Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Biochem Biophys Res Commun ; 546: 130-137, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33582555

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) plays important roles in cancer-associated inflammation by controlling expression of proinflammatory cytokines and chemokines. Recent studies suggest that C/EBPß (CCAAT-enhancer binding protein beta) and STAT3 synergistically stimulate cancer cell proliferation and epithelial-mesenchymal transition. C/EBPß is a leucine-zipper transcription factor that regulates expression of a variety of inflammatory cytokines or chemokines, such as IL-8, G-CSF (granulocyte colony stimulating factor), and GM-CSF (granulocyte macrophage colony stimulating factor) which induce neutrophil infiltration and differentiation. However, molecular mechanisms by which STAT3 and C/EBPß cooperatively interact had not been fully elucidated. In this study, we found that the level of C/EBPß protein, but not that of its mRNA transcript, was decreased in the absence of STAT3 in H-Ras transformed human mammary epithelial (H-Ras MCF10A) cells. In addition, silencing STAT3 dramatically induced ubiquitination of C/EBPß for proteasomal degradation. Furthermore, direct interaction between STAT3 and C/EBPß was confirmed by immunoprecipitation and proximity ligation assays. Taken together, these results suggest that STAT3 stabilizes C/EBPß, thereby promoting cancer-associated inflammation.


Asunto(s)
Mama/patología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Transformación Celular Neoplásica , Células Epiteliales/patología , Genes ras , Factor de Transcripción STAT3/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Línea Celular Transformada , Retroalimentación Fisiológica , Femenino , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Interleucina-8/metabolismo , Neutrófilos/citología , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estabilidad Proteica , Transducción de Señal , Ubiquitinación
2.
Biochim Biophys Acta Mol Cell Res ; 1864(7): 1349-1358, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28476645

RESUMEN

Recent work has shown that deregulation of the transcription factor Myb contributes to the development of leukemia and several other human cancers, making Myb and its cooperation partners attractive targets for drug development. By employing a myeloid Myb-reporter cell line we have identified Withaferin A (WFA), a natural compound that exhibits anti-tumor activities, as an inhibitor of Myb-dependent transcription. Analysis of the inhibitory mechanism of WFA showed that WFA is a significantly more potent inhibitor of C/EBPß, a transcription factor cooperating with Myb in myeloid cells, than of Myb itself. We show that WFA covalently modifies specific cysteine residues of C/EBPß, resulting in the disruption of the interaction of C/EBPß with the co-activator p300. Our work identifies C/EBPß as a novel direct target of WFA and highlights the role of p300 as a crucial co-activator of C/EBPß. The finding that WFA is a potent inhibitor of C/EBPß suggests that inhibition of C/EBPß might contribute to the biological activities of WFA.


Asunto(s)
Antineoplásicos/farmacología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Witanólidos/farmacología , Células 3T3 , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/química , Línea Celular Tumoral , Humanos , Ratones , Unión Proteica , Factores de Transcripción p300-CBP/metabolismo
3.
J Cell Mol Med ; 22(3): 1475-1488, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29266779

RESUMEN

Diabetic cardiomyopathy, a major cardiac complication, contributes to heart remodelling and heart failure. Our previous study discovered that CCAAT/enhancer-binding protein ß (C/EBPß), a transcription factor that belongs to a family of basic leucine zipper transcription factors, interacts with the angiotensin-converting enzyme 2 (ACE2) promoter sequence in other disease models. Here, we aimed to determine the role of C/EBPß in diabetes and whether ACE2 expression is regulated by C/EBPß. A type 1 diabetic mouse model was generated by an intraperitoneal injection of streptozotocin. Diabetic mice were injected with a lentivirus expressing either C/EBPß or sh-C/EBPß or treated with valsartan after 12 weeks to observe the effects of C/EBPß. In vitro, cardiac fibroblasts and cardiomyocytes were treated with high glucose (HG) to investigate the anti-fibrosis, anti-apoptosis and regulatory mechanisms of C/EBPß. C/EBPß expression was down-regulated in diabetic mice and HG-induced cardiac neonatal cells. C/EBPß overexpression significantly attenuated collagen deposition and cardiomyocyte apoptosis by up-regulating ACE2 expression. The molecular mechanism involved the binding of C/EBPß to the ACE2 promoter sequence. Although valsartan, a classic angiotensin receptor blocker, relieved diabetic complications, the up-regulation of ACE2 expression by C/EBPß overexpression may exert greater beneficial effects on patients with diabetic cardiomyopathy.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Diabetes Mellitus Experimental/terapia , Cardiomiopatías Diabéticas/prevención & control , Fibroblastos/metabolismo , Miocitos Cardíacos/metabolismo , Peptidil-Dipeptidasa A/genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Apoptosis/genética , Glucemia/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Línea Celular , Colágeno/antagonistas & inhibidores , Colágeno/genética , Colágeno/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica , Glucosa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Peptidil-Dipeptidasa A/metabolismo , Cultivo Primario de Células , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Estreptozocina , Valsartán/farmacología
4.
Cell Physiol Biochem ; 48(4): 1605-1615, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30071524

RESUMEN

BACKGROUND/AIMS: Interleukin-1ß (IL-1ß) is one of the critical inflammatory factors during atherogenesis. CCAAT/enhancer binding proteins ß (C/EBPß), a regulator of IL-1ß production, recently been evidenced as a key player in the development of atherosclerosis. However, the mechanisms of how C/EBPß regulates the production of IL-1ß are unclear. In this study, we aimed to explore the role of C/EBPß in regulating IL-1ß production in macrophages after oxidized low-density lipoprotein (ox-LDL) exposure and the underlying mechanisms. METHODS: RAW264.7 macrophages were treated with 0, 25, 50 or 100 µg/ml ox-LDL for 12, 24 or 48 h. Small interfering RNAs were used to silence related proteins. The gene and protein expression levels were determined by quantitative real-time polymerase chain reaction or western blot (WB). IL-1ß secretion was assessed by enzyme-linked immunosorbent assay. The cytoplasmic and nuclear proteins were evaluated by nuclear fractionation followed by WB. Localization of p65 was observed by immunofluorescence. The binding activity of p65 to IL-1ß was tested by dual-luciferase reporter assay. RESULTS: Ox-LDL increased IL-1ß production, accompanied with increasing C/EBPß and p65 expression in a dose- and time-dependent manner. Moreover, C/EBPß deficiency in macrophages blocked ox-LDL-induced increases in IL-1ß expression, maturation as well as p65 activation. However, p65 deficiency inhibited the increase in IL-1ß production, but not C/EBPß expression. Dual-luciferase reporter results showed that overexpression of C/EBPß significantly enhanced binding activity of p65 to IL-1ß promoter. In addition, C/EBP 1ß deficiency in macrophages abolished the ox-LDL-induced gene transcription increases of IL-1ß, IL-6, p65 and caspase-1. CONCLUSIONS: Our results demonstrate that C/EBPß acts upstream of NF-κB p65 subunit in ox-LDL-induced IL-1ß production in macrophages and may regulate IL-1ß maturation by promoting caspase-1. C/EBPß may be a promising candidate for the prevention and treatment of atherosclerosis.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Interleucina-1beta/análisis , Lipoproteínas LDL/farmacología , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Células RAW 264.7 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética
5.
Toxicol Appl Pharmacol ; 359: 82-90, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30248417

RESUMEN

Obesity is highly correlated with several metabolic disorders. Adipocyte differentiation is a key process in determining obesogenesis. 14-Deoxy-11,12-didehydroandrographolide (deAND) is a diterpenoid rich in Andrographis paniculata (Burm.f.) Nees., a herbal medicine commonly used to treat colds, infections, and liver diseases. We investigated whether deAND inhibits the adipogenesis of 3T3-L1 cells and the underlying mechanisms. We found that deAND (0-15 µM) dose-dependently inhibits the mRNA and protein expression of peroxisome proliferator-activated receptor γ, sterol regulatory element-binding protein 1c, fatty acid synthase, and stearoyl-CoA desaturase-1. Cellular lipid accumulation was decreased by deAND, and the early phase of adipocyte differentiation was critical for this inhibition. Immunoblotting revealed that deAND attenuated differentiation medium-induced protein kinase A (PKA) and cAMP response element-binding protein (CREB) activation, which leads to down-regulating C/EBPß transcription. Moreover, deAND inhibited ERK- and GSK3ß-mediated C/EBPß transcriptional activity. Flow cytometry analysis showed that deAND impaired the progression of mitotic clonal expansion (MCE) by arresting the cell cycle at the G0/G1 phase, while the expression of cyclin D1, cyclin E, CDK6, and CDK2 was attenuated. deAND increased the phosphorylation of AMPK and raptor, an mTOR-interacting partner, which inhibited the mTOR-driven phosphorylation of P70S6K and eukaryotic translation initiation factor 4E binding protein. In the presence of compound C, deAND modulation of AMPK-mTOR signaling and inhibition of cell cycle regulator expression were reversed. Our results reveal that the anti-adipogenic effect of deAND is likely through inhibition of the PKA-CREB-C/EBPß and AMPK/mTOR pathways, which leads to down-regulating C/EBPß-driven lipogenic protein expression and halting MCE progression.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Diterpenos/farmacología , Mitosis/efectos de los fármacos , Células 3T3-L1 , Activación Metabólica/efectos de los fármacos , Andrographis/química , Animales , Células Clonales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos
6.
Cell Mol Biol (Noisy-le-grand) ; 64(4): 71-77, 2018 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-29631686

RESUMEN

Obesity is global problem that contributes to disease, and is partly caused by fast-food, high-fat diets. Much attention has been focused on developing anti-obesity foods and chemical materials from natural sources. Seaweed has bioactive properties that influence immune activity and have anti-cancer and anti-obesity effects. Laminaria japonica is a widely consumed seaweed, and has been promoted as a health food in Korea. The bioactive properties of L. japonica include anti-cancer, anti-diabetic, and anti-inflammation effects. Most Laminaria japonica are distributed in a simple processing form such as drying, and their availability is very low. Therefore, various types of functional products can be developed if they can be applied to foods through functionalization using fermentation techniques. It is a structural problem that is the most problematic in seaweed processing. In this study, we used fermented Laminaria japonica. To increase physiological activity, fermentation treatment was performed to loosen the structure, thereby increasing the activity of the glycoprotein. First, we screened the anti-obesity potential of an L. japonica fermentation extract (LJF) using 3T3-L1 adipocyte cells. We determined cytotoxicity using an MTS assay and measured LJF for its ability to affect adipogenesis through glucose uptake, triglyceride levels, and Oil Red O staining. We confirmed that LJF inhibited adipocyte differentiation. CCAAT/enhancer-binding proteins α/ß (C/EBP-α/ß) and peroxisome proliferator-activated receptor-γ (PPAR-γ) are involved in the early and late stages of adipocyte differentiation. LJF significantly reduced the expression levels of C/EBP-α/ß and PPAR-γ and decreased the concentration of adiponectin. Thus, our results suggest that LJF inhibits adipogenesis in 3T3-L1 cells, and may be valuable for its anti-obesity effects.


Asunto(s)
Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Mezclas Complejas/farmacología , Laminaria/química , PPAR gamma/genética , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Adiponectina/antagonistas & inhibidores , Adiponectina/genética , Adiponectina/metabolismo , Animales , Fármacos Antiobesidad/química , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Mezclas Complejas/química , Fermentación , Regulación de la Expresión Génica , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Transducción de Señal
7.
PLoS Genet ; 11(9): e1005505, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26372225

RESUMEN

Schmid metaphyseal chondrodysplasia (MCDS) involves dwarfism and growth plate cartilage hypertrophic zone expansion resulting from dominant mutations in the hypertrophic zone collagen, Col10a1. Mouse models phenocopying MCDS through the expression of an exogenous misfolding protein in the endoplasmic reticulum (ER) in hypertrophic chondrocytes have demonstrated the central importance of ER stress in the pathology of MCDS. The resultant unfolded protein response (UPR) in affected chondrocytes involved activation of canonical ER stress sensors, IRE1, ATF6, and PERK with the downstream effect of disrupted chondrocyte differentiation. Here, we investigated the role of the highly conserved IRE1/XBP1 pathway in the pathology of MCDS. Mice with a MCDS collagen X p.N617K knock-in mutation (ColXN617K) were crossed with mice in which Xbp1 was inactivated specifically in cartilage (Xbp1CartΔEx2), generating the compound mutant, C/X. The severity of dwarfism and hypertrophic zone expansion in C/X did not differ significantly from ColXN617K, revealing surprising redundancy for the IRE1/XBP1 UPR pathway in the pathology of MCDS. Transcriptomic analyses of hypertrophic zone cartilage identified differentially expressed gene cohorts in MCDS that are pathologically relevant (XBP1-independent) or pathologically redundant (XBP1-dependent). XBP1-independent gene expression changes included large-scale transcriptional attenuation of genes encoding secreted proteins and disrupted differentiation from proliferative to hypertrophic chondrocytes. Moreover, these changes were consistent with disruption of C/EBP-ß, a master regulator of chondrocyte differentiation, by CHOP, a transcription factor downstream of PERK that inhibits C/EBP proteins, and down-regulation of C/EBP-ß transcriptional co-factors, GADD45-ß and RUNX2. Thus we propose that the pathology of MCDS is underpinned by XBP1 independent UPR-induced dysregulation of C/EBP-ß-mediated chondrocyte differentiation. Our data suggest that modulation of C/EBP-ß activity in MCDS chondrocytes may offer therapeutic opportunities.


Asunto(s)
Enfermedades Óseas/patología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Diferenciación Celular/fisiología , Condrocitos/patología , Proteínas de Unión al ADN/fisiología , Estrés del Retículo Endoplásmico/fisiología , Factores de Transcripción/fisiología , Respuesta de Proteína Desplegada/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box
8.
J Biol Chem ; 291(50): 26098-26108, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27803164

RESUMEN

Recent work has demonstrated pro-oncogenic functions of the transcription factor CCAAT box/enhancer-binding protein ß (C/EBPß) in various tumors, implicating C/EBPß as an interesting target for the development of small-molecule inhibitors. We have previously discovered that the sesquiterpene lactone helenalin acetate, a natural compound known to inhibit NF-κB, is a potent C/EBPß inhibitor. We have now examined the inhibitory mechanism of helenalin acetate in more detail. We demonstrate that helenalin acetate is a significantly more potent inhibitor of C/EBPß than of NF-κB. Our work shows that helenalin acetate inhibits C/EBPß by binding to the N-terminal part of C/EBPß, thereby disrupting the cooperation of C/EBPß with the co-activator p300. C/EBPß is expressed in several isoforms from alternative translational start codons. We have previously demonstrated that helenalin acetate selectively inhibits only the full-length (liver-enriched activating protein* (LAP*)) isoform but not the slightly shorter (LAP) isoform. Consistent with this, helenalin acetate binds to the LAP* but not to the LAP isoform, explaining why its inhibitory activity is selective for LAP*. Although helenalin acetate contains reactive groups that are able to interact covalently with cysteine residues, as exemplified by its effect on NF-κB, the inhibition of C/EBPß by helenalin acetate is not due to irreversible reaction with cysteine residues of C/EBPß. In summary, helenalin acetate is the first highly active small-molecule C/EBPß inhibitor that inhibits C/EBPß by a direct binding mechanism. Its selectivity for the LAP* isoform also makes helenalin acetate an interesting tool to dissect the functions of the LAP* and LAP isoforms.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos Fitogénicos/farmacología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Sesquiterpenos/farmacología , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Células 3T3-L1 , Animales , Antiinflamatorios no Esteroideos/farmacocinética , Antineoplásicos Fitogénicos/farmacocinética , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Sesquiterpenos/farmacocinética , Sesquiterpenos de Guayano , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
9.
J Cell Biochem ; 118(4): 754-763, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27618583

RESUMEN

Increasing apolipoproteinA-I (apoA-I) production may be anti-atherogenic. Thus, there is a need to identify regulatory factors involved. Transcription of apoA-I involves peroxisome-proliferator-activated-receptor-alpha (PPARα) activation, but endoplasmic reticulum (ER) -stress and inflammation also influence apoA-I production. To unravel why PPARα agonist GW7647 increased apoA-I production compared to PPARα agonist fenofibric acid (FeAc) in human hepatocellular carcinoma (HepG2) and colorectal adenocarcinoma (CaCo-2) cells, gene expression profiles were compared. Microarray analyses suggested CCAAT/enhancer-binding-protein-beta (C/EBP-ß) involvement in the FeAc condition. Therefore, C/EBP-ß silencing and isoform-specific overexpression experiments were performed under ER-stressed, inflammatory and non-inflammatory conditions. mRNA expression of C/EBP-ß, ATF3, NF-IL3 and GDF15 were upregulated by FeAc compared to GW7647 in both cell lines, while DDIT3 and DDIT4 mRNA were only upregulated in HepG2 cells. This ER-stress related signature was associated with decreased apoA-I secretion. After ER-stress induction by thapsigargin or FeAc addition, intracellular apoA-I concentrations decreased, while ER-stress marker expression (CHOP, XBP1s, C/EBP-ß) increased. Cytokine addition increased intracellular C/EBP-ß levels and lowered apoA-I concentrations. Although a C/EBP binding place is present in the apoA-I promoter, C/EBP-ß silencing or isoform-specific overexpression did not affect apoA-I production in inflammatory, non-inflammatory and ER-stressed conditions. Therefore, C/EBP-ß is not a target to influence hepatic apoA-I production. J. Cell. Biochem. 118: 754-763, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Butiratos/farmacología , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Fenofibrato/análogos & derivados , PPAR alfa/agonistas , Compuestos de Fenilurea/farmacología , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Células CACO-2 , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fenofibrato/farmacología , Perfilación de la Expresión Génica , Silenciador del Gen , Células Hep G2 , Humanos , Inflamación/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Tapsigargina/farmacología
10.
Lipids Health Dis ; 16(1): 181, 2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28946872

RESUMEN

BACKGROUND: Increased consumption of omega-3 (ω-3) fatty acids found in cold-water fish and fish oil has been reported to protect against obesity. A potential mechanism may be through reduction in adipocyte differentiation. Stearidonic acid (SDA), a plant-based ω-3 fatty acid, has been targeted as a potential surrogate for fish-based fatty acids; however, its role in adipocyte differentiation is unknown. This study was designed to evaluate the effects of SDA on adipocyte differentiation in 3T3-L1 cells. METHODS: 3T3-L1 preadipocytes were differentiated in the presence of SDA or vehicle-control. Cell viability assay was conducted to determine potential toxicity of SDA. Lipid accumulation was measured by Oil Red O staining and triglyceride (TG) quantification in differentiated 3T3-L1 adipocytes. Adipocyte differentiation was evaluated by adipogenic transcription factors and lipid accumulation gene expression by quantitative real-time polymerase chain reaction (qRT-PCR). Fatty acid analysis was conducted by liquid chromatography-mass spectrometry/mass spectrometry (LC-MS/MS). RESULTS: 3T3-L1 cells treated with SDA were viable at concentrations used for all studies. SDA treatment reduced lipid accumulation in 3T3-L1 adipocytes. This anti-adipogenic effect by SDA was a result of down-regulation of mRNA levels of the adipogenic transcription factors CCAAT/enhancer-binding proteins alpha and beta (C/EBPα, C/EBPß), peroxisome proliferator-activated receptor gamma (PPARγ), and sterol-regulatory element binding protein-1c (SREBP-1c). SDA treatment resulted in decreased expression of the lipid accumulation genes adipocyte fatty-acid binding protein (AP2), fatty acid synthase (FAS), stearoyl-CoA desaturase (SCD-1), lipoprotein lipase (LPL), glucose transporter 4 (GLUT4) and phosphoenolpyruvate carboxykinase (PEPCK). The transcriptional activity of PPARγ was found to be decreased with SDA treatment. SDA treatment led to significant EPA enrichment in 3T3-L1 adipocytes compared to vehicle-control. CONCLUSION: These results demonstrated that SDA can suppress adipocyte differentiation and lipid accumulation in 3T3-L1 cells through down-regulation of adipogenic transcription factors and genes associated with lipid accumulation. This study suggests the use of SDA as a dietary treatment for obesity.


Asunto(s)
Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Supervivencia Celular/efectos de los fármacos , Acido Graso Sintasa Tipo I/antagonistas & inhibidores , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Transportador de Glucosa de Tipo 4/antagonistas & inhibidores , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones , PPAR gamma/antagonistas & inhibidores , PPAR gamma/genética , PPAR gamma/metabolismo , Fosfoenolpiruvato Carboxiquinasa (ATP)/antagonistas & inhibidores , Fosfoenolpiruvato Carboxiquinasa (ATP)/genética , Fosfoenolpiruvato Carboxiquinasa (ATP)/metabolismo , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
11.
Toxicol Appl Pharmacol ; 307: 115-122, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27475717

RESUMEN

Andrographolide, a diterpenoid, is the most abundant terpenoid in Andrographis paniculata, a popular Chinese herbal medicine. Andrographolide displays diverse biological activities including hypoglycemia, hypolipidemia, anti-inflammation, and anti-tumorigenesis. Recent evidence indicates that andrographolide displays anti-obesity property by inhibiting lipogenic gene expression, however, the underlying mechanisms remain to be elucidated. In this study, the effects of andrographolide on transcription factor cascade and mitotic clonal expansion in 3T3-L1 preadipocyte differentiation into adipocyte were determined. Andrographolide dose-dependently (0-15µM) inhibited CCAAT/enhancer-binding protein α (C/EBPα) and C/EBPß mRNA and protein expression as well as peroxisome proliferator-activated receptor γ (PPARγ) protein level during the adipogenesis of 3T3-L1 cells. Concomitantly, fatty acid synthase and stearoyl-CoA desaturase expression and lipid accumulation were attenuated by andrographolide. Oil-red O staining further showed that the first 48h after the initiation of differentiation was critical for andrographolide inhibition of adipocyte formation. Andrographolide inhibited the phosphorylation of PKA and the activation of cAMP response element-binding protein (CREB) in response to a differentiation cocktail, which led to attenuated C/EBPß expression. In addition, ERK and GSK3ß-dependent C/EBPß phosphorylation was attenuated by andrographolide. Moreover, andrographolide suppressed cyclin A, cyclin E, and CDK2 expression and impaired the progression of mitotic clonal expansion (MCE) by arresting the cell cycle at the Go/G1 phase. Taken together, these results indicate that andrographolide has a potent anti-obesity action by inhibiting PKA-CREB-mediated C/EBPß expression as well as C/EBPß transcriptional activity, which halts MCE progression and attenuates C/EBPα and PPARγ expression.


Asunto(s)
Adipogénesis/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Diterpenos/farmacología , Células 3T3-L1 , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , PPAR gamma/metabolismo
12.
J Proteome Res ; 13(3): 1307-14, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24450392

RESUMEN

Mitotic clonal expansion (MCE) is one of the important events taking place at the early stage during 3T3-L1 adipocyte differentiation. To investigate the mechanism underlying this process, we carried out a temporal proteomic analysis to profile the dynamic changes in MCE. Using 8-plex-iTRAQ-2DLC-MS/MS analysis, 3152 proteins were quantified during the initial 28 h of 3T3-L1 adipogenesis. Functional analysis was performed on 595 proteins with maximum or minimum quantities at 20 h of adipogenic induction that were potentially involved in MCE, which identified PI3K/AKT/mTOR as the most relevant pathway. Among the 595 proteins, PKM2 (Pyruvate kinase M2), a patterned protein identified as a potential target gene of C/EBPß in our previous work, was selected for further investigation. Network analysis suggested positive correlations among C/EBPß, PIN1, and PKM2, which may be related with the PI3K-AKT pathway. Knockdown of PKM2 with siRNA inhibited both MCE and adipocyte differentiation of 3T3-L1 cells. Moreover, PKM2 was down-regulated at both the mRNA level and the protein level upon the knockdown of C/EBPß. And overexpressed PKM2 can partially restore MCE, although it did not restore terminal adipocyte differentiation, which was inhibited by siC/EBPß. Thus, PKM2, potentially regulated by C/EBPß, is involved in MCE during adipocyte differentiation. The dynamic proteome changes quantified here provide a promising basis for revealing molecular mechanism regulating adipogenesis.


Asunto(s)
Adipocitos/metabolismo , Mitosis , Proteoma/análisis , Células 3T3-L1 , Adipocitos/citología , Animales , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Proliferación Celular , Cromatografía Liquida/métodos , Células Clonales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem
13.
Int J Mol Sci ; 15(10): 17242-55, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25264740

RESUMEN

To further investigate the regulation role of two chemokine genes CCL3 and CCL4 in chondrocytes in response to resistin, human primary chondrocytes and T/C-28a2 cells were cultured. The function of resistin on the chemokine genes, and the expression of C/EBPß, NF-κB isoforms were tested using qPCR. The methods used to investigate timed co-regulation of C/EBPß and NF-κB were NF-κB inhibitor (IKK-NBD) and C/EBPß inhibitor (SB303580) treatments, and subcellular localization, with or without resistin stimulation. Results showed that resistin could increase the up-regulation of chemokine genes independently. Resistin increased the expression of C/EBPß and NF-κB isoforms. C/EBPß regulated basal activity and steadily increased over time up to 24h with resistin. NF-κB was up-regulated upon induction with resistin, peaking at 4 h. C/EBPß and NF-κB co-enhanced the chemokines expression; inhibition of their activity was additive. The timing of activation in chondrocytes was confirmed by subcellular localization of C/EBPß and c-rel. Chondrocytes react to resistin in a non-restricted cell-specific manner, utilizing C/EBPß and NF-κB in a combinatorial regulation of chemokine gene expression. The activity of C/EBPß is augmented by a transient increase in activity of NF-κB, and both transcription factors act independently on the chemokine genes, CCL3 and CCL4. Thus, resistin stimulates CCL3 and CCL4 through combinatorial regulation of C/EBPß and NF-κB in chondrocytes.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Quimiocinas/genética , FN-kappa B/metabolismo , Resistina/farmacología , Regulación hacia Arriba/efectos de los fármacos , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Cartílago Articular/citología , Células Cultivadas , Quimiocina CCL3/genética , Quimiocina CCL3/metabolismo , Quimiocina CCL4/genética , Quimiocina CCL4/metabolismo , Quimiocinas/metabolismo , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Humanos , Inmunohistoquímica , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/metabolismo
14.
J Exp Med ; 204(9): 2233-9, 2007 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-17724128

RESUMEN

Toll-like receptors (TLRs) recognize a variety of microbial components and mediate downstream signal transduction pathways that culminate in the activation of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein (MAP) kinases. Trib1 is reportedly involved in the regulation of NF-kappaB and MAP kinases, as well as gene expression in vitro. To clarify the physiological function of Trib1 in TLR-mediated responses, we generated Trib1-deficient mice by gene targeting. Microarray analysis showed that Trib1-deficient macrophages exhibited a dysregulated expression pattern of lipopolysaccharide-inducible genes, whereas TLR-mediated activation of MAP kinases and NF-kappaB was normal. Trib1 was found to associate with NF-IL6 (also known as CCAAT/enhancer-binding protein beta). NF-IL6-deficient cells showed opposite phenotypes to those in Trib1-deficient cells in terms of TLR-mediated responses. Moreover, overexpression of Trib1 inhibited NF-IL6-dependent gene expression by down-regulating NF-IL6 protein expression. In contrast, Trib1-deficient cells exhibited augmented NF-IL6 DNA-binding activities with increased amounts of NF-IL6 proteins. These results demonstrate that Trib1 is a negative regulator of NF-IL6 protein expression and modulates NF-IL6-dependent gene expression in TLR-mediated signaling.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/genética , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/genética , Animales , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/deficiencia , Femenino , Expresión Génica/efectos de los fármacos , Perfilación de la Expresión Génica , Inmunidad , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos C57BL , Unión Proteica/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
15.
Biochem Biophys Res Commun ; 431(2): 192-6, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23313487

RESUMEN

Recently, it was reported that naturally occurring pentacyclic triterpenoids such as ursolic acid have anti-adiposity property. We studied if acetyl-keto-ß-boswellic acid (AKBA), an established anti-inflammation and anti-cancer pentacyclic triterpenoid which has similar chemical structure to ursolic acid, may modulate adipocyte phenotype. 3T3-L1 murine adipocytes and human subcutaneous adipocytes were treated with AKBA in different concentrations in vitro. AKBA triggered significant lipolysis in 3T3-L1 adipocytes as shown by reduced neutral lipids in cytosol and increased free fatty acids in culture medium. Increased lipolysis by AKBA was accompanied by up-regulation of lipolytic enzymes, adipocyte triglyceride lipase (ATGL) and hormone sensitive lipase (HSL), and a decreased expression of lipid droplet stability regulator perilipin. In addition, AKBA treatment reduced phenotypic markers of mature adipocyte aP2, adiponectin and glut-4 in mature adipocytes. Further studies revealed that AKBA down-regulated PPAR-γ and C/EBP-α expression in a dose and temporal dependent manner in mature adipocytes. In human adipocytes, AKBA likewise mobilized lipolysis accompanied by down-regulation of PPAR-γ2 expression and loss of phenotypic markers of mature adipocytes.


Asunto(s)
Adipocitos/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Lipólisis/efectos de los fármacos , Triterpenos/farmacología , Células 3T3-L1 , Animales , Biomarcadores/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/biosíntesis , Células Cultivadas , Humanos , Ratones , PPAR gamma/antagonistas & inhibidores , PPAR gamma/biosíntesis , Perilipina-1 , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/biosíntesis
16.
Cell Biol Int ; 37(9): 905-16, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23589423

RESUMEN

FoxO1 and C/EBPß are important transcription factors during adipogenic differentiation; however, the potential mechanisms of their inter-regulation in adipogenesis remain unknown. We found that FoxO1 and C/EBPß are abundant in adipose tissues of 3- and 180-day pigs at the mRNA and protein levels. During porcine preadipocyte differentiation, C/EBPß expression increases to the peak at Day 2 and then decreases. In contrast, FoxO1 is lowest on Day 2 and gradually increases. Knockdown of FoxO1 or C/EBPß with lentivirus-mediated shRNA enhances or inhibits lipid accumulation and adipogenic maker (C/EBPα and aP2) expression, respectively. FoxO1 depletion causes a mild decrease of C/EBPß protein level, and C/EBPß interference also inhibits the expression of FoxO1 protein. Knockdown of both FoxO1 and C/EBPß promotes lipid accumulation at Day 8, and increases the adipogenic markers during differentiation in comparison with the controls and the FoxO1 knockdown alone. Co-immunoprecipitation experiments indicate that FoxO1 binds to C/EBPß in adipose tissues and in vitro. In conclusion, the results suggest that FoxO1 and C/EBPß regulate preadipocyte adipogenesis possibly through C/EBPß â†’ FoxO1 → C/EBPß feedback regulatory loop and FoxO1-C/EBPß protein complex.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/genética , Tejido Adiposo/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/genética , Retroalimentación Fisiológica , Factores de Transcripción Forkhead/genética , Adipocitos/citología , Tejido Adiposo/citología , Animales , Animales Recién Nacidos , Secuencia de Bases , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Vectores Genéticos , Lentivirus/genética , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Porcinos
17.
Biochemistry ; 51(5): 952-62, 2012 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-22242598

RESUMEN

Transcription factor C/EBPß is involved in several cellular processes, such as proliferation, differentiation, and energy metabolism. This factor exerts its activity through recruitment of different proteins or protein complexes, including the ATP-dependent chromatin remodeling complex SWI/SNF. The C/EBPß protein is found as three major isoforms, C/EBPß1, -2, and -3. They are generated by translation at alternative AUG initiation codons of a unique mRNA, C/EBPß1 being the full-length isoform. It has been found that C/EBPß1 participates in terminal differentiation processes. Conversely, C/EBPß2 and -3 promote cell proliferation and are involved in malignant progression in a number of tissues. The mechanisms by which C/EBPß2 and -3 promote cell proliferation and tumor progression are not fully understood. In this work, we sought to identify proteins interacting with hC/EBPß using a proteomics approach. We found that all three isoforms interact with hSNF2H and hACF, components of ACF and CHRAC chromatin remodeling complexes, which belong to the imitation switch subfamily. Additional protein-protein interaction studies confirmed this finding and also showed that hC/EBPß directly interacts with hACF1. By overexpressing hC/EBPß, hSNF2H, and hACF1 in HepG2 cells and analyzing variations in expression of cyclin D1 and other C/EBPß target genes, we observed a functional interaction between C/EBPß and SNF2H/ACF1, characterized mainly by suppression of C/EBPß transactivation activity in the presence of SNF2H and ACF1. Consistent with these findings, induction of differentiation of HepG2 cells by 1% DMSO was accompanied by a reduction in the level of cyclin D1 expression and the appearance of hC/EBPß, hSNF2H, and hACF1 on the promoter region of this gene.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/química , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ensamble y Desensamble de Cromatina , Genes de Cambio , Mapeo de Interacción de Proteínas , Proteómica/métodos , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Células HeLa , Células Hep G2 , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/química , Transactivadores/química , Transactivadores/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo
18.
Biochim Biophys Acta ; 1813(8): 1395-403, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21539866

RESUMEN

Inducible nitric oxide synthase (iNOS) and nitric oxide (NO) involve many pathophysiologic conditions. The expression of iNOS is regulated at multiple stages. Presently, the regulatory details of iNOS signaling are still unclear. This study aimed to investigate the regulatory role of C/EBPα and C/EBPß in iNOS signaling pathway. By employing the techniques such as EMSA, ChIP assay, site-directed mutagenesis, and siRNA silencing, the relationship between iNOS and C/EBPα/C/EBPß in rat hepatocytes was clarified. iNOS promoter was the direct transcriptional targets of the C/EBPα, C/EBPß, and NF-κB binding proteins. There was the interactive influence between NF-κB and C/EBPα/C/EBPß. The expression of iNOS was modulated by C/EBPα/C/EBPß transcription factors. Moreover, the iNOS expression mediated glycochenodeoxycholate (GCDC)-induced apoptosis in hepatocytes. C/EBPα/C/EBPß binding proteins could affect the GCDC-induced apoptosis through iNOS cascade. These findings indicate that C/EBPα and C/EBPß regulate the iNOS expression, which may further modify cell responses such as apoptosis and cell survival.


Asunto(s)
Apoptosis/fisiología , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Secuencia de Bases , Sitios de Unión/genética , Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteínas Portadoras/metabolismo , Cartilla de ADN/genética , Ácido Glicoquenodesoxicólico/farmacología , Hepatocitos/efectos de los fármacos , Técnicas In Vitro , Mutagénesis Sitio-Dirigida , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transducción de Señal/fisiología
19.
BMC Complement Altern Med ; 12: 230, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23181522

RESUMEN

BACKGROUND: Centipede grass (CG) originates from China and South America and is reported to contain several C-glycosyl flavones and phenolic constituents, including maysin and luteolin derivatives. This study aimed to investigate, for the first time, the antiobesity activity of CG and its potential molecular mechanism in 3T3-L1 cells. METHODS: To study the effect of CG on adipogenesis, differentiating 3T3-L1 cells were treated every day with CG at various concentrations (0-100 µg/ml) for six days. Oil-red O staining and triglyceride content assay were performed to determine the lipid accumulation in 3T3-L1 cells. The expression of mRNAs or proteins associated with adipogenesis was measured using RT-PCR and Western blotting analysis. We examined the effect of CG on level of phosphorylated Akt in 3T3-L1 cells treated with CG at various concentration s during adipocyte differentiation. RESULTS: Differentiation was investigated with an Oil-red O staining assay using CG-treated 3T3-L1 adipocytes. We found that CG suppressed lipid droplet formation and adipocyte differentiation in 3T3-L1 cells in a dose-dependent manner. Treatment of the 3T3-L1 adipocytes with CG resulted in an attenuation of the expression of adipogenesis-related factors and lipid metabolic genes. The expression of C/EBPα and PPARγ, the central transcriptional regulators of adipogenesis, was decreased by the treatment with CG. The expression of genes involved in lipid metabolism, aP2 were significantly inhibited following the CG treatment. Moreover, the CG treatment down-regulated the phosphorylation levels of Akt and GSK3ß. CONCLUSIONS: Taken collectively, these data indicated that CG exerts antiadipogenic activity by inhibiting the expression of C/EBPß, C/EBPα, and PPARγ and the Akt signaling pathway in 3T3-L1 adipocytes.


Asunto(s)
Adipogénesis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/antagonistas & inhibidores , Expresión Génica/efectos de los fármacos , PPAR gamma/antagonistas & inhibidores , Extractos Vegetales/farmacología , Poaceae , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Fármacos Antiobesidad/farmacología , Proteína alfa Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Flavonoides/farmacología , Glucósidos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Luteolina/farmacología , Ratones , Fosforilación , Polifenoles/farmacología , Transducción de Señal
20.
Mol Cancer Ther ; 21(11): 1632-1644, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36121385

RESUMEN

CCAAT/enhancer binding protein ß (C/EBPß) is a basic leucine zipper (bZIP) family transcription factor, which is upregulated or overactivated in many cancers, resulting in a gene expression profile that drives oncogenesis. C/EBPß dimerization regulates binding to DNA at the canonical TTGCGCAA motif and subsequent transcriptional activity, suggesting that disruption of dimerization represents a powerful approach to inhibit this previously "undruggable" oncogenic target. Here we describe the mechanism of action and antitumor activity of ST101, a novel and selective peptide antagonist of C/EBPß that is currently in clinical evaluation in patients with advanced solid tumors. ST101 binds the leucine zipper domain of C/EBPß, preventing its dimerization and enhancing ubiquitin-proteasome dependent C/EBPß degradation. ST101 exposure attenuates transcription of C/EBPß target genes, including a significant decrease in expression of survival, transcription factors, and cell-cycle-related proteins. The result of ST101 exposure is potent, tumor-specific in vitro cytotoxic activity in cancer cell lines including glioblastoma, breast, melanoma, prostate, and lung cancer, whereas normal human immune and epithelial cells are not impacted. Further, in mouse xenograft models ST101 exposure results in potent tumor growth inhibition or regression, both as a single agent and in combination studies. These data provide the First Disclosure of ST101, and support continued clinical development of ST101 as a novel strategy for targeting C/EBPß-dependent cancers.


Asunto(s)
Antineoplásicos , Proteína beta Potenciadora de Unión a CCAAT , Animales , Humanos , Ratones , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Unión Proteica , Antineoplásicos/farmacología , Neoplasias Experimentales/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA