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










Base de datos
Intervalo de año de publicación
1.
Mar Pollut Bull ; 179: 113684, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35489094

RESUMEN

Ultraviolet (UV) radiation can significantly increase the toxicity of polycyclic aromatic hydrocarbons (PAHs) in crude oil to early life stage (ELS) fishes through photo-induced /photo-enhanced toxicity. However, little is known about the sub-lethal effects and mechanisms of photo-induced PAH toxicity in ELS fishes. The present study investigated apoptosis and global transcriptomic effects in larval red drum (Sciaenops ocellatus) (24-72 h post-fertilization) following co-exposure to oil (0.29-0.30 µg/L ∑PAH50) and UV. Apoptosis was quantified using the TUNEL assay, and transcriptomic effects were assessed using RNA sequencing analysis. Apoptotic fluorescence was greatest in the eyes and skin following 24 and 48 h co-exposure to oil and UV, indicating photo-induced toxicity. Consistent with these phenotypic responses, pathways associated with phototransduction, eye development, and dermatological disease were among the top predicted pathways impacted. The present study is the first to provide global transcriptomic analysis of UV and oil co-exposure in an ELS fish.


Asunto(s)
Perciformes , Contaminación por Petróleo , Petróleo , Hidrocarburos Policíclicos Aromáticos , Contaminantes Químicos del Agua , Animales , Apoptosis , Peces , Larva , Perciformes/fisiología , Petróleo/análisis , Petróleo/toxicidad , Contaminación por Petróleo/análisis , Hidrocarburos Policíclicos Aromáticos/análisis , Transcriptoma , Contaminantes Químicos del Agua/análisis
2.
Neurotoxicol Teratol ; 90: 107071, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35016995

RESUMEN

Epidemiology studies suggest that exposure to ambient air pollution is associated with demyelinating diseases in the central nervous system (CNS), including multiple sclerosis (MS). The pathophysiology of MS results from an autoimmune response involving increased inflammation and demyelination in the CNS, which is higher in young (adult) females. Exposure to traffic-generated air pollution is associated with neuroinflammation and other detrimental outcomes in the CNS; however, its role in the progression of pathologies associated with demyelinating diseases has not yet been fully characterized in a female model. Thus, we investigated the effects of inhalation exposure to mixed vehicle emissions (MVE) in the brains of both ovary-intact (ov+) and ovariectomized (ov-) female Apolipoprotein (ApoE-/-) mice. Ov + and ov- ApoE-/- mice were exposed via whole-body inhalation to either filtered air (FA, controls) or mixed gasoline and diesel vehicle emissions (MVE: 200 PM µg/m3) for 6 h/d, 7 d/wk., for 30 d. We then analyzed MVE-exposure mediated alterations in myelination, the presence of CD4+ and CD8+ T cells, reactive oxygen species (ROS), myelin oligodendrocyte protein (MOG), and expression of estrogen (ERα and ERß) and progesterone (PROA/B) receptors in the CNS. MVE-exposure mediated significant alterations in myelination across multiple regions in the cerebrum, as well as increased CD4+ and CD8+ staining. There was also an increase in ROS production in the CNS of MVE-exposed ov- and ov + ApoE-/- mice. Ov- mice displayed a reduction in cerebral ERα mRNA expression, compared to ov + mice; however, MVE exposure resulted in an even further decrease in ERα expression, while ERß and PRO A/B were unchanged across groups. These findings collectively suggest that inhaled MVE-exposure may mediate estrogen receptor expression alterations associated with increased CD4+/CD8+ infiltration, regional demyelination, and ROS production in the CNS of female ApoE-/- mice.


Asunto(s)
Contaminación del Aire , Enfermedades Desmielinizantes , Contaminación del Aire/efectos adversos , Animales , Apolipoproteínas E/genética , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/genética , Modelos Animales de Enfermedad , Receptor alfa de Estrógeno/genética , Receptor beta de Estrógeno , Femenino , Ratones , Especies Reactivas de Oxígeno , Emisiones de Vehículos/toxicidad
3.
Toxicol Rep ; 8: 846-862, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33948438

RESUMEN

Exposure to air pollution from traffic-generated sources is known to contribute to the etiology of inflammatory diseases, including cardiovascular disease (CVD) and obesity; however, the signaling pathways involved are still under investigation. Dysregulation of the renin-angiotensin system (RAS) can contribute to CVD and alter lipid storage and inflammation in adipose tissue. Our previous exposure studies revealed that traffic-generated emissions increase RAS signaling, further exacerbated by a high-fat diet. Thus, we investigated the hypothesis that exposure to engine emissions increases systemic and local adipocyte RAS signaling, promoting the expression of factors involved in CVD and obesity. Male C57BL/6 mice (6-8 wk old) were fed either a high-fat (HF, n = 16) or low-fat (LF, n = 16) diet, beginning 30d prior to exposures, and then exposed via inhalation to either filtered air (FA, controls) or a mixture of diesel engine + gasoline engine vehicle emissions (MVE: 100 µg PM/m3) via whole-body inhalation for 6 h/d, 7 d/wk, 30d. Endpoints were assessed via immunofluorescence and RT-qPCR. MVE-exposure promoted vascular adhesion factors (VCAM-1, ICAM-1) expression, monocyte/macrophage sequestration, and oxidative stress in the vasculature, associated with increased angiotensin II receptor type 1 (AT1) expression. In the kidney, MVE-exposure promoted the expression of renin, AT1, and AT2 receptors. In adipose tissue, both HF-diet and MVE-exposure mediated increased epididymal fat pad weight and adipocyte hypertrophy, associated with increased angiotensinogen and AT1 receptor expression; however, these outcomes were further exacerbated in the MVE + HF group. MVE-exposure also induced inflammation, monocyte chemoattractant protein (MCP)-1, and leptin, while reducing insulin receptor and glucose transporter, GLUT4, expression in adipose tissue. Our results indicate that MVE-exposure promotes systemic and local adipose RAS signaling, associated with increased expression of factors contributing to CVD and obesity, further exacerbated by HF diet consumption.

4.
Toxicol Lett ; 339: 39-50, 2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33373663

RESUMEN

Traffic-generated air pollutants have been correlated with alterations in blood-brain barrier (BBB) integrity, which is associated with pathologies in the central nervous system (CNS). Much of the existing literature investigating the effects of air pollution in the CNS has predominately been reported in males, with little known regarding the effects in females. As such, this study characterized the effects of inhalation exposure to mixed vehicle emissions (MVE), as well as the presence of female sex hormones, in the CNS of female ApoE-/- mice, which included cohorts of both ovariectomized (ov-) and ovary-intact (ov+) mice. Ov + and ov- were placed on a high-fat diet and randomly grouped to be exposed to either filtered-air (FA) or MVE (200 PM/m3: 50 µg PM/m3 gasoline engine + 150 µg PM/m3 from diesel engine emissions) for 6 h/d, 7d/wk, for 30d. MVE-exposure resulted in altered cerebral microvascular integrity and permeability, as determined by the decreased immunofluorescent expression of tight junction (TJ) proteins, occludin, and claudin-5, and increased IgG extravasation into the cerebral parenchyma, compared to FA controls, regardless of ovary status. Associated with the altered cerebral microvascular integrity, we also observed an increase in matrix metalloproteinases (MMPs) -2/9 activity in the MVE ov+, MVE ov-, and FA ov- groups, compared to FA ov+. There was also elevated expression of intracellular adhesion molecule (ICAM)-1, inflammatory interleukins (IL-1, IL-1ß), and tumor necrosis factor (TNF-α) mRNA in the cerebrum of MVE ov + and MVE ov- animals. IκB kinase (IKK) subunits IKKα and IKKß mRNA expressions were upregulated in the cerebrum of MVE ov- and FA ov- mice. Our findings indicate that MVE exposure mediates altered integrity of the cerebral microvasculature correlated with increased MMP-2/9 activity and inflammatory signaling, regardless of female hormones present.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Encéfalo/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Inflamación/inducido químicamente , Ratones/genética , Microvasos/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Animales , Apolipoproteínas E/efectos de los fármacos , Femenino , Humanos , Modelos Animales , Fragmentos de Péptidos/efectos de los fármacos
5.
Environ Sci Technol ; 54(5): 2843-2850, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32036658

RESUMEN

Polycyclic aromatic hydrocarbons (PAHs) present in crude oil are known to impair visual development in fish. However, the underlying mechanism of PAH-induced toxicity to the visual system of fish is not understood. Embryonic zebrafish (Danio rerio) at 4 h post fertilization were exposed to weathered crude oil and assessed for visual function using an optokinetic response, with subsequent samples taken for immunohistochemistry and gene expression analysis. Cardiotoxicity was also assessed by measuring the heart rate, stroke volume, and cardiac output, as cardiac performance has been proposed to be a contributing factor to eye-associated malformations following oil exposure. Larvae exposed to the highest concentrations of crude oil (89.8 µg/L) exhibited an increased occurrence of bradycardia, though no changes in stroke volume or cardiac output were observed. However, genes important in eye development and phototransduction were downregulated in oil-exposed larvae, with an increased occurrence of cellular apoptosis, reduced neuronal connection, and reduced optokinetic behavioral response in zebrafish larvae.


Asunto(s)
Contaminación por Petróleo , Petróleo , Hidrocarburos Policíclicos Aromáticos , Contaminantes Químicos del Agua , Animales , Apoptosis , Pez Cebra
6.
Environ Res ; 181: 108913, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31753468

RESUMEN

Air pollution exposure is known to contribute to the progression of cardiovascular disease (CVD) and there is increasing evidence that dysbiosis of the gut microbiome may also play a role in the pathogenesis of CVD, including atherosclerosis. To date, the effects of inhaled air pollution mixtures on the intestinal epithelial barrier (IEB), and microbiota profiles are not well characterized, especially in susceptible individuals with comorbidity. Thus, we investigated the effects of inhaled ubiquitous air-pollutants, wood-smoke (WS) and mixed diesel and gasoline vehicle exhaust (MVE) on alterations in the expression of markers of integrity, inflammation, and microbiota profiles in the intestine of atherosclerotic Apolipoprotein E knockout (ApoE-/-) mice. To do this, male 8 wk-old ApoE-/- mice, on a high-fat diet, were exposed to either MVE (300 µg/m3 PM), WS; (∼450 µg/m3 PM), or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-PCR were used to quantify the expression of IEB components and inflammatory factors, including mucin (Muc)-2, tight junction (TJ) proteins, matrix metalloproteinase (MMP)-9, tumor necrosis factor (TNF)-α, and interleukin (IL)-1ß, as well as Toll-like receptor (TLR)-4. Microbial profiling of the intestine was done using Illumina 16S sequencing of V4 16S rRNA PCR amplicons. We observed a decrease in intestinal Muc2 and TJ proteins in both MVE and WS exposures, compared to FA controls, associated with a significant increase in MMP-9, TLR-4, and inflammatory marker expression. Both WS and MVE-exposure resulted in decreased intestinal bacterial diversity, as well as alterations in microbiota profiles, including the Firmicutes: Bacteroidetes ratio at the phylum level. Our findings suggest inhalation exposure to either MVE or WS result in alterations in components involved in mucosal integrity, and also microbiota profiles and diversity, which are associated with increased markers of an inflammatory response.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Apolipoproteínas E , Microbioma Gastrointestinal , Contaminación del Aire , Animales , Inflamación , Intestinos , Masculino , Ratones , Ratones Noqueados , ARN Ribosómico 16S , Emisiones de Vehículos
7.
Toxicol Sci ; 170(2): 525-535, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31132127

RESUMEN

Exposure to traffic-generated pollution is associated with alterations in blood-brain barrier (BBB) integrity and exacerbation of cerebrovascular disorders. Angiotensin (Ang) II signaling through the Ang II type 1 (AT1) receptor is known to promote BBB disruption. We have previously reported that exposure to a mixture of gasoline and diesel vehicle engine emissions (MVE) mediates alterations in cerebral microvasculature of C57Bl/6 mice, which is exacerbated through consumption of a high-fat (HF) diet. Thus, we investigated the hypothesis that inhalation exposure to MVE results in altered central nervous system microvascular integrity mediated by Ang II-AT1 signaling. Three-month-old male C57Bl/6 mice were placed on an HF or low-fat diet and exposed via inhalation to either filtered air (FA) or MVE (100 µg/m3 PM) 6 h/d for 30 days. Exposure to HF+MVE resulted in a significant increase in plasma Ang II and expression of AT1 in the cerebral microvasculature. Results from a BBB coculture study showed that transendothelial electrical resistance was decreased, associated with reduced expression of claudin-5 and occludin when treated with plasma from MVE+HF animals. These effects were attenuated through pretreatment with the AT1 antagonist, Losartan. Our BBB coculture showed increased levels of astrocyte AT1 and decreased expression of aryl hydrocarbon receptor and glutathione peroxidase-1, associated with increased interleukin-6 and transforming growth factor-ß in the astrocyte media, when treated with plasma from MVE-exposed groups. Our results indicate that inhalation exposure to traffic-generated pollutants results in altered BBB integrity, mediated through Ang II-AT1 signaling and inflammation, which is exacerbated by an HF diet.


Asunto(s)
Angiotensina II/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Contaminantes Atmosféricos/toxicidad , Angiotensina II/metabolismo , Animales , Astrocitos/efectos de los fármacos , Circulación Cerebrovascular , Técnicas de Cocultivo , Dieta Alta en Grasa , Expresión Génica/efectos de los fármacos , Inflamación , Exposición por Inhalación/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas de Uniones Estrechas/metabolismo
8.
Toxicol Lett ; 296: 48-62, 2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-30081225

RESUMEN

Exposure to multi-walled carbon nanotubes (MWCNTs) has been associated with detrimental cardiovascular outcomes; however, underlying mechanisms have not yet been fully elucidated. Thus, we investigated alterations in proatherogenic and proinflammatory signaling pathways in C57Bl6/ mice exposed to MWCNTs (1 mg/m3) or filtered air (FA-Controls), via inhalation, for 6 h/day, 14d. Expression of mediators of cholesterol transport, namely the lectin-like oxidized low-density lipoprotein receptor (LOX)-1 and ATP-binding cassette transporter (ABCA)-1, inflammatory markers tumor necrosis factor (TNF)-α and interleukin (IL)-1ß/IL-6, nuclear-factor kappa-light-chain-enhancer of activated B cells (NF-κB), intracellular/vascular adhesion molecule(s) (VCAM-1, ICAM-1), and miRNAs (miR-221/-21/-1), associated with cardiovascular disease (CVD), were analyzed in cardiac tissue and coronary vasculature. Cardiac fibrotic deposition, matrix-metalloproteinases (MMP)-2/9, and reactive oxygen species (ROS) were also assessed. MWCNT-exposure resulted in increased coronary ROS production with concurrent increases in expression of LOX-1, VCAM-1, TNF-α, and MMP-2/9 activity; while ABCA-1 expression was downregulated, compared to FA-Controls. Additionally, trends in fibrotic deposition and induction of cardiac TNF-α, MMP-9, IκB Kinase (IKK)-α/ß, and miR-221 mRNA expression were observed. Analysis using inhibitors for nitric oxide synthase or NADPH oxidase resulted in attenuated coronary ROS production. These findings suggest that subacute inhalation MWCNT-exposure alters expression of cholesterol transporter/receptors, and induces signaling pathways associated with inflammation, oxidative stress, and CVD in wild-type mice.


Asunto(s)
Colesterol/metabolismo , Mediadores de Inflamación/metabolismo , Nanotubos de Carbono/toxicidad , Transducción de Señal/efectos de los fármacos , Transportador 1 de Casete de Unión a ATP/biosíntesis , Administración por Inhalación , Animales , Enfermedades Cardiovasculares/metabolismo , Moléculas de Adhesión Celular/biosíntesis , Colágeno/metabolismo , Interleucinas/biosíntesis , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno , Receptores Depuradores de Clase E/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis
9.
Environ Res ; 160: 449-461, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29073573

RESUMEN

Air pollution-exposure is associated with detrimental outcomes in the central nervous system (CNS) such as cerebrovascular disorders, including stroke, and neurodegenerative diseases. While the mechanisms of these CNS-related outcomes involved have not been fully elucidated, exposure to traffic-generated air pollutants has been associated with altered blood brain barrier (BBB) integrity and permeability. The current study investigated whether inhalation exposure to mixed vehicle emissions (MVE) alters cerebral microvascular integrity in healthy 3 mo old C57BL/6 mice, as well as whether exposure-mediated effects were exacerbated by a high-fat (HF) vs. low-fat (LF) diet. Mice on each diet were randomly assigned to be exposed to either filtered air (FA) or MVE [100PM/m3 vehicle emissions mixture: 30µg PM/m3 gasoline engine + 70µg PM/m3 diesel engine emissions; median size ~ 60nm; particle mass size distribution median of ~ 1µm (range: < 0.5-20µm)] for 6h/d, 7d/wk, for 30d. Using sodium fluorescein as a tracer, we observed a significant increase in BBB permeability in both HF + MVE exposed and HF + FA animals, compared to LF + FA controls. Exposure to HF + MVE also led to a significant increase plasma ox-LDL and ox-LDL scavenger receptors (LOX-1 and CD-36) expression in the cerebral vasculature. Histological analysis revealed decreased expression of TJ protein, claudin-5, associated with increased matrix metalloproteinase (MMP)-9 activity and oxidative stress in the cerebral vasculature of HF + MVE mice, compared to LF + MVE. Such findings indicate that inhalation exposure to traffic-generated pollutants, coupled with a HF diet, results in altered BBB integrity and increased ox-LDL signaling in the cerebral vasculature in a wildtype animal model.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Cerebro/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Emisiones de Vehículos/toxicidad , Animales , Cerebro/irrigación sanguínea , Cerebro/patología , Dieta Alta en Grasa , Masculino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
10.
Inhal Toxicol ; 29(6): 266-281, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28816559

RESUMEN

Recent studies have shown a strong correlation between air pollution-exposure and detrimental outcomes in the central nervous system, including alterations in blood brain barrier (BBB) integrity, neuroinflammation, and neurodegeneration. However, the mechanisms mediating these pathologies have not yet been fully elucidated. We have previously reported that exposure to traffic-generated air pollution results in increased circulating oxidized low-density lipoprotein (oxLDL), associated with alterations in BBB integrity, in atherosclerotic Apolipoprotein E null (ApoE-/-) mice. Thus, we investigated the role of the lectin-like oxLDL receptor (LOX)-1 in mediating these deleterious effects in ApoE-/- mice exposed to a mixture of gasoline and diesel engine exhaust (MVE: 100 PM µg/m3) for 6 h/d, 7d/week, for 30 d by inhalation. Concurrent with exposures, a subset of mice were treated with neutralizing antibodies to LOX-1 (LOX-1 Ab) i.p., or IgG (control) i.p., every other day during exposures. Resulting brain microvascular integrity, tight junction (TJ) protein expression, matrix metalloproteinase (MMP)-9/-2 activity, ROS, and markers of cellular adhesion and monocyte/macrophage sequestration were assessed. MVE-exposure resulted in decreased BBB integrity and alterations in microvascular TJ protein expression, associated with increased LOX-1 expression, MMP-9/-2 activities, and lipid peroxidation, each of which was attenuated with LOX-1 Ab treatment. Furthermore, MVE-exposure induced cerebral microvascular ROS and adhesion molecules, expression of which was not normalized through LOX-1 Ab-treatment. Such findings suggest that alterations in brain microvascular structure and integrity observed with MVE-exposure may be mediated, at least in part, via LOX-1 signaling.


Asunto(s)
Encéfalo/efectos de los fármacos , Microvasos/efectos de los fármacos , Receptores Depuradores de Clase E/metabolismo , Emisiones de Vehículos/toxicidad , Contaminación del Aire/efectos adversos , Animales , Anticuerpos Neutralizantes/farmacología , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Quimiocina CCL2/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Noqueados para ApoE , Microvasos/metabolismo , Permeabilidad , Especies Reactivas de Oxígeno/metabolismo , Receptores Depuradores de Clase E/inmunología , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Inhal Toxicol ; 26(6): 353-60, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24730681

RESUMEN

Cardiovascular health effects of near-roadway pollution appear more substantial than other sources of air pollution. The underlying cause of this phenomenon may simply be concentration-related, but the possibility remains that gases and particulate matter (PM) may physically interact and further enhance systemic vascular toxicity. To test this, we utilized a common hypercholesterolemic mouse model (Apolipoprotein E-null) exposed to mixed vehicle emission (MVE; combined gasoline and diesel exhausts) for 6 h/d × 50 d, with additional permutations of removing PM by filtration and also removing gaseous species from PM by denudation. Several vascular bioassays, including matrix metalloproteinase-9 protein, 3-nitrotyrosine and plasma-induced vasodilatory impairments, highlighted that the whole emissions, containing both particulate and gaseous components, was collectively more potent than MVE-derived PM or gas mixtures, alone. Thus, we conclude that inhalation of fresh whole emissions induce greater systemic vascular toxicity than either the particulate or gas phase alone. These findings lend credence to the hypothesis that the near-roadway environment may have a more focused public health impact due to gas-particle interactions.


Asunto(s)
Material Particulado/toxicidad , Vasodilatación/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Acetilcolina/farmacología , Animales , Líquido del Lavado Bronquioalveolar/química , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Material Particulado/análisis , Tirosina/análogos & derivados , Tirosina/análisis , Emisiones de Vehículos/análisis
12.
Part Fibre Toxicol ; 10: 62, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24344990

RESUMEN

BACKGROUND: Traffic-generated air pollution-exposure is associated with adverse effects in the central nervous system (CNS) in both human exposures and animal models, including neuroinflammation and neurodegeneration. While alterations in the blood brain barrier (BBB) have been implicated as a potential mechanism of air pollution-induced CNS pathologies, pathways involved have not been elucidated. OBJECTIVES: To determine whether inhalation exposure to mixed vehicle exhaust (MVE) mediates alterations in BBB permeability, activation of matrix metalloproteinases (MMP) -2 and -9, and altered tight junction (TJ) protein expression. METHODS: Apolipoprotein (Apo) E(-/-) and C57Bl6 mice were exposed to either MVE (100 µg/m(3) PM) or filtered air (FA) for 6 hr/day for 30 days and resulting BBB permeability, expression of ROS, TJ proteins, markers of neuroinflammation, and MMP activity were assessed. Serum from study mice was applied to an in vitro BBB co-culture model and resulting alterations in transport and permeability were quantified. RESULTS: MVE-exposed Apo E(-/-) mice showed increased BBB permeability, elevated ROS and increased MMP-2 and -9 activity, compared to FA controls. Additionally, cerebral vessels from MVE-exposed mice expressed decreased levels of TJ proteins, occludin and claudin-5, and increased levels of inducible nitric oxide synthase (iNOS) and interleukin (IL)-1ß in the parenchyma. Serum from MVE-exposed animals also resulted in increased in vitro BBB permeability and altered P-glycoprotein transport activity. CONCLUSIONS: These data indicate that inhalation exposure to traffic-generated air pollutants promotes increased MMP activity and degradation of TJ proteins in the cerebral vasculature, resulting in altered BBB permeability and expression of neuroinflammatory markers.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Exposición por Inhalación/efectos adversos , Metaloproteinasas de la Matriz/genética , Proteínas de Uniones Estrechas/genética , Emisiones de Vehículos/toxicidad , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/sangre , Animales , Apolipoproteínas E/genética , Barrera Hematoencefálica/enzimología , Barrera Hematoencefálica/metabolismo , Western Blotting , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inflamación Neurogénica/sangre , Inflamación Neurogénica/inducido químicamente , Inflamación Neurogénica/enzimología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/patología
13.
Am J Physiol Lung Cell Mol Physiol ; 301(3): L315-26, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21685241

RESUMEN

Human immunodeficiency virus (HIV)-associated pulmonary arterial hypertension (PAH) is a serious noninfectious disease involving an aberrant increase in pressure in the blood vessels of the lung, which leads to right ventricular (RV) heart failure and can eventually result in death. A lack of viable animal models of HIV-PAH has limited the identification of signaling pathways involved in HIV-mediated onset and progression of PAH. To determine whether the HIV-1 transgenic (HIV Tg) rat displays pathophysiological end points associated with PAH, we evaluated peak RV systolic pressure (RVSP), RV hypertrophy, pulmonary vessel remodeling, and alterations in gene expression by real-time PCR and microarray. RVSP was measured by RV catheterization via the right jugular vein in 3- and 9-mo-old HIV Tg and age-matched Fischer 344 (control) male rats while under 2% isoflurane anesthesia. RVSP was elevated in the HIV Tg rats (34.2 ± 2.5 mmHg) compared with the F344 controls (21.2 ± 2.5 mmHg), with more significant elevations in the 9-mo-old HIV Tg rats (42.5 ± 3.7 mmHg). We observed significant increases in RV wall thickness in HIV Tg rats compared with controls, both histologically and by echocardiograph measurement. HIV Tg rats also show increased thickening of the pulmonary artery and remodeling of small pulmonary arteries, as well as altered expression of gene pathways associated with PAH. These data represent the first analysis of PAH in HIV Tg rats and suggest that this model will be useful for investigating pathways and identifying potential therapies for HIV-PAH.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Hipertensión Pulmonar Primaria Familiar , Perfilación de la Expresión Génica , Infecciones por VIH/patología , Infecciones por VIH/fisiopatología , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Masculino , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Ratas , Ratas Endogámicas F344 , Ratas Transgénicas , Función Ventricular Derecha
14.
Am J Respir Crit Care Med ; 184(1): 82-91, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21493736

RESUMEN

RATIONALE: To determine vascular signaling pathways involved in inhaled air pollution (vehicular engine emission) exposure-induced exacerbation of atherosclerosis that are associated with onset of clinical cardiovascular events. OBJECTIVES: To elucidate the role of oxidized low-density lipoprotein (oxLDL) and its primary receptor on endothelial cells, the lectin-like oxLDL receptor (LOX-1), in regulation of endothelin-1 expression and matrix metalloproteinase activity associated with inhalational exposure to vehicular engine emissions. METHODS: Atherosclerotic apolipoprotein E knockout mice were exposed by inhalation to filtered air or mixed whole engine emissions (250 µg particulate matter [PM]/m(3) diesel + 50 µg PM/m(3) gasoline exhausts) 6 h/d for 7 days. Concurrently, mice were treated with either mouse IgG or neutralizing antibodies to LOX-1 every other day. Vascular and plasma markers of oxidative stress and expression proatherogenic factors were assessed. In a parallel study, healthy human subjects were exposed to either 100 µg PM/m(3) diesel whole exhaust or high-efficiency particulate air and charcoal-filtered "clean" air (control subjects) for 2 hours, on separate occasions. MEASUREMENTS AND MAIN RESULTS: Mixed emissions exposure increased oxLDL and vascular reactive oxygen species, as well as LOX-1, matrix metalloproteinase-9, and endothelin-1 mRNA expression and also monocyte/macrophage infiltration, each of which was attenuated with LOX-1 antibody treatment. In a parallel study, diesel exhaust exposure in volunteer human subjects induced significant increases in plasma-soluble LOX-1. CONCLUSIONS: These findings demonstrate that acute exposure to vehicular source pollutants results in up-regulation of vascular factors associated with progression of atherosclerosis, endothelin-1, and matrix metalloproteinase-9, mediated through oxLDL-LOX-1 receptor signaling, which may serve as a novel target for future therapy.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Aorta/metabolismo , Receptores Depuradores de Clase E/metabolismo , Emisiones de Vehículos , Adolescente , Adulto , Animales , Anticuerpos Neutralizantes/farmacología , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Endotelina-1/metabolismo , Endotelio Vascular/metabolismo , Humanos , Peroxidación de Lípido , Lipoproteínas LDL/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Receptores Depuradores de Clase E/sangre , Receptores Depuradores de Clase E/inmunología , Transducción de Señal , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Regulación hacia Arriba , Adulto Joven
15.
Arterioscler Thromb Vasc Biol ; 29(4): 511-7, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19150882

RESUMEN

OBJECTIVE: Mechanisms of air pollution-induced exacerbation of cardiovascular disease are currently unknown, thus we examined the roles of vascular endothelin-1 (ET-1) and reactive oxygen species (ROS) in regulating mediators of vascular remodeling, namely matrix metalloproteinases (MMPs), after exposure to vehicle engine emissions. METHODS AND RESULTS: ApoE(-/-) mice were exposed by inhalation to filtered air or gasoline engine exhaust (GEE, 1:12 dilution) 6 hours per day for 1 or 7 days. Concurrently, mice were treated with either ET(A) receptor antagonist BQ-123 (100 ng/kg/d) via osmotic minipumps, Tempol (approximately 41 mg/kg/d, orally), or vehicle. GEE-exposure increased vascular MMP-2 and -9, endothelin-1 (ET-1), tissue inhibitor of metalloproteinases (TIMP)-2 mRNA and ROS levels. Aortic MMP protein and plasma MMP-9 were similarly upregulated. GEE-mediated increases in vascular ROS were attenuated by Tempol-treatment, as were MMP-2 and TIMP-2; whereas BQ-123 ameliorated GEE-induced vascular expression of MMP-9, MMP-2, ROS, and ET-1. In a parallel study, diesel exhaust exposure in volunteer human subjects induced significant increases in plasma ET-1 and MMP-9 expression and activity. CONCLUSIONS: These findings demonstrate that acute exposure to vehicular source air pollutants results in upregulation of circulating and vascular factors associated with progression of atherosclerosis, mediated in part through activation of ET-1-ET(A) receptor pathways.


Asunto(s)
Aorta/efectos de los fármacos , Aterosclerosis/inducido químicamente , Endotelina-1/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Emisiones de Vehículos/toxicidad , Administración Oral , Adolescente , Adulto , Animales , Antioxidantes/administración & dosificación , Aorta/enzimología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/enzimología , Aterosclerosis/genética , Óxidos N-Cíclicos/administración & dosificación , Endotelina-1/sangre , Endotelina-1/genética , Femenino , Humanos , Bombas de Infusión Implantables , Exposición por Inhalación , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/sangre , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Noqueados , Nitratos/metabolismo , Nitritos/metabolismo , Péptidos Cíclicos/administración & dosificación , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Endotelina A/efectos de los fármacos , Receptor de Endotelina A/metabolismo , Marcadores de Spin , Factores de Tiempo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Regulación hacia Arriba , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA