Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Respir Res ; 22(1): 30, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33517896

RESUMO

BACKGROUND: Acute exacerbations of chronic obstructive pulmonary disease (AECOPD) carry significant morbidity and mortality. AECOPD treatment remains limited. High molecular weight hyaluronan (HMW-HA) is a glycosaminoglycan sugar, which is a physiological constituent of the lung extracellular matrix and has notable anti-inflammatory and hydrating properties. RESEARCH QUESTION: We hypothesized that inhaled HMW-HA will improve outcomes in AECOPD. METHODS: We conducted a single center, randomized, placebo-controlled, double-blind study to investigate the effect of inhaled HMW-HA in patients with severe AECOPD necessitating non-invasive positive-pressure ventilation (NIPPV). Primary endpoint was time until liberation from NIPPV. RESULTS: Out of 44 screened patients, 41 were included in the study (21 for placebo and 20 for HMW-HA). Patients treated with HMW-HA had significantly shorter duration of NIPPV. HMW-HA treated patients also had lower measured peak airway pressures on the ventilator and lower systemic inflammation markers after liberation from NIPPV. In vitro testing showed that HMW-HA significantly improved mucociliary transport in air-liquid interface cultures of primary bronchial cells from COPD patients and healthy primary cells exposed to cigarette smoke extract. INTERPRETATION: Inhaled HMW-HA shortens the duration of respiratory failure and need for non-invasive ventilation in patients with AECOPD. Beneficial effects of HMW-HA on mucociliary clearance and inflammation may account for some of the effects (NCT02674880, www.clinicaltrials.gov ).


Assuntos
Ácido Hialurônico/administração & dosagem , Mediadores da Inflamação/metabolismo , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/metabolismo , Insuficiência Respiratória/tratamento farmacológico , Insuficiência Respiratória/metabolismo , Adjuvantes Imunológicos/administração & dosagem , Administração por Inalação , Idoso , Idoso de 80 Anos ou mais , Células Cultivadas , Método Duplo-Cego , Feminino , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Tempo de Internação/tendências , Masculino , Pessoa de Meia-Idade , Peso Molecular , Projetos Piloto , Poluição por Fumaça de Tabaco/efeitos adversos
2.
Int J Nanomedicine ; 9: 4093-105, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25187712

RESUMO

Multiwalled carbon nanotubes (MWCNTs) have seen increasing application in consumer products over the past decade, resulting in an increasing risk of human exposure. While numerous toxicological studies have been performed using acute high doses of various carbonaceous nanomaterials, the effects of longer-term, low doses of MWCNTs remain relatively unexplored. This study examined bronchoscopy-derived healthy human bronchial epithelial cells exposed in submerged culture to noncytotoxic doses of MWCNTs over 7 days. Under these conditions, doses as low as 3 µg/mL caused altered cell morphology, superficially resembling fibroblasts. Electrical impedance of the epithelial monolayer was greatly reduced following MWCNT exposure. However, Western blot and polymerase chain reaction showed no elevated expression of the fibroblast markers, vimentin, α-smooth muscle actin, or fibronectin, indicating that a mechanism other than epithelial-mesenchymal transition may be responsible for the changes. Phalloidin and tubulin immunostaining showed disruption of the cytoskeleton, and confocal imaging showed a reduction of the tight junction proteins, zona occludens 1 and occludin. We propose that MWCNTs interfere with the cytoskeleton of the lung epithelium, which can result in a harmful reduction in barrier function over time, even at noncytotoxic doses.


Assuntos
Brônquios/citologia , Células Epiteliais/efeitos dos fármacos , Nanotubos de Carbono/toxicidade , Morte Celular/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Impedância Elétrica , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Nanotubos de Carbono/química , Junções Íntimas/metabolismo
3.
Part Fibre Toxicol ; 11: 28, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24915862

RESUMO

BACKGROUND: In vivo studies have demonstrated the ability of multi-walled carbon nanotubes (MWCNT) to induce airway remodeling, a key feature of chronic respiratory diseases like asthma and chronic obstructive pulmonary disease. However, the mechanism leading to remodeling is poorly understood. Particularly, there is limited insight about the role of airway epithelial injury in these changes. OBJECTIVES: We investigated the mechanism of MWCNT-induced primary human bronchial epithelial (HBE) cell injury and its contribution in inducing a profibrotic response. METHODS: Primary HBE cells were exposed to thoroughly characterized MWCNTs (1.5-24 µg/mL equivalent to 0.37-6.0 µg/cm2) and MRC-5 human lung fibroblasts were exposed to 1:4 diluted conditioned medium from these cells. Flow cytometry, ELISA, immunostainings/immunoblots and PCR analyses were employed to study cellular mechanisms. RESULTS: MWCNT induced NLRP3 inflammasome dependent pyroptosis in HBE cells in a time- and dose-dependent manner. Cell death and cytokine production were significantly reduced by antioxidants, siRNA to NLRP3, a caspase-1 inhibitor (z-WEHD-FMK) or a cathepsin B inhibitor (CA-074Me). Conditioned medium from MWCNT-treated HBE cells induced significant increase in mRNA expression of pro-fibrotic markers (TIMP-1, Tenascin-C, Procollagen 1, and Osteopontin) in human lung fibroblasts, without a concomitant change in expression of TGF-beta. Induction of pro-fibrotic markers was significantly reduced when IL-1ß, IL-18 and IL-8 neutralizing antibodies were added to the conditioned medium or when conditioned medium from NLRP3 siRNA transfected HBE cells was used. CONCLUSIONS: Taken together these results demonstrate induction of a NLRP3 inflammasome dependent but TGF-beta independent pro-fibrotic response after MWCNT exposure.


Assuntos
Células Epiteliais/patologia , Fibroblastos/patologia , Inflamação/patologia , Nanotubos de Carbono/toxicidade , Fibrose Pulmonar/patologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Meios de Cultivo Condicionados , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Inflamação/induzido quimicamente , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Espécies Reativas de Oxigênio/metabolismo , Suspensões
4.
Free Radic Biol Med ; 60: 98-106, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23376232

RESUMO

Previous studies focused on catalyzed oxidation of (bi)sulfite, leading to the formation of the reactive sulfur trioxide ((•)SO3(-)), peroxymonosulfate ((-)O3SOO(•)), and sulfate (SO4(•-)) anion radicals, which can damage target proteins and oxidize them to protein radicals. It is known that these very reactive sulfur- and oxygen-centered radicals can be formed by oxidation of (bi)sulfite by peroxidases. Myeloperoxidase (MPO), an abundant heme protein secreted from activated neutrophils that play a central role in host defense mechanisms, allergic reactions, and asthma, is a likely candidate for initiating the respiratory damage caused by sulfur dioxide. The objective of this study was to examine the oxidative damage caused by (bi)sulfite-derived free radicals in human neutrophils through formation of protein radicals. We used immuno-spin trapping and confocal microscopy to study the protein oxidations driven by sulfite-derived radicals. We found that the presence of sulfite can cause MPO-catalyzed oxidation of MPO to a protein radical in phorbol 12-myristate 13-acetate-activated human neutrophils. We trapped the MPO-derived radicals in situ using the nitrone spin trap 5,5-dimethyl-1-pyrroline N-oxide and detected them immunologically as nitrone adducts in cells. Our present study demonstrates that myeloperoxidase initiates (bi)sulfite oxidation leading to MPO radical damage, possibly leading to (bi)sulfite-exacerbated allergic reactions.


Assuntos
Radicais Livres/toxicidade , Hipersensibilidade/metabolismo , Neutrófilos/metabolismo , Peroxidase/metabolismo , Sulfitos/toxicidade , Radicais Livres/metabolismo , Humanos , Hipersensibilidade/etiologia , Hipersensibilidade/patologia , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Peroxidase/efeitos dos fármacos , Peróxidos/química , Peróxidos/metabolismo , Peróxidos/toxicidade , Ésteres de Forbol/farmacologia , Proteínas/metabolismo , Detecção de Spin , Sulfatos/química , Sulfatos/metabolismo , Sulfatos/toxicidade , Sulfitos/metabolismo , Óxidos de Enxofre/química , Óxidos de Enxofre/metabolismo , Óxidos de Enxofre/toxicidade
5.
ACS Nano ; 6(7): 5820-9, 2012 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-22717232

RESUMO

Cerium dioxide nanoparticles (CeO(2) NPs) have diversified industrial uses, and novel therapeutic applications are actively being pursued. There is a lack of mechanistic data concerning the effects of CeO(2) NPs on primary human cells. We aimed at characterizing the cytotoxic effects of CeO(2) NPs in human peripheral blood monocytes. CeO(2) NPs and their suspensions were thoroughly characterized, including using transmission electron microscopy (TEM), dynamic light scattering, and zeta potential analysis. Blood from healthy human volunteers was drawn through phlebotomy, and CD14+ cells were isolated. Cells were exposed to CeO(2) NPs (0.5-10 µg/mL) for 20 or 40 h, and mechanisms of cell injury were studied. TEM revealed that CeO(2) NPs are internalized by monocytes and are found either in vesicles or free in the cytoplasm. CeO(2) NP exposure leads to decrease in cell viability, and treated cells exhibit characteristic hallmarks of apoptosis (activation of Bax, loss of mitochondrial membrane potential, DNA fragmentation). CeO(2) NP toxicity is caused by mitochondrial damage and overexpression of apoptosis inducing factor, but is not due to caspase activation or reactive oxygen species production. Moreover, CeO(2) NP exposure leads to autophagy, which is further increased after pharmacological inhibition of tumor suppressor protein p53. Inhibition of autophagy partially reverses cell death by CeO(2) NPs. It is concluded that CeO(2) NPs are toxic to primary human monocytes at relatively low doses.


Assuntos
Cério/toxicidade , Nanopartículas Metálicas/toxicidade , Monócitos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Humanos , Técnicas In Vitro , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Nanopartículas Metálicas/ultraestrutura , Microscopia Eletrônica de Transmissão , Monócitos/metabolismo , Monócitos/patologia , Nanotecnologia , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Proteína X Associada a bcl-2/metabolismo
6.
Int J Nanomedicine ; 7: 1387-97, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22457596

RESUMO

BACKGROUND: Cerium dioxide (CeO(2)) nanoparticles have potential therapeutic applications and are widely used for industrial purposes. However, the effects of these nanoparticles on primary human cells are largely unknown. The ability of nanoparticles to exacerbate pre-existing inflammatory disorders is not well documented for engineered nanoparticles, and is certainly lacking for CeO(2) nanoparticles. We investigated the inflammation-modulating effects of CeO(2) nanoparticles at noncytotoxic concentrations in human peripheral blood monocytes. METHODS: CD14(+) cells were isolated from peripheral blood samples of human volunteers. Cells were exposed to either 0.5 or 1 µg/mL of CeO(2) nanoparticles over a period of 24 or 48 hours with or without lipopolysaccharide (10 ng/mL) prestimulation. Modulation of the inflammatory response was studied by measuring secreted tumor necrosis factor-alpha, interleukin-1beta, macrophage chemotactic protein-1, interferon-gamma, and interferon gamma-induced protein 10. RESULTS: CeO(2) nanoparticle suspensions were thoroughly characterized using dynamic light scattering analysis (194 nm hydrodynamic diameter), zeta potential analysis (-14 mV), and transmission electron microscopy (irregular-shaped particles). Transmission electron microscopy of CD14(+) cells exposed to CeO(2) nanoparticles revealed that these nanoparticles were efficiently internalized by monocytes and were found either in vesicles or free in the cytoplasm. However, no significant differences in secreted cytokine profiles were observed between CeO(2) nanoparticle-treated cells and control cells at noncytotoxic doses. No significant effects of CeO(2) nanoparticle exposure subsequent to lipopolysaccharide priming was observed on cytokine secretion. Moreover, no significant difference in lipopolysaccharide-induced cytokine production was observed after exposure to CeO(2) nanoparticles followed by lipopolysaccharide exposure. CONCLUSION: CeO(2) nanoparticles at noncytotoxic concentrations neither modulate pre-existing inflammation nor prime for subsequent exposure to lipopolysaccharides in human monocytes from healthy subjects.


Assuntos
Cério/farmacologia , Nanopartículas Metálicas/química , Monócitos/efeitos dos fármacos , Análise de Variância , Sobrevivência Celular , Células Cultivadas , Cério/química , Cério/farmacocinética , Citocinas/metabolismo , Histocitoquímica , Humanos , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Lipopolissacarídeos/efeitos adversos , Monócitos/imunologia , Monócitos/patologia , Nanomedicina , Fagocitose
7.
Free Radic Biol Med ; 52(8): 1264-71, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22326772

RESUMO

The objective of this study was to determine the effect of (bi)sulfite (hydrated sulfur dioxide) on human neutrophils and the ability of these immune cells to produce reactive free radicals due to (bi)sulfite oxidation. Myeloperoxidase (MPO) is an abundant heme protein in neutrophils that catalyzes the formation of cytotoxic oxidants implicated in asthma and inflammatory disorders. In this study sulfite ((•)SO(3)(-)) and sulfate (SO(4)(•-)) anion radicals are characterized with the ESR spin-trapping technique using 5,5-dimethyl-1-pyrroline N-oxide (DMPO) in the reaction of (bi)sulfite oxidation by human MPO and human neutrophils via sulfite radical chain reaction chemistry. After treatment with (bi)sulfite, phorbol 12-myristate 13-acetate-stimulated neutrophils produced DMPO-sulfite anion radical, -superoxide, and -hydroxyl radical adducts. The last adduct probably resulted, in part, from the conversion of DMPO-sulfate to DMPO-hydroxyl radical adduct via a nucleophilic substitution reaction of the radical adduct. This anion radical (SO(4)(•-)) is highly reactive and, presumably, can oxidize target proteins to protein radicals, thereby initiating protein oxidation. Therefore, we propose that the potential toxicity of (bi)sulfite during pulmonary inflammation or lung-associated diseases such as asthma may be related to free radical formation.


Assuntos
Espectroscopia de Ressonância de Spin Eletrônica/métodos , Radicais Livres/metabolismo , Ativação de Neutrófilo , Neutrófilos/metabolismo , Sulfitos/metabolismo , Células Cultivadas , Humanos , Neutrófilos/enzimologia , Oxirredução , Peroxidase/antagonistas & inibidores , Peroxidase/metabolismo , Marcadores de Spin
8.
Mol Endocrinol ; 22(9): 2099-115, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18562628

RESUMO

We have studied nonsteroidal ligands of the human androgen receptor (hAR) and have shown elsewhere that when photoactivated by visible light they collide with O2 to yield singlet oxygens (1O2) in vitro. Here we report cell killing after brief light activation (405 nm) of 1,2,3,4-tetrahydro-2,2-dimethyl-6-(trifluoromethyl)-8-pyridono[5,6-g]quinoline (TDPQ) in human prostate tumor cells. TDPQ/AR complexes were required for the death response because AR-positive LNCaP cells were killed, whereas AR-negative PC-3 cells were resistant. Excess dihydrotestosterone (DHT) blocked the TDPQ effect when the two were added together; irradiation of cells containing DHT alone had no effect. When LNCaP AR expression was suppressed using small interfering oligonucleotides targeting AR, photocytotoxicity was diminished. Conversely, stable transfection of hAR into PC-3 cells made the cells photosensitive to TDPQ. Similar results were obtained using a structural isomer of TDPQ, and also the synthetic steroidal AR ligand R1881. Cell death occurred via apoptosis as demonstrated by annexin V immunostaining, nuclear condensation, and caspase inhibition. Death involved oxidative stress, because it was prevented by addition of the antioxidant ascorbic acid during photoactivation. Detection of elevated levels of 8-hydroxy-2'-deoxyguanosine in nuclei of irradiated cells indicated oxidative DNA damage. Apoptosis spread into adjacent nonirradiated cells by direct cell-cell contacts, indicative of a bystander effect. Other photoactivatable ligands are described, implying a general method for ablation of cells bearing specific nuclear hormone receptors.


Assuntos
Apoptose/fisiologia , Receptores Androgênicos/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Antagonistas de Receptores de Andrógenos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Sequência de Bases , Linhagem Celular Tumoral , Dano ao DNA , Di-Hidrotestosterona/farmacologia , Humanos , Ligantes , Masculino , Metribolona/farmacologia , Metribolona/efeitos da radiação , Modelos Biológicos , Estresse Oxidativo , Fotobiologia , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Piridinas/farmacologia , Piridinas/efeitos da radiação , Quinolonas/farmacologia , Quinolonas/efeitos da radiação , RNA Interferente Pequeno/genética , Receptores Androgênicos/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
9.
Am J Pathol ; 167(5): 1221-9, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16251407

RESUMO

The signal transducer and activator of transcription (Stat)-1 mediates growth arrest and apoptosis. We postulated that lung fibrosis characterized by excessive proliferation of lung fibroblasts would be enhanced in Stat1-deficient (Stat1-/-) mice. Two weeks after bleomycin aspiration (3 U/kg), Stat1-/- mice exhibited a more severe fibroproliferative response and significantly elevated total lung collagen compared to wild-type mice. Growth factors [epidermal growth factor (EGF) or platelet-derived growth factor (PDGF)] enhanced [3H]thymidine uptake in lung fibroblasts isolated from Stat1-/- mice compared to wild-type mice. Interferon (IFN)-gamma, which signals growth arrest via Stat1, inhibited EGF- or PDGF-stimulated mitogenesis in wild-type fibroblasts but enhanced [3H]thymidine uptake in Stat1-/- fibroblasts. Moreover, IFN-gamma treatment in the absence of growth factors induced a concentration-dependent increase in [3H]thymidine uptake in Stat1-/- but not wild-type fibroblasts. Mitogen-activated protein kinase (ERK-1/2) phosphorylation in response to PDGF or EGF did not differ among Stat1-/- and wild-type fibroblasts. However, Stat3 phosphorylation induced by PDGF, EGF, or IFN-gamma increased twofold in Stat1-/- fibroblasts compared to wild-type fibroblasts. Our findings indicate that Stat1-/- mice are more susceptible to bleomycin-induced lung fibrosis than wild-type mice due to 1) enhanced fibroblast proliferation in response to growth factors (EGF and PDGF), 2) stimulation of fibroblast growth by a Stat1-independent IFN-gamma signaling pathway, and 3) increased activation of Stat3.


Assuntos
Pulmão/patologia , Fibrose Pulmonar/patologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/fisiologia , Animais , Bleomicina/toxicidade , Western Blotting , Proliferação de Células , Células Cultivadas , Colágeno/análise , Modelos Animais de Doenças , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/patologia , Inibidores do Crescimento/farmacologia , Hidroxiprolina/análise , Interferon gama/farmacologia , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação , Fator de Crescimento Derivado de Plaquetas/farmacologia , Fibrose Pulmonar/genética , Fibrose Pulmonar/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Timidina/metabolismo
10.
FASEB J ; 19(10): 1374-6, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15923396

RESUMO

In cystic fibrosis and chronic bronchitis, airways are chronically injured by exposure to neutrophil elastase (NE). We sought to identify factors required for epithelial repair following NE exposure. Normal human bronchial epithelial cells were treated with NE (50 nM, 22 h) or control vehicle. Following NE treatment, we found a marked and sustained decrease in epithelial proliferation as detected by Ki67 immunostaining. 3H-thymidine incorporation was also initially depressed but increased over 72 h in NE-treated cells, which suggests that DNA synthesis constitutes an early repair process following NE exposure. We hypothesized that ErbB2 receptor tyrosine kinase, a regulator of cancer cell proliferation, was required for epithelial DNA synthesis following NE exposure. Immediately following NE treatment, by flow cytometry analysis, we found a decrease in ErbB2 surface expression. Protein levels of the full-length 185 kD ErbB2 receptor significantly decreased following NE treatment and smaller ErbB2-positive bands, ranging in size from 23 to 40 kD, appeared, which suggests that NE caused ErbB2 degradation. By real-time RT-PCR analysis, we found no change in ErbB2 mRNA expression following NE treatment, which suggests that changes in ErbB2 protein levels were regulated at the post-translational level. Following NE treatment, full-length 185 kD ErbB2 levels increased to pretreatment levels, correlating with the increase in thymidine incorporation during the same time period. Importantly, inhibition of ErbB2 activity with AG825 (5 microM) or Herceptin (3.1 microM), an ErbB2-neutralizing antibody, blocked thymidine incorporation only in NE-treated cells. These results suggest ErbB2 is a critical factor for epithelial recovery following NE exposure.


Assuntos
Brônquios/efeitos dos fármacos , Elastase de Leucócito/toxicidade , Receptor ErbB-2/fisiologia , Benzotiazóis , Brônquios/citologia , Brônquios/fisiologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Receptores ErbB/metabolismo , Humanos , Receptor ErbB-2/antagonistas & inibidores , Tirfostinas/farmacologia
11.
FASEB J ; 18(10): 1132-4, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15155567

RESUMO

Peribronchiolar fibrosis is a prominent feature of airway remodeling in asthma and involves fibroblast growth and collagen deposition. Interleukin-13 (IL-13), a T-helper 2 cytokine, is a key mediator of airway remodeling in asthma, yet the mechanism through which IL-13 promotes fibroblast growth has not been investigated. In this study, we show that IL-13 stimulates the mitogenesis of mouse, rat, and human lung fibroblasts through release of a soluble mitogen that we identified as PDGF-AA. The IL-13-induced growth of human lung fibroblasts was attenuated by an anti-PDGF-AA neutralizing antibody, and IL-13 stimulated human lung fibroblasts to secrete PDGF-AA. Fibroblasts derived from mouse embryos possessing the lethal Patch mutation, which lack the PDGF-Ralpha, showed no mitogenic response to IL-13. However, Patch cells did exhibit IL-13-induced STAT-6 phosphorylation. Stable transfection of the PDGF-Ralpha into Patch cells restored the growth response to PDGF-AA and IL-13. Through the use of lung fibroblasts from STAT-6-deficient mice, we showed that IL-13-induced PDGF-AA release is STAT-6 dependent, but PDGF-AA-induced growth is STAT-6 independent. Finally, we showed that IL-1beta enhanced IL-13-induced mitogenesis of rat lung fibroblasts through up-regulation of the PDGF-Ralpha. Our findings indicate that IL-13 acts in synergy with IL-1beta to stimulate growth by coordinately up-regulating PDGF-AA and the PDGF-Ralpha, respectively.


Assuntos
Interleucina-13/fisiologia , Interleucina-1/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Animais , Becaplermina , Divisão Celular/efeitos dos fármacos , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Meios de Cultivo Condicionados/farmacologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose , Humanos , Interleucina-1/genética , Interleucina-1/farmacologia , Interleucina-13/genética , Interleucina-13/farmacologia , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Ratos , Ratos Sprague-Dawley , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/biossíntese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/deficiência , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Fator de Transcrição STAT6 , Transativadores/fisiologia , Fator de Crescimento Transformador alfa/deficiência
12.
Free Radic Biol Med ; 35(8): 845-55, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-14556849

RESUMO

Vanadium compounds present in air pollution particulate matter activate signal transduction pathways in pulmonary cell types leading to pathological outcomes including aberrant cell proliferation, apoptosis, and cytokine expression. Vanadium has been proposed to activate transcription factors via the generation of hydrogen peroxide (H2O2). We investigated the mechanisms through which vanadium pentoxide (V2O5), the major form of vanadium released from the industrial burning of fuel oil, activated the signal transducer and activator of transcription (STAT)-1. V2O5-induced STAT-1 activation was blocked by catalase and N-acetyl-L-cysteine (NAC), suggesting vanadium-induced generation of H2O2. Surprisingly, however, V2O5 did not increase H2O2 levels released by rat lung myofibroblasts into cell culture supernatants. Instead, these quiescent myofibroblasts spontaneously released micromolar concentrations of H2O2, and the addition of V2O5 reduced H2O2 levels in cell culture supernatants within minutes. V2O5 suppressed H2O2 for as long as 24 h. Differences in the temporal activation of STAT-1 and p38 MAPK were observed following V2O5 or H2O2 treatment, and STAT-1 activation by V2O5 or H2O2 was attenuated by an inhibitor of the EGF receptor tyrosine kinase (AG1478) or p38 MAPK (SB203580). The phosphorylation of p38 MAPK by V2O5 was inhibited by NAC and catalase, yet the EGF receptor inhibitor AG1478 had no effect on V2O5-induced p38 MAPK activation. Collectively, our findings support the novel hypothesis that H2O2 spontaneously generated by myofibroblasts fuels vanadium-induced activation of STAT-1. Moreover, p38 MAPK and EGF receptor activation are required for V2O5-induced STAT-1 activation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Peróxido de Hidrogênio/metabolismo , Pulmão/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transativadores/metabolismo , Compostos de Vanádio/farmacologia , Acetilcisteína/farmacologia , Animais , Carbono/efeitos adversos , Catalase/farmacologia , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Fator de Transcrição STAT1 , Proteínas Quinases p38 Ativadas por Mitógeno
13.
Am J Physiol Lung Cell Mol Physiol ; 284(5): L774-82, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12676768

RESUMO

Vanadium pentoxide (V(2)O(5)) is a transition metal derived from the burning of petrochemicals that causes airway fibrosis and remodeling. Vanadium compounds activate many intracellular signaling pathways via the generation of hydrogen peroxide (H(2)O(2)) or other reactive oxygen species. In this study, we investigated the regulation of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in human lung fibroblasts after V(2)O(5) treatment. V(2)O(5)-induced HB-EGF mRNA expression was abolished by N-acetyl-l-cysteine, suggesting an oxidant-mediated effect. Exogenous H(2)O(2) (>10 microM) mimicked the effect of V(2)O(5) in upregulating HB-EGF expression. Fibroblasts spontaneously released low levels of H(2)O(2) (1-2 microM), and the addition of V(2)O(5) depleted the endogenous H(2)O(2) pool within minutes. V(2)O(5) caused a subsequent increase of H(2)O(2) into the culture medium at 12 h. However, the burst of V(2)O(5)-induced H(2)O(2) occurred after V(2)O(5)-induced HB-EGF mRNA expression at 3 h, indicating that the V(2)O(5)-stimulated H(2)O(2) burst did not mediate HB-EGF expression. Either V(2)O(5) or H(2)O(2) activated ERK-1/2 and p38 MAP kinase. Inhibitors of the ERK-1/2 pathway (PD-98059) or p38 MAP kinase (SB-203580) significantly reduced either V(2)O(5)- or H(2)O(2)-induced HB-EGF expression. These data indicate that vanadium upregulates HB-EGF via ERK and p38 MAP kinases. The induction of HB-EGF is not related to a burst of H(2)O(2) in V(2)O(5) treated cells, yet the action of V(2)O(5) in upregulating HB-EGF is oxidant dependent and could be due to the reaction of V(2)O(5) with endogenous H(2)O(2).


Assuntos
Fator de Crescimento Epidérmico/genética , Fibroblastos/enzimologia , Pulmão/citologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Compostos de Vanádio/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peróxido de Hidrogênio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fibrose Pulmonar/metabolismo , RNA Mensageiro/análise , Proteínas Quinases p38 Ativadas por Mitógeno
14.
Am J Respir Cell Mol Biol ; 27(6): 759-65, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12444037

RESUMO

Myofibroblast proliferation is a central feature of pulmonary fibrogenesis. Several growth factors, including platelet-derived growth factor (PDGF) and epidermal growth factor (EGF), stimulate myofibroblast growth by activating extracellular signal regulated kinases 1 and 2 (ERK1/2). In this report, we demonstrate that PDGF-BB and EGF also activate the p38 mitogen-activated protein (MAP) kinase. Inhibition of p38 activity with the pyridinylimidazole compound SB203580 enhanced both PDGF-BB and EGF-stimulated DNA synthesis in rat lung myofibroblasts. ERK1/2 phosphorylation in response to either PDGF-BB or EGF treatment was significantly increased by pretreatment of cells with SB203580. We also demonstrated that ERK1/2-induced phosphorylation of PHAS-1 substrate was enhanced by inhibition of p38 MAP kinase with SB203580. However, SB203580 did not significantly increase growth factor-induced activation of MEK, the upstream kinase that phosphorylates ERK1/2. p38 MAP kinase was co-immunoprecipitated with ERK-1/2 following growth factor stimulation. Collectively, these data demonstrate that p38 MAP kinase activation negatively regulates PDGF- and EGF-mediated growth responses by directly interacting with ERK1/2 and suppressing its phosphorylation.


Assuntos
Fibroblastos/citologia , Fibroblastos/enzimologia , Substâncias de Crescimento/farmacologia , Pulmão/citologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Inibidores Enzimáticos/farmacologia , Fator de Crescimento Epidérmico/farmacologia , Imidazóis/farmacologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/análise , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/análise , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Fator de Crescimento Derivado de Plaquetas/farmacologia , Testes de Precipitina , Proteínas Serina-Treonina Quinases/metabolismo , Piridinas/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Quinases p38 Ativadas por Mitógeno
15.
Am J Pathol ; 161(2): 459-70, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12163371

RESUMO

The cyclooxygenase (COX)-2 enzyme has been implicated as an important mediator of pulmonary fibrosis. In this study, the lung fibrotic responses were investigated in COX-1 or COX-2-deficient (-/-) mice following vanadium pentoxide (V(2)O(5)) exposure. Lung histology was normal in saline-instilled wild-type and COX-deficient mice. COX-2(-/-), but not COX-1(-/-) or wild-type mice, exhibited severe inflammatory responses by 3 days following V(2)O(5) exposure and developed pulmonary fibrosis 2 weeks post-V(2)O(5) exposure. Western blot analysis and immunohistochemistry showed that COX-1 protein was present in type 2 epithelial cells, bronchial epithelial cells, and airway smooth muscle cells of saline or V(2)O(5)-exposed wild-type and COX-2(-/-) mice. COX-2 protein was present in Clara cells of wild-type and COX-1(-/-) terminal bronchioles and was strongly induced 24 hours after V(2)O(5) exposure. Prostaglandin (PG) E(2) levels in the bronchoalveolar lavage (BAL) fluid from wild-type and COX-1(-/-) mice were significantly up-regulated by V(2)O(5) exposure within 24 hours, whereas PGE(2) was not up-regulated in COX-2(-/-) BAL fluid. Tumor necrosis factor-alpha was elevated in the BAL fluid from all genotypes after V(2)O(5) exposure, but was significantly and chronically elevated in the BAL fluid from COX-2(-/-) mice above wild-type or COX-1(-/-) mice. These findings indicate that the COX-2 enzyme is protective against pulmonary fibrogenesis, and we suggest that COX-2 generation of PGE(2) is an important factor in resolving inflammation.


Assuntos
Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Fibrose Pulmonar/genética , Animais , Ciclo-Oxigenase 2 , Dinoprostona/biossíntese , Feminino , Predisposição Genética para Doença , Imuno-Histoquímica , Isoenzimas/deficiência , Masculino , Camundongos , Camundongos Knockout , Prostaglandina-Endoperóxido Sintases/deficiência , Fibrose Pulmonar/etiologia , Regulação para Cima , Compostos de Vanádio/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...