Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Pharmacol ; 13: 991072, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36386177

RESUMO

Airway cholinergic nerves play a key role in airway physiology and disease. In asthma and other diseases of the respiratory tract, airway cholinergic neurons undergo plasticity and contribute to airway hyperresponsiveness and mucus secretion. We currently lack human in vitro models for airway cholinergic neurons. Here, we aimed to develop a human in vitro model for peripheral cholinergic neurons using human pluripotent stem cell (hPSC) technology. hPSCs were differentiated towards vagal neural crest precursors and subsequently directed towards functional airway cholinergic neurons using the neurotrophin brain-derived neurotrophic factor (BDNF). Cholinergic neurons were characterized by ChAT and VAChT expression, and responded to chemical stimulation with changes in Ca2+ mobilization. To culture these cells, allowing axonal separation from the neuronal cell bodies, a two-compartment PDMS microfluidic chip was subsequently fabricated. The two compartments were connected via microchannels to enable axonal outgrowth. On-chip cell culture did not compromise phenotypical characteristics of the cells compared to standard culture plates. When the hPSC-derived peripheral cholinergic neurons were cultured in the chip, axonal outgrowth was visible, while the somal bodies of the neurons were confined to their compartment. Neurons formed contacts with airway smooth muscle cells cultured in the axonal compartment. The microfluidic chip developed in this study represents a human in vitro platform to model neuro-effector interactions in the airways that may be used for mechanistic studies into neuroplasticity in asthma and other lung diseases.

2.
BMC Pulm Med ; 19(1): 223, 2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-31771541

RESUMO

BACKGROUND: The CAnadian REgistry for Pulmonary Fibrosis (CARE-PF) is a multi-center, prospective registry designed to study the natural history of fibrotic interstitial lung disease (ILD) in adults. The aim of this cross-sectional sub-study was to describe the baseline characteristics, risk factors, and comorbidities of patients enrolled in CARE-PF to date. METHODS: Patients completed study questionnaires and clinical measurements at enrollment and each follow-up visit. Environmental exposures were assessed by patient self-report and comorbidities by the Charlson Comorbidity Index (CCI). Baseline characteristics, exposures, and comorbidities were described for the overall study population and for incident cases, and were compared across ILD subtypes. RESULTS: The full cohort included 1285 patients with ILD (961 incident cases (74.8%)). Diagnoses included connective tissue disease-associated ILD (33.3%), idiopathic pulmonary fibrosis (IPF) (24.7%), unclassifiable ILD (22.3%), chronic hypersensitivity pneumonitis (HP) (7.5%), sarcoidosis (3.2%), non-IPF idiopathic interstitial pneumonias (3.0%, including idiopathic nonspecific interstitial pneumonia (NSIP) in 0.9%), and other ILDs (6.0%). Patient-reported exposures were most frequent amongst chronic HP, but common across all ILD subtypes. The CCI was ≤2 in 81% of patients, with a narrow distribution and range of values. CONCLUSIONS: CTD-ILD, IPF, and unclassifiable ILD made up 80% of ILD diagnoses at ILD referral centers in Canada, while idiopathic NSIP was rare when adhering to recommended diagnostic criteria. CCI had a very narrow distribution across our cohort suggesting it may be a poor discriminator in assessing the impact of comorbidities on patients with ILD.


Assuntos
Alveolite Alérgica Extrínseca/epidemiologia , Exposição Ambiental , Fibrose Pulmonar Idiopática/epidemiologia , Doenças Pulmonares Intersticiais/epidemiologia , Sistema de Registros , Adulto , Idoso , Canadá/epidemiologia , Comorbidade , Doenças do Tecido Conjuntivo/complicações , Estudos Transversais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Risco , Índice de Gravidade de Doença , Inquéritos e Questionários
3.
Sci Rep ; 8(1): 805, 2018 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-29339735

RESUMO

Asthma is an obstructive respiratory disease characterised by chronic inflammation with airway hyperresponsiveness. In asthmatic airways, there is an increase in airway smooth muscle (ASM) cell bulk, which differs from non-asthmatic ASM in characteristics. This study aimed to assess the usefulness of hTERT immortalisation of human ASM cells as a research tool. Specifically we compared proliferative capacity, inflammatory mediator release and extracellular matrix (ECM) production in hTERT immortalised and parent primary ASM cells from asthmatic and non-asthmatic donors. Our studies revealed no significant differences in proliferation, IL-6 and eotaxin-1 production, or CTGF synthesis between donor-matched parent and hTERT immortalised ASM cell lines. However, deposition of ECM proteins fibronectin and fibulin-1 was significantly lower in immortalised ASM cells compared to corresponding primary cells. Notably, previously reported differences in proliferation and inflammatory mediator release between asthmatic and non-asthmatic ASM cells were retained, but excessive ECM protein deposition in asthmatic ASM cells was lost in hTERT ASM cells. This study shows that hTERT immortalised ASM cells mirror primary ASM cells in proliferation and inflammatory profile characteristics. Moreover, we demonstrate both strengths and weaknesses of this immortalised cell model as a representation of primary ASM cells for future asthma pathophysiological research.


Assuntos
Miócitos de Músculo Liso/fisiologia , Fenótipo , Sistema Respiratório/citologia , Telomerase/metabolismo , Asma/patologia , Proliferação de Células , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Fatores Imunológicos/metabolismo , Telomerase/genética
4.
Biochim Biophys Acta Mol Cell Res ; 1865(2): 364-378, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29108912

RESUMO

Autophagy and apoptosis are two major interconnected host cell responses to viral infection, including influenza A virus (IAV). Thus, delineating these events could facilitate the development of better treatment options and provide an effective anti-viral strategy for controlling IAV infection. We used A549 cells and mouse embryonic fibroblasts (MEF) to study the role of virus-induced autophagy and apoptosis, the cross-talk between both pathways, and their relation to IAV infection [ATCC strain A/Puerto Rico/8/34(H1N1) (hereafter; PR8)]. PR8-infected and mock-infected cells were analyzed by immunoblotting, immunofluorescence confocal microscopy, electron microscopy and flow cytometry (FACS). We found that PR8 infection simultaneously induced autophagy and apoptosis in A549 cells. Autophagy was associated with Bax and Bak activation, intrinsic caspase cleavage and subsequent PARP-1 and BID cleavage. Both Bax knockout (KO) and Bax/Bak double knockout MEFs displayed inhibition of virus-induced cytopathology and cell death and diminished virus-mediated caspase activation, suggesting that virus-induced apoptosis is Bax/Bak-dependent. Biochemical inhibition of autophagy induction with 3-methyladenine blocked both virus replication and apoptosis pathways. These effects were replicated using autophagy-refractory Atg3 KO and Atg5 KO cells. Taken together, our data indicate that PR8 infection simultaneously induces autophagy and Bax/caspase-dependent apoptosis, with autophagy playing a role to support PR8 replication, in part, by modulating virus-induced apoptosis.


Assuntos
Apoptose , Autofagia , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/metabolismo , Replicação Viral/fisiologia , Células A549 , Animais , Humanos , Influenza Humana/genética , Influenza Humana/patologia , Camundongos , Camundongos Knockout
5.
Thorax ; 72(1): 74-82, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27325752

RESUMO

BACKGROUND: Asthma affects 300 million people worldwide. In asthma, the major cause of morbidity and mortality is acute airway narrowing, due to airway smooth muscle (ASM) hypercontraction, associated with airway remodelling. However, little is known about the transcriptional differences between healthy and asthmatic ASM cells. OBJECTIVES: To investigate the transcriptional differences between asthmatic and healthy airway smooth muscle cells (ASMC) in culture and investigate the identified targets using in vitro and ex vivo techniques. METHODS: Human asthmatic and healthy ASMC grown in culture were run on Affymetrix_Hugene_1.0_ST microarrays. Identified candidates were confirmed by PCR, and immunohistochemistry. Functional analysis was conducted using in vitro ASMC proliferation, attachment and contraction assays and ex vivo contraction of mouse airways. RESULTS: We suggest a novel role for latrophilin (LPHN) receptors, finding increased expression on ASMC from asthmatics, compared with non-asthmatics in vivo and in vitro, suggesting a role in mediating airway function. A single nucleotide polymorphism in LPHN1 was associated with asthma and with increased LPHN1 expression in lung tissue. When activated, LPHNs regulated ASMC adhesion and proliferation in vitro, and promoted contraction of mouse airways and ASMC. CONCLUSIONS: Given the need for novel inhibitors of airway remodelling and bronchodilators in asthma, the LPHN family may represent promising novel targets for future dual therapeutic intervention.


Assuntos
Asma/genética , Asma/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Peptídeos/genética , Acetilcolina/farmacologia , Animais , Estudos de Casos e Controles , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Masculino , Glicoproteínas de Membrana , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Contração Muscular/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Polimorfismo de Nucleotídeo Único , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Peptídeos/metabolismo , Sistema Respiratório/citologia , Venenos de Aranha/farmacologia , Transcrição Gênica
6.
Paediatr Respir Rev ; 21: 19-26, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27665512

RESUMO

Diabetes is an increasingly common complication of pregnancy. In parallel with this trend, a rise in chronic lung disease in children has been observed in recent decades. While several adverse health outcomes associated with exposure to diabetes in utero have been documented in epidemiological and experimental studies, few have examined the impact of diabetes in pregnancy on offspring lung health and respiratory disease. We provide a comprehensive overview of current literature on this topic, finding suggestive evidence that exposure to diabetes in utero may have adverse effects on lung development. Delayed lung maturation and increased risk of respiratory distress syndrome have been consistently observed among infants born to mothers with diabetes and these findings are also observed in some rodent models of diabetes in pregnancy. Further research is needed to confirm and characterize epidemiologic observations that diabetes in pregnancy may predispose offspring to childhood wheezing illness and asthma. Parallel translational studies in human pregnancy cohorts and experimental models are needed to explore the role of fetal programming and other potential biological mechanisms in this context.


Assuntos
Diabetes Gestacional/epidemiologia , Pulmão/embriologia , Gravidez em Diabéticas/epidemiologia , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Doenças Respiratórias/epidemiologia , Animais , Asma/epidemiologia , Asma/metabolismo , Displasia Broncopulmonar/epidemiologia , Displasia Broncopulmonar/metabolismo , Criança , Diabetes Gestacional/metabolismo , Modelos Animais de Doenças , Feminino , Hérnias Diafragmáticas Congênitas/epidemiologia , Hérnias Diafragmáticas Congênitas/metabolismo , Humanos , Recém-Nascido , Gravidez , Gravidez em Diabéticas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Síndrome do Desconforto Respiratório do Recém-Nascido/epidemiologia , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Sons Respiratórios , Doenças Respiratórias/metabolismo
7.
Clin Exp Allergy ; 46(7): 945-56, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27079765

RESUMO

BACKGROUND: Chronic inflammation, typified by increased expression of IL-17A, together with airway and parenchymal remodelling are features of chronic lung diseases. Emerging evidence suggests that phenotypic heterogeneity of repair and inflammatory capacities of fibroblasts may contribute to the differential structural changes observed in different regions of the lung. OBJECTIVE: To investigate phenotypic differences in parenchymal and bronchial fibroblasts, either in terms of inflammation and remodelling or the ability of these fibroblasts to respond to IL-17A. METHODS: Four groups of primary fibroblasts were used: normal human bronchial fibroblast (NHBF), normal human parenchymal fibroblast (NHPF), COPD human bronchial fibroblast (CHBF) and COPD human parenchymal fibroblast (CHPF). Cytokine and extracellular matrix (ECM) expression were measured at baseline and after stimulation with IL-17A. Actinomycin D was used to measure cytokine mRNA stability. RESULTS: At baseline, we observed higher protein production of IL-6 in NHPF than NHBF, but higher levels of IL-8 and GRO-α in NHBF. IL-17A induced a higher expression of GRO-α (CXCL1) and IL-6 in NHPF than in NHBF, and a higher level of IL-8 expression in NHBF. IL-17A treatment decreased the mRNA stability of IL-6 in NHBF when compared with NHPF. CHPF expressed higher protein levels of fibronectin, collagen-I and collagen-III than CHBF, NHBF and NHPF. IL-17A increased fibronectin and collagen-III protein only in NHPF and collagen-III protein production in CHBF and CHPF. CONCLUSIONS AND CLINICAL RELEVANCE: These findings provide insight into the inflammatory and remodelling processes that may be related to the phenotypic heterogeneity of fibroblasts from airway and parenchymal regions and in their response to IL-17A.


Assuntos
Brônquios/metabolismo , Fibroblastos/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Interleucina-17/metabolismo , Tecido Parenquimatoso/metabolismo , Brônquios/citologia , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular , Fibroblastos/efeitos dos fármacos , Expressão Gênica , Humanos , Interleucina-17/farmacologia , Tecido Parenquimatoso/citologia , Doença Pulmonar Obstrutiva Crônica/etiologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia
8.
Cell Death Dis ; 6: e1944, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26512955

RESUMO

Exposure to metabolic disease during fetal development alters cellular differentiation and perturbs metabolic homeostasis, but the underlying molecular regulators of this phenomenon in muscle cells are not completely understood. To address this, we undertook a computational approach to identify cooperating partners of the myocyte enhancer factor-2 (MEF2) family of transcription factors, known regulators of muscle differentiation and metabolic function. We demonstrate that MEF2 and the serum response factor (SRF) collaboratively regulate the expression of numerous muscle-specific genes, including microRNA-133a (miR-133a). Using tandem mass spectrometry techniques, we identify a conserved phosphorylation motif within the MEF2 and SRF Mcm1 Agamous Deficiens SRF (MADS)-box that regulates miR-133a expression and mitochondrial function in response to a lipotoxic signal. Furthermore, reconstitution of MEF2 function by expression of a neutralizing mutation in this identified phosphorylation motif restores miR-133a expression and mitochondrial membrane potential during lipotoxicity. Mechanistically, we demonstrate that miR-133a regulates mitochondrial function through translational inhibition of a mitophagy and cell death modulating protein, called Nix. Finally, we show that rodents exposed to gestational diabetes during fetal development display muscle diacylglycerol accumulation, concurrent with insulin resistance, reduced miR-133a, and elevated Nix expression, as young adult rats. Given the diverse roles of miR-133a and Nix in regulating mitochondrial function, and proliferation in certain cancers, dysregulation of this genetic pathway may have broad implications involving insulin resistance, cardiovascular disease, and cancer biology.


Assuntos
Diferenciação Celular/genética , Fatores de Transcrição MEF2/química , Mitocôndrias/fisiologia , Fibras Musculares Esqueléticas/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/química , Motivos de Aminoácidos , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Diabetes Gestacional , Feminino , Regulação da Expressão Gênica , Humanos , Fatores de Transcrição MEF2/metabolismo , Fatores de Transcrição MEF2/fisiologia , Potencial da Membrana Mitocondrial/genética , MicroRNAs/metabolismo , Mitocôndrias/genética , Fibras Musculares Esqueléticas/citologia , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/citologia , Miócitos de Músculo Liso/citologia , Fosforilação , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Fator de Resposta Sérica/metabolismo , Fator de Resposta Sérica/fisiologia , Espectrometria de Massas em Tandem
9.
Cell Death Dis ; 6: e1696, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25789971

RESUMO

Transforming growth factor-ß(1) (TGF-ß(1)) is an important regulator of fibrogenesis in heart disease. In many other cellular systems, TGF-ß(1) may also induce autophagy, but a link between its fibrogenic and autophagic effects is unknown. Thus we tested whether or not TGF-ß(1)-induced autophagy has a regulatory function on fibrosis in human atrial myofibroblasts (hATMyofbs). Primary hATMyofbs were treated with TGF-ß(1) to assess for fibrogenic and autophagic responses. Using immunoblotting, immunofluorescence and transmission electron microscopic analyses, we found that TGF-ß(1) promoted collagen type Iα2 and fibronectin synthesis in hATMyofbs and that this was paralleled by an increase in autophagic activation in these cells. Pharmacological inhibition of autophagy by bafilomycin-A1 and 3-methyladenine decreased the fibrotic response in hATMyofb cells. ATG7 knockdown in hATMyofbs and ATG5 knockout (mouse embryonic fibroblast) fibroblasts decreased the fibrotic effect of TGF-ß(1) in experimental versus control cells. Furthermore, using a coronary artery ligation model of myocardial infarction in rats, we observed increases in the levels of protein markers of fibrosis, autophagy and Smad2 phosphorylation in whole scar tissue lysates. Immunohistochemistry for LC3ß indicated the localization of punctate LC3ß with vimentin (a mesenchymal-derived cell marker), ED-A fibronectin and phosphorylated Smad2. These results support the hypothesis that TGF-ß(1)-induced autophagy is required for the fibrogenic response in hATMyofbs.


Assuntos
Autofagia/genética , Fibrose/genética , Átrios do Coração/metabolismo , Miofibroblastos/metabolismo , Fator de Crescimento Transformador beta1/biossíntese , Adenina/administração & dosagem , Adenina/análogos & derivados , Animais , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Fibronectinas/biossíntese , Fibrose/patologia , Átrios do Coração/patologia , Humanos , Macrolídeos/administração & dosagem , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Miofibroblastos/patologia , Cultura Primária de Células , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad2/biossíntese , Proteína Smad2/genética , Fator de Crescimento Transformador beta1/genética
10.
Cell Death Dis ; 3: e330, 2012 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-22717585

RESUMO

3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors (statins) are cholesterol-lowering drugs that exert other cellular effects and underlie their beneficial health effects, including those associated with myocardial remodeling. We recently demonstrated that statins induces apoptosis and autophagy in human lung mesenchymal cells. Here, we extend our knowledge showing that statins simultaneously induces activation of the apoptosis, autophagy and the unfolded protein response (UPR) in primary human atrial fibroblasts (hATF). Thus we tested the degree to which coordination exists between signaling from mitochondria, endoplasmic reticulum and lysosomes during response to simvastatin exposure. Pharmacologic blockade of the activation of ER-dependent cysteine-dependent aspartate-directed protease (caspase)-4 and lysosomal cathepsin-B and -L significantly decreased simvastatin-induced cell death. Simvastatin altered total abundance and the mitochondrial fraction of proapoptotic and antiapoptotic proteins, while c-Jun N-terminal kinase/stress-activated protein kinase mediated effects on B-cell lymphoma 2 expression. Chemical inhibition of autophagy flux with bafilomycin-A1 augmented simvastatin-induced caspase activation, UPR and cell death. In mouse embryonic fibroblasts that are deficient in autophagy protein 5 and refractory to autophagy induction, caspase-7 and UPR were hyper-induced upon treatment with simvastatin. These data demonstrate that mevalonate cascade inhibition-induced death of hATF manifests from a complex mechanism involving co-regulation of apoptosis, autophagy and UPR. Furthermore, autophagy has a crucial role in determining the extent of ER stress, UPR and permissiveness of hATF to cell death induced by statins.


Assuntos
Apoptose , Autofagia , Morte Celular , Estresse do Retículo Endoplasmático , Retículo Endoplasmático/metabolismo , Fibroblastos/efeitos dos fármacos , Ácido Mevalônico/metabolismo , Miocárdio/citologia , Caspase 7/metabolismo , Inibidores de Caspase/farmacologia , Caspases Iniciadoras/metabolismo , Células Cultivadas , Ativação Enzimática , Fibroblastos/metabolismo , Átrios do Coração/citologia , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Ácido Mevalônico/farmacologia , Transdução de Sinais , Sinvastatina/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos
11.
Clin Exp Allergy ; 42(1): 85-94, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22092970

RESUMO

BACKGROUND: Structural cells are an important reservoir of chemokines that coordinate the influx of various immune cells to the lungs of asthmatics. Airway smooth muscle cells (ASMC) are an important source of these chemokines. CCL15 is a recently described chemo-attractant for neutrophils, eosinophils, monocytes and lymphocytes. OBJECTIVE: To determine the production and the regulation of CCL15 by ASMC and to investigate its production in asthmatic airways. METHODS: Human ASMC were obtained from main bronchial airway segments of patients with mild, moderate and severe asthma. To induce chemokine production, cells were incubated with IL-4, IL-13, TNF-α or IFN-γ in presence or absence of dexamethasone, mithramycin A (SP-1 inhibitor) or the IKK-2 inhibitor, AS602868. CCL15 mRNA expression was evaluated by real-time PCR. Immunoreactive CCL15 was detected by immuno-fluorescence and CCL15 protein concentration in the supernatant was measured using ELISA. RESULTS: CCL15 is constitutively expressed in human ASMC and is strongly up-regulated by TNF-α. This up-regulation is inhibited by dexamethasone, mithramycin A and AS602868. TNF-α-induced CCL15 levels can be synergistically enhanced by the presence of IFN-γ, at both the transcriptional and translation level. This synergism is NF-κB-dependent. Asthmatic biopsies demonstrated higher expression of CCL15 compared with non-asthmatic controls. CONCLUSION AND CLINICAL RELEVANCE: Our results show that ASMC are a potent source of CCL15 in the airways and may directly participate in the recruitment of inflammatory cells to asthmatic airways. Targeting the production of CCL15 by ASMC might reduce the inflammatory response within the airways of asthmatic patients.


Assuntos
Asma/fisiopatologia , Brônquios/citologia , Quimiocinas CC/metabolismo , Proteínas Inflamatórias de Macrófagos/metabolismo , Miócitos de Músculo Liso/metabolismo , Regulação para Cima , Adulto , Asma/imunologia , Biópsia , Quimiocinas CC/efeitos dos fármacos , Quimiocinas CC/genética , Quimiocinas CC/imunologia , Feminino , Humanos , Interferon gama/imunologia , Interferon gama/farmacologia , Proteínas Inflamatórias de Macrófagos/efeitos dos fármacos , Proteínas Inflamatórias de Macrófagos/genética , Proteínas Inflamatórias de Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade , Miócitos de Músculo Liso/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/farmacologia
12.
Eur Respir J ; 38(4): 841-50, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21565918

RESUMO

Inhibition of Rho-associated coiled-coil forming kinases (ROCKs) reduces allergic airway responses in mice. The purpose of this study was to determine the roles of the two ROCK isoforms, ROCK1 and ROCK2, in these responses. Wildtype (WT) mice and heterozygous ROCK1 and ROCK2 knockout mice (ROCK1(+/-) and ROCK2(+/-), respectively) were sensitised and challenged with ovalbumin. ROCK expression and activation were assessed by western blotting. Airway responsiveness was measured by forced oscillation. Bronchoalveolar lavage was performed and the lungs were fixed for histological assessment. Compared with WT mice, ROCK1 and ROCK2 expression were 50% lower in lungs of ROCK1(+/-) and ROCK2(+/-) mice, respectively, without changes in the other isoform. In WT lungs, ROCK activation increased after ovalbumin challenge and was sustained for several hours. This activation was reduced in ROCK1(+/-) and ROCK2(+/-) lungs. Airway responsiveness was comparable in WT, ROCK1(+/-), and ROCK2(+/-) mice challenged with PBS. Ovalbumin challenge caused airway hyperresponsiveness in WT, but not ROCK1(+/-) or ROCK2(+/-) mice. Lavage eosinophils and goblet cell hyperplasia were significantly reduced in ovalbumin-challenged ROCK1(+/-) and ROCK2(+/-) versus WT mice. Ovalbumin-induced changes in lavage interleukin-13, interleukin-5 and lymphocytes were also reduced in ROCK1(+/-) mice. In conclusion, both ROCK1 and ROCK2 are important in regulating allergic airway responses.


Assuntos
Hiper-Reatividade Brônquica/imunologia , Hipersensibilidade/imunologia , Quinases Associadas a rho/imunologia , Animais , Hiper-Reatividade Brônquica/genética , Hiper-Reatividade Brônquica/patologia , Líquido da Lavagem Broncoalveolar/imunologia , Células Cultivadas , Técnicas de Cocultura , Células Dendríticas/citologia , Células Dendríticas/imunologia , Eosinófilos/imunologia , Eosinófilos/patologia , Feminino , Expressão Gênica/imunologia , Células Caliciformes/imunologia , Células Caliciformes/patologia , Hipersensibilidade/genética , Hipersensibilidade/patologia , Interleucina-13/imunologia , Interleucina-5/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Mecânica Respiratória/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Quinases Associadas a rho/genética
13.
Br J Pharmacol ; 163(6): 1223-36, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21385177

RESUMO

BACKGROUND AND PURPOSE: Neonatal pulmonary hypertension (PPHN) is characterized by pulmonary vasoconstriction, due in part to dysregulation of the thromboxane prostanoid (TP) receptor. Hypoxia induces TP receptor-mediated hyperresponsiveness, whereas serine phosphorylation mediates desensitization of TP receptors. We hypothesized that prostacyclin (IP) receptor activity induces TP receptor phosphorylation and decreases ligand affinity; that TP receptor sensitization in hypoxic myocytes is due to IP receptor inactivation; and that this would be reversible by the cAMP-specific phosphodiesterase inhibitor milrinone. EXPERIMENTAL APPROACH: We examined functional regulation of TP receptors by serine phosphorylation and effects of IP receptor stimulation and protein kinase A (PKA) activity on TP receptor sensitivity in myocytes from neonatal porcine resistance pulmonary arteries after 72 h hypoxia in vitro. Ca(2+) response curves to U46619 (TP receptor agonist) were determined in hypoxic and normoxic myocytes incubated with or without iloprost (IP receptor agonist), forskolin (adenylyl cyclase activator), H8 (PKA inhibitor) or milrinone. TP and IP receptor saturation binding kinetics were measured in presence of iloprost or 8-bromo-cAMP. KEY RESULTS: Ligand affinity for TP receptors was normalized in vitro by IP receptor signalling intermediates. However, IP receptor affinity was compromised in hypoxic myocytes, decreasing cAMP production. Milrinone normalized TP receptor sensitivity in hypoxic myocytes by restoring PKA-mediated regulatory TP receptor phosphorylation. CONCLUSIONS AND IMPLICATIONS: TP receptor sensitivity and EC(50) for TP receptor agonists was regulated by PKA, as TP receptor serine phosphorylation by PKA down-regulated Ca(2+) mobilization. Hypoxia decreased IP receptor activity and cAMP generation, inducing TP receptor hyperresponsiveness, which was reversed by milrinone.


Assuntos
Milrinona/farmacologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Oxigênio/farmacologia , Artéria Pulmonar/citologia , Receptores de Tromboxanos/metabolismo , Vasodilatadores/farmacologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Iloprosta/farmacologia , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Tromboxanos/agonistas , Receptores de Tromboxanos/antagonistas & inibidores , Transdução de Sinais , Suínos
14.
J Med Genet ; 46(8): 497-510, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19505876

RESUMO

The inactivation of programmed cell death has profound effects not only on the development but also on the overall integrity of multicellular organisms. Beside developmental abnormalities, it may lead to tumorigenesis, autoimmunity, and other serious health problems. Deregulated apoptosis may also be the leading cause of cancer therapy chemoresistance. Caspase family of cysteinyl-proteases plays the key role in the initiation and execution of programmed cell death. This review gives an overview of the role of caspases, their natural modulators like IAPs, FLIPs, and Smac/Diablo in apoptosis and upon inactivation, and also in cancer development. Besides describing the basic mechanisms governing programmed cell death, a large part of this review is dedicated to previous studies that were focused on screening tumours for mutations within caspase genes as well as their regulators. The last part of this review discusses several emerging treatments that involve modulation of caspases and their regulators. Thus, we also highlight caspase cascade modulating experimental anticancer drugs like cFLIP-antagonist CDDO-Me; cIAP1 antagonists OSU-03012 and ME-BS; and XIAP small molecule antagonists 1396-11, 1396-12, 1396-28, triptolide, AEG35156, survivin/Hsp90 antagonist shephedrin, and some of the direct activators of procaspase-3.


Assuntos
Apoptose/genética , Caspases/genética , Mutação , Neoplasias/genética , Neoplasias/patologia , Animais , Inibidores de Caspase , Humanos , Modelos Biológicos , Neoplasias/enzimologia
15.
Eur Respir J ; 34(6): 1436-43, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19460789

RESUMO

Acetylcholine is the primary parasympathetic neurotransmitter in the airways and is known to cause bronchoconstriction and mucus secretion. Recent findings suggest that acetylcholine also regulates aspects of remodelling and inflammation through its action on muscarinic receptors. In the present study, we aimed to determine the effects of muscarinic receptor stimulation on cytokine production by human airway smooth muscle cells (primary and immortalised cell lines). The muscarinic receptor agonists carbachol and methacholine both induced modest effects on basal interleukin (IL)-8 and -6 secretion, whereas the secretion of RANTES, eotaxin, vascular endothelial growth factor-A and monocyte chemoattractant protein-1 was not affected. Secretion of IL-8 and -6 was only observed in immortalised airway smooth muscle cells that express muscarinic M3 receptors. In these cells, methacholine also significantly augmented IL-8 secretion in combination with cigarette smoke extract in a synergistic manner, whereas synergistic effects on IL-6 secretion were not significant. Muscarinic M3 receptors were the primary subtype involved in augmenting cigarette smoke extract-induced IL-8 secretion, as only tiotropium bromide and muscarinic M3 receptor subtype selective antagonists abrogated the effects of methacholine. Collectively, these results indicate that muscarinic M3 receptor stimulation augments cigarette smoke extract-induced cytokine production by airway smooth muscle. This interaction could be of importance in patients with chronic obstructive pulmonary disease.


Assuntos
Interleucina-8/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor Muscarínico M3/metabolismo , Fumar/efeitos adversos , Acetilcolina/metabolismo , Brônquios/metabolismo , Células Cultivadas , Quimiocina CCL5/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Inflamação , Interleucina-6/metabolismo , Cloreto de Metacolina/farmacologia , Neurotransmissores/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/patologia
16.
Eur Respir J ; 30(4): 653-61, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17537779

RESUMO

Chronic inflammation in asthma and chronic obstructive pulmonary disease drives pathological structural remodelling of the airways. Using tiotropium bromide, acetylcholine was recently identified as playing a major regulatory role in airway smooth muscle remodelling in a guinea pig model of ongoing allergic asthma. The aim of the present study was to investigate other aspects of airway remodelling and to compare the effectiveness of tiotropium to the glucocorticosteroid budesonide. Ovalbumin-sensitised guinea pigs were challenged for 12 weeks with aerosolised ovalbumin. The ovalbumin induced airway smooth muscle thickening, hypercontractility of tracheal smooth muscle, increased pulmonary contractile protein (smooth-muscle myosin) abundance, mucous gland hypertrophy, an increase in mucin 5 subtypes A and C (MUC5AC)-positive goblet cell numbers and eosinophilia. It was reported previously that treatment with tiotropium inhibits airway smooth muscle thickening and contractile protein expression, and prevents tracheal hypercontractility. This study demonstrates that tiotropium also fully prevented allergen-induced mucous gland hypertrophy, and partially reduced the increase in MUC5AC-positive goblet cell numbers and eosinophil infiltration. Treatment with budesonide also prevented airway smooth muscle thickening, contractile protein expression, tracheal hypercontractility and mucous gland hypertrophy, and partially reduced MUC5AC-positive goblet cell numbers and eosinophilia. This study demonstrates that tiotropium and budesonide are similarly effective in inhibiting several aspects of airway remodelling, providing further evidence that the beneficial effects of tiotropium bromide might exceed those of bronchodilation.


Assuntos
Alérgenos/química , Budesonida/uso terapêutico , Hipersensibilidade/tratamento farmacológico , Derivados da Escopolamina/uso terapêutico , Corticosteroides/metabolismo , Animais , Broncodilatadores/farmacologia , Budesonida/química , Antagonistas Colinérgicos/farmacologia , Eosinofilia , Matriz Extracelular/metabolismo , Glucocorticoides/química , Cobaias , Humanos , Inflamação , Masculino , Músculo Liso/metabolismo , Ovalbumina/química , Brometo de Tiotrópio , Traqueia/patologia
17.
Allergy ; 62(6): 675-82, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17508973

RESUMO

BACKGROUND: Monoclonal antibodies or soluble receptors have been used to block over-produced endogenous cytokines. However, they have disadvantages of short half-lives, high costs, and possible adverse effects. Using interleukin (IL)-4 as a model target, we sought to develop a novel therapeutic strategy by constructing an IL-4 peptide-based vaccine for blocking IL-4 on a persistent basis, and to evaluate its efficacy in a mouse model of asthma. METHODS: A peptide was selected by antigenic prediction and structure analysis of IL-4/receptor complex. The vaccine was constructed by employing truncated hepatitis B core antigen as carrier with the peptide inserted using gene engineering methods. It was then expressed, purified and identified. Prior to intraperitoneal sensitization and intranasal challenge with ovalbumin, mice were subcutaneously immunized three times with the vaccine, or the carrier or saline as controls. Serum antibodies, inflammatory cells in bronchoalveolar lavage fluids (BALF), lung histology, and responsiveness to inhaled methacholine were analyzed. RESULTS: The vaccine presented as virus-like particles and reacted to polyclonal anti-IL-4 in Western blotting. Vaccinated mice produced high titers of IgG to IL-4. Serum ovalbumin-specific IgE, eosinophil accumulation in BALF, goblet cell hyperplasia, tissue inflammation and methacoline-induced respiratory responses were markedly suppressed in vaccinated mice with statistical significance, as compared with those in the control groups. CONCLUSIONS: Administration of this novel IL-4 vaccine led to an overall decrease in the development of airway allergic inflammatory responses. The results indicate that cytokine peptide-based vaccines hold potential for treatment of asthma and, by extension, other diseases where over-expressed cytokines play a pivotal role in pathogenesis.


Assuntos
Hiper-Reatividade Brônquica/prevenção & controle , Imunoterapia/métodos , Interleucina-4/antagonistas & inibidores , Interleucina-4/imunologia , Vacinas Sintéticas/uso terapêutico , Animais , Asma/fisiopatologia , Asma/prevenção & controle , Autoanticorpos/imunologia , Western Blotting , Hiper-Reatividade Brônquica/sangue , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Engenharia Genética/métodos , Antígenos do Núcleo do Vírus da Hepatite B/imunologia , Imunoglobulina G/sangue , Camundongos , Camundongos Endogâmicos BALB C , Peptídeos/imunologia , Peptídeos/uso terapêutico
18.
Eur Respir J ; 29(5): 834-60, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17470619

RESUMO

Excessive airway obstruction is the cause of symptoms and abnormal lung function in asthma. As airway smooth muscle (ASM) is the effecter controlling airway calibre, it is suspected that dysfunction of ASM contributes to the pathophysiology of asthma. However, the precise role of ASM in the series of events leading to asthmatic symptoms is not clear. It is not certain whether, in asthma, there is a change in the intrinsic properties of ASM, a change in the structure and mechanical properties of the noncontractile components of the airway wall, or a change in the interdependence of the airway wall with the surrounding lung parenchyma. All these potential changes could result from acute or chronic airway inflammation and associated tissue repair and remodelling. Anti-inflammatory therapy, however, does not "cure" asthma, and airway hyperresponsiveness can persist in asthmatics, even in the absence of airway inflammation. This is perhaps because the therapy does not directly address a fundamental abnormality of asthma, that of exaggerated airway narrowing due to excessive shortening of ASM. In the present study, a central role for airway smooth muscle in the pathogenesis of airway hyperresponsiveness in asthma is explored.


Assuntos
Obstrução das Vias Respiratórias/fisiopatologia , Asma/fisiopatologia , Hiper-Reatividade Brônquica/fisiopatologia , Músculo Liso/fisiopatologia , Adaptação Fisiológica , Apoptose , Humanos , Contração Muscular/fisiologia , Testes de Função Respiratória , Mecânica Respiratória
19.
Curr Drug Targets ; 7(5): 525-40, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16719764

RESUMO

Asthma incidence has climbed markedly in the past two decades despite an increased use of medications that suppress airway inflammation and repress contraction of smooth muscle that encircles the airways. Asthmatics exhibit episodes of airway inflammation that potentiates reversible airway smooth muscle spasm. A hallmark diagnostic symptom of asthma is airway hyperresponsiveness to inhaled non-allergic stimuli, such as methacholine, that directly induce airway smooth muscle contraction. Inhaled gluccocorticoids are used for first-line prevention of airway inflammation, and are frequently combined with inhaled beta2-adrenoceptor agonists that can effectively relax airway smooth muscle and restore airway conductance. Leukotriene receptor antagonists and anti-cholinergics can also be used in many patients to ensure optimal control of symptoms. With increasing disease duration irreversible airway restriction develops from inflammation-driven fibro-proliferative airway remodeling that includes increased deposition of extracellular matrix, the accumulation of airway smooth muscle, and increased numbers of myofibroblasts. Mature airway smooth muscle cells are phenotypically plastic, enabling them to subserve contractile, proliferative, migratory and secretory functional responses that contribute to airway remodeling and persistent hyperresponsiveness. This review assesses current understanding of acute and chronic effects of common anti-asthma medications on the diverse phenotype and functional characteristics of airway smooth muscle cells. Furthermore, we describe the significance of these effects in the treatment of asthma symptoms and pathogenesis.


Assuntos
Antiasmáticos/farmacologia , Asma/tratamento farmacológico , Brônquios/fisiologia , Miócitos de Músculo Liso/fisiologia , Traqueia/fisiologia , Agonistas Adrenérgicos beta/farmacologia , Brônquios/efeitos dos fármacos , Hiper-Reatividade Brônquica/fisiopatologia , Proliferação de Células/efeitos dos fármacos , Antagonistas Colinérgicos/farmacologia , Regulação da Expressão Gênica , Glucocorticoides/farmacologia , Humanos , Contração Muscular , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Traqueia/efeitos dos fármacos
20.
Am J Physiol Lung Cell Mol Physiol ; 290(2): L375-84, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16214814

RESUMO

PPHN, caused by perinatal hypoxia or inflammation, is characterized by an increased thromboxane-prostacyclin ratio and pulmonary vasoconstriction. We examined effects of hypoxia on myocyte thromboxane responsiveness. Myocytes from 3rd-6th generation pulmonary arteries of newborn piglets were grown to confluence and synchronized in contractile phenotype by serum deprivation. On the final 3 days of culture, myocytes were exposed to 10% O2 for 3 days; control myocytes from normoxic piglets were cultured in 21% O2. PPHN was induced in newborn piglets by 3-day hypoxic exposure (Fi(O2) 0.10); pulmonary arterial myocytes from these animals were maintained in normoxia. Ca2+ mobilization to thromboxane mimetic U-46619 and ATP was quantified using fura-2 AM. Three-day hypoxic exposure in vitro results in increased basal [Ca2+]i, faster and heightened peak Ca2+ response, and decreased U-46619 EC50. These functional changes persist in myocytes exposed to hypoxia in vivo but cultured in 21% O2. Blockade of Ca2+ entry and store refilling do not alter peak U-46619 Ca2+ responses in hypoxic or normoxic myocytes. Blockade of ryanodine-sensitive or IP3-gated intracellular Ca2+ channels inhibits hypoxic augmentation of peak U-46619 response. Ca2+ response to ryanodine alone is undetectable; ATP-induced Ca2+ mobilization is unaltered by hypoxia, suggesting no independent increase in ryanodine-sensitive or IP3-linked intracellular Ca2+ pool mobilization. We conclude hypoxia has a priming effect on neonatal pulmonary arterial myocytes, resulting in increased resting Ca2+, thromboxane hypersensitivity, and hyperreactivity. We postulate that hypoxia increases agonist-induced TP-R-linked IP3 pathway activation. Myocyte thromboxane hyperresponsiveness persists in culture after removal from the initiating hypoxic stimulus, suggesting altered gene expression.


Assuntos
Hipóxia/fisiopatologia , Células Musculares/efeitos dos fármacos , Síndrome da Persistência do Padrão de Circulação Fetal/fisiopatologia , Receptores de Tromboxanos/agonistas , Vasoconstrição/efeitos dos fármacos , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico/farmacologia , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Ventrículos do Coração/anatomia & histologia , Ventrículos do Coração/patologia , Humanos , Recém-Nascido , Compostos Macrocíclicos , Nifedipino/farmacologia , Tamanho do Órgão , Oxazóis/farmacologia , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Rianodina/farmacologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...