Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Assunto da revista
País de afiliação
Intervalo de ano de publicação
1.
Part Fibre Toxicol ; 10: 61, 2013 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-24330719

RESUMO

OBJECTIVE: Diesel exhaust particulate (DEP), a major component of urban air pollution, has been linked to atherogenesis and precipitation of myocardial infarction. We hypothesized that DEP exposure would increase and destabilise atherosclerotic lesions in apolipoprotein E deficient (ApoE-/-) mice. METHODS: ApoE-/- mice were fed a 'Western diet' (8 weeks) to induce 'complex' atherosclerotic plaques, with parallel experiments in normal chow fed wild-type mice. During the last 4 weeks of feeding, mice received twice weekly instillation (oropharyngeal aspiration) of 35 µL DEP (1 mg/mL, SRM-2975) or vehicle (saline). Atherosclerotic burden was assessed by en-face staining of the thoracic aorta and histological examination of the brachiocephalic artery. RESULTS: Brachiocephalic atherosclerotic plaques were larger in ApoE-/- mice treated with DEP (59 ± 10%) than in controls (32 ± 7%; P = 0.017). In addition, DEP-treated mice had more plaques per section of artery (2.4 ± 0.2 vs 1.8 ± 0.2; P = 0.048) and buried fibrous layers (1.2 ± 0.2 vs 0.4 ± 0.1; P = 0.028). These changes were associated with lung inflammation and increased antioxidant gene expression in the liver, but not with changes in endothelial function, plasma lipids or systemic inflammation. CONCLUSIONS: Increased atherosclerosis is caused by the particulate component of diesel exhaust producing advanced plaques with a potentially more vulnerable phenotype. These results are consistent with the suggestion that removal of the particulate component would reduce the adverse cardiovascular effects of diesel exhaust.


Assuntos
Estresse Oxidativo/efeitos dos fármacos , Material Particulado/toxicidade , Placa Aterosclerótica/induzido quimicamente , Placa Aterosclerótica/patologia , Emissões de Veículos/toxicidade , Remodelação das Vias Aéreas/efeitos dos fármacos , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Apolipoproteínas E/genética , Coagulação Sanguínea/efeitos dos fármacos , Tronco Braquiocefálico/efeitos dos fármacos , Tronco Braquiocefálico/metabolismo , Tronco Braquiocefálico/patologia , Proteína C-Reativa/análise , Modelos Animais de Doenças , Fibrinogênio/análise , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orofaringe/metabolismo , Material Particulado/farmacocinética , Placa Aterosclerótica/sangue , Placa Aterosclerótica/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia
2.
Environ Res ; 115: 1-10, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22507957

RESUMO

BACKGROUND: Cerium oxide (CeO(2)) nanoparticles improve the burning efficiency of fuel, however, little is known about health impacts of altered emissions from the vehicles. METHODS: Atherosclerosis-prone apolipoprotein E knockout (ApoE(-/-)) mice were exposed by inhalation to diluted exhaust (1.7 mg/m(3), 20, 60 or 180 min, 5 day/week, for 4 weeks), from an engine using standard diesel fuel (DE) or the same diesel fuel containing 9 ppm cerium oxide nanoparticles (DCeE). Changes in hematological indices, clinical chemistry, atherosclerotic burden, tissue levels of inflammatory cytokines and pathology of the major organs were assessed. RESULTS: Addition of CeO(2) to fuel resulted in a reduction of the number (30%) and surface area (10%) of the particles in the exhaust, whereas the gaseous co-pollutants were increased (6-8%). There was, however, a trend towards an increased size and complexity of the atherosclerotic plaques following DE exposure, which was not evident in the DCeE group. There were no clear signs of altered hematological or pathological changes induced by either treatment. However, levels of proinflammatory cytokines were modulated in a brain region and liver following DCeE exposure. CONCLUSIONS: These results imply that addition of CeO(2) nanoparticles to fuel decreases the number of particles in exhaust and may reduce atherosclerotic burden associated with exposure to standard diesel fuel. From the extensive assessment of biological parameters performed, the only concerning effect of cerium addition was a slightly raised level of cytokines in a region of the central nervous system. Overall, the use of cerium as a fuel additive may be a potentially useful way to limit the health effects of vehicle exhaust. However, further testing is required to ensure that such an approach is not associated with a chronic inflammatory response which may eventually cause long-term health effects.


Assuntos
Aterosclerose/induzido quimicamente , Cério/toxicidade , Exposição por Inalação/efeitos adversos , Nanopartículas/toxicidade , Material Particulado/toxicidade , Emissões de Veículos/toxicidade , Animais , Aterosclerose/sangue , Aterosclerose/genética , Contagem de Células Sanguíneas , Análise Química do Sangue , Encéfalo/imunologia , Feminino , Hematócrito , Hemoglobinas/metabolismo , Imuno-Histoquímica , Pulmão/imunologia , Masculino , Camundongos , Camundongos Knockout , Tamanho da Partícula , Distribuição Aleatória
3.
Part Fibre Toxicol ; 9: 9, 2012 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-22480168

RESUMO

BACKGROUND: Inhalation of diesel exhaust impairs vascular function in man, by a mechanism that has yet to be fully established. We hypothesised that pulmonary exposure to diesel exhaust particles (DEP) would cause endothelial dysfunction in rats as a consequence of pulmonary and systemic inflammation. METHODS: Wistar rats were exposed to DEP (0.5 mg) or saline vehicle by intratracheal instillation and hind-limb blood flow, blood pressure and heart rate were monitored in situ 6 or 24 h after exposure. Vascular function was tested by administration of the endothelium-dependent vasodilator acetylcholine (ACh) and the endothelium-independent vasodilator sodium nitroprusside (SNP) in vivo and ex vivo in isolated rings of thoracic aorta, femoral and mesenteric artery from DEP exposed rats. Bronchoalveolar lavage fluid (BALF) and blood plasma were collected to assess pulmonary (cell differentials, protein levels & interleukin-6 (IL-6)) and systemic (IL-6), tumour necrosis factor alpha (TNFα) and C-reactive protein (CRP)) inflammation, respectively. RESULTS: DEP instillation increased cell counts, total protein and IL-6 in BALF 6 h after exposure, while levels of IL-6 and TNFα were only raised in blood 24 h after DEP exposure. DEP had no effect on the increased hind-limb blood flow induced by ACh in vivo at 6 or 24 h. However, responses to SNP were impaired at both time points. In contrast, ex vivo responses to ACh and SNP were unaltered in arteries isolated from rats exposed to DEP. CONCLUSIONS: Exposure of rats to DEP induces both pulmonary and systemic inflammation, but does not modify endothelium-dependent vasodilatation. Other mechanisms in vivo limit dilator responses to SNP and these require further investigation.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Material Particulado/toxicidade , Pneumonia/induzido quimicamente , Emissões de Veículos/toxicidade , Animais , Aorta Torácica/efeitos dos fármacos , Contagem de Células Sanguíneas , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Pressão Sanguínea/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Proteína C-Reativa/análise , Diferenciação Celular/efeitos dos fármacos , Fatores Relaxantes Dependentes do Endotélio/farmacologia , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Artéria Femoral/efeitos dos fármacos , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Interleucina-6/análise , Leucócitos/citologia , Leucócitos/efeitos dos fármacos , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Material Particulado/análise , Pneumonia/sangue , Pneumonia/fisiopatologia , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/análise , Vasodilatação/efeitos dos fármacos , Emissões de Veículos/análise
4.
ACS Nano ; 11(5): 4542-4552, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28443337

RESUMO

The development of engineered nanomaterials is growing exponentially, despite concerns over their potential similarities to environmental nanoparticles that are associated with significant cardiorespiratory morbidity and mortality. The mechanisms through which inhalation of nanoparticles could trigger acute cardiovascular events are emerging, but a fundamental unanswered question remains: Do inhaled nanoparticles translocate from the lung in man and directly contribute to the pathogenesis of cardiovascular disease? In complementary clinical and experimental studies, we used gold nanoparticles to evaluate particle translocation, permitting detection by high-resolution inductively coupled mass spectrometry and Raman microscopy. Healthy volunteers were exposed to nanoparticles by acute inhalation, followed by repeated sampling of blood and urine. Gold was detected in the blood and urine within 15 min to 24 h after exposure, and was still present 3 months after exposure. Levels were greater following inhalation of 5 nm (primary diameter) particles compared to 30 nm particles. Studies in mice demonstrated the accumulation in the blood and liver following pulmonary exposure to a broader size range of gold nanoparticles (2-200 nm primary diameter), with translocation markedly greater for particles <10 nm diameter. Gold nanoparticles preferentially accumulated in inflammation-rich vascular lesions of fat-fed apolipoproteinE-deficient mice. Furthermore, following inhalation, gold particles could be detected in surgical specimens of carotid artery disease from patients at risk of stroke. Translocation of inhaled nanoparticles into the systemic circulation and accumulation at sites of vascular inflammation provides a direct mechanism that can explain the link between environmental nanoparticles and cardiovascular disease and has major implications for risk management in the use of engineered nanomaterials.


Assuntos
Nanopartículas Metálicas/administração & dosagem , Doenças Vasculares/metabolismo , Administração por Inalação , Adulto , Animais , Ouro , Voluntários Saudáveis , Humanos , Pulmão/patologia , Masculino , Camundongos , Nanopartículas , Nanoestruturas/análise , Tamanho da Partícula , Doenças Vasculares/terapia
5.
J Am Heart Assoc ; 2(1): e004267, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23525432

RESUMO

BACKGROUND: Urocortin 2 and urocortin 3 are endogenous peptides with an emerging role in cardiovascular pathophysiology. We assessed their pharmacodynamic profile and examined the role of the endothelium in mediating their vasomotor effects in vivo in man. METHODS AND RESULTS: Eighteen healthy male volunteers (23±4 years) were recruited into a series of double-blind, randomized crossover studies using bilateral forearm venous occlusion plethysmography during intra-arterial urocortin 2 (3.6 to 120 pmol/min), urocortin 3 (1.2 to 36 nmol/min), and substance P (2 to 8 pmol/min) in the presence or absence of inhibitors of cyclooxygenase (aspirin), cytochrome P450 metabolites of arachidonic acid (fluconazole), and nitric oxide synthase (L-NMMA). Urocortins 2 and 3 evoked arterial vasodilatation (P<0.0001) without tachyphylaxis but with a slow onset and offset of action. Inhibition of nitric oxide synthase with L-NMMA reduced vasodilatation to substance P and urocortin 2 (P≤0.001 for both) but had little effect on urocortin 3 (P>0.05). Neither aspirin nor fluconazole affected vasodilatation induced by any of the infusions (P>0.05 for all). In the presence of all 3 inhibitors, urocortin 2- and urocortin 3-induced vasodilatation was attenuated (P<0.001 for all) to a greater extent than with L-NMMA alone (P≤0.005). CONCLUSIONS: Urocortins 2 and 3 cause potent and prolonged arterial vasodilatation without tachyphylaxis. These vasomotor responses are at least partly mediated by endothelial nitric oxide and cytochrome P450 metabolites of arachidonic acid. The role of urocortins 2 and 3 remains to be explored in the setting of human heart failure, but they have the potential to have major therapeutic benefits. CLINICAL TRIAL REGISTRATION: http://www.clinicaltrials.gov//. Unique identifier: NCT01096706 and NCT01296607.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Antebraço/irrigação sanguínea , Urocortinas/administração & dosagem , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem , Adulto , Ácido Araquidônico/metabolismo , Pressão Sanguínea/efeitos dos fármacos , Estudos Cross-Over , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/metabolismo , Método Duplo-Cego , Endotélio Vascular/metabolismo , Inibidores Enzimáticos/administração & dosagem , Humanos , Infusões Intra-Arteriais , Masculino , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Escócia , Substância P/administração & dosagem , Fatores de Tempo , Adulto Jovem
7.
Environ Health Perspect ; 117(4): 611-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19440501

RESUMO

BACKGROUND: Diesel exhaust particulate (DEP) is a key arbiter of the adverse cardiovascular effects of air pollution. OBJECTIVES: We assessed the in vitro effects of DEP on vascular function, nitric oxide (NO) availability, and the generation of oxygen-centered free radicals. METHODS: We assessed the direct vascular effects of DEP (10-100 microg/mL) in isolated rat aortic rings using myography. We investigated NO scavenging and oxygen-centered free radical generation using an NO electrode and electron paramagnetic resonance (EPR) with the Tempone-H (1-hydroxyl-2,2,6,6-tetramethyl-4-oxo-piperidine) spin trap, respectively. RESULTS: Acetylcholine-induced relaxation was attenuated by DEP (maximum relaxation reduced from 91 +/- 4% to 49 +/- 6% with 100 microg/mL DEP; p < 0.001) but was restored by superoxide dismutase (SOD; maximum relaxation, 73 +/- 6%; p < 0.001). DEP caused a modest inhibition of relaxation to NO donor drugs, an effect that could be reversed by SOD (p < 0.01). At 10 microg/mL, DEP did not affect verapamil-induced relaxation (p = 0.73), but at 100 microg/mL DEP inhibited relaxation (p < 0.001) by a mechanism independent of SOD. NO concentrations generated by 2-(N,N-diethylamino)-diazenolate-2-oxide (DEA/NO; 10 microM) were reduced by DEP (100 microg/mL; from 5.2 +/- 0.4 to 3.3 +/- 0.4 microM; p = 0.002). Free radical generation was increased by DEP (10 microg/mL; 9-fold increase in EPR spectra; p = 0.004) in a manner that could be attenuated by SOD (p = 0.015). CONCLUSIONS: DEP caused oxidative stress through the generation of oxygen-centered free radicals that reduced the bioavailability of endothelium-derived NO without prior interaction with the lung or vascular tissue. These findings provide a mechanism for the adverse cardiovascular effects of particulate air pollution.


Assuntos
Poluentes Atmosféricos/toxicidade , Endotélio Vascular/efeitos dos fármacos , Óxido Nítrico/metabolismo , Material Particulado/toxicidade , Superóxidos/metabolismo , Emissões de Veículos/toxicidade , Animais , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiologia , Sequestradores de Radicais Livres/toxicidade , Técnicas In Vitro , Masculino , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar , Superóxido Dismutase/metabolismo , Vasodilatação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA