Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Inhal Toxicol ; 35(3-4): 86-100, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35037817

RESUMEN

OBJECTIVE: Environmental exposures exacerbate age-related pathologies, such as cardiovascular and neurodegenerative diseases. Nanoparticulates, and specifically carbon nanomaterials, are a fast-growing contributor to the category of inhalable pollutants, whose risks to health are only now being unraveled. The current study assessed the exacerbating effect of age on multiwalled-carbon nanotube (MWCNT) exposure in young and old C57BL/6 and ApoE-/- mice. MATERIALS AND METHODS: Female C57BL/6 and apolipoprotein E-deficient (ApoE-/-) mice, aged 8 weeks and 15 months, were exposed to 0 or 40 µg MWCNT via oropharyngeal aspiration. Pulmonary inflammation, inflammatory bioactivity of serum, and neurometabolic changes were assessed at 24 h post-exposure. RESULTS: Pulmonary neutrophil infiltration was induced by MWCNT in bronchoalveolar lavage fluid in both C57BL/6 and ApoE-/-. Macrophage counts decreased with MWCNT exposure in ApoE-/- mice but were unaffected by exposure in C57BL/6 mice. Older mice appeared to have greater MWCNT-induced total protein in lavage fluid. BALF cytokines and chemokines were elevated with MWCNT exposure, but CCL2, CXCL1, and CXCL10 showed reduced responses to MWCNT in older mice. However, no significant serum inflammatory bioactivity was detected. Cerebellar metabolic changes in response to MWCNT were modest, but age and strain significantly influenced metabolite profiles assessed. ApoE-/- mice and older mice exhibited less robust metabolite changes in response to exposure, suggesting a reduced health reserve. CONCLUSIONS: Age influences the pulmonary and neurological responses to short-term MWCNT exposure. However, with only the model of moderate aging (15 months) in this study, the responses appeared modest compared to inhaled toxicant impacts in more advanced aging models.


Asunto(s)
Nanotubos de Carbono , Femenino , Animales , Ratones , Nanotubos de Carbono/toxicidad , Ratones Endogámicos C57BL , Pulmón , Líquido del Lavado Bronquioalveolar , Inflamación/patología , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Apolipoproteínas E/farmacología , Apolipoproteínas/metabolismo , Apolipoproteínas/farmacología , Exposición por Inhalación/efectos adversos
2.
Part Fibre Toxicol ; 18(1): 34, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496918

RESUMEN

BACKGROUND: Multiwalled carbon nanotubes (MWCNT) are an increasingly utilized engineered nanomaterial that pose the potential for significant risk of exposure-related health outcomes. The mechanism(s) underlying MWCNT-induced toxicity to extrapulmonary sites are still being defined. MWCNT-induced serum-borne bioactivity appears to dysregulate systemic endothelial cell function. The serum compositional changes after MWCNT exposure have been identified as a surge of fragmented endogenous peptides, likely derived from matrix metalloproteinase (MMP) activity. In the present study, we utilize a broad-spectrum MMP inhibitor, Marimastat, along with a previously described oropharyngeal aspiration model of MWCNT administration to investigate the role of MMPs in MWCNT-derived serum peptide generation and endothelial bioactivity. RESULTS: C57BL/6 mice were treated with Marimastat or vehicle by oropharyngeal aspiration 1 h prior to MWCNT treatment. Pulmonary neutrophil infiltration and total bronchoalveolar lavage fluid protein increased independent of MMP blockade. The lung cytokine profile similarly increased following MWCNT exposure for major inflammatory markers (IL-1ß, IL-6, and TNF-α), with minimal impact from MMP inhibition. However, serum peptidomic analysis revealed differential peptide compositional profiles, with MMP blockade abrogating MWCNT-derived serum peptide fragments. The serum, in turn, exhibited differential potency in terms of inflammatory bioactivity when incubated with primary murine cerebrovascular endothelial cells. Serum from MWCNT-treated mice led to inflammatory responses in endothelial cells that were significantly blunted with serum from Marimastat-treated mice. CONCLUSIONS: Thus, MWCNT exposure induced pulmonary inflammation that was largely independent of MMP activity but generated circulating bioactive peptides through predominantly MMP-dependent pathways. This MWCNT-induced lung-derived bioactivity caused pathological consequences of endothelial inflammation and barrier disruption.


Asunto(s)
Nanotubos de Carbono , Neumonía , Animales , Líquido del Lavado Bronquioalveolar , Células Endoteliales , Ácidos Hidroxámicos , Pulmón , Inhibidores de la Metaloproteinasa de la Matriz/toxicidad , Ratones , Ratones Endogámicos C57BL , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente
3.
J Toxicol Environ Health A ; 84(12): 503-517, 2021 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-33682625

RESUMEN

The Southwestern United States has a legacy of industrial mining due to the presence of rich mineral ore deposits. The relationship between environmental inhaled particulate matter (PM) exposures and neurological outcomes within an autoimmune context is understudied. The aim of this study was to compare two regionally-relevant dusts from high-priority abandoned mine-sites, Claim 28 PM, from Blue Gap Tachee, AZ and St. Anthony mine PM, from the Pueblo of Laguna, NM and to expose autoimmune-prone mice (NZBWF1/J). Mice were randomly assigned to one of three groups (n = 8/group): DM (dispersion media, control), Claim 28 PM, or St. Anthony PM, subjected to oropharyngeal aspiration of (100 µg/50 µl), once per week for a total of 4 consecutive doses. A battery of immunological and neurological endpoints was assessed at 24 weeks of age including: bronchoalveolar lavage cell counts, lung gene expression, brain immunohistochemistry, behavioral tasks and serum autoimmune biomarkers. Bronchoalveolar lavage results demonstrated a significant increase in number of polymorphonuclear neutrophils following Claim 28 and St. Anthony mine PM aspiration. Lung mRNA expression showed significant upregulation in CCL-2 and IL-1ß following St. Anthony mine PM aspiration. In addition, neuroinflammation was present in both Claim 28 and St. Anthony mine-site derived PM exposure groups. Behavioral tasks resulted in significant deficits as determined by Y-maze new arm frequency following Claim 28 aspiration. Neutrophil elastase was significantly upregulated in the St. Anthony mine exposure group. Interestingly, there were no significant changes in serum autoantigens suggesting systemic inflammatory effects may be mediated through other molecular mechanisms following low-dose PM exposures.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Polvo/análisis , Encefalitis/fisiopatología , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Material Particulado/toxicidad , Neumonía/fisiopatología , Animales , Arizona , Enfermedades Autoinmunes/etiología , Biomarcadores/metabolismo , Modelos Animales de Enfermedad , Polvo/inmunología , Encefalitis/inducido químicamente , Femenino , Exposición por Inhalación/efectos adversos , Ratones , Minería , New Mexico , Tamaño de la Partícula , Neumonía/inducido químicamente , Distribución Aleatoria
4.
Toxicol Appl Pharmacol ; 305: 40-45, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27286659

RESUMEN

Ozone (O3)-related cardiorespiratory effects are a growing public health concern. Ground level O3 can exacerbate pre-existing respiratory conditions; however, research regarding therapeutic interventions to reduce O3-induced lung injury is limited. In patients with chronic obstructive pulmonary disease, hypoxia-associated pulmonary hypertension (HPH) is a frequent comorbidity that is difficult to treat clinically, yet associated with increased mortality and frequency of exacerbations. In this study, we hypothesized that established HPH would confer vulnerability to acute O3 pulmonary toxicity. Additionally, we tested whether improvement of pulmonary endothelial barrier integrity via rho-kinase inhibition could mitigate pulmonary inflammation and injury. To determine if O3 exacerbated HPH, male C57BL/6 mice were subject to either 3 weeks continuous normoxia (20.9% O2) or hypoxia (10.0% O2), followed by a 4-h exposure to either 1ppm O3 or filtered air (FA). As an additional experimental intervention fasudil (20mg/kg) was administered intraperitoneally prior to and after O3 exposures. As expected, hypoxia significantly increased right ventricular pressure and hypertrophy. O3 exposure in normoxic mice caused lung inflammation but not injury, as indicated by increased cellularity and edema in the lung. However, in hypoxic mice, O3 exposure led to increased inflammation and edema, along with a profound increase in airway hyperresponsiveness to methacholine. Fasudil administration resulted in reduced O3-induced lung injury via the enhancement of pulmonary endothelial barrier integrity. These results indicate that increased pulmonary vascular pressure may enhance lung injury, inflammation and edema when exposed to pollutants, and that enhancement of pulmonary endothelial barrier integrity may alleviate such vulnerability.


Asunto(s)
1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , Contaminantes Atmosféricos/toxicidad , Lesión Pulmonar/tratamiento farmacológico , Ozono/toxicidad , Inhibidores de Proteínas Quinasas/uso terapéutico , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/uso terapéutico , Animales , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/tratamiento farmacológico , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Líquido del Lavado Bronquioalveolar/citología , Recuento de Células , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/inducido químicamente , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Hipoxia/tratamiento farmacológico , Hipoxia/patología , Hipoxia/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Lesión Pulmonar/fisiopatología , Masculino , Ratones Endogámicos C57BL , Tamaño de los Órganos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Función Ventricular Derecha/efectos de los fármacos , Presión Ventricular/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores
5.
J Toxicol Environ Health A ; 74(13): 828-37, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21598168

RESUMEN

Epidemiological studies have associated traffic-related airborne pollution with adverse cardiovascular outcomes. Nitric oxide (NO) is a common component of fresh diesel and gasoline engine emissions that rapidly transforms both in the atmosphere and once inhaled. Because of this rapid transformation, limited information is available in terms of potential human exposures and adverse health effects. Young rats were exposed to whole diesel emissions (DE) adjusted to 300 µg/m(3) of particulate matter (containing 3.5 ppm NO) or 0, 3, or 10 ppm NO as a positive control. Animals were also pre-injected (ip) with either saline or N-acetylcysteine (NAC), a precursor of glutathione. Predictably, pure NO exposures led to a concentration-dependent increase in plasma nitrates compared to controls, which lasted for roughly 4 h postexposure. Whole DE exposure for 1 h also led to a doubling of plasma NOx. NAC injection increased the levels of plasma nitrates and nitrites (NOx) in the DE exposure group. Inhibition of nitric oxide symthase (NOS) by N(G)-nitro-L-arginine (L-NNA) did not block the rise in plasma NOx, demonstrating that the increase was entirely due to exogenous sources. Both DE and pure NO exposures paradoxically led to elevated eNOS expression in aortic tissue. Furthermore, coronary arterioles from NO-exposed animals exhibited greater constriction to endothelin-1 compared to controls, consistent with a derangement of the NOS system. Thus, NO may be an important contributor to traffic-related cardiovascular morbidity, although further research is necessary for proper hazard identification.


Asunto(s)
Vasos Coronarios/química , Nitratos/sangre , Nitritos/sangre , S-Nitrosotioles/análisis , Emisiones de Vehículos/toxicidad , Animales , Aorta/química , Vasos Coronarios/metabolismo , Relación Dosis-Respuesta a Droga , Glutatión/metabolismo , Exposición por Inhalación/efectos adversos , Masculino , Nitratos/análisis , Óxido Nítrico/efectos adversos , Óxido Nítrico/sangre , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , Nitritos/análisis , Ratas , Ratas Sprague-Dawley , S-Nitrosotioles/sangre
6.
Toxicol Sci ; 184(2): 286-299, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34498067

RESUMEN

Inhalation of tungsten particulates is a relevant route of exposure in occupational and military settings. Exposure to tungsten alloys is associated with increased incidence of lung pathologies, including interstitial lung disease and cancer. We have demonstrated, oral exposure to soluble tungsten enhances breast cancer metastasis to the lungs through changes in the surrounding microenvironment. However, more research is required to investigate if changes in the lung microenvironment, following tungsten particulate exposure, can drive tumorigenesis or metastasis to the lung niche. This study examined if inhalation to environmentally relevant concentrations of tungsten particulates caused acute damage to the microenvironment in the lungs and/or systemically using a whole-body inhalation system. Twenty-four female BALB/c mice were exposed to Filtered Air, 0.60 mg/m3, or 1.7 mg/m3 tungsten particulates (<1 µm) for 4 h. Tissue samples were collected at days 1 and 7 post-exposure. Tungsten accumulation in the lungs persisted up to 7 days post-exposure and produced acute changes to the lung microenvironment including increased macrophage and neutrophil infiltration, increased levels of proinflammatory cytokines interleukin 1 beta and C-X-C motif chemokine ligand 1, and an increased percentage of activated fibroblasts (alpha-smooth muscle actin+). Exposure to tungsten also resulted in systemic effects on the bone, including tungsten deposition and transient increases in gene expression of proinflammatory cytokines. Taken together, acute whole-body inhalation of tungsten particulates, at levels commonly observed in occupational and military settings, resulted in changes to the lung and bone microenvironments that may promote tumorigenesis or metastasis and be important molecular drivers of other tungsten-associated lung pathologies such as interstitial lung disease.


Asunto(s)
Pulmón , Tungsteno , Administración por Inhalación , Animales , Polvo , Femenino , Exposición por Inhalación/efectos adversos , Pulmón/patología , Ratones , Infiltración Neutrófila , Tungsteno/metabolismo , Tungsteno/toxicidad
7.
Cardiovasc Toxicol ; 19(5): 401-411, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30963444

RESUMEN

Ambient particulate matter (PM) is associated with increased mortality and morbidity, an effect influenced by the metal components of the PM. We characterized five sediment samples obtained near a tungsten-molybdenum ore-processing complex in Zakamensk, Russia for elemental composition and PM toxicity with regard to pulmonary, vascular, and neurological outcomes. Elemental and trace metals analysis of complete sediment and PM10 (the respirable fraction, < 10 µm mass mean aerodynamic diameter) were performed using inductively coupled plasma optical emission spectrometry (ICP-OES) and mass spectrometry (ICP-MS). Sediment samples and PM10 consisted largely of silicon and iron and silicon and sodium, respectively. Trace metals including manganese and uranium in the complete sediment, as well as copper and lead in the PM10 were observed. Notably, metal concentrations were approximately 10 × higher in the PM10 than in the sediment. Exposure to 100 µg of PM10 via oropharyngeal aspiration in C56BL/6 mice resulted in pulmonary inflammation across all groups. In addition, mice exposed to three of the five PM10 samples exhibited impaired endothelial-dependent relaxation, and correlative analysis revealed associations between pulmonary inflammation and levels of lead and cadmium. A tendency for elevated cortical ccl2 and Tnf-α mRNA expression was induced by all samples and significant upregulation was noted following exposure to PM10 samples Z3 and Z4, respectively. Cortical Nqo1 mRNA levels were significantly upregulated in mice exposed to PM10 Z2. In conclusion, pulmonary exposure to PM samples from the Zakamensk region sediments induced varied pulmonary and systemic effects that may be influenced by elemental PM composition. Further investigation is needed to pinpoint putative drivers of neurological outcomes.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Aorta Torácica/efectos de los fármacos , Cadmio/toxicidad , Corteza Cerebral/efectos de los fármacos , Polvo , Plomo/toxicidad , Minería , Material Particulado/toxicidad , Neumonía/inducido químicamente , Animales , Aorta Torácica/fisiopatología , Corteza Cerebral/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Exposición por Inhalación , Masculino , Ratones Endogámicos C57BL , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Neumonía/genética , Neumonía/metabolismo , Medición de Riesgo , Siberia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Vasodilatación/efectos de los fármacos
8.
Toxicol Sci ; 146(2): 244-53, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25962394

RESUMEN

Ambient ozone (O3) levels are associated with cardiovascular morbidity and mortality, but the underlying pathophysiological mechanisms driving extrapulmonary toxicity remain unclear. This study examined the coronary vascular bed of rats in terms of constrictive and dilatory responses to known agonists following a single O3 inhalation exposure. In addition, serum from exposed rats was used in ex vivo preparations to examine whether bioactivity and toxic effects of inhaled O3 could be conveyed to extrapulmonary systems via the circulation. We found that 24 h following inhalation of 1 ppm O3, isolated coronary vessels exhibited greater basal tone and constricted to a greater degree to serotonin stimulation. Vasodilation to acetylcholine (ACh) was markedly diminished in coronary arteries from O3-exposed rats, compared with filtered air-exposed controls. Dilation to ACh was restored by combined superoxide dismutase and catalase treatment, and also by NADPH oxidase inhibition. When dilute (10%) serum from exposed rats was perfused into the lumen of coronary arteries from unexposed, naïve rats, the O3-induced reduction in vasodilatory response to ACh was partially recapitulated. Furthermore, following O3 inhalation, serum exhibited a nitric oxide scavenging capacity, which may partially explain blunted ACh-mediated vasodilatory responses. Thus, bioactivity from inhalation exposures may be due to compositional changes of the circulation. These studies shed light on possible mechanisms of action that may explain O3-associated cardiac morbidity and mortality in humans.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ozono/toxicidad , Vasodilatación/efectos de los fármacos , Acetilcolina/administración & dosificación , Animales , Sangre , Vasos Coronarios/metabolismo , Exposición por Inhalación , Masculino , Ratones , Ozono/administración & dosificación , Ratas , Ratas Sprague-Dawley , Serotonina/administración & dosificación
9.
PLoS One ; 9(5): e97084, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24811453

RESUMEN

Exposure to chronic hypoxia (CH) induces elevated pulmonary artery pressure/resistance, leading to an eventual maladaptive right ventricular hypertrophy (RVH). Muscle RING finger-1 (MuRF1) is a muscle-specific ubiquitin ligase that mediates myocyte atrophy and has been shown to play a role in left ventricular hypertrophy and altered cardiac bioenergetics in pressure overloaded hearts. However, little is known about the contribution of MuRF1 impacting RVH in the setting of CH. Therefore, we hypothesized that MuRF1 deletion would enhance RVH compared to their wild-type littermates, while cardiac-specific overexpression would reduce hypertrophy following CH-induced pulmonary hypertension. We assessed right ventricular systolic pressure (RVSP), right ventricle to left ventricle plus septal weight ratio (RV/LV+S) and hematocrit (Hct) following a 3-wk isobaric CH exposure. Additionally, we conducted dual-isotope SPECT/CT imaging with cardiac function agent 201Tl-chloride and cell death agent 99mTc-annexin V. Predictably, CH induced pulmonary hypertension, measured by increased RVSP, RV/LV+S and Hct in WT mice compared to normoxic WT mice. Normoxic WT and MuRF1-null mice exhibited no significant differences in RVSP, RV/LV+S or Hct. CH-induced increases in RVSP were also similar between WT and MuRF1-null mice; however, RV/LV+S and Hct were significantly elevated in CH-exposed MuRF1-null mice compared to WT. In cardiac-specific MuRF1 overexpressing mice, RV/LV+S increased significantly due to CH exposure, even greater than in WT mice. This remodeling appeared eccentric, maladaptive and led to reduced systemic perfusion. In conclusion, these results are consistent with an atrophic role for MuRF1 regulating the magnitude of right ventricular hypertrophy following CH-induction of pulmonary hypertension.


Asunto(s)
Adaptación Fisiológica , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Proteínas Musculares/metabolismo , Fenotipo , Ubiquitina-Proteína Ligasas/metabolismo , Remodelación Ventricular , Animales , Hipoxia de la Célula , Regulación de la Expresión Génica , Hematócrito , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/complicaciones , Ratones , Proteínas Musculares/deficiencia , Músculo Esquelético/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Flujo Sanguíneo Regional , Factores de Tiempo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/deficiencia
10.
Toxicol Sci ; 134(2): 304-11, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23650127

RESUMEN

Inhaled pollutants induce the release of vasoactive factors into the systemic circulation, but little information is available regarding the nature of these factors or their receptors. The pattern recognition receptor CD36 interacts with many damage-related circulating molecules, leading to activation of endothelial cells and promoting vascular inflammation; therefore, we hypothesized that CD36 plays a pivotal role in mediating cross talk between inhaled ozone (O3)-induced circulating factors and systemic vascular dysfunction. O3 exposure (1 ppm × 4h) induced lung inflammation in wild-type (WT) mice, which was absent in the CD36 deficient (CD36(-/-)) mice. Acetylcholine (ACh)-evoked vasorelaxation was impaired in isolated aortas from O3-exposed WT mice but not in vessels from CD36(-/-) mice. To delineate whether vascular impairments were caused by lung inflammation or CD36-mediated generation of circulating factors, naïve aortas were treated with diluted serum from control or O3-exposed WT mice, which recapitulated the impairments of vasorelaxation observed after inhalation exposures. Aortas from CD36(-/-) mice were insensitive to the effects of O3-induced circulating factors, with robust vasorelaxation responses in the presence of serum from O3-exposed WT mice. Lung inflammation was not a requirement for production of circulating vasoactive factors, as serum from O3-exposed CD36(-/-) mice could inhibit vasorelaxation in naïve WT aortas. These results suggest that O3 inhalation induces the release of circulating bioactive factors capable of impairing vasorelaxation to ACh via a CD36-dependent signaling mechanism. Although lung inflammatory and systemic vascular effects were both dependent on CD36, the presence of circulating factors appears to be independent of CD36 and inflammatory responses.


Asunto(s)
Antígenos CD36/inmunología , Endotelio Vascular/efectos de los fármacos , Ozono/toxicidad , Animales , Líquido del Lavado Bronquioalveolar , Endotelio Vascular/inmunología , Endotelio Vascular/fisiopatología , Femenino , Exposición por Inhalación , Ratones , Ratones Endogámicos C57BL
11.
Cardiovasc Toxicol ; 13(4): 364-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23959951

RESUMEN

Usage of nicotine-only formulations, such as transdermal patches, nicotine gum, or electronic nicotine delivery systems is increasing, as they are perceived as healthier alternatives to traditional cigarettes. Unfortunately, there is little data available on the effect of isolated nicotine on myocardial and aortic remodeling, especially in the setting of cardiovascular disease risk factors, such as hypertension. We hypothesized that nicotine would exacerbate cardiovascular remodeling induced by angiotensin-II (Ang II) treatment. Subcutaneous osmotic minipumps were implanted to administer Ang II, Nic, nicotine plus Ang II or saline to C57Bl/6 mice for 4 weeks. Heart weights were increased by all treatments, with control < nicotine < Ang II < nicotine + Ang II. Activity levels of matrix metalloproteinase-2 mirrored these changes and demonstrated clear additivity between nicotine and Ang II. Histopathological analysis of aortas revealed that mice receiving combined nicotine and Ang II treatment induced significant hypertrophy compared to all other groups. This study reveals possible cardiotoxic interactions between nicotine and a common model of systemic hypertension. Safety testing of novel nicotine delivery devices should consider that hypertension is a common impetus to begin smoking cessation therapy, and potential interactions should be more thoroughly studied.


Asunto(s)
Modelos Animales de Enfermedad , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Nicotina/administración & dosificación , Remodelación Ventricular/efectos de los fármacos , Angiotensina II/administración & dosificación , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Hipertensión/fisiopatología , Bombas de Infusión , Masculino , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Remodelación Ventricular/fisiología
12.
Vascul Pharmacol ; 56(1-2): 64-73, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22146233

RESUMEN

Arterial remodeling contributes to elevated pulmonary artery (PA) pressures and right ventricular hypertrophy seen in pulmonary hypertension (PH). Resveratrol, a sirtuin-1 (SIRT1) pathway activator, can prevent the development of PH in a commonly used animal model, but it is unclear whether it can reverse established PH pathophysiology. Furthermore, atrophic ubiquitin ligases, such as atrogin-1 and MuRF-1, are known to be induced by SIRT1 activators but have not been characterized in hypertrophic vascular disease. Therefore, we hypothesized that monocrotaline (MCT)-induced PH would attenuate atrophy pathways in the PA while, conversely, SIRT1 activation (resveratrol) would reverse indices of PH and restore atrophic gene expression. Thus, we injected Sprague-Dawley rats with MCT (50 mg/kg i.p.) or saline at Day 0, and then treated with oral resveratrol or sildenafil from days 28-42 post-MCT injection. Oral resveratrol attenuated established MCT-induced PH indices, including right ventricular systolic pressure, right ventricular hypertrophy, and medial thickening of intrapulmonary arteries. Resveratrol also normalized PA atrogin-1 mRNA expression, which was significantly reduced by MCT. In cultured human PA smooth muscle cells (hPASMC), resveratrol significantly inhibited PDGF-stimulated proliferation and cellular hypertrophy, which was also associated with improvements in atrogin-1 levels. In addition, SIRT1 inhibition augmented hPASMC proliferation, as assessed by DNA mass, and suppressed atrogin mRNA expression. These findings demonstrate an inverse relationship between indices of PH and PA atrogin expression that is SIRT1 dependent and may reflect a novel role for SIRT1 in PASMCs opposing cellular hypertrophy and proliferation.


Asunto(s)
Hipertensión Pulmonar/tratamiento farmacológico , Hipertrofia Ventricular Derecha/tratamiento farmacológico , Monocrotalina/farmacología , Proteínas Musculares/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Proteínas Ligasas SKP Cullina F-box/metabolismo , Estilbenos/farmacología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Remodelación de las Vías Aéreas (Respiratorias)/genética , Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Interacciones Farmacológicas , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia/tratamiento farmacológico , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/fisiopatología , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/fisiopatología , Canal de Potasio Kv1.5/metabolismo , Masculino , Proteínas Musculares/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Resveratrol , Proteínas Ligasas SKP Cullina F-box/genética , Sirtuina 1/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética , Remodelación Ventricular/fisiología
13.
Cardiovasc Toxicol ; 12(3): 226-34, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22311109

RESUMEN

Atrophic signaling elements of the ubiquitin-proteasome system (UPS) are involved in skeletal muscle wasting as well as pressure overload models of heart failure. In our prior experiments, we demonstrated a transcriptional downregulation of atrophy-inducing vascular E3 ubiquitin ligases in a toxic model of pulmonary hypertension where pulmonary artery and right ventricle (RV) hypertrophy are evident. Given the numerous reports of glucocorticoid activation of the UPS and the negative regulator of muscle mass, myostatin, we investigated the efficacy of dexamethasone to reverse monocrotaline (MCT)-induced pulmonary hypertension and augment atrogin-1 expression in both pulmonary arteries and myocardium. Dexamethasone caused significant reductions in body weight in combination with MCT. As predicted, MCT-induced pulmonary hypertension was evident by increases in RV systolic pressure, right ventricle to left ventricle plus septal weight ratios (RV/LVS) and arterial remodeling. MCT treatment significantly reduced both RV and PA atrogin-1 expression. Dexamethasone treatment reversed the MCT-induced pathological indices and restored RV atrogin-1 expression, but did not impact atrogin-1 expression in pulmonary arteries. Myostatin was poorly expressed in pulmonary arteries compared to the RV, and dexamethasone treatment increase RV myostatin in controls but not MCT-treated rats. These findings suggest that mechanisms independent of myostatin/atrogin-1 are responsible for glucocorticoid efficacy in this model of pulmonary hypertension.


Asunto(s)
Dexametasona/farmacología , Expresión Génica , Glucocorticoides/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Monocrotalina/toxicidad , Proteínas Musculares/genética , Proteínas Ligasas SKP Cullina F-box/genética , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Antagonismo de Drogas , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/inducido químicamente , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Masculino , Proteínas Musculares/metabolismo , Miostatina/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Ligasas SKP Cullina F-box/metabolismo
14.
Toxicol Appl Pharmacol ; 230(3): 346-51, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18455212

RESUMEN

Environmental air pollution is associated with adverse cardiovascular events, including increased hospital admissions due to heart failure and myocardial infarction. The exact mechanism(s) by which air pollution affects the heart and vasculature is currently unknown. Recent studies have found that exposure to air pollution enhances arterial vasoconstriction in humans and animal models. Work in our laboratory has shown that diesel emissions (DE) enhance vasoconstriction of mouse coronary arteries. Thus, we hypothesized that DE could enhance vasoconstriction in arteries and veins through uncoupling of endothelial nitric oxide synthase (eNOS). To test this hypothesis, we first bubbled DE through a physiological saline solution and exposed isolated mesenteric veins. Second, we exposed animals, whole body, to DE at 350 microg/m(3) for 4 h, after which mesenteric arteries and veins were isolated. Results from these experiments show that saline bubbled with DE as well as inhaled DE enhances vasoconstriction in veins but not arteries. Exposure to several representative volatile organic compounds found in the DE-exposed saline did not enhance arterial constriction. L-nitro-arginine-methyl-ester (L-NAME), an eNOS inhibitor, normalized the control vessels to the DE-exposed vessels implicating an uncoupling of eNOS as a mechanism for enhanced vasoconstriction. The principal conclusions of this research are 1) veins exhibit endothelial dysfunction following in vivo and ex vivo exposures to DE, 2) veins appear to be more sensitive to DE effects than arteries, and 3) DE components most likely induce endothelial dysfunction through the uncoupling of eNOS.


Asunto(s)
Venas Mesentéricas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/fisiología , Vasoconstricción/efectos de los fármacos , Emisiones de Vehículos/toxicidad , Animales , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Venas Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA