Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Transl Med ; 10(9): 1329-1342, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34008349

RESUMEN

The conventional planar culture of adherent cells is inefficient for large-scale manufacturing of cell and gene therapy products. We developed a facile and efficient bead-to-bead cell-transfer method for serial subculture and large-scale expansion of human mesenchymal stem cells (hMSCs) with microcarriers in bioreactors. We first compared culture medium with and without nucleosides and found the former maintained the expression of surface markers of hMSCs during their prolonged culture and enabled faster cell proliferation. Subsequently, we developed our bead-to-bead cell transfer method to subculture hMSCs and found that intermittent agitation after adding fresh microcarriers to cell-populated microcarriers could promote spontaneous cell migration to fresh microcarriers, reduce microcarrier aggregation, and improve cell yield. This method enabled serial subculture of hMSCs in spinner flasks from passage 4 to passage 9 without using proteolytic enzymes, which showed faster cell proliferation than the serial planar cultures undergoing multiple enzyme treatment. Finally, we used the medium containing nucleosides and our bead-to-bead cell transfer method for cell culture scale-up from 4- to 50-L cultures in single-use bioreactors. We achieved a 242-fold increase in the number of cells to 1.45 × 1010 after 27-day culture and found that the cells harvested from the bioreactors maintained proliferation ability, expression of their surface markers, tri-lineage differentiation potential and immunomodulatory property. This study shows the promotive effect of nucleosides on hMSC expansion and the potential of using our bead-to-bead transfer method for larger-scale manufacturing of hMSCs for cell therapy.


Asunto(s)
Células Madre Mesenquimatosas , Reactores Biológicos , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Proliferación Celular , Medios de Cultivo , Humanos
2.
Oncol Rep ; 42(4): 1507-1516, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31524271

RESUMEN

CUB domain­containing protein 1 (CDCP1) is phosphorylated by Src family kinases (SFK), and is thought to serve an important role in tumor metastasis through downstream signaling subsequent to its interaction with protein kinase C δ. The present study investigated the mechanisms of activation for CDCP1 signaling, and demonstrated that CDCP1 is able to activate SFK via a homophilic complex of the extracellular complement C1r/C1s, urchin embryonic growth factor, bone morphogenetic protein 1 (CUB) 2 domain. Deletion of the extracellular CDCP1 region abolished homophilic complex formation of CDCP1 and the ability to promote cancer cell migration. When the culture medium was supplemented with recombinant CUB2 domain protein fused with maltose binding protein (rMBP­CUB2), CDCP1 homophilic complex formation was effectively inhibited. rMBP­CUB2 also inhibited SFK activation and the migratory capacity of invasive human lung adenocarcinoma A549 cells, and human pancreatic BxPC3 cells. These findings demonstrated a novel function for the extracellular CUB2 domain of CDCP1, promoting cancer cell migration via SFK activation on the plasma membrane. It was also indicated that the region blocking the homophilic binding site may be a potential therapeutic target against CDCP1­dependent tumor invasion.

3.
Biochem Biophys Res Commun ; 495(1): 1292-1299, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29162448

RESUMEN

Development of resistance against temozolomide (TMZ) in glioblastoma (GBM) after continuous treatment with TMZ is one of the critical problems in clinical GBM therapy. Intracellular cholesterol regulates cancer cell biology, but whether intracellular cholesterol is involved in TMZ resistance of GBM cells remains unclear. The involvement of intracellular cholesterol in acquired resistance against TMZ in GBM cells was investigated. Intracellular cholesterol levels were measured in human U251 MG cells with acquired TMZ resistance (U251-R cells) and TMZ-sensitive control U251 MG cells (U251-Con cells), and found that the intracellular cholesterol level was significantly lower in U251-R cells than in U251-Con cells. In addition, treatment by intracellular cholesterol remover, methyl-beta cyclodextrin (MßCD), or intracellular cholesterol inducer, soluble cholesterol (Chol), regulated TMZ-induced U251-Con cell death in line with changes in intracellular cholesterol level. Involvement of death receptor 5 (DR5), a death receptor localized in the plasma membrane, was evaluated. TMZ without or with MßCD and/or Chol caused accumulation of DR5 into the plasma membrane lipid raft and formed a complex with caspase-8, an extrinsic caspase cascade inducer, reflected in the induction of cell death. In addition, treatment with caspase-8 inhibitor or knockdown of DR5 dramatically suppressed U251-Con cell death induced by combination treatment with TMZ, MßCD, and Chol. Combined treatment of Chol with TMZ reversed the TMZ resistance of U251-R cells and another GBM cell model with acquired TMZ resistance, whereas clinical antihypercholesterolemia agents at physiological concentrations suppressed TMZ-induced cell death of U251-Con cells. These findings suggest that intracellular cholesterol level affects TMZ treatment of GBM mediated via a DR5-caspase-8 mechanism.


Asunto(s)
Caspasa 8/metabolismo , Colesterol/metabolismo , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Microdominios de Membrana/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Antineoplásicos Alquilantes/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Dacarbazina/administración & dosificación , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos , Glioblastoma/patología , Humanos , Líquido Intracelular/metabolismo , Microdominios de Membrana/efectos de los fármacos , Temozolomida
4.
Cancer Sci ; 108(5): 1049-1057, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28256037

RESUMEN

CUB domain-containing protein-1 (CDCP1) is a trans-membrane protein predominantly expressed in various cancer cells and involved in tumor progression. CDCP1 is phosphorylated at tyrosine residues in the intracellular domain by Src family kinases and recruits PKCδ to the plasma membrane through tyrosine phosphorylation-dependent association with the C2 domain of PKCδ, which in turn induces a survival signal in an anchorage-independent condition. In this study, we used our cell-free screening system to identify a small compound, glycoconjugated palladium complex (Pd-Oqn), which significantly inhibited the interaction between the C2 domain of PKCδ and phosphorylated CDCP1. Immunoprecipitation assays demonstrated that Pd-Oqn hindered the intercellular interaction of phosphorylated CDCP1 with PKCδ and also suppressed the phosphorylation of PKCδ but not that of ERK or AKT. In addition, Pd-Oqn inhibited the colony formation of gastric adenocarcinoma 44As3 cells in soft agar as well as their invasion. In mouse models, Pd-Oqn markedly reduced the peritoneal dissemination of gastric adenocarcinoma cells and the tumor growth of pancreatic cancer orthotopic xenografts. These results suggest that the novel compound Pd-Oqn reduces tumor metastasis and growth by inhibiting the association between CDCP1 and PKCδ, thus potentially representing a promising candidate among therapeutic reagents targeting protein-protein interaction.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Metástasis de la Neoplasia/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Proteína Quinasa C-delta/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Células A549 , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Sci Rep ; 6: 38762, 2016 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-27929116

RESUMEN

Apoptosis inhibitor of macrophage (AIM, encoded by cd5l) is a multi-functional circulating protein that has a beneficial role in the regulation of a broad range of diseases, some of which are ameliorated by AIM administration in mice. In blood, AIM is stabilized by association with IgM pentamers and maintains its high circulating levels. The mechanism regulating the excessive accumulation of blood AIM remains unknown, although it is important, since a constitutive increase in AIM levels promotes chronic inflammation. Here we found a physiological AIM-cleavage process that induces destabilization of AIM and its excretion in urine. In blood, IgM-free AIM appeared to be cleaved and reduced in size approximately 10 kDa. Cleaved AIM was unable to bind to IgM and was selectively filtered by the glomerulus, thereby excreted in urine. Amino acid substitution at the cleavage site resulted in no renal excretion of AIM. Interestingly, cleaved AIM retained a comparable potency with full-length AIM in facilitating the clearance of dead cell debris in injured kidney, which is a key response in the recovery of acute kidney injury. Identification of AIM-cleavage and resulting functional modification could be the basis for designing safe and efficient AIM therapy for various diseases.


Asunto(s)
Riñón/metabolismo , Receptores Depuradores/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Humanos , Ratones , Proteolisis , Ratas
6.
Biochem Biophys Res Commun ; 468(1-2): 240-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26518652

RESUMEN

Temozolomide (TMZ) is one of the few effective anticancer agents against gliomas. However, acquisition of TMZ resistance or adaptation by gliomas is currently a crucial problem, especially increased invasiveness which is critical for the determination of clinical prognosis. This study investigated the molecular regulatory mechanisms of TMZ resistance in gliomas involved in invasiveness, particularly invadopodia formation, a molecular complex formed at the invasive front to cause extracellular matrix degradation during cellular local invasion. The TMZ-resistant clone of the U343 MG human glioma cell line (U343-R cells) was established. U343-R cells demonstrated higher invadopodia formation compared with U343 cells without TMZ resistance (U343-Con cells). Immunoblot analysis of DNA damage-related mitogen-activated protein kinase signals found increased phosphorylation of c-Jun terminal kinase (JNK) and higher activation of its downstream signaling in U343-R cells compared with U343-Con cells. Treatment of U343-R cells with specific inhibitors of JNK or siRNA targeting JNK suppressed up-regulation of invadopodia formation. In addition, paxillin, one of the known JNK effectors which is phosphorylated and affects cell migration, was phosphorylated at serine 178 in JNK activity-dependent manner. Expression of paxillin with mutation of the serine 178 phosphorylation site in U343-R cells blocked invadopodia formation. The present findings suggest that increased formation of invadopodia in U343-R cells is mediated by hyperactivation of JNK-paxillin signaling, and both JNK and paxillin might become targets of novel therapies against TMZ-resistant gliomas.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Dacarbazina/análogos & derivados , Resistencia a Antineoplásicos , Glioma/tratamiento farmacológico , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Paxillin/metabolismo , Podosomas/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Dacarbazina/farmacología , Glioma/metabolismo , Glioma/patología , Humanos , Fosforilación/efectos de los fármacos , Podosomas/metabolismo , Podosomas/patología , Transducción de Señal/efectos de los fármacos , Temozolomida
7.
Mol Cancer Res ; 12(10): 1449-59, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24939643

RESUMEN

UNLABELLED: Involvement of Ras in cancer initiation is known, but recent evidence indicates a role in cancer progression, including metastasis and invasion; however, the mechanism is still unknown. In this study, it was determined that human lung cancer cells with Ras mutations, among other popular mutations, showed significantly higher expression of CUB domain-containing protein 1 (CDCP1) than those without. Furthermore, activated Ras clearly induced CDCP1, whereas CDCP1 knockdown or inhibition of CDCP1 phosphorylation by Src-directed therapy abrogated anoikis resistance, migration, and invasion induced by activated-Ras. Activation of MMP2 and secretion of MMP9, in a model of Ras-induced invasion, was found to be regulated through induction of phosphorylated CDCP1. Thus, CDCP1 is required for the functional link between Ras and Src signaling during the multistage development of human malignant tumors, highlighting CDCP1 as a potent target for treatment in the broad spectrum of human cancers associated with these oncogenes. IMPLICATIONS: CDCP1 protein induced by oncogenic Ras/Erk signaling is essential for Ras-mediated metastatic potential of cancer cells.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Genes ras , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Anoicis , Antígenos de Neoplasias , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Mutación/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/genética , Familia-src Quinasas/metabolismo
8.
Nat Commun ; 5: 3147, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24445679

RESUMEN

MafB is a transcription factor that induces myelomonocytic differentiation. However, the precise role of MafB in the pathogenic function of macrophages has never been clarified. Here we demonstrate that MafB promotes hyperlipidemic atherosclerosis by suppressing foam-cell apoptosis. Our data show that MafB is predominantly expressed in foam cells found within atherosclerotic lesions, where MafB mediates the oxidized LDL-activated LXR/RXR-induced expression of apoptosis inhibitor of macrophages (AIM). In the absence of MafB, activated LXR/RXR fails to induce the expression of AIM, a protein that is normally responsible for protecting macrophages from apoptosis; thus, Mafb-deficient macrophages are prone to apoptosis. Haematopoietic reconstitution with Mafb-deficient fetal liver cells in recipient LDL receptor-deficient hyperlipidemic mice revealed accelerated foam-cell apoptosis, which subsequently led to the attenuation of the early atherogenic lesion. These findings represent the first evidence that the macrophage-affiliated MafB transcription factor participates in the acceleration of atherogenesis.


Asunto(s)
Apoptosis , Aterosclerosis/fisiopatología , Células Espumosas/patología , Factor de Transcripción MafB/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Aterosclerosis/patología , Secuencia de Bases , Humanos , Receptores X del Hígado , Factor de Transcripción MafB/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores Nucleares Huérfanos/metabolismo , Receptores Inmunológicos/genética , Receptores Depuradores , Receptores X Retinoide/metabolismo , Homología de Secuencia de Ácido Nucleico
9.
Nat Commun ; 4: 1836, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23673621

RESUMEN

Peptidylarginine deiminase 4 (PAD4) functions as a transcriptional coregulator by catalyzing the conversion of histone H3 arginine residues to citrulline residues. Although the high level of PAD4 expression in bone marrow cells suggests its involvement in haematopoiesis, its precise contribution remains unclear. Here we show that PAD4, which is highly expressed in lineage(-) Sca-1(+) c-Kit(+) (LSK) cells of mouse bone marrow compared with other progenitor cells, controls c-myc expression by catalyzing the citrullination of histone H3 on its promoter. Furthermore, PAD4 is associated with lymphoid enhancer-binding factor 1 and histone deacetylase 1 at the upstream region of the c-myc gene. Supporting these findings, LSK cells, especially multipotent progenitors, in PAD4-deficient mice show increased proliferation in a cell-autonomous fashion compared with those in wild-type mice. Together, our results strongly suggest that PAD4 regulates the proliferation of multipotent progenitors in the bone marrow by controlling c-myc expression.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Hidrolasas/metabolismo , Células Madre Multipotentes/citología , Células Madre Multipotentes/enzimología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Recuento de Células , Núcleo Celular/enzimología , Proliferación Celular , Células HEK293 , Histona Desacetilasas/metabolismo , Humanos , Hidrolasas/deficiencia , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Transporte de Proteínas , Arginina Deiminasa Proteína-Tipo 4 , Proteínas Proto-Oncogénicas c-myc/genética
10.
Cell Rep ; 3(4): 1187-98, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23562157

RESUMEN

Natural immunoglobulin M (IgM) is reactive to autoantigens and is believed to be important for autoimmunity. Blood pentameric IgM loaded with antigens forms a large immune complex (IC) that contains various elements, including apoptosis inhibitor of macrophage (AIM). Here we demonstrate that this IgM-AIM association contributes to autoantibody production under obese conditions. In mice fed a high-fat diet, natural IgM increased through B cell TLR4 stimulation. AIM associated with IgM and protected AIM from renal excretion, increasing blood AIM levels along with the obesity-induced IgM augmentation. Meanwhile, the AIM association inhibited IgM binding to the Fcα/µ receptor on splenic follicular dendritic cells, thereby protecting the IgM IC from Fcα/µ receptor-mediated internalization. This supported IgM-dependent autoantigen presentation to B cells, stimulating IgG autoantibody production. Accordingly, in obese AIM-deficient (AIM(-/-)) mice, the increase of multiple IgG autoantibodies observed in obese wild-type mice was abrogated. Thus, the AIM-IgM association plays a critical role in the obesity-associated autoimmune process.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autoanticuerpos/metabolismo , Células Dendríticas Foliculares/metabolismo , Inmunoglobulina M/metabolismo , Receptores Inmunológicos/metabolismo , Animales , Complejo Antígeno-Anticuerpo/sangre , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Células Dendríticas Foliculares/inmunología , Dieta Alta en Grasa , Células HEK293 , Humanos , Inmunoglobulina G/metabolismo , Inmunoglobulina M/química , Inmunoglobulina M/inmunología , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Obesidad/patología , Células Plasmáticas/metabolismo , Unión Proteica , Receptores Fc/genética , Receptores Fc/metabolismo , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Depuradores , Receptor Toll-Like 4/metabolismo
12.
Biochem Biophys Res Commun ; 422(3): 476-81, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22579686

RESUMEN

Under fasting conditions, triacylglycerol in adipose tissue undergoes lipolysis to supply fatty acids as energy substrates. Such lipolysis is regulated by hormones, which activate lipases via stimulation of specific signalling cascades. We previously showed that macrophage-derived soluble protein, AIM induces obesity-associated lipolysis, triggering chronic inflammation in fat tissue which causes insulin resistance. However, the mechanism of how AIM mediates lipolysis remains unknown. Here we show that AIM induces lipolysis in a manner distinct from that of hormone-dependent lipolysis, without activation or augmentation of lipases. In vivo and in vitro, AIM did not enhance phosphorylation of hormone-sensitive lipase (HSL) in adipocytes, a hallmark of hormone-dependent lipolysis activation. Similarly, adipose tissue from obese AIM-deficient and wild-type mice showed comparable HSL phosphorylation. Consistent with the suppressive effect of AIM on fatty acid synthase activity, the amount of saturated and unsaturated fatty acids was reduced in adipocytes treated with AIM. This response ablated transcriptional activity of peroxisome proliferator-activated receptor (PPARγ), leading to diminished gene expression of lipid-droplet coating proteins including fat-specific protein 27 (FSP27) and Perilipin, which are indispensable for triacylglycerol storage in adipocytes. Accordingly, the lipolytic effect of AIM was overcome by a PPARγ-agonist or forced expression of FSP27, while it was synergized by a PPARγ-antagonist. Overall, distinct modes of lipolysis appear to take place in different physiological situations; one is a supportive response against nutritional deprivation achieved by enhancing lipase activity, and the other is a pathological consequence of obesity, causing subclinical inflammation and metabolic disorders, mediated by abolishing droplet-coating proteins.


Asunto(s)
Adipocitos/metabolismo , Proteínas Reguladoras de la Apoptosis/fisiología , Lipólisis , Receptores Inmunológicos/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Portadoras/metabolismo , Ácidos Grasos/antagonistas & inhibidores , Ácidos Grasos/biosíntesis , Ratones , PPAR gamma/agonistas , PPAR gamma/metabolismo , Perilipina-1 , Fosfoproteínas/metabolismo , Fosforilación , Proteínas/metabolismo , Receptores Inmunológicos/genética , Receptores Depuradores , Esterol Esterasa/metabolismo
13.
EMBO J ; 30(5): 823-34, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21285944

RESUMEN

Somatic cells can be reprogrammed to an ES-like state to create induced pluripotent stem cells (iPSCs) by ectopic expression of four transcription factors, Oct4, Sox2, Klf4 and cMyc. Here, we show that cellular microRNAs (miRNAs) regulate iPSC generation. Knock-down of key microRNA pathway proteins resulted in significant decreases in reprogramming efficiency. Three miRNA clusters, miR-17∼92, miR-106b∼25 and miR-106a∼363, were shown to be highly induced during early reprogramming stages. Several miRNAs, including miR-93 and miR-106b, which have very similar seed regions, greatly enhanced iPSC induction and modulated mesenchymal-to-epithelial transition step in the initiation stage of reprogramming, and inhibiting these miRNAs significantly decreased reprogramming efficiency. Moreover, miR-iPSC clones reached the fully reprogrammed state. Further analysis revealed that Tgfbr2 and p21 are directly targeted by these miRNAs and that siRNA knock-down of both genes indeed enhanced iPSC induction. Here, for the first time, we demonstrate that miR-93 and its family members directly target TGF-ß receptor II to enhance iPSC generation. Overall, we demonstrate that miRNAs function in the reprogramming process and that iPSC induction efficiency can be greatly enhanced by modulating miRNA levels in cells.


Asunto(s)
Reprogramación Celular , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , MicroARNs/fisiología , Procesamiento Postranscripcional del ARN , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Desdiferenciación Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Transición Epitelial-Mesenquimal , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Factor 4 Similar a Kruppel , Luciferasas/metabolismo , Ratones , Ratones Desnudos , Proteínas Nucleares/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Interferente Pequeño , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Teratoma/genética , Teratoma/metabolismo , Teratoma/patología , Transcripción Genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
14.
Cell Metab ; 11(6): 479-92, 2010 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-20519120

RESUMEN

Macrophages infiltrate adipose tissue in obesity and are involved in the induction of inflammation, thereby contributing to the development of obesity-associated metabolic disorders. Here, we show that the macrophage-derived soluble protein AIM is endocytosed into adipocytes via CD36. Within adipocytes, AIM associates with cytosolic fatty acid synthase (FAS), thereby decreasing FAS activity. This decreases lipid droplet size, stimulating the efflux of free fatty acids and glycerol from adipocytes. As an additional consequence of FAS inhibition, AIM prevents preadipocyte maturation. In vivo, the increase in adipocyte size and fat weight induced by high-fat diet (HFD) was accelerated in AIM-deficient (AIM(-)(/-)) mice compared to AIM(+/+) mice. Moreover, injection of recombinant AIM in AIM(-)(/-) mice suppresses the increase in fat mass induced by HFD. Interestingly, metabolic rates are comparable in AIM(-)(/-) and AIM(+/+) mice, suggesting that AIM specifically influences adipocyte status. Thus, this AIM function in adipocytes may be physiologically relevant to obesity progression.


Asunto(s)
Adipocitos/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Endocitosis , Ácido Graso Sintasas/metabolismo , Macrófagos/metabolismo , Receptores Inmunológicos/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/farmacología , Antígenos CD36/metabolismo , Grasas de la Dieta , Ácido Graso Sintasas/antagonistas & inhibidores , Metabolismo de los Lípidos , Macrófagos/inmunología , Ratones , Ratones Noqueados , Obesidad/etiología , Receptores Inmunológicos/genética , Receptores Depuradores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
15.
Biochem Biophys Res Commun ; 391(4): 1708-13, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-20043882

RESUMEN

Insulin secretion and glucose transport are the major mechanisms to balance glucose homeostasis. Recently, we found that the death effector domain-containing DEDD inhibits cyclin-dependent kinase-1 (Cdk1) function, thereby preventing Cdk1-dependent inhibitory phosphorylation of S6 kinase-1 (S6K1), downstream of phosphatidylinositol 3-kinase (PI3K), which overall results in maintenance of S6K1 activity. Here we newly show that DEDD forms a complex with Akt and heat-shock protein 90 (Hsp90), and supports the stability of both proteins. Hence, in DEDD(-/-) mice, Akt protein levels are diminished in skeletal muscles and adipose tissues, which interferes with the translocation of glucose-transporter 4 (GLUT4) upon insulin stimulation, leading to inefficient incorporation of glucose in these organs. Interestingly, as for the activation of S6K1, suppression of Cdk1 is involved in the stabilization of Akt protein by DEDD, since diminishment of Cdk1 in DEDD(-/-) cells via siRNA expression or treatment with a Cdk1-inhibitor, increases both Akt and Hsp90 protein levels. Such multifaceted involvement of DEDD in glucose homeostasis by supporting both insulin secretion (via maintenance of S6K1 activity) and glucose uptake (via stabilizing Akt protein), may suggest an association of DEDD-deficiency with the pathogenesis of type 2 diabetes mellitus.


Asunto(s)
Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Glucosa/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Insulina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Tejido Adiposo/metabolismo , Animales , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Diabetes Mellitus Tipo 2/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Ratones , Ratones Mutantes , Músculo Esquelético/metabolismo , Estabilidad Proteica , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
16.
J Biochem ; 146(4): 471-9, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19564157

RESUMEN

Peptidylarginine deiminases (PADs) consist of five enzymes which are widely distributed in human and rodent tissues. The two types of enzymes are found in human peripheral blood cells; PAD4 mainly in granulocytes and monocytes and PAD2 in lymphocytes and macrophages. Little is known about the regulation of PAD expression in macrophages. Here, we report that PAD2 is expressed in human monocytic leukaemia THP-1 cells during differentiation into macrophages by 12-O-tetradecanoylphorbol-13-acetate. During this differentiation, the levels of PAD2 mRNA and protein increased concomitantly, indicating the transcriptional regulation of PAD2 gene expression in the cells. The treatment of THP-1-derived macrophages with calcium ionophore A23187 generated vimentin deimination and resulted in the disruption of vimentin filament organization. We discuss the possible role of vimentin deimination in cell physiology.


Asunto(s)
Hidrolasas/genética , Macrófagos/metabolismo , Diferenciación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Hidrolasas/metabolismo , Macrófagos/efectos de los fármacos , Arginina Deiminasa Proteína-Tipo 2 , Desiminasas de la Arginina Proteica , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas
17.
Cancer Res ; 67(19): 9568-76, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17909068

RESUMEN

Colon cancers have been shown to develop after accumulation of multiple genetic and epigenetic alterations with changes in global gene expression profiles, contributing to the establishment of widely diverse phenotypes. Transcriptional and posttranscriptional regulation of gene expression by small RNA species, such as the small interfering RNA and microRNA and the RNA-induced silencing complex (RISC), is currently drawing major interest with regard to cancer development. SND1, also called Tudor-SN and p100 and recently reported to be a component of RISC, is among the list of highly expressed genes in human colon cancers. In the present study, we showed remarkable up-regulation of SND1 mRNA in human colon cancer tissues, even in early-stage lesions, and also in colon cancer cell lines. When mouse Snd1 was stably overexpressed in IEC6 rat intestinal epithelial cells, contact inhibition was lost and cell growth was promoted, even after the cells became confluent. Intriguingly, IEC6 cells with high levels of Snd1 also showed an altered distribution of E-cadherin from the cell membrane to the cytoplasm, suggesting loss of cellular polarity. Furthermore, the adenomatous polyposis coli (Apc) protein was coincidentally down-regulated, with no significant changes in the Apc mRNA level. Immunohistochemical analysis using chemically induced colonic lesions developed in rats revealed overexpression of Snd1 not only in colon cancers but also in aberrant crypt foci, putative precancerous lesions of the colon. Up-regulation of SND1 may thus occur at a very early stage in colon carcinogenesis and contribute to the posttranscriptional regulation of key players in colon cancer development, including APC and beta-catenin.


Asunto(s)
Neoplasias del Colon/genética , Proteínas Nucleares/genética , Complejo Silenciador Inducido por ARN/genética , Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Procesos de Crecimiento Celular/fisiología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Inhibición de Contacto/genética , Endonucleasas , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Células 3T3 NIH , Estadificación de Neoplasias , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Complejo Silenciador Inducido por ARN/biosíntesis , Complejo Silenciador Inducido por ARN/metabolismo , Ratas , Regulación hacia Arriba
18.
Biochem Biophys Res Commun ; 358(2): 608-14, 2007 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-17502110

RESUMEN

The biological function of hnRNP family proteins is widely diverse and involved in pre-mRNA processing, transcriptional regulation, recombination, and telomere maintenance. In the course of our study on the elucidation of biological functions of minisatellite DNA, we isolated several nuclear proteins that bind to the mouse minisatellite Pc-1, which consists of a tandem array of d(GGCAG) repeats, from NIH3T3 cells. One of the minisatellite binding proteins, MNBP-A, which binds to a single-stranded G-rich strand of the Pc-1 repeat, was proven identical to the hnRNP A3. Recombinant hnRNP A3 was demonstrated to bind to the single-stranded telomeric d(TTAGGG) repeat with much higher affinity than the d(GGCAG) repeat. Binding of hnRNP A3 to the single-stranded telomeric repeat protected the repeat from nuclease attack, and inhibited both telomerase reaction and DNA synthesis in vitro. These results suggest a possible biological role of hnRNP A3 in the stable maintenance of telomere repeats.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/química , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Proteínas de Unión a Telómeros/química , Proteínas de Unión a Telómeros/genética , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Datos de Secuencia Molecular , Unión Proteica
19.
Mutat Res ; 598(1-2): 120-31, 2006 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-16513142

RESUMEN

Mammalian genomes contain several types of repetitive sequences. Some of these sequences are implicated in various specific cellular events, including meiotic recombination, chromosomal breaks and transcriptional regulation, and also in several human disorders. In this review, we document the formation of DNA secondary structures by the G-rich repetitive sequences that have been found in several minisatellites, telomeres and in various triplet repeats, and report their effects on in vitro DNA synthesis. d(GGCAG) repeats in the mouse minisatellite Pc-1 were demonstrated to form an intra-molecular folded-back quadruplex structure (also called a G4' structure) by NMR and CD spectrum analyses. d(TTAGGG) telomere repeats and d(CGG) triplet repeats were also shown to form G4' and other unspecified higher order structures, respectively. In vitro DNA synthesis was substantially arrested within the repeats, and this could be responsible for the preferential mutability of the G-rich repetitive sequences. Electrophoretic mobility shift assays using NIH3T3 cell extracts revealed heterogeneous nuclear ribonucleoprotein (hnRNP) A1 and A3, which were tightly and specifically bound to d(GGCAG) and d(TTAGGG) repeats with K(d) values in the order of nM. HnRNP A1 unfolded the G4' structure formed in the d(GGCAG)(n) and d(TTAGGG)(n) repeat regions, and also resolved the higher order structure formed by d(CGG) triplet repeats. Furthermore, DNA synthesis arrest at the secondary structures of d(GGCAG) repeats, telomeres and d(CGG) triplet repeats was efficiently repressed by the addition of hnRNP A1. High expression of hnRNPs may contribute to the maintenance of G-rich repetitive sequences, including telomere repeats, and may also participate in ensuring the stability of the genome in cells with enhanced proliferation. Transcriptional regulation of genes, such as c-myc and insulin, by G4 sequences found in the promoter regions could be an intriguing field of research and help further elucidate the biological functions of the hnRNP family of proteins in human diseases.


Asunto(s)
ADN/química , Guanina , Células 3T3 , Animales , Secuencia de Bases , Dicroismo Circular , Ratones , Peso Molecular , Conformación de Ácido Nucleico , Polimorfismo Genético , Secuencias Repetidas en Tándem , Repeticiones de Trinucleótidos
20.
Nat Struct Mol Biol ; 11(8): 777-83, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15247907

RESUMEN

Peptidylarginine deiminase 4 (PAD4) is a Ca(2+)-dependent enzyme that catalyzes the conversion of protein arginine residues to citrulline. Its gene is a susceptibility locus for rheumatoid arthritis. Here we present the crystal structure of Ca(2+)-free wild-type PAD4, which shows that the polypeptide chain adopts an elongated fold in which the N-terminal domain forms two immunoglobulin-like subdomains, and the C-terminal domain forms an alpha/beta propeller structure. Five Ca(2+)-binding sites, none of which adopt an EF-hand motif, were identified in the structure of a Ca(2+)-bound inactive mutant with and without bound substrate. These structural data indicate that Ca(2+) binding induces conformational changes that generate the active site cleft. Our findings identify a novel mechanism for enzyme activation by Ca(2+) ions, and are important for understanding the mechanism of protein citrullination and for developing PAD-inhibiting drugs for the treatment of rheumatoid arthritis.


Asunto(s)
Calcio/química , Hidrolasas/química , Secuencias de Aminoácidos , Arginina/química , Sitios de Unión , Cadmio/química , Calcio/metabolismo , Catálisis , Citrulina/química , Cristalografía por Rayos X , Activación Enzimática , Humanos , Hidrolasas/metabolismo , Hidrólisis , Iones , Modelos Químicos , Modelos Moleculares , Mutagénesis , Mutación , Polimorfismo de Nucleótido Simple , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Arginina Deiminasa Proteína-Tipo 4 , Desiminasas de la Arginina Proteica , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...