Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-36293494

RESUMEN

Patients with high-risk non-metastatic renal cell carcinoma (RCC) are at risk of metastatic relapse following nephrectomy. Cabozantinib (CZ), a potent multitarget tyrosine kinase inhibitor, interferes with angiogenesis and immunosuppression associated with surgery-induced metastasis. Here, we explored the therapeutic potential of CZ-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (CZ-PLGA-NPs) as an adjuvant strategy for targeting post-nephrectomy metastasis. A clinically relevant subline recapitulating post-nephrectomy lung metastasis of high-risk human RCC, namely Renca-SRLu5-Luc, was established through in vivo serial selection of luciferase-expressing murine RCC Renca-Luc cells. CZ was encapsulated into PLGA-NPs via the conventional single emulsion technique. The multifaceted preclinical antimetastatic efficacy of CZ-PLGA-NPs was assessed in Renca-SRLu5-Luc cells. CZ-PLGA-NPs with a smooth surface displayed desirable physicochemical properties, good CZ encapsulation efficiency, as well as controlled and sustained CZ release. CZ-PLGA-NPs exhibited remarkable dose-dependent toxicity against Renca-SRLu5-Luc cells by inducing G2/M cell cycle arrest and apoptosis. CZ-PLGA-NPs attenuated in vitro colony formation, migration, and invasion by abrogating AKT and ERK1/2 activation. An intravenous injection of CZ-PLGA-NPs markedly reduced lung metastatic burden and prolonged lifespan with favorable safety in the Renca-SRLu5-Luc experimental lung metastasis model. The novel CZ-PLGA-NPs system with multifaceted antimetastatic effects and alleviating off-target toxicity potential is a promising adjunctive agent for patients with surgically resected high-risk RCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Neoplasias Pulmonares , Nanopartículas , Humanos , Ratones , Animales , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ácido Láctico/química , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/cirugía , Portadores de Fármacos/química , Emulsiones , Proteínas Proto-Oncogénicas c-akt , Nanopartículas/química , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/cirugía , Inhibidores de Proteínas Quinasas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/cirugía , Tamaño de la Partícula
2.
Curr Issues Mol Biol ; 43(1): 226-239, 2021 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-34071865

RESUMEN

Due to the highly immunogenic nature of renal cell carcinoma (RCC), the tumor microenvironment (TME) is enriched with various innate and adaptive immune subsets. In particular, gamma-delta (γδ) T cells can act as potent attractive mediators of adoptive cell transfer immunotherapy because of their unique properties such as non-reliance on major histocompatibility complex expression, their ability to infiltrate human tumors and recognize tumor antigens, relative insensitivity to immune checkpoint molecules, and broad tumor cytotoxicity. Therefore, it is now critical to better characterize human γδ T-cell subsets and their mechanisms in RCCs, especially the stage of differentiation. In this study, we aimed to identify γδ T cells that might have adaptive responses against RCC progression. We characterized γδ T cells in peripheral blood and tumor-infiltrating lymphocytes (TILs) in freshly resected tumor specimens from 20 RCC patients. Furthermore, we performed a gene set enrichment analysis on RNA-sequencing data from The Cancer Genome Atlas (TCGA) derived from normal kidneys and RCC tumors to ascertain the association between γδ T-cell infiltration and anti-cancer immune activity. Notably, RCC-infiltrating CD3low Vγ9Vδ1 T cells with a terminally differentiated effector memory phenotype with up-regulated activation/exhaustion molecules were newly detected as predominant TILs, and the cytotoxic activity of these cells against RCC was confirmed in vitro. In an additional analysis of the TCGA RCC dataset, γδ T-cell enrichment scores correlated strongly with those for CTLs, Th1 cells, "exhausted" T cells, and M1 macrophages, suggesting active involvement of γδ T cells in anti-tumor rather than pro-tumor activity, and Vδ1 cells were more abundant than Vδ2 or Vδ3 cells in RCC tumor samples. Thus, we posit that Vγ9Vδ1 T cells may represent an excellent candidate for adoptive immunotherapy in RCC patients with a high risk of relapse after surgery.


Asunto(s)
Complejo CD3/inmunología , Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Complejo CD3/genética , Complejo CD3/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Citometría de Flujo/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Persona de Mediana Edad , RNA-Seq/métodos , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
3.
Sci Rep ; 11(1): 10145, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33980886

RESUMEN

Glioblastoma (GBM) is an aggressive brain tumor with a strong tendency of relapse and resistance to chemotherapy, but we currently lack non-toxic agents that effectively treat GBM. In this study, high-throughput screening of FDA-approved drugs was performed to identify safe and effective molecules and test their effect on GBM cell lines, LN229, U87 and T98G. Cough suppressants, oxelaidin and butamirate inhibited GBM growth. A Ras family GTPase, Ras-related associated with diabetes (RRAD), contributes to activation of STAT3, which is essential for survival and growth of many cancer types. Interestingly, oxelaidin and butamirate did not affect proliferation in RRAD negative GBM cells. Docking simulation analyses revealed selective interactions between oxelaidin and RRAD. The mechanism by which butamirate and oxelaidin inhibits GBM cell growth involves the suppression of STAT3 transcriptional activity, leading to down-regulation of cyclin D1 and survivin. In addition, components of RRAD-associated signaling cascades, including p-EGFR, p-Akt, and p-STAT3, were inhibited upon oxelaidin treatment. Intraperitoneal administration of oxelaidin or butamirate markedly suppressed tumor growth in a glioblastoma xenograft mouse model without significant adverse effects. Our collective findings indicate that oxelaidin and butamirate exert anti-tumor effects in glioblastoma, supporting its utility as a novel therapeutic candidate for glioblastoma.


Asunto(s)
Antineoplásicos/farmacología , Antitusígenos/farmacología , Fenilbutiratos/farmacología , Animales , Antineoplásicos/química , Antitusígenos/química , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Reposicionamiento de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Ratones , Modelos Moleculares , Fenilbutiratos/química , Unión Proteica , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Sci Rep ; 11(1): 21984, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753990

RESUMEN

Abnormal expression of insulin gene enhancer-binding protein 1 (ISL1) has been demonstrated to be closely associated with cancer development and progression in several cancers. However, little is known about ISL1 expression in metastatic castration-resistant prostate cancer (CRPC). ISL1 has also been recognized as a positive modulator of epithelial-mesenchymal transition (EMT). In this study, we focused on ISL1 which showed maximum upregulation at the mRNA level in the enzalutamide-resistant cell line. Accordingly, we found that ISL1 was overexpressed in enzalutamide-resistant C4-2B cells and its expression was significantly related to EMT. Our findings reveal the important role of ISL1 in androgen receptor (AR)-dependent prostate cancer cell growth; ISL1 knockdown reduced the AR activity and cell growth. ISL1 knockdown using small-interfering RNA inhibited AR, PSA, and EMT-related protein expression in C4-2B ENZR cells. In addition, knock-down ISL1 reduced the levels of AKT and p65 phosphorylation in C4-2B ENZR cells and these suggest that knock-down ISL1 suppresses EMT in part by targeting the AKT/NF-κB pathway. Further, ISL1 downregulation could effectively inhibit tumor growth in a human CRPC xenograft model. Together, the present study shows that downregulation of ISL1 expression is necessary for overcoming enzalutamide resistance and improving the survival of CRPC patients.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Proteínas con Homeodominio LIM/fisiología , Nitrilos/uso terapéutico , Feniltiohidantoína/uso terapéutico , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/fisiopatología , Factores de Transcripción/fisiología , Antagonistas de Andrógenos/farmacología , Animales , Benzamidas/farmacología , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Humanos , Masculino , Ratones , Nitrilos/farmacología , Feniltiohidantoína/farmacología , Neoplasias de la Próstata Resistentes a la Castración/patología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Cell Biol ; 26(12): 4553-63, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738321

RESUMEN

Activating signal cointegrator 2 (ASC-2) is a transcriptional coactivator of many nuclear receptors (NRs) and other transcription factors and contains two NR-interacting LXXLL motifs (NR boxes). In the pancreas, ASC-2 is expressed only in the endocrine cells of the islets of Langerhans, but not in the exocrine cells. Thus, we examined the potential role of ASC-2 in insulin secretion from pancreatic beta-cells. Overexpressed ASC-2 increased glucose-elicited insulin secretion, whereas insulin secretion was decreased in islets from ASC-2+/- mice. DN1 and DN2 are two dominant-negative fragments of ASC-2 that contain NR boxes 1 and 2, respectively, and block the interactions of cognate NRs with the endogenous ASC-2. Primary rat islets ectopically expressing DN1 or DN2 exhibited decreased insulin secretion. Furthermore, relative to the wild type, ASC-2+/- mice showed reduced islet mass and number, which correlated with increased apoptosis and decreased proliferation of ASC-2+/- islets. These results suggest that ASC-2 regulates insulin secretion and beta-cell survival and that the regulatory role of ASC-2 in insulin secretion appears to involve, at least in part, its interaction with NRs via its two NR boxes.


Asunto(s)
Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Islotes Pancreáticos/citología , Islotes Pancreáticos/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Apoptosis , Secuencia de Bases , Proliferación Celular , ADN/genética , Expresión Génica , Glucosa/metabolismo , Glucosa/farmacología , Técnicas In Vitro , Secreción de Insulina , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Islotes Pancreáticos/efectos de los fármacos , Ratones , Ratones Mutantes , Ratones Transgénicos , Coactivadores de Receptor Nuclear , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas OLETF , Ratas Mutantes , Ratas Sprague-Dawley
6.
Mol Endocrinol ; 19(7): 1711-9, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15764585

RESUMEN

Activating signal cointegrator-2 (ASC-2) is a recently isolated transcriptional coactivator protein for a variety of different transcription factors, including many members of the nuclear receptor superfamily. In this report, we demonstrate that ASC-2 also serves as a coactivator of the xenobiotic nuclear receptor constitutive androstane receptor (CAR). First, transcriptional activation by CAR was enhanced by cotransfected ASC-2 in CV-1 and HeLa cells. In contrast, CAR transactivation was significantly impaired in HepG2 cells stably expressing specific small interfering RNA directed against ASC-2. Consistent with these results, chromatin immunoprecipitation experiments revealed that ASC-2 is recruited to the known CAR target genes in a ligand-dependent manner. Secondly, CAR specifically interacted with the first LXXLL motif of ASC-2, and these interactions were stimulated by CAR agonist 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene and repressed by CAR inverse agonist androstanol, suggesting that this motif may mediate the interactions of ASC-2 and CAR in vivo. In support of this idea, DN1, a fragment of ASC-2 encompassing the first LXXLL motif, suppressed CAR transactivation, and coexpressed ASC-2 but not other LXXLL-type coactivators such as thyroid hormone receptor-associated protein 220 reversed this repression. Finally, CAR was recently found to play a pivotal role in effecting the severe acetaminophen-induced liver damage. Interestingly, transgenic mice expressing DN1 were resistant to the acetaminophen-induced hepatotoxicity and expression of a series of the known CAR target genes was specifically repressed in these transgenic mice. Taken together, these results strongly suggest that ASC-2 is a bona fide coactivator of the xenobiotic nuclear receptor CAR and mediate the specific xenobiotic response by CAR in vivo.


Asunto(s)
Acetaminofén/toxicidad , Péptidos y Proteínas de Señalización Intracelular/fisiología , Hígado/efectos de los fármacos , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/fisiología , Factores de Transcripción/metabolismo , Acetaminofén/metabolismo , Secuencias de Aminoácidos , Animales , Hidrocarburo de Aril Hidroxilasas/genética , Inmunoprecipitación de Cromatina , Receptor de Androstano Constitutivo , Citocromo P-450 CYP2B6 , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/genética , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Transgénicos , Coactivadores de Receptor Nuclear , Oxidorreductasas N-Desmetilantes/genética , Mapeo de Interacción de Proteínas , Interferencia de ARN , Receptores Citoplasmáticos y Nucleares/genética , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Xenobióticos/metabolismo , Xenobióticos/toxicidad
7.
Mol Cancer Ther ; 13(12): 3049-61, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25313011

RESUMEN

Glioblastoma multiforme (GBM) is an extremely aggressive brain cancer with a median survival of less than 2 years. GBM is characterized by abnormal activation of receptor tyrosine kinase and constitutively activated STAT3. Although EGFR phosphorylation and STAT3 activation are essential for the maintenance of GBM cancer stem cells, the molecular mechanism underlying endosome-mediated STAT3 activation is not fully understood. In the current study, we showed that GTP-binding protein RRAD (RAS associated with diabetes, RAD) physically associates with EGFR, and EEA1, enhancing the stability and endosome-associated nuclear translocation of EGFR. Functionally, RRAD contributes to the activation of STAT3 and expression of the stem cell factors OCT4, NANOG, and SOX2, thereby enhancing self-renewing ability, tumor sphere formation, EMT, and in vivo tumorigenesis. Most importantly, RRAD contributes to poor survival in patients with GBM. RRAD expression is correlated with temozolomide resistance, and, conversely, depletion of RRAD leads to sensitization of highly temozolomide-resistant GBM cells. Our data collectively support a novel function of RRAD in STAT3 activation and provide evidence that RRAD acts as a positive regulator in the EGFR signaling pathway. These results demonstrate a critical role for RRAD in GBM tumorigenesis and provide a rationale for the development of pharmacologic inhibitors of RRAD in GBM.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas ras/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Dacarbazina/farmacología , Endosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Pronóstico , Estabilidad Proteica , Transporte de Proteínas , Esferoides Celulares , Temozolomida , Células Tumorales Cultivadas , Proteínas ras/genética
8.
Leuk Res ; 36(9): 1172-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22658652

RESUMEN

To understand the molecular mechanism(s) underlying bortezomib resistance, we sought to identify potential target genes that were differentially expressed in bortezomib-resistant leukemia cells versus parental controls. Microarray analysis revealed that the mRNA levels of Rad (Ras associated with diabetes) were higher in the bortezomib-resistant Jurkat (Jurkat-R) cells than in the parental control cells. The importance of Rad for bortezomib resistance was supported by three observations. First, Rad knockdown overcame bortezomib resistance and induced mitochondrial apoptosis via Noxa/Bcl-2 modulation. Second, Rad decreased cell death in response to bortezomib. Third, leukemia and lymphoma cell lines (K-562, Raji, IM-9 and Jurkat-R) with elevated Rad expression levels showed higher degrees of bortezomib resistance versus those (Sup-B15, JVM-2, U266 and Jurkat) with low Rad expression levels (r=0.48, P=0.0004). Thus, Rad over expression could be a molecular target to improve bortezomib sensitivity in human leukemia and lymphoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Borónicos/uso terapéutico , Leucemia/genética , Linfoma/genética , Mitocondrias/efectos de los fármacos , Pirazinas/uso terapéutico , Proteínas ras/genética , Antineoplásicos/uso terapéutico , Apoptosis/genética , Bortezomib , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Células Jurkat , Células K562 , Leucemia/tratamiento farmacológico , Leucemia/patología , Linfoma/tratamiento farmacológico , Linfoma/patología , Mitocondrias/patología , ARN Interferente Pequeño/farmacología , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología , Proteínas ras/antagonistas & inhibidores , Proteínas ras/fisiología
9.
FEBS Lett ; 584(8): 1469-75, 2010 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-20211623

RESUMEN

Previous studies have shown that testisin promotes malignant transformation in cancer cells. To define the mechanism of testisin-induced carcinogenesis, we performed yeast two-hybrid analysis and identified maspin, a tumor suppressor protein, as a testisin-interacting molecule. The direct interaction and cytoplasmic co-localization of testisin with maspin was confirmed by immunoprecipitation and confocal analysis, respectively. In cervical cancer cells, maspin modulated cell death and invasion; however, these effects were inhibited by testisin in parallel experiments. Of interest, the doxorubicin resistance was dramatically reduced by testisin knockdown (P=0.016). Moreover, testisin was found to be over-expressed in cervical cancer samples as compared to matched normal cervical tissues. Thus, we postulate that testisin may promote carcinogenesis by inhibiting tumor suppressor activity of maspin.


Asunto(s)
Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Serpinas/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Apoptosis , Caspasa 3/metabolismo , Línea Celular Tumoral , Activación Enzimática , Femenino , Proteínas Ligadas a GPI , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Invasividad Neoplásica , Unión Proteica , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
10.
Cancer Res ; 70(11): 4357-65, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20460530

RESUMEN

Ras associated with diabetes (Rad) is a Ras-related GTPase that promotes cell growth by accelerating cell cycle transitions. Rad knockdown induced cell cycle arrest and premature senescence without additional cellular stress in multiple cancer cell lines, indicating that Rad expression might be critical for the cell cycle in these cells. To investigate the precise function of Rad in this process, we used human Rad as bait in a yeast two-hybrid screening system and sought Rad-interacting proteins. We identified the Grap2 and cyclin D interacting protein (GCIP)/DIP1/CCNDBP1/HHM, a cell cycle-inhibitory molecule, as a binding partner of Rad. Further analyses revealed that Rad binds directly to GCIP in vitro and coimmunoprecipitates with GCIP from cell lysates. Rad translocates GCIP from the nucleus to the cytoplasm, thereby inhibiting the tumor suppressor activity of GCIP, which occurs in the nucleus. Furthermore, in the presence of Rad, GCIP loses its ability to reduce retinoblastoma phosphorylation and inhibit cyclin D1 activity. The function of Rad in transformation is also evidenced by increased telomerase activity and colony formation according to Rad expression level. In vivo tumorigenesis analyses revealed that tumors derived from Rad knockdown cells were significantly smaller than those from control cells (P = 0.0131) and the preestablished tumors are reduced in size after the injection of siRad (P = 0.0064). Therefore, we propose for the first time that Rad may promote carcinogenesis at least in part by inhibiting GCIP-mediated tumor suppression.


Asunto(s)
Doxorrubicina/farmacología , Neoplasias/patología , Factores de Transcripción/fisiología , Proteínas ras/fisiología , Animales , Línea Celular Tumoral , Senescencia Celular/efectos de los fármacos , Senescencia Celular/fisiología , Ciclina D/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos , Femenino , Células HeLa , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fosforilación , Proteína de Retinoblastoma/metabolismo , Telomerasa/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteínas ras/biosíntesis , Proteínas ras/genética
11.
Mol Endocrinol ; 23(7): 966-74, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19342446

RESUMEN

Activating signal cointegrator-2 (ASC-2) functions as a transcriptional coactivator of many nuclear receptors and also plays important roles in the physiology of the liver and pancreas by interacting with liver X receptors (LXRs), which antagonize the development of atherosclerosis. This study was undertaken to establish the specific function of ASC-2 in macrophages and atherogenesis. Intriguingly, ASC-2 was more highly expressed in macrophages than in the liver and pancreas. To inhibit LXR-specific activity of ASC-2, we used DN2, which contains the C-terminal LXXLL motif of ASC-2 and thereby acts as an LXR-specific, dominant-negative mutant of ASC-2. In DN2-overexpressing transgenic macrophages, cellular cholesterol content was higher and cholesterol efflux lower than in control macrophages. DN2 reduced LXR ligand-dependent increases in the levels of ABCA1, ABCG1, and apolipoprotein E (apoE) transcripts as well as the activity of luciferase reporters driven by the LXR response elements (LXREs) of ABCA1, ABCG1, and apoE genes. These inhibitory effects of DN2 were reversed by overexpression of ASC-2. Chromatin immunoprecipitation analysis demonstrated that ASC-2 was recruited to the LXREs of the ABCA1, ABCG1, and apoE genes in a ligand-dependent manner and that DN2 interfered with the recruitment of ASC-2 to these LXREs. Furthermore, low-density lipoprotein receptor (LDLR)-null mice receiving bone marrow transplantation from DN2-transgenic mice showed accelerated atherogenesis when administered a high-fat diet. Taken together, these results indicate that suppression of the LXR-specific activity of ASC-2 results in both defective cholesterol metabolism in macrophages and accelerated atherogenesis, suggesting that ASC-2 is an antiatherogenic coactivator of LXRs in macrophages.


Asunto(s)
Aterosclerosis/genética , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Macrófagos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Transactivadores/fisiología , Transportador 1 de Casete de Unión a ATP , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Apolipoproteínas E/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipoproteínas/genética , Receptores X del Hígado , Masculino , Ratones , Ratones Noqueados , Proteínas Mutantes/metabolismo , Proteínas Mutantes/fisiología , Coactivadores de Receptor Nuclear , Receptores Nucleares Huérfanos , Unión Proteica , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Transactivadores/genética , Transactivadores/metabolismo , Activación Transcripcional/genética , Activación Transcripcional/fisiología
12.
Biotechnol Lett ; 26(9): 699-703, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15195967

RESUMEN

RNA interference has become a powerful tool for silencing of gene expression in mammals and plants. To determine the effect of Smad3 on transforming growth factor-beta signaling, we constructed a small interfering RNA (siRNA) targeted to Smad3. This siRNA inhibited expression of the endogenous Smad3 leading to the prevention of nuclear localization of Smad3. Further, Smad3 siRNA prevented not only anti-proliferative activity of TGF-beta1 but also TGF-beta1-inducible promoter activity.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ARN Interferente Pequeño , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Citoplasma/metabolismo , Luciferasas/metabolismo , Ratones , Microscopía Fluorescente , Plásmidos/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteína smad3 , Timidina/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA