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











Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 29(2): 335-347.e5, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30318339

RESUMO

Urban particulate matter air pollution induces the release of pro-inflammatory cytokines including interleukin-6 (IL-6) from alveolar macrophages, resulting in an increase in thrombosis. Here, we report that metformin provides protection in this murine model. Treatment of mice with metformin or exposure of murine or human alveolar macrophages to metformin prevented the particulate matter-induced generation of complex III mitochondrial reactive oxygen species, which were necessary for the opening of calcium release-activated channels (CRAC) and release of IL-6. Targeted genetic deletion of electron transport or CRAC channels in alveolar macrophages in mice prevented particulate matter-induced acceleration of arterial thrombosis. These findings suggest metformin as a potential therapy to prevent some of the premature deaths attributable to air pollution exposure worldwide.


Assuntos
Poluição do Ar/efeitos adversos , Pneumopatias/tratamento farmacológico , Macrófagos Alveolares/metabolismo , Metformina/farmacologia , Mitocôndrias/metabolismo , Material Particulado/toxicidade , Trombose/tratamento farmacológico , Animais , Linhagem Celular , Citocinas/metabolismo , Transporte de Elétrons , Humanos , Interleucina-6/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Espécies Reativas de Oxigênio/metabolismo
2.
Am J Respir Crit Care Med ; 199(12): 1517-1536, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30554520

RESUMO

Rationale: The contributions of diverse cell populations in the human lung to pulmonary fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal changes within individual cell populations during pulmonary fibrosis that are important for disease pathogenesis. Objectives: To determine whether single-cell RNA sequencing can reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other cell types in lung tissue from subjects with pulmonary fibrosis compared with control subjects. Methods: We performed single-cell RNA sequencing on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and on one bronchoscopic cryobiospy sample from a patient with idiopathic pulmonary fibrosis. We validated these data using in situ RNA hybridization, immunohistochemistry, and bulk RNA-sequencing on flow-sorted cells from 22 additional subjects. Measurements and Main Results: We identified a distinct, novel population of profibrotic alveolar macrophages exclusively in patients with fibrosis. Within epithelial cells, the expression of genes involved in Wnt secretion and response was restricted to nonoverlapping cells. We identified rare cell populations including airway stem cells and senescent cells emerging during pulmonary fibrosis. We developed a web-based tool to explore these data. Conclusions: We generated a single-cell atlas of pulmonary fibrosis. Using this atlas, we demonstrated heterogeneity within alveolar macrophages and epithelial cells from subjects with pulmonary fibrosis. These results support the feasibility of discovery-based approaches using next-generation sequencing technologies to identify signaling pathways for targeting in the development of personalized therapies for patients with pulmonary fibrosis.


Assuntos
Células Cultivadas/patologia , Células Epiteliais/patologia , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Análise de Sequência de RNA , Células-Tronco/patologia , Transcriptoma , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino
3.
Am J Respir Cell Mol Biol ; 52(4): 503-12, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25188360

RESUMO

Laminins are heterotrimeric proteins that are secreted by the alveolar epithelium into the basement membrane, and their expression is altered in extracellular matrices from patients with pulmonary fibrosis. In a small number of patients with pulmonary fibrosis, we found that the normal basement membrane distribution of the α3 laminin subunit was lost in fibrotic regions of the lung. To determine if these changes play a causal role in the development of fibrosis, we generated mice lacking the α3 laminin subunit specifically in the lung epithelium by crossing mice expressing Cre recombinase driven by the surfactant protein C promoter (SPC-Cre) with mice expressing floxed alleles encoding the α3 laminin gene (Lama3(fl/fl)). These mice exhibited no developmental abnormalities in the lungs up to 6 months of age, but, compared with control mice, had worsened mortality, increased inflammation, and increased fibrosis after the intratracheal administration of bleomycin. Similarly, the severity of fibrosis induced by an adenovirus encoding an active form of transforming growth factor-ß was worse in mice deficient in α3 laminin in the lung. Taken together, our results suggest that the loss of α3 laminin in the lung epithelium does not affect lung development, but plays a causal role in the development of fibrosis in response to bleomycin or adenovirally delivered transforming growth factor-ß. Thus, we speculate that the loss of the normal basement membrane organization of α3 laminin that we observe in fibrotic regions from the lungs of patients with pulmonary fibrosis contributes to their disease progression.


Assuntos
Laminina/metabolismo , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Bleomicina , Humanos , Pulmão/patologia , Camundongos Transgênicos , Alvéolos Pulmonares/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fator de Crescimento Transformador beta/fisiologia
4.
FEBS Lett ; 588(24): 4686-93, 2014 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-25447523

RESUMO

ß2-Adrenergic agonists have been shown to regulate Na,K-ATPase in the alveolar epithelium by recruiting Na,K-ATPase-containing vesicles to the plasma membrane of alveolar epithelial cells (AEC). Here, we provide evidence that ß2-agonists induce store-operated calcium entry (SOCE) in AECs. This calcium entry is necessary for ß2-agonist-induced recruitment of Na,K-ATPase to the plasma membrane of AECs. Specifically, we show that ß2-agonists induce SOCE via stromal interaction molecule 1 (STIM1)-associated calcium release-activated calcium (CRAC) channels. We also demonstrate that the magnitude of SOCE affects the abundance of Na,K-ATPase at the plasma membrane of AECs.


Assuntos
Antagonistas de Receptores Adrenérgicos beta 2/farmacologia , Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Adenilil Ciclases/metabolismo , Albuterol/farmacologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Transporte Proteico/efeitos dos fármacos , Alvéolos Pulmonares/citologia , Ratos , Molécula 1 de Interação Estromal
5.
J Clin Invest ; 124(7): 2935-46, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24865431

RESUMO

Acute exposure to particulate matter (PM) air pollution causes thrombotic cardiovascular events, leading to increased mortality rates; however, the link between PM and cardiovascular dysfunction is not completely understood. We have previously shown that the release of IL-6 from alveolar macrophages is required for a prothrombotic state and acceleration of thrombosis following exposure to PM. Here, we determined that PM exposure results in the systemic release of catecholamines, which engage the ß2-adrenergic receptor (ß2AR) on murine alveolar macrophages and augment the release of IL-6. In mice, ß2AR signaling promoted the development of a prothrombotic state that was sufficient to accelerate arterial thrombosis. In primary human alveolar macrophages, administration of a ß2AR agonist augmented IL-6 release, while the addition of a beta blocker inhibited PM-induced IL-6 release. Genetic loss or pharmacologic inhibition of the ß2AR on murine alveolar macrophages attenuated PM-induced IL-6 release and prothrombotic state. Furthermore, exogenous ß2AR agonist therapy further augmented these responses in alveolar macrophages through generation of mitochondrial ROS and subsequent increase of adenylyl cyclase activity. Together, these results link the activation of the sympathetic nervous system by ß2AR signaling with metabolism, lung inflammation, and an enhanced susceptibility to thrombotic cardiovascular events.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/administração & dosagem , Agonistas de Receptores Adrenérgicos beta 2/efeitos adversos , Interleucina-6/biossíntese , Material Particulado/administração & dosagem , Material Particulado/efeitos adversos , Trombose/etiologia , Adenilil Ciclases/biossíntese , Animais , Antitrombina III/biossíntese , Líquido da Lavagem Broncoalveolar/química , Catecolaminas/biossíntese , Colforsina/administração & dosagem , Humanos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/fisiologia , Masculino , Camundongos , Camundongos Knockout , Peptídeo Hidrolases/biossíntese , Propranolol/administração & dosagem , Propranolol/efeitos adversos , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta/deficiência , Receptores Adrenérgicos beta/genética , Receptores Adrenérgicos beta 1/deficiência , Receptores Adrenérgicos beta 1/genética
6.
Elife ; 3: e02242, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24843020

RESUMO

Recent epidemiological and laboratory-based studies suggest that the anti-diabetic drug metformin prevents cancer progression. How metformin diminishes tumor growth is not fully understood. In this study, we report that in human cancer cells, metformin inhibits mitochondrial complex I (NADH dehydrogenase) activity and cellular respiration. Metformin inhibited cellular proliferation in the presence of glucose, but induced cell death upon glucose deprivation, indicating that cancer cells rely exclusively on glycolysis for survival in the presence of metformin. Metformin also reduced hypoxic activation of hypoxia-inducible factor 1 (HIF-1). All of these effects of metformin were reversed when the metformin-resistant Saccharomyces cerevisiae NADH dehydrogenase NDI1 was overexpressed. In vivo, the administration of metformin to mice inhibited the growth of control human cancer cells but not those expressing NDI1. Thus, we have demonstrated that metformin's inhibitory effects on cancer progression are cancer cell autonomous and depend on its ability to inhibit mitochondrial complex I.DOI: http://dx.doi.org/10.7554/eLife.02242.001.


Assuntos
Carcinogênese , Complexo I de Transporte de Elétrons/efeitos dos fármacos , Metformina/farmacologia , Neoplasias/enzimologia , Linhagem Celular Tumoral , Humanos , Neoplasias/patologia
7.
PLoS One ; 7(4): e35788, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22536437

RESUMO

RATIONALE: HMG-CoA reductase inhibitors such as rosuvastatin may have immunomodulatory and anti-inflammatory effects that may reduce the severity of influenza A infection. We hypothesized that rosuvastatin would decrease viral replication, attenuate lung injury, and improve mortality following influenza A infection in mice. METHODS: C57Bl/6 mice were treated daily with rosuvastatin (10 mg/kg/day) supplemented in chow (or control chow) beginning three days prior to infection with either A//Udorn/72 [H3N2] or A/WSN/33 [H1N1] influenza A virus (1×10(5) pfu/mouse). Plaque assays were used to examine the effect of rosuvastatin on viral replication in vitro and in the lungs of infected mice. We measured cell count with differential, protein and cytokines in the bronchoalveolar lavage (BAL) fluid, histologic evidence of lung injury, and wet-to-dry ratio on Day 1, 2, 4, and 6. We also recorded daily weights and mortality. RESULTS: The administration of rosuvastatin had no effect on viral clearance of influenza A after infection. Weight loss, lung inflammation and lung injury severity were similar in the rosuvastatin and control treated mice. In the mice infected with influenza A (A/WSN/33), mortality was unaffected by treatment with rosuvastatin. CONCLUSIONS: Statins did not alter the replication of influenza A in vitro or enhance its clearance from the lung in vivo. Statins neither attenuated the severity of influenza A-induced lung injury nor had an effect on influenza A-related mortality. Our data suggest that the association between HMG CoA reductase inhibitors and improved outcomes in patients with sepsis and pneumonia are not attributable to their effects on influenza A infection.


Assuntos
Antivirais/farmacologia , Fluorbenzenos/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Infecções por Orthomyxoviridae/tratamento farmacológico , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Animais , Líquido da Lavagem Broncoalveolar/química , Linhagem Celular , Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Pneumonia/tratamento farmacológico , Pneumonia/metabolismo , Pneumonia/virologia , Rosuvastatina Cálcica , Replicação Viral/efeitos dos fármacos
8.
Sci Rep ; 2: 275, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22355787

RESUMO

Exposure of human populations to chronically elevated levels of ambient particulate matter air pollution < 2.5 µm in diameter (PM(2.5)) has been associated with an increase in lung cancer incidence. Over 70% of lung cancer cell lines exhibit promoter methylation of the tumor suppressor p16, an epigenetic modification that reduces its expression. We exposed mice to concentrated ambient PM(2.5) via inhalation, 8 hours daily for 3 weeks and exposed primary murine alveolar epithelial cells to daily doses of fine urban PM (5 µg/cm(2)). In both mice and alveolar epithelial cells, PM exposure increased ROS production, expression of the DNA methyltransferase 1 (DNMT1), and methylation of the p16 promoter. In alveolar epithelial cells, increased transcription of DNMT1 and methylation of the p16 promoter were inhibited by a mitochondrially targeted antioxidant and a JNK inhibitor. These findings provide a potential mechanism by which PM exposure increases the risk of lung cancer.

9.
PLoS One ; 7(1): e30448, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22272351

RESUMO

OBJECTIVE: Alcohol intake increases the risk of acute lung injury (ALI) and the acute respiratory distress syndrome (ARDS) and is associated with poor outcomes in patients who develop these syndromes. No specific therapies are currently available to treat or decrease the risk of ARDS in patients with alcoholism. We have recently shown increased levels of lung adenosine inhibit alveolar fluid clearance, an important predictor of outcome in patients with ARDS. We hypothesized that alcohol might worsen lung injury by increasing lung adenosine levels, resulting in impaired active Na(+) transport in the lung. METHODS: We treated wild-type mice with alcohol administered i.p. to achieve blood alcohol levels associated with moderate to severe intoxication and measured the rate of alveolar fluid clearance and Na,K-ATPase expression in peripheral lung tissue and assessed the effect of alcohol on survival during exposure to hyperoxia. We used primary rat alveolar type II cells to investigate the mechanisms by which alcohol regulates alveolar Na(+) transport. RESULTS: Exposure to alcohol reduced alveolar fluid clearance, downregulated Na,K-ATPase in the lung tissue and worsened hyperoxia-induced lung injury. Alcohol caused an increase in BAL fluid adenosine levels. A similar increase in lung adenosine levels was observed after exposure to hyperoxia. In primary rat alveolar type II cells alcohol and adenosine decreased the abundance of the Na,K-ATPase at the basolateral membrane via a mechanism that required activation of the AMPK. CONCLUSIONS: Alcohol decreases alveolar fluid clearance and impairs survival from acute lung injury. Alcohol induced increases in lung adenosine levels may be responsible for reduction in alveolar fluid clearance and associated worsening of lung injury.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Etanol/toxicidade , Alvéolos Pulmonares/efeitos dos fármacos , Receptor A1 de Adenosina/metabolismo , Sódio/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/mortalidade , Adenosina/metabolismo , Animais , Western Blotting , Líquido da Lavagem Broncoalveolar/química , Células Cultivadas , Depressores do Sistema Nervoso Central/toxicidade , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Hiperóxia , Transporte de Íons/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/metabolismo , Análise de Sobrevida , Taxa de Sobrevida
10.
J Cell Sci ; 124(Pt 17): 2927-37, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21878500

RESUMO

Laminins are heterotrimeric glycoproteins of the extracellular matrix that are secreted by epithelial cells and which are crucial for the normal structure and function of the basement membrane. We have generated a mouse harboring a conditional knockout of α3 laminin (Lama3(fl/fl)), one of the main laminin subunits in the lung basement membrane. At 60 days after intratracheal treatment of adult Lama3(fl/fl) mice with an adenovirus encoding Cre recombinase (Ad-Cre), the protein abundance of α3 laminin in whole lung homogenates was more than 50% lower than that in control-treated mice, suggesting a relatively long half-life for the protein in the lung. Upon exposure to an injurious ventilation strategy (tidal volume of 35 ml per kg of body weight for 2 hours), the mice with a knockdown of the α3 laminin subunit had less severe injury, as shown by lung mechanics, histology, alveolar capillary permeability and survival when compared with Ad-Null-treated mice. Knockdown of the α3 laminin subunit resulted in evidence of lung inflammation. However, this did not account for their resistance to mechanical ventilation. Rather, the loss of α3 laminin was associated with a significant increase in the collagen content of the lungs. We conclude that the loss of α3 laminin in the alveolar epithelium results in an increase in lung collagen, which confers resistance to mechanical injury.


Assuntos
Laminina/deficiência , Pulmão/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Adenoviridae/genética , Animais , Colágeno Tipo I/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Laminina/química , Laminina/genética , Laminina/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/patologia , Respiração com Pressão Positiva , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Alvéolos Pulmonares/fisiologia , Lesão Pulmonar Induzida por Ventilação Mecânica/genética , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia
11.
Part Fibre Toxicol ; 8: 19, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21658250

RESUMO

BACKGROUND: Exposure to particulate matter (PM) air pollution may be an important environmental factor leading to exacerbations of inflammatory illnesses in the GI tract. PM can gain access to the gastrointestinal (GI) tract via swallowing of air or secretions from the upper airways or mucociliary clearance of inhaled particles. METHODS: We measured PM-induced cell death and mitochondrial ROS generation in Caco-2 cells stably expressing oxidant sensitive GFP localized to mitochondria in the absence or presence of an antioxidant. C57BL/6 mice were exposed to a very high dose of urban PM from Washington, DC (200 µg/mouse) or saline via gastric gavage and small bowel and colonic tissue were harvested for histologic evaluation, and RNA isolation up to 48 hours. Permeability to 4 kD dextran was measured at 48 hours. RESULTS: PM induced mitochondrial ROS generation and cell death in Caco-2 cells. PM also caused oxidant-dependent NF-κB activation, disruption of tight junctions and increased permeability of Caco-2 monolayers. Mice exposed to PM had increased intestinal permeability compared with PBS treated mice. In the small bowel, colocalization of the tight junction protein, ZO-1 was lower in the PM treated animals. In the small bowel and colon, PM exposed mice had higher levels of IL-6 mRNA and reduced levels of ZO-1 mRNA. Increased apoptosis was observed in the colon of PM exposed mice. CONCLUSIONS: Exposure to high doses of urban PM causes oxidant dependent GI epithelial cell death, disruption of tight junction proteins, inflammation and increased permeability in the gut in vitro and in vivo. These PM-induced changes may contribute to exacerbations of inflammatory disorders of the gut.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Oxidantes/farmacologia , Material Particulado/farmacologia , Poluição do Ar , Animais , Células CACO-2/citologia , Células CACO-2/efeitos dos fármacos , Células CACO-2/fisiologia , Morte Celular/efeitos dos fármacos , District of Columbia , Impedância Elétrica , Trato Gastrointestinal/citologia , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Ocludina , Tamanho da Partícula , Material Particulado/administração & dosagem , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/ultraestrutura , Proteína da Zônula de Oclusão-1
12.
PLoS One ; 6(4): e18525, 2011 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-21494547

RESUMO

BACKGROUND: Exposure of human populations to ambient particulate matter (PM) air pollution significantly contributes to the mortality attributable to ischemic cardiovascular events. We reported that mice treated with intratracheally instilled PM develop a prothrombotic state that requires the release of IL-6 by alveolar macrophages. We sought to determine whether exposure of mice to PM increases the levels of PAI-1, a major regulator of thrombolysis, via a similar or distinct mechanism. METHODS AND PRINCIPAL FINDINGS: Adult, male C57BL/6 and IL-6 knock out (IL-6(-/-)) mice were exposed to either concentrated ambient PM less than 2.5 µm (CAPs) or filtered air 8 hours daily for 3 days or were exposed to either urban particulate matter or PBS via intratracheal instillation and examined 24 hours later. Exposure to CAPs or urban PM resulted in the IL-6 dependent activation of coagulation in the lung and systemically. PAI-1 mRNA and protein levels were higher in the lung and adipose tissue of mice treated with CAPs or PM compared with filtered air or PBS controls. The increase in PAI-1 was similar in wild-type and IL-6(-/-) mice but was absent in mice treated with etanercept, a TNF-α inhibitor. Treatment with etanercept did not prevent the PM-induced tendency toward thrombus formation. CONCLUSIONS: Mice exposed to inhaled PM exhibited a TNF-α-dependent increase in PAI-1 and an IL-6-dependent activation of coagulation. These results suggest that multiple mechanisms link PM-induced lung inflammation with the development of a prothrombotic state.


Assuntos
Coagulação Sanguínea/fisiologia , Material Particulado/efeitos adversos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Pneumonia/sangue , Pneumonia/metabolismo , Tecido Adiposo Branco/metabolismo , Animais , Linhagem Celular Tumoral , Cidades , Fibrina/metabolismo , Humanos , Exposição por Inalação , Interleucina-6 , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Tamanho da Partícula , Inibidor 1 de Ativador de Plasminogênio/genética , Pneumonia/patologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/metabolismo
13.
Nano Lett ; 10(5): 1664-70, 2010 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-20377197

RESUMO

Excitement surrounding the attractive physical and chemical characteristics of single walled carbon nanotubes (SWCNTs) has been tempered by concerns regarding their potential health risks. Here we consider the lung toxicity of nanoscale dispersed SWCNTs (mean diameter approximately 1 nm). Because dispersion of the SWCNTs increases their aspect ratio relative to as-produced aggregates, we directly test the prevailing hypothesis that lung toxicity associated with SWCNTs compared with other carbon structures is attributable to the large aspect ratio of the individual particles. Thirty days after their intratracheal administration to mice, the granuloma-like structures with mild fibrosis in the large airways observed in mice treated with aggregated SWCNTs were absent in mice treated with nanoscale dispersed SWCNTs. Examination of lung sections from mice treated with nanoscale dispersed SWCNTs revealed uptake of the SWCNTs by macrophages and gradual clearance over time. We conclude that the toxicity of SWCNTs in vivo is attributable to aggregation of the nanomaterial rather than the large aspect ratio of the individual nanotubes. Biocompatible nanoscale dispersion provides a scalable method to generate purified preparations of SWCNTs with minimal toxicity, thus allowing them to be used safely in commercial and biomedical applications.


Assuntos
Materiais Biocompatíveis/toxicidade , Nanotubos/química , Nanotubos/toxicidade , Traqueia/efeitos dos fármacos , Traqueia/patologia , Traqueíte/induzido quimicamente , Traqueíte/patologia , Animais , Coloides/química , Coloides/toxicidade , Cristalização/métodos , Teste de Materiais , Camundongos , Camundongos Endogâmicos C57BL , Nanotubos/ultraestrutura , Tamanho da Partícula
14.
FASEB J ; 23(7): 2055-64, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19237507

RESUMO

Elevated ambient levels of particulate matter air pollution are associated with excess daily mortality, largely attributable to increased rates of cardiovascular events. We have previously reported that particulate matter induces p53-dependent apoptosis in primary human alveolar epithelial cells. Activation of the intrinsic apoptotic pathway by p53 often requires the transcription of the proapoptotic Bcl-2 proteins Noxa, Puma, or both. In this study, we exposed alveolar epithelial cells in culture and mice to fine particulate matter <2.5 microm in diameter (PM(2.5)) collected from the ambient air in Washington, D. C. Exposure to PM(2.5) induced apoptosis in primary alveolar epithelial cells from wild-type but not Noxa(-/-) mice. Twenty-four hours after the intratracheal instillation of PM(2.5), wild-type mice showed increased apoptosis in the lung and increased levels of mRNA encoding Noxa but not Puma. These changes were associated with increased permeability of the alveolar-capillary membrane and inflammation. All of these findings were absent or attenuated in Noxa(-/-) animals. We conclude that PM(2.5)-induced cell death requires Noxa both in vitro and in vivo and that Noxa-dependent cell death might contribute to PM-induced alveolar epithelial dysfunction and the resulting inflammatory response.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Inflamação/induzido quimicamente , Pneumopatias/patologia , Material Particulado/efeitos adversos , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Animais , Proteínas Reguladoras de Apoptose/fisiologia , Morte Celular , Camundongos , Camundongos Knockout , Tamanho da Partícula , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/análise , Proteínas Supressoras de Tumor/análise , Regulação para Cima
15.
J Biol Chem ; 284(4): 2176-86, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-19033436

RESUMO

We have previously reported that airborne particulate matter air pollution (PM) activates the intrinsic apoptotic pathway in alveolar epithelial cells through a pathway that requires the mitochondrial generation of reactive oxygen species (ROS) and the activation of p53. We sought to examine the source of mitochondrial oxidant production and the molecular links between ROS generation and the activation of p53 in response to PM exposure. Using a mitochondrially targeted ratiometric sensor (Ro-GFP) in cells lacking mitochondrial DNA (rho0 cells) and cells stably expressing a small hairpin RNA directed against the Rieske iron-sulfur protein, we show that site III of the mitochondrial electron transport chain is primarily responsible for fine PM (PM2.5)-induced oxidant production. In alveolar epithelial cells, the overexpression of SOD1 prevented the PM2.5-induced ROS generation from the mitochondria and prevented cell death. Infection of mice with an adenovirus encoding SOD1 prevented the PM2.5-induced death of alveolar epithelial cells and the associated increase in alveolar-capillary permeability. Treatment with PM2.5 resulted in the ROS-mediated activation of the oxidant-sensitive kinase ASK1 and its downstream kinase JNK. Murine embryonic fibroblasts from ASK1 knock-out mice, alveolar epithelial cells transfected with dominant negative constructs against ASK1, and pharmacologic inhibition of JNK with SP600125 (25 microM) prevented the PM2.5-induced phosphorylation of p53 and cell death. We conclude that particulate matter air pollution induces the generation of ROS primarily from site III of the mitochondrial electron transport chain and that these ROS activate the intrinsic apoptotic pathway through ASK1, JNK, and p53.


Assuntos
Poluentes Atmosféricos/farmacologia , Apoptose/efeitos dos fármacos , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Células Epiteliais/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Mitocôndrias/efeitos dos fármacos , Alvéolos Pulmonares/citologia , Animais , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Oxidantes/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1
16.
Am J Respir Cell Mol Biol ; 39(6): 666-72, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18556591

RESUMO

Lung cells are exposed to cyclic stretch during normal respiration and during positive pressure mechanical ventilation administered to support gas exchange. Dystroglycan is a ubiquitously expressed matrix receptor that is required for normal basement membrane formation during embryogenesis and for maintaining the function of skeletal muscle myocytes and neurons where it links cells to matrix. We previously reported that equibiaxial stretch of primary alveolar epithelial cells activated the MAP kinase pathway ERK1/2 through a mechanism that required an interaction between dystroglycan and matrix. We determined whether this mechanism of mechanotransduction activates other signaling cascades in lung epithelium. Exposure of rat epithelial alveolar type II cells (AEC) to cyclic mechanical stretch resulted in activation of 5' AMP-activated protein kinase (AMPK). This response was not affected by pretreatment of AEC with the ERK inhibitor PD98059 but was inhibited by knockdown in dystroglycan expression. Moreover, production of reactive oxygen species was enhanced in mechanically stimulated AEC in which dystroglycan was knocked down. This enhancement was reversed by treatment of AEC with an AMPK activator. Activation of AMPK was also observed in lung homogenates from mice after 15 minutes of noninjurious mechanical ventilation. Furthermore, knockdown of dystroglycan in the lungs of mice using an adenovirus encoding a dystroglycan shRNA prevented the stretch-induced activation of AMPK. These results suggest that exposure to cyclic stretch activates the metabolic sensing pathway AMPK in the lung epithelium and supports a novel role for dystroglycan in this mechanotransduction.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Distroglicanas/metabolismo , Pulmão/enzimologia , Adenoviridae , Animais , Ativação Enzimática , Células Epiteliais/citologia , Células Epiteliais/enzimologia , Pulmão/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/enzimologia , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Respiração Artificial , Estresse Mecânico
17.
Am J Respir Crit Care Med ; 174(11): 1229-38, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16946128

RESUMO

RATIONALE: Exposure to particulate matter (PM) causes lung cancer by mechanisms that are unknown, but p53 dysfunction is implicated. OBJECTIVE: We determined whether p53 is required for PM-induced apoptosis in both human and rodent alveolar type (AT) 2 cells. METHODS: A well-characterized form of urban PM was used to determine whether it induces mitochondrial dysfunction (mitochondrial membrane potential change [DeltaPsi m] and caspase-9 activation), p53 protein and mRNA expression, and apoptosis (DNA fragmentation and annexin V staining) in vitro using A549 cells and primary isolated human and rat AT2 cells. The role of p53 was assessed using inhibitors of p53-dependent transcription, pifithrin-alpha, and a genetic approach (overexpressing E6 or dominant negative p53). In mice, the in vivo effects of PM causing p53 expression and apoptosis were assessed 72 h after a single PM intratracheal instillation. MEASUREMENTS AND MAIN RESULTS: PM-induced apoptosis in A549 cells was characterized by increased p53 mRNA and protein expression, mitochondrial translocation of Bax and p53, a reduction in DeltaPsi m, and caspase-9 activation, and these effects were blocked by inhibiting p53-dependent transcription. Similar findings were noted in primary isolated human and rat AT2 cells. A549-rho degrees cells that are incapable of mitochondrial reactive oxygen species production were protected against PM-induced DeltaPsi m, p53 expression, and apoptosis. In mice, PM induced p53 expression and apoptosis at the bronchoalveolar duct junctions. CONCLUSIONS: These data suggest a novel interaction between p53 and the mitochondria in mediating PM-induced apoptosis that is relevant to the pathogenesis of lung cancer from air pollution.


Assuntos
Apoptose/fisiologia , Genes p53/fisiologia , Mitocôndrias/fisiologia , Material Particulado/farmacologia , Alvéolos Pulmonares/fisiologia , Animais , Benzotiazóis/farmacologia , Células Cultivadas , Fragmentação do DNA , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Mitocôndrias/efeitos dos fármacos , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio , Tolueno/análogos & derivados , Tolueno/farmacologia , Transcrição Gênica/efeitos dos fármacos , Translocação Genética/fisiologia
18.
Am J Respir Cell Mol Biol ; 34(6): 670-6, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16439801

RESUMO

Ambient particulate matter is increasingly recognized as a significant contributor to human cardiopulmonary morbidity and mortality in the United States and worldwide. We sought to determine whether exposure to ambient particulate matter would alter alveolar fluid clearance in mice. Mice were exposed to a range of doses of a well-characterized particulate matter collected from the ambient air in Düsseldorf, Germany through a single intratracheal instillation, and alveolar fluid clearance and measurements of lung injury were made. Exposure to even very low doses of particulate matter (10 microg) resulted in a significant reduction in alveolar fluid clearance that was maximal 24 h after the exposure, with complete resolution after 7 d. This was paralleled by a decrease in lung Na,K-ATPase activity. To investigate the mechanism of this effect, we measured plasma membrane Na,K-ATPase abundance in A549 cells and Na,K-ATPase activity in primary rat alveolar type II cells after exposure to particulate matter in the presence or absence of the combined superoxide dismutase and catalase mimetic EUK-134 (5 microM). Membrane but not total protein abundance of the Na,K-ATPase was decreased after exposure to particulate matter, as was Na,K-ATPase activity. This decrease was prevented by the combined superoxide dismutase/catalase mimetic EUK-134. The intratracheal instillation of particulate matter results in alveolar epithelial injury and decreased alveolar fluid clearance, conceivably due to downregulation of the Na,K-ATPase.


Assuntos
Poluentes Atmosféricos/toxicidade , Água Extravascular Pulmonar/metabolismo , Alvéolos Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/farmacologia , Linhagem Celular , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Células Epiteliais/enzimologia , Células Epiteliais/ultraestrutura , Proteínas de Fluorescência Verde , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Compostos Organometálicos/farmacologia , Alvéolos Pulmonares/enzimologia , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Salicilatos/farmacologia , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Transfecção
19.
Am J Respir Cell Mol Biol ; 34(4): 443-52, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16357363

RESUMO

Asbestos causes pulmonary toxicity in part by generating reactive oxygen species that cause DNA damage. We previously showed that the mitochondria-regulated (intrinsic) death pathway mediates alveolar epithelial cell (AEC) DNA damage and apoptosis. Because p53 regulates the DNA damage response in part by inducing intrinsic cell death, we determined whether p53-dependent transcriptional activity mediates asbestos-induced AEC mitochondrial dysfunction and apoptosis. We show that inhibitors of p53-dependent transcriptional activation (pifithrin and type 16-E6 protein) block asbestos-induced AEC mitochondrial membrane potential change (DeltaPsim), caspase 9 activation, and apoptosis. We demonstrate that asbestos activates p53 promoter activity, mRNA levels, protein expression, and Bax and p53 mitochondrial translocation. Further, pifithrin, E6, phytic acid, or rho(0)-A549 cells (cells incapable of mitochondrial reactive oxygen species production) block asbestos-induced p53 activation. Finally, we show that asbestos augments p53 expression in cells at the bronchoalveolar duct junctions of rat lungs and that phytic acid prevents this. These data suggest that p53-dependent transcription pathways mediate asbestos-induced AEC mitochondria-regulated apoptosis. This suggests an important interactive effect between p53 and the mitochondria in the pathogenesis of asbestos-induced pulmonary toxicity that may have broader implications for our understanding of pulmonary fibrosis and lung cancer.


Assuntos
Apoptose , Amianto Amosita/toxicidade , Células Epiteliais/metabolismo , Membranas Mitocondriais/fisiologia , Alvéolos Pulmonares/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Animais , Asbestose/metabolismo , Benzotiazóis , Caspase 9 , Caspases/metabolismo , Linhagem Celular , Ativação Enzimática , Células Epiteliais/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Humanos , Quelantes de Ferro/farmacologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Potenciais da Membrana , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Ácido Fítico/farmacologia , Regiões Promotoras Genéticas , Transporte Proteico , Alvéolos Pulmonares/citologia , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Tiazóis/farmacologia , Tolueno/análogos & derivados , Tolueno/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA