Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
1.
Int J Mol Sci ; 22(19)2021 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-34638570

RESUMEN

The microbial biodegradation of new PLA and PCL materials containing birch tar (1-10% v/v) was investigated. Product of dry distillation of birch bark (Betula pendula Roth) was added to polymeric materials to obtain films with antimicrobial properties. The subject of the study was the course of enzymatic degradation of a biodegradable polymer with antibacterial properties. The results show that the type of the material, tar concentration, and the environment influenced the hydrolytic activity of potential biofilm degraders. In the presence of PCL films, the enzyme activities were higher (except for α-D-glucosidase) compared to PLA films. The highest concentration of birch tar (10% v/v) decreased the activity of hydrolases produced by microorganisms to the most significant extent; however, SEM analysis showed the presence of a biofilm even on plastics with the highest tar content. Based on the results of the biological oxygen demand (BOD), the new materials can be classified as biodegradable but, the biodegradation process was less efficient when compared to plastics without the addition of birch tar.


Asunto(s)
Antiinfecciosos/química , Betula/química , Plásticos Biodegradables/química , Poliésteres/química , Breas/química , Aminopeptidasas/metabolismo , Antiinfecciosos/farmacología , Bacterias/efectos de los fármacos , Bacterias/enzimología , Betula/microbiología , Plásticos Biodegradables/farmacología , Biopelículas , Análisis de la Demanda Biológica de Oxígeno , Destilación , Pruebas de Enzimas , Esterasas/metabolismo , Lipasa/metabolismo , Corteza de la Planta/química , Corteza de la Planta/microbiología , Poliésteres/metabolismo , Breas/farmacología , alfa-Glucosidasas/metabolismo , beta-Glucosidasa/metabolismo
2.
J Cell Mol Med ; 25(15): 7122-7134, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34160882

RESUMEN

Traumatic brain injury (TBI) is a major reason of cerebrovascular and neurological damage. Premorbid conditions such as tobacco smoking (TS) can worsen post-TBI injuries by promoting vascular endothelial impairments. Indeed, TS-induced oxidative stress (OS) and inflammation can hamper the blood-brain barrier (BBB) endothelium. This study evaluated the subsequence of chronic TS exposure on BBB endothelial cells in an established in vitro model of traumatic cell injury. Experiments were conducted on confluent TS-exposed mouse brain microvascular endothelial cells (mBMEC-P5) following scratch injury. The expression of BBB integrity-associated tight junction (TJ) proteins was assessed by immunofluorescence imaging (IF), Western blotting (WB) and quantitative RT-PCR. We evaluated reactive oxygen species (ROS) generation, the nuclear factor 2-related (Nrf2) with its downstream effectors and several inflammatory markers. Thrombomodulin expression was used to assess the endothelial haemostatic response to injury and TS exposure. Our results show that TS significantly decreased Nrf2, thrombomodulin and TJ expression in the BBB endothelium injury models while increased OS and inflammation compared to parallel TS-free cultures. These data suggest that chronic TS exposure exacerbates traumatic endothelial injury and abrogates the protective antioxidative cell responses. The downstream effect was a more significant decline of BBB endothelial viability, which could aggravate subsequent neurological impairments.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/metabolismo , Breas/toxicidad , Contaminación por Humo de Tabaco/efectos adversos , Animales , Barrera Hematoencefálica/citología , Barrera Hematoencefálica/metabolismo , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Breas/farmacología , Trombomodulina/genética , Trombomodulina/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
3.
Chem Res Toxicol ; 34(6): 1588-1603, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34033466

RESUMEN

Widespread smoke from wildfires and biomass burning contributes to air pollution and the deterioration of air quality and human health. A common and major emission of biomass burning, often found in collected smoke particles, is spherical wood tar particles, also known as "tar balls". However, the toxicity of wood tar particles and the mechanisms that govern their health impacts and the impact of their complicated chemical matrix are not fully elucidated. To address these questions, we generated wood tar material from wood pyrolysis and isolated two main subfractions: water-soluble and organic-soluble fractions. The chemical characteristics as well as the cytotoxicity, oxidative damage, and DNA damage mechanisms were investigated after exposure of A549 and BEAS-2B lung epithelial cells to wood tar. Our results suggest that both wood tar subfractions reduce cell viability in exposed lung cells; however, these fractions have different modes of action that are related to their physicochemical properties. Exposure to the water-soluble wood tar fraction increased total reactive oxygen species production in the cells, decreased mitochondrial membrane potential (MMP), and induced oxidative damage and cell death, probably through apoptosis. Exposure to the organic-soluble fraction increased superoxide anion production, with a sharp decrease in MMP. DNA damage is a significant process that may explain the course of toxicity of the organic-soluble fraction. For both subfractions, exposure caused cell cycle alterations in the G2/M phase that were induced by upregulation of p21 and p16. Collectively, both subfractions of wood tar are toxic. The water-soluble fraction contains chemicals (such as phenolic compounds) that induce a strong oxidative stress response and penetrate living cells more easily. The organic-soluble fraction contained more polycyclic aromatic hydrocarbons (PAHs) and oxygenated PAHs and induced genotoxic processes, such as DNA damage.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Células Epiteliales/efectos de los fármacos , Pulmón/efectos de los fármacos , Extractos Vegetales/farmacología , Breas/farmacología , Madera/química , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Biomasa , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Solubilidad , Breas/química , Breas/aislamiento & purificación , Células Tumorales Cultivadas , Agua/química
4.
In Vivo ; 32(3): 493-497, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29695551

RESUMEN

We investigated the anti-metastatic action of nicotine- and tar-removed cigarette smoke extract (CSE) on highly metastatic mouse Colon-26 cells using syngeneic BALB/c mice. Colon-26 cells were injected into the spleen of mice, cells were grown in the spleen as the primary lesion, and some metastasized from the spleen to liver and established a metastatic lesion. CSE (10, 30, and 100%) was intraperitoneally administered daily to the mice for 14 days after tumor inoculation. As a result, the relative spleen weights of CSE-administered mice did not differ significantly from those of the control mice. However, the relative liver weights of CSE 30%-administered mice significantly decreased compared to control mice. In order to identify the active component in CSE, we examined the action of methyl vinyl ketone (MVK) on the invasiveness of Colon-26 cells. MVK significantly reduced the invasiveness of cells. MVK may be a candidate active component of CSE.


Asunto(s)
Humo/efectos adversos , Fumar , Breas/química , Breas/farmacología , Animales , Butanonas/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Nicotina/farmacología , Neoplasias del Recto
5.
Int J Mol Sci ; 19(4)2018 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-29649105

RESUMEN

Atopic dermatitis (AD) is a common inflammatory skin disease. Recent studies have revealed the involvement of T helper (Th)2 cytokines including Interleukin 4 (IL-4) in the pathogenesis of AD. Since epidermal Langerhans cells (LCs) and dermal myeloid dendritic cells (DCs) produce CCL17 and CCL22 that chemoattract Th2 cells, interfering with CCL17 and CCL22 production from LCs and dermal myeloid DCs may be beneficial in the treatment of AD. To investigate this, we stimulated murine bone marrow-derived DCs (BMDCs) with IL-4. IL-4 stimulation produced Ccl17 and Ccl22, which was attenuated by soybean tar Glyteer, a known aryl hydrocarbon receptor (Ahr) activator. Notably, Glyteer treatment blocked the nuclear translocation of Stat6 induced by IL-4 stimulation, suggesting that this treatment impairs the IL-4/Stat6 signaling pathway in BMDCs. Unexpectedly, Glyteer treatment did not potently upregulate the expression of Cyp1a1, a specific Ahr-responsive gene, suggesting that its inhibitory machinery for Ccl17 and Ccl22 expression is likely to operate in an Ahr-independent manner. These findings indicate that Glyteer may exhibit therapeutic potential for AD by downregulating the CCL17 and CCL22 production from DCs in a Th2-deviated microenvironment.


Asunto(s)
Células de la Médula Ósea/citología , Células Dendríticas/citología , Regulación hacia Abajo , Interleucina-4/farmacología , Transducción de Señal/efectos de los fármacos , Breas/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/inmunología , Técnicas de Cultivo de Célula , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL17/metabolismo , Quimiocina CCL22/metabolismo , Medios de Cultivo/química , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/inmunología , Humanos , Ratones , Modelos Biológicos , Factor de Transcripción STAT6/metabolismo
6.
Cell Death Dis ; 8(7): e2931, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28703805

RESUMEN

Filaggrin (FLG) mutation is a well-confirmed genetic aberration in atopic dermatitis (AD). Genome-wide association studies on AD have revealed other susceptibility genes, for example, Ovo-like 1 (OVOL1). Nonetheless, the relation between FLG and OVOL1 is unclear. Because aryl hydrocarbon receptor (AHR; a ligand-activated transcription factor), plays a role in FLG expression in keratinocytes, we hypothesized that AHR regulates FLG expression via OVOL1. To demonstrate this mechanism, we analyzed FLG expression in OVOL1-overexpressing or OVOL1-knockdown normal human epidermal keratinocytes (NHEKs). Furthermore, we tested whether AHR activation by 6-formylindolo(3,2-b)carbazole (FICZ), an endogenous AHR ligand, or Glyteer, clinically used soybean tar, upregulates FLG and OVOL1 expression in NHEKs. We found that (1) OVOL1 regulates FLG expression; (2) AHR activation upregulates OVOL1; and (3) AHR activation upregulates FLG via OVOL1. Moreover, nuclear translocation of OVOL1 was less pronounced in AD skin compared with normal skin. IL-4-treated NHEKs, an in vitro AD skin model, also showed inhibition of the OVOL1 nuclear translocation, which was restored by FICZ and Glyteer. Thus, targeting the AHR-OVOL1-FLG axis may provide new therapeutics for AD.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Dermatitis Atópica/patología , Proteínas de Filamentos Intermediarios/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factores de Transcripción/metabolismo , Carbazoles/farmacología , Técnicas de Cultivo de Célula , Línea Celular , Núcleo Celular/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dermatitis Atópica/metabolismo , Proteínas Filagrina , Humanos , Interleucina-4/farmacología , Proteínas de Filamentos Intermediarios/genética , Queratinocitos/citología , Queratinocitos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Receptores de Hidrocarburo de Aril/genética , Breas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transglutaminasas/genética , Transglutaminasas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
7.
J Dermatol ; 42(2): 171-80, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25482884

RESUMEN

Soybean tar Glyteer (Gly) has been widely used for the treatment of various inflammatory skin diseases in Japan since 1924 as an alternative to coal tar remedy. Recently, coal tar has been shown to induce barrier repair in atopic dermatitis via aryl hydrocarbon receptor (AhR). In this study, we demonstrated that Gly activated AhR by inducing its cytoplasmic to nuclear translocation in keratinocytes. The AhR ligation by Gly was biologically active, with significant and dose-dependent upregulation of CYP1A1 expression, which is a specific marker for AhR activation. Gly upregulated the expression of filaggrin in an AhR-dependent manner because its enhancing effect was completely abrogated in AhR-knockdown keratinocytes. T-helper (Th)2 cytokines inhibited the expression of filaggrin; however, Gly completely restored the Th2-mediated inhibition of filaggrin expression. Furthermore, Gly coordinately upregulated a series of epidermal differentiation complex genes, including involucrin, loricrin and hornerin. In addition, Gly exhibited potent antioxidant activity through the activation of nuclear factor-erythroid 2-related factor-2 (Nrf2) and downstream antioxidant enzymes such as NAD(P)H: quinone oxidoreductase 1 (Nqo1), which actually inhibited the generation of reactive oxygen species in keratinocytes treated with tumor necrosis factor-α or benzo[α]pyrene. In conclusion, antioxidant Gly rescues the downregulated expression of filaggrin (and plausibly other barrier proteins) in a Th2-skewed milieu via AhR activation, which may partly explain its empirical anti-inflammatory therapeutic effects.


Asunto(s)
Antioxidantes/farmacología , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Proteínas de Filamentos Intermediarios/genética , Receptores de Hidrocarburo de Aril/metabolismo , Breas/farmacología , Células Th2/metabolismo , Benzo(a)pireno/farmacología , Proteínas de Unión al Calcio/genética , Línea Celular , Citocromo P-450 CYP1A1/genética , Regulación hacia Abajo/efectos de los fármacos , Proteínas Filagrina , Humanos , Queratinocitos , Proteínas de la Membrana/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Precursores de Proteínas/genética , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología
8.
Toxicology ; 314(1): 1-10, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-23981515

RESUMEN

Smoking is a major risk factor for atherosclerotic vascular diseases, but the mechanism for its genesis is unknown. We have recently shown that the gas phase of cigarette smoke (nicotine- and tar-free cigarette smoke extract; CSE) likely to reach the systemic circulation contains stable substances which cause cytotoxicity like plasma membrane damage and cell death in cultured cells, and also that the plasma membrane damage is caused through sequential activation of protein kinase C (PKC) and NADPH oxidase (NOX) and the resulting generation of reactive oxygen species (PKC/NOX-dependent mechanism), whereas cell death is caused through PKC/NOX-dependent and -independent mechanisms. To identify these stable substances, the CSE was prepared by passing the main-stream smoke of 10 cigarettes through a Cambridge glass fiber filter, trapping of the smoke in a vessel cooled at -80°C, and subsequent dissolution in 10ml of water. The CSE was fractionated into nine fractions using reversed-phase HPLC, and each fraction was screened for cytotoxicity in cultured cells, using propidium iodide uptake assay for cell membrane damage and MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] reduction assay for cell viability. The cytotoxicity was positive in two of the nine fractions (Fr2 and Fr5). After extraction of the active fractions into dichloromethane, GC/MS analysis identified 2-cyclopenten-1-one (CPO) in Fr5 but none in Fr2. After derivatization of the active fractions with O-(2,3,4,5,6-pentafluorobenzyl) hydroxylamine hydrochloride, GC/MS analysis identified acrolein, acetone and propionaldehyde in Fr2, and methyl vinyl ketone (MVK) in Fr5. After 4-h incubation, authentic acrolein and MVK induced concentration-dependent cytotoxicity with EC50 values of 75.9±8.2 and 47.0±8.0µM (mean±SEM; n=3), respectively, whereas acetone, propionaldehyde and CPO were without effect. However, after 24-h incubation, CPO induced concentration-dependent cytotoxicity with an EC50 value of 264.0±16.9µM (n=3). The concentrations of acrolein, MVK and CPO in the CSE were 3368±334, 2429±123 and 392.9±31.8µM (n=4), respectively, which were higher than the cytotoxic concentrations. The cytotoxicity of acrolein and MVK consisted of plasma membrane damage and decreased cell viability: the plasma membrane damage was totally prevented by treatment with an inhibitor of PKC or NOX, whereas the decreased cell viability was only partially prevented by these inhibitors. The cytotoxicity of CPO consisted only of decreased cell viability, which was totally resistant to these inhibitors. These results show that acrolein and MVK are responsible for the acute cytotoxicity of the CSE through PKC/NOX-dependent and -independent mechanisms, whereas CPO is responsible for the delayed cytotoxicity of the CSE through a PKC/NOX-independent mechanism.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Contaminación por Humo de Tabaco/efectos adversos , Contaminación por Humo de Tabaco/análisis , Butanonas/química , Butanonas/toxicidad , Línea Celular Tumoral , Sistema Libre de Células , Cromatografía Líquida de Alta Presión , Ciclopentanos/química , Ciclopentanos/toxicidad , Interpretación Estadística de Datos , Cromatografía de Gases y Espectrometría de Masas , Humanos , Hidroxilaminas , Indicadores y Reactivos , Espectrometría de Masas , NADPH Oxidasas/antagonistas & inhibidores , Nicotina/química , Proteína Quinasa C/antagonistas & inhibidores , Breas/farmacología
9.
Pneumologie ; 67(4): 223-7, 2013 Apr.
Artículo en Alemán | MEDLINE | ID: mdl-23479400

RESUMEN

BACKGROUND: The effect of cigarette smoke (CS) on the phagocytosis of alveolar macrophages is discussed controversially on the basis of in vitro experiments. In this short report we describe the in vivo observations that we have performed. METHODS: For this purpose mice were exposed to CS for three consecutive days. One day later the fluorescent microspheres were administered intratracheally and the lung surface was investigated using long-distance fluorescence microscopy. RESULTS: We found that the numbers of neutrophils which engulfed particles was increased in the CS group as compared to controls. The overall phagocytic activity was not significantly different after CS exposure. CONCLUSIONS: In conclusion the phagocytosis of alveolar macrophages and neutrophils after short time CS exposure was not affected. Further investigations will need to look for the effects of long-term CS exposure and the phagocytosis of living bacteria.


Asunto(s)
Modelos Animales de Enfermedad , Macrófagos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Alveolos Pulmonares/patología , Alveolos Pulmonares/fisiopatología , Fumar/fisiopatología , Breas/farmacología , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/efectos de los fármacos
10.
Drug Metab Pharmacokinet ; 27(2): 200-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22076448

RESUMEN

Aryl hydrocarbon receptor (AhR) activators have been shown to induce members of the cytochrome P450 (P450) 1 family. Here we demonstrate that the AhR activators induce CYP3A4 through human pregnane X receptor (PXR). AhR activators, polycyclic aromatic hydrocarbons (PAHs) and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) increased CYP3A4 reporter activity and CYP3A4 mRNA expression in HepG2 cells. The CYP3A4 reporter activity was also increased by treatment with cigarette tar. The increased CYP3A4 reporter activity was clearly knocked down by the introduction of human PXR-small interfering RNA, but not by that of human AhR-small interfering RNA. The CYP3A4 reporter activity enhanced by overexpression of human PXR was further increased by treatment with PAHs and TCDD as well as by treatment with rifampicin. These results suggest that PAHs contained in cigarette smoke induce CYP3A4 in human liver.


Asunto(s)
Citocromo P-450 CYP3A/biosíntesis , Hidrocarburos Policíclicos Aromáticos/farmacología , Receptores de Esteroides/fisiología , Breas/farmacología , Transcripción Genética/efectos de los fármacos , Citocromo P-450 CYP3A/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inducción Enzimática/efectos de los fármacos , Inducción Enzimática/fisiología , Células Hep G2 , Humanos , Receptor X de Pregnano , Transcripción Genética/fisiología
11.
Respir Res ; 12: 149, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22070100

RESUMEN

BACKGROUND: Cigarette smoke is a major risk factor for chronic obstructive pulmonary disease (COPD), an inflammatory lung disorder. COPD is characterized by an increase in CD8(+) T cells within the central and peripheral airways. We hypothesized that the CD8(+) T cells in COPD patients have increased Toll-like receptor (TLR) expression compared to control subjects due to the exposure of cigarette smoke in the airways. METHODS: Endobronchial biopsies and peripheral blood were obtained from COPD patients and control subjects. TLR4 and TLR9 expression was assessed by immunostaining of lung tissue and flow cytometry of the peripheral blood. CD8(+) T cells isolated from peripheral blood were treated with or without cigarette smoke condensate (CSC) as well as TLR4 and TLR9 inhibitors. PCR and western blotting were used to determine TLR4 and TLR9 expression, while cytokine secretion from these cells was detected using electrochemiluminescence technology. RESULTS: No difference was observed in the overall expression of TLR4 and TLR9 in the lung tissue and peripheral blood of COPD patients compared to control subjects. However, COPD patients had increased TLR4 and TLR9 expression on lung CD8(+) T cells. Exposure of CD8(+) T cells to CSC resulted in an increase of TLR4 and TLR9 protein expression. CSC exposure also caused the activation of CD8(+) T cells, resulting in the production of IL-1ß, IL-6, IL-10, IL-12p70, TNFα and IFNγ. Furthermore, inhibition of TLR4 or TLR9 significantly attenuated the production of TNFα and IL-10. CONCLUSIONS: Our results demonstrate increased expression of TLR4 and TLR9 on lung CD8(+) T cells in COPD. CD8(+) T cells exposed to CSC increased TLR4 and TLR9 levels and increased cytokine production. These results provide a new perspective on the role of CD8(+) T cells in COPD.


Asunto(s)
Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Citocinas/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Breas/farmacología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 9/inmunología , Anciano , Células Cultivadas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/patología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
12.
Ann Biomed Eng ; 36(7): 1142-51, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18452059

RESUMEN

Cigarette smoke has been shown to increase platelet activation and endothelial cell (EC) adhesion molecule expression. In the present study, we utilized a hemodynamic shearing device (HSD) to investigate the above effects in vitro in a combined system of platelets and cultured HUVECs (Human Umblical Vein ECs) under physiological shear stress. We investigated the alteration of E-selectin expression on ECs upon exposure to: (1) platelets and nicotine-free smoke extract (NFE), (2) platelets alone, (3) NFE alone, under physiological shear stress. We additionally confirmed the protective effect of nicotine on platelet activation. We found that: (i) surface expression of E-selectin on ECs was significantly increased upon simultaneous exposure of ECs and platelets to NFE relative to exposure of ECs to either platelets or NFE alone (p < 0.05). (ii) Platelet activation was significantly increased in the presence of NFE (p < 0.05). (iii) Nicotine (200 nM) when added to NFE, significantly reduced platelet activation due to NFE (p < 0.05), an effect additionally confirmed by conventional cigarette extracts which contain nicotine (p < 0.05). We therefore conclude that: (a) NFE and platelets additively increase EC E-selectin surface expression, and (b) nicotine modulates platelet activation regardless of ECs.


Asunto(s)
Cadherinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Nicotina/farmacología , Activación Plaquetaria/fisiología , Humo , Breas/farmacología , Velocidad del Flujo Sanguíneo/fisiología , Presión Sanguínea/fisiología , Células Cultivadas , Humanos , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/fisiología , Activación Plaquetaria/efectos de los fármacos
13.
Respir Res ; 9: 7, 2008 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-18208591

RESUMEN

BACKGROUND: Bronchioles are critical zones in cigarette smoke (CS)-induced lung inflammation. However, there have been few studies on the in vivo dynamics of cytokine gene expression in bronchiolar epithelial cells in response to CS. METHODS: We subjected C57BL/6J mice to CS (whole body exposure, 90 min/day) for various periods, and used laser capture microdissection to isolate bronchiolar epithelial cells for analysis of mRNA by quantitative reverse transcription-polymerase chain reaction. RESULTS: We detected enhanced expression of keratinocyte-derived chemokine (KC), macrophage inflammatory protein-2 (MIP-2), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta) by bronchial epithelial cells after 10 consecutive days of CS exposure. This was mirrored by increases in neutrophils and KC, MIP-2, TNF-alpha, and IL-1beta proteins in the bronchoalveolar lavage (BAL) fluid. The initial inhalation of CS resulted in rapid and robust upregulation of KC and MIP-2 with concomitant DNA oxidation within 1 hr, followed by a return to control values within 3 hrs. In contrast, after CS exposure for 10 days, this initial surge was not observed. As the CS exposure was extended to 4, 12, 18 and 24 weeks, the bronchiolar KC and MIP-2 expression and their levels in BAL fluid were relatively dampened compared to those at 10 days. However, neutrophils in BAL fluid continuously increased up to 24 weeks, suggesting that neutrophil accumulation as a result of long-term CS exposure became independent of KC and MIP-2. CONCLUSION: These findings indicate variable patterns of bronchiolar epithelial cytokine expression depending on the duration of CS exposure, and that complex mechanisms govern bronchiolar molecular dynamics in vivo.


Asunto(s)
Bronquios/metabolismo , Quimiocinas/metabolismo , Exposición por Inhalación , Humo , Breas/farmacología , Contaminación por Humo de Tabaco , Animales , Bronquios/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL
14.
Arch Biochem Biophys ; 465(2): 336-46, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17707332

RESUMEN

Beta-carotene (betaC) supplementation in smokers was unexpectedly associated with increased incidence of lung cancer versus smoking alone. We performed a study in A/J mice to explore possible betaC/cigarette smoke (CS) interactions potentially influencing lung cancer risk in smokers. A/J mice received a diet containing 120 or 600 ppm betaC for six weeks, and exposed to mainstream CS (140 mg total suspended particulates/m(3)) during the last two weeks. Lung transcriptomics analysis revealed that CS induced drug metabolism, oxidative stress, extracellular matrix (ECM) degradation, inflammation markers, and apoptosis. betaC reduced CS-induced inflammation markers and ECM degradation. betaC modulated the CS effect on apoptosis without a clear pro- or anti-apoptotic trend. betaC alone induced only minor changes of gene expression. In conclusion, betaC/CS interactions caused gene regulations in lungs. CS was the main effector. The gene regulations overall did not indicate that betaC exacerbated CS effects. Dose-dependency of betaC effects was minor and not detectable by genome-wide data mining.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Pulmón/metabolismo , Proteoma/metabolismo , Breas/farmacología , Contaminación por Humo de Tabaco , Factores de Transcripción/metabolismo , beta Caroteno/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Exposición a Riesgos Ambientales , Regulación de la Expresión Génica/efectos de los fármacos , Pulmón/efectos de los fármacos , Masculino , Ratones
15.
Heart ; 93(10): 1274-7, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17502323

RESUMEN

BACKGROUND: Smoking is the most prevalent and most preventable risk factor for cardiovascular diseases. Smoking low-tar, low-nicotine cigarettes (light cigarettes) would be expected to be less hazardous than smoking regular cigarettes owing to the lower nicotine and tar yield. OBJECTIVE: To compare the chronic and acute effects of light cigarette and regular cigarette smoking on coronary flow velocity reserve (CFVR). METHODS: 20 regular cigarette smokers (mean (SD) age 24.8 (5.0)), 20 light cigarette smokers (mean age 25.6 (6.4)), and 22 non-smoker healthy volunteers (mean age 25.1 (4.2)) were included. First, each subject underwent echocardiographic examination, including CFVR measurement, after a 12 hour fasting and smokeless period. Two days later, each subject smoked two of their normal cigarettes in a closed room within 15 minutes. Finally, within 20-30 minutes, each subject underwent an echocardiographic examination, including CFVR measurement. RESULTS: Mean (SD) CFVR values were similar in light cigarette and regular cigarette smokers and significantly lower than in the controls (2.68 (0.50), 2.65 (0.61), 3.11 (0.53), p = 0.013). Before and after smoking a paired t test showed that smoking two light cigarettes acutely decreased the CFVR from 2.68 (0.50) to 2.05 (0.43) (p = 0.001), and smoking of two regular cigarettes acutely decreased CFVR from 2.65 (0.61) to 2.18 (0.48) (p = 0.001). CONCLUSION: Smoking low-tar, low-nicotine cigarettes impairs the CFVR as severely as smoking regular cigarettes. CFVR values are similar in light cigarette and regular cigarette smokers and significantly lower than in controls.


Asunto(s)
Enfermedad Coronaria/etiología , Fumar/efectos adversos , Enfermedad Aguda , Adulto , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Velocidad del Flujo Sanguíneo/fisiología , Monóxido de Carbono/análisis , Monóxido de Carbono/farmacología , Enfermedad Crónica , Circulación Coronaria/efectos de los fármacos , Circulación Coronaria/fisiología , Enfermedad Coronaria/fisiopatología , Ecocardiografía , Femenino , Estimulantes Ganglionares/análisis , Estimulantes Ganglionares/farmacología , Humanos , Masculino , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Nicotina/análisis , Nicotina/farmacología , Factores de Riesgo , Fumar/fisiopatología , Breas/análisis , Breas/farmacología
16.
Respir Res ; 8: 10, 2007 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-17288615

RESUMEN

BACKGROUND: Cigarette smoke disrupts the protective barrier established by the airway epithelium through direct damage to the epithelial cells, leading to cell death. Since the morphology of the airway epithelium of smokers does not typically demonstrate necrosis, the most likely mechanism for epithelial cell death in response to cigarette smoke is apoptosis. We hypothesized that cigarette smoke directly up-regulates expression of apoptotic genes, which could play a role in airway epithelial apoptosis. METHODS: Microarray analysis of airway epithelium obtained by bronchoscopy on matched cohorts of 13 phenotypically normal smokers and 9 non-smokers was used to identify specific genes modulated by smoking that were associated with apoptosis. Among the up-regulated apoptotic genes was pirin (3.1-fold, p < 0.002), an iron-binding nuclear protein and transcription cofactor. In vitro studies using human bronchial cells exposed to cigarette smoke extract (CSE) and an adenovirus vector encoding the pirin cDNA (AdPirin) were performed to test the direct effect of cigarette smoke on pirin expression and the effect of pirin expression on apoptosis. RESULTS: Quantitative TaqMan RT-PCR confirmed a 2-fold increase in pirin expression in the airway epithelium of smokers compared to non-smokers (p < 0.02). CSE applied to primary human bronchial epithelial cell cultures demonstrated that pirin mRNA levels increase in a time-and concentration-dependent manner (p < 0.03, all conditions compared to controls). Overexpression of pirin, using the vector AdPirin, in human bronchial epithelial cells was associated with an increase in the number of apoptotic cells assessed by both TUNEL assay (5-fold, p < 0.01) and ELISA for cytoplasmic nucleosomes (19.3-fold, p < 0.01) compared to control adenovirus vector. CONCLUSION: These observations suggest that up-regulation of pirin may represent one mechanism by which cigarette smoke induces apoptosis in the airway epithelium, an observation that has implications for the pathogenesis of cigarette smoke-induced diseases.


Asunto(s)
Bronquios/citología , Bronquios/metabolismo , Proteínas Portadoras/metabolismo , Células Epiteliales/metabolismo , Proteínas Nucleares/metabolismo , Fumar/metabolismo , Breas/farmacología , Apoptosis/efectos de los fármacos , Bronquios/efectos de los fármacos , Dioxigenasas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Humanos , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo , Regulación hacia Arriba/efectos de los fármacos
17.
Am J Physiol Heart Circ Physiol ; 292(1): H76-82, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16920799

RESUMEN

Cigarette smoke (CS) is the most important source of preventable morbidity and mortality in the United States. Recent clinical studies have suggested that, in addition to being a major cardiovascular risk factor, CS promotes the progression of kidney disease. The mechanisms by which CS promotes the progression of chronic kidney disease have not been elucidated. Here we demonstrate for the first time that human mesangial cells (MCs) are endowed with the nicotinic ACh receptors (nAChRs) alpha4, alpha5, alpha7, beta2, beta3, and beta4. Studies performed in other cell types have shown that these nAChRs are ionotropic receptors that function as agonist-regulated Ca(2+) channels. Nicotine induced MC proliferation in a dose-dependent manner. At 10 (-7) M, a concentration found in the plasma of active smokers, nicotine induced MC proliferation [control, 1,328 +/- 50 vs. nicotine, 2,761 +/- 90 counts/minute (cpm); P < 0.05] and increased the synthesis of fibronectin (50%), a critical matrix component involved in the progression of chronic kidney disease. We and others have shown that, in response to PKC activation, MC synthesize reactive oxygen species (ROS) via NADPH oxidase. In the current studies we demonstrate that PKC inhibition as well as diphenyleneiodonium and apocynin, two inhibitors of NADPH oxidase, prevented the effects of nicotine on MC proliferation and fibronectin production, hence establishing ROS as second messengers of the actions of nicotine. Furthermore, nicotine increased the production of ROS as assessed by 2',7'-dichlorofluorescein diacetate fluorescence [control, 184.4 +/- 26 vs. nicotine, 281.5 +/- 26 arbitrary fluorescence units (AFU); n = 5 experiments, P < 0.05]. These studies unveil previously unrecognized mechanisms that indict nicotine, a component of CS, as an agent that may accelerate and promote the progression of kidney disease.


Asunto(s)
Enfermedades Renales/etiología , Células Mesangiales/efectos de los fármacos , Células Mesangiales/fisiología , Nicotina/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Receptores Nicotínicos/metabolismo , Fumar/efectos adversos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Enfermedades Renales/fisiopatología , Células Mesangiales/citología , Fumar/fisiopatología , Breas/farmacología
18.
Biochem Biophys Res Commun ; 353(1): 121-6, 2007 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-17173857

RESUMEN

We have previously reported that cigarette smoke can induce DNA damage in human lung cells without leading to apoptosis or necrosis. In this study, we report that STAT3 is required for the survival of human bronchial epithelial cells (HBECs) following cigarette smoke-induced DNA damage. Cigarette smoke extract (CSE) exposure increases STAT3 phosphorylation (Tyr 705) and DNA binding activity in HBECs. CSE also stimulates IL-6 release and mRNA expression. Anti-IL-6 neutralizing antibody partially blocks STAT3 activation and renders the cells sensitive to CSE-induced DNA damage. Suppression of STAT3 by siRNA results in severe DNA damage and cell death in response to CSE exposure. These findings suggest that STAT3 mediates HBEC survival in response to CSE-induced DNA damage, at least in part, through the IL-6/STAT3 signaling pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Bronquios/citología , Bronquios/fisiología , Células Epiteliales/citología , Células Epiteliales/fisiología , Factor de Transcripción STAT3/metabolismo , Breas/farmacología , Apoptosis/fisiología , Bronquios/efectos de los fármacos , Línea Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Humanos , Contaminación por Humo de Tabaco
19.
Respir Res ; 6: 140, 2005 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-16309548

RESUMEN

BACKGROUND: Increased airway epithelial proliferation is frequently observed in smokers. To elucidate the molecular mechanisms leading to these epithelial changes, we studied the effect of cigarette smoke condensate (CSC) on cell proliferation, wound closure and mitogen activated protein kinase (MAPK) activation. We also studied whether modulation of intracellular glutathione/thiol levels could attenuate CSC-induced cell proliferation. METHODS: Cells of the bronchial epithelial cell line NCI-H292 and subcultures of primary bronchial epithelial cells were used for the present study. The effect of CSC on epithelial proliferation was assessed using 5-bromo-2-deoxyuridine (BrdU) incorporation. Modulation of epithelial wound repair was studied by analysis of closure of 3 mm circular scrape wounds during 72 hours of culture. Wound closure was calculated from digital images obtained at 24 h intervals. Activation of mitogen-activated protein kinases was assessed by Western blotting using phospho-specific antibodies. RESULTS: At low concentrations CSC increased proliferation of NCI-H292 cells, whereas high concentrations were inhibitory as a result of cytotoxicity. Low concentrations of CSC also increased epithelial wound closure of both NCI-H292 and PBEC, whereas at high concentrations closure was inhibited. At low, mitogenic concentrations, CSC caused persistent activation of ERK1/2, a MAPK involved in cell proliferation. Inhibition of cell proliferation by high concentrations of CSC was associated with activation of the pro-apoptotic MAP kinases p38 and JNK. Modulation of intracellular glutathione (GSH)/thiol levels using N-acetyl-L-cysteine, GSH or buthionine sulphoximine (BSO), demonstrated that both the stimulatory and the inhibitory effects of CSC were regulated in part by intracellular GSH levels. CONCLUSION: These results indicate that CSC may increase cell proliferation and wound closure dependent on the local concentration of cigarette smoke and the anti-oxidant status. These findings are consistent with increased epithelial proliferation in smokers, and may provide further insight in the development of lung cancer.


Asunto(s)
Bronquios/metabolismo , Bronquios/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Glutatión/metabolismo , Breas/farmacología , Cicatrización de Heridas/efectos de los fármacos , Bronquios/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
20.
Respir Res ; 6: 108, 2005 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-16168067

RESUMEN

Exhaled nitric oxide (eNO) is decreased by cigarette smoking. The hypothesis that oxides of nitrogen (NOX) in cigarette smoke solution (CSS) may exert a negative feedback mechanism upon NO release from epithelial (AEC, A549, and NHTBE) and basophilic cells (RBL-2H3) was tested in vitro. CSS inhibited both NO production and degranulation (measured as release of beta-hexosaminidase) in a dose-dependent manner from RBL-2H3 cells. Inhibition of NO production by CSS in AEC, A549, and NHTBE cells was also dose-dependent. In addition, CSS decreased expression of NOS mRNA and protein expression. The addition of NO inhibitors and scavengers did not, however, reverse the effects of CSS, nor did a NO donor (SNP) or nicotine mimic CSS. N-acetyl-cysteine, partially reversed the inhibition of beta-hexosaminidase release suggesting CSS may act via oxidative free radicals. Thus, some of the inhibitory effects of CSS appear to be via oxidative free radicals rather than a NOX-related negative feedback.


Asunto(s)
Degranulación de la Célula/efectos de los fármacos , Células Epiteliales/metabolismo , Mastocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Mucosa Respiratoria/metabolismo , Breas/farmacología , Tráquea/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Mastocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo III , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Tráquea/citología , Tráquea/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...