Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Cell Mol Med ; 26(4): 1060-1070, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34984827

RESUMEN

Berberine (BBR), an isoquinoline alkaloid, is used to treat gastrointestinal disorders as an herbal medicine in China. The aim of this study was to investigate the anti-inflammatory activities of BBR in a mouse model with acute graft-versus-host disease (aGVHD). Mice were intravenously injected with bone marrow cells from donors combined with splenocytes to develop aGVHD. The body weight, survival rate and clinical scores were monitored. Then the levels of inflammatory cytokines, histological changes (lung, liver and colon), colonic mucosal barrier and gut microbiota were analysed. Moreover, the toll-like receptor 4 (TLR4)/myeloid differentiation primary response gene 88 (Myd88)/nuclear factor-κB signalling pathway, NLRP3 inflammasome and its cytokines' expressions were determined. The results showed that the gavage of BBR lessened GVHD-induced weight loss, high mortality and clinical scores, inhibited inflammation and target organs damages and prevented GVHD-indued colonic barrier damage. Additionally, BBR modulated gut microbiota, suppressed the activation of the TLR4 signaling pathway and inhibited NLRP3 inflammasome and its cytokine release. This study indicated that BBR might be a potential therapy for aGVHD through NLRP3 inflammasome inhibition.


Asunto(s)
Berberina , Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped , Animales , Berberina/farmacología , Berberina/uso terapéutico , Colon/patología , Enfermedad Injerto contra Huésped/patología , Inflamasomas/metabolismo , Ratones , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Toll-Like 4/metabolismo
2.
Bioorg Med Chem ; 30: 115954, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33360197

RESUMEN

Acute respiratory distress syndrome/chronic obstructive pulmonary disease (ARDS/COPD) is a diffuse inflammatory injury of the lung that is characterized by respiratory distress and vascular leakage and is caused by various factors. Although the treatment strategy for ARDS/COPD continues to be improved, it still lacks effective drugs. MCC950 is a potent and selective inhibitor ofthe nucleotide-binding oligomerization domain-like-receptor family pyrin domain-containing 3 (NLRP3) inflammasome. However, there have been no reports on the effects of MCC950 on lipopolysaccharide (LPS)-induced lung inflammation in mice. The objective of the present study was to evaluate the effects of MCC950 (given either intranasally or intraperitoneally) on inhibiting LPS-induced lung inflammation in mice. Acute lung inflammation was induced by intratracheal administration of LPS in ICR mice. The results showed that MCC950 at 50 mg/kg efficiently suppressed neutrophil lymphocytes (p < 0.001) and macrophage accumulation (p < 0.01) in bronchoalveolar lavage fluid (BALF) in LPS-instilled mice. In addition, hematoxylin and eosin (H&E) staining revealed that MCC950 at 50 mg/kg significantly inhibited pathological progress in the lung tissues (p < 0.01). Furthermore, treatment with MCC950 substantially reduced mRNA expression of IL-1ß, IL-8, TGF-ß1, and MMP-9 and also reduced protein levels of IL-1ß, IL-18 and caspase-1 at 24 h after LPS instillation. The results of the present study indicate that MCC950 effectively inhibits LPS-induced lung inflammation in vivo, which can be considered for clinical translation.


Asunto(s)
Furanos/farmacología , Indenos/farmacología , Lipopolisacáridos/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Neumonía/tratamiento farmacológico , Sulfonamidas/farmacología , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos ICR , Estructura Molecular , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Neumonía/inducido químicamente , Neumonía/metabolismo , Relación Estructura-Actividad
3.
Toxicol Lett ; 310: 61-69, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31018152

RESUMEN

PM2.5 is the main particulate air pollutant that is capable of inducing airway injury. Previous studies have indicated that Rac1 is involved in cigarette smoke-induced lung inflammation and lipopolysaccharide-mediated pulmonary injury. However, the contribution of Rac1 activity to PM2.5-induced lung inflammation remains largely unclear. Here, we investigated the regulation of Rac1 in PM2.5-induced inflammation in mouse airways and human bronchial epithelial cells (16HBE). The lungs of mice exposed to PM2.5 showed increased IL-1ß expression and an accumulation of inflammatory cells, thereby indicating high Rac1 activity. The exposure of 16HBE cells to PM2.5 resulted in elevated Rac1 levels, as well as an increased release of IL-1ß. Particularly, the selective inhibition of Rac1 ameliorated the IL-1ß release and inflammation in model lungs. Histological assessment showed that treatment with a Rac1 inhibitor, NSC23766, reduced the infiltration of neutrophils and macrophages into the airway lumen. Moreover, the selective inhibition or knockdown of Rac1 decreased IL-1ß release in 16HBE cells induced by PM2.5, which correlated with PM2.5-induced Rac1-regulated AKT signaling. Our data suggest an important role for Rac1 in the pathological alterations associated with PM2.5-mediated lung inflammation. Rac1 may be a promising therapeutic target for the treatment of the inflammatory diseases induced by PM2.5 inhalation.


Asunto(s)
Aminoquinolinas/farmacología , Antiinflamatorios/farmacología , Pulmón/efectos de los fármacos , Neuropéptidos/antagonistas & inhibidores , Material Particulado/toxicidad , Neumonía/prevención & control , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Animales , Línea Celular , Humanos , Exposición por Inhalación/efectos adversos , Interleucina-1beta/metabolismo , Pulmón/enzimología , Pulmón/patología , Masculino , Ratones Endogámicos ICR , Neuropéptidos/metabolismo , Tamaño de la Partícula , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/patología , Interferencia de ARN , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
4.
Biochim Biophys Acta Mol Basis Dis ; 1863(7): 1778-1788, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28428003

RESUMEN

Cigarette smoke (CS) is a major risk factor for the development of chronic obstructive pulmonary disease (COPD). Our previous studies have indicated that Rac1 is involved in lipopolysaccharide-induced pulmonary injury and CS-mediated epithelial-mesenchymal transition. However, the contribution of Rac1 activity to CS-induced lung inflammation remains not fully clear. In this study, we investigated the regulation of Rac1 in CS-induced pulmonary inflammation. Mice or 16HBE cells were exposed to CS or cigarette smoke extract (CSE) to induce acute inflammation. The lungs of mice exposed to CS showed an increase in the release of interleukin-6 (IL-6) and keratinocyte-derived chemokine (KC), as well as an accumulation of inflammatory cells, indicating high Rac1 activity. The exposure of 16HBE cells to CSE resulted in elevated Rac1 levels, as well as increased release of IL-6 and interleukin-8 (IL-8). Selective inhibition of Rac1 ameliorated the release of IL-6 and KC as well as inflammation in the lungs of CS-exposed mice. Histological assessment showed that treatment with a Rac1 inhibitor, NSC23766, led to a decrease in CD68 and CD11b positive cells and the infiltration of neutrophils and macrophages into the alveolar spaces. Selective inhibition or knockdown of Rac1 decreased IL-6 and IL-8 release in 16HBE cells induced by CSE, which correlated with CSE-induced Rac1-regulated Erk1/2 mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription-3 (STAT3) signaling. Our data suggest an important role for Rac1 in the pathological alterations associated with CS-mediated inflammation. Rac1 may be a promising therapeutic target for the treatment of CS-induced pulmonary inflammation.


Asunto(s)
Fumar Cigarrillos/efectos adversos , Pulmón/metabolismo , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuropéptidos/metabolismo , Neumonía/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Fumar Cigarrillos/genética , Fumar Cigarrillos/metabolismo , Citocinas/genética , Citocinas/metabolismo , Inflamación/etiología , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Pulmón/patología , Ratones , Proteína Quinasa 3 Activada por Mitógenos/genética , Neuropéptidos/genética , Infiltración Neutrófila/genética , Neutrófilos/metabolismo , Neutrófilos/patología , Neumonía/etiología , Neumonía/genética , Neumonía/patología , Factor de Transcripción STAT3/genética , Proteína de Unión al GTP rac1/genética
5.
Eur J Pharmacol ; 775: 138-48, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26872986

RESUMEN

Ambroxol, a metabolite of bromhexine, is shown to exert several pharmacological activities, including secretolytic, anti-inflammatory and antioxidant actions. Oral and intravenous administration of ambroxol is useful for the airway inflammatory diseases. However, little is known about its potential in inhalation therapy for lipopolysaccharide (LPS)-induced mucous hypersecretion and inflammatory response. In the present study, we compared the pharmacological effects of ambroxol by inhalation with intravenous administration and preliminarily explored its mechanism of action. Our results demonstrated that ambroxol administered by inhalation inhibited MUC5AC expression, reduced glycosaminoglycan levels, enhanced the function of mucociliary clearance and promoted sputum excretion, suggesting that ambroxol increases expectoration of sputum by reducing its viscosity. Moreover, ambroxol significantly alleviated LPS-induced the influx of inflammatory cells and the extracellular signal-regulated kinase 1/2 (Erk 1/2) expression in lung tissues, and inhibited increases in the mRNA expression of the pro-inflammatory cytokines tumor necrosis factor (TNF)-α, CCL-2 (monocyte chemotactic protein-1), KC (keratinocyte cell protein) and interleukin (IL)-1ß in lung tissues. The secretolytic and anti-inflammatory effects of inhaled ambroxol at a dose of 7.5 mg/ml was comparable to that of ambroxol at 20 mg/ml i.v. and dexamethasone at 0.5 mg/kg i.p. In addition, we found that ambroxol dose-dependently inhibited LPS-induced increases in the mRNA expression of MUC5AC, TNF-α, and IL-1ß in human bronchial epithelial cell (NCI-H292) by inhibiting the Erk signaling pathway. These results demonstrate the beneficial effects of ambroxol in inhalation therapy for the airway inflammatory diseases.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Ambroxol , Antiinflamatorios , Expectorantes , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Administración por Inhalación , Ambroxol/administración & dosificación , Ambroxol/farmacología , Ambroxol/uso terapéutico , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Línea Celular , Citocinas/genética , Expectorantes/administración & dosificación , Expectorantes/farmacología , Expectorantes/uso terapéutico , Femenino , Glicosaminoglicanos/metabolismo , Humanos , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Ratones Endogámicos ICR , Mucina 5AC/genética , Depuración Mucociliar/efectos de los fármacos , Moco/metabolismo , ARN Mensajero/metabolismo
6.
PLoS One ; 10(6): e0128278, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26035589

RESUMEN

Cytochrome P-450 epoxygenase (EPOX)-derived epoxyeicosatrienoic acids (EETs), 5-lipoxygenase (5-LO), and leukotriene B4 (LTB4), the product of 5-LO, all play a pivotal role in the vascular inflammatory process. We have previously shown that EETs can alleviate oxidized low-density lipoprotein (ox-LDL)-induced endothelial inflammation in primary rat pulmonary artery endothelial cells (RPAECs). Here, we investigated whether ox-LDL can promote LTB4 production through the 5-LO pathway. We further explored how exogenous EETs influence ox-LDL-induced LTB4 production and activity. We found that treatment with ox-LDL increased the production of LTB4 and further led to the expression and release of both monocyte chemoattractant protein-1 (MCP-1/CCL2) and intercellular adhesion molecule-1 (ICAM-1). All of the above ox-LDL-induced changes were attenuated by the presence of 11,12-EET and 14,15-EET, as these molecules inhibited the 5-LO pathway. Furthermore, the LTB4 receptor 1 (BLT1 receptor) antagonist U75302 attenuated ox-LDL-induced ICAM-1 and MCP-1/CCL2 expression and production, whereas LY255283, a LTB4 receptor 2 (BLT2 receptor) antagonist, produced no such effects. Moreover, in RPAECs, we demonstrated that the increased expression of 5-LO and BLT1 following ox-LDL treatment resulted from the activation of nuclear factor-κB (NF-κB) via the p38 mitogen-activated protein kinase (MAPK) pathway. Our results indicated that EETs suppress ox-LDL-induced LTB4 production and subsequent inflammatory responses by downregulating the 5-LO/BLT1 receptor pathway, in which p38 MAPK phosphorylation activates NF-κB. These results suggest that the metabolism of arachidonic acid via the 5-LO and EPOX pathways may present a mutual constraint on the physiological regulation of vascular endothelial cells.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Araquidonato 5-Lipooxigenasa/química , Leucotrieno B4/metabolismo , Lipoproteínas LDL/farmacología , Arteria Pulmonar/metabolismo , Receptores de Leucotrieno B4/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Western Blotting , Células Cultivadas , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Masculino , Fosforilación/efectos de los fármacos , Arteria Pulmonar/citología , Arteria Pulmonar/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Leucotrieno B4/genética , Receptores de Leucotrieno B4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vasodilatadores/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
Drug Dev Res ; 76(3): 123-31, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25958838

RESUMEN

Bencycloquidium bromide (BCQB), a novel M3 receptor antagonist, alleviates airway hyperresponsiveness, inflammation, and airway remodeling in a murine model of asthma. The aim of this study was to investigate the anti-inflammatory activity of inhaled BCQB in a cigarette smoke (CS)-induced model of acute lung inflammation. Mice exposed to CS developed chronic obstructive pulmonary disease (COPD). Inhalation of BCQB suppressed the accumulation of neutrophils and macrophages in airways and lung and also inhibited the CS-induced increases in mRNA levels of keratinocyte-derived chemokine, monocyte chemotactic protein-1, tumor necrosis factor-alpha, and interleukin-1ß in lung and protein expression levels in bronchoalveolar lavage fluid. Moreover, BCQB (300 µg/ml) inhibited the CS-induced changes in superoxide dismutase and myeloperoxidase activities in the lungs. Our study suggests that BCQB might be a potential therapy for inflammation in CS-induced pulmonary diseases, including COPD.


Asunto(s)
Compuestos Bicíclicos Heterocíclicos con Puentes/administración & dosificación , Modelos Animales de Enfermedad , Nicotiana/efectos adversos , Neumonía/tratamiento farmacológico , Receptor Muscarínico M3/antagonistas & inhibidores , Humo/efectos adversos , Administración por Inhalación , Animales , Relación Dosis-Respuesta a Droga , Femenino , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos ICR , Neumonía/metabolismo , Neumonía/patología , Fumar/efectos adversos , Fumar/metabolismo , Fumar/patología , Resultado del Tratamiento
8.
Biomed Res Int ; 2015: 786104, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26000303

RESUMEN

We assessed the effect of a novel and selective phosphodiesterase 4 (PDE4) inhibitor, ciclamilast, on chronic inflammation in adjuvant-induced arthritis (AIA), a rat model of rheumatoid arthritis (RA), and acute inflammation in the rat and mouse model of carrageenan-induced paw edema and peritonitis. Our results showed that daily oral administration of ciclamilast at 1, 3, and 10 mg/kg dose-dependently inhibited the increase in hind paw volume of rats with AIA. The inhibition of paw edema was associated with inhibition of both the production of cytokines such as TNF-α, IL-1ß, and IL-6 and cell infiltration assessed in subcutaneous paw tissue. Moreover, there was significantly less tissue destruction in the ciclamilast-treated rats compared to the vehicle-treated rats, as assessed by radiographic analysis and histopathological evaluation. In the two acute inflammation models, ciclamilast inhibited carrageenan-induced paw edema in rats and inflammatory cell migration into the peritoneal cavity in mice in a dose-dependent manner. These results not only suggest that ciclamilast, as a disease-modifying antirheumatic drug (DMARD), can attenuate RA but also provide proof of principle that a PDE4 inhibitor may be useful for the treatment of arthritis.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Benzamidas/uso terapéutico , Piridinas/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Artritis Experimental/complicaciones , Artritis Experimental/diagnóstico por imagen , Artritis Experimental/patología , Benzamidas/farmacología , Peso Corporal/efectos de los fármacos , Carragenina , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos ICR , Tamaño de los Órganos/efectos de los fármacos , Peritonitis/inducido químicamente , Peritonitis/complicaciones , Peritonitis/tratamiento farmacológico , Peritonitis/patología , Piridinas/farmacología , Radiografía , Ratas Sprague-Dawley , Tejido Subcutáneo/efectos de los fármacos
9.
AAPS J ; 17(3): 711-22, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25762447

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a fatal lung disease. The current standard treatment with glucocorticoids (GCs) leads to many adverse effects, and its effectiveness is questionable. Thus, it is critical and urgent to find new drug(s) for treatment of IPF. Baicalin (BAI) is an attractive candidate for this purpose. Herein, utilizing shotgun lipidomics, we revealed that IPF could lead to a lipid disorder of the liver in an animal model induced by bleomycin and confirmed through histopathological studies of the lung. Lipidomics further demonstrated that this disorder could virtually be corrected after treatment with BAI, but not with dexamethasone (DEX) (a commonly used GC for treatment of IPF). In contrast, the treatment with DEX did not improve IPF but led to tremendous alterations in hepatic lipidomes and accumulation of fat in the liver, which was very different from the lipid disorder induced by IPF. The underpinning mechanisms of the IPF-resultant lipid disorder and DEX-induced lipotoxicity as revealed by shotgun lipidomics were extensively discussed. Taken together, the current study showed that IPF could lead to hepatic lipid disorder, which can be treated with BAI, and demonstrated that lipidomics could be a powerful tool for drug screening.


Asunto(s)
Hígado Graso/tratamiento farmacológico , Flavonoides/farmacología , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Hepatopatías/tratamiento farmacológico , Animales , Dexametasona/farmacología , Dexametasona/toxicidad , Hígado Graso/etiología , Hígado Graso/patología , Glucocorticoides/farmacología , Glucocorticoides/toxicidad , Fibrosis Pulmonar Idiopática/complicaciones , Lípidos/química , Hepatopatías/etiología , Hepatopatías/patología , Masculino , Ratones , Ratones Endogámicos ICR
10.
Toxicol Mech Methods ; 24(8): 575-83, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25134437

RESUMEN

Cigarette smoke contains reactive oxygen (ROS) that can cause oxidative stress. It increases the number of apoptotic and necrotic lung cells and further induces the development of chronic airway disease. In this study, we investigated the effects of cigarette smoke extract (CSE) on apoptosis in human bronchial epithelial cells (BEAS-2B). CSE exposure induced ROS generation and p38 mitogen-activated protein kinase (MAPK) activation that are associated with the activation of apoptosis-regulating signal kinase 1 (ASK-1). N-acetylcysteine (a general antioxidant) attenuated the CSE-induced ASK-1 and p38 MAPK activation and cell apoptosis, suggesting a triggering role of ROS in ASK-1/p38 MAPK activation during apoptotic progression. In contrast, the inhibition and knockdown of p38 attenuated the expression of anti-oxidant master NF-E2-related factor 2 (Nrf-2) and CSE-induced apoptosis, suggesting that p38 MAPK modulates Nrf-2 expression and presumably prevents cell apoptosis. Taken together, the data presented in this manuscript demonstrate that the ROS-dependent ASK-1/p38 signaling cascade regulates CSE-induced BEAS-2B cell apoptosis. In addition, anti-oxidative Nrf-2 is also up-regulated by the ROS/p38 signaling cascade in this progression.


Asunto(s)
Apoptosis/efectos de los fármacos , Bronquios/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Factor 2 Relacionado con NF-E2/agonistas , Mucosa Respiratoria/efectos de los fármacos , Fumar/efectos adversos , Regulación hacia Arriba/efectos de los fármacos , Acetilcisteína/farmacología , Elementos de Respuesta Antioxidante/efectos de los fármacos , Antioxidantes/farmacología , Bronquios/enzimología , Bronquios/metabolismo , Línea Celular , Mezclas Complejas/antagonistas & inhibidores , Mezclas Complejas/toxicidad , Activación Enzimática/efectos de los fármacos , Silenciador del Gen , Humanos , MAP Quinasa Quinasa Quinasa 5/antagonistas & inhibidores , MAP Quinasa Quinasa Quinasa 5/química , MAP Quinasa Quinasa Quinasa 5/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Factor 2 Relacionado con NF-E2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/enzimología , Mucosa Respiratoria/metabolismo , Humo , Productos de Tabaco , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/química , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Biochim Biophys Acta ; 1840(6): 1838-49, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24508121

RESUMEN

BACKGROUND: Epithelial-mesenchymal transition (EMT) is the major pathophysiological process in lung fibrosis observed in chronic obstructive pulmonary disease (COPD) and lung cancer. Smoking is a risk factor for developing EMT, yet the mechanism remains largely unknown. In this study, we investigated the role of Rac1 in cigarette smoke (CS) induced EMT. METHODS: EMT was induced in mice and pulmonary epithelial cells by exposure of CS and cigarette smoke extract (CSE) respectively. RESULTS: Treatment of pulmonary epithelial cells with CSE elevated Rac1 expression associated with increased TGF-ß1 release. Blocking TGF-ß pathway restrained CSE-induced changes in EMT-related markers. Pharmacological inhibition or knockdown of Rac1 decreased the CSE exposure induced TGF-ß1 release and ameliorated CSE-induced EMT. In CS-exposed mice, pharmacological inhibition of Rac1 reduced TGF-ß1 release and prevented aberrations in expression of EMT markers, suggesting that Rac1 is a critical signaling molecule for induction of CS-stimulated EMT. Furthermore, Rac1 inhibition or knockdown abrogated CSE-induced Smad2 and Akt (PKB, protein kinase B) activation in pulmonary epithelial cells. Inhibition of Smad2, PI3K (phosphatidylinositol 3-kinase) or Akt suppressed CSE-induced changes in epithelial and mesenchymal marker expression. CONCLUSIONS AND GENERAL SIGNIFICANCE: Altogether, these data suggest that CS initiates EMT through Rac1/Smad2 and Rac1/PI3K/Akt signaling pathway. Our data provide new insights into the fundamental basis of EMT and suggest a possible new course of therapy for COPD and lung cancer.


Asunto(s)
Transición Epitelial-Mesenquimal , Neuropéptidos/fisiología , Nicotiana/efectos adversos , Alveolos Pulmonares/patología , Humo/efectos adversos , Proteína de Unión al GTP rac1/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteína Smad2/fisiología , Factor de Crecimiento Transformador beta1/análisis , Factor de Crecimiento Transformador beta1/biosíntesis
12.
Eur J Pharmacol ; 727: 43-51, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24486707

RESUMEN

Oxidized low-density lipoprotein (Ox-LDL) is associated with atherosclerotic events through the modulation of arachidonic acid (AA) metabolism and activation of inflammatory signaling. Cytochrome P450 (CYP) epoxygenase-derived epoxyeicosatrienoic acids (EETs) mitigate inflammation through nuclear factor-κB (NF-κB). In this study, we explored the effects and mechanisms of exogenous EETs on the ox-LDL-induced inflammation of pulmonary artery endothelial cells (PAECs), which were cultured from rat pulmonary arteries. We determined that pre-treatment with 11,12-EET or 14,15-EET attenuated the ox-LDL-induced expression and release of intercellular adhesion molecule-1 (ICAM-1), E-selectin, and monocyte chemoattractant protein-1 (MCP-1) in a concentration-dependent manner. In addition, the ox-LDL-induced expression of CYP2J4 was upregulated by 11,12-EET and 14,15-EET (1µM). Furthermore, the endothelial receptor of lectin-like oxidized low-density lipoprotein (LOX-1) was downregulated in PAECs treated with EETs. The inflammatory responses evoked by ox-LDL (100µg/mL) were blocked by pharmacological inhibitors of Erk1/2 mitogen-activated protein kinase (MAPK) (U0126), p38 MAPK (SB203580), and NF-κB (PDTC). In addition, we confirmed that 11,12-EET suppresses phosphorylation of p38, degradation of IκBα, and activation of NF-κB (p65), whereas 14,15-EET can significantly suppress the phosphorylation of p38 and Erk1/2. Our results indicate that EETs exert beneficial effects on ox-LDL-induced inflammation primarily through the inhibition of LOX-1 receptor upregulation, MAPK phosphorylation, and NF-κB activation and through the upregulation of CYP2J4 expression. This study helps focus the current understanding of the contribution of EETs to the regulation of the inflammation of pulmonary vascular endothelial cells. Furthermore, the therapeutic potential of targeting the EET pathway in pulmonary vascular disease will be highlighted.


Asunto(s)
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Antiinflamatorios/farmacología , Células Endoteliales/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Inflamación/prevención & control , Lipoproteínas LDL/metabolismo , Arteria Pulmonar/efectos de los fármacos , Receptores Depuradores de Clase E/efectos de los fármacos , Ácido 8,11,14-Eicosatrienoico/farmacología , Animales , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Familia 2 del Citocromo P450 , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Activación Enzimática , Inflamación/genética , Inflamación/metabolismo , Masculino , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , Arteria Pulmonar/metabolismo , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptores Depuradores de Clase E/metabolismo , Transducción de Señal/efectos de los fármacos
13.
BMC Complement Altern Med ; 13: 86, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23587180

RESUMEN

BACKGROUND: Neurotoxin-Nna (NT), an analgesic peptide separated from the venom of Naja naja atra, has reported to have an exceptional specificity to block transmission of the nerve impulse by binding to the α- subunit of the nicotinic acetylcholine receptor in the membrane. However, little information is available on the anti-inflammatory effects of NT. Therefore, the anti-inflammatory activity of Neurotoxin-Nna was investigated in this study. METHODS: The anti-inflammatory effects of NT were evaluated by measuring its influence on several crucial factors in inflammatory pathways, including total antioxidant activity, antinociceptive effects in vivo, nuclear factor kappa B (NF-κB), polymorphonuclear cells (PMN), inducible nitric oxide synthase (iNOS), adhesion molecule (ICAM-1) and tactile hyperalgesia. RESULTS: NT treatment decreased the levels of tumor necrosis factor alpha (TNF-α) and interleukin 1 beta (IL-1ß). NT treatment decreased the total antioxidant status (TAOS) and reduced CFA-induced tactile hyperalgesia in a dose-dependent manner. NT significantly inhibited regulation of NF-kappaB activation and the production of IL-1ß, TNF-α, iNOS and CAM-1. Moreover, NT suppressed infiltration of PMN. CONCLUSIONS: Our results showed that NT reduced CFA-induced tactile hyperalgesia through inhibition inflammatory pathways in experimental inflammatory rats.


Asunto(s)
Antiinflamatorios/administración & dosificación , Proteínas Neurotóxicas de Elápidos/administración & dosificación , Venenos Elapídicos/química , Elapidae , Hiperalgesia/tratamiento farmacológico , Péptidos/administración & dosificación , Animales , Antiinflamatorios/química , Antiinflamatorios/aislamiento & purificación , Proteínas Neurotóxicas de Elápidos/química , Proteínas Neurotóxicas de Elápidos/aislamiento & purificación , Femenino , Humanos , Hiperalgesia/genética , Hiperalgesia/inmunología , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Masculino , FN-kappa B/genética , FN-kappa B/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Péptidos/química , Péptidos/aislamiento & purificación , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
Artículo en Inglés | MEDLINE | ID: mdl-24454488

RESUMEN

Caudatin 3-O-ß-D-cymaropyranosyl-(1 → 4)-ß-D-oleandropyranosyl-(1 → 4)-ß-D-cymaropyranosyl-(1 → 4)-ß-D-cymaropyranoside (CGII) is one of the C21-steroidal glycosides isolated from the roots of Cynanchum auriculatum ROYLE ex WIGHT. This study aimed to determine the cell growth, cell proliferation, and apoptotic cell death of human gastric cancer cells after CGII treatment. MTT assay was used to determine cell growth; fluorescence-activated cell sorting analysis was used to evaluate cell cycle distribution and apoptotic cell death. Immunoblotting was applied for measuring the expression of proteins involved in the cell cycle progression. The activities of caspase-3, -8, and -9 were detected by colorimetric caspase activity assays. CGII inhibited cell growth of human gastric cancer SGC-7901 cells in a concentration- and time-dependent manner. Treatment of SGC-7901 cells with CGII resulted in G1 phase cell cycle arrest, accompanied with decreased expression of cyclin D1 and cyclin-dependent kinases 4 and 6. CGII induced cell apoptosis and activated caspase-3, caspase-8, and caspase-9. In contrast, pan-caspase inhibitor z-VAD-fmk partially abolished the CGII-induced growth inhibition of SGC-7901 cells. In conclusion, CGII inhibits cell growth of human gastric cancer cells by inducing G1 phase cell cycle arrest and caspase-dependent apoptosis cascades.

15.
Drug Dev Ind Pharm ; 38(1): 123-8, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21721852

RESUMEN

BACKGROUND: Neurotoxin-II (NT-II), an analgesic peptide which was separated from the venom of Naja naja atra, is endowed an exceptional specificity of action that block transmission of the nerve impulse by binding to the acetylcholine receptor in the membrane. However, it has limited permeability across the blood-brain barrier (BBB) after intravenously (i.v.) injection. METHODS: In this study, we explored the potential application of nanoparticles overcoated with polysorbate 80 (P-80-NP) as drug carrier system for the nasal delivery of NT and the antinociceptive properties of NT-loaded P-80-NP (NT-P-NP) were also evaluated. RESULTS: The brain delivery of NT-II could be enhanced with nanoparticles coated with polysorbate-80 through intranasally (i.n.) administration. Compared with NT-II solution, NT-P-NP exhibited sustained release in vitro and higher concentrations of NT-II in the brain. The antinociceptive animal testing also revealed that intranasal delivery of NT-loaded nanoparticle coated with polysorbate-80 were able to promote better biodistribution of the drug into the brain. CONCLUSION: The nanoparticles overcoated with polysorbate-80 were capable of transporting the loaded drug across the BBB after intranasal administration.


Asunto(s)
Analgésicos/administración & dosificación , Analgésicos/química , Portadores de Fármacos/química , Nanopartículas/administración & dosificación , Nanopartículas/química , Neurotoxinas/administración & dosificación , Neurotoxinas/química , Administración Intranasal , Analgésicos/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Excipientes/química , Femenino , Masculino , Ratones , Tamaño de la Partícula , Permeabilidad , Polisorbatos/química
16.
Peptides ; 32(7): 1526-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21600257

RESUMEN

Neurotoxin-1 (NT) is an analgesic peptide which is endowed an exceptional specificity of action that blocks transmission of the nerve impulse. The aim of this study was to evaluate the potential application of nanoparticles technology as drug carrier system for the nasal delivery of NT. Mice were administered intranasally (i.n.) with NT (NT-P-NP), free NT solution (F-NT) and intravenously (i.v.) with NT (IV-NT) respectively. The NT levels in animal brain and antinociceptive activity of NT were analyzed. The result on brain transport showed that nanoparticles could exert enhanced delivery of NT into the brain significantly after i.n. administration. The results of antinociceptive activity showed that NT-P-NP increased immobility in the open-field test, both phases of formalin test were significantly inhibited by the NT-P-NP and NT-P-NP significantly inhibited the reaction time to thermal stimuli at 60 and 90 min. Both NT-P-NP and IV-NT were able to inhibit constrictions in acetic acid-induced writhing reaction. These data suggest that NT-loaded nanoparticles coated with polysorbate-80 could generate a significant improvement of drug levels in the brain. Intranasal administration of Neurotoxin-1 entrapped in nanoparticles coated with polysorbate-80 is an attractive alternative to intravenous administration.


Asunto(s)
Administración Intranasal , Analgésicos , Encéfalo/efectos de los fármacos , Portadores de Fármacos , Neurotoxinas , Dolor/tratamiento farmacológico , Analgésicos/química , Analgésicos/farmacología , Animales , Encéfalo/fisiología , Materiales Biocompatibles Revestidos/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacología , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Infusiones Intravenosas , Ratones , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Nanopartículas/química , Neurotoxinas/química , Neurotoxinas/farmacología , Dolor/fisiopatología , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/psicología , Percepción del Dolor/efectos de los fármacos , Percepción del Dolor/fisiología , Polisorbatos/química
17.
Respir Res ; 11: 39, 2010 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-20403205

RESUMEN

BACKGROUND: Basic and clinical studies suggest that hypothalamic-pituitary-adrenal (HPA) axis is the neuroendocrine-immune pathway that functionally regulates the chronic inflammatory disease including asthma. Our previous studies showed corresponding changes of cytokines and leukotriene B4 (LTB4) between brain and lung tissues in antigen-challenged asthmatic rats. Here, we investigated how the increased LTB4 level in brain interacts with HPA axis in regulating antigen-induced asthmatic response in sensitized rats. METHODS: Ovalbumin-sensitized rats were challenged by inhalation of antigen. Rats received vehicle, LTB4 or U75302 (a selective LTB4 BLT1 receptor inhibitor) was given via intracerebroventricular injection (i.c.v) 30 min before challenge. Lung resistance (RL) and dynamic lung compliance (Cdyn) were measured before and after antigen challenge. Inflammatory response in lung tissue was assessed 24 h after challenge. Expression of CRH mRNA and protein in hypothalamus were evaluated by RT-PCR and Western Blot, and plasma levels of adrenocorticotropic hormone (ACTH) and corticosterone (CORT) were measured using the ELISA kits. RESULTS: Antigen challenge decreased pulmonary function and induced airway inflammation, evoked HPA axis response in sensitized rats. Administration of LTB4 via i.c.v markedly attenuated airway contraction and inflammation. Meanwhile, LTB4 via i.c.v markedly increased CORT and ACTH level in plasma before antigen challenge, and followed by further increases in CORT and ACTH levels in plasma after antigen challenge in sensitized rats. Expression of CRH mRNA and protein in hypothalamus were also significantly increased by LTB4 via i.c.v in sensitized rats after antigen challenge. These effect were completely blocked by pre-treatment with BLT1 receptor antagonist U75302 (10 ng), but not by BLT2 antagonist LY255283. CONCLUSIONS: LTB4 administered via i.c.v down-regulates the airway contraction response and inflammation through activation of the HPA axis via its BLT1 receptor. This study expands our concept of the regulatory role of intracranial inflammatory mediators in inflammatory diseases including asthma. The favourable effects of LTB4 on the HPA axis may help to explain the phenomenon of self-relief after an asthmatic attack.


Asunto(s)
Asma/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Leucotrieno B4/metabolismo , Pulmón/metabolismo , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Leucotrieno B4/metabolismo , Hormona Adrenocorticotrópica/sangre , Resistencia de las Vías Respiratorias , Animales , Asma/inmunología , Asma/fisiopatología , Western Blotting , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Alcoholes Grasos/administración & dosificación , Femenino , Glicoles/administración & dosificación , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipotálamo-Hipofisario/inmunología , Hipotálamo/inmunología , Hipotálamo/metabolismo , Mediadores de Inflamación/metabolismo , Inyecciones Intraventriculares , Leucotrieno B4/administración & dosificación , Pulmón/inmunología , Pulmón/fisiopatología , Rendimiento Pulmonar , Masculino , Ovalbúmina , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/inmunología , Eosinofilia Pulmonar/inmunología , Eosinofilia Pulmonar/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Leucotrieno B4/agonistas , Receptores de Leucotrieno B4/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Zhonghua Zhong Liu Za Zhi ; 31(2): 90-4, 2009 Feb.
Artículo en Chino | MEDLINE | ID: mdl-19538881

RESUMEN

OBJECTIVE: To investigate the inhibitory effects of an antisense PC cell derived growth factor (PCDGF) vector on proliferation and invasion of highly malignant ovarian cancer cell lines Sw626 and A2780 cells, and preliminarily explore the related mechanisms. METHODS: MTT assay and Boyden chamber in vitro invasion assay were employed to detect the changes of proliferation and invasion ability in the Sw626 and A2780 cells transfected with anti-sense PCDGF. The expression levels of cyclin D1 and CDK4 proteins before and after transfection were detected by Western blotting. The effects on the expression and activity of MMP-2 were evaluated by quantitative RT-PCR and zymography, respectively. RESULTS: Comparing with the blank group, the proliferation inhibition rate of the Sw626 and A2780 cells transfected with anti-sense PCDGF was 72.9% and 70.9%, respectively, and the invasion ability was inhibited by 62.9% and 59.0%, respectively. The levels of cyclin D1 and CDK4 protein expression in antisense PCDGF transfected cells were 0.38 +/- 0.08 and 0.37 +/- 0.13, respectively, all significantly lower than 0.84 +/- 0.11 and 0.64 +/- 0.11, respectively, in the blank group (P < 0.01). The MMP-2 mRNA expression level in antisense PCDGF transfected cell group was 0.66 +/- 0.11, not significantly decreased in comparison with 0.89 +/- 0.09 in the blank group (P > 0.05), but the activity of MMP-2 was inhibited significantly. CONCLUSION: The antisense PCDGF vector may inhibit markedly the proliferation and invasion of highly malignant ovarian cancer cells, and partially reverses their malignant phenotype. It seems to be related with down-regulating the expression of cyclin D1 and CDK4 and inhibiting the activity of MMP-2. Our findings indicate that PCDGF may become a new target for antisense gene therapy of ovarian cancer.


Asunto(s)
Adhesión Celular , Proliferación Celular , ADN sin Sentido , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias Ováricas/patología , Línea Celular Tumoral , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo , Femenino , Vectores Genéticos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica , Neoplasias Ováricas/metabolismo , Progranulinas , ARN Mensajero/metabolismo , Transfección
19.
Int Immunopharmacol ; 8(12): 1639-45, 2008 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-18721904

RESUMEN

Clinically sublingual immunotherapy (SLIT) by using allergen extracts effectively alleviates the symptoms of allergic rhinitis and asthma. Supposed that oral administration of high-dose of allergen extracts imitates SLIT and may prevent IgE-related responses in allergic diseases, we investigated the effects of oral administration of allergen extracts from Dermatophagoides farinae (Derf) on allergen-induced inflammation and airway hyperresponsiveness (AHR) in a model of asthmatic rat. After administration to the specific Derf-sensitized rats with Derfdrop solution containing Derf1 and Derf2 extracts derived from Derf, the effects of Derfdrop on AHR, inflammatory cell accumulation, cytokine production in the bronchoalveolar lavage fluid and lung tissue, as well as serum IgE and IgG levels were investigated. Results indicated that Derfdrop not only dose-dependently prevented the AHR in response to methacholine, but also significantly reduced the serum total and allergen-specific IgE levels, all the maximal effects were achieved at dose of 5 mg/kg/d, and were as comparable as those of dexamethasone at dose of 1.0 mg/kg/d. Furthermore, oral administration of Derfdrop not only dose-dependently elevated allergen-specific serum IgG levels and reduced total and allergen-specific IgE levels, but also normalized the imbalance between the Th1 cytokine, IFN-gamma and Th2 cytokine, IL-4. Finally, oral administration of Derfdrop significantly reduced Goblet cell hyperplasia and eosinophilia in the Derf-sensitized allergic rat model. These data suggest that Derfdrop effectively improves specific allergen-induced inflammation and AHR in Derf-sensitized and -challenged rats and provide with the rationale for clinical SLIT by using Derfdrop in a specific allergen-induced asthma.


Asunto(s)
Asma/terapia , Hiperreactividad Bronquial/terapia , Dermatophagoides farinae/inmunología , Desensibilización Inmunológica , Administración Oral , Animales , Eosinofilia/prevención & control , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Interferón gamma/biosíntesis , Interleucina-4/biosíntesis , Ratas , Ratas Sprague-Dawley
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 37(4): 340-4, 2008 07.
Artículo en Chino | MEDLINE | ID: mdl-18705005

RESUMEN

OBJECTIVE: To determine the inhibitory effects of BIO-1211, a very late antigen-4 (vla-4) antagonist, on bronchoconstriction and neutrophil adhesion in rats. METHODS: For evaluating ovalbumin-induced bronchoconstriction in the sensitized rats, the changes in lung resistance (RL) and lung dynamic compliance (C(dyn)) were determined after antigen challenge. Neutrophils from the rats were used to determine fibronectin and serum-induced cell adhesion. The effect of BIO-1211 on wheezing was determined after inhalation of histamine and acetylcholine in guinea pigs. RESULT: BIO-1211 aerosol at 1, 3 and 10 mg/ml significantly inhibited the changes in lung resistance and lung dynamic compliance after antigen challenge in the sensitized rats in a dose-dependent manner. BIO-1211 at 25, 50, 100 and 200 microgram/ml inhibited the fibronectin-induced neutrophil adhesion by 23.5%, 24.6%, 61.4% and 58.1%, respectively, and serum-induced adhesion by 29.9%, 35.9%, 35.3% and 15.4%, respectively. Inhalation of 10 mg/ml BIO-1211 did not show any protection against histamine and acetylcholine-induced bronchoconstriction. CONCLUSION: BIO-1211 inhibits bronchoconstriction and neutrophil adhesion, which may be associated with its effect against bronchoconstriction in rats.


Asunto(s)
Asma/tratamiento farmacológico , Broncoconstricción/efectos de los fármacos , Broncodilatadores/farmacología , Neutrófilos/citología , Oligopéptidos/farmacología , Administración por Inhalación , Animales , Asma/fisiopatología , Broncoconstricción/fisiología , Broncodilatadores/administración & dosificación , Adhesión Celular/efectos de los fármacos , Femenino , Cobayas , Masculino , Neutrófilos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...