Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Clin Pharmacol Drug Dev ; 13(6): 665-671, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38523487

RESUMO

Tozorakimab is a high-affinity human immunoglobulin G1 monoclonal antibody that neutralizes interleukin (IL)-33, an IL-1 family cytokine. This phase 1, single-center, randomized, double-blind, placebo-controlled, single ascending dose study (NCT05070312) evaluated tozorakimab in a healthy Chinese population. Outcomes included the characterization of the pharmacokinetic (PK) profile and immunogenicity of tozorakimab. Safety outcomes included treatment-emergent adverse events (TEAEs) and clinical laboratory, electrocardiogram, and vital sign parameters. Healthy, non-smoking, male, and female Chinese participants aged 18-45 years with a body mass index 19-24 kg/m2 were enrolled. In total, 36 participants across 2 cohorts of 18 participants were randomized 2:1 to receive a single subcutaneous dose of tozorakimab (300 mg [2 mL] or 600 mg [4 mL]) or matching placebo (2 or 4 mL). Tozorakimab showed dose-dependent serum PK concentrations with an approximate monophasic distribution in serum over time and a maximum observed peak concentration of 20.1 and 33.7 µg/mL in the 300- and 600-mg cohorts, respectively. No treatment-emergent anti-drug antibodies for tozorakimab were observed in any of the participants. There were no clinically relevant trends in the occurrence of TEAEs across the treatment groups. There were no clinically relevant trends over time in clinical laboratory (hematology, clinical chemistry, and urinalysis), electrocardiogram, or vital sign parameters in any treatment group. Overall, tozorakimab demonstrated dose-dependent systemic exposure in healthy Chinese participants and was well tolerated, with no safety concerns identified in this study.


Assuntos
Anticorpos Monoclonais Humanizados , Povo Asiático , Relação Dose-Resposta a Droga , Voluntários Saudáveis , Humanos , Método Duplo-Cego , Feminino , Masculino , Adulto , Injeções Subcutâneas , Adulto Jovem , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Pessoa de Meia-Idade , Adolescente , China , População do Leste Asiático
2.
Clin Pharmacol Ther ; 115(3): 565-575, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38115209

RESUMO

Tozorakimab is a human monoclonal antibody that neutralizes interleukin (IL)-33. IL-33 is a broad-acting epithelial "alarmin" cytokine upregulated in lung tissue of patients with chronic obstructive pulmonary disease (COPD). This first-in-human, phase I, randomized, double-blind, placebo-controlled study (NCT03096795) evaluated the safety, tolerability, pharmacokinetics (PKs), immunogenicity, target engagement, and pharmacodynamics (PDs) of tozorakimab. This was a 3-part study. In part 1, 56 healthy participants with a history of mild atopy received single escalating doses of either intravenous or subcutaneous tozorakimab or placebo. In part 2, 24 patients with mild COPD received multiple ascending doses of subcutaneous tozorakimab or placebo. In part 3, 8 healthy Japanese participants received a single intravenous dose of tozorakimab or placebo. The safety data collected included treatment-emergent adverse events (TEAEs), vital signs, and clinical laboratory parameters. Biological samples for PKs, immunogenicity, target engagement, and PD biomarker analyses were collected. No meaningful differences in the frequencies of TEAEs were observed between the tozorakimab and placebo arms. Three tozorakimab-treated participants with COPD experienced treatment-emergent serious adverse events. Subcutaneous or intravenous tozorakimab demonstrated linear, time-independent PKs with a mean half-life of 11.7-17.3 days. Treatment-emergent anti-drug antibody frequency was low. Engagement of tozorakimab with endogenous IL-33 in serum and nasal airways was demonstrated. Tozorakimab significantly reduced serum IL-5 and IL-13 levels in patients with COPD compared with placebo. Overall, tozorakimab was well tolerated, with a linear, time-independent serum PK profile. Additionally, biomarker studies demonstrated proof of mechanism. Overall, these data support the further clinical development of tozorakimab in COPD and other inflammatory diseases.


Assuntos
Interleucina-33 , Doença Pulmonar Obstrutiva Crônica , Adulto , Humanos , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Anticorpos Monoclonais/efeitos adversos , Citocinas , Método Duplo-Cego , Biomarcadores , Voluntários Saudáveis
3.
ERJ Open Res ; 9(5)2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37868151

RESUMO

Background: Increased serum interleukin (IL)-33 predicts poor outcomes in patients hospitalised with coronavirus disease 2019 (COVID-19). We examined the efficacy and safety of tozorakimab, a monoclonal antibody that neutralises IL-33, in improving outcomes in ACCORD-2 (EudraCT: 2020-001736-95). Methods: ACCORD-2 was an open-label, phase 2a study in adults hospitalised with COVID-19. Patients were randomised 1:1 to tozorakimab 300 mg plus standard of care (SoC) or SoC alone. The primary end-point was time to clinical response (sustained clinical improvement of ≥2 points on the World Health Organization ordinal scale, discharge from hospital or fit for discharge) by day 29. Other end-points included death or respiratory failure, mortality and intensive care unit admission by day 29, and safety. Serum IL-33/soluble stimulated-2 (sST2) complex levels were measured by high-sensitivity immunoassay. Results: Efficacy analyses included 97 patients (tozorakimab+SoC, n=53; SoC, n=44). Median time to clinical response did not differ between the tozorakimab and SoC arms (8.0 and 9.5 days, respectively; HR 0.96, 80% CI 0.70-1.31; one-sided p=0.33). Tozorakimab was well tolerated and the OR for risk of death or respiratory failure with treatment versus SoC was 0.55 (80% CI 0.27-1.12; p=0.26), while the OR was 0.31 (80% CI 0.09-1.06) in patents with high baseline serum IL-33/sST2 complex levels. Conclusions: Overall, ACCORD-2 results suggest that tozorakimab could be a novel therapy for patients hospitalised with COVID-19, warranting further investigation in confirmatory phase 3 studies.

4.
Sci Transl Med ; 14(671): eabl5849, 2022 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-36383685

RESUMO

Acute cardiorespiratory breathlessness accounts for one in eight of all emergency hospitalizations. Early, noninvasive diagnostic testing is a clinical priority that allows rapid triage and treatment. Here, we sought to find and replicate diagnostic breath volatile organic compound (VOC) biomarkers of acute cardiorespiratory disease and understand breath metabolite network enrichment in acute disease, with a view to gaining mechanistic insight of breath biochemical derangements. We collected and analyzed exhaled breath samples from 277 participants presenting acute cardiorespiratory exacerbations and aged-matched healthy volunteers. Topological data analysis phenotypes differentiated acute disease from health and acute cardiorespiratory exacerbation subtypes (acute heart failure, acute asthma, acute chronic obstructive pulmonary disease, and community-acquired pneumonia). A multibiomarker score (101 breath biomarkers) demonstrated good diagnostic sensitivity and specificity (≥80%) in both discovery and replication sets and was associated with all-cause mortality at 2 years. In addition, VOC biomarker scores differentiated metabolic subgroups of cardiorespiratory exacerbation. Louvain clustering of VOCs coupled with metabolite enrichment and similarity assessment revealed highly specific enrichment patterns in all acute disease subgroups, for example, selective enrichment of correlated C5-7 hydrocarbons and C3-5 carbonyls in heart failure and selective depletion of correlated aldehydes in acute asthma. This study identified breath VOCs that differentiate acute cardiorespiratory exacerbations and associated subtypes and metabolic clusters of disease-associated VOCs.


Assuntos
Asma , Insuficiência Cardíaca , Compostos Orgânicos Voláteis , Humanos , Testes Respiratórios , Compostos Orgânicos Voláteis/análise , Doença Aguda , Dispneia/diagnóstico , Asma/diagnóstico , Biomarcadores/metabolismo , Insuficiência Cardíaca/diagnóstico
5.
Pharmacol Res Perspect ; 10(5): e01004, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36036654

RESUMO

Altered physiology caused by critical illness may change midazolam pharmacokinetics and thereby result in adverse reactions and outcomes in this vulnerable patient population. This study set out to determine which critical illness-related factors impact midazolam pharmacokinetics in children using population modeling. This was an observational, prospective, controlled study of children receiving IV midazolam as part of routine care. Children recruited into the study were either critically-ill receiving continuous infusions of midazolam or otherwise well, admitted for elective day-case surgery (control) who received a single IV bolus dose of midazolam. The primary outcome was to determine the population pharmacokinetics and identify covariates that influence midazolam disposition during critical illness. Thirty-five patients were recruited into the critically ill arm of the study, and 54 children into the control arm. Blood samples for assessing midazolam and 1-OH-midazolam concentrations were collected opportunistically (critically ill arm) and in pre-set time windows (control arm). Pharmacokinetic modeling demonstrated a significant change in midazolam clearance with acute inflammation (measured using C-Reactive Protein), cardio-vascular status, and weight. Simulations predict that elevated C-Reactive Protein and compromised cardiovascular function in critically ill children result in midazolam concentrations up to 10-fold higher than in healthy children. The extremely high concentrations of midazolam observed in some critically-ill children indicate that the current therapeutic dosing regimen for midazolam can lead to over-dosing. Clinicians should be aware of this risk and intensify monitoring for oversedation in such patients.


Assuntos
Estado Terminal , Midazolam , Proteína C-Reativa , Criança , Humanos , Inflamação/tratamento farmacológico , Estudos Prospectivos
6.
PLoS One ; 15(11): e0240964, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33166307

RESUMO

BACKGROUND: Allergic asthma is a heterogenous disorder predominantly driven by a type 2 inflammatory response to aeroallergens. Therapeutic modulation to rebalance these type 2 responses may offer clinical benefit for allergic respiratory inflammatory diseases, with the potential for disease modification. GSK2245035, a selective toll-like receptor-7 agonist, preferentially stimulates the induction of type 1 interferon alpha, reducing type 2 responses. OBJECTIVE: This study investigated whether intranasal GSK2245035 reduced allergen-induced bronchial reactivity in mild allergic asthma. METHODS: This double-blind, placebo-controlled, parallel-group Phase IIa trial randomized (1:1) participants with mild allergic asthma to intranasal GSK2245035 20 ng or placebo once weekly for 8 weeks; follow-up was conducted 1, 4, and 12 weeks after treatment. Allergen-induced late asthmatic response 1 week after treatment was measured as minimum and weighted mean forced expiratory volume in 1 second (FEV1) 4-10 hours following bronchial allergen challenge (primary endpoint). Pharmacodynamic and allergic biomarkers, and adverse events, were assessed. A Bayesian analysis framework was used; a posterior probability >0.7 denoted primary endpoint success. RESULTS: Thirty-six participants were randomized (GSK2245035, n = 22; placebo, n = 14). The percentage attenuation in late asthmatic response was -4.6% (posterior probability: 0.385) and -10.5% (posterior probability: 0.303) for minimum and weighted mean FEV1, respectively. Type 2 responses were confirmed by changes in lung function, eosinophils (blood and sputum), interleukin-5 (sputum) and fractional exhaled nitric oxide biomarkers pre- and post-bronchial allergen challenge. However, no treatment effect was observed. Adverse events were reported by 10/14 (71%) and 21/22 (95%) participants in the placebo and GSK2245035 groups, respectively; headache was the most common. CONCLUSIONS AND CLINICAL RELEVANCE: Although target engagement was observed, weekly intranasal GSK2245035 20 ng for 8 weeks did not substantially attenuate the late asthmatic response in participants with mild allergic asthma. Overall, treatment was well tolerated.


Assuntos
Adenina/análogos & derivados , Asma/tratamento farmacológico , Piperidinas/administração & dosagem , Receptor 7 Toll-Like/agonistas , Adenina/administração & dosagem , Adenina/efeitos adversos , Administração Intranasal , Adulto , Alérgenos/administração & dosagem , Antiasmáticos/administração & dosagem , Antiasmáticos/efeitos adversos , Asma/imunologia , Asma/fisiopatologia , Testes de Provocação Brônquica , Método Duplo-Cego , Feminino , Volume Expiratório Forçado/efeitos dos fármacos , Humanos , Interferon-alfa/biossíntese , Masculino , Pessoa de Meia-Idade , Piperidinas/efeitos adversos , Estudo de Prova de Conceito , Rinite Alérgica/tratamento farmacológico , Rinite Alérgica/imunologia , Rinite Alérgica/fisiopatologia , Adulto Jovem
7.
Bioanalysis ; 11(19): 1737-1754, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31617393

RESUMO

Aim: Collection and quantitative analysis in dry blood using volumetric absorptive microsampling (VAMS™) potentially offers significant advantages over conventional wet whole blood analysis. This manuscript explores their use for pediatric sampling and explores additional considerations for the validation of the bioanalytical method. Results: HPLC-MS/MS methods for the determination of midazolam and its major metabolite 1-OH midazolam in both whole wet blood, and dry blood collected on VAMS were developed, validated, and used to support an observational clinical study to compare pharmacokinetic parameters in pediatric patients. Conclusion: Validation data met internationally accepted guideline criteria. A strong correlation was observed in calculated concentrations between wet and dry test samples, indicating that VAMS is a suitable technique for use in pediatric clinical studies.


Assuntos
Coleta de Amostras Sanguíneas/métodos , Teste em Amostras de Sangue Seco/métodos , Hipnóticos e Sedativos/sangue , Midazolam/sangue , Adulto , Criança , Cromatografia Líquida de Alta Pressão/métodos , Humanos , Limite de Detecção , Espectrometria de Massas em Tandem/métodos
8.
BMJ Open ; 9(3): e025486, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30852546

RESUMO

INTRODUCTION: Patients presenting with acute undifferentiated breathlessness are commonly encountered in admissions units across the UK. Existing blood biomarkers have clinical utility in distinguishing patients with single organ pathologies but have poor discriminatory power in multifactorial presentations. Evaluation of volatile organic compounds (VOCs) in exhaled breath offers the potential to develop biomarkers of disease states that underpin acute cardiorespiratory breathlessness, owing to their proximity to the cardiorespiratory system. To date, there has been no systematic evaluation of VOC in acute cardiorespiratory breathlessness. The proposed study will seek to use both offline and online VOC technologies to evaluate the predictive value of VOC in identifying common conditions that present with acute cardiorespiratory breathlessness. METHODS AND ANALYSIS: A prospective real-world observational study carried out across three acute admissions units within Leicestershire. Participants with self-reported acute breathlessness, with a confirmed primary diagnosis of either acute heart failure, community-acquired pneumonia and acute exacerbation of asthma or chronic obstructive pulmonary disease will be recruited within 24 hours of admission. Additionally, school-age children admitted with severe asthma will be evaluated. All participants will undergo breath sampling on admission and on recovery following discharge. A range of online technologies including: proton transfer reaction mass spectrometry, gas chromatography ion mobility spectrometry, atmospheric pressure chemical ionisation-mass spectrometry and offline technologies including gas chromatography mass spectroscopy and comprehensive two-dimensional gas chromatography-mass spectrometry will be used for VOC discovery and replication. For offline technologies, a standardised CE-marked breath sampling device (ReCIVA) will be used. All recruited participants will be characterised using existing blood biomarkers including C reactive protein, brain-derived natriuretic peptide, troponin-I and blood eosinophil levels and further evaluated using a range of standardised questionnaires, lung function testing, sputum cell counts and other diagnostic tests pertinent to acute disease. ETHICS AND DISSEMINATION: The National Research Ethics Service Committee East Midlands has approved the study protocol (REC number: 16/LO/1747). Integrated Research Approval System (IRAS) 198921. Findings will be presented at academic conferences and published in peer-reviewed scientific journals. Dissemination will be facilitated via a partnership with the East Midlands Academic Health Sciences Network and via interaction with all UK-funded Medical Research Council and Engineering and Physical Sciences Research Council molecular pathology nodes. TRIAL REGISTRATION NUMBER: NCT03672994.


Assuntos
Doenças Cardiovasculares/diagnóstico , Dispneia/diagnóstico , Estudos Multicêntricos como Assunto/métodos , Estudos Observacionais como Assunto/métodos , Compostos Orgânicos Voláteis/análise , Doença Aguda , Adulto , Testes Respiratórios , Coleta de Dados , Diagnóstico Diferencial , Expiração , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Estudos Prospectivos , Doenças Respiratórias/diagnóstico , Tamanho da Amostra , Escarro
9.
J Asthma ; 55(11): 1205-1213, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29211599

RESUMO

OBJECTIVES: A novel gas chromatography-mass spectrometry (GC-MS) method has been developed to quantify salbutamol in micro-volumes (10 µL) of blood. A potential application is paediatric therapeutic dose monitoring (TDM) in acute severe asthma. METHODS: At presentation, the children receive multiple doses of salbutamol (inhaled, nebulised and occasionally intravenous) but it is difficult to distinguish children who do not respond to treatment because of inadequate concentrations from those with toxicity, as symptoms are similar. A comparison was made between traditional dried blood spots (DBS) and the newly developed technique volumetric absorptive micro-sampling (VAMS), with specific investigation into the effect of drying time on analyte recovery. RESULTS: For both sampling techniques, the final assay demonstrated good precision and accuracy across the concentration range tested (3-100 ng/mL), including both the normal therapeutic and toxic range. The method was developed to comply with FDA guidelines with precision and accuracy ≤15% for all concentrations, except the limit of quantification (5 ng/mL) where they were ≤20%. VAMS offered advantages in sampling ease and reduced GC-MS interference. The assay was successfully applied to the quantification of blood salbutamol concentrations in three healthy volunteers dosed with 1 mg salbutamol by inhalation. CONCLUSIONS: This demonstrated its potential for use in paediatric TDM studies, where in the acute situation considerably higher doses of salbutamol will have been administered. This is the first time that a TDM method for salbutamol has been carried out using VAMS and offers all the advantages provided by DBS, whilst eliminating the inherent sampling volume inaccuracies of traditional DBS collection.


Assuntos
Albuterol/administração & dosagem , Albuterol/uso terapêutico , Asma/tratamento farmacológico , Monitoramento de Medicamentos/métodos , Doença Aguda , Administração por Inalação , Coleta de Amostras Sanguíneas , Criança , Relação Dose-Resposta a Droga , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Índice de Gravidade de Doença
11.
Pediatr Res ; 81(2): 376-383, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27925619

RESUMO

BACKGROUND: Premature infants are at increased risk for airway diseases, such as wheezing and asthma, because of early exposure to risk factors including hyperoxia. As in adult asthma, airway remodeling and increased extracellular matrix (ECM) deposition is involved. METHODS: We assessed the impact of 24-72 h of moderate hyperoxia (50%) on human fetal airway smooth muscle (fASM) ECM deposition through western blot, modified in-cell western, and zymography techniques. RESULTS: Hyperoxia exposure significantly increased collagen I and collagen III deposition, increased pro- and cleaved matrix metalloproteinase 9 (MMP9) activity, and decreased endogenous MMP inhibitor, TIMP1, expression. Hyperoxia-induced change in caveolin-1 (CAV1) expression was assessed as a potential mechanism for the changes in ECM deposition. CAV1 expression was decreased following hyperoxia. Supplementation of CAV1 activity with caveolar scaffolding domain (CSD) peptide abrogated the hyperoxia-mediated ECM changes. CONCLUSION: These results demonstrate that moderate hyperoxia enhances ECM deposition in developing airways by altering the balance between MMPs and their inhibitors (TIMPs), and by increasing collagen deposition. These effects are partly mediated by a hyperoxia-induced decrease in CAV1 expression. In conjunction with prior data demonstrating increased fASM proliferation with hyperoxia, these data further demonstrate that hyperoxia is an important instigator of remodeling in developing airways.


Assuntos
Matriz Extracelular/metabolismo , Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Sistema Respiratório/embriologia , Remodelação das Vias Aéreas , Asma/terapia , Caveolina 1/metabolismo , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Fenótipo , RNA Interferente Pequeno/metabolismo , Sistema Respiratório/efeitos dos fármacos , Fatores de Risco , Transdução de Sinais , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/metabolismo
12.
Bioanalysis ; 8(11): 1173-82, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27212021

RESUMO

AIM: Excipients are used to overcome the chemical, physical and microbiological challenges posed by developing formulated medicines. Both methyl and propyl paraben are commonly used in pediatric liquid formulations. There is no data on systemic exposure to parabens in neonates. The European Study of Neonatal Exposure to Excipients project has investigated this. Results & methodology: DBS sampling was used to collect opportunistic blood samples. Parabens were extracted from the DBS and analyzed using a validated LC-MS/MS assay. DISCUSSION & CONCLUSION: The above assay was applied to analyze neonatal DBS samples. The blood concentrations of parabens in neonates confirm systemic exposure to parabens following administration of routine medicines.


Assuntos
Teste em Amostras de Sangue Seco/métodos , Parabenos/análise , Conservantes Farmacêuticos/análise , Espectrometria de Massas em Tandem/métodos , Cromatografia Líquida/métodos , Humanos , Recém-Nascido , Limite de Detecção , Extração em Fase Sólida/métodos
13.
Am J Physiol Lung Cell Mol Physiol ; 310(2): L202-11, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26589477

RESUMO

Viral infections, such as respiratory syncytial virus and rhinovirus, adversely affect neonatal and pediatric populations, resulting in significant lung morbidity, including acute asthma exacerbation. Studies in adults have demonstrated that human airway smooth muscle (ASM) cells modulate inflammation through their ability to secrete inflammatory cytokines and chemokines. The role of ASM in the developing airway during infection remains undefined. In our study, we used human fetal ASM cells as an in vitro model to examine the effect of Toll-like receptor (TLR) agonists on chemokine secretion. We found that fetal ASM express multiple TLRs, including TLR3 and TLR4, which are implicated in the pathogenesis of respiratory syncytial virus and rhinovirus infection. Cells were treated with TLR agonists, polyinosinic-polycytidylic acid [poly(I:C)] (TLR3 agonist), lipopolysaccharide (TLR4 agonist), or R848 (TLR7/8 agonist), and IL-8 and chemokine (C-C motif) ligand 5 (CCL5) secretion were evaluated. Interestingly, poly(I:C), but neither lipopolysaccharide nor R848, increased IL-8 and chemokine (C-C motif) ligand 5 secretion. Examination of signaling pathways suggested that the poly(I:C) effects in fetal ASM involve TLR and ERK signaling, in addition to another major inflammatory pathway, NF-κB. Moreover, there are variations between fetal and adult ASM with respect to poly(I:C) effects on signaling pathways. Pharmacological inhibition suggested that ERK pathways mediate poly(I:C) effects. Overall, our data show that poly(I:C) initiates activation of proinflammatory pathways in developing ASM, which may contribute to immune responses to infection and exacerbation of asthma.


Assuntos
Quimiocinas/metabolismo , Pulmão/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor 3 Toll-Like/metabolismo , Células Cultivadas , Citocinas/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Pediatr Res ; 78(6): 650-6, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26331770

RESUMO

BACKGROUND: Adult human airway smooth muscle (ASM) produce cytokines involved in recruitment and survival of leukocytes within airway walls. Cytokine generation by adult ASM is glucocorticoid-sensitive. Whether developing lung ASM produces cytokines in a glucocorticoid-sensitive fashion is unknown. METHODS: Cultured fetal human ASM cells stimulated with TNF-α (0-20 ng/ml) were incubated with TNF-α receptor-blocking antibodies, fluticasone (1 and 100 nm), or vehicle. Supernatants and cells were assayed for the production of CCL5, CXCL10, and CXCL8 mRNA and protein and glucocorticoid receptor phosphorylation. RESULTS: CCL5, CXCL10, and CXCL8 mRNA and protein production by fetal ASM cell was significantly and dose-dependently following TNF-α treatment. Cytokine mRNA and protein production were effectively blocked by TNF-α R1 and R2 receptor neutralizing antibodies but variably inhibited by fluticasone. TNF-α-induced TNF-R1 and R2 receptor mRNA expression was only partially attenuated by fluticasone. Glucocorticoid receptor phosphorylation at serine (Ser) 211 but not at Ser 226 was enhanced by fluticasone. CONCLUSION: Production of CCL5, CXCL10, and CXCL8 by fetal ASM appears to involve pathways that are both qualitatively and mechanistically distinct to those described for adult ASM. The findings imply developing ASM has potential to recruit leukocyte into airways and, therefore, of relevance to childhood airway diseases.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Citocinas/metabolismo , Fluticasona/farmacologia , Glucocorticoides/farmacologia , Pulmão/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Anticorpos/farmacologia , Células Cultivadas , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Citocinas/imunologia , Relação Dose-Resposta a Droga , Idade Gestacional , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Pulmão/embriologia , Pulmão/imunologia , Pulmão/metabolismo , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/metabolismo , Fosforilação , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/efeitos dos fármacos , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Serina , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
15.
Biochim Biophys Acta ; 1853(10 Pt A): 2506-14, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26112987

RESUMO

Moderate hyperoxic exposure in preterm infants contributes to subsequent airway dysfunction and to risk of developing recurrent wheeze and asthma. The regulatory mechanisms that can contribute to hyperoxia-induced airway dysfunction are still under investigation. Recent studies in mice show that hyperoxia increases brain-derived neurotrophic factor (BDNF), a growth factor that increases airway smooth muscle (ASM) proliferation and contractility. We assessed the mechanisms underlying effects of moderate hyperoxia (50% O2) on BDNF expression and secretion in developing human ASM. Hyperoxia increased BDNF secretion, but did not alter endogenous BDNF mRNA or intracellular protein levels. Exposure to hyperoxia significantly increased [Ca2+]i responses to histamine, an effect blunted by the BDNF chelator TrkB-Fc. Hyperoxia also increased ASM cAMP levels, associated with reduced PDE4 activity, but did not alter protein kinase A (PKA) activity or adenylyl cyclase mRNA levels. However, 50% O2 increased expression of Epac2, which is activated by cAMP and can regulate protein secretion. Silencing RNA studies indicated that Epac2, but not Epac1, is important for hyperoxia-induced BDNF secretion, while PKA inhibition did not influence BDNF secretion. In turn, BDNF had autocrine effects of enhancing ASM cAMP levels, an effect inhibited by TrkB and BDNF siRNAs. Together, these novel studies suggest that hyperoxia can modulate BDNF secretion, via cAMP-mediated Epac2 activation in ASM, resulting in a positive feedback effect of BDNF-mediated elevation in cAMP levels. The potential functional role of this pathway is to sustain BDNF secretion following hyperoxic stimulus, leading to enhanced ASM contractility and proliferation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Brônquios/metabolismo , AMP Cíclico/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Traqueia/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Brônquios/patologia , Sinalização do Cálcio/genética , Células Cultivadas , AMP Cíclico/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Hiperóxia/genética , Hiperóxia/metabolismo , Hiperóxia/patologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Músculo Liso/patologia , Miócitos de Músculo Liso/patologia , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Receptor trkB , Traqueia/patologia
16.
J Infect Dis ; 211(10): 1677-85, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25429101

RESUMO

Impaired immunity and tissue hypoxia-ischemia are strongly linked with Streptococcus pneumoniae pathogenesis in patients with sickle cell anemia. Perfluorocarbon emulsions (PFCEs) have high O2-dissolving capacity and can alleviate tissue hypoxia. Here, we evaluate the effects of intravenous PFCE therapy in transgenic sickle cell (HbSS) mice infected with S. pneumoniae. HbSS and C57BL/6 (control) mice intravenously infected with S. pneumoniae were treated intravenously with PFCE or phosphate-buffered saline (PBS) and then managed in either air/O2 (FiO2 proportion, 50%; hereafter referred to as the PFCE-O2 and PBS-O2 groups) or air only (hereafter, the PFCE-air and PBS-air groups) gas mixtures. Lungs were processed for leukocyte and bacterial counts and cytokine measurements. HbSS mice developed severe pneumococcal infection significantly faster than C57BL/6 mice (Kaplan-Maier analysis, P < .05). PFCE-O2-treated HbSS mice had significantly better survival at 72 hours than HBSS mice treated with PFCE-air, PBS-O2, or PBS-air (P < .05). PFCE-O2-treated HbSS mice also had significantly lower pulmonary leukocyte counts, lower interleukin 1ß and interferon γ levels, and higher interleukin 10 levels than PFCE-air-treated HbSS mice. Clearance of S. pneumoniae from lungs of HbSS mice or C57BL/6 mice was not altered by PFCE treatment. Improved survival of PFCE-O2-treated HbSS mice infected with S. pneumoniae is associated with altered pulmonary inflammation but not enhanced bacterial clearance.


Assuntos
Anemia Falciforme/complicações , Emulsões/uso terapêutico , Fluorocarbonos/uso terapêutico , Oxigenoterapia Hiperbárica , Infecções Pneumocócicas/tratamento farmacológico , Administração Intravenosa , Animais , Carga Bacteriana , Citocinas/análise , Modelos Animais de Doenças , Leucócitos/imunologia , Pulmão/patologia , Camundongos Endogâmicos C57BL , Streptococcus pneumoniae/isolamento & purificação , Análise de Sobrevida
17.
J Cell Physiol ; 230(6): 1189-98, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25204635

RESUMO

Asthma in the pediatric population remains a significant contributor to morbidity and increasing healthcare costs. Vitamin D3 insufficiency and deficiency have been associated with development of asthma. Recent studies in models of adult airway diseases suggest that the bioactive Vitamin D3 metabolite, calcitriol (1,25-dihydroxyvitamin D3 ; 1,25(OH)2 D3 ), modulates responses to inflammation; however, this concept has not been explored in developing airways in the context of pediatric asthma. We used human fetal airway smooth muscle (ASM) cells as a model of the early postnatal airway to explore how calcitriol modulates remodeling induced by pro-inflammatory cytokines. Cells were pre-treated with calcitriol and then exposed to TNFα or TGFß for up to 72 h. Matrix metalloproteinase (MMP) activity, production of extracellular matrix (ECM), and cell proliferation were assessed. Calcitriol attenuated TNFα enhancement of MMP-9 expression and activity. Additionally, calcitriol attenuated TNFα and TGFß-induced collagen III expression and deposition, and separately, inhibited proliferation of fetal ASM cells induced by either inflammatory mediator. Analysis of signaling pathways suggested that calcitriol effects in fetal ASM involve ERK signaling, but not other major inflammatory pathways. Overall, our data demonstrate that calcitriol can blunt multiple effects of TNFα and TGFß in developing airway, and point to a potentially novel approach to alleviating structural changes in inflammatory airway diseases of childhood.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Asma/metabolismo , Calcitriol/deficiência , Citocinas/metabolismo , Miócitos de Músculo Liso/metabolismo , Deficiência de Vitamina D/metabolismo , Proliferação de Células/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Músculo Liso/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L141-6, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25399436

RESUMO

Supplemental oxygen, used to treat hypoxia in preterm and term neonates, increases the risk of neonatal lung diseases, such as bronchopulmonary dysplasia (BPD) and asthma. There is a known sex predilection for BPD, but the underlying mechanisms are not clear. We tested the hypothesis that altered, local estradiol following hyperoxia contributes to pathophysiological changes observed in immature lung. In human fetal airway smooth muscle (fASM) cells exposed to normoxia or hyperoxia, we measured the expression of proteins involved in estrogen metabolism and cell proliferation responses to estradiol. In fASM cells, CYP1a1 expression was increased by hyperoxia, whereas hyperoxia-induced enhancement of cell proliferation was blunted by estradiol. Pharmacological studies indicated that these effects were attributable to upregulation of CYP1a1 and subsequent increased metabolism of estradiol to a downstream intermediate 2-methoxyestradiol. Microarray analysis of mouse lung exposed to 14 days of hyperoxia showed the most significant alteration in CYP1a1 expression, with minimal changes in expression of five other genes related to estrogen receptors, synthesis, and metabolism. Our novel results on estradiol metabolism in fetal and early postnatal lung in the context of hyperoxia indicate CYP1a1 as a potential mechanism for the protective effect of estradiol in hyperoxia-exposed immature lung, which may help explain the sex difference in neonatal lung diseases.


Assuntos
Citocromo P-450 CYP1A1/biossíntese , Estradiol/metabolismo , Hiperóxia/fisiopatologia , Pulmão/embriologia , 2-Metoxiestradiol , Animais , Apoptose , Aromatase/biossíntese , Asma/epidemiologia , Displasia Broncopulmonar/epidemiologia , Catecol O-Metiltransferase/biossíntese , Hipóxia Celular/fisiologia , Proliferação de Células , Células Cultivadas , Citocromo P-450 CYP1B1/biossíntese , Estradiol/análogos & derivados , Estradiol/biossíntese , Receptor alfa de Estrogênio/biossíntese , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/biossíntese , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Humanos , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Endogâmicos ICR , Músculo Liso/metabolismo , Oxigênio/metabolismo , RNA Mensageiro/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Receptores de Estrogênio/biossíntese , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Fatores Sexuais , Regulação para Cima
19.
Am J Physiol Lung Cell Mol Physiol ; 307(12): L978-86, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25344066

RESUMO

Cigarette smoke is a common environmental insult associated with increased risk of developing airway diseases such as wheezing and asthma in neonates and children. In adults, asthma involves airway remodeling characterized by increased airway smooth muscle (ASM) cell proliferation and increased extracellular matrix (ECM) deposition, as well as airway hyperreactivity. The effects of cigarette smoke on remodeling and contractility in the developing airway are not well-elucidated. In this study, we used canalicular-stage (18-20 wk gestational age) human fetal airway smooth muscle (fASM) cells as an in vitro model of the immature airway. fASM cells were exposed to cigarette smoke extract (CSE; 0.5-1.5% for 24-72 h), and cell proliferation, ECM deposition, and intracellular calcium ([Ca(2+)]i) responses to agonist (histamine 10 µM) were used to evaluate effects on remodeling and hyperreactivity. CSE significantly increased cell proliferation and deposition of ECM molecules collagen I, collagen III, and fibronectin. In contrast, [Ca(2+)]i responses were not significantly affected by CSE. Analysis of key signaling pathways demonstrated significant increase in extracellular signal-related kinase (ERK) and p38 activation with CSE. Inhibition of ERK or p38 signaling prevented CSE-mediated changes in proliferation, whereas only ERK inhibition attenuated the CSE-mediated increase in ECM deposition. Overall, these results demonstrate that cigarette smoke may enhance remodeling in developing human ASM through hyperplasia and ECM production, thus contributing to development of neonatal and pediatric airway disease.


Assuntos
Proliferação de Células , Matriz Extracelular/metabolismo , Modelos Biológicos , Músculo Liso/metabolismo , Sistema Respiratório/metabolismo , Fumar/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio , Colágeno Tipo I/biossíntese , Colágeno Tipo III/biossíntese , Matriz Extracelular/patologia , Feto/metabolismo , Feto/patologia , Humanos , Sistema de Sinalização das MAP Quinases , Músculo Liso/patologia , Sistema Respiratório/patologia , Fumar/efeitos adversos , Fumar/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
20.
Lancet Respir Med ; 2(10): 796-803, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25212745

RESUMO

BACKGROUND: The effectiveness of intermittent montelukast for wheeze in young children is unclear. We aimed to assess whether intermittent montelukast is better than placebo for treatment of wheeze in this age group. Because copy numbers of the Sp1-binding motif in the arachidonate 5-lipoxygenase (ALOX5) gene promoter (either 5/5, 5/x, or x/x, where x does not equal 5) modifies response to montelukast in adults, we stratified by this genotype. METHODS: We did this multicentre, parallel-group, randomised, placebo-controlled trial between Oct 1, 2010, and Dec 20, 2013, at 21 primary care sites and 41 secondary care sites in England and Scotland. Children aged 10 months to 5 years with two or more wheeze episodes were allocated to either a 5/5 or 5/x+x/x ALOX5 promoter genotype stratum, then randomly assigned (1:1) via a permuted block schedule (size ten), to receive intermittent montelukast or placebo given by parents at each wheeze episode over a 12 month period. Clinical investigators and parents were masked to treatment group and genotype strata. The primary outcome was number of unscheduled medical attendances for wheezing episodes. Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT01142505. FINDINGS: We randomly assigned 1358 children to receive montelukast (n=669) or placebo (n=677). Consent was withdrawn for 12 (1%) children. Primary outcome data were available for 1308 (96%) children. There was no difference in unscheduled medical attendances for wheezing episodes between children in the montelukast and placebo groups (mean 2·0 [SD 2·6] vs 2·3 [2·7]; incidence rate ratio [IRR] 0·88, 95% CI: 0·77-1·01; p=0·06). Compared with placebo, unscheduled medical attendances for wheezing episodes were reduced in children given montelukast in the 5/5 stratum (2·0 [2·7] vs 2·4 [3·0]; IRR 0·80, 95% CI 0·68-0·95; p=0·01), but not in those in the 5/x+x/x stratum (2·0 [2·5] vs 2·0 [2·3]; 1·03, 0·83-1·29; p=0·79, pinteraction=0·08). We recorded one serious adverse event, which was a skin reaction in a child allocated to placebo. INTERPRETATION: Our findings show no clear benefit of intermittent montelukast in young children with wheeze. However, the 5/5 ALOX5 promoter genotype might identify a montelukast-responsive subgroup. FUNDING: Medical Research Council (UK) and National Institute for Health Research.


Assuntos
Acetatos/uso terapêutico , Antiasmáticos/uso terapêutico , Asma/tratamento farmacológico , Quinolinas/uso terapêutico , Sons Respiratórios/efeitos dos fármacos , Acetatos/administração & dosagem , Agendamento de Consultas , Araquidonato 5-Lipoxigenase/efeitos dos fármacos , Pré-Escolar , Ciclopropanos , Cisteína/urina , Esquema de Medicação , Feminino , Genótipo , Humanos , Lactente , Leucotrienos/urina , Masculino , Quinolinas/administração & dosagem , Sulfetos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...