Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Toxics ; 12(4)2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38668515

RESUMO

The capacity of particulate matter (PM) to enhance and stimulate the expression of pro-inflammatory mediators has been previously demonstrated in non-antigen-presenting cells (human bronchial epithelia). Nonetheless, many proposed mechanisms for this are extrapolated from known canonical molecular pathways. This work evaluates a possible mechanism for inflammatory exacerbation after exposure to PM2.5 (from Puerto Rico) and CuSO4, using human bronchial epithelial cells (BEAS-2B) as a model. The induction of CIITA, MHCII genes, and various pro-inflammatory mediators was investigated. Among these, the phosphorylation of STAT1 Y701 was significantly induced after 4 h of PM2.5 exposure, concurrent with a slight increase in CIITA and HLA-DRα mRNA levels. INFγ mRNA levels remained low amidst exposure time, while IL-6 levels significantly increased at earlier times. IL-8 remained low, as expected from attenuation by IL-6 in the known INFγ-independent inflammation pathway. The effects of CuSO4 showed an increase in HLA-DRα expression after 8 h, an increase in STAT1 at 1 h, and RF1 at 8 h We hypothesize and show evidence that an inflammatory response due to PM2.5 extract exposure in human bronchial epithelia can be induced early via an alternate non-canonical pathway in the absence of INFγ.

2.
Toxics ; 12(1)2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38251037

RESUMO

Graphene-based nanomaterials (GBNs) consist of a single or few layers of graphene sheets or modified graphene including pristine graphene, graphene nanosheets (GNS), graphene oxide (GO), reduced graphene oxide (rGO), as well as graphene modified with various functional groups or chemicals (e.g., hydroxyl, carboxyl, and polyethylene glycol), which are frequently used in industrial and biomedical applications owing to their exceptional physicochemical properties. Given the widespread production and extensive application of GBNs, they can be disseminated in a wide range of environmental mediums, such as air, water, food, and soil. GBNs can enter the human body through various routes such as inhalation, ingestion, dermal penetration, injection, and implantation in biomedical applications, and the majority of GBNs tend to accumulate in the respiratory system. GBNs inhaled and substantially deposited in the human respiratory tract may impair lung defenses and clearance, resulting in the formation of granulomas and pulmonary fibrosis. However, the specific toxicity of the respiratory system caused by different GBNs, their influencing factors, and the underlying mechanisms remain relatively scarce. This review summarizes recent advances in the exposure, metabolism, toxicity and potential mechanisms, current limitations, and future perspectives of various GBNs in the respiratory system.

3.
R Soc Open Sci ; 10(4): 221426, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37063998

RESUMO

Annually, an estimated seven million deaths are linked to exposure to airborne pollutants. Despite extensive epidemiological evidence supporting clear associations between poor air quality and a range of short- and long-term health effects, there are considerable gaps in our understanding of the specific mechanisms by which pollutant exposure induces adverse biological responses at the cellular and tissue levels. The development of more complex, predictive, in vitro respiratory models, including two- and three-dimensional cell cultures, spheroids, organoids and tissue cultures, along with more realistic aerosol exposure systems, offers new opportunities to investigate the cytotoxic effects of airborne particulates under controlled laboratory conditions. Parallel advances in high-resolution microscopy have resulted in a range of in vitro imaging tools capable of visualizing and analysing biological systems across unprecedented scales of length, time and complexity. This article considers state-of-the-art in vitro respiratory models and aerosol exposure systems and how they can be interrogated using high-resolution microscopy techniques to investigate cell-pollutant interactions, from the uptake and trafficking of particles to structural and functional modification of subcellular organelles and cells. These data can provide a mechanistic basis from which to advance our understanding of the health effects of airborne particulate pollution and develop improved mitigation measures.

4.
Altern Lab Anim ; 50(4): 293-309, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35938181

RESUMO

The use of reconstituted human airway (RHuA) epithelial tissues to assess functional endpoints is highly relevant in respiratory toxicology, but standardised methods are lacking. In June 2015, the Institute for In Vitro Sciences (IIVS) held a technical workshop to evaluate the potential for standardisation of methods, including ciliary beat frequency (CBF). The applicability of a protocol suggested in the workshop was assessed in a multi-laboratory ring study. This report summarises the findings, and uses the similarities and differences identified between the laboratories to make recommendations for researchers in the absence of a validated method. Two software platforms for the assessment of CBF were used - Sisson-Ammons Video Analysis (SAVA; Ammons Engineering, Clio, MI, USA) and ciliaFA (National Institutes of Health, Bethesda, MD, USA). Both were utilised for multiple read temperatures, one objective strength (10×) and up to four video captures per tissue, to assess their utility. Two commercial RHuA tissue cultures were used: MucilAir™ (Epithelix, Geneva, Switzerland) and EpiAirway™ (MatTek, Ashland, MA, USA). IL-13 and procaterol were used to induce CBF-specific responses as positive controls. Further testing addressed the impact of tissue acclimation duration, the number of capture fields and objective strengths on baseline CBF readings. Both SAVA and ciliaFA reliably collected CBF data. However, ciliaFA failed to generate accurate CBF measurements above ∼10 Hz. The positive controls were effective, but were subject to inter-laboratory variability. CBF endpoints were generally uniform across replicate tissues, objective strengths and laboratories. Longer tissue acclimation increased the percentage active area, but had minimal impact on CBF. Taken together, these findings support the development and validation of a standardised CBF measurement protocol.


Assuntos
Cílios , Depuração Mucociliar , Epitélio , Humanos , Laboratórios , Software , Estados Unidos
5.
Artigo em Inglês | MEDLINE | ID: mdl-34798932

RESUMO

Nanoclays and graphene oxide nanomaterials represent a class of materials sharing similar shapes constituted of high aspect ratio platelets. The increased production of these materials for various industrial applications increases the risk of occupational exposure, consequently with elevated risk of adverse reactions and development of pulmonary diseases, including lung cancer. In this study, pro-inflammatory responses and genotoxicity were assessed in alveolar epithelial cells (A549) and activated THP-1 macrophages (THP-1a) after exposure to three nanoclays; a pristine (Bentonite) and two surface modified (benzalkonium chloride-coated Nanofil9, and dialkyldimethyl-ammonium-coated NanofilSE3000); graphene oxide (GO) and reduced graphene oxide (r-GO) nanomaterials. The pro-inflammatory response in terms of IL-8 expression was strongest in cells exposed to Bentonite, whereas surface modification resulted in decreased toxicity in both cell lines when exposed to Nanofil9 and NanofilSE3000. GO and r-GO induced a pro-inflammatory response in A549 cells, while no effect was detected with the two nanomaterials on THP-1a cells. The pro-inflammatory response was strongly correlated with in vivo inflammation in mice after intra-tracheal instillation when doses were normalized against surface area. Genotoxicity was assessed as DNA strand breaks, using the alkaline comet assay. In A549 cells, an increase in DNA strand breaks was detected only in cells exposed to Bentonite, whereas Bentonite, NanofilSE3000 and GO caused an increased level of genotoxicity in THP-1a cells. Genotoxicity in THP-1a cells was concordant with the DNA damage in bronchoalveolar lavage fluid cells following 1 and 3 days after intra-tracheal instillation in mice. In conclusion, this study shows that surface modification of pristine nanoclays reduces the inflammatory and genotoxic response in A549 and THP-1a cells, and these in vitro models show comparable toxicity to what seen in previous mouse studies with the same materials.


Assuntos
Argila , Dano ao DNA , Grafite , Nanoestruturas , Células A549 , Animais , Bentonita , Grafite/toxicidade , Humanos , Pulmão , Camundongos , Nanoestruturas/toxicidade , Células THP-1
6.
Regul Toxicol Pharmacol ; 125: 105022, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34333067

RESUMO

The aim of the study was to use multiple in vitro assays to assess the effects of a model irritant, sodium dodecyl sulphate (SDS) (≤10 mM (0.29 %, w/v)), on an in vitro model of the airway, MucilAir™. The use of MucilAir™ in recovery studies was also explored. A 24 h exposure increased IL-8 release at an SDS concentration ≥0.63 mM (0.018 %, w/v). Mucin secretion increased and transepithelial electrical resistance (TEER) decreased at SDS concentrations ≥1.25 mM (0.04 %, w/v). Cytotoxicity (lactate dehydrogenase (LDH) release into basolateral chamber) was observed at SDS concentrations of ≥2.5 mM (0.07 %, w/v). The sensitivity of the assays was IL-8 release > TEER = mucin secretion > LDH release. After 7 days, full or partial recovery was observed for intermediate concentrations of SDS using all assays but not at 5 and 10 mM SDS. Morphologically, erosion and cell loss were observed at these concentrations. Resazurin metabolism at 7 days tended to decrease in a dose-dependent manner at SDS concentrations above 2.5 mM (0.07 %, w/v). Together, these data support a No Observable Effect Level of 0.31 mM (0.009 % w/v) SDS and the use of MucilAir™ as a relevant model for airway toxicity studies.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico , Dodecilsulfato de Sódio/toxicidade , Administração por Inalação , Adulto , Alternativas aos Testes com Animais , Técnicas de Cultura de Células , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Interleucina-8/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Mucinas/efeitos dos fármacos , Medição de Risco , Fatores de Tempo
7.
Basic Clin Pharmacol Toxicol ; 128(2): 204-212, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33176395

RESUMO

The coronavirus responsible for COVID-19, SARS-CoV-2, utilizes a viral membrane spike protein for host cell entry. For the virus to engage in host membrane fusion, SARS-CoV-2 utilizes the human transmembrane surface protease, TMPRSS2, to cleave and activate the spike protein. Camostat mesylate, an orally available well-known serine protease inhibitor, is a potent inhibitor of TMPRSS2 and has been hypothesized as a potential antiviral drug against COVID-19. In vitro human cell and animal studies have shown that camostat mesylate inhibits virus-cell membrane fusion and hence viral replication. In mice, camostat mesylate treatment during acute infection with influenza, also dependent on TMPRSS2, leads to a reduced viral load. The decreased viral load may be associated with an improved patient outcome. Because camostat mesylate is administered as an oral drug, it may be used in outpatients as well as inpatients at all disease stages of SARS-CoV-2 infection if it is shown to be an effective antiviral agent. Clinical trials are currently ongoing to test whether this well-known drug could be repurposed and utilized to combat the current pandemic. In the following, we will review current knowledge on camostat mesylate mode of action, potential benefits as an antiviral agent and ongoing clinical trials.


Assuntos
Antivirais/uso terapêutico , Tratamento Farmacológico da COVID-19 , Ésteres/uso terapêutico , Guanidinas/uso terapêutico , Inibidores de Serina Proteinase/uso terapêutico , Animais , Antivirais/administração & dosagem , Antivirais/efeitos adversos , Reposicionamento de Medicamentos , Ésteres/administração & dosagem , Ésteres/efeitos adversos , Guanidinas/administração & dosagem , Guanidinas/efeitos adversos , Humanos , Camundongos , Segurança do Paciente , Serina Endopeptidases/efeitos dos fármacos , Inibidores de Serina Proteinase/administração & dosagem , Inibidores de Serina Proteinase/efeitos adversos
8.
Pharmacol Res Perspect ; 7(5): e00516, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31467680

RESUMO

Epinephrine HFA (Primatene® Mist) is a newly formulated asthma metered dose inhaler developed to replace the previous Primatene® Mist CFC. The formulation of Epinephrine HFA contains thymol, a substance recognized to be safe by the FDA. Although the content of thymol contained in Epinephrine HFA is much lower compared to many common foods and medications available, there are no known nonclinical data about the chronic toxicity of thymol through inhalation. Two sequential 6-month studies of identical design were conducted to assess the chronic toxicity of inhaled thymol in mice. Four treatment groups, (a) Air; (b) vehicle control; (c) Article-1 (thymol 0.1%); and (d) Article-2 (thymol 0.5%) were assessed in 128 mice for 26 weeks. The mice were sacrificed at the end of the treatment period and a histopathologic evaluation was performed with respect to lungs, bronchial lymph nodes, nasal passages/nasopharynx, and trachea. Forty-five pathologic assessment parameters (PAPs) were evaluated. In total, 5591 data points from 487 mouse organs were assessed. Chronic toxicity index was calculated for 16 PAPs that had multiple histopathologic abnormal observations. The t tests were conducted for these 16 PAPs (Articles-1 and 2 versus Air and vehicle control, respectively), and all P-values were greater than .05 indicating no significant differences between all treatment groups. An evaluation was also conducted for 25 PAPs that had only a very small number of pathologic abnormalities. No significant differences for chronic toxicity were found when comparing mice under long-term repeated exposure of high doses of inhaled thymol and mice that inhaled no thymol.


Assuntos
Epinefrina/química , Pulmão/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Timol/toxicidade , Administração por Inalação , Animais , Estudos de Casos e Controles , Feminino , Pulmão/citologia , Masculino , Camundongos , Modelos Animais , Sistema Respiratório/citologia , Testes de Toxicidade Crônica
9.
Cardiovasc Toxicol ; 19(5): 401-411, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30963444

RESUMO

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.


Assuntos
Poluentes Atmosféricos/toxicidade , Aorta Torácica/efeitos dos fármacos , Cádmio/toxicidade , Córtex Cerebral/efeitos dos fármacos , Poeira , Chumbo/toxicidade , Mineração , Material Particulado/toxicidade , Pneumonia/induzido quimicamente , Animais , Aorta Torácica/fisiopatologia , Córtex Cerebral/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Exposição por Inalação , Masculino , Camundongos Endogâmicos C57BL , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Tamanho da Partícula , Pneumonia/genética , Pneumonia/metabolismo , Medição de Risco , Sibéria , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Vasodilatação/efeitos dos fármacos
10.
Toxicol In Vitro ; 43: 76-86, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28572013

RESUMO

Worldwide, over 4 million premature deaths each year are attributed to the burning of biomass fuels for cooking and heating. Epidemiological studies associate household air pollution with lung diseases, including chronic obstructive pulmonary disease, lung cancer, and respiratory infections. Animal dung, a biomass fuel used by economically vulnerable populations, generates more toxic compounds per mass burned than other biomass fuels. The type of animal dung used varies widely depending on local agro-geography. There are currently neither standardized experimental systems for dung biomass smoke research nor studies assessing the health impacts of different types of dung smoke. Here, we used a novel reproducible exposure system to assess outcomes related to inflammation and respiratory infections in human airway cells exposed to six different types of dung biomass smoke. We report that dung biomass smoke, regardless of species, is pro-inflammatory and activates the aryl hydrocarbon receptor and JNK transcription factors; however, dung smoke also suppresses interferon responses after a challenge with a viral mimetic. These effects are consistent with epidemiological data, and suggest a mechanism by which the combustion of animal dung can directly cause lung diseases, promote increased susceptibility to infection, and contribute to the global health problem of household air pollution.


Assuntos
Esterco , Fumaça/efeitos adversos , Animais , Biomassa , Linhagem Celular , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , MAP Quinase Quinase 4/metabolismo , Poli I-C/farmacologia , Receptores de Hidrocarboneto Arílico/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição RelA/metabolismo
11.
Inhal Toxicol ; 29(2): 65-74, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28330429

RESUMO

This study examined the real-time exposure-response effects of aerosolized carfentanil (CRF) on opioid-induced toxicity, respiratory dynamics and cardiac function in mice. Unrestrained, conscious male CD-1 mice (25-30 g) were exposed to 0.4 or 4.0 mg/m3 of aerosolized CRF for 15 min (Ct = 6 or 60 mg min/m3) in a whole-body plethysmograph chamber. Minute volume (MV), core body temperature (Tc), mean arterial blood pressure (MAP) and heart rate (HR) were evaluated in animals exposed to CRF or sterile H2O. Loss of consciousness and Straub tail were observed in before 1 min following initiation of exposure to 6 or 60 mg min/m3 of CRF. Clinical signs of opioid-induced toxicity were observed in a dose-dependent manner. Exposure to 6 or 60 mg min/m3 of CRF resulted in significant decrease in MV as compared to the controls. MAP, HR and Tc decreased 24 h in animals exposed to either 6 or 60 mg min/m3 of CRF as compared to the controls. Post-exposure administration of naloxone (NX, 0.05 mg/kg, i.m.) did not increase the MV of animals exposed to CRF to control levels within 24 h, but decreased clinical signs of opioid-induced toxicity and the duration of respiratory depression. This is the first study to evaluate real-time respiratory dynamics and cardiac function during exposure and up to 24 h post-exposure to CRF. The evaluation of toxicological signs and respiratory dynamics following exposure to CRF will be useful in the development of therapeutic strategies to counteract the ongoing threat of abuse and overuse of opioids and their synthetic variants.


Assuntos
Analgésicos Opioides/toxicidade , Fentanila/análogos & derivados , Naloxona/uso terapêutico , Antagonistas de Entorpecentes/uso terapêutico , Administração por Inalação , Aerossóis , Animais , Temperatura Corporal/efeitos dos fármacos , Fentanila/toxicidade , Frequência Cardíaca/efeitos dos fármacos , Masculino , Camundongos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/tratamento farmacológico , Inconsciência/induzido quimicamente , Inconsciência/tratamento farmacológico
12.
Inhal Toxicol ; 29(1): 32-41, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28183203

RESUMO

Acute respiratory dynamics and histopathology of the lungs and trachea following inhaled exposure to ammonia were investigated. Respiratory dynamic parameters were collected from male Sprague-Dawley rats (300-350 g) during (20 min) and 24 h (10 min) after inhalation exposure for 20 min to 9000, 20,000, and 23,000 ppm of ammonia in a head-only exposure system. Body weight loss, analysis of blood cells, and lungs and trachea histopathology were assessed 1, 3, and 24 h following inhalation exposure to 20,000 ppm of ammonia. Prominent decreases in minute volume (MV) and tidal volume (TV) were observed during and 24 h post-exposure in all ammonia-exposed animals. Inspiratory time (IT) and expiratory time (ET) followed similar patterns and decreased significantly during the exposure and then increased at 24 h post-exposure in all ammonia-exposed animals in comparison to air-exposed controls. Peak inspiratory (PIF) and expiratory flow (PEF) significantly decreased during the exposure to all ammonia doses, while at 24 h post-exposure they remained significantly decreased following exposure to 20,000 and 23,000 ppm. Exposure to 20,000 ppm of ammonia resulted in body weight loss at 1 and 3 h post-exposure; weight loss was significant at 24 h compared to controls. Exposure to 20,000 ppm of ammonia for 20 min resulted in increases in the total blood cell counts of white blood cells, neutrophils, and platelets at 1, 3, and 24 h post-exposure. Histopathologic evaluation of the lungs and trachea tissue of animals exposed to 20,000 ppm of ammonia at 1, 3, and 24 h post-exposure revealed various morphological changes, including alveolar, bronchial, and tracheal edema, epithelial necrosis, and exudate consisting of fibrin, hemorrhage, and inflammatory cells. The various alterations in respiratory dynamics and damage to the respiratory system observed in this study further emphasize ammonia-induced respiratory toxicity and the relevance of efficacious medical countermeasure strategies.


Assuntos
Amônia/toxicidade , Pulmão/efeitos dos fármacos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Administração por Inalação , Animais , Peso Corporal/efeitos dos fármacos , Contagem de Leucócitos , Pulmão/patologia , Masculino , Ratos Sprague-Dawley , Traqueia/efeitos dos fármacos , Traqueia/patologia
13.
Am J Physiol Lung Cell Mol Physiol ; 311(6): L1222-L1233, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27836898

RESUMO

Animal dung is a biomass fuel burned by vulnerable populations who cannot afford cleaner sources of energy, such as wood and gas, for cooking and heating their homes. Exposure to biomass smoke is the leading environmental risk for mortality, with over 4,000,000 deaths each year worldwide attributed to indoor air pollution from biomass smoke. Biomass smoke inhalation is epidemiologically associated with pulmonary diseases, including chronic obstructive pulmonary disease (COPD), lung cancer, and respiratory infections, especially in low and middle-income countries. Yet, few studies have examined the mechanisms of dung biomass smoke-induced inflammatory responses in human lung cells. Here, we tested the hypothesis that dung biomass smoke causes inflammatory responses in human lung cells through signaling pathways involved in acute and chronic lung inflammation. Primary human small airway epithelial cells (SAECs) were exposed to dung smoke at the air-liquid interface using a newly developed, automated, and reproducible dung biomass smoke generation system. The examination of inflammatory signaling showed that dung biomass smoke increased the production of several proinflammatory cytokines and enzymes in SAECs through activation of the activator protein (AP)-1 and arylhydrocarbon receptor (AhR) but not nuclear factor-κB (NF-κB) pathways. We propose that the inflammatory responses of lung cells exposed to dung biomass smoke contribute to the development of respiratory diseases.


Assuntos
Biomassa , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Inflamação/metabolismo , Inflamação/patologia , Pulmão/patologia , Transdução de Sinais , Fumaça/efeitos adversos , Animais , Compostos Azo/farmacologia , Ciclo-Oxigenase 2/metabolismo , Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Cavalos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Material Particulado/análise , Pirazóis/farmacologia , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Receptores de Hidrocarboneto Arílico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição AP-1/metabolismo
14.
Inhal Toxicol ; 28(2): 71-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26821737

RESUMO

This study examined acute toxicity and lung injury following inhalation exposure to ammonia. Male Sprague-Dawley rats (300-350 g) were exposed to 9000, 20,000, 23,000, 26,000, 30,000 or 35,000 ppm of ammonia for 20 min in a custom head-out exposure system. The exposure atmosphere, which attained steady state within 3 min for all ammonia concentrations, was monitored and verified using a Fourier transform infrared spectroscopy (FTIR) gas analyzer. Animals exposed to ammonia resulted in dose-dependent increases in observed signs of intoxication, including increased chewing and licking, ocular irritation, salivation, lacrimation, oronasal secretion and labored breathing. The LCt50 of ammonia within this head-out inhalation exposure model was determined by probit analysis to be 23,672 ppm (16,489 mg/m(3)) for the 20 min exposure in male rats. Exposure to 20,000 or 23,000 ppm of ammonia resulted in significant body weight loss 24-h post-exposure. Lung edema increased in all ammonia-exposed animal groups and was significant following exposure to 9000 ppm. Bronchoalveolar fluid (BALF) protein concentrations significantly increased following exposure to 20,000 or 23,000 ppm of ammonia in comparison to controls. BAL cell (BALC) death and total cell counts increased in animals exposed to 20,000 or 23,000 ppm of ammonia in comparison to controls. Differential cell counts of white blood cells, neutrophils and platelets from blood and BALF were significantly increased following exposure to 23,000 ppm of ammonia. The following studies describe the validation of a head-out inhalation exposure model for the determination of acute ammonia-induced toxicity; this model will be used for the development and evaluation of potential therapies that provide protection against respiratory and systemic toxicological effects.


Assuntos
Amônia/toxicidade , Lesão Pulmonar/patologia , Pulmão/efeitos dos fármacos , Amônia/administração & dosagem , Animais , Líquido da Lavagem Broncoalveolar/citologia , Exposição por Inalação , Masculino , Neutrófilos , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
Toxicol Lett ; 244: 21-27, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26562769

RESUMO

Phosgene (CG), a toxic inhalation and industrial hazard, causes bronchoconstriction, vasoconstriction and associated pathological effects that could be life threatening. Ion channels of the transient receptor potential (TRP) family have been identified to act as specific chemosensory molecules in the respiratory tract in the detection, control of adaptive responses and initiation of detrimental signaling cascades upon exposure to various toxic inhalation hazards (TIH); their activation due to TIH exposure may result in broncho- and vasoconstriction. We studied changes in the regulation of intracellular free Ca(2+) concentration ([Ca(2+)]i) in cultures of human bronchial smooth muscle cells (BSMC) and human pulmonary microvascular endothelial cells (HPMEC) exposed to CG (16ppm, 8min), using an air/liquid interface exposure system. CG increased [Ca(2+)]i (p<0.05) in both cell types, The CG-induced [Ca(2+)]i was blocked (p<0.05) by two types of TRP channel blockers, SKF-96365, a general TRP channel blocker, and RR, a general TRPV (vanilloid type) blocker, in both BSMC and HPMEC. These effects correlate with the in vivo efficacies of these compounds to protect against lung injury and 24h lethality from whole body CG inhalation exposure in mice (8-10ppm×20min). Thus the TRP channel mechanism appears to be a potential target for intervention in CG toxicity.


Assuntos
Antídotos/farmacologia , Brônquios/efeitos dos fármacos , Substâncias para a Guerra Química/toxicidade , Células Endoteliais/efeitos dos fármacos , Moduladores de Transporte de Membrana/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosgênio/toxicidade , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Animais , Brônquios/metabolismo , Brônquios/patologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Imidazóis/farmacologia , Exposição por Inalação , Masculino , Camundongos , Terapia de Alvo Molecular , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/metabolismo
16.
Toxicol Sci ; 147(2): 466-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26149495

RESUMO

The study objective was to assess age-related changes in glutathione (GSH) adaptive response to cigarette smoke (CS) exposure. Older cigarette smokers show a decline (67%) in lung epithelial lining fluid (ELF) GSH and a 1.8-fold decreased GSH adaptive response to cigarette smoking with a concomitant elevation (47%) of exhaled nitric oxide compared with younger smokers. In order to isolate the changes in tissue GSH from other age-related effects, pharmacological inhibition of the rate limiting step in GSH synthesis was employed to examine the lung's response to CS exposure in young mice. The γ-glutamylcysteine ligase inhibitor L-buthionine-sulfoximine (BSO) was administered in the drinking water (20 mM) to decrease by half the in vivo GSH levels to those found in aged mice and humans. Mice were then exposed to CS (3 h/day) for 5 or 15 days. Biochemical analysis of the ELF and lung tissue revealed an inhibition of the CS-induced GSH adaptive response by BSO with a concurrent increase in mixed protein-GSH disulfides indicating increased cysteine oxidation. The prevention of the GSH adaptive response led to an increase in pro-inflammatory cytokines present in the lung. Airspace enlargement is a hallmark of lung emphysema and was observed in mice treated with BSO and exposed to CS for as little as 15 days, whereas these types of changes normally take up to 6 months in this model. BSO treatment potentiated both lung elastase and matrix metalloproteinase activity in the CS group. These data suggest that age-related decline in the GSH adaptive response can markedly accelerate many of the factors thought to drive CS-induced emphysema.


Assuntos
Glutationa/deficiência , Inflamação/induzido quimicamente , Sistema Respiratório/efeitos dos fármacos , Fumar/efeitos adversos , Adolescente , Adulto , Fatores Etários , Idoso , Animais , Líquido da Lavagem Broncoalveolar/química , Citocinas/análise , Glutationa/análise , Glutationa/metabolismo , Glutationa/fisiologia , Humanos , Inflamação/fisiopatologia , Pulmão/química , Pulmão/efeitos dos fármacos , Pulmão/fisiopatologia , Masculino , Metaloproteinases da Matriz/análise , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Sistema Respiratório/fisiopatologia , Adulto Jovem
17.
Food Chem Toxicol ; 84 Suppl: S50-6, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26206495

RESUMO

The use of this material under current use conditions is supported by the existing information. This material was evaluated for genotoxicity, repeated dose toxicity, developmental toxicity, reproductive toxicity, local respiratory toxicity, phototoxicity, skin sensitization potential as well as environmental safety. Repeated dose, developmental, and reproductive toxicities were determined to have the most conservative systemic exposure derived NO[A]EL of 50 mg/kg/day, based on OECD gavage toxicity studies in rats, that resulted in a MOE of 4545455 after considering 100% absorption from skin contact and inhalation. A MOE of >100 is deemed acceptable.


Assuntos
Pentanóis/toxicidade , Perfumes/toxicidade , Testes de Toxicidade , Animais , Qualidade de Produtos para o Consumidor , Dano ao DNA , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Determinação de Ponto Final , Humanos , Nível de Efeito Adverso não Observado , Pentanóis/química , Perfumes/química , Ratos , Medição de Risco
18.
Inhal Toxicol ; 27(9): 432-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26207672

RESUMO

Respiratory dynamics were investigated in head-out plethysmography chambers following inhalational exposure to soman in untreated, non-anesthetized rats. A multipass saturator cell was used to generate 520, 560 and 600 mg × min/m(3) of soman vapor in a customized inhalational exposure system. Various respiratory dynamic parameters were collected from male Sprague-Dawley rats (300--350 g) during (20 min) and 24 h (10 min) after inhalational exposure. Signs of CWNA-induced cholinergic crisis were observed in all soman-exposed animals. Percentage body weight loss and lung edema were observed in all soman-exposed animals, with significant increases in both at 24 h following exposure to 600 mg × min/m(3). Exposure to soman resulted in increases in respiratory frequency (RF) in animals exposed to 560 and 600 mg × min/m(3) with significant increases following exposure to 560 mg × min/m(3) at 24 h. No significant alterations in inspiratory time (IT) or expiratory time (ET) were observed in soman-exposed animals 24 h post-exposure. Prominent increases in tidal volume (TV) and minute volume (MV) were observed at 24 h post-exposure in animals exposed to 600 mg × min/m(3). Peak inspiratory (PIF) and expiratory flow (PEF) followed similar patterns and increased 24 h post-exposure to 600 mg × min/m(3) of soman. Results demonstrate that inhalational exposure to 600 mg × min/m(3) soman produces notable alterations in various respiratory dynamic parameters at 24 h. The following multitude of physiological changes in respiratory dynamics highlights the need to develop countermeasures that protect against respiratory toxicity and lung injury.


Assuntos
Inibidores da Colinesterase/toxicidade , Intoxicação por Gás/fisiopatologia , Agentes Neurotóxicos/toxicidade , Intoxicação por Organofosfatos/fisiopatologia , Mucosa Respiratória/efeitos dos fármacos , Sistema Respiratório/efeitos dos fármacos , Soman/toxicidade , Administração por Inalação , Animais , Biomarcadores , Fluorocarbonos/administração & dosagem , Fluorocarbonos/efeitos adversos , Masculino , Veículos Farmacêuticos/administração & dosagem , Veículos Farmacêuticos/efeitos adversos , Edema Pulmonar/etiologia , Ventilação Pulmonar/efeitos dos fármacos , Ratos Sprague-Dawley , Mucosa Respiratória/metabolismo , Mucosa Respiratória/fisiopatologia , Taxa Respiratória/efeitos dos fármacos , Sistema Respiratório/fisiopatologia , Rinite/etiologia , Convulsões/etiologia , Volume de Ventilação Pulmonar/efeitos dos fármacos , Volatilização , Redução de Peso/efeitos dos fármacos
20.
Toxicol Sci ; 146(2): 244-53, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25962394

RESUMO

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.


Assuntos
Vasos Coronários/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ozônio/toxicidade , Vasodilatação/efeitos dos fármacos , Acetilcolina/administração & dosagem , Animais , Sangue , Vasos Coronários/metabolismo , Exposição por Inalação , Masculino , Camundongos , Ozônio/administração & dosagem , Ratos , Ratos Sprague-Dawley , Serotonina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA