Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Biochem ; 118(8): 2219-2230, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28067406

RESUMEN

Ninjurin1 (Ninj1) is a cell surface protein known as a homophilic adhesion molecule. Previous studies have shown a trans-interaction of Ninj1 between immune cells and endothelial cells; however, little is known about Ninj1 modification and structure in the cis-interaction. We showed that Ninj1 assembles into a homomeric complex via a cis-interaction mediated by the intracellular region and N-glycosylation at Asn60 . We identified cis-interaction between Ninj1 proteins using CFP- and YFP-tagged Ninj1 by Förster resonance energy transfer using a confocal microscope and fluorescence-activated cell sorter. We further observed the Ninj1 homomeric complexes composed of two to six monomeric Ninj1 molecules by a formaldehyde cross-linking assay. Co-immunoprecipitation assays with epitope-tagged truncated Ninj1 suggested that the intracellular region encompassing Leu101 -Ala110 participates in Ninj1 homomer assembly. Ninj1 N-glycosylation was characterized by treatment of tunicamycin and substitution of Asn to Gln or Ala. Fluorescence-activated cell sorting-based Förster resonance energy transfer assays further demonstrated that N-glycosylation is indispensable for the Ninj1 cis-interaction, and a formaldehyde cross-linking assay confirmed that interruption of N-glycosylation by Asn substitution disrupted Ninj1 homomeric complex formation. In silico analysis revealed that Ninj1 is highly conserved in vertebrates and that the conserved sequence contains an N-glycosylation motif and cis-interacting intracellular region, which participate in Ninj1 homomer assembly. Taken together, these data show that Ninj1 assembles into a homomeric protein complex and that N-glycosylation is a prerequisite for Ninj1 homomer assembly. J. Cell. Biochem. 118: 2219-2230, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/química , Moléculas de Adhesión Celular Neuronal/metabolismo , Factores de Crecimiento Nervioso/química , Factores de Crecimiento Nervioso/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Glicosilación/efectos de los fármacos , Células HEK293 , Humanos , Immunoblotting , Inmunoprecipitación , Masculino , Ratones , Ratones Noqueados , Microscopía Confocal , Células 3T3 NIH , Factores de Crecimiento Nervioso/genética , Multimerización de Proteína/efectos de los fármacos , Multimerización de Proteína/genética , Tunicamicina/farmacología
2.
J Biol Chem ; 289(6): 3328-38, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24347169

RESUMEN

Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26-37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Movimiento Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Macrófagos/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Células de la Médula Ósea/patología , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Macrófagos/patología , Ratones , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Esclerosis Múltiple/terapia , Factores de Crecimiento Nervioso/antagonistas & inhibidores , Factores de Crecimiento Nervioso/genética
3.
J Biol Chem ; 289(32): 21926-36, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24917672

RESUMEN

Ninjurin1 is involved in the pathogenesis of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, by mediating leukocyte extravasation, a process that depends on homotypic binding. However, the precise regulatory mechanisms of Ninjurin1 during inflammation are largely undefined. We therefore examined the pro-migratory function of Ninjurin1 and its regulatory mechanisms in macrophages. Interestingly, Ninjurin1-deficient bone marrow-derived macrophages exhibited reduced membrane protrusion formation and dynamics, resulting in the impairment of cell motility. Furthermore, exogenous Ninjurin1 was distributed at the membrane of filopodial structures in Raw264.7 macrophage cells. In Raw264.7 cells, RNA interference of Ninjurin1 reduced the number of filopodial projections, whereas overexpression of Ninjurin1 facilitated their formation and thus promoted cell motility. Ninjurin1-induced filopodial protrusion formation required the activation of Rac1. In Raw264.7 cells penetrating an MBEC4 endothelial cell monolayer, Ninjurin1 was localized to the membrane of protrusions and promoted their formation, suggesting that Ninjurin1-induced protrusive activity contributed to transendothelial migration. Taking these data together, we conclude that Ninjurin1 enhances macrophage motility and consequent extravasation of immune cells through the regulation of protrusive membrane dynamics. We expect these findings to provide insight into the understanding of immune responses mediated by Ninjurin1.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/fisiología , Movimiento Celular/fisiología , Macrófagos/fisiología , Factores de Crecimiento Nervioso/fisiología , Animales , Adhesión Celular/fisiología , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Línea Celular , Membrana Celular/fisiología , Células Cultivadas , Células Endoteliales/fisiología , Técnicas de Silenciamiento del Gen , Inflamación/etiología , Inflamación/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Neuropéptidos/metabolismo , Seudópodos/fisiología , Interferencia de ARN , Proteína de Unión al GTP rac1/metabolismo
4.
J Pineal Res ; 55(3): 294-303, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23869429

RESUMEN

Aberrant expression of inducible nitric oxide synthase (iNOS) in macrophages, which has been reported to be suppressed by melatonin, has an important contribution in the development of pathological inflammation. Visfatin, an adipokine, regulates the expression of various inflammatory factors, leading to inflammation; however, the influence of visfatin on iNOS-driven processes in macrophages is unclear. Here, we report the assessment of the role of visfatin in the regulation of iNOS gene expression in macrophages. Our data show that the levels of iNOS protein in peritoneal macrophages as well as nitric oxide (NO) in blood plasma were significantly lower after lipopolysaccharide treatment in visfatin(+/-) mice than those in the WT mice. In addition, visfatin increases iNOS mRNA and protein levels in RAW 264.7 cells, along with increasing production of NO. The enhancement of iNOS expression was prevented by treating the cells with inhibitors of the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT3), nuclear factor (NF)-κB, extracellular signal-regulated kinase 1/2, and c-Jun N-terminal kinase pathways. Our results also show that visfatin-induced iNOS expression and NO production were significantly inhibited by melatonin, an effect that was closely associated with a reduction in phosphorylated JAK2/STAT3 levels and with the inhibition of p65 translocation into nucleus. In conclusion, our data show, for the first time, that melatonin suppresses visfatin-induced iNOS upregulation in macrophages by inhibiting the STAT3 and NF-κB pathways. Moreover, our data suggest that melatonin could be therapeutically useful for attenuating the development of visfatin-iNOS axis-associated diseases.


Asunto(s)
Antioxidantes/farmacología , Citocinas/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Macrófagos Peritoneales/enzimología , Melatonina/farmacología , Nicotinamida Fosforribosiltransferasa/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Óxido Nítrico/biosíntesis , Animales , Línea Celular , Citocinas/genética , Regulación Enzimológica de la Expresión Génica/genética , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Janus Quinasa 2/metabolismo , Janus Quinasa 3/genética , Janus Quinasa 3/metabolismo , Ratones , Ratones Mutantes , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Nicotinamida Fosforribosiltransferasa/genética , Óxido Nítrico/genética , Óxido Nítrico Sintasa de Tipo II/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
5.
Korean J Physiol Pharmacol ; 17(4): 291-7, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23946688

RESUMEN

Notch1 has been reported to be highly expressed in triple-negative and other subtypes of breast cancer. Mutant p53 (R280K) is overexpressed in MDA-MB-231 triple-negative human breast cancer cells. The present study aimed to determine whether the mutant p53 can be a potent transcriptional activator of the Notch1 in MDA-MB-231 cells, and explore the role of this mutant p53-Notch1 axis in curcumin-induced apoptosis. We found that curcumin treatment resulted in an induction of apoptosis in MDA-MB-231 cells, together with downregulation of Notch1 and its downstream target, Hes1. This reduction in Notch1 expression was determined to be due to the decreased activity of endogenous mutant p53. We confirmed the suppressive effect of curcumin on Notch1 transcription by performing a Notch1 promoter-driven reporter assay and identified a putative p53-binding site in the Notch1 promoter by EMSA and chromatin immunoprecipitation analysis. Overexpression of mutant p53 increased Notch1 promoter activity, whereas knockdown of mutant p53 by small interfering RNA suppressed Notch1 expression, leading to the induction of cellular apoptosis. Moreover, curcumin-induced apoptosis was further enhanced by the knockdown of Notch1 or mutant p53, but it was decreased by the overexpression of active Notch1. Taken together, our results demonstrate, for the first time, that Notch1 is a transcriptional target of mutant p53 in breast cancer cells and suggest that the targeting of mutant p53 and/or Notch1 may be combined with a chemotherapeutic strategy to improve the response of breast cancer cells to curcumin.

6.
Biochem Biophys Res Commun ; 428(4): 438-44, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23142597

RESUMEN

Ninjurin1 is known as an adhesion molecule promoting leukocyte trafficking under inflammatory conditions. However, the posttranslational modifications of Ninjurin1 are poorly understood. Herein, we defined the proteolytic cleavage of Ninjurin1 and its functions. HEK293T cells overexpressing the C- or N-terminus tagging mouse Ninjurin1 plasmid produced additional cleaved forms of Ninjurin1 in the lysates or conditioned media (CM). Two custom-made anti-Ninjurin1 antibodies, Ab(1-15) or Ab(139-152), specific to the N- or C-terminal regions of Ninjurin1 revealed the presence of its shedding fragments in the mouse liver and kidney lysates. Furthermore, Matrix Metalloproteinase (MMP) 9 was responsible for Ninjurin1 cleavage between Leu(56) and Leu(57). Interestingly, the soluble N-terminal Ninjurin1 fragment has structural similarity with well-known chemokines. Indeed, the CM from HEK293T cells overexpressing the GFP-mNinj1 plasmid was able to attract Raw264.7 cells in trans-well assay. Collectively, we suggest that the N-terminal ectodomain of mouse Ninjurin1, which may act as a chemoattractant, is cleaved by MMP9.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/química , Factores Quimiotácticos/química , Metaloproteinasa 9 de la Matriz/química , Factores de Crecimiento Nervioso/química , Secuencia de Aminoácidos , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Quimiocinas/química , Quimiocinas/genética , Quimiocinas/metabolismo , Factores Quimiotácticos/genética , Factores Quimiotácticos/metabolismo , Células HEK293 , Humanos , Riñón/metabolismo , Leucina/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
7.
Biochim Biophys Acta ; 1793(11): 1759-67, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19751774

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) acts as a mediator and biomarker in endothelial activation. We have recently shown that a novel adipokine visfatin promotes endothelial angiogenesis. The present study was to determine whether visfatin affects STAT3 activity and to explore the potential target gene(s). Here, we found that visfatin induced the activation of STAT3, as characterized by increased tyrosine phosphorylation, nuclear translocation, and DNA-binding activity in human endothelial cells. In addition, visfatin significantly upregulated mRNA and protein levels of endothelial interleukin-6 (IL-6), which was blocked by a specific inhibitor of STAT3 signaling and by the transfection of siRNA specific for STAT3. Furthermore, visfatin-induced angiogenesis was reduced by the inhibition of STAT3 signaling or neutralization of IL-6 function, as measured by tube formation, rat aortic ring assay, and mouse Matrigel plug assay. Taken together, our results provide the first example of STAT3-dependent endothelial IL-6 induction by visfatin and of the role of IL-6 in mediating visfatin-induced angiogenesis.


Asunto(s)
Citocinas/metabolismo , Endotelio Vascular/metabolismo , Interleucina-6/biosíntesis , Neovascularización Fisiológica/fisiología , Nicotinamida Fosforribosiltransferasa/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Células Cultivadas , Citocinas/farmacología , Endotelio Vascular/citología , Humanos , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Nicotinamida Fosforribosiltransferasa/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
8.
J Cell Biochem ; 110(1): 97-103, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20225274

RESUMEN

Cleidocranial dysplasia (CCD) is caused by haploinsufficiency in RUNX2 function. We have previously identified a series of RUNX2 mutations in Korean CCD patients, including a novel R131G missense mutation in the Runt-homology domain. Here, we examine the functional consequences of the RUNX2(R131G) mutation, which could potentially affect DNA binding, nuclear localization signal, and/or heterodimerization with core-binding factor-beta (CBF-beta). Immunofluorescence microscopy and western blot analysis with subcellular fractions show that RUNX2(R131G) is localized in the nucleus. Immunoprecipitation analysis reveals that heterodimerization with CBF-beta is retained. However, precipitation assays with biotinylated oligonucleotides and reporter gene assays with RUNX2 responsive promoters together reveal that DNA-binding activity and consequently the transactivation of potential of RUNX2(R131G) is abrogated. We conclude that loss of DNA binding, but not nuclear localization or CBF-beta heterodimerization, causes RUNX2 haploinsufficiency in patients with the RUNX2(R131G) mutation. Retention of specific functions including nuclear localization and binding to CBF-beta of the RUNX2(R131G) mutation may render the mutant protein an effective competitor that interferes with wild-type function.


Asunto(s)
Sustitución de Aminoácidos/genética , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , ADN/metabolismo , Mutación/efectos de los fármacos , Secuencias de Aminoácidos , Animales , Células CHO , Núcleo Celular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/química , Cricetinae , Cricetulus , Células HeLa , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Unión Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Transporte de Proteínas , Activación Transcripcional/genética
9.
Blood ; 112(9): 3777-87, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18695000

RESUMEN

The heterodimeric transcription factor RUNX1/PEBP2-beta (also known as AML1/CBF-beta) is essential for definitive hematopoiesis. Here, we show that interaction with PEBP2-beta leads to the phosphorylation of RUNX1, which in turn induces p300 phosphorylation. This is mediated by homeodomain interacting kinase 2 (HIPK2), targeting Ser(249), Ser(273), and Thr(276) in RUNX1, in a manner that is also dependent on the RUNX1 PY motif. Importantly, we observed the in vitro disruption of this phosphorylation cascade by multiple leukemogenic genetic defects targeting RUNX1/CBFB. In particular, the oncogenic protein PEBP2-beta-SMMHC prevents RUNX1/p300 phosphorylation by sequestering HIPK2 to mislocalized RUNX1/beta-SMMHC complexes. Therefore, phosphorylation of RUNX1 appears a critical step in its association with and phosphorylation of p300, and its disruption may be a common theme in RUNX1-associated leukemogenesis.


Asunto(s)
Factor de Unión a CCAAT/metabolismo , Proteínas Portadoras/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Leucemia/etiología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Secuencia de Bases , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Subunidad alfa 2 del Factor de Unión al Sitio Principal/química , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/química , Cartilla de ADN/genética , Humanos , Células K562 , Leucemia/genética , Leucemia/metabolismo , Ratones , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Fosforilación , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia , Factores de Transcripción p300-CBP/química
10.
Biochem Biophys Res Commun ; 387(2): 342-7, 2009 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-19595669

RESUMEN

Aspirin has been reported to induce apoptosis in various cancer cell lines. However, the apoptotic effects of aspirin on human brain tumor cells are not well understood. Here, we have assessed the effect of aspirin on human gliobalstoma cell line A172 and found that aspirin induced the apoptosis of A172 cells, as determined by TUNEL assay, FACS analysis, and Hoechst staining. The underlying mechanism of this effect consists of reduction in the level of phosphorylated STAT3 (Tyr705), a transcription factor required for survival of A172 cells. Moreover, the expression of STAT3 target genes such as Cyclin D1, XIAP, and Bcl-2 that are essential for cell growth and survival was apparently attenuated after aspirin treatment. We also showed that the expression and secretion of interleukin-6 (IL-6), leading to STAT3 phosphorylation, was inhibited by aspirin. When administered exogenous IL-6 to aspirin-treated A172 cells, the phosphorylation of STAT3 and cellular apoptosis were restrained compared to aspirin only-treated cells. Taken together, our results indicate that aspirin causes apoptosis via down-regulation of IL-6-dependent STAT3 signaling, suggesting that aspirin could be therapeutically useful for a potential anti-glioblastoma therapeutic approach.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Aspirina/farmacología , Glioblastoma/metabolismo , Interleucina-6/antagonistas & inhibidores , Factor de Transcripción STAT3/antagonistas & inhibidores , Antineoplásicos/antagonistas & inhibidores , Aspirina/antagonistas & inhibidores , Línea Celular Tumoral , Humanos , Interleucina-6/farmacología , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tirosina/metabolismo
11.
Biochem Biophys Res Commun ; 379(2): 206-11, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19100714

RESUMEN

Adipokines have been known to act as angiogenic regulators in the process of angiogenesis. Recently, we have demonstrated that visfatin, a novel adipokine, has angiogenic activity. However, little has been reported on the underlying mechanism of visfatin-induced angiogenesis. In this study, we report that visfatin-induced angiogenesis is mediated by endothelial fibroblast growth factor-2 (FGF-2). Visfatin increased the levels of FGF-2 mRNA and protein in human endothelial cells. The enhancement in FGF-2 expression was prevented by an inhibitor of the extracellular signal-regulated kinase 1/2 (Erk1/2) pathway. Furthermore, visfatin-induced angiogenesis was reduced by inhibition of FGF-2 receptor kinase or by neutralization of FGF-2 function. Taken together, our results indicate that visfatin-induced endothelial angiogenesis is composed largely of two sequential steps: the induction of Erk1/2-dependent FGF-2 gene expression by visfatin and the subsequent FGF-2-induced angiogenesis. These data further suggest an integral role for visfatin-FGF-2 signaling axis in modulating endothelial angiogenesis.


Asunto(s)
Citocinas/metabolismo , Endotelio Vascular/enzimología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Fisiológica , Nicotinamida Fosforribosiltransferasa/metabolismo , Animales , Factor 2 de Crecimiento de Fibroblastos/genética , Humanos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
12.
Mol Cell Biol ; 26(12): 4474-88, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16738314

RESUMEN

Genes involved in the transforming growth factor beta (TGF-beta) signaling pathway are frequently altered in several types of cancers, and a gastric tumor suppressor RUNX3 appears to be an integral component of this pathway. We reported previously that apoptosis is notably reduced in Runx3-/- gastric epithelial cells. In the present study, we show that a proapoptotic gene Bim was transcriptionally activated by RUNX3 in the gastric cancer cell lines SNU16 and SNU719 treated with TGF-beta. The human Bim promoter contains RUNX sites, which are required for its activation. Furthermore, a dominant negative form of RUNX3 comprised of amino acids 1 to 187 increased tumorigenicity of SNU16 by inhibiting Bim expression. In Runx3-/- mouse gastric epithelium, Bim was down-regulated, and apoptosis was reduced to the same extent as that in Bim-/- gastric epithelium. We confirmed comparable expression of TGF-beta1 and TGF-beta receptors between wild-type and Runx3-/- gastric epithelia and reduction of Bim in TGF-beta1-/- stomach. These results demonstrate that RUNX3 is responsible for transcriptional up-regulation of Bim in TGF-beta-induced apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Mucosa Gástrica/citología , Mucosa Gástrica/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , ADN/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mucosa Gástrica/efectos de los fármacos , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Smad/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
13.
Biochim Biophys Acta ; 1773(9): 1462-72, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17586065

RESUMEN

Angiogenesis is important for adult tissue regeneration as well as normal development. Vascular endothelial growth factor (VEGF) is a unique potent angiogenic factor, and plays an essential role in regulating angiogenesis during embryonic development, normal tissue growth, and tissue regeneration. Recent evidence shows that nerve growth factor (NGF) also plays a role as an angiogenic regulator as well as a well-known neurotrophic factor. The aim of this study was to investigate whether thymus regeneration accompanies reparative angiogenesis and also to evaluate whether the thymic expression of VEGF is regulated by NGF in vivo and in vitro. Here, we show that high VEGF mRNA and protein levels are concomitant with reparative angiogenesis that occurs dramatically during regeneration following acute involution induced by cyclophosphamide (CY) in the rat thymus. Fluorescent thymus angiography using FITC-dextran showed that thymic regeneration is associated with a much denser capillary network compared with normal control thymus. Furthermore, the expressions of NGF and TrkA were highly increased during thymic regeneration. We also show that NGF mediates thymic epithelial induction of VEGF expression in vitro and in vivo. Taken together, our results suggest that NGF-mediated VEGF up-regulation in thymic epithelial cells may contribute to reparative angiogenesis during thymic regeneration in adult.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Regeneración , Timo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Angiografía , Animales , Células Cultivadas , Ciclofosfamida/toxicidad , Fluoresceína-5-Isotiocianato , Técnica del Anticuerpo Fluorescente Indirecta , Colorantes Fluorescentes , Inmunohistoquímica , Masculino , Neovascularización Fisiológica , Proteínas/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Organismos Libres de Patógenos Específicos , Timo/citología , Timo/patología , Timo/fisiología
14.
J Cell Biochem ; 105(1): 236-44, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18570183

RESUMEN

The adenoviral gene, termed early region 1A (E1A), is crucial for transformation and has been used very effectively as a tool to determine the molecular mechanisms that underlie the basis of cellular transformation. pRb, p107, p130, p300/CBP, p400, TRRAP, and CtBP were identified to be E1A-binding proteins and their roles in cellular transformation have been established. Although the major function of E1A is considered to be the regulation of gene expression that is critical for differentiation and cell cycle exit, one of the most significant questions relating to E1A transformation is how E1A mediates this regulation. RUNX3 is a transcription factor that was first described as a gastric cancer tumor suppressor but is now known to be involved in many different cancers. Exogenous expression of RUNX3 strongly inhibits the growth of cells. Here, we show that the adenovirus oncoprotein E1A interacts with RUNX3 in vitro and in vivo. RUNX3 interacts with the N-terminus (amino acids 2-29) of E1A, which is known to interact with p300/CBP, p400, and TRRAP. E1A interacts directly with the Runt domain of RUNX3 but does not interfere with CBFbeta-RUNX3 interactions. In addition, E1A inhibits the transactivation activity of RUNX3 on the p21(WAF1/CIP1) promoter. Consistent with these observations, the growth inhibition induced by RUNX3 is reduced by E1A. These results demonstrate that E1A specifically binds to RUNX3 and inactivates its transactivation activity. We propose that one of the mechanisms for the oncogenic activity of E1A is the inhibition of RUNX3, similar to that of RB and p300/CBP.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Activación Transcripcional/genética , Proteínas E1A de Adenovirus/genética , Factor de Unión a CCAAT/metabolismo , Línea Celular , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Humanos , Unión Proteica
15.
Stem Cells ; 25(12): 2976-86, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17823240

RESUMEN

The RUNX1/AML1 gene encodes a transcription factor essential for the generation of hematopoietic stem cells and is frequently targeted in human leukemia. In human RUNX1-related leukemias, the RAS pathway is often concurrently mutated, but the mechanism of the synergism remains elusive. Here, we found that inactivation of Runx1 in mouse bone marrow cells results in an increase in the stem/progenitor cell fraction due to suppression of apoptosis and elevated expression of the polycomb gene Bmi-1, which is important for stem cell self-renewal. Introduction of oncogenic N-RAS into wild-type cells, in contrast, reduced the stem/progenitor cell fraction because of senescence, apoptosis, and differentiation. Such detrimental events presumably occurred because of the cellular fail-safe program, although hyperproliferation was initially induced by an oncogenic stimulus. Runx1 insufficiency appears to impair such a fail-safe mechanism, particularly in the stem/progenitor cells, thereby supporting the clonal maintenance of leukemia-initiating cells expressing an activated oncogene. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Genes ras/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Leucemia Experimental/metabolismo , Leucemia Experimental/prevención & control , Animales , Apoptosis/genética , Supervivencia Celular/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Subunidad alfa 2 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Leucemia Experimental/genética , Leucemia Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo
16.
Exp Mol Med ; 50(4): 1-13, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29700280

RESUMEN

Liver fibrosis can be reversed by removing its causative injuries; however, the molecular mechanisms mediating the resolution of liver fibrogenesis are poorly understood. We investigate the role of a scaffold protein, A-Kinase Anchoring Protein 12 (AKAP12), during liver fibrosis onset, and resolution. Biliary fibrogenesis and fibrosis resolution was induced in wild-type (WT) or AKAP12-deficient C57BL/6 mice through different feeding regimens with 0.1% 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-containing chow. AKAP12 expression in portal fibroblasts (PFs) and liver sinusoidal endothelial cells (LSECs) gradually decreased as fibrosis progressed but was restored after cessation of the fibrotic challenge. Histological analysis of human liver specimens with varying degrees of fibrosis of different etiologies revealed that AKAP12 expression diminishes in hepatic fibrosis from its early stages onward. AKAP12 KO mice displayed reduced fibrosis resolution in a DDC-induced biliary fibrosis model, which was accompanied by impaired normalization of myofibroblasts and capillarized sinusoids. RNA sequencing of the liver transcriptome revealed that genes related to ECM accumulation and vascular remodeling were mostly elevated in AKAP12 KO samples. Gene ontology (GO) and bioinformatic pathway analyses identified that the differentially expressed genes were significantly enriched in GO categories and pathways, such as the adenosine 3',5'-cyclic monophosphate (cAMP) pathway. Knockdown of the AKAP12 gene in cultured primary PFs revealed that AKAP12 inhibited PF activation in association with the adenosine 3',5'-cyclic monophosphate (cAMP) pathway. Moreover, AKAP12 knockdown in LSECs led to enhanced angiogenesis, endothelin-1 expression and alterations in laminin composition. Collectively, this study demonstrates that AKAP12-mediated regulation of PFs and LSECs has a central role in resolving hepatic fibrosis.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/genética , Proteínas de Ciclo Celular/genética , Fibroblastos/patología , Regulación de la Expresión Génica , Cirrosis Hepática/genética , Hígado/patología , Proteínas de Anclaje a la Quinasa A/análisis , Animales , Proteínas de Ciclo Celular/análisis , Línea Celular , Fibroblastos/metabolismo , Humanos , Hígado/citología , Hígado/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Transcriptoma
17.
Biochim Biophys Acta ; 1763(3): 282-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16516312

RESUMEN

The marked induction of glial fibrillary acidic protein (GFAP) has been observed in astrocytes during neuropathological processes accompanying reactive gliosis; however, the precise molecular mechanism(s) underlying this GFAP induction remains poorly resolved. Therefore, in this study, we examined whether the change of nuclear factor-kappa B (NF-kappaB) activity can influence GFAP expression levels. Aspirin, widely used to prevent NF-kappaB activity, reduced the levels of GFAP mRNA and protein in human astroglial cells including human glioblastoma A172 cells and primary human brain astrocyte cells (HBAs). Furthermore, aspirin inhibited the effects of hypoxic injury on the up-regulation of GFAP expression in HBAs. We confirmed the repressive effect of aspirin on GFAP transcription by GFAP promoter-driven reporter assay and found that one NF-kappaB binding site conserved in the mouse and human GFAP gene promoters is critical for this effect. To further delineate whether NF-kappaB is directly involved in the regulation of GFAP gene expression, we transfected A172 cells with an expression vector encoding a super-repressor IkappaBalpha protein (IkappaBalpha-SR) to specifically inhibit NF-kappaB activity and found the marked reduction of GFAP protein levels in IkappaBalpha-SR-transfectant cells. Taken together, our results suggest that NF-kappaB may play pivotal roles in GFAP gene expression.


Asunto(s)
Aspirina/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , FN-kappa B/antagonistas & inhibidores , Regulación hacia Arriba/efectos de los fármacos , Astrocitos/citología , Hipoxia de la Célula/efectos de los fármacos , Regulación hacia Abajo/genética , Humanos , Quinasa I-kappa B/metabolismo , FN-kappa B/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
18.
Oncotarget ; 8(34): 57216-57230, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28915666

RESUMEN

Aurora kinase A (AuA) is a prerequisite for centrosome maturation, separation, and mitotic spindle assembly, thus, it is essential for cell cycle regulation. Overexpression of AuA is implicated in poor prognosis of many types of cancer. However, the regulatory mechanisms underlying the functions of AuA are still not fully understood. Here, we report that AuA colocalizes with arrest defective protein 1 (ARD1) acetyltransferase during cell division and cell migration. Additionally, AuA is acetylated by ARD1 at lysine residues at positions 75 and 125. The double mutations at K75/K125 abolished the kinase activity of AuA. Moreover, the double mutant AuA exhibited diminished ability to promote cell proliferation and cell migration. Mechanistic studies revealed that AuA acetylation at K75/K125 promoted cell proliferation via activation of cyclin E/CDK2 and cyclin B1. In addition, AuA acetylation stimulated cell migration by activating the p38/AKT/MMP-2 pathway. Our findings indicate that ARD1-mediated acetylation of AuA enhances cell proliferation and migration, and probably contributes to cancer development.

19.
Mol Neurobiol ; 54(9): 7353-7368, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-27815839

RESUMEN

Over the last few decades, molecular neurobiology has uncovered many genes whose deficiency in mice results in behavioral traits associated with human neuropsychiatric disorders such as autism, obsessive-compulsive disorder (OCD), and schizophrenia. However, the etiology of these common diseases remains enigmatic with the potential involvement of a battery of genes. Here, we report abnormal behavioral phenotypes of mice deficient in a cell adhesion molecule Ninjurin 1 (Ninj1), which are relevant to repetitive and anxiety behaviors of neuropsychiatric disorders. Ninj1 knockout (KO) mice exhibit compulsive grooming-induced hair loss and self-made lesions as well as increased anxiety-like behaviors. Histological analysis reveals that Ninj1 is predominantly expressed in cortico-thalamic circuits, and neuron-specific Ninj1 conditional KO mice manifest aberrant phenotypes similar to the global Ninj1 KO mice. Notably, the brains of Ninj1 KO mice display altered synaptic transmission in thalamic neurons as well as a reduced number of functional synapses. Moreover, the disruption of Ninj1 leads to glutamatergic abnormalities, including increased ionotropic glutamate receptors but reduced glutamate levels. Furthermore, chronic treatment with fluoxetine, a drug reportedly ameliorates compulsive behaviors in mice, prevents progression of hair loss and alleviates the compulsive grooming and anxiety-like behavior of Ninj1 KO mice. Collectively, our results suggest that Ninj1 could be involved in neuropsychiatric disorders associated with impairments of repetitive and anxiety behaviors.


Asunto(s)
Ansiedad/genética , Ansiedad/metabolismo , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Conducta Compulsiva/genética , Conducta Compulsiva/metabolismo , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Animales , Ansiedad/psicología , Células Cultivadas , Conducta Compulsiva/psicología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Oncotarget ; 8(40): 68517-68529, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978134

RESUMEN

SAM domain and HD domain containing protein 1 (SAMHD1) is a deoxynucleotide triphosphohydrolase (dNTPase) that inhibits retroviruses by depleting intracellular deoxynucleotide triphosphates (dNTPs) in non-cycling myeloid cells. Although SAMHD1 is expressed ubiquitously throughout the human body, the molecular mechanisms regulating its enzymatic activity and function in non-immune cells are relatively unexplored. Here, we demonstrate that the dNTPase activity of SAMHD1 is regulated by acetylation, which promotes cell cycle progression in cancer cells. SAMHD1 is acetylated at residue lysine 405 (K405) in vitro and in vivo by an acetylatransferase, arrest defective protein 1 (ARD1). Acetylated SAMHD1 wildtype proteins have enhanced dNTPase activity in vitro, whereas non-acetylated arginine substituted mutants (K405R) do not. K405R mutant expressing cancer cells have reduced G1/S transition and slower proliferation compared to wildtype. SAMHD1 acetylation levels are strongest during the G1 phase, indicating a role during G1 phase. Collectively, these findings suggest that SAMHD1 acetylation enhances its dNTPase activity and promotes cancer cell proliferation. Therefore, SAMHD1 acetylation may be a potent therapeutic target for cancer treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA