Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(10)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38791252

RESUMEN

Leptin is an obesity-related hormone that plays an important role in breast cancer progression. Vasculogenic mimicry (VM) refers to the formation of vascular channels lined by tumor cells. This study aimed to investigate the relationship between leptin and VM in human breast cancer cells. VM was measured by a 3D culture assay. Signal transducers and activators of transcription 3 (STAT3) signaling, aquaporin-1 (AQP1), and the expression of VM-related proteins, including vascular endothelial cadherin (VE-cadherin), twist, matrix metalloproteinase-2 (MMP-2), and laminin subunit 5 gamma-2 (LAMC2), were examined by Western blot. AQP1 mRNA was analyzed by a reverse transcriptase-polymerase chain reaction (RT-PCR). Leptin increased VM and upregulated phospho-STAT3, VE-cadherin, twist, MMP-2, and LAMC2. These effects were inhibited by the leptin receptor-blocking peptide, Ob-R BP, and the STAT3 inhibitor, AG490. A positive correlation between leptin and AQP1 mRNA was observed and was confirmed by RT-PCR. Leptin upregulated AQP1 expression, which was blocked by Ob-R BP and AG490. AQP1 overexpression increased VM and the expression of VM-related proteins. AQP1 silencing inhibited leptin-induced VM and the expression of VM-related proteins. Thus, these results showed that leptin facilitates VM in breast cancer cells via the Ob-R/STAT3 pathway and that AQP1 is a key mediator in leptin-induced VM.


Asunto(s)
Neoplasias de la Mama , Leptina , Neovascularización Patológica , Femenino , Humanos , Antígenos CD , Acuaporina 1/metabolismo , Acuaporina 1/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Cadherinas/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Laminina/metabolismo , Leptina/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Células MCF-7 , Neovascularización Patológica/metabolismo , Neovascularización Patológica/genética , Transducción de Señal , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética
2.
Int J Mol Sci ; 24(4)2023 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-36834665

RESUMEN

Apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) is a multifunctional protein involved in DNA repair and redox regulation. The redox activity of APE1/Ref-1 is involved in inflammatory responses and regulation of DNA binding of transcription factors related to cell survival pathways. However, the effect of APE1/Ref-1 on adipogenic transcription factor regulation remains unknown. In this study, we investigated the effect of APE1/Ref-1 on the regulation of adipocyte differentiation in 3T3-L1 cells. During adipocyte differentiation, APE1/Ref-1 expression significantly decreased with the increased expression of adipogenic transcription factors such as CCAAT/enhancer binding protein (C/EBP)-α and peroxisome proliferator-activated receptor (PPAR)-γ, and the adipocyte differentiation marker adipocyte protein 2 (aP2) in a time-dependent manner. However, APE1/Ref-1 overexpression inhibited C/EBP-α, PPAR-γ, and aP2 expression, which was upregulated during adipocyte differentiation. In contrast, silencing APE1/Ref-1 or redox inhibition of APE1/Ref-1 using E3330 increased the mRNA and protein levels of C/EBP-α, PPAR-γ, and aP2 during adipocyte differentiation. These results suggest that APE1/Ref-1 inhibits adipocyte differentiation by regulating adipogenic transcription factors, suggesting that APE1/Ref-1 is a potential therapeutic target for regulating adipocyte differentiation.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma , Factores de Transcripción , Animales , Ratones , Células 3T3-L1 , Adipocitos/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Receptores Activados del Proliferador del Peroxisoma/metabolismo , PPAR gamma/metabolismo , Factores de Transcripción/metabolismo
3.
Int J Mol Sci ; 24(8)2023 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-37108616

RESUMEN

Mitochondrial oxidative phosphorylation (OXPHOS) system dysfunction in cancer cells has been exploited as a target for anti-cancer therapeutic intervention. The downregulation of CR6-interacting factor 1 (CRIF1), an essential mito-ribosomal factor, can impair mitochondrial function in various cell types. In this study, we investigated whether CRIF1 deficiency induced by siRNA and siRNA nanoparticles could suppress MCF-7 breast cancer growth and tumor development, respectively. Our results showed that CRIF1 silencing decreased the assembly of mitochondrial OXPHOS complexes I and II, which induced mitochondrial dysfunction, mitochondrial reactive oxygen species (ROS) production, mitochondrial membrane potential depolarization, and excessive mitochondrial fission. CRIF1 inhibition reduced p53-induced glycolysis and apoptosis regulator (TIGAR) expression, as well as NADPH synthesis, leading to additional increases in ROS production. The downregulation of CRIF1 suppressed cell proliferation and inhibited cell migration through the induction of G0/G1 phase cell cycle arrest in MCF-7 breast cancer cells. Similarly, the intratumoral injection of CRIF1 siRNA-encapsulated PLGA nanoparticles inhibited tumor growth, downregulated the assembly of mitochondrial OXPHOS complexes I and II, and induced the expression of cell cycle protein markers (p53, p21, and p16) in MCF-7 xenograft mice. Thus, the inhibition of mitochondrial OXPHOS protein synthesis through CRIF1 deletion destroyed mitochondrial function, leading to elevated ROS levels and inducing antitumor effects in MCF-7 cells.


Asunto(s)
Neoplasias de la Mama , Animales , Femenino , Humanos , Ratones , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias de la Mama/genética , Proteínas de Ciclo Celular/metabolismo , Células MCF-7 , Monoéster Fosfórico Hidrolasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , ARN Interferente Pequeño/genética , Proteína p53 Supresora de Tumor , Polietilenglicoles/química , Nanopartículas
4.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163245

RESUMEN

Sp1 transcription factor regulates genes involved in various phenomena of tumor progression. Vasculogenic mimicry (VM) is the alternative neovascularization by aggressive tumor cells. However, there is no evidence of the relationship between Sp1 and VM. This study investigated whether and how Sp1 plays a crucial role in the process of VM in human prostate cancer (PCa) cell lines, PC-3 and DU145. A cell viability assay and three-dimensional culture VM tube formation assay were performed. Protein and mRNA expression levels were detected by Western blot and reverse transcriptase-polymerase chain reaction, respectively. The nuclear twist expression was observed by immunofluorescence assay. A co-immunoprecipitation assay was performed. Mithramycin A (MiA) and Sp1 siRNA significantly decreased serum-induced VM, whereas Sp1 overexpression caused a significant induction of VM. Serum-upregulated vascular endothelial cadherin (VE-cadherin) protein and mRNA expression levels were decreased after MiA treatment or Sp1 silencing. The protein expression and the nuclear localization of twist were increased by serum, which was effectively inhibited after MiA treatment or Sp1 silencing. The interaction between Sp1 and twist was reduced by MiA. On the contrary, Sp1 overexpression enhanced VE-cadherin and twist expressions. Serum phosphorylated AKT and raised matrix metalloproteinase-2 (MMP-2) and laminin subunit 5 gamma-2 (LAMC2) expressions. MiA or Sp1 silencing impaired these effects. However, Sp1 overexpression upregulated phosphor-AKT, MMP-2 and LAMC2 expressions. Serum-upregulated Sp1 was significantly reduced by an AKT inhibitor, wortmannin. These results demonstrate that Sp1 mediates VM formation through interacting with the twist/VE-cadherin/AKT pathway in human PCa cells.


Asunto(s)
Neovascularización Patológica/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Factor de Transcripción Sp1/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Masculino , Neovascularización Patológica/patología , Células PC-3 , ARN Mensajero/metabolismo , Regulación hacia Arriba/fisiología
5.
Int J Mol Sci ; 21(2)2020 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936664

RESUMEN

Vasculogenic mimicry (VM) is the alternative process of forming vessel-like networks by aggressive tumor cells, and it has an important role in tumor survival, growth, and metastasis. Epigallocatechin-3-gallate (EGCG) is well known to have diverse bioactivities including anti-cancer effects. However, the efficacy of EGCG on VM is elusive. In this study, we explored whether and how EGCG affects VM in human prostate cancer (PCa) PC-3 cells. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Invasive and VM formation abilities were assessed by an invasion assay and a three-dimensional (3D) culture VM tube formation assay, respectively. Western blots were carried out. An immunofluorescence assay was performed to detect nuclear twist expression. EGCG effectively inhibited the invasive ability, as well as tubular channel formation, without affecting cell viability. EGCG significantly downregulated the expression of vascular endothelial cadherin (VE-cadherin) and its transcription factor, twist, N-cadherin, vimentin, phosphor-AKT, and AKT, but not phospho-erythropoietin-producing hepatocellular receptor A2 (EphA2) and EphA2. In addition, EGCG diminished the nuclear localization of twist. Treatment with SC79, an AKT activator, effectively rescued EGCG-inhibited VM formation. These results demonstrated for the first time that EGCG causes marked suppression of VM through inhibiting the twist/VE-cadherin/AKT pathway in human PCa PC-3 cells.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Catequina/análogos & derivados , Neovascularización Patológica/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias de la Próstata/irrigación sanguínea , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteína 1 Relacionada con Twist/metabolismo , Catequina/química , Catequina/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Masculino , Invasividad Neoplásica , Transducción de Señal/efectos de los fármacos
6.
Int J Mol Sci ; 20(5)2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30845749

RESUMEN

Fomes fomentarius, an edible mushroom, is known to have anti-cancer, anti-inflammatory, and anti-diabetes effects. However, the underlying anti-cancer mechanism of F. fomentarius is unknown. To determine the molecular mechanism of the anti-cancer effects of F. fomentarius, various methods were used including fluorescence-activated cell sorting, Western blotting, migration, and crystal violet assays. F. fomentarius ethanol extract (FFE) decreased cell viability in six cancer cell lines (MDA-MB-231, MCF-7, A549, H460, DU145, and PC-3). FFE decreased the migration of MDA-MB-231 cells without causing cell toxicity. Furthermore, FFE attenuated the expression of matrix metalloproteinase-9 and phosphorylation of Akt as well as increased E-cadherin in MDA-MB-231 cells. FFE arrested the S and G2/M populations by inhibiting the expression of cell cycle regulatory proteins such as cyclin-dependent kinase 2, cyclin A/E, and S-phase kinase-associated protein 2. FFE increased the sub-G1 population and expression of cleaved caspase-9, -3, and cleaved poly adenosine diphosphate (ADP-ribose) polymerase at 72 h and suppressed B-cell lymphoma 2. Interestingly, FFE and AKT inhibitors showed similar effects in MDA-MB-231 cells. Additionally, FFE contained betulin which inhibited p-AKT in MDA-MB-231 cells. Our findings demonstrate that FFE inhibits cell motility and growth and induces apoptosis by inhibiting the phsphoinositide 3- kinase /AKT pathway and caspase activation.


Asunto(s)
Productos Biológicos/farmacología , Neoplasias de la Mama/metabolismo , Coriolaceae/química , Etanol/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Metaloproteinasa 9 de la Matriz/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos
7.
Int J Mol Sci ; 20(17)2019 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-31480681

RESUMEN

The main function of the skin is to protect the body from the external environment. The barrier function of the skin is mainly provided by the stratum corneum, which consists of corneocytes bound with the corneodesmosomes and lamellar lipids. Skin barrier proteins like loricrin and filaggrin also contribute to the skin barrier function. In various skin diseases, skin barrier dysfunction is a common symptom, and skin irritants like detergents or surfactants could also perturb skin barrier function. Many efforts have been made to develop strategies to improve skin barrier function. Here, we investigated whether the microfluidized lysates of Lactobacillus rhamnosus (LR), one of the most widely used probiotic species for various health benefits, may improve the skin barrier function in a reconstructed human epidermis, Keraskin™. Application of LR lysate on Keraskin™ increased the expression of tight junction proteins; claudin 1 and occludin as determined by immunofluorescence analysis, and skin barrier proteins; loricrin and filaggrin as determined by immunohistochemistry and immunofluorescence analysis and qPCR. Also, the cytotoxicity of a skin irritant, sodium lauryl sulfate (SLS), was alleviated by the pretreatment of LR lysate. The skin barrier protective effects of LR lysate could be further demonstrated by the attenuation of SLS-enhanced dye-penetration. LR lysate also attenuated the destruction of desmosomes after SLS treatment. Collectively, we demonstrated that LR lysate has protective effects on the skin barrier, which could expand the utility of probiotics to skin-moisturization ingredients.


Asunto(s)
Epidermis/efectos de los fármacos , Lacticaseibacillus rhamnosus/metabolismo , Modelos Biológicos , Probióticos/farmacología , Administración Tópica , Anticuerpos/farmacología , Biomarcadores/metabolismo , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Desmosomas/efectos de los fármacos , Desmosomas/metabolismo , Desmosomas/ultraestructura , Epidermis/patología , Proteínas Filagrina , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Irritantes/toxicidad , Proteínas de la Membrana/metabolismo , Permeabilidad , Rodaminas/metabolismo , Proteínas de Uniones Estrechas/metabolismo
8.
Int J Mol Sci ; 20(13)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261750

RESUMEN

Acetylation of nuclear apurinic/apyrimidinic endonuclease-1/redox factor-1 (APE1/Ref-1) is associated with its extracellular secretion, despite the lack of an N-terminal protein secretion signal. In this study, we investigated plasma membrane targeting and translocation of APE1/Ref-1 in HEK293T cells with enhanced acetylation. While APE1/Ref-1 targeting was not affected by inhibition of the endoplasmic reticulum/Golgi-dependent secretion, its secretion was reduced by inhibitors of ATP-binding cassette (ABC) transporters, and siRNA-mediated down-regulation of ABC transporter A1. The association between APE1/Ref-1 and ABCA1 transporter was confirmed by proximal ligation assay and immunoprecipitation experiments. An APE1/Ref-1 construct with mutated acetylation sites (K6/K7R) showed reduced co-localization with ABC transporter A1. Exposure of trichostatin A (TSA) induced the acetylation of APE1/Ref-1, which translocated into membrane fraction. Taken together, acetylation of APE1/Ref-1 is considered to be necessary for its extracellular targeting via non-classical secretory pathway using the ABCA1 transporter.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Vías Secretoras , Acetilación , Secuencias de Aminoácidos , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Células HEK293 , Humanos , Ácidos Hidroxámicos/farmacología , Mutación , Unión Proteica , Procesamiento Proteico-Postraduccional , Transporte de Proteínas/efectos de los fármacos
9.
BMC Complement Altern Med ; 18(1): 242, 2018 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-30165848

RESUMEN

BACKGROUND: Rhus verniciflua Stokes is an Asian tree species that is used as a food supplement and traditional medicine in Korea. However, its use is restricted by its potential to cause allergy. Thus, allergen-free R. verniciflua extracts are currently being marketed as a functional health food in Korea. In the present study, three different allergen-free R. verniciflua extracts (DRVE, FRVE, and FFRVE) were produced by detoxification of R. verniciflua, and their properties and constituents were compared. METHODS: The main components and properties (antibacterial, antioxidant, anticancer, and hepatic lipogenesis inhibitory effects) of the three allergen-free extracts were compared. Moreover, the major phenolic constituents of R. verniciflua, including gallic acid, fustin, fisetin, and quercetin, were analyzed in the three extracts. RESULTS: DRVE was superior to the two other extracts with regard to antioxidant activity, while FRVE was superior with regard to antimicrobial activity and suppression of hepatic lipogenesis. FRVE exhibited lipid-lowering effects by lowering sterol regulatory element-binding protein 1 and triglyceride levels, and promoting the activation of peroxisome proliferator-activated receptor and AMP-activated protein kinase in an in vitro model of non-alcoholic fatty liver. CONCLUSIONS: Overall, our findings demonstrate various differences among the three extracts. This suggests that functional and bioactive compounds present in R. verniciflua could be altered by the detoxification process, and this property could be considered in the development of functional health foods in the future.


Asunto(s)
Extractos Vegetales , Rhus/química , Antiinfecciosos/análisis , Antiinfecciosos/química , Antiinfecciosos/farmacología , Antioxidantes/análisis , Antioxidantes/química , Antioxidantes/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fermentación , Flavonoides/análisis , Flavonoides/química , Flavonoles , Humanos , Lipogénesis/efectos de los fármacos , Modelos Biológicos , Enfermedad del Hígado Graso no Alcohólico , Fenoles/análisis , Fenoles/química , Extractos Vegetales/análisis , Extractos Vegetales/química , Extractos Vegetales/farmacología
10.
Int J Mol Sci ; 19(3)2018 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-29534512

RESUMEN

Anthocyanins, the most prevalent flavonoids in red/purple fruits and vegetables, are known to improve immune responses and reduce chronic disease risks. In this study, the anti-inflammatory activities of an anthocyanin-rich extract from red Chinese cabbage (ArCC) were shown based on its inhibitory effects in cultured endothelial cells and hyperlipidemic apolipoprotein E-deficient mice. ArCC treatment suppressed monocyte adhesion to tumor necrosis factor-α-stimulated endothelial cells. This was validated by ArCC's ability to downregulate the expression and transcription of endothelial adhesion molecules, determined by immunoblot and luciferase promoter assays, respectively. The regulation of adhesion molecules was accompanied by transcriptional inhibition of nuclear factor-κB, which restricted cytoplasmic localization as shown by immunocytochemistry. Administration of ArCC (150 or 300 mg/kg/day) inhibited aortic inflammation in hyperlipidemic apolipoprotein E-deficient mice, as shown by in vivo imaging. Immunohistochemistry and plasma analysis showed that the aortas from these mice exhibited markedly lower leukocyte infiltration, reduced plaque formation, and lower concentrations of blood inflammatory cytokines than those observed in the control mice. The results suggest that the consumption of anthocyanin-rich red Chinese cabbage is closely correlated with lowering the risk of vascular inflammatory diseases.


Asunto(s)
Antocianinas/análisis , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Brassica/química , Endotelio Vascular/efectos de los fármacos , Extractos Vegetales/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular Tumoral , Citocinas/sangre , Endotelio Vascular/metabolismo , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Masculino , Ratones , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
11.
Bioorg Med Chem Lett ; 27(9): 1914-1918, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28359791

RESUMEN

Lung cancer is the second most commonly diagnosed cancer and the leading cause of cancer deaths in both men and women in the United States. It has been recently demonstrated that osteopontin (OPN) effectively inhibits cofilin activity through the focal adhesion kinase (FAK)/AKT/Rho-associated kinase (ROCK) pathway to induce the invasion of human non-small cell lung cancer (NSCLC) cells. Plumbagin was isolated from the roots of the medicinal plant Plumbago zeylanica L. and has been reported to possess anticancer activities. However, the molecular mechanisms by which plumbagin inhibits the invasion of cancer cells is still unclear. In this study, the anti-invasive and anti-metastatic mechanisms of plumbagin were investigated in OPN-treated NSCLC A549 cells. OPN effectively induced the motility and invasion of NSCLC A549 cells and H1299 cells, which was strongly suppressed by plumbagin with no evidence of cytotoxicity. In addition, lamellipodia formation at the leading edge of cells by OPN was dramatically decreased in plumbagin-treated cells. Plumbagin caused an effective inhibition in OPN-induced the expression of ROCK1 as well as the phosphorylation of LIM kinase 1 and 2 (LIMK1/2), and cofilin. OPN-induced the phosphorylation of FAK and AKT was impaired without affecting their total forms by plumbagin treatment. OPN facilitated metastatic lung colonization, which was effectively suppressed in plumbagin-treated mice. Taken together, these results suggest that plumbagin reduces OPN-induced the invasion of NSCLC A549 cells, which resulted from inhibiting the ROCK pathway mediated by the FAK/AKT pathway and suppresses lung metastasis in vivo.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Naftoquinonas/uso terapéutico , Osteopontina/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , Células A549 , Animales , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Naftoquinonas/química , Naftoquinonas/farmacología , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Plumbaginaceae/química , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
12.
Int J Mol Sci ; 18(10)2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28946662

RESUMEN

Vascular calcification plays a role in the pathogenesis of atherosclerosis, diabetes, and chronic kidney disease; however, the role of apurinic/apyrimidinic endonuclease 1/redox factor-1 (APE1/Ref-1) in inorganic phosphate (Pi)-induced vascular smooth muscle cell (VSMC) calcification remains unknown. In this study, we investigated the possible role of APE1/Ref-1 in Pi-induced VSMC calcification. We observed that Pi decreased endogenous APE1/Ref-1 expression and promoter activity in VSMCs, and that adenoviral overexpression of APE1/Ref-1 inhibited Pi-induced calcification in VSMCs and in an ex vivo organ culture of a rat aorta. However, a redox mutant of APE1/Ref-1(C65A/C93A) did not reduce Pi-induced calcification in VSMCs, suggesting APE1/Ref-1-mediated redox function against vascular calcification. Additionally, APE1/Ref-1 overexpression inhibited Pi-induced intracellular and mitochondrial reactive oxygen species production, and APE1/Ref-1 overexpression resulted in decreased Pi-induced lactate dehydrogenase activity, pro-apoptotic Bax levels, and increased anti-apoptotic Bcl-2 protein levels. Furthermore, APE1/Ref-1 inhibited Pi-induced osteoblastic differentiation associated with alkaline phosphatase activity and inhibited Pi-exposure-induced loss of the smooth muscle phenotype. Our findings provided valuable insights into the redox function of APE1/Ref-1 in preventing Pi-induced VSMC calcification by inhibiting oxidative stress and osteoblastic differentiation, resulting in prevention of altered osteoblastic phenotypes in VSMCs.


Asunto(s)
Calcificación Fisiológica/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/metabolismo , Osteoblastos/metabolismo , Fenotipo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Masculino , Mitocondrias/metabolismo , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Oxidación-Reducción , Fosfatos/metabolismo , Fosfatos/farmacología , Interferencia de ARN , Ratas , Especies Reactivas de Oxígeno/metabolismo , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
13.
J Nat Prod ; 79(1): 156-60, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26681550

RESUMEN

Lung cancer is the most frequently diagnosed cancer and the leading cause of cancer deaths in the United States and Korea. We have previously demonstrated that osteopontin (OPN) induces cell invasion through inactivating cofilin. Inactivation of cofilin is mediated by the FAK/AKT/Rho-associated kinase (ROCK) pathway in human nonsmall cell lung cancer (NSCLC) cells. Zerumbone (1) has been shown to exert anticancer activities. In this study, whether and how 1 affects OPN-induced cell invasion was determined in NSCLC A549 cells. Results from Boyden chamber assays suggested that OPN induced invasion of A549 cells and that 1 strongly suppressed this activity without affecting cell viability. Compound 1 effectively inhibited OPN-induced protein expression of ROCK1, the phosphorylation of LIM kinase 1 and 2 (LIMK1/2), and cofilin. In addition, immunofluorescence staining showed that OPN caused a significant increase in lamellipodia formation at the leading edge of cells. However, 1 dramatically decreased OPN-induced lamellipodia formation. Compound 1 impaired OPN-induced phosphorylation of FAK and AKT, as determined by Western blot analysis. Taken together, these results suggest that 1 causes considerable suppression of OPN-induced cell invasion through inhibiting the FAK/AKT/ROCK pathway in NSCLC A549 cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Osteopontina/farmacología , Sesquiterpenos/farmacología , Western Blotting , Zingiber officinale/química , Humanos , Estructura Molecular , Fosforilación , República de Corea , Quinasas Asociadas a rho
14.
BMC Complement Altern Med ; 16: 51, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26846328

RESUMEN

BACKGROUND: In this study, we investigated the anti-obesity and anti-hyperlipidemic mechanisms of lambertianic acid (LA) isolated from Pinus koraiensis leaves and the ethanol extract of Pinus koraiensis leaves (EPK), both in vitro and in vivo. METHODS: Differentiated 3T3L-1 cells were treated with EPK (25 or 50 µg/mL) or LA (200 µM) and analyzed by western blotting or RT-PCR. In vitro, lipid accumulation of adipocytes was observed using Oil-Red-O staining and triglyceride analysis. The contribution of AMPK to anti-obesity activity was assessed by siRNA-mediated AMPK knockdown. After AMPK silencing, expression of AMPK was observed by western blotting. To confirm the in vitro activity, an animal study was conducted by administering a normal diet, HFD, and EPK for 6 weeks. Obesity-related physiological parameters and protein levels were measured. RESULTS: LA induced the expression of p-AMPK and inhibited PPARγ, C/EBP α, adiponectin, FAS, SREBP-1, and HMGCR expression. EPK containing LA significantly decreased lipid accumulation and triglyceride levels in the differentiated 3 T3-L1 cells. EPK treatment suppressed the expression of adipogenic transcription factors, FABP, GPDH, and cholesterol-synthesis-related factors in the differentiated 3 T3-L1 cells. EPK increased the expression of p-AMPK. The effects of EPK were reversed on inhibiting AMPK by using AMPK siRNA and compound C. In vivo analysis showed that body weight gain, serum triglyceride, total cholesterol, LDL cholesterol and AI value in the EPK treatment group were lower than those in the HFD control group. EPK induced the expression of p-AMPK and inhibited PPARγ in liver and adipose tissue. CONCLUSIONS: Overall, the results suggest that EPK containing LA exerts significant anti-obesity and cholesterol-lowering effects by activating AMPK.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fármacos Antiobesidad/farmacología , Ácidos Carboxílicos/farmacología , Naftalenos/farmacología , Pinus/química , Extractos Vegetales/farmacología , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Ácidos Carboxílicos/aislamiento & purificación , Colesterol/sangre , Dieta Alta en Grasa , Activación Enzimática/efectos de los fármacos , Lípidos/sangre , Hígado/efectos de los fármacos , Masculino , Ratones , Estructura Molecular , Naftalenos/aislamiento & purificación , Hojas de la Planta/química , Ratas Sprague-Dawley , Triglicéridos/metabolismo
15.
Int J Mol Sci ; 17(7)2016 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-27399684

RESUMEN

Lambertianic acid (LA) is known to have anti-allergic and antibacterial effects. However, the anticancer activities and mechanism of action of LA have not been investigated. Therefore, the anticancer effects and mechanism of LA are investigated in this study. LA decreased not only AR protein levels, but also cellular and secretory levels of PSA. Furthermore, LA inhibited nuclear translocation of the AR induced by mibolerone. LA suppressed cell proliferation by inducing G1 arrest, downregulating CDK4/6 and cyclin D1 and activating p53 and its downstream molecules, p21 and p27. LA induced apoptosis and the expression of related proteins, including cleaved caspase-9 and -3, c-PARP and BAX, and inhibited BCl-2. The role of AR in LA-induced apoptosis was assessed by using siRNA. Collectively, these findings suggest that LA exerts the anticancer effect by inhibiting AR and is a valuable therapeutic agent in prostate cancer treatment.


Asunto(s)
Antineoplásicos/toxicidad , Ácidos Carboxílicos/toxicidad , Proliferación Celular/efectos de los fármacos , Naftalenos/toxicidad , Receptores Androgénicos/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Ciclina D1/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Nandrolona/análogos & derivados , Nandrolona/farmacología , Antígeno Prostático Específico/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
16.
Bioorg Med Chem Lett ; 25(9): 1956-60, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25817589

RESUMEN

Osteopontin (OPN) is involved in tumor progression such as invasion and metastasis, and poor prognosis of lung cancer. However, how OPN affects the invasive behavior of lung cancer is not well defined. Here, we examined the underlying molecular mechanism of OPN-induced invasion in human non-small cell lung cancer (NSCLC) cell lines including A549 cells. OPN markedly increased the phosphorylation of LIM kinase 1 and 2 (LIMK1/2), and cofilin without affecting their total forms. The expression of Rho-associated kinase 1 (ROCK1), but not p21-activated kinase 1 and 2 (PAK1/2), was increased by OPN treatment as an upstream effector of LIMK/cofilin. The phosphorylation of cofilin by OPN was suppressed when cells were pretreated with ROCK inhibitor Y27632 by Western blot. Moreover, it verified that OPN inactivated cofilin through ROCK signaling in other NSCLC cell lines. OPN induced the phosphorylation of FAK and AKT. FAK inhibitor FAKi-14 and PI3K inhibitor wortmannin decreased the expressions of ROCK1, and phosphorylation of LIMK1/2 and cofilin. OPN caused a significant increase in the lamellipodia formation and cell invasion, and these are suppressed by FAK inhibitor FAKi-14, PI3K inhibitor wortmannin and ROCK inhibitor Y27632. Taken together, these results suggest that OPN triggers ROCK signaling mediated by FAK/PI3K/AKT pathway, which in turn induces the lamellipodia formation to allow the invasion of lung cancer cells through inactivating cofilin.


Asunto(s)
Factores Despolimerizantes de la Actina/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Osteopontina/metabolismo , Transducción de Señal , Quinasas Asociadas a rho/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Invasividad Neoplásica , Osteopontina/farmacología , Embarazo , Transducción de Señal/efectos de los fármacos
17.
Bioorg Med Chem Lett ; 24(11): 2560-4, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24768446

RESUMEN

Among many signals to regulate hypoxia inducible factor 1α (HIF-1α), sphingosine kinase 1 (SPHK1) is also involved in various biological activities such as cell growth, survival, invasion, angiogenesis, and carcinogenesis. Thus, in the present study, molecular mechanisms of coumestrol were investigated on the SPHK1 and HIF-1α signaling pathway in hypoxic PC-3 prostate cancer cells. Coumestrol significantly suppressed SPHK1 activity and accumulation of HIF-1α in a time- and concentration-dependent manner in hypoxic PC-3 cells. In addition, coumestrol inhibited the phosphorylation status of AKT and glycogen synthase kinase-3ß (GSK 3ß) signaling involved in cancer metabolism. Furthermore, SPHK1 siRNA transfection, sphigosine kinase inhibitor (SKI), reactive oxygen species (ROS) enhanced the inhibitory effect of coumestrol on the accumulation of HIF-1α and the expression of pAKT and pGSK 3ß in hypoxic PC-3 cells by combination index. Overall, our findings suggest that coumestrol suppresses the accumulation of HIF-1α via suppression of SPHK1 pathway in hypoxic PC-3 cells.


Asunto(s)
Cumestrol/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Fosfotransferasas (Aceptor de Grupo Alcohol)/antagonistas & inhibidores , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Línea Celular Tumoral , Cumestrol/química , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad
18.
Bioorg Med Chem Lett ; 24(4): 1176-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24440300

RESUMEN

Breast cancer is the most common malignancy among females, and cancer invasion and metastasis are the leading causes of cancer death in breast cancer patients. Pterostilbene, a naturally occurring dimethylether analogue of resveratrol, has been demonstrated to possess anti-cancer effects. However, inhibitory effects of pterostilbene on cell migration and invasion and its underlying mechanisms are not fully understood. In this study, we investigated the anti-invasive mechanisms of pterostilbene in human breast cancer cell line MDA-MB-231 cells. Pterostilbene effectively inhibited serum-induced migration and invasion without affecting the viability of breast cancer cells. The mRNA expression and activity of urokinase-type plasminogen activator (uPA) were markedly reduced by pterostilbene treatment. Moreover, pterostilbene attenuated nuclear factor κB (NF-κB) transcriptional activity and DNA binding of NF-κB on uPA promoter. In addition, pterostilbene significantly impaired the activity of Rac1 and the expression of WASP-family verprolin-homologous protein-2 (WAVE-2) and actin-related protein 2/3 (Arp2/3). Overall, these results suggest that pterostilbene caused considerable suppression of cell migration and invasion through blocking NF-κB-mediated uPA expression and Rac1/WAVE/Arp2/3 pathway.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina/antagonistas & inhibidores , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Invasividad Neoplásica/prevención & control , Estilbenos/farmacología , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores , Familia de Proteínas del Síndrome de Wiskott-Aldrich/antagonistas & inhibidores , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Conformación Molecular , Invasividad Neoplásica/patología , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Familia de Proteínas del Síndrome de Wiskott-Aldrich/metabolismo , Proteína de Unión al GTP rac1/metabolismo
19.
J Nat Prod ; 77(5): 1135-9, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24828286

RESUMEN

The invasive behavior of cancer cells resulting in metastasis is the major cause of cancer-related deaths. Rhapontigenin (1) has various biological activities including anticancer activities. However, whether and how 1 affects cancer invasion has never been explored. Here, we examined the anti-invasive effects of 1 and its underlying molecular mechanisms in the highly invasive human breast cancer cell line designated MDA-MB-231. At noncytotoxic concentrations, 1 strongly suppressed serum-induced cell migration and invasion as judged by Boyden chamber analysis and wound-healing assays, respectively. Compound 1 strikingly reduced Rac1 activity as judged by both absorbance-based and pull-down assays. In addition, its downstream effectors such as WASP-family verprolin homologous proteins 2 (WAVE-2) and p21-activated kinase 1 (PAK1) signaling cascades were attenuated after treatment with 1. Immunofluorescence staining showed that 1 diminished lamellipodia formation at the leading edge of cells. Finally, 1 decreased the phosphorylation of phosphoinisitide-3-kinase (PI3K) and AKT. Rac1 activity was inhibited by the PI3K inhibitor wortmannin. Taken together, these results suggest that 1 suppresses breast cancer cell migration and invasion, which is involved in inhibiting the PI3K-dependent Rac1 signaling pathway.


Asunto(s)
Inhibidores de las Quinasa Fosfoinosítidos-3 , Estilbenos/farmacología , Androstadienos/farmacología , Neoplasias de la Mama/metabolismo , Movimiento Celular/efectos de los fármacos , Femenino , Humanos , Estructura Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estilbenos/química , Vía de Señalización Wnt/efectos de los fármacos , Wortmanina
20.
BMC Complement Altern Med ; 14: 275, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25074784

RESUMEN

BACKGROUND: The essential oil of Pinus koraiensis (EOPK) is biologically active compound obtained from the leaves of P. koraiensis. The goal of this study was to investigate the anti-cancer mechanism of EOPK in HCT116 colorectal cancer cells. METHODS: HCT116 cell proliferation was assessed by conducting crystal violet and BrdU assays. To assess the effects of EOPK on cell migration, we performed a wound-healing assay. Further, the contribution of PAK1 to EOPK-induced AKT and extracellular signal-regulated kinase (ERK) suppression was assessed by siRNA-mediated PAK1 knockdown. Changes to the expression and phosphorylation of PAK1 and its effectors were determined by western blotting, and changes to the actin cytoskeleton were determined by performing an immunofluorescence assay. RESULTS: EOPK significantly decreased HCT116 cell proliferation and migration, and induced G1 arrest without affecting normal cells. Additionally, EOPK suppressed the expression of PAK1, and decreased ERK and AKT phosphorylation in HCT116 cells. Finally, EOPK suppressed ß-catenin, cyclin D1, and CDK4/6 expression. CONCLUSIONS: Our studies indicate that EOPK significantly reduced proliferation and migration of colorectal cancer cells. Furthermore, EOPK suppressed PAK1 expression in a dose-dependent manner, and this suppression of PAK1 led to inhibition of ERK, AKT, and ß-catenin activities. Our findings suggest that EOPK exerts its anticancer activity via the inhibition of PAK1 expression, suggesting it may be a potent chemotherapeutic agent for colorectal cancer.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Aceites Volátiles/farmacología , Pinus/química , Extractos Vegetales/farmacología , Quinasas p21 Activadas/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Neoplasias Colorrectales/patología , Células HCT116 , Humanos , Ratones , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Quinasas p21 Activadas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA