Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biochem Biophys Res Commun ; 525(2): 313-318, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32087973

RESUMEN

For achieving efficient cancer treatment, it is important to elucidate the mechanism responsible for the accumulation of nanoparticles in tumor tissue. Recent studies suggest that nanoparticles are not delivered merely through gaps between tumor endothelial cells. We previously reported that the maturation of the vascular structure by the vascular endothelial cell growth factor receptor 2 (VEGFR2) using a previously developed siRNA delivery technology (RGD-MEND) significantly enhanced the accumulation of nanoparticles in types of cancers that area vessel-rich (renal cell carcinoma). This result was completely inconsistent with the generally accepted theory of the enhanced permeability and retention (EPR) effect. We hypothesized that a caveolin-1 (Cav1)-mediated transcellular route would be involved with the penetration of nanoparticles into tumor vasculature. To reveal the exact mechanism responsible for this enhancement, we observed the delivery of long-circulating liposomes (LPs) after Cav1 was co-suppressed by RGD-MEND with VEGFR2. The enhanced delivery of LPs by siRNA against VEGFR2 (siVEGFR2) was accompanied by the elevated expression of the Cav1 protein. In addition, Cav1 knockdown by siRNA against Cav1 (siCav1) canceled the enhanced delivery of LPs by siVEGFR2. The injection of siCav1 had no effect on the formation of alpha smooth muscle actin or vascular endothelial cell adhesion molecules. These results suggest that a Cav1-induced transcellular route and not a paracellular route, at least partially, contributes to the accumulation of nanoparticles in tumors.


Asunto(s)
Caveolina 1/fisiología , Liposomas/metabolismo , Neoplasias/metabolismo , Transcitosis , Animales , Carcinoma de Células Renales/irrigación sanguínea , Carcinoma de Células Renales/metabolismo , Caveolina 1/efectos de los fármacos , Humanos , Nanopartículas/metabolismo , Neoplasias/irrigación sanguínea , ARN Interferente Pequeño/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
2.
Mol Cancer Res ; 18(4): 644-656, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31900313

RESUMEN

The antibody-drug conjugate trastuzumab-emtansine (T-DM1) offers an additional treatment option for patients with HER2-amplified tumors. However, primary and acquired resistance is a limiting factor in a significant subset of patients. Hypoxia, a hallmark of cancer, regulates the trafficking of several receptor proteins with potential implications for tumor targeting. Here, we have investigated how hypoxic conditions may regulate T-DM1 treatment efficacy in breast cancer. The therapeutic effect of T-DM1 and its metabolites was evaluated in conjunction with biochemical, flow cytometry, and high-resolution imaging studies to elucidate the functional and mechanistic aspects of hypoxic regulation. HER2 and caveolin-1 expression was investigated in a well-annotated breast cancer cohort. We find that hypoxia fosters relative resistance to T-DM1 in HER2+ cells (SKBR3 and BT474). This effect was not a result of deregulated HER2 expression or resistance to emtansine and its metabolites. Instead, we show that hypoxia-induced translocation of caveolin-1 from cytoplasmic vesicles to the plasma membrane contributes to deficient trastuzumab internalization and T-DM1 chemosensitivity. Caveolin-1 depletion mimicked the hypoxic situation, indicating that vesicular caveolin-1 is indispensable for trastuzumab uptake and T-DM1 cytotoxicity. In vitro studies suggested that HER2 and caveolin-1 are not coregulated, which was supported by IHC analysis in patient tumors. We find that phosphorylation-deficient caveolin-1 inhibits trastuzumab internalization and T-DM1 cytotoxicity, suggesting a specific role for caveolin-1 phosphorylation in HER2 trafficking. IMPLICATIONS: Together, our data for the first time identify hypoxic regulation of caveolin-1 as a resistance mechanism to T-DM1 with potential implications for individualized treatment of breast cancer.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Caveolina 1/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Maitansina/uso terapéutico , Trastuzumab/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Neoplasias de la Mama/patología , Femenino , Humanos , Maitansina/farmacología , Transfección , Trastuzumab/farmacología
3.
Zhonghua Jie He He Hu Xi Za Zhi ; 42(11): 845-851, 2019 Nov 12.
Artículo en Chino | MEDLINE | ID: mdl-31694095

RESUMEN

Objective: To explore the role of S100A8, the receptor for advanced glycation endproducts (RAGE) and Caveolin-1 in neutrophilic asthmatic rats, and to further study the intervention of roxithromycin and the possible mechanisms. Methods: Male Brown Norway rats were randomly assigned to a control group, an asthma group and a Roxithromycin group. The asthmatic rat model was established by intraperitoneal injection of ovalbumin (OVA) and Freund's complete adjuvant (FCA) mixture, and aerosol inhalation of OVA. Rats in the Roxithromycin group were given roxithromycin injection 30 mg/kg 30 minutes before each challenge. Rats in the control and the asthma groups were replaced with equal volumes of saline, respectively. Bronchoalveolar lavage fluid (BALF) neutrophil percentage (Neu%) and pathological changes of pulmonary tissue (hematoxylin-eosin, HE staining) were measured to confirm the establishment of asthmatic models. The concentration of inflammatory cytokines and S100A8 were quantified by enzyme-linked immunosorbent assay (ELISA), and the expression of Caveolin-1 and RAGE at protein levels were detected by immunohistochemistry and Western blot. Results: Neu% in BALF of the asthma group was significantly higher than those of the control group, and Neu% in the Roxithromycin group was lower than the asthma group (all P<0.01). Pulmonary histology revealed that there were a large number of inflammatory cells infiltrated in the bronchial and perivascular, pulmonary interstitial and alveolar spaces, and the bronchial wall and smooth muscles were thickened obviously in the asthma group. Rats in the Roxithromycin group showed milder inflammation and airway remodeling change than the asthma group. There was no obvious pathological damage in the control group. The concentration of IL-6 and IL-17 in BALF and serum of rats in the asthma group were significantly higher than those in the control group (P<0.01), and Roxithromycin inhibited the high expression of these cytokines (P<0.05). The expression of S100A8 and RAGE in the asthma group were significantly higher than those in the control group [(20.6±4.4) vs (7.1±2.0) ng/L; (885±118) vs (462±102) ng/L; (14.2±1.7) vs (7.6±1.8) ng/L; (774±166) vs (406±69) ng/L, all P<0.05], and Roxithromycin inhibited the high expression of these proteins [(14.3±3.7) vs (20.6±4.4) ng/L; (650±53) vs (885±118) ng/L; (10.4±1.2) vs (14.2±1.7) ng/L; (560±64) vs (728±72) ng/L] (all P<0.05). Meanwhile, the expression of Caveolin-1 in the asthma group was significantly lower than that in the control group (P<0.01), and Roxithromycin up-regulated its expression (P<0.01). Correlation analysis showed that there was a significantly positive correlation between the expression of S100A8 and RAGE (r=0.706, P<0.01), while there was a significantly negative correlation between the expression of S100A8 and Caveolin-1 (r=-0.775, P<0.01), and between the expression of Caveolin-1 and RAGE (r=-0.919, P<0.01). Conclusion: S100A8 and Caveolin-1 may play an important role in neutrophilic asthma via RAGE, and Roxithromycin may exerts anti-inflammatory effects and inhibition of airway remodeling partly through this signaling pathway.


Asunto(s)
Antibacterianos/farmacología , Asma/tratamiento farmacológico , Calgranulina A/efectos de los fármacos , Caveolina 1/efectos de los fármacos , Roxitromicina/farmacología , Remodelación de las Vías Aéreas (Respiratorias) , Animales , Antibacterianos/administración & dosificación , Western Blotting , Líquido del Lavado Bronquioalveolar , Calgranulina A/metabolismo , Caveolina 1/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Pulmón/fisiopatología , Masculino , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Ovalbúmina , Ratas , Receptor para Productos Finales de Glicación Avanzada , Roxitromicina/administración & dosificación
4.
Acta Biochim Pol ; 65(1): 111-118, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29549671

RESUMEN

This study was undertaken to verify whether simvastatin modulates Cav-1/eNOS expression, and if this modulation is associated with changes in pro- and anti-inflammatory cytokine and Toll-like receptor 4 (TLR4) level in abdominal aortic aneurysm (AAA). It is a 1:2 case-control study of non-statin (n=12) and simvastatin-treated patients (n=24) who underwent open AAA repair. Simvastatin treatment decreased Cav-1 (p<0.05) and increased eNOS expression (p<0.01) in the AAA wall. These changes might be dose dependent. The changes in Cav-1 and eNOS were associated with a trend towards decreased IL-6 and IL-17 concentration (p>0.05) and increased IL-10 concentration (p=0.055); however, TLR4 expression was unaffected, suggesting that simvastatin influences Cav-1 and eNOS in the AAA wall by other mechanisms. Simvastatin may modulate Cav-1 and eNOS expression in the aneurysmal wall, indicating a potentially beneficial role for statins in AAA patients.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Caveolina 1/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Simvastatina/uso terapéutico , Anciano , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Aneurisma de la Aorta Abdominal/cirugía , Estudios de Casos y Controles , Caveolina 1/efectos de los fármacos , Citocinas/efectos de los fármacos , Femenino , Humanos , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Masculino , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Simvastatina/farmacología , Receptor Toll-Like 4/efectos de los fármacos
5.
Life Sci ; 192: 270-277, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129771

RESUMEN

Tumor metastasis leads to a poor prognosis in breast cancer, yet the mechanisms remain unclear. Docosahexaenoic acid (DHA) extracted from Antarctic krill is an optical isomer of common DHA and has a much stronger anti-neoplastic effect. In this work, the migration and invasion abilities of MCF-7 cells treated with low concentrations of Antarctic krill DHA were evaluated. Low concentrations of Antarctic krill DHA significantly reduced the numbers of migrating and invasive MCF-7 cells, whereas the cell numbers decreased slowly in the CD95-silenced MCF-7 cells, which implies that CD95 might be involved in cell migration and invasion. Additionally, co-immunoprecipitation and Western blotting demonstrated that Antarctic krill DHA induced the accumulation of CD95 and caveolin-1 interaction, resulting in the down-regulation of MMP2 expression through the FAK/SRC/PI3K/AKT signaling pathway. In conclusion, Antarctic krill DHA enhanced the interaction between CD95 and caveolin-1, which may led to an inhibitory effect on cell migration and invasion via the FAK/SRC/PI3K/AKT signaling pathway. Our study indicates that Antarctic krill DHA has great potential for tumor therapy and has revealed a new metastatic mechanism mediated by the interaction of CD95 with caveolin-1.


Asunto(s)
Caveolina 1/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Euphausiacea/química , Invasividad Neoplásica , Receptor fas/efectos de los fármacos , Animales , Regiones Antárticas , Recuento de Células , Supervivencia Celular , Femenino , Humanos , Células MCF-7 , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metástasis de la Neoplasia , Transducción de Señal/efectos de los fármacos
6.
Psychopharmacology (Berl) ; 234(7): 1069-1077, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28233032

RESUMEN

OBJECTIVE: Here, we present the data indicating that chronic treatment with fluoxetine regulates Cav-1/PTEN/PI3K/AKT/GSK-3ß signalling pathway and glycogen content in primary cultures of astrocytes with bi-phasic concentration dependence. RESULTS: At lower concentrations, fluoxetine downregulates gene expression of Cav-1, decreases membrane content of PTEN, increases activity of PI3K/AKT, and elevates GSK-3ß phosphorylation thus suppressing its activity. At higher concentrations, fluoxetine acts in an inverse fashion. As expected, fluoxetine at lower concentrations increased while at higher concentrations decreased glycogen content in astrocytes. CONCLUSIONS: Our findings indicate that bi-phasic regulation of glycogen content via Cav-1/PTEN/PI3K/AKT/GSK-3ß pathway by fluoxetine may be responsible for both therapeutic and side effects of the drug.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Caveolina 1/efectos de los fármacos , Fluoxetina/farmacología , Glucógeno Sintasa Quinasa 3 beta/efectos de los fármacos , Glucógeno/metabolismo , Fosfohidrolasa PTEN/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Fosfatidilinositol 3-Quinasa Clase I , Técnicas de Silenciamiento del Gen , Ratones , Fosforilación , Cultivo Primario de Células , ARN Interferente Pequeño/farmacología
7.
Future Microbiol ; 11: 1227-1248, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26986451

RESUMEN

AIM: To investigate the detailed mechanism of Japanese encephalitis virus (JEV) cell entry. MATERIALS & METHODS: Utilize a siRNA library targeting cellular membrane trafficking genes to identify key molecules that mediate JEV entry into human neuronal cells. RESULTS: JEV enters human neuronal cells by caveolin-1-mediated endocytosis, which depends on a two-step regulation of actin cytoskeleton remodeling triggered by RhoA and Rac1: RhoA activation promoted the phosphorylation of caveolin-1, and then Rac1 activation facilitated caveolin-associated viral internalization. Specifically, virus attachment activates the EGFR-PI3K signaling pathway, thereby leading to RhoA activation. CONCLUSION: This work provides a detailed picture of the entry route and intricate cellular events following the entry of JEV into human neuronal cells, and promotes a better understanding of JEV entry.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/virología , Caveolina 1/metabolismo , Virus de la Encefalitis Japonesa (Especie)/metabolismo , Virus de la Encefalitis Japonesa (Especie)/fisiología , Internalización del Virus/efectos de los fármacos , Citoesqueleto de Actina/efectos de los fármacos , Factores Despolimerizantes de la Actina/metabolismo , Factores Despolimerizantes de la Actina/farmacología , Animales , Caveolina 1/efectos de los fármacos , Caveolina 1/genética , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Membrana Celular/virología , Colesterol/metabolismo , Cricetinae , Dinamina II/genética , Dinamina II/metabolismo , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/patogenicidad , Encefalitis Japonesa/virología , Endocitosis/fisiología , Receptores ErbB/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/fisiología , Humanos , Estadios del Ciclo de Vida/fisiología , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Neuronas/virología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas/farmacología , ARN Interferente Pequeño/genética , Transducción de Señal , Transfección , Acoplamiento Viral , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/farmacología , Proteína de Unión al GTP rhoA/metabolismo , Proteína de Unión al GTP rhoA/farmacología
8.
Am J Obstet Gynecol ; 211(5): 496.e1-496.e13, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24909339

RESUMEN

OBJECTIVE: Uterine leiomyomas, the most common neoplasms of the female genital tract, are benign tumors of the uterus arising from the smooth muscle cells (SMCs) of the myometrium with an involvement of estrogen. Caveolin-1 (Cav-1), a major protein component in caveolae membrane lipid rafts, is down-regulated in several estrogen-related cancer cells, and overexpression of Cav-1 inhibits proliferation of cancer cells and vascular SMCs as well. Therefore, we hypothesize that Cav-1 is down-regulated in human uterine leiomyoma. RESULTS: Western blot using tissues from clinical patients showed that Cav-1 expression was significantly lower or undetectable in uterine leiomyoma compared with their matched myometrium (P < .001). This finding was confirmed by immunohistochemistry and confocal microscopy. The cav-1 mRNA level in uterine leiomyomas was also significantly lower as detected by reverse transcription-quantitative polymerase chain reaction analysis (P = .001). To further study the underlying mechanism, we performed primary cell culture, and found that the expression of Cav-1 remained low in cultured leiomyoma SMCs (P = .009). Serum withdrawal did not change Cav-1 expression in leiomyoma SMCs, but increased expression in myometrial SMCs (P = .006). 17-ß estradiol inhibited the expression of Cav-1 protein (P = .047) and mRNA (P = .007) in leiomyoma SMCs, whereas it stimulated expression in myometrial SMCs (P = .043). 17-ß estradiol, although activating the mitogen-activated protein kinase pathway in both SMCs, did not stimulate their proliferation. CONCLUSION: We conclude that human uterine leiomyomas in vitro express low levels of Cav-1, which may result from estrogen inhibition. This effect of estrogen may contribute to the pathogenesis of uterine leiomyoma. Further studies in vivo are needed to verify these results.


Asunto(s)
Caveolina 1/genética , Leiomioma/genética , Miocitos del Músculo Liso/metabolismo , Miometrio/metabolismo , ARN Mensajero/genética , Neoplasias Uterinas/genética , Adulto , Western Blotting , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Células Cultivadas , Estradiol/farmacología , Estrógenos/farmacología , Femenino , Perfilación de la Expresión Génica , Humanos , Leiomioma/metabolismo , Persona de Mediana Edad , Miocitos del Músculo Liso/efectos de los fármacos , Miometrio/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Neoplasias Uterinas/metabolismo
9.
Asian Pac J Cancer Prev ; 14(9): 4983-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24175763

RESUMEN

OBJECTIVES: To establish a taxol-resistant cell line of human ovarian carcinoma (A2780/Taxol) and investigate its biological features. METHODS: The drug-resistant cell line (A2780/Taxol) was established by continuous stepwise selection with increasing concentrations of Taxol. Cell morphology was assessed by microscopy and growth curves were generated with in vitro and in vivo tumor xenograft models. With rhodamine123 (Rh123) assays, cell cycle distribution and the apoptotic rate were analyzed by flow cytometry (FCM). Drug resistance-related and signal associated proteins, including P-gp, MRPs, caveolin-1, PKC-α, Akt, ERK1/2, were detected by Western blotting. RESULTS: A2780/Taxol cells were established with stable resistance to taxol. The drug resistance index (RI) was 430.7. Cross-resistance to other drugs was also shown, but there was no significant change to radioresistance. Compared with parental cells, A2780/Taxol cells were significantly heteromorphous, with a significant delay in population doubling time and reduced uptake of Rh123 (p < 0.01). In vivo, tumor take by A2780 cells was 80%, and tumor volume increased gradually. In contrast, with A2780/Taxol cells in xenograft models there was no tumor development. FCM analysis revealed that A2780/Taxol cells had a higher percentage of G0/G1 and lower S phase, but no changes of G2 phase and the apoptosis rate. Expression of P-gp, MRP1, MRP2, BCRP, LRP, caveolin-1, PKC-α, Phospho-ERK1/2 and Phospho-JNK protein was significantly up-regulated, while Akt and p38 MARK protein expression was not changed in A2780/Taxol cells. CONCLUSION: The A2780/Taxol cell line is an ideal model to investigate the mechanism of muti-drug resistance related to overexpression of drug-resistance associated proteins and activation of the PKC-α/ERK (JNK) signaling pathway.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma/genética , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas/genética , Paclitaxel/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Carcinoma/metabolismo , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Múltiples Medicamentos/fisiología , Resistencia a Antineoplásicos/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/efectos de los fármacos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/metabolismo , Proteína Quinasa C-alfa/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Am J Respir Crit Care Med ; 188(7): 831-41, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23924348

RESUMEN

RATIONALE: Alveolar transforming growth factor (TGF)-ß1 signaling and expression of TGF-ß1 target genes are increased in patients with idiopathic pulmonary fibrosis (IPF) and in animal models of pulmonary fibrosis. Internalization and degradation of TGF-ß receptor TßRI inhibits TGF-ß signaling and could attenuate development of experimental lung fibrosis. OBJECTIVES: To demonstrate that after experimental lung injury, human syndecan-2 confers antifibrotic effects by inhibiting TGF-ß1 signaling in alveolar epithelial cells. METHODS: Microarray assays were performed to identify genes differentially expressed in alveolar macrophages of patients with IPF versus control subjects. Transgenic mice that constitutively overexpress human syndecan-2 in macrophages were developed to test the antifibrotic properties of syndecan-2. In vitro assays were performed to determine syndecan-2-dependent changes in epithelial cell TGF-ß1 signaling, TGF-ß1, and TßRI internalization and apoptosis. Wild-type mice were treated with recombinant human syndecan-2 during the fibrotic phase of bleomycin-induced lung injury. MEASUREMENTS AND MAIN RESULTS: We observed significant increases in alveolar macrophage syndecan-2 levels in patients with IPF. Macrophage-specific overexpression of human syndecan-2 in transgenic mice conferred antifibrotic effects after lung injury by inhibiting TGF-ß1 signaling and downstream expression of TGF-ß1 target genes, reducing extracellular matrix production and alveolar epithelial cell apoptosis. In vitro, syndecan-2 promoted caveolin-1-dependent internalization of TGF-ß1 and TßRI in alveolar epithelial cells, which inhibited TGF-ß1 signaling and epithelial cell apoptosis. Therapeutic administration of human syndecan-2 abrogated lung fibrosis in mice. CONCLUSIONS: Alveolar macrophage syndecan-2 exerts antifibrotic effects by promoting caveolin-1-dependent TGF-ß1 and TßRI internalization and inhibiting TGF-ß1 signaling in alveolar epithelial cells. Hence, molecules that facilitate TßRI degradation via endocytosis represent potential therapies for pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/inducido químicamente , Macrófagos Alveolares/efectos de los fármacos , Sindecano-2/uso terapéutico , Factor de Crecimiento Transformador beta1/efectos de los fármacos , Animales , Apoptosis , Bleomicina/administración & dosificación , Lavado Broncoalveolar , Caveolina 1/efectos de los fármacos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Marcadores Genéticos , Humanos , Hidroxiprolina/análisis , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/genética , Técnicas In Vitro , Ratones , Ratones Transgénicos , Transducción de Señal , Sindecano-2/fisiología , Análisis de Matrices Tisulares , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/fisiología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
11.
J Biomed Biotechnol ; 2011: 625380, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22162638

RESUMEN

We evaluated the in vitro effects of estradiol, progesterone, and testosterone on the molting process, which is the initial and crucial step in the development of the muscular larvae (ML or L1) to adult worm. Testosterone had no significative effect on the molting rate of the parasite, however, progesterone decreased the molting rate about a 50% in a concentration- and time-independent pattern, while estradiol had a slight effect (10%). The gene expression of caveolin-1, a specific gene used as a marker of parasite development, showed that progesterone and estradiol downregulated its expression, while protein expression was unaffected. By using flow citometry, a possible protein that is recognized by a commercial antiprogesterone receptor antibody was detected. These findings may have strong implications in the host-parasite coevolution, in the sex-associated susceptibility to this infection and could point out to possibilities to use antihormones to inhibit parasite development.


Asunto(s)
Expresión Génica/fisiología , Hormonas Esteroides Gonadales/metabolismo , Helmintiasis/parasitología , Interacciones Huésped-Parásitos/fisiología , Muda/fisiología , Receptores de Progesterona/metabolismo , Trichinella spiralis/crecimiento & desarrollo , Animales , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Estradiol/metabolismo , Estradiol/farmacología , Femenino , Expresión Génica/efectos de los fármacos , Hormonas Esteroides Gonadales/farmacología , Helmintiasis/tratamiento farmacológico , Humanos , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Muda/efectos de los fármacos , Progesterona/metabolismo , Progesterona/farmacología , Receptores de Progesterona/efectos de los fármacos , Testosterona/metabolismo , Testosterona/farmacología , Trichinella spiralis/efectos de los fármacos
12.
Vascul Pharmacol ; 52(5-6): 236-42, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20197113

RESUMEN

High soy (HS) diets are neuroprotective and promote vascular dilatation in the periphery. We hypothesized that an HS diet would promote vascular dilatation in the cerebrovasculature by mimicking estradiol's actions on the endothelial nitric oxide synthase (eNOS) system including increasing eNOS expression and decreasing caveolin-1 expression to increase nitric oxide (NO) production. Ovariectomized rats were fed HS or a soy-free diet (SF)+/-low physiological estradiol (E2) for 4weeks. Neither E2 nor HS altered middle cerebral artery (MCA) structure or vascular responses to acetylcholine, serotonin, or phenylephrine. Estradiol enhanced bradykinin-induced relaxation in an eNOS-dependent manner. Although E2 and HS increased eNOS mRNA expression in the brain and cerebrovasculature, they had no effect on eNOS protein expression or phosphorylation in the MCA. However, E2 decreased caveolin-1 protein in the MCA. In MCAs neither E2 nor HS altered estrogen receptor (ER) alpha expression, but E2 did reduce ER beta levels. These data suggest that HS diets have no effect on vascular NO production, and that E2 may modulate basal NO production by reducing the expression of caveolin-1, an allosteric inhibitor of NOS activity. However, the effects of E2 and HS on the cerebrovasculature are small and may not underlie their protective actions in pathological states.


Asunto(s)
Glycine max/química , Isoflavonas/farmacología , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Caveolina 1/efectos de los fármacos , Caveolina 1/genética , Dieta , Estradiol/farmacología , Receptor beta de Estrógeno/efectos de los fármacos , Receptor beta de Estrógeno/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Isoflavonas/administración & dosificación , Arteria Cerebral Media/efectos de los fármacos , Arteria Cerebral Media/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ovariectomía , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
13.
J Neurosci ; 30(4): 1417-25, 2010 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-20107068

RESUMEN

Increased brain expression of vascular endothelial growth factor (VEGF) is associated with neurological disease, brain injury, and blood-brain barrier (BBB) dysfunction. However, the specific effect of VEGF on the efflux transporter P-glycoprotein, a critical component of the BBB, is not known. Using isolated rat brain capillaries and in situ rat brain perfusion, we determined the effect of VEGF exposure on P-glycoprotein activity in vitro and in vivo. In isolated capillaries, VEGF acutely and reversibly decreased P-glycoprotein transport activity without decreasing transporter protein expression or opening tight junctions. This effect was blocked by inhibitors of the VEGF receptor flk-1 and Src kinase, but not by inhibitors of phosphatidylinositol-3-kinase or protein kinase C. VEGF also increased Tyr-14 phosphorylation of caveolin-1, and this was blocked by the Src inhibitor PP2. Pharmacological activation of Src kinase activity mimicked the effects of VEGF on P-glycoprotein activity and Tyr-14 phosphorylation of caveolin-1. In vivo, intracerebroventricular injection of VEGF increased brain distribution of P-glycoprotein substrates morphine and verapamil, but not the tight junction marker, sucrose; this effect was blocked by PP2. These findings indicate that VEGF decreases P-glycoprotein activity via activation of flk-1 and Src, and suggest Src-mediated phosphorylation of caveolin-1 may play a role in downregulation of P-glycoprotein activity. These findings also imply that P-glycoprotein activity is acutely diminished in pathological conditions associated with increased brain VEGF expression and that BBB VEGF/Src signaling could be targeted to acutely modulate P-glycoprotein activity and thus improve brain drug delivery.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/fisiología , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Arterias Cerebrales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Capilares/efectos de los fármacos , Capilares/metabolismo , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Arterias Cerebrales/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos/farmacología , Inyecciones Intraventriculares , Masculino , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Morfina/farmacocinética , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Verapamilo/farmacocinética , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
14.
Int J Oncol ; 35(5): 1037-43, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19787257

RESUMEN

Progression of colon cancer is associated with the up-regulation of cyclooxygenase-2 (COX-2) and hydroxymethyl glutaryl CoA reductase (HMG-R). Clinical and preclinical evidence shows that a combination of COX-2 and HMG-R inhibitors provide additive/synergistic chemopreventive effects against colorectal cancer. However, the mechanism by which statins and NSAIDs inhibit cancer growth is not yet fully understood. We aimed to identify critical molecules and signal pathways modulated by a combination of lovastatin and celecoxib in the human HCT-116 colon cancer cell line. HCT-116 cells were exposed to 50 microM celecoxib, 25 microM lovastatin or a combination of both to assess their effect in modulating caveolin-1 expression and its down-stream signaling pathways. Our results suggest that a combination of lovastatin and/or celecoxib suppressed caveolin-1 expression and membrane localization profoundly when compared to either agent alone. Lovastatin and/or celecoxib also inhibited caveolin-1-dependent cell survival signals mediated through Akt activation as well as its down-stream effectors such as phosphorylated ERK and STAT3 in HCT-116 cells. Treatment with lovastatin or celecoxib decreased the levels of cyclin D1, CDK2, pRb and E2F1, while the combination treatment showed more pronounced suppression. In addition, lovastatin and celecoxib also decreased the amount of cholesterol rich cytoplasmic lipid bodies (storehouses of esteridied arachidonates) by 80%, while the combination showed a complete inhibition. Overall, our data suggest that a combination of COX-2 and HMG-R inhibitors synergistically inhibits caveolin-1 and its associated signaling pathways.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Caveolina 1/efectos de los fármacos , Neoplasias del Colon/prevención & control , Lovastatina/administración & dosificación , Pirazoles/administración & dosificación , Transducción de Señal/efectos de los fármacos , Sulfonamidas/administración & dosificación , Apoptosis/efectos de los fármacos , Western Blotting , Caveolina 1/metabolismo , Celecoxib , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Sinergismo Farmacológico , Técnica del Anticuerpo Fluorescente , Humanos , Transducción de Señal/fisiología
15.
Hypertension ; 54(5): 1070-6, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19752292

RESUMEN

In moderate sodium-replete states, dopamine 1-like receptors (D1R/D5R) are responsible for regulating >50% of renal sodium excretion. This is partly mediated by internalization and inactivation of NaKATPase, when associated with adapter protein 2. We used dopaminergic stimulation via fenoldopam (D1-like receptor agonist) to study the interaction among D1-like receptors, caveolin-1 (CAV1), and the G protein-coupled receptor kinase type 4 in cultured human renal proximal tubule cells (RPTCs). We compared 2 groups of RPTCs, 1 of cell lines that were isolated from normal subjects (nRPTCs) and a second group of cell lines that have D1-like receptors that are uncoupled (uncoupled RPTCs) from adenylyl cyclase second messengers. In nRPTCs, fenoldopam increased the plasma membrane expression of D1R (10.0-fold) and CAV1 (1.3-fold) and markedly decreased G protein-coupled receptor kinase type 4 by 94+/-8%; no effects were seen in uncoupled RPTCs. Fenoldopam also increased the association of adapter protein 2 and NaKATPase by 53+/-9% in nRPTCs but not in uncoupled RPTCs. When CAV1 expression was reduced by 86.0+/-8.5% using small interfering RNA, restimulation of the D1-like receptors with fenoldopam in nRPTCs resulted in only a 7+/-9% increase in association between adapter protein 2 and NaKATPase. Basal CAV1 expression and association with G protein-coupled receptor kinase type 4 was decreased in uncoupled RPTCs (58+/-5% decrease in association) relative to nRPTCs. We conclude that the scaffolding protein CAV1 is necessary for the association of D1-like receptors with G protein-coupled receptor kinase type 4 and the adapter protein 2-associated reduction in plasma membrane NaKATPase.


Asunto(s)
Caveolina 1/metabolismo , Fenoldopam/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Receptores de Dopamina D1/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Análisis de Varianza , Caveolina 1/efectos de los fármacos , Células Cultivadas , AMP Cíclico/metabolismo , Ensayo de Inmunoadsorción Enzimática , Quinasa 4 del Receptor Acoplado a Proteína-G/análisis , Quinasa 4 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Túbulos Renales Proximales/citología , Microscopía Confocal , Probabilidad , Receptores de Dopamina D1/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/efectos de los fármacos
16.
J Neurochem ; 110(1): 194-207, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19457127

RESUMEN

Cellular prion protein (PrP(c)) is a ubiquitous glycoprotein, whose physiological role is poorly characterized. It has been suggested that PrP(c) participates in neuritogenesis, neuroprotection, copper metabolism, and signal transduction. In this study we detailed the intracellular events induced by PrP(c) antibody-mediated cross-linking in PC12 cells. We found a Fyn-dependent activation of the Ras-Raf pathway, which leads to a rapid and transient phosphorylation of extracellular regulated kinases. In addition, this activation cascade relies on the engagement of integrins, and involves focal adhesion kinase activation. We demonstrated the tyrosine phosphorylation of caveolin-1 as a consequence of PrP(c) stimulation, and showed that phosphocaveolin-1 scaffolds and coordinates protein complexes involved in PrP(c)-dependent signaling. Moreover, we found that caveolin-1 phosphorylation, is a mechanism for recruiting the C-terminal Src kinase and inactivating Fyn, so as to terminate cell signaling. Furthermore our data support a significant role for PrP(c) as a response mediator in neuritogenesis and cell differentiation.


Asunto(s)
Caveolina 1/metabolismo , Diferenciación Celular/fisiología , Neuritas/metabolismo , Neurogénesis/fisiología , Proteínas PrPC/metabolismo , Transducción de Señal/fisiología , Animales , Encéfalo/citología , Encéfalo/metabolismo , Proteína Tirosina Quinasa CSK , Caveolina 1/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Integrinas/metabolismo , Neuritas/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Células PC12 , Fosforilación/efectos de los fármacos , Proteínas PrPC/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Ratas , Sistemas de Mensajero Secundario/fisiología , Transducción de Señal/efectos de los fármacos , Quinasas raf/metabolismo , Proteínas ras/metabolismo , Familia-src Quinasas
17.
Cancer Detect Prev ; 32(4): 319-28, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19186008

RESUMEN

BACKGROUND: The purpose of our study was to show the apoptotic and anti-proliferative effects of phytosterols as distinct from cholesterol effects on prostate cancer cell lines, and also their differential expression of caveolin-1, and a prostate specific gene, PCGEM1. METHODS: PC-3 and DU145 cells were treated with sterols (cholesterol and phytosterols) for 48h, followed by trypan blue dye exclusion measurement of cytotoxicity and MTT cell proliferation assays, respectively. Cell cycle analysis was carried out microscopically, and by propidium iodide uptake using flow cytometry. Sterol induction of oncogenic gene expression was evaluated by RT-PCR. Apoptotic cells were identified by immunocytochemistry using DNA fragmentation method, and by annexin V adhesion using flow cytometry. RESULTS: Physiological doses (16microM) of these sterols were not cytotoxic in these cells. Cholesterol-enrichment promoted mitosis (54 and 61% by microscopy; 40.8 and 34.08% by FACS analysis in PC-3 and DU145, respectively) and cell growth (P<0.05), while phytosterols suppressed mitosis (29 and 35% by microscopy; 27.71 and 17.37% by FACS analysis in PC-3 and DU145, respectively), and significantly induced tumor-suppression (P<0.05) and apoptosis. We demonstrated for the first time that cholesterols upregulated the expression of PCGEM1 even in androgen-insensitive prostate cancer cell lines. Phytosterols reversed this effect, while upregulating the expression of caveolin-1, a known mediator of androgen-dependent proto-oncogene signals that presumably control growth and anti-apoptosis. CONCLUSIONS: Phytosterol inhibition of PCGEM1 and cell growth and the overexpression of caveolin-1, suggests that poor disease prognosis anchors on the ability of caveolin-1 to regulate downstream oncogene(s) and apoptosis genes. Sterol intake may contribute to the disparity in incidence of prostate cancer, and elucidation of the mechanism for modulation of growth and apoptosis signaling may reveal potential targets for cancer prevention and/or chemotherapeutic intervention. Sterol regulation of PCGEM1 expression suggests its potential as biomarker for prediction of neoplasms that would be responsive to chemoprevention by phytosterols.


Asunto(s)
Colesterol/farmacología , Fitosteroles/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , ARN no Traducido/efectos de los fármacos , Apoptosis/efectos de los fármacos , Caveolina 1/efectos de los fármacos , Caveolina 1/genética , Caveolina 1/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral/citología , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colesterol/metabolismo , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Fitosteroles/metabolismo , Proto-Oncogenes Mas , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo
18.
Free Radic Biol Med ; 45(7): 1027-34, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18694817

RESUMEN

Endothelial dysfunction and impaired angiogenesis constitute a hallmark of hypercholesterolemia. This study was designed to examine the effects of resveratrol, an antioxidant with lipid-lowering properties similar to those of statins, on neovascularization along with caveolar interaction with proangiogenic molecules in hypercholesterolemic rats. Animals were divided into: rats maintained on a normal diet (control group); rats maintained on a 5% high-cholesterol diet for 8 weeks (HC group); and rats maintained on a 5% high-cholesterol diet for 8 weeks and administered resveratrol (20 mg/kg) orally for 2 weeks (HCR group). Myocardial infarction was induced by ligating the left anterior descending artery. Herein we examined a novel method for stimulating myocardial angiogenesis by pharmacological preconditioning with resveratrol at both the capillary and arteriolar levels and the potential role of hemeoxygenase-1, endothelial nitric oxide synthase and caveolin-1 in mediating such a response. We also investigated the functional relevance of such treatment by assessing whether the induced neovascularization can help preserve left ventricle-contractile functional reserve in the setting of a chronic hypercholesterolemic condition. Four weeks after sham surgery and left anterior descending artery occlusion, rats underwent echocardiographic evaluation, which revealed improvement in ejection fraction and fractional shortening in the HCR group compared with the HC group. Left ventricular tissue sections displayed increased capillary and arteriolar density in the HCR group compared with the HC group. Western blot analysis revealed downregulation of vascular endothelial growth factor and hemeoxygenase-1 and increased association of caveolin-1 eNOS in the HC group, decreasing the availability of eNOS to the system; which was reversed with resveratrol treatment in the HCR group. This study was further validated in cardiac-specific hemeoxygenase-1-overexpressed mice assuming molecular cross-talk between the targets. Hence, our data identified potential regulators that primarily attenuate endothelial dysfunction by resveratrol therapy in hypercholesterolemic myocardium.


Asunto(s)
Antioxidantes/farmacología , Corazón/efectos de los fármacos , Hipercolesterolemia/tratamiento farmacológico , Neovascularización Fisiológica/efectos de los fármacos , Estilbenos/farmacología , Animales , Western Blotting , Caveolina 1/efectos de los fármacos , Caveolina 1/metabolismo , Circulación Coronaria/efectos de los fármacos , Hemo-Oxigenasa 1/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Hipercolesterolemia/fisiopatología , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Sprague-Dawley , Resveratrol , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Transplantation ; 85(9): 1311-7, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18475189

RESUMEN

BACKGROUND: BK virus (BKV), a human polyomavirus, causes BKV nephritis, which often leads to graft loss after renal transplantation. Currently, the only efficient therapy against BKV nephritis seems to be a reduction or change of immunosuppressive agents, but this may increase the inherent risk of rejection. Here, we report the ability of 3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitor (statin), which is routinely used to treat hypercholesterolemia, to repress BKV entry pathways in human renal proximal tubular epithelial cells (HRPTEC) and, correspondently, prevent BKV infection. METHODS: HRPTEC were co-incubated with BKV and pravastatin. Then the percentage of HRPTEC infected with BKV by immunofluorescent analysis and large T-antigen expression which suggested BKV infection by Western blots was assessed in the absence and presence of pravastatin. The distribution of purified and labeled BKV particles in the presence and absence of pravastatin was also investigated. RESULTS: Both the percentage of BKV infected cells and the large T-antigen expression were significantly decreased in HRPTEC pretreated and co-incubated with pravastatin. However, when pravastatin was added 72 hr after BKV infection it failed to decrease percentage of BKV infected cells. It is likely, that pravastatin's inhibitory effect is explained by depletion of caveolin-1, a critical element of caveolae. BKV enters HRPTEC by caveolar-mediated endocytosis. We provide evidence that pravastatin dramatically decreased caveolin-1 expression in HRPTEC and interfered with internalization of labeled BKV particles. CONCLUSIONS: Our data suggest that pravastatin, acting through depletion of caveolin-1, prevented caveolar-dependent BKV internalization and repressed BKV infection of HRPTEC.


Asunto(s)
Virus BK/efectos de los fármacos , Células Epiteliales/virología , Túbulos Renales Proximales/virología , Pravastatina/farmacología , Caveolina 1/antagonistas & inhibidores , Caveolina 1/efectos de los fármacos , Humanos , Infecciones por Polyomavirus/prevención & control , Infecciones Tumorales por Virus/prevención & control
20.
FASEB J ; 22(7): 2185-97, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18296501

RESUMEN

Epidemiological studies correlate moderate red wine consumption to reduced incidence of cardiovascular disease. Resveratrol is a polyphenolic compound in red wine that has cardioprotective effects in rodents. Although endothelial cell (EC) studies indicate that micromolar resveratrol has diverse biological activities, these concentrations are not physiologically relevant because human oral ingestion provides only brief exposure to nanomolar plasma levels. Previously, we reported that nanomolar resveratrol activated ERK1/2 signaling in bovine aortic ECs (BAECs). The goal of this study was to determine the mechanisms by which nanomolar resveratrol rapidly activates endothelial nitric oxide synthase (eNOS) in human umbilical vein ECs (HUVECs). We report for the first time that resveratrol increased interaction between estrogen receptor alpha (ER alpha), caveolin-1 (Cav-1) and c-Src, and increased phosphorylation of Cav-1, c-Src, and eNOS. Pretreatment with the lipid raft disruptor beta-methyl cyclodextrin or G alpha inhibitor pertussis toxin blocked resveratrol- and E(2)-induced eNOS activation and NO production. Depletion of endogenous ER alpha, not ERbeta, by siRNA attenuated resveratrol- and E(2)-induced ERK1/2, Src, and eNOS phosphorylation. Our data demonstrate that nanomolar resveratrol induces ER alpha-Cav-1-c-SRC interaction, resulting in NO production through a G alpha-protein-coupled mechanism. This study provides important new insights into mechanisms for the beneficial effects of resveratrol in ECs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Caveolina 1/fisiología , Endotelio Vascular/fisiología , Receptor alfa de Estrógeno/fisiología , Estilbenos/farmacología , Venas Umbilicales/fisiología , Familia-src Quinasas/fisiología , Caveolina 1/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Etanol/farmacología , Humanos , Cinética , Fosforilación , Resveratrol , Venas Umbilicales/efectos de los fármacos , Familia-src Quinasas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA