Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 9(1): 11974, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31427606

RESUMEN

Accumulating evidence indicates that inflammation participates in the pathophysiological progress from insulin resistance, obesity, metabolic abnormalities, and type 2 diabetes mellitus. Our previous study reveals that interleukin-4 (IL-4) inhibits adipogenesis and promotes lipolysis to decrease lipid deposits by enhancing the activity of hormone sensitive lipase (HSL). The present study further dissects and characterizes the molecular mechanism of IL-4 in regulating HSL expression and lipolytic activity in the terminal differentiated 3T3-L1 mature adipocytes. Our results showed that IL-4 increased cAMP which then enhanced PKA activity and subsequent phosphorylation of HSL and perilipin. The phosphorylated HSL (p-HSL) translocated from cytoplasm to the surface of lipid droplets and exhibited lipolytic function. After being phosphorylated, p-perilipin also facilitated lipolysis through interacting with p-HSL. The in vitro findings were further verified by in vivo study in which IL-4 exhibited pro-lipolytic activity and enhanced HSL activity. In summary, the net outcome of IL-4 treatment is to reduce lipid storage by promoting lipolysis through enhancing HSL activity via cAMP/PKA pathway, the major route leading to lipolysis.


Asunto(s)
Interleucina-4/metabolismo , Metabolismo de los Lípidos , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-4/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Ratones , Modelos Biológicos , Fosforilación , Proteolisis/efectos de los fármacos
2.
Eur J Pharmacol ; 840: 79-88, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30287153

RESUMEN

Escin (ß-escin) is used as traditional folk medicine. The anti-tumour effects of escin have been demonstrated in vitro in certain cell lines, but its effect on bladder cancer has not been well investigated. In this study, the apoptotic activity of escin dissolved in dimethyl sulfoxide (DMSO) in bladder cancer cells and normal peripheral blood mononuclear cells (PBMC) and SV-HUC1 cells (controls) was determined. Cell cytotoxicity was assessed using the MTT assay. Cell cycle, Reactive oxygen species (ROS) generation, annexin V-FITC staining (for detecting early apoptosis), and changes in mitochondrial membrane potential were evaluated using flow cytometry. Expression of apoptosis-related proteins such as Fas (CD95) death receptor/FADD (Fas-associated protein with death domain) and BCL2 family of proteins was assessed using immunoblotting. Escin dose-dependently inhibited the growth of human bladder cancer cells, and showed IC50 of ~40 µM. The cell population in the sub-G1 phase, annexin-V staining, Fas expression, ratio of BAX/BCL2, cleavage of activated caspase-3/-8/-9, increase in poly (ADP-ribose) polymerase (PARP) levels, and suppression of nuclear factor kappa B (NF-κB) were observed after 24 h of escin treatment. Escin decreased mitochondrial membrane potential and increased cytochrome C release via generation of reactive oxygen species, which led to apoptosis of bladder cancer cells. Furthermore, escin effectively inhibited bladder tumour growth in a xenograft mouse model. Together, these results demonstrate that escin induces apoptosis in human bladder cancer cells through the Fas death receptor and mitochondrial pathways and inhibits bladder tumour growth. Escin is a potential chemotherapeutic agent for bladder cancer.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Escina/farmacología , Neoplasias de la Vejiga Urinaria/patología , Animales , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Muerte Celular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Anticancer Res ; 38(6): 3435-3445, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29848694

RESUMEN

BACKGROUND/AIM: Pyrimethamine (PYR), an anti-malarial drug is known to inhibit various types of human cancer cells. The aim of this study was to investigate the anti-tumour effects of pyrimethamine (PYR) and its underlying molecular mechanisms using the human NSCLC cell line A549. MATERIALS AND METHODS: PYR was dissolved in dimethyl sulfoxide to determine its apoptotic activity on A549 cells. Cell viability was determined by the MTT assay. Cell cycle, mitochondrial membrane potential, and Annexin V-FITC early apoptosis detection were evaluated by flow cytometry. Cyclin-dependent kinase (CDK) and Bcl-2 family protein expression was determined by western blotting. RESULTS: PYR reduced cell viability percentage and induced G0/G1 arrest, which was associated with down-regulation of cyclins D1 and E, CDK4, and CDK2, and up-regulation of p21. PYR induced sub-G1 accumulation, Annexin-V binding, caspase-9 and -3 activation, poly (ADPribose) polymerase cleavage, and mitochondrial dysfunction in A549 cells. Moreover, PYR effectively inhibited NSCLC tumour growth in an A549 xenograft model. CONCLUSION: PYR demonstrated anti-tumour effects on NSCLC in vitro and in vivo, indicating its therapeutic potential against human NSCLC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Pirimetamina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Células A549 , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos
4.
Oncol Rep ; 37(6): 3606-3616, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28498480

RESUMEN

Miconazole (MIC), an antifungal agent, diplays anti­tumorigenic activity in various types of human cancers, including bladder cancer, yet its mechanism of antitumor action is not well understood. In the present study, we demonstrated that, in a cell viability assay, MIC had a cytotoxic effect on human T24, J82 and TSGH-8301 bladder cancer cells in a dose- and time­dependent manner, but did not exhibit significant toxicity toward human peripheral blood mononuclear cells. Cell cycle analysis revealed that MIC at concentrations of 25 and 50 µM significantly caused G0/G1 arrest in the TSGH-8301 and T24 cells, respectively. DNA fragmentation, mitochondrial membrane potential and western blot analyses showed that MIC inhibited the growth of these cells by both mitochondrial­mediated and death receptor (DR5)­mediated apoptosis pathways. Specifically, MIC increased the protein levels of p21 and p27, but decreased the expression of cyclin E1, CDK2 and CDK4. MIC augmented the expression of DR5, cleaved forms of caspase-3 -8 and -9, poly(ADP­ribose) polymerase and Bax, decreased the expression of Bcl-2 but increased cytosol levels of cytochrome c. Our results suggest that MIC inhibits the growth of bladder cancer cells through induction of G0/G1 arrest and apoptosis via activation of both the extrinsic and intrinsic apoptotic pathways. MIC is a potential chemotherapeutic agent for treating bladder cancer in humans.


Asunto(s)
Apoptosis/efectos de los fármacos , Miconazol/administración & dosificación , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteínas de Neoplasias/genética , Transducción de Señal/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
5.
Oncol Rep ; 37(2): 1002-1010, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28075477

RESUMEN

Escin, a natural pentacyclic triterpenoid compound, exhibits antitumor effects on various types of human cancer cells, but its effect on human renal cancer cells has not been fully elucidated. In the present study, we demonstrated that escin elicits cytotoxic effects on human renal cancer cells (786-O and Caki-1) in a dose-dependent manner, as determined by MTT assay. Escin induced G2/M arrest, and then increased the sub-G1 population, Annexin V binding, activation of caspase-9/-3, cleavage of poly(ADP-ribose) polymerase (PARP) and Bax protein. Escin also decreased the anti-apoptotic protein levels of Bcl-2, X-linked inhibitor of apoptosis protein and survivin. In addition, escin induced reactive oxygen species (ROS) generation, leading to mitochondrial membrane potential dysfunction and inducing apoptosis in 786-O renal cancer cells, which were suppressed by antioxidants, such as NAC. Collectively, our results suggest that escin induces apoptosis via the intrinsic-mitochondrial apoptosis pathway through G2/M arrest and ROS generation in human renal cancer cells. Escin appears to have potential as a clinically useful chemotherapeutic agent for human renal cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Escina/farmacología , Neoplasias Renales/tratamiento farmacológico , Especies Reactivas de Oxígeno/metabolismo , Antineoplásicos/farmacología , Caspasas/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
6.
Eur J Pharmacol ; 761: 309-20, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26086857

RESUMEN

Nortriptyline (NTP), an antidepressant, has antitumor effects on some human cancer cells, but its effect on human bladder cancer cells is not known. In this study, we used a cell viability assay to demonstrate that NTP is cytotoxic to human TCCSUP and mouse MBT-2 bladder cancer cells in a concentration and time-dependent manner. We also performed cell cycle analysis, annexin V and mitochondrial membrane potential assays, and Western blot analysis to show that NTP inhibits cell growth in these cells by inducing both mitochondria-mediated and death receptor-mediated apoptosis. Specifically, NTP increases the expression of Fas, FasL, FADD, Bax, Bak, and cleaved forms of caspase-3, caspase-8, caspase-9, and poly(ADP-ribose) polymerase. In addition, NTP decreases the expression of Bcl-2, Bcl-xL, BH3 interacting domain death agonist, X-linked inhibitor of apoptosis protein, and survivin. Furthermore, NTP-induced apoptosis is associated with reactive oxygen species (ROS) production, which can be reduced by antioxidants, such as N-acetyl-L-cysteine. Finally, we showed that NTP suppresses tumor growth in mice inoculated with MBT-2 cells. Collectively, our results suggest that NTP induces both intrinsic and extrinsic apoptosis in human and mouse bladder cancer cells and that it may be a clinically useful chemotherapeutic agent for bladder cancer in humans.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Nortriptilina/farmacología , Receptores de Muerte Celular/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Masculino , Ratones Endogámicos C3H , Mitocondrias/metabolismo , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Muerte Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
7.
Artículo en Inglés | MEDLINE | ID: mdl-21760824

RESUMEN

Calocedrus formosana (Florin) bark acetone/ethylacetate extracts are known to exert an antitumor effect on some human cancer cell lines, but the mechanism is yet to be defined. The aim of this study was to determine the effects of Florin leaf methanol extracts on the growth and apoptosis of human bladder cancer cell lines. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay showed that the growth of these bladder cancer cells was potently inhibited by the Florin leaf extracts. The cell cycle of these extract-treated cells (TCCSUP cells) was arrested at the G2/M phase as determined by flow cytometry. Western blot analysis revealed the increases of cyclin B1 and Cdc2 kinase levels, alone with the decrease of phosphorylated Cdc2 kinase, after treating these cells with the extracts. An immunofluorescence assessment of ß-tubulin showed decreased levels of polymerized tubulin in treated cells. However, the proteolytic cleavage of poly ADP-ribose polymerase and the activation of caspase-3/-8/-9 were all increased upon treatments of extracts. The concurrent increase of Bax and decrease of Bcl-2 levels indicated that the extracts could induce apoptosis in these treated cells. Taken together, these results suggest that the Florin leaf extracts may be an effective antibladder cancer agent.

8.
Int J Toxicol ; 30(1): 35-46, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21224419

RESUMEN

Aristolochic acid (AA) and tetrandrine (TET) are the major bioactive components in Chinese herbs used for weight loss. The nephropathy caused by the 2 Chinese herbs has not been simultaneously investigated. The aim of this study was to examine the potential nephrotoxicity of AA and TET using Madin-Darby canine kidney (MDCK) cells and mice. The results showed that TET was more potent than AA in inhibiting MDCK cell growth via inducing apoptosis, as determined by annexin-V staining, 4', 6'-diamino-2-phenylindole (DAPI) staining, DNA fragmentation, and caspase 3 activity. Mice treated with AA (10 mg/kg) by intraperitoneal administration for 3 months showed nephrotoxicity, elevated blood urea nitrogen, and increased renal tubular injuries. In contrast, mice treated with 50 mg/kg of TET in the same time period had moderate hydropic degeneration of the distal tubules in the kidneys. These results suggest that TET is more cytotoxic than AA in MDCK cells but shows less nephrotoxic than AA in mice.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Bencilisoquinolinas/toxicidad , Medicamentos Herbarios Chinos/toxicidad , Riñón/efectos de los fármacos , Nefritis Intersticial/inducido químicamente , Animales , Apoptosis/efectos de los fármacos , Ácidos Aristolóquicos/administración & dosificación , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fragmentación del ADN , Perros , Inyecciones Intraperitoneales , Riñón/patología , Riñón/fisiopatología , Pruebas de Función Renal , Longevidad/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Nefritis Intersticial/patología
9.
Urol Res ; 30(5): 282-8, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12389115

RESUMEN

We examined a human urothelial cancer T24 cell line, which was exposed to clinically achievable concentrations of Taxol and detected the lethal effect of Taxol as measured by a cytotoxic dose-response curve. Marked nuclear condensation and the fragmentation of chromatin were observed by DAPI stain, DNA ladder formation, and flow cytometry at an LC(90)concentration of 0.8 microg/ml Taxol, which also induced a G2/M arrest. In response to Taxol-treatment, caspase-9 activity increased at 8 h, and both caspase-2 and -3 activities were increased twofold relative to control cultures at 16 h. Moreover, treatment with the broad-spectrum caspase inhibitor (z-VAD-fmk) or the caspase-9 specific inhibitor (z-LEHD-fmk) effectively protected T24 cells against Taxol-triggered apoptosis. Furthermore, the phosphorylation of Bcl-2 and Bcl-X(L) proteins in Taxol treated cells was detected at 8 h. In contrast, Taxol had no effect on the levels of Fas and FasL proteins and neither antagonistic, anti-Fas antibody affected Taxol-induced apoptosis. These results suggest that, following the phosphorylation of Bcl-2 and Bcl-X(L)proteins, Taxol-induced apoptosis is induced through the mitochondria-dependent pathway in T24 cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/fisiología , Mitocondrias/fisiología , Paclitaxel/farmacología , Neoplasias de la Vejiga Urinaria/fisiopatología , Caspasas/metabolismo , División Celular/efectos de los fármacos , Proteína Ligando Fas , Humanos , Glicoproteínas de Membrana/metabolismo , Mitosis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología , Proteína bcl-X , Receptor fas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...