Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Small ; 18(52): e2203259, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36373669

RESUMEN

The toxicity of boron nitride nanotubes (BNNTs) has been the subject of conflicting reports, likely due to differences in the residuals and impurities that can make up to 30-60% of the material produced based on the manufacturing processes and purification employed. Four BNNTs manufactured by induction thermal plasma process with a gradient of BNNT purity levels achieved through sequential gas purification, water and solvent washing, allowed assessing the influence of these residuals/impurities on the toxicity profile of BNNTs. Extensive characterization including infrared and X-ray spectroscopy, thermogravimetric analysis, size, charge, surface area, and density captured the alteration in physicochemical properties as the material went through sequential purification. The material from each step is screened using acellular and in vitro assays for evaluating general toxicity, mechanisms of toxicity, and macrophage function. As the material increased in purity, there are more high-aspect-ratio particulates and a corresponding distinct increase in cytotoxicity, nuclear factor-κB transcription, and inflammasome activation. There is no alteration in macrophage function after BNNT exposure with all purity grades. The cytotoxicity and mechanism of screening clustered with the purity grade of BNNTs, illustrating that greater purity of BNNT corresponds to greater toxicity.


Asunto(s)
Compuestos de Boro , Nanotubos , Compuestos de Boro/toxicidad , Compuestos de Boro/química , Macrófagos , Nanotubos/toxicidad , Nanotubos/química
2.
Inhal Toxicol ; 32(1): 24-38, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-32028803

RESUMEN

Objective: In this study, we compared in vitro and in vivo bioactivity of nitrogen-doped multi-walled carbon nanotubes (NDMWCNT) to MWCNT to test the hypothesis that nitrogen doping would alter bioactivity.Materials and Methods: High-resolution transmission electron microscopy (TEM) confirmed the multilayer structure of MWCNT with an average layer distance of 0.36 nm, which was not altered by nitrogen doping: the nanomaterials had similar widths and lengths. In vitro studies with THP-1 cells and alveolar macrophages from C57BL/6 mice demonstrated that NDMWCNT were less cytotoxic and stimulated less IL-1ß release compared to MWCNT. For in vivo studies, male C57BL/6J mice received a single dose of dispersion medium (DM), 2.5, 10 or 40 µg/mouse of NDMWCNT, or 40 µg/mouse of MWCNT by oropharyngeal aspiration. Animals were euthanized between 1 and 7 days post-exposure for whole lung lavage (WLL) studies.Results and Discussion: NDMWCNT caused time- and dose-dependent pulmonary inflammation. However, it was less than that caused by MWCNT. Activation of the NLRP3 inflammasome was assessed in particle-exposed mice by determining cytokine production in WLL fluid at 1 day post-exposure. Compared to DM-exposed mice, IL-1ß and IL-18 were significantly increased in MWCNT- and NDMWCNT-exposed mice, but the increase caused by NDMWCNT was less than MWCNT. At 56 days post-exposure, histopathology determined lung fibrosis in MWCNT-exposed mice was greater than NDMWCNT-exposed mice.Conclusions: These data indicate nitrogen doping of MWCNT decreases their bioactivity, as reflected with lower in vitro and in vivo toxicity inflammation and lung disease. The lower activation of the NLRP3 inflammasome may be responsible. Abbreviations: NDMWCNT: nitrogen-doped multi-walled carbon nanotubes; MWCNT: multi-walled carbon nanotubes; TEM: transmission electron microscopy; HRTEM: high resolution transmission electron microscopy; IL-1ß: interleukin-1ß; DM: dispersion medium; WLL: whole lung lavage; IL-18: interleukin-18; GSD: geometric standard deviation; XPS: X-ray photoelectron spectroscopy; SEM: standard error of the mean; PMA: phorbol 12-myristate 13-acetate; LPS: lipopolysacharride; LDH: lactate dehydrogenase; AM: alveolar macrophage; PMN: polymorphonuclear leukocyte.


Asunto(s)
Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Nitrógeno/toxicidad , Neumonía/inducido químicamente , Animales , Líquido del Lavado Bronquioalveolar/química , Citocinas/análisis , Relación Dosis-Respuesta a Droga , Humanos , Inflamasomas/inmunología , Inflamasomas/metabolismo , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Masculino , Ratones Endogámicos C57BL , Nanotubos de Carbono/química , Nitrógeno/química , Tamaño de la Partícula , Neumonía/inmunología , Neumonía/patología , Propiedades de Superficie , Células THP-1 , Factores de Tiempo
3.
Int J Mol Sci ; 20(11)2019 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-31146342

RESUMEN

As the demand for multi-walled carbon nanotube (MWCNT) incorporation into industrial and biomedical applications increases, so does the potential for unintentional pulmonary MWCNT exposure, particularly among workers during manufacturing. Pulmonary exposure to MWCNTs raises the potential for development of lung inflammation, fibrosis, and cancer among those exposed; however, there are currently no effective biomarkers for detecting lung fibrosis or predicting the risk of lung cancer resulting from MWCNT exposure. To uncover potential mRNAs and miRNAs that could be used as markers of exposure, this study compared in vivo mRNA and miRNA expression in lung tissue and blood of mice exposed to MWCNTs with in vitro mRNA and miRNA expression from a co-culture model of human lung epithelial and microvascular cells, a system previously shown to have a higher overall genome-scale correlation with mRNA expression in mouse lungs than either cell type grown separately. Concordant mRNAs and miRNAs identified by this study could be used to drive future studies confirming human biomarkers of MWCNT exposure. These potential biomarkers could be used to assess overall worker health and predict the occurrence of MWCNT-induced diseases.


Asunto(s)
Enfermedades Pulmonares/sangre , Pulmón/metabolismo , MicroARNs/sangre , Nanotubos de Carbono/toxicidad , ARN Mensajero/sangre , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Línea Celular , Células Cultivadas , Humanos , Pulmón/efectos de los fármacos , Enfermedades Pulmonares/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Exposición Profesional , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Int J Toxicol ; 37(4): 276-284, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29916280

RESUMEN

Respiratory exposure to multiwalled carbon nanotubes (MWCNT) or asbestos results in fibrosis; however, the mechanisms to reach this end point may be different. A previous study by our group identified pulmonary effects and significantly altered messenger RNA (mRNA) signaling pathways following exposure to 1, 10, 40, and 80 µg MWCNT and 120 µg crocidolite asbestos on mouse lungs over time at 1-month, 6-month, and 1-year postexposure following pulmonary aspiration. As a continuation to the above study, this current study took an in-depth look at the signaling pathways involved in fibrosis development at a single time point, 1 year, and exposure, 40 µg MWCNT, the lowest exposure at which fibrosis was pathologically evident. The 120 µg asbestos exposure was included to compare MWCNT-induced fibrosis with asbestos-induced fibrosis. A previously validated computational model was used to identify mRNAs with expression profiles matching the fibrosis pathology patterns from exposed mouse lungs. mRNAs that matched the pathology patterns were then input into ingenuity pathway analysis to determine potential signaling pathways and physiological disease functions inherent to MWCNT and asbestos exposure. Both MWCNT and asbestos exposure induced changes in mouse lungs regarding gene expression, cell proliferation, and survival, while MWCNT uniquely induced alterations in pathways involved in oxidative phosphorylation, mitochondrial dysfunction, and transcription. Asbestos exposure produced unique alterations in pathways involved in sustained inflammation. Although typically considered similar due to scale and fiber-like appearance, the different compositional properties inherent to either MWCNT or asbestos may play a role in their ability to induce fibrosis after pulmonary exposure.


Asunto(s)
Asbesto Crocidolita/toxicidad , Nanotubos de Carbono/toxicidad , Fibrosis Pulmonar/inducido químicamente , Administración por Inhalación , Animales , Expresión Génica/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , ARN Mensajero/metabolismo
5.
Part Fibre Toxicol ; 13(1): 34, 2016 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-27328692

RESUMEN

BACKGROUND: Graphene, a monolayer of carbon, is an engineered nanomaterial (ENM) with physical and chemical properties that may offer application advantages over other carbonaceous ENMs, such as carbon nanotubes (CNT). The goal of this study was to comparatively assess pulmonary and systemic toxicity of graphite nanoplates, a member of the graphene-based nanomaterial family, with respect to nanoplate size. METHODS: Three sizes of graphite nanoplates [20 µm lateral (Gr20), 5 µm lateral (Gr5), and <2 µm lateral (Gr1)] ranging from 8-25 nm in thickness were characterized for difference in surface area, structure,, zeta potential, and agglomeration in dispersion medium, the vehicle for in vivo studies. Mice were exposed by pharyngeal aspiration to these 3 sizes of graphite nanoplates at doses of 4 or 40 µg/mouse, or to carbon black (CB) as a carbonaceous control material. At 4 h, 1 day, 7 days, 1 month, and 2 months post-exposure, bronchoalveolar lavage was performed to collect fluid and cells for analysis of lung injury and inflammation. Particle clearance, histopathology and gene expression in lung tissue were evaluated. In addition, protein levels and gene expression were measured in blood, heart, aorta and liver to assess systemic responses. RESULTS: All Gr samples were found to be similarly composed of two graphite structures and agglomerated to varying degrees in DM in proportion to the lateral dimension. Surface area for Gr1 was approximately 7-fold greater than Gr5 and Gr20, but was less reactive reactive per m(2). At the low dose, none of the Gr materials induced toxicity. At the high dose, Gr20 and Gr5 exposure increased indices of lung inflammation and injury in lavage fluid and tissue gene expression to a greater degree and duration than Gr1 and CB. Gr5 and Gr20 showed no or minimal lung epithelial hypertrophy and hyperplasia, and no development of fibrosis by 2 months post-exposure. In addition, the aorta and liver inflammatory and acute phase genes were transiently elevated in Gr5 and Gr20, relative to Gr1. CONCLUSIONS: Pulmonary and systemic toxicity of graphite nanoplates may be dependent on lateral size and/or surface reactivity, with the graphite nanoplates > 5 µm laterally inducing greater toxicity which peaked at the early time points post-exposure relative to the 1-2 µm graphite nanoplate.


Asunto(s)
Grafito/toxicidad , Pulmón/efectos de los fármacos , Nanopartículas , Nanoestructuras/toxicidad , Animales , Líquido del Lavado Bronquioalveolar , Pulmón/metabolismo , Ratones , Microscopía Electrónica de Rastreo , ARN Mensajero/metabolismo
6.
J Toxicol Environ Health A ; 79(8): 352-66, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27092743

RESUMEN

Pulmonary exposure to multiwalled carbon nanotubes (MWCNT) induces an inflammatory and rapid fibrotic response, although the long-term signaling mechanisms are unknown. The aim of this study was to examine the effects of 1, 10, 40, or 80 µg MWCNT administered by pharyngeal aspiration on bronchoalveolar lavage (BAL) fluid for polymorphonuclear cell (PMN) infiltration, lactate dehydrogenase (LDH) activity, and lung histopathology for inflammatory and fibrotic responses in mouse lungs 1 mo, 6 mo, and 1 yr postexposure. Further, a 120-µg crocidolite asbestos group was incorporated as a positive control for comparative purposes. Results showed that MWCNT increased BAL fluid LDH activity and PMN infiltration in a dose-dependent manner at all three postexposure times. Asbestos exposure elevated LDH activity at all 3 postexposure times and PMN infiltration at 1 mo and 6 mo postexposure. Pathological changes in the lung, the presence of MWCNT or asbestos, and fibrosis were noted at 40 and 80 µg MWCNT and in asbestos-exposed mice at 1 yr postexposure. To determine potential signaling pathways involved with MWCNT-associated pathological changes in comparison to asbestos, up- and down-regulated gene expression was determined in lung tissue at 1 yr postexposure. Exposure to MWCNT tended to favor those pathways involved in immune responses, specifically T-cell responses, whereas exposure to asbestos tended to favor pathways involved in oxygen species production, electron transport, and cancer. Data indicate that MWCNT are biopersistent in the lung and induce inflammatory and fibrotic pathological alterations similar to those of crocidolite asbestos, but may reach these endpoints by different mechanisms.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Asbesto Crocidolita/toxicidad , Exposición por Inhalación , Pulmón/efectos de los fármacos , Pulmón/patología , Nanotubos de Carbono/toxicidad , Animales , Líquido del Lavado Bronquioalveolar/química , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , L-Lactato Deshidrogenasa/genética , L-Lactato Deshidrogenasa/metabolismo , Pulmón/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Factores de Tiempo
7.
Arch Toxicol ; 89(4): 621-33, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25510677

RESUMEN

Multi-walled carbon nanotubes (MWCNT) are new materials with a wide range of industrial and commercial applications. However, their nano-scaled size and fiber-like shape render them respirable and potentially fibrogenic if inhaled into the lungs. To understand MWCNT fibrogenesis, we analyzed the pathologic and molecular aspects of the early phase response to MWCNT in mouse lungs. MWCNT induced rapid and pronounced lesions in the lungs characterized by increased cellularity and formation of fibrotic foci, most notably near where MWCNT deposited, within 14 days post-exposure. Deposition of collagen fibers was markedly increased in the alveolar septa and fibrotic foci, accompanied by elevated expression of fibrotic genes Col1a1, Col1a2, and Fn1 at both mRNA and protein levels. Fibrosis was induced rapidly at 40 µg, wherein fibrotic changes were detected on day 1 and reached a maximal intensity on day 7 through day 14. Induction of fibrosis was dose-dependent at the dose range of 5-40 µg, 7 days post-exposure. MWCNT elicited rapid and prominent infiltrations of neutrophils and macrophages alongside fibrosis implicating acute inflammation in the fibrotic response. At the molecular level, MWCNT induced elevated expression of proinflammatory cytokines TNFα, IL1α, IL1ß, IL6, and CCL2 in lung tissues as well as the bronchoalveolar lavage fluid, in a dose- and time-dependent manner. MWCNT also increased the expression of fibrogenic growth factors TGF-ß1 and PDGF-A in the lungs significantly. These findings underscore the interplay between acute inflammation and the early fibrotic response in the initiation and propagation of pulmonary fibrosis induced by MWCNT.


Asunto(s)
Exposición por Inhalación/efectos adversos , Pulmón/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Fibrosis Pulmonar , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Citocinas/análisis , Fibronectinas/genética , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Factores de Tiempo
8.
Nanotoxicology ; 18(1): 69-86, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38420937

RESUMEN

In the lung, carcinogenesis is a multi-stage process that includes initiation by a genotoxic agent, promotion that expands the population of cells with damaged DNA to form a tumor, and progression from benign to malignant neoplasms. We have previously shown that Mitsui-7, a long and rigid multi-walled carbon nanotube (MWCNT), promotes pulmonary carcinogenesis in a mouse model. To investigate the potential exposure threshold and dose-response for tumor promotion by this MWCNT, 3-methylcholanthrene (MC) initiated (10 µg/g, i.p., once) or vehicle (corn oil) treated B6C3F1 mice were exposed by inhalation to filtered air or MWCNT (5 mg/m3) for 5 h/day for 0, 2, 5, or 10 days and were followed for 17 months post-exposure for evidence of lung tumors. Pulmonary neoplasia incidence in MC-initiated mice significantly increased with each MWCNT exposure duration. Exposure to either MC or MWCNT alone did not affect pulmonary neoplasia incidence compared with vehicle controls. Lung tumor multiplicity in MC-initiated mice also significantly increased with each MWCNT exposure duration. Thus, a significantly higher lung tumor multiplicity was observed after a 10-day MWCNT exposure than following a 2-day exposure. Both bronchioloalveolar adenoma and bronchioloalveolar adenocarcinoma multiplicity in MC-initiated mice were significantly increased following 5- and 10-day MWCNT exposure, while a 2-day MWCNT exposure in MC-initiated mice significantly increased the multiplicity of adenomas but not adenocarcinomas. In this study, even the lowest MWCNT exposure promoted lung tumors in MC-initiated mice. Our findings indicate that exposure to this MWCNT strongly promotes pulmonary carcinogenesis.


Asunto(s)
Neoplasias Pulmonares , Pulmón , Ratones , Animales , Pulmón/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Ratones Endogámicos , Transformación Celular Neoplásica , Carcinogénesis/inducido químicamente , Carcinogénesis/patología , Exposición por Inhalación , Ratones Endogámicos C57BL
9.
Toxicol Appl Pharmacol ; 272(2): 476-89, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23845593

RESUMEN

The fibrous shape and biopersistence of multi-walled carbon nanotubes (MWCNT) have raised concern over their potential toxicity after pulmonary exposure. As in vivo exposure to MWCNT produced a transient inflammatory and progressive fibrotic response, this study sought to identify significant biological processes associated with lung inflammation and fibrosis pathology data, based upon whole genome mRNA expression, bronchoaveolar lavage scores, and morphometric analysis from C57BL/6J mice exposed by pharyngeal aspiration to 0, 10, 20, 40, or 80 µg MWCNT at 1, 7, 28, or 56 days post-exposure. Using a novel computational model employing non-negative matrix factorization and Monte Carlo Markov Chain simulation, significant biological processes with expression similar to MWCNT-induced lung inflammation and fibrosis pathology data in mice were identified. A subset of genes in these processes was determined to be functionally related to either fibrosis or inflammation by Ingenuity Pathway Analysis and was used to determine potential significant signaling cascades. Two genes determined to be functionally related to inflammation and fibrosis, vascular endothelial growth factor A (vegfa) and C-C motif chemokine 2 (ccl2), were confirmed by in vitro studies of mRNA and protein expression in small airway epithelial cells exposed to MWCNT as concordant with in vivo expression. This study identified that the novel computational model was sufficient to determine biological processes strongly associated with the pathology of lung inflammation and fibrosis and could identify potential toxicity signaling pathways and mechanisms of MWCNT exposure which could be used for future animal studies to support human risk assessment and intervention efforts.


Asunto(s)
Biología Computacional/métodos , Contaminantes Ambientales/toxicidad , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente , Fibrosis Pulmonar/inducido químicamente , Transcriptoma , Animales , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Biología Computacional/estadística & datos numéricos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Perfilación de la Expresión Génica , Exposición por Inhalación , Masculino , Cadenas de Markov , Ratones , Ratones Endogámicos C57BL , Método de Montecarlo , Neumonía/genética , Neumonía/inmunología , Neumonía/patología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Transducción de Señal/efectos de los fármacos
10.
Exp Lung Res ; 39(1): 48-57, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23282019

RESUMEN

An association has been observed between indoor mold contamination and lung allergy and asthma. This relationship is not fully understood. 1→3-ß-Glucan is the major cell wall component of fungi and a good marker of fungi exposure. The objective was to evaluate the adjuvant effect of zymosan, a crude yeast cell wall preparation of 1→3-ß-glucan, during ovalbumin (OVA) sensitization in an allergy model. BALB/c mice were sensitized by pharyngeal aspiration with saline, 50 µg of OVA, or OVA with 1, 10, 50, or 75 µg of zymosan on days 0, 7, and 14. One week after sensitization, each sensitized animal group was challenged with an aspiration dose of 50 µg of OVA once a week for 2 weeks. At 1 day after the last aspiration, bronchoalveolar lavage fluid and blood was collected, and markers of lung allergy and inflammation were assessed. An adjuvant effect of zymosan on OVA allergy during sensitization was observed as indicated by significant elevations in lung eosinophils, serum OVA-specific IgE, and lung IL-5 in the groups sensitized with zymosan and OVA. Pulmonary treatment with zymosan also amplified lung inflammation. Elevations were observed in lung neutrophils, TNF-α, and parameters of lung injury in the groups primed with both zymosan and OVA. In nearly all parameters, a non-linear dose-response relationship was observed in the groups primed with OVA and zymosan. The optimum adjuvant dose of zymosan was 10 µg. This study demonstrated an adjuvant effect of zymosan when exposures occurred during the sensitization phase in an OVA-induced allergy model in BALB/c mice.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Asma/tratamiento farmacológico , Modelos Animales de Enfermedad , Hipersensibilidad a las Drogas/tratamiento farmacológico , Zimosan/farmacología , Administración por Inhalación , Animales , Asma/etiología , Asma/inmunología , Líquido del Lavado Bronquioalveolar/química , Relación Dosis-Respuesta Inmunológica , Hipersensibilidad a las Drogas/etiología , Hipersensibilidad a las Drogas/inmunología , Quimioterapia Combinada , Eosinófilos/efectos de los fármacos , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Interleucina-5/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/toxicidad , Organismos Libres de Patógenos Específicos
11.
Part Fibre Toxicol ; 10: 33, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23895460

RESUMEN

BACKGROUND: Prior studies have demonstrated a rapid and progressive acute phase response to bolus aspiration of multi-walled carbon nanotubes (MWCNTs). In this study we sought to test the hypothesis that inhalation exposure to MWCNT produces a fibrotic response and that the response is chronically persistent. To address the hypothesis that inhaled MWCNTs cause persistent morphologic changes, male C57BL/6 J mice were exposed in a whole-body inhalation system to a MWCNT aerosol and the fibrotic response in the alveolar region examined at up to 336 days after termination of exposure. METHODS: Inhalation exposure was to a 5 mg/m3 MWCNT aerosol for 5 hours/day for 12 days (4 times/week for 3 weeks). At the end of inhalation exposures, lungs were either lavaged for analysis of bronchoalveolar lavage (BAL) or preserved by vascular perfusion of fixative while inflated with air at 1, 14, 84, 168 and 336 days post inhalation exposure. Separate, clean-air control groups were also studied. Light microscopy, enhanced darkfield microscopy and field emission electron microscopy (FESEM) of tissue sections were used to analyze the distribution of lung burden following inhalation exposure. Morphometric measurements of Sirius Red staining for fibrillar collagen were used to assess the connective tissue response. Serial section analysis of enhanced darkfield microscope images was used to examine the redistribution of MWCNT fibers within the lungs during the post-exposure period. RESULTS: At day 1 post-exposure 84 ± 3 and 16 ± 2 percent of the lung burden (Mean ± S.E., N = 5) were in the alveolar and airway regions, respectively. Initial distribution within the alveolar region was 56 ± 5, 7 ± 4 and 20 ± 3 percent of lung burden in alveolar macrophages, alveolar airspaces and alveolar tissue, respectively. Clearance reduced the alveolar macrophage burden of MWCNTs by 35 percent between 1 and 168 days post-exposure, while the content of MWCNTs in the alveolar tissue increased by 63 percent. Large MWCNT structures containing greater than 4 fibers were 53.6 percent of the initial lung burden and accounted for the majority of the decline with clearance, while lung burden of singlet MWCNT was essentially unchanged. The mean linear intercept of alveolar airspace, a measure of the expansion of the lungs, was not significantly different between groups. Pulmonary inflammation and damage, measured as the number of polymorphnuclear leukocytes (PMNs) or lactate dehydrogenase activity (LDH) and albumin in BAL, increased rapidly (1 day post) after inhalation of MWCNTs and declined slowly with time post-exposure. The fibrillar collagen in the alveolar region of MWCNT-exposed mice demonstrated a progressive increase in thickness over time (0.17 ± 0.02, 0.22 ± 0.02, 0.26 ± 0.03, 0.25 ± 0.02 and 0.29 ± 0.01 microns for 1, 14, 84, 168 and 336 days post-exposure) and was significantly different from clean-air controls (0.16 ± 0.02) at 84 and (0.15 ± 0.02) at 336 days post-exposure. CONCLUSIONS: Despite the relatively low fraction of the lung burden being delivered to the alveolar tissue, the average thickness of connective tissue in the alveolar region increased by 70% in the 336 days after inhalation exposure. These results demonstrate that inhaled MWCNTs deposit and are retained within the alveolar tissue where they produce a progressive and persistent fibrotic response up to 336 days post-exposure.


Asunto(s)
Exposición por Inhalación/efectos adversos , Nanotubos de Carbono/toxicidad , Alveolos Pulmonares/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Aerosoles , Albúminas/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Colágenos Fibrilares/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Masculino , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/ultraestructura , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Factores de Tiempo
12.
J Toxicol Environ Health A ; 75(18): 1129-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22891886

RESUMEN

Concerns over the potential for multiwalled carbon nanotubes (MWCNT) to induce lung carcinogenesis have emerged. This study sought to (1) identify gene expression signatures in the mouse lungs following pharyngeal aspiration of well-dispersed MWCNT and (2) determine if these genes were associated with human lung cancer risk and progression. Genome-wide mRNA expression profiles were analyzed in mouse lungs (n = 160) exposed to 0, 10, 20, 40, or 80 µg of MWCNT by pharyngeal aspiration at 1, 7, 28, and 56 d postexposure. By using pairwise statistical analysis of microarray (SAM) and linear modeling, 24 genes were selected, which have significant changes in at least two time points, have a more than 1.5-fold change at all doses, and are significant in the linear model for the dose or the interaction of time and dose. Additionally, a 38-gene set was identified as related to cancer from 330 genes differentially expressed at d 56 postexposure in functional pathway analysis. Using the expression profiles of the cancer-related gene set in 8 mice at d 56 postexposure to 10 µg of MWCNT, a nearest centroid classification accurately predicts human lung cancer survival with a significant hazard ratio in training set (n = 256) and test set (n = 186). Furthermore, both gene signatures were associated with human lung cancer risk (n = 164) with significant odds ratios. These results may lead to development of a surveillance approach for early detection of lung cancer and prognosis associated with MWCNT in the workplace.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Exposición por Inhalación/efectos adversos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Nanotubos de Carbono/efectos adversos , Medición de Riesgo/métodos , Adulto , Anciano , Animales , Inteligencia Artificial , Biomarcadores de Tumor/genética , Estudios de Cohortes , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/epidemiología , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Nanotubos de Carbono/química , Estadificación de Neoplasias , Valor Predictivo de las Pruebas , Pronóstico , Estudios Retrospectivos , Organismos Libres de Patógenos Específicos
13.
Front Immunol ; 11: 1186, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32595644

RESUMEN

Pulmonary exposure to certain engineered nanomaterials (ENMs) causes chronic lesions like fibrosis and cancer in animal models as a result of unresolved inflammation. Resolution of inflammation involves the time-dependent biosynthesis of lipid mediators (LMs)-in particular, specialized pro-resolving mediators (SPMs). To understand how ENM-induced pulmonary inflammation is resolved, we analyzed the inflammatory and pro-resolving responses to fibrogenic multi-walled carbon nanotubes (MWCNTs, Mitsui-7) and low-toxicity fullerenes (fullerene C60, C60F). Pharyngeal aspiration of MWCNTs at 40 µg/mouse or C60F at a dose above 640 µg/mouse elicited pulmonary effects in B6C3F1 mice. Both ENMs stimulated acute inflammation, predominated by neutrophils, in the lung at day 1, which transitioned to histiocytic inflammation by day 7. By day 28, the lesion in MWCNT-exposed mice progressed to fibrotic granulomas, whereas it remained as alveolar histiocytosis in C60F-exposed mice. Flow cytometric profiling of whole lung lavage (WLL) cells revealed that neutrophil recruitment was the greatest at day 1 and declined to 36.6% of that level in MWCNT- and 16.8% in C60F-treated mice by day 7, and to basal levels by day 28, suggesting a rapid initiation phase and an extended resolution phase. Both ENMs induced high levels of proinflammatory leukotriene B4 (LTB4) and prostaglandin E2 (PGE2) with peaks at day 1, and high levels of SPMs resolvin D1 (RvD1) and E1 (RvE1) with peaks at day 7. MWCNTs and C60F induced time-dependent polarization of M1 macrophages with a peak at day 1 and subsequently of M2 macrophages with a peak at day 7 in the lung, accompanied by elevated levels of type 1 or type 2 cytokines, respectively. M1 macrophages exhibited preferential induction of arachidonate 5-lipoxygenase activating protein (ALOX5AP), whereas M2 macrophages had a high level expression of arachidonate 15-lipoxygenase (ALOX15). Polarization of macrophages in vitro differentially induced ALOX5AP in M1 macrophages or ALOX15 in M2 macrophages resulting in increased preferential biosynthesis of proinflammatory LMs or SPMs. MWCNTs increased the M1- or M2-specific production of LMs accordingly. These findings support a mechanism by which persistent ENM-induced neutrophilic inflammation is actively resolved through time-dependent polarization of macrophages and enhanced biosynthesis of specialized LMs via distinct ALOX pathways.


Asunto(s)
Fulerenos/toxicidad , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente , Neumonía/inmunología , Animales , Macrófagos/efectos de los fármacos , Ratones , Neumonía/patología
14.
J Toxicol Environ Health A ; 70(2): 118-27, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17365572

RESUMEN

Arginase induction was reported in several inflammatory lung diseases, suggesting that this may be a common feature underlying the pathophysiology of such diseases. As little is known regarding arginase expression in silicosis, the induction and cellular localization of arginase were elucidated in lungs of Sprague-Dawley rats 24 h following exposure to varying doses of silica by intratracheal instillation. Arginase expression was evaluated by activity assay, quantification of arginase I and arginase II mRNA levels using real-time polymerase chain reaction (PCR), and immunohistochemistry. Analyses of cells and fluid obtained by bronchoalveolar lavage (BAL) showed that markers of pulmonary inflammation, tissue damage, activation of alveolar macrophages (AM) and NO production were significantly increased by all silica doses. Arginase activity was increased also in AMs isolated from BAL fluid of silica-treated rats. Silica produced two- and three-fold increases in arginase activity of whole lung at doses of 1 and 5 mg/100 g body weight, respectively. Levels of arginase I mRNA, but not of arginase II mRNA, were similarly elevated. In control lungs, arginase I immunoreactivity was observed only in AMs sparsely dispersed throughout the lung; no inducible nitric oxide synthase (iNOS) immunoreactivity was detected. In silica-treated lungs, arginase I and iNOS were co-expressed in most AMs that were abundantly clustered at inflammatory foci. The rapid induction of arginase I expression in inflammatory lung cells, similar to induction of arginase in other inflammatory lung diseases, implicates elevated arginase activity as a factor in the development of lung damage following exposure to silica.


Asunto(s)
Arginasa/metabolismo , Pulmón/efectos de los fármacos , Dióxido de Silicio/toxicidad , Albúminas/metabolismo , Animales , Arginasa/genética , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Recuento de Leucocitos , Pulmón/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Masculino , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
15.
Nanotoxicology ; 11(8): 1040-1058, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29094619

RESUMEN

Boron nitride nanotubes (BNNTs) are an emerging engineered nanomaterial attracting significant attention due to superior electrical, chemical and thermal properties. Currently, the toxicity profile of this material is largely unknown. Commercial grade BNNTs are composed of a mixture (BNNT-M) of ∼50-60% BNNTs, and ∼40-50% impurities of boron and hexagonal boron nitride. We performed acute in vitro and in vivo studies with commercial grade BNNT-M, dispersed by sonication in vehicle, in comparison to the extensively studied multiwalled carbon nanotube-7 (MWCNT-7). THP-1 wild-type and NLRP3-deficient human monocytic cells were exposed to 0-100 µg/ml and C57BL/6 J male mice were treated with 40 µg of BNNT-M for in vitro and in vivo studies, respectively. In vitro, BNNT-M induced a dose-dependent increase in cytotoxicity and oxidative stress. This was confirmed in vivo following acute exposure increase in bronchoalveolar lavage levels of lactate dehydrogenase, pulmonary polymorphonuclear cell influx, loss in mitochondrial membrane potential and augmented levels of 4-hydroxynonenal. Uptake of this material caused lysosomal destabilization, pyroptosis and inflammasome activation, corroborated by an increase in cathepsin B, caspase 1, increased protein levels of IL-1ß and IL-18 both in vitro and in vivo. Attenuation of these effects in NLRP3-deficient THP-1 cells confirmed NLRP3-dependent inflammasome activation by BNNT-M. BNNT-M induced a similar profile of inflammatory pulmonary protein production when compared to MWCNT-7. Functionally, pretreatment with BNNT-M caused suppression in bacterial uptake by THP-1 cells, an effect that was mirrored in challenged alveolar macrophages collected from exposed mice and attenuated with NLRP3 deficiency. Analysis of cytokines secreted by LPS-challenged alveolar macrophages collected after in vivo exposure to dispersions of BNNT-M showed a differential macrophage response. The observed results demonstrated acute inflammation and toxicity in vitro and in vivo following exposure to sonicated BNNT-M was in part due to NLRP3 inflammasome activation.


Asunto(s)
Compuestos de Boro/toxicidad , Pulmón/efectos de los fármacos , Nanotubos/toxicidad , Estrés Oxidativo/efectos de los fármacos , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Inflamación , Pulmón/metabolismo , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Tamaño de la Partícula , Piroptosis/efectos de los fármacos
16.
Toxicology ; 328: 66-74, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25511174

RESUMEN

There is a current interest in reducing the in vivo toxicity testing of nanomaterials in animals by increasing toxicity testing using in vitro cellular assays; however, toxicological results are seldom concordant between in vivo and in vitro models. This study compared global multi-walled carbon nanotube (MWCNT)-induced gene expression from human lung epithelial and microvascular endothelial cells in monoculture and coculture with gene expression from mouse lungs exposed to MWCNT. Using a cutoff of 10% false discovery rate and 1.5 fold change, we determined that there were more concordant genes (gene expression both up- or downregulated in vivo and in vitro) expressed in both cell types in coculture than in monoculture. When reduced to only those genes involved in inflammation and fibrosis, known outcomes of in vivo MWCNT exposure, there were more disease-related concordant genes expressed in coculture than monoculture. Additionally, different cellular signaling pathways are activated in response to MWCNT dependent upon culturing conditions. As coculture gene expression better correlated with in vivo gene expression, we suggest that cellular cocultures may offer enhanced in vitro models for nanoparticle risk assessment and the reduction of in vivo toxicological testing.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Marcadores Genéticos , Humanos , Exposición por Inhalación/efectos adversos , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Reproducibilidad de los Resultados , Medición de Riesgo
17.
Toxicol Sci ; 144(1): 51-64, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25527334

RESUMEN

Multi-walled carbon nanotubes (MWCNTs) are known for their transient inflammatory and progressive fibrotic pulmonary effects; however, the mechanisms underlying these pathologies are unknown. In this study, we used time-series microarray data of global lung mRNA and miRNA expression isolated from C57BL/6J mice exposed by pharyngeal aspiration to vehicle or 10, 20, 40, or 80 µg MWCNT at 1, 7, 28, or 56 days post-exposure to determine miRNA and mRNA regulatory networks that are potentially involved in MWCNT-induced inflammatory and fibrotic lung etiology. Using a non-negative matrix factorization method, we determined mRNAs and miRNAs with expression profiles associated with pathology patterns of MWCNT-induced inflammation (based on bronchoalveolar lavage score) and fibrosis (based on Sirius Red staining measured with quantitative morphometric analysis). Potential binding targets between pathology-related mRNAs and miRNAs were identified using Ingenuity Pathway Analysis and the miRTarBase, miRecords, and TargetScan databases. Using these experimentally validated and predicted binding targets, we were able to build molecular signaling networks that are potentially reflective of and play a role in MWCNT-induced lung inflammatory and fibrotic pathology. As understanding the regulatory networks between mRNAs and miRNAs in different disease states would be beneficial for understanding the complex mechanisms of pathogenesis, these identified genes and pathways may be useful for determining biomarkers of MWCNT-induced lung inflammation and fibrosis for early detection of disease.


Asunto(s)
Redes Reguladoras de Genes , Marcadores Genéticos , Pulmón/metabolismo , MicroARNs/genética , Nanotubos de Carbono , Neumonía/genética , Fibrosis Pulmonar/genética , ARN Mensajero/genética , Animales , Biología Computacional , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Exposición por Inhalación , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Neumonía/inducido químicamente , Neumonía/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo
18.
Cardiovasc Toxicol ; 13(4): 323-37, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23645470

RESUMEN

Cerium dioxide nanoparticles (CeO2 NPs) are an engineered nanomaterial (ENM) that possesses unique catalytic, oxidative, and reductive properties. Currently, CeO2 NPs are being used as a fuel catalyst but these properties are also utilized in the development of potential drug treatments for radiation and stroke protection. These uses of CeO2 NPs present a risk for human exposure; however, to date, no studies have investigated the effects of CeO2 NPs on the microcirculation following pulmonary exposure. Previous studies in our laboratory with other nanomaterials have shown impairments in normal microvascular function after pulmonary exposures. Therefore, we predicted that CeO2 NP exposure would cause microvascular dysfunction that is dependent on the tissue bed and dose. Twenty-four-hour post-exposure to CeO2 NPs (0-400 µg), mesenteric, and coronary arterioles was isolated and microvascular function was assessed. Our results provided evidence that pulmonary CeO2 NP exposure impairs endothelium-dependent and endothelium-independent arteriolar dilation in a dose-dependent manner. The CeO2 NP exposure dose which causes a 50 % impairment in arteriolar function (EC50) was calculated and ranged from 15 to 100 µg depending on the chemical agonist and microvascular bed. Microvascular assessments with acetylcholine revealed a 33-75 % reduction in function following exposure. Additionally, there was a greater sensitivity to CeO2 NP exposure in the mesenteric microvasculature due to the 40 % decrease in the calculated EC50 compared to the coronary microvasculature EC50. CeO2 NP exposure increased mean arterial pressure in some groups. Taken together, these observed microvascular changes may likely have detrimental effects on local blood flow regulation and contribute to cardiovascular dysfunction associated with particle exposure.


Asunto(s)
Cerio/toxicidad , Vasos Coronarios/efectos de los fármacos , Pulmón/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Nanopartículas/toxicidad , Vasodilatación/efectos de los fármacos , Animales , Arteriolas/efectos de los fármacos , Arteriolas/fisiología , Vasos Coronarios/fisiología , Relación Dosis-Respuesta a Droga , Humanos , Pulmón/irrigación sanguínea , Pulmón/patología , Masculino , Arterias Mesentéricas/fisiología , Técnicas de Cultivo de Órganos , Ratas Sprague-Dawley , Vasoconstricción/efectos de los fármacos , Vasoconstricción/fisiología , Vasodilatación/fisiología
19.
Toxicol Sci ; 131(1): 179-93, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22956629

RESUMEN

Three anatase titanium dioxide (TiO(2)) nanoparticles (NPs) were prepared; nanospheres (NSs), short nanobelts (NB1), and long nanobelts (NB2). These NPs were used to investigate the effect of NP shape and length on lung toxicity. Mice were exposed (0-30 µg per mouse) by pharyngeal aspiration and pulmonary toxicity was assessed over a 112-day time course. Whole lung lavage data indicated that NB1- and NB2-exposed mice, but not NS-exposed mice, had significant dose- and time-dependent pulmonary inflammation and damage. Histopathological analyses at 112 days postexposure determined no interstitial fibrosis in any NS-exposed mice, an increased incidence in 30 µg NB1-exposed mice, and significant interstitial fibrosis in 30 µg NB2-exposed mice. At 112 days postexposure, lung burden of NS was decreased by 96.4% and NB2 by 80.5% from initial deposition levels. At 112 days postexposure, enhanced dark field microscopy determined that alveolar macro- phages were the dominant deposition site, but a fraction of NB1 and NB2 was observed in the alveolar interstitial spaces. For the 30 µg exposure groups at 112 days postexposure, confocal micro- scopy and immunofluorescent staining demonstrated that retained NB2 but not NS were present in the interstitium subjacent to the terminal bronchiole near the normal location of the smallest lymphatic capillaries in the lung. These lymphatic capillaries play a critical role in particle clearance, and the accumulation of NB2, but not NS, suggests possible impaired lymphatic clearance by the high aspect ratio particles. In summary, our data indicate that TiO(2) NP shape alters pulmonary responses, with severity of responses being ranked as NS < NB1 < NB2.


Asunto(s)
Contaminantes Ambientales/toxicidad , Nanopartículas/toxicidad , Neumonía por Aspiración/inducido químicamente , Fibrosis Pulmonar/inducido químicamente , Titanio/toxicidad , Animales , Carga Corporal (Radioterapia) , Relación Dosis-Respuesta a Droga , Contaminantes Ambientales/química , Contaminantes Ambientales/farmacocinética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Nanopartículas/química , Nanopartículas/ultraestructura , Nanosferas/química , Nanosferas/toxicidad , Nanosferas/ultraestructura , Tamaño de la Partícula , Neumonía por Aspiración/metabolismo , Neumonía por Aspiración/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Factores de Tiempo , Titanio/química , Titanio/farmacocinética
20.
Nanotoxicology ; 7(7): 1179-94, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22881873

RESUMEN

This study investigated the in vivo pulmonary toxicity of inhaled multi-walled carbon nanotubes (MWCNT). Mice-inhaled aerosolized MWCNT (10 mg/m³, 5 h/day) for 2, 4, 8 or 12 days. MWCNT lung burden was linearly related to exposure duration. MWCNT-induced pulmonary inflammation was assessed by determining whole lung lavage (WLL) polymorphonuclear leukocytes (PMN). Lung cytotoxicity was assessed by WLL fluid LDH activities. WLL fluid albumin concentrations were determined as a marker of alveolar air-blood barrier integrity. These parameters significantly increased in MWCNT-exposed mice versus controls and were dose-dependent. Histopathologic alterations identified in the lung included (1) bronciolocentric inflammation, (2) bronchiolar epithelial hyperplasia and hypertrophy, (3) fibrosis, (4) vascular changes and (5) rare pleural penetration. MWCNT translocated to the lymph node where the deep paracortex was expanded after 8 or 12 days. Acute inhalation of MWCNT induced dose-dependent pulmonary inflammation and damage with rapid development of pulmonary fibrosis, and also demonstrated that MWCNT can reach the pleura after inhalation exposure.


Asunto(s)
Pulmón/efectos de los fármacos , Pulmón/patología , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente , Neumonía/patología , Aerosoles , Albúminas/análisis , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Supervivencia Celular/efectos de los fármacos , Citocinas/análisis , Espectroscopía de Resonancia por Spin del Electrón , Fibrosis , Exposición por Inhalación , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Nanotubos de Carbono/química , Neutrófilos/efectos de los fármacos , Tamaño de la Partícula , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda