Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Immunol ; 190(2): 531-8, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23248261

RESUMEN

IL-33 is elevated in afflicted tissues of patients with mast cell (MC)-dependent chronic allergic diseases. Based on its acute effects on mouse MCs, IL-33 is thought to play a role in the pathogenesis of allergic disease through MC activation. However, the manifestations of prolonged IL-33 exposure on human MC function, which best reflect the conditions associated with chronic allergic disease, are unknown. In this study, we found that long-term exposure of human and mouse MCs to IL-33 results in a substantial reduction of MC activation in response to Ag. This reduction required >72 h exposure to IL-33 for onset and 1-2 wk for reversion following IL-33 removal. This hyporesponsive phenotype was determined to be a consequence of MyD88-dependent attenuation of signaling processes necessary for MC activation, including Ag-mediated calcium mobilization and cytoskeletal reorganization, potentially as a consequence of downregulation of the expression of phospholipase Cγ(1) and Hck. These findings suggest that IL-33 may play a protective, rather than a causative, role in MC activation under chronic conditions and, furthermore, reveal regulated plasticity in the MC activation phenotype. The ability to downregulate MC activation in this manner may provide alternative approaches for treatment of MC-driven disease.


Asunto(s)
Terapia de Inmunosupresión , Interleucinas/inmunología , Mastocitos/inmunología , Mastocitos/metabolismo , Fenotipo , Actinas/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Humanos , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/farmacología , Mastocitos/efectos de los fármacos , Ratones , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Fosfolipasa C gamma/genética , Fosfolipasa C gamma/inmunología , Fosfolipasa C gamma/metabolismo , Multimerización de Proteína/efectos de los fármacos , Proteínas Proto-Oncogénicas c-hck/genética , Proteínas Proto-Oncogénicas c-hck/inmunología , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología
2.
Small ; 8(18): 2904-12, 2012 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-22777948

RESUMEN

Concern about the use of nanomaterials has increased significantly in recent years due to potentially hazardous impacts on human health. Mast cells are critical for innate and adaptive immune responses, often modulating allergic and pathogenic conditions. Mast cells are well known to act in response to danger signals through a variety of receptors and pathways including IL-33 and the IL-1-like receptor ST2. Here, the involvement of mast cells and the IL-33/ST2 axis in pulmonary and cardiovascular responses to multi-walled carbon nanotube (MWCNT) exposure are examined. Toxicological effects of MWCNTs are observed only in mice with a sufficient population of mast cells and are not observed when mast cells are absent or incapable of responding to IL-33. Our findings establish for the first time that mast cells and the IL-33/ST2 axis orchestrates adverse pulmonary and cardiovascular responses to an engineered nanomaterial, giving insight into a previously unknown mechanism of toxicity. This novel mechanism of toxicity could be used for assessing the safety of engineered nanomaterials and provides a realistic therapeutic target for potential nanoparticle induced toxicities.


Asunto(s)
Interleucinas/metabolismo , Mastocitos/metabolismo , Nanotubos de Carbono/toxicidad , Receptores de Interleucina/metabolismo , Animales , Femenino , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Mastocitos/citología , Ratones , Ratones Endogámicos BALB C
3.
Part Fibre Toxicol ; 9: 38, 2012 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-23072542

RESUMEN

BACKGROUND: The exceptional physical-chemical properties of carbon nanotubes have lead to their use in diverse commercial and biomedical applications. However, their utilization has raised concerns about human exposure that may predispose individuals to adverse health risks. The present study investigated the susceptibility to cardiac ischemic injury following a single exposure to various forms of multi-walled carbon nanotubes (MWCNTs). It was hypothesized that oropharyngeal aspiration of MWCNTs exacerbates myocardial ischemia and reperfusion injury (I/R injury). METHODS: Oropharyngeal aspiration was performed on male C57BL/6J mice with a single amount of MWCNT (0.01 - 100 µg) suspended in 100 µL of a surfactant saline (SS) solution. Three forms of MWCNTs were used in this study: unmodified, commercial grade (C-grade), and functionalized forms that were modified either by acid treatment (carboxylated, COOH) or nitrogenation (N-doped) and a SS vehicle. The pulmonary inflammation, serum cytokine profile and cardiac ischemic/reperfusion (I/R) injury were assessed at 1, 7 and 28 days post-aspiration. RESULTS: Pulmonary response to MWCNT oropharyngeal aspiration assessed by bronchoalveolar lavage fluid (BALF) revealed modest increases in protein and inflammatory cell recruitment. Lung histology showed modest tissue inflammation as compared to the SS group. Serum levels of eotaxin were significantly elevated in the carboxylated MWCNT aspirated mice 1 day post exposure. Oropharyngeal aspiration of all three forms of MWCNTs resulted in a time and/or dose-dependent exacerbation of myocardial infarction. The severity of myocardial injury varied with the form of MWCNTs used. The N-doped MWCNT produced the greatest expansion of the infarct at any time point and required a log concentration lower to establish a no effect level. The expansion of the I/R injury remained significantly elevated at 28 days following aspiration of the COOH and N-doped forms, but not the C-grade as compared to SS. CONCLUSION: Our results suggest that oropharyngeal aspiration of MWCNT promotes increased susceptibility of cardiac tissue to ischemia/reperfusion injury without a significant pulmonary inflammatory response. The cardiac injury effects were observed at low concentrations of MWCNTs and presence of MWCNTs may pose a significant risk to the cardiovascular system.


Asunto(s)
Pulmón/efectos de los fármacos , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Nanotubos de Carbono/toxicidad , Neumonía/inducido químicamente , Administración por Inhalación , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Ácidos Carboxílicos/química , Quimiocina CCL11/sangre , Susceptibilidad a Enfermedades/inducido químicamente , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/patología , Relación Dosis-Respuesta a Droga , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Nanotubos de Carbono/clasificación , Nitrógeno/química , Neumonía/metabolismo , Neumonía/patología
4.
Arch Toxicol ; 85(2): 135-41, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20549194

RESUMEN

Brevetoxins (PbTx) are sodium channel neurotoxins produced by the marine dinoflagellate Karenia brevis during red tide blooms. Inhalation of PbTx in normal individuals and individuals with pre-existing airways disease results in adverse airway symptoms including bronchoconstriction. In animal models of allergic inflammation, inhalation of PbTx results in a histamine H1-mediated bronchoconstriction suggestive of mast cell activation. How mast cells would respond directly to PbTx is unknown. We thus explored the activation of mouse bone marrow-derived mast cells (BMMCs) following exposure to purified PbTx-2. Following in vitro exposure to PbTx-2, we examined cellular viability, mast cell degranulation (ß-hexosaminidase release), intracellular Ca²+ and Na+ flux, and the production of inflammatory mediators (IL-6). PbTx-2 induced significant cellular toxicity within 24 h as measured by LDH release and Annexin-V staining. However, within 1 h of exposure, PbTx-2 induced BMMC degranulation and an increase in IL-6 mRNA expression independent of the high-affinity IgE receptor (FcεRI) stimulation. Activation of BMMCs by PbTx-2 was associated with altered intracellular Ca²+ and Na+ levels. Brevenal, a naturally produced compound that antagonizes the activity of PbTx, prevented changes in intracellular Na+ levels but did not alter activation of BMMCs by PbTx-2. These findings demonstrate that PbTx-2 activates mast cells independent of FcεRI providing insight into critical events in the pathogenesis and a potential therapeutic target in brevetoxin-induced airway symptoms.


Asunto(s)
Toxinas Marinas/toxicidad , Mastocitos/efectos de los fármacos , Mastocitos/fisiología , Neurotoxinas/toxicidad , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea , Señalización del Calcio , Degranulación de la Célula , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Éteres/farmacología , Inmunoglobulina E/metabolismo , Interleucina-6/metabolismo , Transporte Iónico/efectos de los fármacos , Toxinas Marinas/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Neurotoxinas/antagonistas & inhibidores , Oxocinas , Polímeros/farmacología , ARN Mensajero/metabolismo , Receptores de IgE/agonistas , Sodio/metabolismo , Bloqueadores de los Canales de Sodio/antagonistas & inhibidores , Bloqueadores de los Canales de Sodio/toxicidad
5.
Cancer Res ; 67(10): 4716-24, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17510399

RESUMEN

The antiangiogenic protein angiostatin inhibits ATP synthase on the endothelial cell surface, blocking cellular proliferation. To examine the specificity of this interaction, we generated monoclonal antibodies (mAb) directed against ATP synthase. mAb directed against the beta-catalytic subunit of ATP synthase (MAb3D5AB1) inhibits the activity of the F(1) domain of ATP synthase and recognizes the catalytic beta-subunit of ATP synthase. We located the antibody recognition site of MAb3D5AB1 in domains containing the active site of the beta-subunit. MAb3D5AB1 also binds to purified Escherichia coli F(1) with an affinity 25-fold higher than the affinity of angiostatin for this protein. MAb3D5AB1 inhibits the hydrolytic activity of F(1) ATP synthase at lower concentrations than angiostatin. Like angiostatin, MAb3D5AB1 inhibits ATP generation by ATP synthase on the endothelial cell surface in acidic conditions, the typical tumor microenvironment where cell surface ATP synthase exhibits greater activity. MAb3D5AB1 disrupts tube formation and decreases intracellular pH in endothelial cells exposed to low extracellular pH. Neither angiostatin nor MAb3D5AB1 showed an antiangiogenic effect in the corneal neovascularization assay; however, both were effective in the low-pH environment of the chicken chorioallantoic membrane assay. Thus, MAb3D5AB1 shows angiostatin-like properties superior to angiostatin and may be exploited in cancer chemotherapy.


Asunto(s)
Angiostatinas/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , ATPasas de Translocación de Protón Mitocondriales/inmunología , Adenosina Trifosfato/biosíntesis , Animales , Anticuerpos Monoclonales/metabolismo , Sitios de Unión de Anticuerpos , Materiales Biomiméticos , Dominio Catalítico/inmunología , Bovinos , Membrana Corioalantoides/irrigación sanguínea , Neovascularización de la Córnea/tratamiento farmacológico , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Mapeo Epitopo , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , ATPasas de Translocación de Protón Mitocondriales/antagonistas & inhibidores , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Modelos Moleculares , Neovascularización Fisiológica/efectos de los fármacos , Ratas , Ratas Endogámicas F344
6.
Environ Health Perspect ; 121(6): 676-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23649427

RESUMEN

BACKGROUND: Engineered nanomaterials (ENMs) have potential benefits, but they also present safety concerns for human health. Interlaboratory studies in rodents using standardized protocols are needed to assess ENM toxicity. METHODS: Four laboratories evaluated lung responses in C57BL/6 mice to ENMs delivered by oropharyngeal aspiration (OPA), and three labs evaluated Sprague-Dawley (SD) or Fisher 344 (F344) rats following intratracheal instillation (IT). ENMs tested included three forms of titanium dioxide (TiO2) [anatase/rutile spheres (TiO2-P25), anatase spheres (TiO2-A), and anatase nanobelts (TiO2-NBs)] and three forms of multiwalled carbon nanotubes (MWCNTs) [original (O), purified (P), and carboxylic acid "functionalized" (F)]. One day after treatment, bronchoalveolar lavage fluid was collected to determine differential cell counts, lactate dehydrogenase (LDH), and protein. Lungs were fixed for histopathology. Responses were also examined at 7 days (TiO2 forms) and 21 days (MWCNTs) after treatment. RESULTS: TiO2-A, TiO2-P25, and TiO2-NB caused significant neutrophilia in mice at 1 day in three of four labs. TiO2-NB caused neutrophilia in rats at 1 day in two of three labs, and TiO2-P25 and TiO2-A had no significant effect in any of the labs. Inflammation induced by TiO2 in mice and rats resolved by day 7. All MWCNT types caused neutrophilia at 1 day in three of four mouse labs and in all rat labs. Three of four labs observed similar histopathology to O-MWCNTs and TiO2-NBs in mice. CONCLUSIONS: ENMs produced similar patterns of neutrophilia and pathology in rats and mice. Although interlaboratory variability was found in the degree of neutrophilia caused by the three types of TiO2 nanoparticles, similar findings of relative potency for the three types of MWCNTs were found across all laboratories, thus providing greater confidence in these interlaboratory comparisons.


Asunto(s)
Pulmón/efectos de los fármacos , Nanopartículas/toxicidad , Nanotubos de Carbono/toxicidad , Titanio/toxicidad , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , National Institute of Environmental Health Sciences (U.S.) , Neutrófilos/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Estados Unidos
7.
Toxicology ; 302(2-3): 114-22, 2012 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-23026733

RESUMEN

The use and production of multi-walled carbon nanotubes (MWCNTs) have significantly increased over the last decade due to their versatility in numerous applications. Their unique physical and chemical properties make them desirable for various biomedical applications, but the same properties also raise concerns about their safety to human health, particularly at the cellular level. The vascular endothelium could be exposed to nanomaterials either by direct intravenous administration in nanomedicine or by translocation following inhalational exposure in an occupational setting. We hypothesized that direct exposure to MWCNTs will increase the expression of inflammatory markers in human aortic endothelial cells (HAEC). We also investigated the effect of the route of exposure on activation by changing the suspension medium of the MWCNTs. HAEC were treated in vitro with MWCNTs (1 or 10 µg/cm(2)) suspended in either cell culture medium [(M)-MWCNTs] or 10% clinical grade pulmonary surfactant [(S)-MWCNTs]. The zeta potential of the (S)-MWCNTs was significantly more negative than the (M)-MWCNTs suggesting a more stable suspension. Treatment of HAEC with (S)-MWCNTs; as compared to (M)-MWCNTs resulted in a significantly higher up-regulation of mRNA transcripts for cell adhesion molecules VCAM1, SELE, ICAM1 and the chemokine CCL2. Time dependent changes in VCAM1 and CCL2 protein levels were confirmed by immunofluorescence, flow cytometry and ELISA. A label free quantitative mass spectrometry proteomic analysis was utilized to compare protein expression patterns between the two suspensions of MWCNTs. We identified significant expression changes in >200 unique proteins in MWCNT treated HAEC. However, the two suspensions of MWCNTs resulted in different protein expression patterns with the eIF2 pathway as the only common pathway identified between the two suspensions. These data suggest that direct exposure to MWCNTs induces acute inflammatory and protein expression changes in HAEC, which is influenced by the type of media used for suspension of MWCNTs and their resulting zeta potential.


Asunto(s)
Medios de Cultivo/química , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/citología , Nanotubos de Carbono/química , Aorta/citología , Aorta/efectos de los fármacos , Aorta/patología , Adhesión Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Células Endoteliales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Exposición por Inhalación , Nanotubos de Carbono/toxicidad , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
8.
Nanotoxicology ; 5(4): 531-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21043986

RESUMEN

Cerium oxide (CeO2) represents an important nanomaterial with wide ranging applications. However, little is known regarding how CeO2 exposure may influence pulmonary or systemic inflammation. Furthermore, how mast cells would influence inflammatory responses to a nanoparticle exposure is unknown. We thus compared pulmonary and cardiovascular responses between C57BL/6 and B6.Cg-Kit(W-sh) mast cell deficient mice following CeO2 nanoparticle instillation. C57BL/6 mice instilled with CeO2 exhibited mild pulmonary inflammation. However, B6.Cg-Kit(W-sh) mice did not display a similar degree of inflammation following CeO2 instillation. Moreover, C57BL/6 mice instilled with CeO2 exhibited altered aortic vascular responses to adenosine and an increase in myocardial ischemia/reperfusion injury which was absent in B6.Cg-Kit(W-sh) mice. In vitro CeO2 exposure resulted in increased production of PGD2, TNF-α, IL-6 and osteopontin by cultured mast cells. These findings demonstrate that CeO2 nanoparticles activate mast cells contributing to pulmonary inflammation, impairment of vascular relaxation and exacerbation of myocardial ischemia/reperfusion injury.


Asunto(s)
Cerio/toxicidad , Mastocitos/metabolismo , Nanopartículas del Metal/toxicidad , Daño por Reperfusión/metabolismo , Adenosina/farmacología , Análisis de Varianza , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Líquido del Lavado Bronquioalveolar/citología , Quimiocina CCL3 , Regulación de la Expresión Génica/efectos de los fármacos , Histocitoquímica , Interleucina-10 , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/química , Masculino , Mastocitos/patología , Nanopartículas del Metal/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio , Miocardio/química , Osteopontina/genética , Osteopontina/metabolismo , Tamaño de la Partícula , Neumonía/inducido químicamente , Neumonía/metabolismo , Neumonía/patología , Prostaglandina D2 , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA