Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
País como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Inhal Toxicol ; 28(9): 383-92, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27240593

RESUMO

BACKGROUND: Inhaled ozone (O3) has been demonstrated as a harmful pollutant and associated with chronic inflammatory diseases such as diabetes and vascular disorders. However, the underlying mechanisms by which O3 mediates harmful effects are poorly understood. OBJECTIVES: To investigate the effect of O3 exposure on glucose intolerance, immune activation and underlying mechanisms in a genetically susceptible mouse model. METHODS: Diabetes-prone KK mice were exposed to filtered air (FA), or O3 (0.5 ppm) for 13 consecutive weekdays (4 h/day). Insulin tolerance test (ITT) was performed following the last exposure. Plasma insulin, adiponectin, and leptin were measured by ELISA. Pathologic changes were examined by H&E and Oil-Red-O staining. Inflammatory responses were detected using flow cytometry and real-time PCR. RESULTS: KK mice exposed to O3 displayed an impaired insulin response. Plasma insulin and leptin levels were reduced in O3-exposed mice. Three-week exposure to O3 induced lung inflammation and increased monocytes/macrophages in both blood and visceral adipose tissue. Inflammatory monocytes/macrophages increased both systemically and locally. CD4 + T cell activation was also enhanced by the exposure of O3 although the relative percentage of CD4 + T cell decreased in blood and adipose tissue. Multiple inflammatory genes including CXCL-11, IFN-γ, TNFα, IL-12, and iNOS were up-regulated in visceral adipose tissue. Furthermore, the expression of oxidative stress-related genes such as Cox4, Cox5a, Scd1, Nrf1, and Nrf2, increased in visceral adipose tissue of O3-exposed mice. CONCLUSIONS: Repeated O3 inhalation induces oxidative stress, adipose inflammation and insulin resistance.


Assuntos
Poluentes Atmosféricos/toxicidade , Imunidade Inata/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Resistência à Insulina/genética , Ozônio/toxicidade , Animais , Expressão Gênica/efeitos dos fármacos , Imunidade Inata/genética , Insulina/sangue , Leptina/sangue , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos Endogâmicos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética
2.
Part Fibre Toxicol ; 10: 26, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23815813

RESUMO

BACKGROUND: With the increase in production and use of engineered nanoparticles (NP; ≤ 100 nm), safety concerns have risen about the potential health effects of occupational or environmental NP exposure. Results of animal toxicology studies suggest that inhalation of NP may cause pulmonary injury with subsequent acute or chronic inflammation. People with chronic respiratory diseases like asthma or allergic rhinitis may be even more susceptible to toxic effects of inhaled NP. Few studies, however, have investigated adverse effects of inhaled NP that may enhance the development of allergic airway disease. METHODS: We investigated the potential of polyethylene glycol coated amorphous silica NP (SNP; 90 nm diameter) to promote allergic airway disease when co-exposed during sensitization with an allergen. BALB/c mice were sensitized by intranasal instillation with 0.02% ovalbumin (OVA; allergen) or saline (control), and co-exposed to 0, 10, 100, or 400 µg of SNP. OVA-sensitized mice were then challenged intranasally with 0.5% OVA 14 and 15 days after sensitization, and all animals were sacrificed a day after the last OVA challenge. Blood and bronchoalveolar lavage fluid (BALF) were collected, and pulmonary tissue was processed for histopathology and biochemical and molecular analyses. RESULTS: Co-exposure to SNP during OVA sensitization caused a dose-dependent enhancement of allergic airway disease upon challenge with OVA alone. This adjuvant-like effect was manifested by significantly greater OVA-specific serum IgE, airway eosinophil infiltration, mucous cell metaplasia, and Th2 and Th17 cytokine gene and protein expression, as compared to mice that were sensitized to OVA without SNP. In saline controls, SNP exposure did cause a moderate increase in airway neutrophils at the highest doses. CONCLUSIONS: These results suggest that airway exposure to engineered SNP could enhance allergen sensitization and foster greater manifestation of allergic airway disease upon secondary allergen exposures. Whereas SNP caused innate immune responses at high doses in non-allergic mice, the adjuvant effects of SNP were found at lower doses in allergic mice and were Th2/Th17 related. In conclusion, these findings in mice suggest that individuals exposed to SNP might be more prone to manifest allergic airway disease, due to adjuvant-like properties of SNP.


Assuntos
Pulmão/efeitos dos fármacos , Nanopartículas/toxicidade , Ovalbumina , Hipersensibilidade Respiratória/induzido quimicamente , Dióxido de Silício/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Imunoglobulina E/sangue , Mediadores da Inflamação/metabolismo , Exposição por Inalação/efeitos adversos , Pulmão/imunologia , Pulmão/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos Endogâmicos BALB C , Infiltração de Neutrófilos/efeitos dos fármacos , Tamanho da Partícula , Eosinofilia Pulmonar/sangue , Eosinofilia Pulmonar/induzido quimicamente , Eosinofilia Pulmonar/imunologia , Hipersensibilidade Respiratória/sangue , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/imunologia , Medição de Risco , Células Th17/efeitos dos fármacos , Células Th17/imunologia , Células Th17/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Fatores de Tempo
3.
Part Fibre Toxicol ; 10: 43, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23968387

RESUMO

BACKGROUND: Inflammation and oxidative stress play critical roles in the pathogenesis of inhaled air pollutant-mediated metabolic disease. Inflammation in the adipose tissues niches are widely believed to exert important effects on organ dysfunction. Recent data from both human and animal models suggest a role for inflammation and oxidative stress in epicardial adipose tissue (EAT) as a risk factor for the development of cardiovascular disease. We hypothesized that inhalational exposure to concentrated ambient fine particulates (CAPs) and ozone (O3) exaggerates inflammation and oxidative stress in EAT and perirenal adipose tissue (PAT). METHODS: Eight- week-old Male Sprague-Dawley rats were fed a normal diet (ND) or high fructose diet (HFr) for 8 weeks, and then exposed to ambient AIR, CAPs at a mean of 356 µg/m3, O3 at 0.485 ppm, or CAPs (441 µg/m3) + O3 (0.497 ppm) in Dearborn, MI, 8 hours/day, 5 days/week, for 9 days over 2 weeks. RESULTS: EAT and PAT showed whitish color in gross, and less mitochondria, higher mRNA expression of white adipose specific and lower brown adipose specific genes than in brown adipose tissues. Exposure to CAPs and O3 resulted in the increase of macrophage infiltration in both EAT and PAT of HFr groups. Proinflammatory genes of Tnf-α, Mcp-1 and leptin were significantly upregulated while IL-10 and adiponectin, known as antiinflammatory genes, were reduced after the exposures. CAPs and O3 exposures also induced an increase in inducible nitric oxide synthase (iNOS) protein expression, and decrease in mitochondrial area in EAT and PAT. We also found significant increases in macrophages of HFr-O3 rats. The synergetic interaction of HFr and dirty air exposure on the inflammation was found in most of the experiments. Surprisingly, exposure to CAPs or O3 induced more significant inflammation and oxidative stress than co-exposure of CAPs and O3 in EAT and PAT. CONCLUSION: EAT and PAT are both white adipose tissues. Short-term exposure to CAPs and O3, especially with high fructose diet, induced inflammation and oxidative stress in EAT and PAT in rats. These findings may provide a link between air-pollution exposure and accelerated susceptibility to cardiovascular disease and metabolic complications.


Assuntos
Tecido Adiposo Branco/efeitos dos fármacos , Carboidratos da Dieta , Frutose , Exposição por Inalação/efeitos adversos , Ozônio/toxicidade , Paniculite/induzido quimicamente , Material Particulado/toxicidade , Adipocinas/genética , Adipocinas/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/ultraestrutura , Tecido Adiposo Branco/metabolismo , Tecido Adiposo Branco/ultraestrutura , Animais , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Rim , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Paniculite/genética , Paniculite/metabolismo , Paniculite/patologia , Pericárdio , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Medição de Risco , Fatores de Tempo
4.
Am J Physiol Lung Cell Mol Physiol ; 302(5): L455-62, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22180657

RESUMO

Allergic airway disease is characterized by a T helper type 2 cell-mediated airway inflammation and airway hyperresponsiveness. Little is known about the role of hypoxia-mediated signaling in the progression of the disease. To address this knowledge gap, a mouse model was created in which doxycycline exposure induces the functional deletion of hypoxia inducible factor-1α from alveolar type II and Clara cells of the lung. When hypoxia inducible factor-1α deletion was induced during the early postnatal development period of the lung, the mice displayed an enhanced response to the ovalbumin model of allergic airway disease. These hypoxia inducible factor-1α-deficient mice exhibit increased cellular infiltrates, eosinophilia in the lavage fluid and parenchyma, and T helper type 2 cytokines, as compared with ovalbumin-treated control mice. Moreover, these hypoxia inducible factor-1α-deficient mice display increased airway resistance when compared with their control counterparts. Interestingly, if the loss of hypoxia inducible factor-1α was induced in early adulthood, the exacerbated phenotype was not observed. Taken together, these results suggest that epithelial hypoxia inducible factor-1α plays an important role in establishing the innate immunity of the lung and epithelial-specific deficiency in the transcription factor, during early postnatal development, increases the severity of inflammation and functional airway resistance, following ovalbumin challenge. Finally, these results might explain some of the chronic respiratory pathology observed in premature infants, especially those that receive supplemental oxygen. This early hyperoxic exposure, from normal ambient and supplemental oxygen, would presumably inhibit normal hypoxia inducible factor-1α signaling, mimicking the functional deletion described.


Assuntos
Asma/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/metabolismo , Resistência das Vias Respiratórias/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Asma/induzido quimicamente , Asma/imunologia , Líquido da Lavagem Broncoalveolar/química , Broncoconstritores/farmacologia , Contagem de Células , Citocinas/metabolismo , Proteína Básica Maior de Eosinófilos/metabolismo , Eosinófilos/metabolismo , Eosinófilos/patologia , Feminino , Expressão Gênica , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão/patologia , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Transgênicos , Ovalbumina , Distribuição Aleatória
5.
Part Fibre Toxicol ; 8: 20, 2011 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-21745393

RESUMO

BACKGROUND: Prior studies have demonstrated a link between air pollution and metabolic diseases such as type II diabetes. Changes in adipose tissue and its mitochondrial content/function are closely associated with the development of insulin resistance and attendant metabolic complications. We investigated changes in adipose tissue structure and function in brown and white adipose depots in response to chronic ambient air pollutant exposure in a rodent model. METHODS: Male ApoE knockout (ApoE-/-) mice inhaled concentrated fine ambient PM (PM < 2.5 µm in aerodynamic diameter; PM2.5) or filtered air (FA) for 6 hours/day, 5 days/week, for 2 months. We examined superoxide production by dihydroethidium staining; inflammatory responses by immunohistochemistry; and changes in white and brown adipocyte-specific gene profiles by real-time PCR and mitochondria by transmission electron microscopy in response to PM2.5 exposure in different adipose depots of ApoE-/- mice to understand responses to chronic inhalational stimuli. RESULTS: Exposure to PM2.5 induced an increase in the production of reactive oxygen species (ROS) in brown adipose depots. Additionally, exposure to PM2.5 decreased expression of uncoupling protein 1 in brown adipose tissue as measured by immunohistochemistry and Western blot. Mitochondrial number was significantly reduced in white (WAT) and brown adipose tissues (BAT), while mitochondrial size was also reduced in BAT. In BAT, PM2.5 exposure down-regulated brown adipocyte-specific genes, while white adipocyte-specific genes were differentially up-regulated. CONCLUSIONS: PM2.5 exposure triggers oxidative stress in BAT, and results in key alterations in mitochondrial gene expression and mitochondrial alterations that are pronounced in BAT. We postulate that exposure to PM2.5 may induce imbalance between white and brown adipose tissue functionality and thereby predispose to metabolic dysfunction.


Assuntos
Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/fisiologia , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/fisiologia , Expressão Gênica/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Material Particulado/farmacologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Branco/citologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Metabolismo Energético/efeitos dos fármacos , Humanos , Canais Iônicos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Tamanho da Partícula , Material Particulado/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Desacopladora 1
6.
Proteomics ; 10(3): 520-31, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20029843

RESUMO

Ambient particulate matter (PM) from air pollution is associated with exacerbation of asthma. The immunological basis for the adjuvant effects of PM is still not well understood. The generation of ROS and the resulting oxidative stress has been identified as one of the major mechanisms. Using a new intranasal sensitization model in which ambient PM is used as an adjuvant to enhance allergic inflammation (Li et al., Environ. Health Perspect. 2009, 117, 1116-1123), a proteomics approach was applied to study the adjuvant effects of ambient PM. The enhanced in vivo adjuvant effect of ultrafine particles correlates with a higher in vitro oxidant potential and a higher content of redox-cycling organic chemicals. Bronchoalveolar lavage fluid proteins from normal and sensitized mice were resolved by 2-DE, and identified by MS. Polymeric immunoglobulin receptor, complement C3, neutrophil gelatinase-associated lipocalin, chitinase 3-like protein 3, chitinase 3-like protein 4, and acidic mammalian chitinase demonstrated significantly enhanced up-regulation by UFP with a polycyclic aromatic hydrocarbon content and a higher oxidant potential. These proteins may be the important specific elements targeted by PM in air pollution through the ability to generate ROS in the immune system, and may be involved in allergen sensitization and asthma pathogenesis.


Assuntos
Adjuvantes Imunológicos/metabolismo , Líquido da Lavagem Broncoalveolar/imunologia , Exposição por Inalação/efeitos adversos , Material Particulado/imunologia , Proteoma/imunologia , Poluição do Ar , Animais , Feminino , Inflamação/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Tamanho da Partícula , Proteômica/métodos
7.
Am J Physiol Lung Cell Mol Physiol ; 298(2): L139-47, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19915160

RESUMO

Hypoxia plays an important role in development, cellular homeostasis, and pathological conditions, such as cancer and stroke. There is also growing evidence that hypoxia is an important modulator of the inflammatory process. Hypoxia-inducible factors (HIFs) are a family of proteins that regulate the cellular response to oxygen deficit, and loss of HIFs impairs inflammatory cell function. There is little known, however, about the role of epithelial-derived HIF signaling in modulating inflammation. Cobalt is capable of eliciting an allergic response and promoting HIF signaling. To characterize the inflammatory function of epithelial-derived HIF in response to inhaled cobalt, a conditional lung-specific HIF1alpha, the most ubiquitously expressed HIF, deletion mouse, was created. Control mice showed classic signs of metal-induced injury following cobalt exposure, including fibrosis and neutrophil infiltration. In contrast, HIF1alpha-deficient mice displayed a Th2 response that resembled asthma, including increased eosinophilic infiltration, mucus cell metaplasia, and chitinase-like protein expression. The results suggest that epithelial-derived HIF signaling has a critical role in establishing a tissue's inflammatory response, and compromised HIF1alpha signaling biases the tissue towards a Th2-mediated reaction.


Assuntos
Cobalto , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inflamação/induzido quimicamente , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Animais , Asma/induzido quimicamente , Asma/imunologia , Peso Corporal , Líquido da Lavagem Broncoalveolar/citologia , Cobalto/imunologia , Cobalto/farmacologia , Citocinas/imunologia , Perfilação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Inflamação/imunologia , Pulmão/citologia , Pulmão/patologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células Th2/imunologia
8.
Am J Physiol Lung Cell Mol Physiol ; 299(3): L374-83, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20562226

RESUMO

We have previously demonstrated that intranasal administration of ambient ultrafine particles (UFP) acts as an adjuvant for primary allergic sensitization to ovalbumin (OVA) in Balb/c mice. It is important to find out whether inhaled UFP exert the same effect on the secondary immune response as a way of explaining asthma flares in already-sensitized individuals due to traffic exposure near a freeway. The objective of this study is to determine whether inhalation exposure to ambient UFP near an urban freeway could enhance the secondary immune response to OVA in already-sensitized mice. Prior OVA-sensitized animals were exposed to concentrated ambient UFP at the time of secondary OVA challenge in our mobile animal laboratory in Los Angeles. OVA-specific antibody production, airway morphometry, allergic airway inflammation, cytokine gene expression, and oxidative stress marker were assessed. As few as five ambient UFP exposures were sufficient to promote the OVA recall immune response, including generating allergic airway inflammation in smaller and more distal airways compared with the adjuvant effect of intranasally instilled UFP on the primary immune response. The secondary immune response was characterized by the T helper 2 and IL-17 cytokine gene expression in the lung. In summary, our results demonstrated that inhalation of prooxidative ambient UFP could effectively boost the secondary immune response to an experimental allergen, indicating that vehicular traffic exposure could exacerbate allergic inflammation in already-sensitized subjects.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Formação de Anticorpos/efeitos dos fármacos , Material Particulado/administração & dosagem , Emissões de Veículos , Administração por Inalação , Animais , Asma/fisiopatologia , Citocinas/metabolismo , Feminino , Expressão Gênica , Hipersensibilidade/complicações , Inflamação/etiologia , Inflamação/patologia , Inflamação/fisiopatologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Sistema Respiratório/imunologia , Doenças Respiratórias/etiologia , Doenças Respiratórias/patologia , Doenças Respiratórias/fisiopatologia , Índice de Gravidade de Doença
9.
Toxicol Sci ; 137(2): 447-57, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24218148

RESUMO

Hard metal lung disease (HMLD) is an occupational lung disease specific to inhalation of cobalt-containing particles whose mechanism is largely unknown. Cobalt is a known hypoxia mimic and stabilizer of the alpha subunits of hypoxia-inducible factors (HIFs). Previous work revealed that though HIF1α contrib utes to cobalt toxicity in vitro, loss of HIF1α in the alveolar epithelial cells does not provide in vivo protection from cobalt-induced lung inflammation. HIF1α and HIF2α show unique tissue expression profiles, and HIF2α is known to be the predominant HIF mRNA isoform in the adult lung. Thus, if HIF2α activation by cobalt contributes to pathophysiology of HMLD, we hypothesized that loss of HIF2α in lung epithelium would provide protection from cobalt-induced inflammation. Mice with HIF2α-deficiency in Club and alveolar type II epithelial cells (ATIIs) (HIF2α(Δ/Δ)) were exposed to cobalt (60 µg/day) or saline using a subacute occupational exposure model. Bronchoalveolar lavage cellularity, cytokines, qRT-PCR, and histopathology were analyzed. Results show that loss of HIF2α leads to enhanced eosinophilic inflammation and increased goblet cell metaplasia. Additionally, control mice demonstrated a mild recovery from cobalt-induced lung injury compared with HIF2α(Δ/Δ) mice, suggesting a role for epithelial HIF2α in repair mechanisms. The expression of important cytokines, such as interleukin (IL)-5 and IL-10, displayed significant differences following cobalt exposure when HIF2α(Δ/Δ) and control mice were compared. In summary, our data suggest that although loss of HIF2α does not afford protection from cobalt-induced lung inflammation, epithelial HIF2α signaling does play an important role in modulating the inflammatory and repair response in the lung.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Cobalto/toxicidade , Lesão Pulmonar/induzido quimicamente , Alvéolos Pulmonares/efeitos dos fármacos , Eosinofilia Pulmonar/induzido quimicamente , Mucosa Respiratória/efeitos dos fármacos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Doxiciclina/farmacologia , Perfilação da Expressão Gênica , Imuno-Histoquímica , Lesão Pulmonar/complicações , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Camundongos , Camundongos Knockout , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Eosinofilia Pulmonar/etiologia , Eosinofilia Pulmonar/imunologia , Eosinofilia Pulmonar/patologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/patologia
10.
Environ Health Perspect ; 121(6): 676-82, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23649427

RESUMO

BACKGROUND: Engineered nanomaterials (ENMs) have potential benefits, but they also present safety concerns for human health. Interlaboratory studies in rodents using standardized protocols are needed to assess ENM toxicity. METHODS: Four laboratories evaluated lung responses in C57BL/6 mice to ENMs delivered by oropharyngeal aspiration (OPA), and three labs evaluated Sprague-Dawley (SD) or Fisher 344 (F344) rats following intratracheal instillation (IT). ENMs tested included three forms of titanium dioxide (TiO2) [anatase/rutile spheres (TiO2-P25), anatase spheres (TiO2-A), and anatase nanobelts (TiO2-NBs)] and three forms of multiwalled carbon nanotubes (MWCNTs) [original (O), purified (P), and carboxylic acid "functionalized" (F)]. One day after treatment, bronchoalveolar lavage fluid was collected to determine differential cell counts, lactate dehydrogenase (LDH), and protein. Lungs were fixed for histopathology. Responses were also examined at 7 days (TiO2 forms) and 21 days (MWCNTs) after treatment. RESULTS: TiO2-A, TiO2-P25, and TiO2-NB caused significant neutrophilia in mice at 1 day in three of four labs. TiO2-NB caused neutrophilia in rats at 1 day in two of three labs, and TiO2-P25 and TiO2-A had no significant effect in any of the labs. Inflammation induced by TiO2 in mice and rats resolved by day 7. All MWCNT types caused neutrophilia at 1 day in three of four mouse labs and in all rat labs. Three of four labs observed similar histopathology to O-MWCNTs and TiO2-NBs in mice. CONCLUSIONS: ENMs produced similar patterns of neutrophilia and pathology in rats and mice. Although interlaboratory variability was found in the degree of neutrophilia caused by the three types of TiO2 nanoparticles, similar findings of relative potency for the three types of MWCNTs were found across all laboratories, thus providing greater confidence in these interlaboratory comparisons.


Assuntos
Pulmão/efeitos dos fármacos , Nanopartículas/toxicidade , Nanotubos de Carbono/toxicidade , Titânio/toxicidade , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , National Institute of Environmental Health Sciences (U.S.) , Neutrófilos/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Estados Unidos
11.
Environ Health Perspect ; 117(7): 1116-23, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19654922

RESUMO

BACKGROUND: It has been demonstrated that ambient particulate matter (PM) can act as an adjuvant for allergic sensitization. Redox-active organic chemicals on the particle surface play an important role in PM adverse health effects and may determine the adjuvant effect of different particle types according to their potential to perturb redox equilibrium in the immune system. OBJECTIVES: We determined whether the adjuvant effect of ambient fine particles versus ultrafine particles (UFPs) is correlated to their prooxidant potential. METHODS: We have established an intranasal sensitization model that uses ambient PM as a potential adjuvant for sensitization to ovalbumin (OVA), which enhances the capacity for secondary OVA challenge to induce allergic airway inflammation. RESULTS: UFPs with a greater polycyclic aromatic hydrocarbon (PAH) content and higher oxidant potential enhanced OVA sensitization more readily than did fine particles. This manifests as enhanced allergic inflammation upon secondary OVA challenge, leading to eosinophilic inflammation and mucoid hyperplasia starting at the nasal turbinates all the way down to the small pulmonary airways. The thiol antioxidant N-acetyl cysteine was able to suppress some of these sensitization events. CONCLUSIONS: The adjuvant effects of ambient UFP is determined by their oxidant potential, which likely plays a role in changing the redox equilibrium in the mucosal immune system.


Assuntos
Ovalbumina/imunologia , Material Particulado/imunologia , Material Particulado/toxicidade , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/imunologia , Adjuvantes Imunológicos , Animais , Feminino , Imuno-Histoquímica , Técnicas In Vitro , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/metabolismo , Nariz/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos
12.
Toxicol Sci ; 108(1): 207-21, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19176365

RESUMO

The interaction between air particulates and genetic susceptibility has been implicated in the pathogenesis of asthma. The overall objective of this study was to determine the effects of inhalation exposure to environmentally relevant concentrated air particulates (CAPs) on the lungs of ovalbumin (ova) sensitized and challenged Brown Norway rats. Changes in gene expression were compared with lung tissue histopathology, morphometry, and biochemical and cellular parameters in bronchoalveolar lavage fluid (BALF). Ova challenge was responsible for the preponderance of gene expression changes, related largely to inflammation. CAPs exposure alone resulted in no significant gene expression changes, but CAPs and ova-exposed rodents exhibited an enhanced effect relative to ova alone with differentially expressed genes primarily related to inflammation and airway remodeling. Gene expression data was consistent with the biochemical and cellular analyses of the BALF, the pulmonary pathology, and morphometric changes when comparing the CAPs-ova group to the air-saline or CAPs-saline group. However, the gene expression data were more sensitive than the BALF cell type and number for assessing the effects of CAPs and ova versus the ova challenge alone. In addition, the gene expression results provided some additional insight into the TGF-beta-mediated molecular processes underlying these changes. The broad-based histopathology and functional genomic analyses demonstrate that exposure to CAPs exacerbates rodents with allergic inflammation induced by an allergen and suggests that asthmatics may be at increased risk for air pollution effects.


Assuntos
Poluentes Atmosféricos/toxicidade , Alérgenos/administração & dosagem , Expressão Gênica/efeitos dos fármacos , Pulmão/metabolismo , Ovalbumina/administração & dosagem , Material Particulado/toxicidade , Alérgenos/imunologia , Análise de Variância , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Broncopneumonia/metabolismo , Broncopneumonia/patologia , Inflamação/imunologia , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ovalbumina/imunologia , Material Particulado/administração & dosagem , Análise de Componente Principal , Ratos , Hipersensibilidade Respiratória/metabolismo , Hipersensibilidade Respiratória/patologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa