Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
J Toxicol Environ Health A ; 84(12): 503-517, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33682625

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Dust/analysis , Encephalitis/physiopathology , Learning/drug effects , Memory/drug effects , Particulate Matter/toxicity , Pneumonia/physiopathology , Animals , Arizona , Autoimmune Diseases/etiology , Biomarkers/metabolism , Disease Models, Animal , Dust/immunology , Encephalitis/chemically induced , Female , Inhalation Exposure/adverse effects , Mice , Mining , New Mexico , Particle Size , Pneumonia/chemically induced , Random Allocation
2.
J Toxicol Environ Health A ; 84(1): 31-48, 2021 01 02.
Article in English | MEDLINE | ID: mdl-33050837

ABSTRACT

Thousands of abandoned uranium mines (AUMs) exist in the western United States. Due to improper remediation, windblown dusts generated from AUMs are of significant community concern. A mobile inhalation lab was sited near an AUM of high community concern ("Claim 28") with three primary objectives: to (1) determine the composition of the regional ambient particulate matter (PM), (2) assess meteorological characteristics (wind speed and direction), and (3) assess immunological and physiological responses of mice after exposures to concentrated ambient PM (or CAPs). C57BL/6 and apolipoprotein E-null (ApoE-/-) mice were exposed to CAPs in AirCARE1 located approximately 1 km to the SW of Claim 28, for 1 or 28 days for 4 hr/day at approximately 80 µg/m3 CAPs. Bronchoalveolar lavage fluid (BALF) analysis revealed a significant influx of neutrophils after a single-day exposure in C57BL/6 mice (average PM2.5 concentration = 68 µg/m3). Lungs from mice exposed for 1 day exhibited modest increases in Tnfa and Tgfb mRNA levels in the CAPs exposure group compared to filtered air (FA). Lungs from mice exposed for 28 days exhibited reduced Tgfb (C57BL/6) and Tnfa (ApoE-/-) mRNA levels. Wind direction was typically moving from SW to NE (away from the community) and, while detectable in all samples, uranium concentrations in the PM2.5 fraction were not markedly different from published-reported values. Overall, exposure to CAPs in the region of the Blue GAP Tachee's Claim-28 uranium mine demonstrated little evidence of overt pulmonary injury or inflammation or ambient air contamination attributed to uranium or vanadium.


Subject(s)
Air Pollutants/toxicity , Inhalation Exposure/adverse effects , Mining , Particulate Matter/toxicity , Uranium , Animals , Female , Male , Mice , Mice, Inbred C57BL , Toxicity Tests, Acute , Toxicity Tests, Subchronic
3.
Part Fibre Toxicol ; 17(1): 29, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32611356

ABSTRACT

BACKGROUND: Commercial uranium mining on the Navajo Nation has subjected communities on tribal lands in the Southwestern United States to exposures from residual environmental contamination. Vascular health effects from these ongoing exposures are an active area of study. There is an association between residential mine-site proximity and circulating biomarkers in residents, however, the contribution of mine-site derived wind-blown dusts on vascular and other health outcomes is unknown. To assess neurovascular effects of mine-site derived dusts, we exposed mice using a novel exposure paradigm, the AirCARE1 mobile inhalation laboratory, located 2 km from an abandoned uranium mine, Claim 28 in Blue Gap Tachee, AZ. Mice were exposed to filtered air (FA) (n = 6) or concentrated ambient particulate matter (CAPs) (n = 5) for 2 wks for 4 h per day. RESULTS: To assess miRNA differential expression in cultured mouse cerebrovascular cells following particulate matter (PM) exposure (average: 96.6 ± 60.4 µg/m3 for all 4 h exposures), the serum cumulative inflammatory potential (SCIP) assay was employed. MiRNA sequencing was then performed in cultured mouse cerebrovascular endothelial cells (mCECs) to evaluate transcriptional changes. Results indicated 27 highly differentially expressed (p < 0.01) murine miRNAs, as measured in the SCIP assay. Gene ontology (GO) pathway analysis revealed notable alterations in GO enrichment related to the cytoplasm, protein binding and the cytosol, while significant KEGG pathways involved pathways in cancer, axon guidance and Wnt signaling. Expression of these 27 identified, differentially expressed murine miRNAs were then evaluated in the serum. Nine of these miRNAs (~ 30%) were significantly altered in the serum and 8 of those miRNAs demonstrated the same directional change (either upregulation or downregulation) as cellular miRNAs, as measured in the SCIP assay. Significantly upregulated miRNAs in the CAPs exposure group included miRNAs in the let-7a family. Overexpression of mmu-let-7a via transfection experiments, suggested that this miRNA may mediate mCEC barrier integrity following dust exposure. CONCLUSIONS: Our data suggest that mCEC miRNAs as measured in the SCIP assay show similarity to serum-borne miRNAs, as approximately 30% of highly differentially expressed cellular miRNAs in the SCIP assay were also found in the serum. While translocation of miRNAs via exosomes or an alternative mechanism is certainly possible, other yet-to-be-identified factors in the serum may be responsible for significant miRNA differential expression in endothelium following inhaled exposures. Additionally, the most highly upregulated murine miRNAs in the CAPs exposure group were in the let-7a family. These miRNAs play a prominent role in cell growth and differentiation and based on our transfection experiments, mmu-let-7a may contribute to cerebrovascular mCEC alterations following inhaled dust exposure.


Subject(s)
Air Pollutants/toxicity , Particulate Matter/toxicity , Animals , Biomarkers/blood , Cell Differentiation , Cell Proliferation , Endothelium , Inhalation Exposure , Mice , MicroRNAs , Southwestern United States , Uranium
4.
Cardiovasc Toxicol ; 20(3): 211-221, 2020 06.
Article in English | MEDLINE | ID: mdl-31410643

ABSTRACT

Roadside proximity and exposure to mixed vehicular emissions (MVE) have been linked to adverse pulmonary and vascular outcomes. However, because of the complex nature of the contribution of particulate matter (PM) versus gases, it is difficult to decipher the precise causative factors regarding PM and the copollutant gaseous fraction. To this end, C57BL/6 and apolipoprotein E knockout mice (ApoE-/-) were exposed to either filtered air (FA), fine particulate (FP), FP+gases (FP+G), ultrafine particulate (UFP), or UFP+gases (UFP+G). Two different timeframes were employed: 1-day (acute) or 30-day (subchronic) exposures. Examined biological endpoints included aortic vasoreactivity, aortic lesion quantification, and aortic mRNA expression. Impairments in vasorelaxation were observed following acute exposure to FP+G in C57BL/6 animals and FP, UFP, and UFP+G in ApoE-/- animals. These effects were completely abrogated or markedly reduced following subchronic exposure. Aortic lesion quantification in ApoE-/- animals indicated a significant increase in atheroma size in the UFP-, FP-, and FP+G-exposed groups. Additionally, ApoE-/- mice demonstrated a significant fold increase in TNFα expression following FP+G exposure and ET-1 following UFP exposure. Interestingly, C57BL/6 aortic gene expression varied widely across exposure groups. TNFα decreased significantly following FP exposure and CCL-5 decreased in the UFP-, FP-, and FP+G-exposed groups. Conversely, ET-1, CCL-2, and CXCL-1 were all significantly upregulated in the FP+G group. These findings suggest that gas-particle interactions may play a role in vascular toxicity, but the contribution of surface area is not clear.


Subject(s)
Aorta/drug effects , Aortic Diseases/chemically induced , Atherosclerosis/chemically induced , Inhalation Exposure/adverse effects , Particulate Matter/toxicity , Vehicle Emissions/toxicity , Animals , Aorta/metabolism , Aorta/pathology , Aorta/physiopathology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Aortic Diseases/physiopathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/physiopathology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Disease Models, Animal , Endothelin-1/genetics , Endothelin-1/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Risk Assessment , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects
5.
Cardiovasc Toxicol ; 19(5): 401-411, 2019 10.
Article in English | MEDLINE | ID: mdl-30963444

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Aorta, Thoracic/drug effects , Cadmium/toxicity , Cerebral Cortex/drug effects , Dust , Lead/toxicity , Mining , Particulate Matter/toxicity , Pneumonia/chemically induced , Animals , Aorta, Thoracic/physiopathology , Cerebral Cortex/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Inhalation Exposure , Male , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Particle Size , Pneumonia/genetics , Pneumonia/metabolism , Risk Assessment , Siberia , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vasodilation/drug effects
6.
Toxicol Sci ; 164(1): 101-114, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29660078

ABSTRACT

Exposure to windblown particulate matter (PM) arising from legacy uranium (U) mine sites in the Navajo Nation may pose a human health hazard due to their potentially high metal content, including U and vanadium (V). To assess the toxic impact of PM derived from Claim 28 (a priority U mine) compared with background PM, and consider the putative role of metal species U and V. Two representative sediment samples from Navajo Nation sites (Background PM and Claim 28 PM) were obtained, characterized in terms of chemistry and morphology, and fractioned to the respirable (≤ 10 µm) fraction. Mice were dosed with either PM sample, uranyl acetate, or vanadyl sulfate via aspiration (100 µg), with assessments of pulmonary and vascular toxicity 24 h later. Particulate matter samples were also examined for in vitro effects on cytotoxicity, oxidative stress, phagocytosis, and inflammasome induction. Claim 28 PM10 was highly enriched with U and V and exhibited a unique nanoparticle ultrastructure compared with background PM10. Claim 28 PM10 exhibited enhanced pulmonary and vascular toxicity relative to background PM10. Both U and V exhibited complementary pulmonary inflammatory potential, with U driving a classical inflammatory cytokine profile (elevated interleukin [IL]-1ß, tumor necrosis factor-α, and keratinocyte chemoattractant/human growth-regulated oncogene) while V preferentially induced a different cytokine pattern (elevated IL-5, IL-6, and IL-10). Claim 28 PM10 was more potent than background PM10 in terms of in vitro cytotoxicity, impairment of phagocytosis, and oxidative stress responses. Resuspended PM10 derived from U mine waste exhibit greater cardiopulmonary toxicity than background dusts. Rigorous exposure assessment is needed to gauge the regional health risks imparted by these unremediated sites.


Subject(s)
Heart/drug effects , Inhalation Exposure/adverse effects , Lung/drug effects , Nanoparticles/toxicity , Particulate Matter/toxicity , Uranium/toxicity , Vanadium Compounds/toxicity , Animals , Bronchoalveolar Lavage Fluid/immunology , Cell Survival/drug effects , Cytokines/analysis , Geologic Sediments/chemistry , Humans , Lung/immunology , Male , Mice, Inbred C57BL , Mining , Nanoparticles/analysis , Oxidative Stress/drug effects , Particle Size , Particulate Matter/analysis , THP-1 Cells , Uranium/analysis , Vanadium Compounds/analysis , Vasodilation/drug effects
7.
Toxicol Sci ; 163(1): 123-139, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29385576

ABSTRACT

The role of environmental stressors, particularly exposure to air pollution, in the development of neurodegenerative disease remains underappreciated. We examined the neurological effects of acute ozone (O3) exposure in aged mice, where increased blood-brain barrier (BBB) permeability may confer vulnerability to neuroinflammatory outcomes. C57BL/6 male mice, aged 8-10 weeks or 12-18 months were exposed to either filtered air or 1.0 ppm O3 for 4 h; animals received a single IP injection of sodium fluorescein (FSCN) 20 h postexposure. One-hour post-FSCN injection, animals were transcardially perfused for immunohistochemical analysis of BBB permeability. ß-amyloid protein expression was assessed via ELISA. Flow cytometric characterization of infiltrating immune cells, including neutrophils, macrophages, and microglia populations was performed 20 h post-O3 exposure. Flow cytometry analysis of brains revealed increased microglia "activation" and presentation of CD11b, F4/80, and MHCII in aged animals relative to younger ones; these age-induced differences were potentiated by acute O3 exposure. Cortical and limbic regions in aged brains had increased reactive microgliosis and ß-amyloid protein expression after O3 insult. The aged cerebellum was particularly vulnerable to acute O3 exposure with increased populations of infiltrating neutrophils, peripheral macrophages/monocytes, and Ly6C+ inflammatory monocytes after insult, which were not significantly increased in the young cerebellum. O3 exposure increased the penetration of FSCN beyond the BBB, the infiltration of peripheral immune cells, and reactive gliosis of microglia. Thus, the aged BBB is vulnerable to insult and becomes highly penetrable in response to O3 exposure, leading to greater neuroinflammatory outcomes.


Subject(s)
Aging/drug effects , Air Pollutants/toxicity , Blood-Brain Barrier/drug effects , Neurogenic Inflammation/chemically induced , Ozone/toxicity , Aging/immunology , Air Pollutants/pharmacokinetics , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/immunology , Blood-Brain Barrier/metabolism , Capillary Permeability , Cerebellum/drug effects , Cerebellum/immunology , Cerebellum/metabolism , Male , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , Neurogenic Inflammation/immunology , Neurogenic Inflammation/metabolism , Neutrophil Infiltration/drug effects , Ozone/pharmacokinetics
8.
Proc Natl Acad Sci U S A ; 114(10): E1968-E1976, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223486

ABSTRACT

Pulmonary exposure to multiwalled carbon nanotubes (MWCNTs) causes indirect systemic inflammation through unknown pathways. MWCNTs translocate only minimally from the lungs into the systemic circulation, suggesting that extrapulmonary toxicity may be caused indirectly by lung-derived factors entering the circulation. To assess a role for MWCNT-induced circulating factors in driving neuroinflammatory outcomes, mice were acutely exposed to MWCNTs (10 or 40 µg/mouse) via oropharyngeal aspiration. At 4 h after MWCNT exposure, broad disruption of the blood-brain barrier (BBB) was observed across the capillary bed with the small molecule fluorescein, concomitant with reactive astrocytosis. However, pronounced BBB permeation was noted, with frank albumin leakage around larger vessels (>10 µm), overlain by a dose-dependent astroglial scar-like formation and recruitment of phagocytic microglia. As affirmed by elevated inflammatory marker transcription, MWCNT-induced BBB disruption and neuroinflammation were abrogated by pretreatment with the rho kinase inhibitor fasudil. Serum from MWCNT-exposed mice induced expression of adhesion molecules in primary murine cerebrovascular endothelial cells and, in a wound-healing in vitro assay, impaired cell motility and cytokinesis. Serum thrombospondin-1 level was significantly increased after MWCNT exposure, and mice lacking the endogenous receptor CD36 were protected from the neuroinflammatory and BBB permeability effects of MWCNTs. In conclusion, acute pulmonary exposure to MWCNTs causes neuroinflammatory responses that are dependent on the disruption of BBB integrity.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Blood-Brain Barrier/drug effects , Drug Carriers/adverse effects , Encephalitis/prevention & control , Nanotubes, Carbon/adverse effects , Protein Kinase Inhibitors/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Administration, Inhalation , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , CD36 Antigens/deficiency , CD36 Antigens/genetics , Cell Movement/drug effects , Encephalitis/chemically induced , Encephalitis/genetics , Encephalitis/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fluorescein/pharmacokinetics , Fluorescent Dyes/pharmacokinetics , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , rho-Associated Kinases/genetics
9.
Part Fibre Toxicol ; 13(1): 64, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27906023

ABSTRACT

BACKGROUND: Deleterious consequences of exposure to traffic emissions may derive from interactions between carbonaceous particulate matter (PM) and gaseous components in a manner that is dependent on the surface area or complexity of the particles. To determine the validity of this hypothesis, we examined pulmonary and neurological inflammatory outcomes in C57BL/6 and apolipoprotein E knockout (ApoE-/-) male mice after acute and chronic exposure to vehicle engine-derived particulate matter, generated as ultrafine (UFP) and fine (FP) sizes, with additional exposures using UFP or FP combined with gaseous copollutants derived from fresh gasoline and diesel emissions, labeled as UFP + G and FP + G. RESULTS: The UFP and UFP + G exposure groups resulted in the most profound pulmonary and neuroinflammatory effects. Phagocytosis of UFP + G particles via resident alveolar macrophages was substantial in both mouse strains, particularly after chronic exposure, with concurrent increased proinflammatory cytokine expression of CXCL1 and TNFα in the bronchial lavage fluid. In the acute exposure paradigm, only UFP and UFP + G induced significant changes in pulmonary inflammation and only in the ApoE-/- animals. Similarly, acute exposure to UFP and UFP + G increased the expression of several cytokines in the hippocampus of ApoE-/- mice including Il-1ß, IL-6, Tgf-ß and Tnf-α and in the hippocampus of C57BL/6 mice including Ccl5, Cxcl1, Il-1ß, and Tnf-α. Interestingly, Il-6 and Tgf-ß expression were decreased in the C57BL/6 hippocampus after acute exposure. Chronic exposure to UFP + G increased expression of Ccl5, Cxcl1, Il-6, and Tgf-ß in the ApoE-/- hippocampus, but this effect was minimal in the C57BL/6 mice, suggesting compensatory mechanisms to manage neuroinflammation in this strain. CONCLUSIONS: Inflammatory responses the lung and brain were most substantial in ApoE-/- animals exposed to UFP + G, suggesting that the surface area-dependent interaction of gases and particles is an important determinant of toxic responses. As such, freshly generated UFP, in the presence of combustion-derived gas phase pollutants, may be a greater health hazard than would be predicted from PM concentration, alone, lending support for epidemiological findings of adverse neurological outcomes associated with roadway proximity.


Subject(s)
Inflammation/chemically induced , Lung/drug effects , Vehicle Emissions/toxicity , Animals , Apolipoproteins E/genetics , Body Weight , Bronchoalveolar Lavage Fluid , Cytokines/biosynthesis , Inhalation Exposure , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Surface Properties
10.
Atherosclerosis ; 254: 59-66, 2016 11.
Article in English | MEDLINE | ID: mdl-27693879

ABSTRACT

BACKGROUND AND AIMS: Obstructive sleep apnea (OSA) is characterized by intermittent airway obstruction and systemic hypoxia during sleep, which can contribute to an increase in reactive oxygen species, vascular remodeling, vasoconstriction and ultimately cardiovascular disease. Continuous positive airway pressure (CPAP) is a clinical therapy that maintains airway patency and mitigates several symptoms of OSA. However, it is currently unknown whether CPAP therapy also reduces the overall inflammatory potential in the circulation; to address this in an unbiased manner, we applied a novel endothelial biosensor approach, the serum cumulative inflammatory potential (SCIP) assay. METHODS: We studied healthy controls (n = 7), OSA subjects receiving no treatment, (OSA controls) (n = 7) and OSA subjects receiving CPAP for 3 months (n = 8). Serum was obtained from OSA subjects before and after CPAP or no treatment. A battery of quantitative and functional assays was performed to assess the serum inflammatory potential, in terms of endothelial responses. For the SCIP assay, human coronary artery endothelial cells (hCAECs) were incubated with 5% serum in media from individual subjects for 4 h. qPCR was performed to assess endothelial inflammatory transcript (ICAM-1, VCAM-1, IL-8, P-selectin, CCL5, and CXCL12) responses to serum. Additionally, transendothelial resistance was measured in serum-incubated hCAECs following leukocyte challenge. RESULTS: hCAECs exhibited significant increases in VCAM-1, ICAM-1, IL-8 and P-selectin mRNA when incubated with serum from OSA patients compared to serum from healthy control subjects. Furthermore, compared to no treatment, serum from CPAP-treated individuals was less potent at inducing inflammatory gene expression in the SCIP assay. Similarly, in a leukocyte adhesion assay, naïve cells treated with serum from patients who received CPAP exhibited improved endothelial barrier function than cells treated with OSA control serum. CONCLUSIONS: OSA results in greater serum inflammatory potential, thereby driving endothelial activation and dysfunction.


Subject(s)
Continuous Positive Airway Pressure/methods , Coronary Vessels/pathology , Endothelial Cells/cytology , Inflammation/blood , Sleep Apnea, Obstructive/blood , Adult , Biosensing Techniques , Case-Control Studies , Cell Adhesion , Cohort Studies , Humans , Hypoxia/blood , Hypoxia/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-8/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Male , Middle Aged , P-Selectin/metabolism , Risk Factors , Vascular Cell Adhesion Molecule-1/metabolism
11.
Toxicol Appl Pharmacol ; 305: 40-45, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27286659

ABSTRACT

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.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Air Pollutants/toxicity , Lung Injury/drug therapy , Ozone/toxicity , Protein Kinase Inhibitors/therapeutic use , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use , Animals , Bronchial Hyperreactivity/chemically induced , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Cell Count , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/chemically induced , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/complications , Hypoxia/drug therapy , Hypoxia/pathology , Hypoxia/physiopathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Lung Injury/chemically induced , Lung Injury/pathology , Lung Injury/physiopathology , Male , Mice, Inbred C57BL , Organ Size/drug effects , Protein Kinase Inhibitors/pharmacology , Ventricular Function, Right/drug effects , Ventricular Pressure/drug effects , rho-Associated Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL