Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Intern Med ; 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38462518

RESUMO

Objective We aimed to investigate the relationship between tortuosity of the extracranial internal carotid artery (ICA) or vertebral artery (VA) and vascular risk factors among residents of Asahikawa, northeast Japan. Methods We retrospectively surveyed participants of "brain dock" medical brain checkups, which involved magnetic resonance imaging and angiography. We measured the tortuosity of the ICA and VA, and evaluated vascular risk factors based on medical interviews, questionnaires, and medical records. Results A total of 218 participants were enrolled in the study. ICA tortuosity (right and left) was significantly correlated with age [odds ratio (OR): 2.452, 95% confidence interval (CI): 1.695-3.548, p<0.001]. A more pronounced correlation was observed in females than in males (OR: 1.678, 95% CI: 1.004-2.807, p=0.048). VA tortuosity (right and left) was significantly correlated with age (OR: 1.786, 95% CI: 1.250-2.550, p=0.001) and smoking history (OR: 2.140, 95% CI: 1.235-3.707, p=0.007), and was more pronounced in females than in males (OR: 1.864, 95% CI: 1.107-3.137, p=0.019). Conclusion ICA tortuosity was correlated with age, while VA tortuosity was correlated with age and smoking history. ICA and VA tortuosity were more pronounced in females than in males.

2.
J Environ Manage ; 180: 551-6, 2016 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-27322817

RESUMO

In this work, we propose a novel approach to dye decomposition under subcritical water conditions using a continuous-flow tubular reactor coated with thin layer of PdO as a catalyst. Remazole Brilliant Blue R was used as an example of synthetic dyes. Hydrogen peroxide was used as an environmental-friendly oxidant as it leaves no residues after treatment. The effect of temperature, pressure and dye concentration on total organic carbon (TOC) removal were studied. 99.9% of TOC removal was achieved at 300 °C and 10 MPa pressure within a short residence time of 3.2 s. This method provided an efficient and rapid process that has a potential for treating a wide range of textile wastewaters.


Assuntos
Antraquinonas/química , Corantes/química , Poluentes Químicos da Água/química , Peróxido de Hidrogênio/química , Oxidantes/química , Pressão , Temperatura , Eliminação de Resíduos Líquidos/métodos
3.
Transfusion ; 54(12): 3097-107, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24965098

RESUMO

BACKGROUND: Transfusion-related acute lung injury (TRALI) is a life-threatening complication of blood transfusion. Antibodies against human leukocyte antigens in donors' plasma are the major causes of TRALI. Several animal models of TRALI have been developed, and the mechanism underlying TRALI development has been extensively investigated using rodent models. Although sheep models of nonimmune TRALI have been developed, large-animal models of antibody-mediated TRALI are not yet available. STUDY DESIGN AND METHODS: To develop a swine model of TRALI, male Clawn strain miniature pigs were used. A monoclonal antibody (MoAb) against swine leukocyte antigens (SLAs) Class I (4G8, 0.3 or 1.0 mg/kg body weight [BW]) and a control antibody (1.0 mg/kg BW) were injected into the peripheral vein after priming with or without 1 µg/kg BW lipopolysaccharide (LPS; n = 3 each). Lung injury was assessed using PaO2 /FiO2 (P/F) ratio and by chest X-ray imaging. Histopathologic analysis was also conducted. RESULTS: Lung injury could be induced by injecting 4G8 at an amount of 1.0 mg/kg BW, after LPS. The P/F ratio 90 minutes after the administration of 4G8 significantly decreased (p < 0.05). Bilateral infiltration was shown in chest X-ray imaging. Lung injury was confirmed by histopathologic analysis. CONCLUSION: Lung injury in pigs was successfully induced by anti-SLA MoAb. Priming with LPS is a prerequisite for inducing lung injury and the amount of the antibody is a critical condition.


Assuntos
Lesão Pulmonar Aguda , Anticorpos Monoclonais Murinos/toxicidade , Transfusão de Sangue , Modelos Animais de Doenças , Antígenos de Histocompatibilidade Classe I/imunologia , Lipopolissacarídeos/toxicidade , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/diagnóstico por imagem , Lesão Pulmonar Aguda/fisiopatologia , Animais , Anticorpos Monoclonais Murinos/imunologia , Humanos , Pulmão/diagnóstico por imagem , Pulmão/fisiopatologia , Masculino , Radiografia , Testes de Função Respiratória , Suínos , Porco Miniatura
4.
Beilstein J Org Chem ; 9: 1156-63, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23843908

RESUMO

The inner surface of a metallic tube (i.d. 0.5 mm) was coated with a palladium (Pd)-based thin metallic layer by flow electroless plating. Simultaneous plating of Pd and silver (Ag) from their electroless-plating solution produced a mixed distributed bimetallic layer. Preferential acid leaching of Ag from the Pd-Ag layer produced a porous Pd surface. Hydrogenation of p-nitrophenol was examined in the presence of formic acid simply by passing the reaction solution through the catalytic tubular reactors. p-Aminophenol was the sole product of hydrogenation. No side reaction occurred. Reaction conversion with respect to p-nitrophenol was dependent on the catalyst layer type, the temperature, pH, amount of formic acid, and the residence time. A porous and oxidized Pd (PdO) surface gave the best reaction conversion among the catalytic reactors examined. p-Nitrophenol was converted quantitatively to p-aminophenol within 15 s of residence time in the porous PdO reactor at 40 °C. Evolution of carbon dioxide (CO2) was observed during the reaction, although hydrogen (H2) was not found in the gas phase. Dehydrogenation of formic acid did not occur to any practical degree in the absence of p-nitrophenol. Consequently, the nitro group was reduced via hydrogen transfer from formic acid to p-nitrophenol and not by hydrogen generated by dehydrogenation of formic acid.

5.
Int Arch Allergy Immunol ; 155(2): 119-28, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21196756

RESUMO

BACKGROUND: Airway remodeling is an important feature of chronic airway disease, but the mechanisms involved remain unclear. Recently, epithelial mesenchymal transition (EMT) was reported to be associated with tissue fibrosis. TGF-ß1, which is a potent inducer of EMT, is thought to be related to the pathogenesis of airway remodeling. We investigated whether TGF-ß1 and/or TNF-α induce EMT in bronchial epithelial cells. METHODS: Cultured BEAS-2B cells and primary normal human bronchial epithelial cells (NHBE) were treated with TGF-ß1 and/or TNF-α. Morphological changes and the expression of EMT-related markers were evaluated by immunocytochemical staining. Expressions of EMT-related markers, extracellular matrix (ECM) components (collagen type I and versican), and TGF-ß receptors I, II, and III were analyzed by quantitative RT-PCR. Migration was evaluated using the Boyden chamber technique. RESULTS: The TGF-ß1-induced EMT in BEAS-2B cells was demonstrated on the basis of morphological changes and the downregulation of E-cadherin. Costimulation with TNF-α enhanced the TGF-ß1-induced morphological changes and increased vimentin expression. Treatment with TGF-ß1 increased the expression of collagen type I and versican. EMT induced with TGF-ß1 plus TNF-α promoted cell migration. Stimulation of NHBE with TGF-ß1 led to EMT. CONCLUSION: TGF-ß1 induced EMT in BEAS-2B cells, and costimulation with TNF-α enhanced the EMT. As a result of the EMT process, BEAS-2B cells acquired functions of mesenchymal cells. In addition, TGF-ß1 treatment induced EMT in NHBE as shown by changes in EMT-related markers. Bronchial epithelial cells might contribute to airway remodeling through EMT.


Assuntos
Brônquios/citologia , Células Epiteliais/metabolismo , Receptor Cross-Talk , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Remodelação das Vias Aéreas , Biomarcadores/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Transformada , Movimento Celular/imunologia , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Transição Epitelial-Mesenquimal/imunologia , Humanos , Receptor Cross-Talk/imunologia , Fator de Crescimento Transformador beta/imunologia , Fator de Necrose Tumoral alfa/imunologia , Versicanas/genética , Versicanas/metabolismo , Vimentina/genética , Vimentina/metabolismo
6.
J Oleo Sci ; 59(10): 557-62, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20877149

RESUMO

Novel micro swirl mixers were developed to synthesize nanoparticles, and the effect of their mixing performance on the characteristics of the synthesized nanoparticles was determined. The results were compared with those obtained using simple T-shaped mixers under the same reaction conditions. The synthesis of NiO, whose characteristics depend on the mixing performance of the mixer, was chosen as a model reaction. Initial investigations highlighted that the average particle size decreased from 32 to 23 to 20 nm as the inner diameter of the swirl mixers was decreased from 3.2 mm (Swirl mixer, SM-3.2) to 0.8 mm (Micro swirl mixer, MSM-0.8) to 0.5 mm (Micro swirl mixer, MSM-0.5), respectively. On the other hand, a similar decrease in the average particle size from 34 to 20 nm was observed with a decrease in the inner diameter of the T-shaped mixers from 1.3 mm (Tee union, T-1.3) to 0.3 mm (Micro tee union, T-0.3), respectively. Further, narrow particle size distributions were observed with a decrease in the inner diameter of each mixer. Furthermore, a computational fluid dynamics (CFD) simulation indicated an excellent mixing mechanism, which contributed to the improvement in the heating rate and the formation of nanoparticles of smaller size with a narrow particle size distribution. The result presented here indicates that the micro swirl mixers produce high-quality metal oxide nanoparticles. The size of the obtained particles with improved size distributions was comparable to that of the particles obtained using the T-shaped mixers, although the inner diameter of the swirl mixers was larger. Therefore, preliminary evidence suggests that the swirl flow mixers have the ability to produce rapid and homogeneous fluid mixing, thus controlling the particle size.


Assuntos
Microfluídica/instrumentação , Nanopartículas/química , Níquel/química , Temperatura , Desenho de Equipamento , Microfluídica/métodos , Simulação de Dinâmica Molecular , Tamanho da Partícula , Propriedades de Superfície
7.
Exp Cell Res ; 316(20): 3501-11, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-20850432

RESUMO

Low concentrations of cigarette smoke induced DNA damage and repair without leading to apoptosis in human bronchial epithelial cells. Higher concentrations of cigarette smoke, however, could induce either apoptosis or necrosis. The current study demonstrated that 15% cigarette smoke extract (CSE) induced apoptosis as evidenced by DNA content profiling (17.8±2.1% vs 10.2±1.6% of control, p<0.05), LIVE/DEAD staining (60.2±2.1% viable cells in CSE-treated vs 86.5±2.3% in control cells, p<0.05), and COMET assay (24.3±0.6% of Apoptotic Index in the cells treated with CSE vs 4.7±0.6% of control, P<0.05). Hepatocyte growth factor (HGF) significantly blocked the cigarette smoke-induced apoptosis as shown by DNA profiling (10.8±1.5% of CSE+HGF, p<0.05), LIVE/DEAD staining (78.5±1.2% in CSE+HGF treated cells, p<0.05), and COMET assay (Apoptotic Index: 10.0±0.8% in CSE+HGF treated cells, P<0.05). This protective effect of HGF on CSE-induced apoptosis was abolished by PI3K inhibitors, wortmannin and LY294002, and by introduction of the dominant negative AKT into the cells. Furthermore, CSE plus HGF could induce phosphorylation of AKT Thr 308 and the pro-apoptotic protein, BAD. These results suggest that HGF modulates cell survival in response to cigarette smoke exposure through the PI3K/AKT signaling pathway.


Assuntos
Apoptose/efeitos dos fármacos , Brônquios/citologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Fator de Crescimento de Hepatócito/farmacologia , Nicotiana/química , Fumaça/efeitos adversos , Proteínas Reguladoras de Apoptose/metabolismo , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Humanos , Necrose/induzido quimicamente , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo , Proteína bcl-X/metabolismo
8.
Anal Chim Acta ; 674(2): 234-8, 2010 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-20678635

RESUMO

Facile and specific methods to quantify a nucleobase in biological samples are of great importance for diagnosing disorders in nucleic acid metabolism. In the present study, a novel fluorogenic reaction specific for uracil has been developed. The reaction was carried out in an alkaline medium containing benzamidoxime and K(3)[Fe(CN)(6)] which were heated for 2.0 min. Under the optimum reaction conditions, strong fluorescence was produced only from uracil, not from other many biogenic compounds tested such as cytosine, thymine, adenine, guanine, nucleobases, nucleosides, nucleotides, amino acids, saccharides, creatine, creatinine and urea. The sensitivity of this method was compared with a known fluorogenic reaction using phenacylbromide which does not react with uracil but reacts with cytosine, adenine and their analogues. The proposed uracil-specific reaction showed approximately 400-fold higher sensitivity than the phenacylbromide reaction. The lower detection limit of uracil by the present method was 100 pmol mL(-1), and a good linearity of the calibration curve was obtained up to 100 nmol mL(-1) uracil. Due to its high sensitivity and specificity, the quantitative determination of uracil was possible by the proposed fluorimetric method.


Assuntos
Fluorometria/métodos , Uracila/metabolismo , Acetofenonas/metabolismo , Benzamidinas/metabolismo , Citosina/metabolismo , Limite de Detecção
9.
Exp Lung Res ; 36(1): 12-24, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20128678

RESUMO

Recently, epithelial-mesenchymal transition (EMT) has been reported to contribute to tissue fibrosis through enhanced transforming growth factor (TGF)-beta1 signaling. Tumor necrosis factor (TNF)-alpha has also been implicated in tissue fibrosis. Therefore, the authors investigated whether TNF-alpha affected TGF-beta1-induced EMT. Cultured alveolar epithelial cells (A549 cells) were stimulated with TGF-beta1 (5 ng/mL), with/without TNF-alpha (10 ng/mL). TGF-beta1 induced EMT of A549 cells, with loss of E-cadherin and acquisition of vimentin. Combination of TNF-alpha with TGF-beta1 enhanced EMT, causing morphological changes, while quantitative polymerase chain reaction (PCR) showed suppression of E-cadherin mRNA and expression of vimentin mRNA. In addition, the gel contraction method revealed that cells that had undergone EMT acquired cell contractility, which is a feature of mesenchymal cells. Stimulation with TGF-beta1 induced cell contraction, as did TNF-alpha. Moreover, costimulation with TGF-beta1 and TNF-alpha enhanced the cell contraction. Although IFN-gamma suppressed spontaneous cell contraction, it did not suppress cell contraction, which was induced by TGF-beta1. In conclusion, TNF-alpha enhances not only EMT but also cell contraction induced by TGF-beta1. EMT might contribute to tissue fibrosis through induction of cell contraction.


Assuntos
Adenocarcinoma Bronquioloalveolar/patologia , Desdiferenciação Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Linhagem Celular Tumoral , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Fibrose/etiologia , Humanos , Células-Tronco Mesenquimais/patologia
10.
Am J Respir Cell Mol Biol ; 43(6): 635-40, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20042711

RESUMO

Exposure to cigarette smoke is associated with airway epithelial mucus cell hyperplasia and a decrease in cilia and ciliated cells. Few models have addressed the long-term effects of chronic cigarette smoke exposure on ciliated epithelial cells. Our previous in vitro studies showed that cigarette smoke decreases ciliary beat frequency (CBF) via the activation of protein kinase C (PKC). We hypothesized that chronic cigarette smoke exposure in an in vivo model would decrease airway epithelial cell ciliary beating in a PKC-dependent manner. We exposed C57BL/6 mice to whole-body cigarette smoke 2 hours/day, 5 days/week for up to 1 year. Tracheal epithelial cell CBF and the number of motile cells were measured after necropsy in cut tracheal rings, using high-speed digital video microscopy. Tracheal epithelial PKC was assayed according to direct kinase activity. At 6 weeks and 3 months of smoke exposure, the baseline CBF was slightly elevated (~1 Hz) versus control mice, with no change in ß-agonist-stimulated CBF between control mice and cigarette smoke-exposed mice. By 6 months of smoke exposure, the baseline CBF was significantly decreased (2-3 Hz) versus control mice, and a ß-agonist failed to stimulate increased CBF. The loss of ß-agonist-increased CBF continued at 9 months and 12 months of smoke exposure, and the baseline CBF was significantly decreased to less than one third of the control rate. In addition to CBF, ciliated cell numbers significantly decreased in response to smoke over time, with a significant loss of tracheal ciliated cells occurring between 6 and 12 months. In parallel with the slowing of CBF, significant PKC activation from cytosol to the membrane of tracheal epithelial cells was detected in mice exposed to smoke for 6-12 months.


Assuntos
Cílios/patologia , Células Epiteliais/patologia , Fumar/efeitos adversos , Acetilação , Animais , Cílios/enzimologia , Células Epiteliais/enzimologia , Feminino , Imuno-Histoquímica , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Proteína Quinase C/metabolismo , Fatores de Tempo , Traqueia/metabolismo , Traqueia/patologia , Tubulina (Proteína)/metabolismo
11.
Mol Cell Biochem ; 337(1-2): 77-81, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19851834

RESUMO

Histamine is a potent mediator in allergic inflammatory processes and is released by basophils and mast cells. The aim of this study was to investigate the effect of histamine on in vitro migration of human fetal lung fibroblasts (HFL-1) to human plasma fibronectin (HFn), a chemoattractant. Using the blindwell chamber technique, histamine alone had no chemotactic activity. However, histamine augmented HFn-induced HFL-1 migration at concentrations ranging between 0 and 10(-7) M (290.6 +/- 20.8%) (P < 0.05). The concentration-response was bell-shaped. The effect of histamine increased with time. The stimulatory effect of histamine on HFL-1 migration was inhibited by an H4 receptor antagonist, JNJ7777120 (10(-5) M). Histamine's effect was also inhibited by pertussis toxin (50 ng/ml), showing that the effect was mediated by the H4 receptor. This study demonstrated that histamine has the potential to stimulate human lung fibroblast migration, and thus may contribute to regulation of wound healing and the development of fibrotic disorders of the lung.


Assuntos
Movimento Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Histamina/farmacologia , Pulmão/efeitos dos fármacos , Ensaios de Migração Celular , Células Cultivadas , Difenidramina/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Fibroblastos/fisiologia , Fibronectinas/farmacologia , Antagonistas dos Receptores Histamínicos/farmacologia , Humanos , Indóis/farmacologia , Piperazinas/farmacologia , Ranitidina/farmacologia , Receptores Histamínicos/metabolismo , Receptores Histamínicos/fisiologia , Cicatrização/efeitos dos fármacos
12.
Inflammation ; 32(6): 387-92, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19728063

RESUMO

Fibroblasts are important cells that are involved in modulation of fibrosis after injuries. In some uncontrollable inflammatory processes, excess fibroblasts migrate around the small airway. The pathogenesis of chronic obstructive pulmonary disease is related to fibrosis around the small airways. The aim of the current study was to investigate the effect of procaterol, a second-generation beta (2)-agonist, on migration of human fetal lung fibroblasts (HFL-1) induced by human plasma fibronectin (HFn). Using the blindwell chamber technique, 10(-8) M procaterol inhibited migration of HFL-1 (control, 100%; 10(-8) M, 73.2 +/- 4.9%; n = 6, p < 0.05). The inhibitory effect of procaterol was concentration-dependent. Although a beta 2-receptor inhibitor, ICI 181551, blocked the inhibitory effect of procaterol, a beta 1-receptor inhibitor, atenolol, did not. Because a cyclic AMP-dependent protein kinase (PKA) inhibitor, KT5720, blocked the effect of procaterol, the cyclic AMP-PKA pathway may be involved in the migration inhibitory process. Procaterol, which is prescribed mainly for treatment of bronchial asthma, might be a useful drug for inhibiting lung fibrosis following injuries to the lung.


Assuntos
Inibição de Migração Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Pulmão/citologia , Pulmão/efeitos dos fármacos , Procaterol/farmacologia , Inibição de Migração Celular/fisiologia , Células Cultivadas , Fibroblastos/fisiologia , Fibrose , Humanos , Mediadores da Inflamação/farmacologia , Pulmão/fisiologia , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/patologia
13.
Pulm Pharmacol Ther ; 22(6): 487-91, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19393328

RESUMO

BACKGROUND: Excessive production of TGF-beta(1) plays a key role in the tissue remodeling or fibrotic process observed in bronchial asthma, chronic pulmonary disease (COPD), and idiopathic pulmonary fibrosis (IPF). TGF-beta(1) has been reported to decrease the intracellular glutathione level and stimulate the production of reactive oxygen species. OBJECTIVES: The aim of this study was to evaluate whether the antioxidant N-acetyl-l-cysteine (NAC) can affect TGF-beta(1)-mediated tissue remodeling in fibroblasts or modulate the production of fibronectin and vascular endothelial growth factor (VEGF) which are believed to be important mediators of tissue repair and remodeling. METHODS: To accomplish this, human fetal lung fibroblasts (HFL-1) were used to assess the effect of NAC on the TGF-beta(1)-mediated contraction of floating gels and the TGF-beta(1)-induced mediator production. In addition, the effect of NAC on the TGF-beta(1)-induced differentiation to myofibroblasts was evaluated by assessing alpha-smooth muscle actin (alpha-SMA) expression. RESULTS: NAC significantly abolished the TGF-beta(1)-augmented gel contraction (at 3mM, gel size 63.4+/-2.6% vs. 39.1+/-4.1%; p<0.01) compared with control in a concentration-dependent manner. NAC also significantly inhibited the TGF-beta(1)-augmented fibronectin (p<0.01) and VEGF (p<0.01) production in the media of both the three-dimensional gel and monolayer culture. Furthermore, NAC reversed the TGF-beta(1)-stimulated alpha-SMA expression (p<0.01). CONCLUSION: These results suggest that NAC can affect the TGF-beta(1)-induced tissue remodeling or fibrotic process in vitro.


Assuntos
Acetilcisteína/farmacologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/toxicidade , Actinas/biossíntese , Actinas/genética , Animais , Western Blotting , Células Cultivadas , Colágeno Tipo I/farmacologia , Ensaio de Imunoadsorção Enzimática , Fibronectinas/biossíntese , Fibronectinas/genética , Fibrose , Humanos , Ratos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética
14.
Am J Physiol Lung Cell Mol Physiol ; 296(6): L959-69, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19304913

RESUMO

Fibrotic diseases are characterized by the accumulation of extracellular matrix together with distortion and disruption of tissue architecture. Phosphodiesterase (PDE)4 inhibitors, by preventing the breakdown of cAMP, can inhibit fibroblast functions and may be able to mitigate tissue remodeling. Transforming growth factor (TGF)-beta1, a mediator of fibrosis, can potentially modulate cAMP by altering PGE(2) metabolism. The present study assessed whether PDE4 inhibitors functionally antagonize the profibrotic activity of fibroblasts stimulated by TGF-beta1. The PDE4 inhibitors roflumilast and rolipram both inhibited fibroblast-mediated contraction of three-dimensional collagen gels and fibroblast chemotaxis toward fibronectin in the widely studied human fetal lung fibroblast strain HFL-1 and several strains of fibroblasts from adult human lung. Roflumilast was approximately 10-fold more potent than rolipram. There was a trend for PDE4 inhibitors to inhibit more in the presence of TGF-beta1 (0.05 < P < 0.08). The effect of the PDE4 inhibitors was mediated through cAMP-stimulated protein kinase A (PKA), although a PKA-independent effect on gel contraction was also observed. The effect of PDE4 inhibitors depended on fibroblast production of PGE(2) and TGF-beta1-induced PGE(2) production. PDE4 inhibitors together with TGF-beta1 resulted in augmented PGE(2) production together with increased expression of COX mRNA and protein. The present study supports the concept that PDE4 inhibitors may attenuate fibroblast activities that can lead to fibrosis and that PDE4 inhibitors may be particularly effective in the presence of TGF-beta1-induced fibroblast stimulation.


Assuntos
Aminopiridinas/farmacologia , Benzamidas/farmacologia , Dinoprostona/metabolismo , Fibroblastos/efeitos dos fármacos , Inibidores da Fosfodiesterase 4 , Rolipram/farmacologia , Fator de Crescimento Transformador beta1/farmacologia , Adulto , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Colágeno/fisiologia , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/genética , Ciclopropanos/farmacologia , Sinergismo Farmacológico , Fibroblastos/citologia , Fibroblastos/enzimologia , Fibronectinas/metabolismo , Humanos , Pulmão/citologia , Inibidores de Fosfodiesterase/farmacologia , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/metabolismo
15.
Am J Respir Crit Care Med ; 178(3): 248-60, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18467512

RESUMO

RATIONALE: Fibroblasts are believed to be the major cells responsible for the production and maintenance of extracellular matrix. Alterations in fibroblast functional capacity, therefore, could play a role in the pathogenesis of pulmonary emphysema, which is characterized by inadequate maintenance of tissue structure. OBJECTIVES: To evaluate the hypothesis that deficient fibroblast repair characterizes cells obtained from individuals with chronic obstructive pulmonary disease (COPD) compared with control subjects. METHODS: Fibroblasts were cultured from lung tissue obtained from individuals undergoing thoracotomy and were characterized in vitro. MEASUREMENTS AND MAIN RESULTS: Fibroblasts from individuals with COPD, defined by reduced FEV(1), manifested reduced chemotaxis toward fibronectin and reduced contraction of three-dimensional collagen gels, two bioassays associated with fibroblast repair function. At least two mechanisms appear to account for these differences. Prostaglandin E (PGE), a known inhibitor of fibroblast repair functions, was produced in increased amount by fibroblasts from subjects with COPD, which also expressed increased amounts of the receptors EP2 and EP4, both of which signal through cyclic AMP. Incubation of fibroblasts with indomethacin or with the PKA inhibitor KT-5720 partially restored COPD subject fibroblast function. In addition, fibroblasts from subjects with COPD produced more transforming growth factor (TGF)-beta1, but manifested reduced response to TGF-beta1. The functional alterations in fibroblasts correlated with both lung function assessed by FEV(1) and, for the data available, with severity of emphysema assessed by Dl(CO). CONCLUSIONS: Fibroblasts from individuals with COPD have reduced capability to sustain tissue repair, which suggests that this may be one mechanism that contributes to the development of emphysema.


Assuntos
Quimiotaxia/fisiologia , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Idoso , Estudos de Casos e Controles , Células Cultivadas , Dinoprostona/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fator de Crescimento Transformador beta1/metabolismo
16.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1226-32, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18424619

RESUMO

Prostacyclin is a short-lived metabolite of arachidonic acid that is produced by several cells in the lung and prominently by endothelial cells. It increases intracellular cAMP levels activating downstream signaling thus regulating vascular mesenchymal cell functions. The alveolar wall contains a rich capillary network as well as a population of mesenchymal cells, i.e., fibroblasts. The current study evaluated the hypothesis that prostacyclin may mediate signaling between endothelial and mesenchymal cells in the alveolar wall by assessing the ability of prostacyclin analogs to modulate fibroblast release of VEGF. To accomplish this study, human lung fibroblasts were cultured in routine culture on plastic support and in three-dimensional collagen gels with or without three prostacyclin analogs, carbaprostacyclin, iloprost, and beraprost, and the production of VEGF was evaluated by ELISA and quantitative real-time PCR. Iloprost and beraprost significantly stimulated VEGF mRNA levels and protein release in a concentration-dependent manner. These effects were blocked by the adenylate cyclase inhibitor SQ-22536 and by the protein kinase A (PKA) inhibitor KT-5720 and were reproduced by a direct PKA activator but not by an activator of exchange protein directly activated by cAMP (Epac), indicating that cAMP-activated PKA signaling mediated the effect. Since VEGF serves to maintain the pulmonary microvasculature, the current study suggests that prostacyclin is part of a bidirectional signaling network between the mesenchymal and vascular cells of the alveolar wall. Prostacyclin analogs, therefore, have the potential to modulate the maintenance of the pulmonary microcirculation by driving the production of VEGF from lung fibroblasts.


Assuntos
Fibroblastos/metabolismo , Prostaglandinas I/farmacologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adenina/análogos & derivados , Adenina/farmacologia , Inibidores de Adenilil Ciclases , Adenilil Ciclases/fisiologia , Adulto , Carbazóis/farmacologia , Técnicas de Cultura de Células , Células Cultivadas , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Fibroblastos/efeitos dos fármacos , Humanos , Iloprosta/farmacologia , Pulmão/citologia , Pirróis/farmacologia , RNA Mensageiro/metabolismo , Estimulação Química
17.
Am J Respir Cell Mol Biol ; 39(3): 356-63, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18367729

RESUMO

Migration of fibroblasts plays an essential role in tissue repair after injury. Sphingosine 1-phosphate (S1P) is a multifunctional mediator released by many cells that can be released in inflammation and after injury. This study evaluated the effect of S1P on fibroblast chemotaxis toward fibronectin. S1P alone did not affect fibroblast migration, but S1P enhanced fibronectin-directed chemotaxis in a concentration-dependent manner. The effect of S1P was not mimicked by dihydro (dh) S1P or the S1P(1) receptor agonist SEW2871. S1P augmentation of fibroblast chemotaxis, however, was completely blocked by JTE-013, an S1P(2) antagonist, but not by suramin, an S1P(3) antagonist. Suppression of the S1P(2) receptor by small interfering (si)RNA also completely blocked S1P augmentation of fibroblast chemotaxis to fibronectin. S1P stimulated Rho activation and focal adhesion kinase (FAK) phosphorylation, and these were also significantly inhibited by the S1P(2) receptor antagonist (JTE-013) or by S1P(2) siRNA. Further, the potentiation of S1P signaling was blocked by the Rho-kinase inhibitor Y-27632 in a concentration-dependent manner. Inhibition of FAK with siRNA reduced basal chemotaxis toward fibronectin slightly but significantly, and almost completely blocked S1P augmented chemotaxis. These results suggest that S1P-augmented fibroblast chemotaxis toward fibronectin depends on the S1P(2) receptor and requires Rho and Rho-kinase, and FAK phosphorylation. By augmenting fibroblast recruitment, S1P has the potential to modulate tissue repair after injury. The pathways by which S1P mediates this effect, therefore, represent a potential therapeutic target to affect tissue repair and remodeling.


Assuntos
Quimiotaxia , Fibroblastos/fisiologia , Pulmão/fisiologia , Lisofosfolipídeos/fisiologia , Receptores de Lisoesfingolipídeo/fisiologia , Esfingosina/análogos & derivados , Sequência de Bases , Western Blotting , Células Cultivadas , Primers do DNA , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/farmacologia , Humanos , Imunoprecipitação , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Lisofosfolipídeos/farmacologia , Interferência de RNA , Regeneração/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Esfingosina/farmacologia , Esfingosina/fisiologia
18.
Toxicology ; 241(1-2): 58-65, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-17897767

RESUMO

Cigarette smoking is the major environmental risk factor for bladder cancer in humans. Aromatic amines, potent DNA-reactive bladder carcinogens present in cigarette smoke, contribute significantly. However, increased cell proliferation, caused by direct mitogenesis or in response to cytotoxicity, may also play a role since urothelial hyperplasia has been observed in human cigarette smokers. We examined the urothelial effects of cigarette smoke (whole body inhalation exposure (Teague) system) in female C57BL/6 mice at various times in two studies, including reversibility evaluations. In both studies, no urothelial hyperplasia was observed by light microscopy in any group. However, in study 1, the Ki-67 labeling index (LI) of the urothelium was significantly increased in the smoke exposed group compared to controls through 3 months, but was not present at 6, 9 or 12 months even with continued exposures. In the groups that discontinued smoke exposure, it returned to the same levels as controls or lower. In study 2, the bromodeoxyuridine LI was similar to controls on day 1 but significantly increased at 5 days in the smoke exposed group. In the group that discontinued smoke exposure for 2 days, the LI was increased compared to controls but not significantly. Superficial urothelial cell cytotoxicity and necrosis were detectable by scanning electron microscopy at 5 days. Changes in LI of submucosal endothelial cells generally followed those of the urothelium and effects were reversible upon cessation of exposure. The increased urothelial proliferation appeared to be due to superficial cell cytotoxicity with consequent regeneration.


Assuntos
Células Endoteliais/patologia , Células Epiteliais/patologia , Fumar/patologia , Bexiga Urinária/patologia , Animais , Antimetabólitos , Bromodesoxiuridina , Sobrevivência Celular/efeitos dos fármacos , Feminino , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Mitógenos/toxicidade , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo
19.
Am J Respir Cell Mol Biol ; 37(4): 424-30, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17575074

RESUMO

Asthmatic airway remodeling is characterized by goblet cell hyperplasia, angiogenesis, smooth muscle hypertrophy, and subepithelial fibrosis. This study evaluated whether acquired changes in fibroblast phenotype could contribute to this remodeling. Airway and parenchymal fibroblasts from control or chronically ovalbumin (OVA)-sensitized and challenged "asthmatic" mice were assessed for several functions related to repair and remodeling +/- exogenous transforming growth factor (TGF)-beta. All OVA-challenged mouse fibroblasts demonstrated augmented gel contraction (P < 0.05) and chemotaxis (P < 0.05); increased TGF-beta(1) (P < 0.05), fibronectin (P < 0.05), and vascular endothelial growth factor (P < 0.05) release; and expressed more alpha-smooth muscle actin (P < 0.05). TGF-beta(1) stimulated both control and asthmatic fibroblasts, which retained all differences from control fibroblasts for all features(P < 0.05, all comparisons). Parenchymal fibroblasts proliferated more rapidly (P < 0.05), while airway fibroblasts proliferated similarly compared with control fibroblasts (P = 0.25). Thus, in this animal model, OVA-challenged mouse fibroblasts acquire a distinct phenotype that differs from control fibroblasts. The augmented profibrotic activity and mediator release of asthmatic fibroblasts could contribute to airway remodeling in asthma.


Assuntos
Asma/patologia , Testes de Provocação Brônquica , Fibroblastos/patologia , Pulmão/patologia , Ovalbumina/farmacologia , Actinas/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibronectinas/metabolismo , Géis , Pulmão/citologia , Pulmão/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Am J Respir Cell Mol Biol ; 37(1): 121-8, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17379849

RESUMO

Apoptosis of lung structural cells is crucial in the process of normal tissue repair. Insufficient apoptosis of lung fibroblasts may contribute to the development of fibrosis. Since the CC chemokine ligand 2 (CCL2) is associated with fibrotic disease and the cytokine IL-6 blocks apoptosis in many cell types, we hypothesized that CCL2 may contribute to the development of lung fibrosis by inducing IL-6, which, in turn, inhibits fibroblast apoptosis. Fibroblasts were cultured in the presence of CCL2, which stimulated IL-6 production and mRNA expression in a concentration-dependent manner (250-1,000 ng/ml). This effect was mediated through the ERK1/2 signaling pathway. In addition, through a feedback loop, the secreted IL-6 activated the fibroblasts as evidenced by immunoblotting for phosphorylated STAT3. CCL2 reduced fibroblast apoptosis induced by staurosporin as detected by DNA content profiling (53.6 +/- 10.8%, P < 0.05) and apoptosis induced by serum starvation as detected by COMET assay (Tail moment: 36.6 +/- 9.9 of control versus 3.6 +/- 1.4 of CCL2, P < 0.01). In the presence of anti-IL-6 neutralizing antibody, however, this anti-apoptotic effect of CCL2 was eliminated. These data suggest that CCL2 mediates fibroblast survival by inhibiting apoptosis through IL-6/STAT3 signaling and provides a novel mechanism through which CCL2 may contribute to the development and maintenance of lung fibrosis.


Assuntos
Quimiocina CCL2/fisiologia , Fibroblastos/metabolismo , Interleucina-6/fisiologia , Fator de Transcrição STAT3/fisiologia , Apoptose , Sobrevivência Celular , Ensaio Cometa , Relação Dose-Resposta a Droga , Fibrose , Citometria de Fluxo , Humanos , Interleucina-6/metabolismo , Modelos Biológicos , Fosforilação , Ligação Proteica , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA