Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biomolecules ; 14(1)2024 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-38254705

RESUMEN

The low bioavailability of most phytochemicals limits their anticancer effects in humans. The present study was designed to test whether combining arctigenin (Arc), a lignan mainly from the seed of Arctium lappa, with green tea (GT) and quercetin (Q) enhances the chemopreventive effect on prostate cancer. We performed in vitro proliferation studies on different cell lines. We observed a strong synergistic anti-proliferative effect of GT+Q+Arc in exposing androgen-sensitive human prostate cancer LNCaP cells. The pre-malignant WPE1-NA22 cell line was more sensitive to this combination. No cytotoxicity was observed in normal prostate epithelial PrEC cells. For an in vivo study, 3-week-old, prostate-specific PTEN (phosphatase and tensin homolog) knockout mice were treated with GT+Q, Arc, GT+Q+Arc, or the control daily until 16 weeks of age. In vivo imaging using prostate-specific membrane antigen (PSMA) probes demonstrated that the prostate tumorigenesis was significantly inhibited by 40% (GT+Q), 60% (Arc at 30 mg/kg bw), and 90% (GT+Q+Arc) compared to the control. A pathological examination showed that all control mice developed invasive prostate adenocarcinoma. In contrast, the primary lesion in the GT+Q and Arc alone groups was high-grade prostatic intraepithelial neoplasia (PIN), with low-grade PIN in the GT+Q+Arc group. The combined effect of GT+Q+Arc was associated with an increased inhibition of the androgen receptor, the PI3K/Akt pathway, Ki67 expression, and angiogenesis. This study demonstrates that combining Arc with GT and Q was highly effective in prostate cancer chemoprevention. These results warrant clinical trials to confirm the efficacy of this combination in humans.


Asunto(s)
Furanos , Lignanos , Neoplasias de la Próstata , Animales , Masculino , Ratones , Quimioprevención , Lignanos/farmacología , Lignanos/uso terapéutico , Ratones Noqueados , Fosfatidilinositol 3-Quinasas , Próstata/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/prevención & control , Quercetina/farmacología , Quercetina/uso terapéutico , Tensinas , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo ,
2.
Sci Rep ; 11(1): 518, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436815

RESUMEN

Patients with chronic kidney disease (CKD) have elevated circulating levels of trimethylamine N-oxide (TMAO), a metabolite derived from gut microbes and associated with cardiovascular diseases. High circulating levels of TMAO and its dietary precursor, choline, predict increased risk for development of CKD in apparently healthy subjects, and studies in mice fed TMAO or choline suggest that TMAO can contribute to kidney impairment and renal fibrosis. Here we examined the interactions between TMAO, kidney disease, and cardiovascular disease in mouse models. We observed that while female hyperlipidemic apoE KO mice fed a 0.2% adenine diet for 14 weeks developed CKD with elevated plasma levels of TMAO, provision of a non-lethal inhibitor of gut microbial trimethylamine (TMA) production, iodomethylcholine (IMC), significantly reduced multiple markers of renal injury (plasma creatinine, cystatin C, FGF23, and TMAO), reduced histopathologic evidence of fibrosis, and markedly attenuated development of microalbuminuria. In addition, while the adenine-induced CKD model significantly increased heart weight, a surrogate marker for myocardial hypertrophy, this was largely prevented by IMC supplementation. Surprisingly, adenine feeding did not increase atherosclerosis and significantly decreased the expression of inflammatory genes in the aorta compared to the control groups, effects unrelated to TMAO levels. Our data demonstrate that inhibition of TMAO production attenuated CKD development and cardiac hypertrophy in mice, suggesting that TMAO reduction may be a novel strategy in treating CKD and its cardiovascular disease complications.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Metilaminas/efectos adversos , Metilaminas/metabolismo , Insuficiencia Renal Crónica/etiología , Adenina/administración & dosificación , Adenina/efectos adversos , Albuminuria/etiología , Animales , Cardiomegalia/etiología , Cardiomegalia/prevención & control , Colina/administración & dosificación , Colina/efectos adversos , Colina/análogos & derivados , Colina/farmacología , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Fibrosis , Riñón/patología , Metilaminas/administración & dosificación , Ratones , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/prevención & control
3.
J Exp Clin Cancer Res ; 35: 73, 2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-27151407

RESUMEN

BACKGROUND: Chemotherapy with docetaxel (Doc) remains the standard treatment for metastatic and castration-resistance prostate cancer (CRPC). However, the clinical success of Doc is limited by its chemoresistance and side effects. This study investigated whether natural products green tea (GT) and quercetin (Q) enhance the therapeutic efficacy of Doc in CRPC in mouse models. METHODS: Male severe combined immunodeficiency (SCID) mice (n = 10 per group) were inoculated with androgen-independent prostate cancer PC-3 cells subcutaneously. When tumors were established the intervention started. Mice were administered with GT + Q, Doc 5 mg/kg (LD), GT + Q + LD Doc, Doc 10 mg/kg (HD) or control. The concentration of GT polyphenols in brewed tea administered as drinking water was 0.07% and Q was supplemented in diet at 0.4%. Doc was intravenously injected weekly for 4 weeks, GT and Q given throughout the study. RESULTS: GT + Q or LD Doc slightly inhibited tumor growth compared to control. However, the combination of GT and Q with LD Doc significantly enhanced the potency of Doc 2-fold and reduced tumor growth by 62% compared to LD Doc in 7-weeks intervention. A decrease of Ki67 and increase of cleaved caspase 7 were observed in tumors by the mixture, along with lowered blood concentrations of growth factors like VEGF and EGF. The mixture significantly elevated the levels of tumor suppressor mir15a and mir330 in tumor tissues. An increased risk of liver toxicity was only observed with HD Doc treatment. CONCLUSIONS: These results provide a promising regimen to enhance the therapeutic effect of Doc in a less toxic manner.


Asunto(s)
Polifenoles/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Quercetina/administración & dosificación , Taxoides/administración & dosificación , Té/química , Animales , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Docetaxel , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones SCID , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Extractos Vegetales/farmacología , Polifenoles/farmacología , Neoplasias de la Próstata/metabolismo , Quercetina/farmacología , Taxoides/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Nutr Biochem ; 25(1): 73-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24314868

RESUMEN

The chemopreventive activity of green tea (GT) is limited by the low bioavailability and extensive methylation of GT polyphenols (GTPs) in vivo. We determined whether a methylation inhibitor quercetin (Q) will enhance the chemoprevention of prostate cancer in vivo. Androgen-sensitive LAPC-4 prostate cancer cells were injected subcutaneously into severe combined immunodeficiency (SCID) mice one week before the intervention. The concentration of GTPs in brewed tea administered as drinking water was 0.07% and Q was supplemented in diet at 0.2% or 0.4%. After 6-weeks of intervention tumor growth was inhibited by 3% (0.2% Q), 15% (0.4% Q), 21% (GT), 28% (GT+0.2% Q) and 45% (GT+0.4% Q) compared to control. The concentration of non-methylated GTPs was significantly increased in tumor tissue with GT+0.4% Q treatment compared to GT alone, and was associated with a decreased protein expression of catechol-O-methyltransferase and multidrug resistance-associated protein (MRP)-1. The combination treatment was also associated with a significant increase in the inhibition of proliferation, androgen receptor and phosphatidylinositol 3-kinase/Akt signaling, and stimulation of apoptosis. The combined effect of GT+0.4% Q on tumor inhibition was further confirmed in another experiment where the intervention started prior to tumor inoculation. These results provide a novel regimen by combining GT and Q to improve chemoprevention in a non-toxic manner and warrant future studies in humans.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Quercetina/farmacología , Té/química , Animales , Apoptosis/efectos de los fármacos , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioprevención , Masculino , Ratones , Ratones SCID , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Polifenoles/farmacología , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Nutr Biochem ; 23(11): 1537-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22405694

RESUMEN

It has been demonstrated in various animal models that the oral administration of green tea (GT) extracts in drinking water can inhibit tumor growth, but the effects of brewed GT on factors promoting tumor growth, including oxidant damage of DNA and protein, angiogenesis and DNA methylation, have not been tested in an animal model. To explore these potential mechanisms, brewed GT was administered instead of drinking water to male severe combined immunodeficiency (SCID) mice with androgen-dependent human LAPC4 prostate cancer cell subcutaneous xenografts. Tumor volume was decreased significantly in mice consuming GT, and tumor size was significantly correlated with GT polyphenol (GTP) content in tumor tissue. There was a significant reduction in hypoxia-inducible factor 1-alpha and vascular endothelial growth factor protein expression. GT consumption significantly reduced oxidative DNA and protein damage in tumor tissue as determined by 8-hydroxydeoxyguanosine/deoxyguanosine ratio and protein carbonyl assay, respectively. Methylation is known to inhibit antioxidative enzymes such as glutathione S-transferase pi to permit reactive oxygen species promotion of tumor growth. GT inhibited tumor 5-cytosine DNA methyltransferase 1 mRNA and protein expression significantly, which may contribute to the inhibition of tumor growth by reactivation of antioxidative enzymes. This study advances our understanding of tumor growth inhibition by brewed GT in an animal model by demonstrating tissue localization of GTPs in correlation with inhibition of tumor growth. Our results suggest that the inhibition of tumor growth is due to GTP-mediated inhibition of oxidative stress and angiogenesis in the LAPC4 xenograft prostate tumor in SCID mice.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Polifenoles/farmacología , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/tratamiento farmacológico , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Camellia sinensis , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/genética , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análisis , Desoxiguanosina/metabolismo , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/patología , Masculino , Ratones , Ratones SCID , Extractos Vegetales/farmacología , Polifenoles/farmacocinética , Neoplasias de la Próstata/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Heart Rhythm ; 8(3): 439-47, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21056123

RESUMEN

BACKGROUND: Contact mapping of the ventricle with NAVX has not been validated. OBJECTIVE: This study sought to compare the accuracy of infarct mapping between NAVX and CARTO using a histopathologic gold standard. METHODS: A closed-chest porcine infarction model was created by circumflex artery occlusion. After 4 to 12 weeks, 7 subjects underwent high-density endocardial and epicardial mapping using CARTO (Biosense Webster, Diamond Bar, California) and NAVX (St. Jude Medical, St. Paul, Minnesota) mapping systems. After mapping, animals were euthanized and histopathologic examination was used to quantify areas of scar at depths of 1 to 4 mm. RESULTS: Using a single-point acquisition approach with CARTO and a multipoint acquisition approach with the NAVX, endocardial and epicardial maps created using CARTO consisted of 360 ± 121 points performed in 54 ± 23 minutes/361 ± 90 points in 41 ± 13 minutes compared with 697 ± 132 points in 35 ± 8 minutes/1,303 ± 207 points in 30 ± 11 minutes using NAVX. At a 2-mm depth from the endocardial and epicardial surface, the mean scar area quantified by histopathology was 9.5 ± 8.7 cm(2) and 6.2 ± 4.8 cm(2), respectively. Correlation between histopathology and electroanatomic maps was excellent (r = 0.88, CARTO and 0.92, NAVX). Correlation between scar area determined by CARTO and NAVX was good (r = 0.88, P <.0001). CONCLUSION: Balloon occlusion-reperfusion of the circumflex artery creates a localized patchy infarction in the inferolateral wall. Multipolar mapping achieves higher density in a shorter period of time and increases the detection of late potentials. A strong correlation between CARTO and NAVX exists, and both systems demonstrate good correlation with histopathologic quantification of scar.


Asunto(s)
Mapeo del Potencial de Superficie Corporal/métodos , Técnicas Electrofisiológicas Cardíacas/métodos , Animales , Cicatriz/patología , Cicatriz/fisiopatología , Modelos Animales de Enfermedad , Electrocardiografía , Endocardio/fisiopatología , Mapeo Epicárdico , Infarto del Miocardio/fisiopatología , Reperfusión Miocárdica , Pericardio/fisiopatología , Porcinos
7.
J Biol Chem ; 278(44): 43787-96, 2003 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-12920119

RESUMEN

Parathyroid hormone (PTH) regulates extracellular calcium homeostasis through the type 1 PTH receptor (PTH1R) expressed in kidney and bone. The PTH1R undergoes beta-arrestin/dynamin-mediated endocytosis in response to the biologically active forms of PTH, PTH-(1-34), and PTH-(1-84). We now show that amino-truncated forms of PTH that do not activate the PTH1R nonetheless induce PTH1R internalization in a cell-specific pattern. Activation-independent PTH1R endocytosis proceeds through a distinct arrestin-independent mechanism that is operative in cells lacking the adaptor protein Na/H exchange regulatory factor 1 (NHERF1) (ezrin-binding protein 50). Using a combination of radioligand binding experiments and quantitative, live cell confocal microscopy of fluorescently tagged PTH1Rs, we show that in kidney distal tubule cells and rat osteosarcoma cells, which lack NHERF1, the synthetic antagonist PTH-(7-34) and naturally circulating PTH-(7-84) induce internalization of PTH1R in a beta-arrestin-independent but dynamin-dependent manner. Expression of NHERF1 in these cells inhibited antagonist-induced endocytosis. Conversely, expression of dominant-negative forms of NHERF1 conferred internalization sensitivity to PTH-(7-34) in cells expressing NHERF1. Mutation of the PTH1R PDZ-binding motif abrogated interaction of the receptor with NHERF1. These mutated receptors were fully functional but were now internalized in response to PTH-(7-34) even in NHERF1-expressing cells. Removing the NHERF1 ERM domain or inhibiting actin polymerization allowed otherwise inactive ligands to internalize the PTH1R. These results demonstrate that NHERF1 acts as a molecular switch that legislates the conditional efficacy of PTH fragments. Distinct endocytic pathways are determined by NHERF1 that are operative for the PTH1R in kidney and bone cells.


Asunto(s)
Fosfoproteínas/fisiología , Receptor de Hormona Paratiroídea Tipo 1/metabolismo , Actinas/metabolismo , Secuencias de Aminoácidos , Animales , Arrestinas/metabolismo , Huesos/metabolismo , Línea Celular , AMP Cíclico/metabolismo , Citoesqueleto/metabolismo , ADN Complementario/metabolismo , Dinaminas/metabolismo , Endocitosis , Genes Dominantes , Humanos , Immunoblotting , Fosfatos de Inositol/metabolismo , Riñón/metabolismo , Ligandos , Ratones , Microscopía Confocal , Microscopía Fluorescente , Mutación , Fosfoproteínas/metabolismo , Pruebas de Precipitina , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Intercambiadores de Sodio-Hidrógeno , Factores de Tiempo , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA