: Compared with vehicle treatment, administration of BI113823 reduced neutrophil and macrophage infiltration, reduced cytokine production, attenuated intestinal mucosal hyperpermeability, prevented hemodynamic derangement, and improved cardiac output. Furthermore, administration of BI113823 reduced inducible nitric oxide synthase expression and the injury score in the lung and attenuated nuclear factor ĸB activation and apoptosis in the liver. Treatment with BI113823 also reduced plasma levels of cardiac troponin, aspartate aminotransferase, alanine aminotransferase, urea, and lactate, as well as proteinuria. Finally, administration of BI113823 improved the 7-day survival rate following CLP in rats. CONCLUSIONS: Administration of BI113823 reduced systemic and tissue inflammatory responses, prevented hemodynamic derangement, attenuated multiorgan injury, and improved overall survival.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antagonistas de Receptor B1 da Bradicinina/uso terapêutico , Inflamação/patologia , Inflamação/prevenção & controle , Sepse/tratamento farmacológico , Sepse/patologia , Animais , Modelos Animais de Doenças , Masculino , Ratos Wistar , Análise de Sobrevida , Resultado do Tratamento
13.
Crit Care Med ; 43(11): e499-507, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26468713

RESUMO

OBJECTIVES: This study was undertaken to examine the effects of BI113823, a potent small molecule orally active nonpeptide B1 receptor antagonist, in an experimental model of endotoxin-induced direct lung injury in mice and indirect lung injury and survival in cecal ligation and puncture-induced polymicrobial sepsis in rats. DESIGN: Experimental, prospective study. SETTING: University research laboratory. SUBJECTS: Male BALB/c mice and male Wistar rats. INTERVENTIONS: Series 1: acute lung injury was induced in mice by intratracheal injection of lipopolysaccharide. Mice were then randomly assigned to receive treatment of vehicle, BI113823, or dexamethasone. Bronchoalveolar lavage fluid and lung tissues were analyzed for inflammatory cell influx and various histologic variables. Series 2: sepsis was induced by cecal ligation and puncture in anesthetized rats. Animals were then randomly assigned to receive treatment of vehicle or BI113823. Experiments were terminated at 20 hours and 7 days following cecal ligation and puncture, respectively. MEASUREMENTS AND MAIN RESULTS: Series 1: treatment with BI113823 significantly reduced lipopolysaccharide-induced neutrophil influx in bronchoalveolar lavage fluid. The BI113823 group had significantly lower lung vascular permeability, lung water content, myeloperoxidase activity, lung apoptosis and lung injury scores, total protein content, and tumor necrosis factor-α and interleukin-1ß levels compared with vehicle controls. In addition, nuclear factor-κB phosphorylation, nuclear translocation, and cyclooxygenase-2 and inducible nitric oxide synthase expression in the lung were attenuated in BI113823-treated animals compared with vehicle controls. Series 2: BI113823 significantly reduced sepsis-induced macrophage recruitment, protein content, and tumor necrosis factor-α and interleukin-1ß levels in lavage fluid and also reduced lung water content and plasma levels of tumor necrosis factor-α and interleukin-6 compared with vehicle controls. Most importantly, treatment with BI113823 significantly improved survival following severe sepsis in rats. CONCLUSIONS: Administration of B1 receptor antagonist BI113823 significantly reduced endotoxin-induced direct lung injury and also reduced sepsis-induced lung inflammatory response. Most importantly, BI113823 improved survival following severe polymicrobial sepsis.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Bradicinina/antagonistas & inibidores , Citocinas/metabolismo , Dexametasona/uso terapêutico , Macrófagos/efeitos dos fármacos , Lesão Pulmonar Aguda/mortalidade , Lesão Pulmonar Aguda/fisiopatologia , Animais , Western Blotting , Bradicinina/administração & dosagem , Líquido da Lavagem Broncoalveolar/citologia , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/metabolismo , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Distribuição Aleatória , Ratos , Ratos Wistar , Valores de Referência , Taxa de Sobrevida , Resultado do Tratamento
14.
PLoS One ; 10(7): e0132810, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26225825

RESUMO

Cigarette smoking (CS) aggravates post-traumatic acute lung injury and increases ventilator-induced lung injury due to more severe tissue inflammation and apoptosis. Hyper-inflammation after chest trauma is due to the physical damage, the drop in alveolar PO2, and the consecutive hypoxemia and tissue hypoxia. Therefore, we tested the hypotheses that 1) CS exposure prior to blunt chest trauma causes more severe post-traumatic inflammation and thereby aggravates lung injury, and that 2) hyperoxia may attenuate this effect. Immediately after blast wave-induced blunt chest trauma, mice (n=32) with or without 3-4 weeks of CS exposure underwent 4 hours of pressure-controlled, thoraco-pulmonary compliance-titrated, lung-protective mechanical ventilation with air or 100% O2. Hemodynamics, lung mechanics, gas exchange, and acid-base status were measured together with blood and tissue cytokine and chemokine concentrations, heme oxygenase-1 (HO-1), activated caspase-3, and hypoxia-inducible factor 1-α (HIF-1α) expression, nuclear factor-κB (NF-κB) activation, nitrotyrosine formation, purinergic receptor 2X4 (P2XR4) and 2X7 (P2XR7) expression, and histological scoring. CS exposure prior to chest trauma lead to higher pulmonary compliance and lower PaO2 and Horovitz-index, associated with increased tissue IL-18 and blood MCP-1 concentrations, a 2-4-fold higher inflammatory cell infiltration, and more pronounced alveolar membrane thickening. This effect coincided with increased activated caspase-3, nitrotyrosine, P2XR4, and P2XR7 expression, NF-κB activation, and reduced HIF-1α expression. Hyperoxia did not further affect lung mechanics, gas exchange, pulmonary and systemic cytokine and chemokine concentrations, or histological scoring, except for some patchy alveolar edema in CS exposed mice. However, hyperoxia attenuated tissue HIF-1α, nitrotyrosine, P2XR7, and P2XR4 expression, while it increased HO-1 formation in CS exposed mice. Overall, CS exposure aggravated post-traumatic inflammation, nitrosative stress and thereby organ dysfunction and injury; short-term, lung-protective, hyperoxic mechanical ventilation have no major beneficial effect despite attenuation of nitrosative stress, possibly due to compensation of by regional alveolar hypoxia and/or consecutive hypoxemia, resulting in down-regulation of HIF-1α expression.


Assuntos
Respiração Artificial/efeitos adversos , Fumar/efeitos adversos , Traumatismos Torácicos/complicações , Traumatismos Torácicos/terapia , Ferimentos não Penetrantes/complicações , Ferimentos não Penetrantes/terapia , Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/fisiopatologia , Lesão Pulmonar Aguda/terapia , Animais , Modelos Animais de Doenças , Feminino , Hiperóxia/complicações , Hiperóxia/patologia , Hiperóxia/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Doença Pulmonar Obstrutiva Crônica/terapia , Espécies Reativas de Nitrogênio/metabolismo , Receptores Purinérgicos P2X/metabolismo , Traumatismos Torácicos/fisiopatologia , Ferimentos não Penetrantes/fisiopatologia
15.
Shock ; 43(2): 140-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25526372

RESUMO

Blunt chest trauma causes pulmonary and systemic inflammation. It is still a matter of debate whether the long-term course of this inflammatory response is associated with persistent impairment of lung function. We hypothesized that an increase of inflammatory biomarkers may still be present at later time points after blunt chest trauma, eventually, despite normalized lung mechanics and gas exchange. Anesthetized spontaneously breathing male C57BL/6J mice underwent a blast wave-induced blunt chest trauma or sham procedure. Twelve and 24 h later, blood gases and lung mechanics were measured, together with blood, bronchoalveolar lavage (BAL), and tissue cytokine concentrations (multiplex cytokine kit); heme oxygenase 1 (HO-1), activated caspase-3, Bcl-xL, and Bax expression (Western blotting); nuclear factor-κB activation (electrophoretic mobility shift assay); nitrotyrosine formation; and purinergic (P2XR4 and P2XR7) receptor expression (immunohistochemistry). Histological damage was assessed by hematoxylin and eosin and periodic acid-Schiff staining. High-resolution respirometry allowed assessing mitochondrial respiration in diaphragm biopsies. Chest trauma significantly increased tissue and BAL cytokine levels, associated with a significant increase in HO-1, purinergic receptor expression, and tissue nitrotyrosine formation. In contrast, lung mechanics, gas exchange, and histological damage did not show any significant difference between sham and trauma groups. Activation of the immune response remains present at later time points after murine blunt chest trauma. Discordance of the increased local inflammatory response and preserved pulmonary function may be explained by a dissociation of the immune response and lung function, such as previously suggested after experimental sepsis.


Assuntos
Traumatismos Torácicos/fisiopatologia , Ferimentos não Penetrantes/fisiopatologia , Animais , Apoptose/fisiologia , Respiração Celular/fisiologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Pulmão/imunologia , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Mitocôndrias Musculares/fisiologia , Troca Gasosa Pulmonar/fisiologia , Receptores Purinérgicos/biossíntese , Mecânica Respiratória/fisiologia , Traumatismos Torácicos/imunologia , Traumatismos Torácicos/patologia , Tirosina/análogos & derivados , Tirosina/biossíntese , Ferimentos não Penetrantes/imunologia , Ferimentos não Penetrantes/patologia
16.
Antioxid Redox Signal ; 17(10): 1393-406, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22578329

RESUMO

AIMS: Prostaglandin endoperoxide H(2) synthase (PGHS) is a well-known target for peroxynitrite-mediated nitration. In several experimental macrophage models, however, the relatively late onset of nitration failed to coincide with the early peak of endogenous peroxynitrite formation. In the present work, we aimed to identify an alternative, peroxynitrite-independent mechanism, responsible for the observed nitration and inactivation of PGHS-2 in an inflammatory cell model. RESULTS: In primary rat alveolar macrophages stimulated with lipopolysaccharide (LPS), PGHS-2 activity was suppressed after 12 h, although the prostaglandin endoperoxide H(2) synthase (PGHS-2) protein was still present. This coincided with a nitration of the enzyme. Coincubation with a nitric oxide synthase-2 (NOS-2) inhibitor preserved PGHS-2 nitration and at the same time restored thromboxane A(2) (TxA(2)) synthesis in the cells. Formation of reactive oxygen species (ROS) was maximal at 4 h and then returned to baseline levels. Nitrite (NO(2)(-)) production occurred later than ROS generation. This rendered generation of peroxynitrite and the nitration of PGHS-2 unlikely. We found that the nitrating agent was formed from NO(2)(-), independent from superoxide ((•)O(2)(-)). Purified PGHS-2 treated with NO(2)(-) was selectively nitrated on the active site Tyr(371), as identified by mass spectrometry (MS). Exposure to peroxynitrite resulted in the nitration not only of Tyr(371), but also of other tyrosines (Tyr). INNOVATION AND CONCLUSION: The data presented here point to an autocatalytic nitration of PGHS-2 by NO(2)(-), catalyzed by the enzyme's endogenous peroxidase activity and indicate a potential involvement of this mechanism in the termination of prostanoid formation under inflammatory conditions.


Assuntos
Macrófagos Alveolares/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Prostaglandinas/metabolismo , Animais , Ratos , Espécies Reativas de Oxigênio/metabolismo
17.
Exp Cell Res ; 317(1): 42-50, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20858487

RESUMO

The question whether epidermal growth factor (EGF)-induced receptor endocytosis requires the prior autophosphorylation via the EGF receptor (EGFR) kinase domain has been a matter of long-standing debate. In the airway epithelial cell line NCI-H292, the EGFR kinase domain inhibitor BIBW 2948 BS was found to inhibit both autophosphorylation and subsequent internalization of the endogenous EGFR with similar IC50 values. Applying an ex vivo EGFR internalization assay in a clinical study, the in vivo effect of inhalatively administered BIBW 2948 BS was determined directly at the targeted receptor in airway tissues from COPD patients. In these experiments, the in vivo inhibition of the EGFR kinase domain prevented the EGF-induced internalization of EGFR.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Biópsia , Técnicas de Cultura de Células , Células Cultivadas , Método Duplo-Cego , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Masculino , Fosforilação/efeitos dos fármacos , Placebos , Inibidores de Proteínas Quinases/uso terapêutico , Transporte Proteico/efeitos dos fármacos , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
18.
Biochem Biophys Res Commun ; 334(1): 254-62, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15993846

RESUMO

TARC (CCL17) and MDC (CCL22) are well-known chemoattractants for Th2 cells. Here, we evaluated the role of both chemokines for cigarette smoke-induced airway inflammation. The expression profiles of MDC, TARC, and their receptor CCR4 were analyzed in models of acute and chronic cigarette smoke-induced airway inflammation that is characterized by a Th1 immune response. The results were compared to the expression of both chemokines in models of idiopathic pulmonary fibrosis and acute asthma, which are associated with a Th2 immune response. The expression of MDC and TARC was found to be elevated in all lung inflammation models. In contrast to the findings in the asthma and lung fibrosis models, the increased expression of MDC and TARC in the cigarette-smoke model was not associated with an increased infiltration of Th2 cells into smoke-treated lungs. Our data indicate that instead of Th2 cells, airway epithelial cells expressing CCR4 might be the principal targets for MDC and TARC released from alveolar macrophages during cigarette smoke-induced airway inflammation.


Assuntos
Brônquios/imunologia , Quimiocinas CC/imunologia , Macrófagos/imunologia , Pneumonia/etiologia , Pneumonia/imunologia , Mucosa Respiratória/imunologia , Fumar/efeitos adversos , Compostos de Alúmen , Animais , Asma/etiologia , Asma/imunologia , Bleomicina , Brônquios/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL22 , Doença Crônica , Modelos Animais de Doenças , Fibrose/etiologia , Fibrose/imunologia , Regulação da Expressão Gênica/imunologia , Macrófagos/efeitos dos fármacos , Masculino , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Mucosa Respiratória/efeitos dos fármacos , Alcatrões/toxicidade
19.
Eur J Pharmacol ; 491(2-3): 191-4, 2004 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-15140636

RESUMO

Substance P is speculated to be a key mediator of itching in atopic dermatitis, possibly acting via the tachykinin NK1 receptor. Thus, we examined the effect of a tachykinin NK1 antagonist, BIIF 1149 CL, on scratching behaviour in a picrylchloride-induced dermatitis model in NC/Nga mice. BIIF 1149 CL ((S)-N-[2-[3,5-bis(trifluoromethyl) phenyl]ethyl]-4-(cyclopropylmethyl)-N-methyl-alpha-phenyl-1-piperazineacetamide, monohydrochloride, monohydrate) at a dose of 100 mg/kg, p.o., significantly inhibited scratching behaviour immediately after administration, and the effect continued up to 6 h. The results suggest that clinical trials of tachykinin NK1 antagonists for the treatment of itching in atopic dermatitis patients would be warranted.


Assuntos
Comportamento Animal/efeitos dos fármacos , Dermatite Atópica/tratamento farmacológico , Modelos Animais de Doenças , Prurido/tratamento farmacológico , Substância P/antagonistas & inibidores , Animais , Dermatite Atópica/induzido quimicamente , Masculino , Camundongos , Antagonistas dos Receptores de Neurocinina-1 , Prurido/induzido quimicamente , Receptores da Neurocinina-1/fisiologia , Substância P/toxicidade , Taquicininas/farmacologia , Taquicininas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA