Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Ecotoxicol Environ Saf ; 261: 115094, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37285676

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is currently plaguing the population at pandemic proportions and is expected to become more prevalent over the next decade. Recent epidemiological studies have demonstrated a correlation between the manifestation of NAFLD and ambient air pollution levels, which is exacerbated by other risk factors, such as diabetes, dyslipidemia, obesity, and hypertension. Exposure to airborne particulate matter has also been associated with inflammation, hepatic lipid accumulation, oxidative stress, fibrosis, and hepatocyte injury. While prolonged consumption of a high-fat (HF) diet is associated with NAFLD, little is known regarding the effects of inhaled traffic-generated air pollution, a ubiquitous environmental pollutant, on the pathogenesis of NAFLD. Therefore, we investigated the hypothesis that exposure to a mixture of gasoline and diesel engine emissions (MVE), coupled with the concurrent consumption of a HF diet, promotes the development of a NAFLD phenotype within the liver. Three-month-old male C57Bl/6 mice were placed on either a low-fat or HF diet and exposed via whole-body inhalation to either filtered (FA) air or MVE (30 µg PM/m3 gasoline engine emissions + 70 µg PM/m3 diesel engine emissions) 6 hr/day for 30 days. Histology revealed mild microvesicular steatosis and hepatocyte hypertrophy in response to MVE exposure alone, compared to FA controls, yielding a classification of "borderline NASH" under the criteria of the modified NAFLD active score (NAS) system. As anticipated, animals on a HF diet exhibited moderate steatosis; however, we also observed inflammatory infiltrates, hepatocyte hypertrophy, and increased lipid accumulation, with the combined effect of HF diet and MVE exposure. Our results indicate that inhalation exposure to traffic-generated air pollution initiates hepatocyte injury and further exacerbates lipid accumulation and hepatocyte injury induced by the consumption of a HF diet, thereby contributing to the progression of NAFLD-related pathologies.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Mice , Male , Non-alcoholic Fatty Liver Disease/etiology , Vehicle Emissions/toxicity , Gasoline , Diet, High-Fat/adverse effects , Liver , Lipids , Mice, Inbred C57BL
3.
Environ Sci Pollut Res Int ; 30(8): 21990-21999, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36280635

ABSTRACT

The pronephros (early-stage kidney) is an important osmoregulatory organ, and the onset of its function occurs relatively early in some teleost fishes. As such, any defects in kidney development and function are likely associated with a decreased ability to osmoregulate. Previous work has shown that early-life stage (ELS) zebrafish (Danio rerio) acutely exposed to Deepwater Horizon (DWH) crude oil exhibit transcriptional changes in key genes involved in pronephros development and function, as well as pronephric morphological defects and whole-animal osmoregulatory impairment. The objective of this study was to examine the acute effects of crude oil exposure during zebrafish ELS on pronephros function by assessing its fluid clearance capacity and glomerular filtration integrity. Following a 72-h exposure to control conditions, 20% or 40% dilutions of high-energy water-accommodated fractions (HEWAF) of DWH crude oil, zebrafish were injected into the common cardinal vein either with fluorescein-labeled (FITC) 70-kDa dextran to assess glomerular filtration integrity or with FITC-inulin to assess pronephric clearance capacity. Fluorescence was quantified after the injections at predetermined time intervals by fluorescence microscopy. The results demonstrated a diminished pronephric fluid clearance capacity and failed glomerular perfusion when larvae were exposed to 40% HEWAF dilutions, whereas only a reduced glomerular filtration selectivity was observed in zebrafish previously exposed to the 20% HEWAF dilution.


Subject(s)
Petroleum Pollution , Petroleum , Water Pollutants, Chemical , Animals , Zebrafish/genetics , Petroleum/toxicity , Kidney/chemistry , Larva , Water Pollutants, Chemical/analysis
4.
Cells ; 11(9)2022 04 25.
Article in English | MEDLINE | ID: mdl-35563751

ABSTRACT

Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.


Subject(s)
Probiotics , Vehicle Emissions , Adjuvants, Immunologic , Animals , Claudin-3 , Diet, High-Fat/adverse effects , Immunologic Factors , Inflammation , Intestines , Male , Mice , Mice, Inbred C57BL , Occludin , Probiotics/pharmacology , Tumor Necrosis Factor-alpha
5.
Mar Pollut Bull ; 179: 113684, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35489094

ABSTRACT

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.


Subject(s)
Perciformes , Petroleum Pollution , Petroleum , Polycyclic Aromatic Hydrocarbons , Water Pollutants, Chemical , Animals , Apoptosis , Fishes , Larva , Perciformes/physiology , Petroleum/analysis , Petroleum/toxicity , Petroleum Pollution/analysis , Polycyclic Aromatic Hydrocarbons/analysis , Transcriptome , Water Pollutants, Chemical/analysis
6.
Part Fibre Toxicol ; 19(1): 10, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35135577

ABSTRACT

BACKGROUND: The gut microbiota plays a vital role in host homeostasis and is associated with inflammation and cardiovascular disease (CVD) risk. Exposure to particulate matter (PM) is a known mediator of inflammation and CVD and is reported to promote dysbiosis and decreased intestinal integrity. However, the role of inhaled traffic-generated PM on the gut microbiome and its corresponding systemic effects are not well-characterized. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) alters the gut microbiome and promotes microbial-related inflammation and CVD biomarkers. 4-6-week-old male C57Bl/6 mice on either a low-fat (LF, 10% fat) or high-fat (HF, 45% fat) diet were exposed via oropharyngeal aspiration to 35 µg DEP suspended in 35 µl saline or saline only (CON) 2x/week for 30 days. To determine whether probiotics could prevent diet or DEP exposure mediated alterations in the gut microbiome or systemic outcomes, a subset of animals on the HF diet were treated orally with 0.3 g/day (~ 7.5 × 108 CFU/day) of Winclove Ecologic® Barrier probiotics throughout the study. RESULTS: Our results show that inhaled DEP exposure alters gut microbial profiles, including reducing Actinobacteria and expanding Verrucomicrobia and Proteobacteria. We observed increased circulating LPS, altered circulating cytokines (IL-1α, IL-3, IL-13, IL-15, G-CSF, LIF, MIP-2, and TNF-α), and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in DEP-exposed and/or HF diet mice. Furthermore, probiotics attenuated the observed reduction of Actinobacteria and expansion of Proteobacteria in DEP-exposed and HF-diet mice. Probiotics mitigated circulating cytokines (IL-3, IL-13, G-CSF, RANTES, and TNF- α) and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in respect to DEP-exposure and/or HF diet. CONCLUSION: Key findings of this study are that inhaled DEP exposure alters small intestinal microbial profiles that play a role in systemic inflammation and early CVD biomarkers. Probiotic treatment in this study was fundamental in understanding the role of inhaled DEP on the microbiome and related systemic inflammatory and CVD biomarkers.


Subject(s)
Cardiovascular Diseases , Microbiota , Animals , Biomarkers , Cardiovascular Diseases/chemically induced , Cytokines , Granulocyte Colony-Stimulating Factor , Inflammation/chemically induced , Interleukin-13 , Interleukin-3 , Male , Mice , Mice, Inbred C57BL , Particulate Matter , Plasminogen Activator Inhibitor 1 , Thrombomodulin , Vehicle Emissions/toxicity
7.
Neurotoxicol Teratol ; 90: 107071, 2022.
Article in English | MEDLINE | ID: mdl-35016995

ABSTRACT

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.


Subject(s)
Air Pollution , Demyelinating Diseases , Air Pollution/adverse effects , Animals , Apolipoproteins E/genetics , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Disease Models, Animal , Estrogen Receptor alpha/genetics , Estrogen Receptor beta , Female , Mice , Reactive Oxygen Species , Vehicle Emissions/toxicity
8.
J Neurosci Methods ; 365: 109387, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34662591

ABSTRACT

BACKGROUND: Conventional methods for individually housing, training, and testing rodents in behavioral assays can impose constraints that may limit some kinds of experimental external validity, preempt environmental enrichment, impose heavy experimenter time burdens that limit high-throughput data collection, and negatively impact animal welfare. NEW METHOD: To address these issues, we created a simple apparatus for automatically collecting individually identified data with rodents in social and/or enriched housing. RESULTS: We validated this "One Rat Turnstyle" (ORT) apparatus by utilizing it to automatically teach socially housed rats to individually press a lever without experimenter intervention or shaping. Results confirmed the feasibility and reliability of the apparatus, with almost all rats learning to move through the ORT and press a lever for sugar water by the end of the experiment. Rats had lower fecal cortisol when engaging with the ORT than with experimenter conducted daily behavioral training sessions. COMPARISON WITH EXISTING METHOD(S): The ORT is less electronically complex and more scalable compared to previous similar ideas. It requires only a 3d printer and the purchase of few parts. It is also designed to allow animals to quickly learn how to utilize it by minimizing passthrough time. CONCLUSIONS: Rats passed through the ORT both quickly and efficiently, self-administering reasonably timed behavioral sessions throughout the day. This success demonstrates that the ORT can enable the collection of both traditional and innovative, self-paced data in the context of socially housed animals, and may contribute to expanded, ecologically valid modelling.


Subject(s)
Housing, Animal , Rodentia , Animal Welfare , Animals , Behavior, Animal , Rats , Reproducibility of Results
9.
Environ Toxicol ; 37(3): 457-467, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34792841

ABSTRACT

Silver nanoparticles (AgNPs) have become increasingly popular in the biomedical field over the last few decades due to its proven antibacterial property. Previous scientific studies have reported that one of the major organs responsible for detoxification of AgNPs is the liver. The liver is also the primary organ responsible for secretion of angiotensinogen (AGT), a key signaling molecule involved in the renin-angiotensin system (RAS), which plays an important role in maintaining cardiac output and vascular pressure. The aim of this study was to assess any potential changes in the RAS-associated gene signaling, inflammatory response, and hepatocellular toxicity resulting from AgNP exposure. To do this, 6-week-old, male Wistar rats were exposed to a subacute inhalation exposure of AgNP (200 ppb/days over 4 h/days exposure, for 5 d) and their livers were analyzed for alterations in RAS components, inflammation, and oxidative stress. Real time qPCR analysis showed that AgNP-exposure resulted in a significant increase in hepatic AGT, angiotensin converting enzyme (ACE)-1, and ACE-2 mRNA expression. Expression of inflammatory markers interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α were also upregulated with AgNP-exposure, compared to controls. Furthermore AgNP-exposure mediated a significant increase in hepatic expression of catalase, and superoxide dismutase, and oxidative stress, as assessed via 8-Oxo-2'-deoxyguanosine staining. Increased oxidative stress was associated with increased monocyte/macrophage-2 staining in the liver of AgNP-exposed rats. Such findings indicate that subacute inhalation exposure to AgNPs mediate increased hepatic RAS signaling, associated with inflammation, macrophage infiltration, and oxidative stress.


Subject(s)
Metal Nanoparticles , Silver , Animals , Inflammation/chemically induced , Inhalation Exposure/adverse effects , Male , Metal Nanoparticles/toxicity , Oxidative Stress , Rats , Rats, Wistar , Renin-Angiotensin System
10.
Toxicol Rep ; 8: 846-862, 2021.
Article in English | MEDLINE | ID: mdl-33948438

ABSTRACT

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.

11.
Ecotoxicol Environ Saf ; 213: 112035, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33581487

ABSTRACT

Air pollution has been documented to contribute to severe respiratory diseases like asthma and chronic obstructive pulmonary disorder (COPD). Although these diseases demonstrate a shift in the lung microbiota towards Proteobacteria, the effects of traffic generated emissions on lung microbiota profiles have not been well-characterized. Thus, we investigated the hypothesis that exposure to traffic-generated emissions can alter lung microbiota and immune defenses. Since a large population of the Western world consumes a diet rich in fats, we sought to investigate the synergistic effects of mixed vehicle emissions and high-fat diet consumption. We exposed 3-month-old male C57Bl/6 mice placed either on regular chow (LF) or a high-fat (HF: 45% kcal fat) diet to mixed emissions (ME: 30 µg PM/m3 gasoline engine emissions+70 µg PM/m3 diesel engine emissions) or filtered air (FA) for 6 h/d, 7 d/wk for 30 days. Levels of pulmonary immunoglobulins IgA, IgG, and IgM were analyzed by ELISA, and lung microbial profiling was done using qPCR and Illumina 16 S sequencing. We observed a significant decrease in lung IgA in the ME-exposed animals, compared to the FA-exposed animals, both fed a HF diet. Our results also revealed a significant decrease in lung IgG in the ME-exposed animals both on the LF diet and HF diet, in comparison to the FA-exposed animals. We also observed an expansion of Enterobacteriaceae belonging to the Proteobacteria phylum in the ME-exposed groups on the HF diet. Collectively, we show that the combined effects of ME and HF diet result in decreased immune surveillance and lung bacterial dysbiosis, which is of significance in lung diseases.


Subject(s)
Lung/microbiology , Proteobacteria , Vehicle Emissions/toxicity , Air Pollution , Animals , Diet, High-Fat , Dysbiosis , Male , Mice , Mice, Inbred C57BL , Microbiota
12.
Part Fibre Toxicol ; 18(1): 3, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33419468

ABSTRACT

BACKGROUND: Exposure to traffic-generated emissions is associated with the development and exacerbation of inflammatory lung disorders such as chronic obstructive pulmonary disorder (COPD) and idiopathic pulmonary fibrosis (IPF). Although many lung diseases show an expansion of Proteobacteria, the role of traffic-generated particulate matter pollutants on the lung microbiota has not been well-characterized. Thus, we investigated the hypothesis that exposure to diesel exhaust particles (DEP) can alter commensal lung microbiota, thereby promoting alterations in the lung's immune and inflammatory responses. We aimed to understand whether diet might also contribute to the alteration of the commensal lung microbiome, either alone or related to exposure. To do this, we used male C57Bl/6 mice (4-6-week-old) on either regular chow (LF) or high-fat (HF) diet (45% kcal fat), randomly assigned to be exposed via oropharyngeal aspiration to 35 µg DEP, suspended in 35 µl 0.9% sterile saline or sterile saline only (control) twice a week for 30 days. A separate group of study animals on the HF diet was concurrently treated with 0.3 g/day of Winclove Ecologic® Barrier probiotics in their drinking water throughout the study. RESULTS: Our results show that DEP-exposure increases lung tumor necrosis factor (TNF)-α, interleukin (IL)-10, Toll-like receptor (TLR)-2, TLR-4, and the nuclear factor kappa B (NF-κB) histologically and by RT-qPCR, as well as Immunoglobulin A (IgA) and Immunoglobulin G (IgG) in the bronchoalveolar lavage fluid (BALF), as quantified by ELISA. We also observed an increase in macrophage infiltration and peroxynitrite, a marker of reactive oxygen species (ROS) + reactive nitrogen species (RNS), immunofluorescence staining in the lungs of DEP-exposed and HF-diet animals, which was further exacerbated by concurrent DEP-exposure and HF-diet consumption. Histological examinations revealed enhanced inflammation and collagen deposition in the lungs DEP-exposed mice, regardless of diet. We observed an expansion of Proteobacteria, by qPCR of bacterial 16S rRNA, in the BALF of DEP-exposed mice on the HF diet, which was diminished with probiotic-treatment. CONCLUSIONS: Our findings suggest that exposure to DEP causes persistent and sustained inflammation and bacterial alterations in a ROS-RNS mediated fashion, which is exacerbated by concurrent consumption of an HF diet.


Subject(s)
Diet, High-Fat , Vehicle Emissions , Animals , Bronchoalveolar Lavage Fluid , Inflammation , Lung , Male , Mice , Mice, Inbred C57BL , Nitrogen , Particulate Matter/toxicity , RNA, Ribosomal, 16S , Reactive Nitrogen Species , Reactive Oxygen Species , Vehicle Emissions/toxicity
13.
Toxicol Lett ; 339: 39-50, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33373663

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Brain/drug effects , Central Nervous System/drug effects , Inflammation/chemically induced , Mice/genetics , Microvessels/drug effects , Vehicle Emissions/toxicity , Animals , Apolipoproteins E/drug effects , Female , Humans , Models, Animal , Peptide Fragments/drug effects
14.
J Appl Toxicol ; 41(6): 972-986, 2021 06.
Article in English | MEDLINE | ID: mdl-33029829

ABSTRACT

Silver nanoparticles (AgNPs) have become crucial players in the field of medicine and various other industries. AgNPs have a wide array of applications, which includes production of electronic goods, cosmetics, synthesis of dyes, and printing inks, as well as targeted delivery of drugs to specialized cells inside the body. Even though humans readily come in contact with these particles, the organ-specific accumulation and resulting mechanisms of toxicity induced by inhaled AgNPs are still under investigation. The goal of this study was to determine the organ distribution of inhaled AgNPs and investigate the resulting systemic toxicity. To do this, male Wistar rats were exposed by inhalation to AgNPs for 4 hr/day (200 parts per billion/day) for five consecutive days. The nanoparticles were generated using a laser ablation technique using a soft-landing ion mobility (SLIM) instrument. Inductively coupled plasma mass spectrometric (ICP-MS) analysis showed organ-specific accumulation of the nanoparticles, with the highest concentration present in the lungs, followed by the liver and kidneys. Nanoparticle distribution was characterized in the organs using scanning electron microscopy (SEM) and matrix-assisted laser desorption/ionization mass spectrometric (MALDI-MS) imaging. Bone marrow cytotoxicity assay of the cells from the femur of rats showed micronuclei formation and signs of cellular cytotoxicity. Moreover, rats displayed increased levels of circulating lactate and glutathione disulphide (GSSG), as determined by liquid chromatography-mass spectrometry (LC-MS) analysis. Collectively, our observations suggest that inhaled subacute exposure to AgNP results in accumulation of AgNPs in the lungs, liver, and kidneys, preferentially, as well as mediates induced systemic toxicity.


Subject(s)
Metal Nanoparticles/toxicity , Silver/toxicity , Animals , Inhalation Exposure/analysis , Liver/drug effects , Lung/drug effects , Male , Microscopy, Electron, Scanning , Particle Size , Rats , Rats, Wistar , Silver/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Toxicity Tests, Subacute
15.
J Alzheimers Dis ; 78(4): 1453-1471, 2020.
Article in English | MEDLINE | ID: mdl-33164937

ABSTRACT

BACKGROUND: Multiple studies report a strong correlation between traffic-generated air pollution-exposure and detrimental outcomes in the central nervous system (CNS), including Alzheimer's disease (AD). Incidence of AD is rapidly increasing and, worldwide, many live in regions where pollutants exceed regulatory standards. Thus, it is imperative to identify environmental pollutants that contribute to AD, and the mechanisms involved. OBJECTIVE: We investigated the effects of mixed gasoline and diesel engine emissions (MVE) on the expression of factors involved in progression of AD in the hippocampus and cerebrum in a young versus aged mouse model. METHODS: Young (2 months old) and aged (18 months old) male C57BL/6 mice were exposed to either MVE (300µg/m3 PM) or filtered air (FA) for 6 h/d, 7 d/wk, for 50 d. Immunofluorescence and RT-qPCR were used to quantify oxidative stress (8-OHdG) and expression of amyloid-ß protein precursor (AßPP), ß secretase (BACE1), amyloid-ß (Aß), aryl hydrocarbon receptor (AhR), cytochrome P450 (CYP) 1B1, angiotensin-converting enzyme (ACE1), and angiotensin II type 1 (AT1) receptor in the cerebrum and hippocampus, in addition to cerebral microvascular tight junction (TJ) protein expression. RESULTS: We observed age-related increases in oxidative stress, AhR, CYP1B1, Aß, BACE1, and AT1 receptor in the CA1 region of the hippocampus, and elevation of cerebral AßPP, AhR, and CYP1B1 mRNA, associated with decreased cerebral microvascular TJ protein claudin-5. MVE-exposure resulted in further promotion of oxidative stress, and significant increases in AhR, CYP1B1, BACE1, ACE1, and Aß, compared to the young and aged FA-exposed mice. CONCLUSION: Such findings suggest that MVE-exposure exacerbates the expression of factors in the CNS associated with AD pathogenesis in aged populations.


Subject(s)
Alzheimer Disease/genetics , Oxidative Stress/genetics , Vehicle Emissions , 8-Hydroxy-2'-Deoxyguanosine/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cerebrum/metabolism , Cytochrome P-450 CYP1B1/genetics , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Peptidyl-Dipeptidase A/genetics , Receptor, Angiotensin, Type 1/genetics , Receptors, Aryl Hydrocarbon/genetics , Tight Junction Proteins/genetics , Traffic-Related Pollution , Transcriptome/genetics
17.
Environ Sci Technol ; 54(5): 2843-2850, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32036658

ABSTRACT

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.


Subject(s)
Petroleum Pollution , Petroleum , Polycyclic Aromatic Hydrocarbons , Water Pollutants, Chemical , Animals , Apoptosis , Zebrafish
18.
Sci Rep ; 10(1): 1143, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980673

ABSTRACT

Neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are characterized by increased protein aggregation in the brain, progressive neuronal loss, increased inflammation, and neurogenesis impairment. We analyzed the effects of a new purine derivative drug, PDD005, in attenuating mechanisms involved in the pathogenesis of neurodegenerative diseases, using both in vivo and in vitro models. We show that PDD005 is distributed to the brain and can rescue cognitive deficits associated with aging in mice. Treatment with PDD005 prevents impairment of neurogenesis by increasing sex-determining region Y-box 2, nestin, and also enhances synaptic function through upregulation of synaptophysin and postsynaptic density protein 95. PDD005 treatment also reduced neuro-inflammation by decreasing interleukin-1ß expression, activation of astrocytes, and microglia. We identified prohibitin as a potential target in mediating the therapeutic effects of PDD005 for the treatment of cognitive deficit in aging mice. Additionally, in the current study, glycogen synthase kinase appears to attenuate tau pathology.


Subject(s)
Cognition Disorders/prevention & control , Hippocampus/drug effects , Molecular Targeted Therapy , Nerve Tissue Proteins/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Repressor Proteins/antagonists & inhibitors , Tauopathies/prevention & control , Aging/psychology , Animals , Blood-Brain Barrier , Brain/metabolism , Cells, Cultured , Cognition Disorders/drug therapy , Donepezil/pharmacology , Drug Evaluation, Preclinical , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Glycogen Synthase Kinase 3 beta/biosynthesis , Glycogen Synthase Kinase 3 beta/genetics , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurogenesis/drug effects , Neuroglia/drug effects , Neuronal Plasticity/drug effects , Neuroprotective Agents/pharmacokinetics , Phosphorylation/drug effects , Prohibitins , Protein Processing, Post-Translational/drug effects , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Tauopathies/drug therapy , tau Proteins/metabolism
19.
Environ Res ; 181: 108913, 2020 02.
Article in English | MEDLINE | ID: mdl-31753468

ABSTRACT

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.


Subject(s)
Air Pollutants/toxicity , Apolipoproteins E , Gastrointestinal Microbiome , Air Pollution , Animals , Inflammation , Intestines , Male , Mice , Mice, Knockout , RNA, Ribosomal, 16S , Vehicle Emissions
20.
Toxicol Sci ; 170(2): 525-535, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31132127

ABSTRACT

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.


Subject(s)
Angiotensin II/drug effects , Blood-Brain Barrier/drug effects , Central Nervous System/drug effects , Receptor, Angiotensin, Type 1/drug effects , Renin-Angiotensin System/drug effects , Vehicle Emissions/toxicity , Air Pollutants/toxicity , Angiotensin II/metabolism , Animals , Astrocytes/drug effects , Cerebrovascular Circulation , Coculture Techniques , Diet, High-Fat , Gene Expression/drug effects , Inflammation , Inhalation Exposure/adverse effects , Male , Mice , Mice, Inbred C57BL , Microvessels/metabolism , Receptor, Angiotensin, Type 1/metabolism , Tight Junction Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...