Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(52): 33628-33638, 2020 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-33318192

RESUMEN

Retinoblastoma (Rb) is the most prevalent intraocular malignancy in children, with a worldwide survival rate <30%. We have developed a cancerous model of Rb in retinal organoids derived from genetically engineered human embryonic stem cells (hESCs) with a biallelic mutagenesis of the RB1 gene. These organoid Rbs exhibit properties highly consistent with Rb tumorigenesis, transcriptome, and genome-wide methylation. Single-cell sequencing analysis suggests that Rb originated from ARR3-positive maturing cone precursors during development, which was further validated by immunostaining. Notably, we found that the PI3K-Akt pathway was aberrantly deregulated and its activator spleen tyrosine kinase (SYK) was significantly up-regulated. In addition, SYK inhibitors led to remarkable cell apoptosis in cancerous organoids. In conclusion, we have established an organoid Rb model derived from genetically engineered hESCs in a dish that has enabled us to trace the cell of origin and to test novel candidate therapeutic agents for human Rb, shedding light on the development and therapeutics of other malignancies.


Asunto(s)
Células Madre Embrionarias Humanas/patología , Organoides/patología , Retinoblastoma/patología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Carcinogénesis/patología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones Endogámicos NOD , Mutagénesis/genética , Mutación/genética , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transcriptoma/genética
2.
Front Cell Dev Biol ; 8: 128, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32211407

RESUMEN

Although an increasing number of disease genes have been identified, the exact cellular mechanisms of retinitis pigmentosa (RP) remain largely unclear. Retinal organoids (ROs) derived from the induced pluripotent stem cells (iPSCs) of patients provide a potential but unvalidated platform for deciphering disease mechanisms and an advantageous tool for preclinical testing of new treatments. Notably, early-onset RP has been extensively recapitulated by patient-iPSC-derived ROs. However, it remains a challenge to model late-onset disease in a dish due to its chronicity, complexity, and instability. Here, we generated ROs from late-onset RP proband-derived iPSCs harboring a PDE6B mutation. Transcriptome analysis revealed a remarkably distinct gene expression profile in the patient ROs at differentiation day (D) 230. Changes in the expression genes regulating cGMP hydrolysis prompted the elevation of cGMP levels, which was verified by a cGMP enzyme-linked immunosorbent assay (ELISA) in patient ROs. Furthermore, significantly higher cGMP levels in patient ROs than in control ROs at D193 and D230 might lead to impaired formation of synaptic connections and the connecting cilium in photoreceptor cells. In this study, we established the first late-onset RP model with a consistent phenotype using an in vitro cell culture system and provided new insights into the PDE6B-related mechanism of RP.

3.
J Proteome Res ; 13(3): 1307-14, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24450392

RESUMEN

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


Asunto(s)
Adipocitos/metabolismo , Mitosis , Proteoma/análisis , Células 3T3-L1 , Adipocitos/citología , Animales , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Proliferación Celular , Cromatografía Liquida/métodos , Células Clonales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Quinasa/antagonistas & inhibidores , Piruvato Quinasa/genética , Piruvato Quinasa/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem
4.
Mol Cell Biol ; 34(3): 315-24, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24216764

RESUMEN

Adipogenesis is a multistep process by which 3T3-L1 preadipocytes differentiate into mature adipocytes through mitotic clonal expansion (MCE) and terminal differentiation. The CCAAT/enhancer-binding protein ß (C/EBPß) is an important transcription factor that takes part in both of these processes. C/EBPß not only transactivates C/EBPα and the peroxisome proliferator-activated receptor γ (PPARγ), which cause 3T3-L1 preadipocytes to enter terminal adipocyte differentiation, but also is required to activate cell cycle genes necessary for MCE. The identification of potential cofactors of C/EBPß will help to explain how C/EBPß undertakes these specialized roles during the different stages of adipogenesis. In this study, we found that activating transcription factor 5 (ATF5) can bind to the promoter of C/EBPα via its direct interaction with C/EBPß (which is mediated via the p300-dependent acetylation of ATF5), leading to enhanced C/EBPß transactivation of C/EBPα. We also show that p300 is important for the interaction of ATF5 with C/EBPß as well as for the binding activity of this complex on the C/EBPα promoter. Consistent with these findings, overexpression of ATF5 and an acetylated ATF5 mimic both promoted 3T3-L1 adipocyte differentiation, whereas short interfering RNA-mediated ATF5 downregulation inhibited this process. Furthermore, we show that the elevated expression of ATF5 is correlated with an obese phenotype in both mice and humans. In summary, we have identified ATF5 as a new cofactor of C/EBPß and examined how C/EBPß and ATF5 (acetylated by a p300-dependent mechanism) regulate the transcription of C/EBPα.


Asunto(s)
Factores de Transcripción Activadores/metabolismo , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/fisiología , Proteína p300 Asociada a E1A/metabolismo , Células 3T3-L1 , Acetilación , Factores de Transcripción Activadores/genética , Adipogénesis/genética , Adipogénesis/fisiología , Animales , Western Blotting , Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Diferenciación Celular/genética , Proteína p300 Asociada a E1A/genética , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Activación Transcripcional
5.
Am J Physiol Endocrinol Metab ; 304(9): E990-8, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23512806

RESUMEN

In 3T3-L1 preadipocyte differentiation, the CCAAT/enhancer-binding protein-ß (C/EBPß) is an important early transcription factor that activates cell cycle genes during mitotic clonal expansion (MCE), sequentially activating peroxisome proliferator-activated receptor-γ (PPARγ) and C/EBPα during terminal differentiation. Although C/EBPß acquires its DNA binding activity via dual phosphorylation at about 12-16 h postinduction, the expression of PPARγ and C/EBPα is not induced until 36-72 h. The delayed expression of PPARγ and C/EBPα ensures the progression of MCE, but the mechanism responsible for the delay remains elusive. We provide evidence that G9a, a major euchromatic methyltransferase, is transactivated by C/EBPß and represses PPARγ and C/EBPα through H3K9 dimethylation of their promoters during MCE. Inhibitor- or siRNA-mediated G9a downregulation modestly enhances PPARγ and C/EBPα expression and adipogenesis in 3T3-L1 preadipocytes. Conversely, forced expression of G9a impairs the accumulation of triglycerides. Thus, this study elucidates an epigenetic mechanism for the delayed expression of PPARγ and C/EBPα.


Asunto(s)
Adipocitos/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/biosíntesis , Proteína beta Potenciadora de Unión a CCAAT/genética , N-Metiltransferasa de Histona-Lisina/biosíntesis , N-Metiltransferasa de Histona-Lisina/genética , Células 3T3 , Adipogénesis/genética , Adipogénesis/fisiología , Animales , Compuestos Azo , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inmunoprecipitación de Cromatina , Colorantes , Remoción de Radical Alquila , Histonas/metabolismo , Metilación , Ratones , Mitosis/genética , Mitosis/fisiología , PPAR gamma/metabolismo , Plásmidos/genética , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Transactivadores , Activación Transcripcional/genética , Activación Transcripcional/fisiología
6.
Proc Natl Acad Sci U S A ; 110(9): E798-807, 2013 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-23388637

RESUMEN

Expression of bone morphogenetic protein 4 (BMP4) in adipocytes of white adipose tissue (WAT) produces "white adipocytes" with characteristics of brown fat and leads to a reduction of adiposity and its metabolic complications. Although BMP4 is known to induce commitment of pluripotent stem cells to the adipocyte lineage by producing cells that possess the characteristics of preadipocytes, its effects on the mature white adipocyte phenotype and function were unknown. Forced expression of a BMP4 transgene in white adipocytes of mice gives rise to reduced WAT mass and white adipocyte size along with an increased number of a white adipocyte cell types with brown adipocyte characteristics comparable to those of beige or brite adipocytes. These changes correlate closely with increased energy expenditure, improved insulin sensitivity, and protection against diet-induced obesity and diabetes. Conversely, BMP4-deficient mice exhibit enlarged white adipocyte morphology and impaired insulin sensitivity. We identify peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC1α) as the target of BMP signaling required for these brown fat-like changes in WAT. This effect of BMP4 on WAT appears to extend to human adipose tissue, because the level of expression of BMP4 in WAT correlates inversely with body mass index. These findings provide a genetic and metabolic basis for BMP4's role in altering insulin sensitivity by affecting WAT development.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Metabolismo Energético , Glucosa/metabolismo , Homeostasis , Células 3T3-L1 , Factor de Transcripción Activador 2/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/enzimología , Adipocitos Blancos/patología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/patología , Tejido Adiposo Pardo/ultraestructura , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/enzimología , Tejido Adiposo Blanco/ultraestructura , Animales , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Proteínas de Unión a Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Insulina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Tamaño de los Órganos/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Fenotipo , Delgadez/metabolismo , Delgadez/patología , Transactivadores/metabolismo , Factores de Transcripción , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Biol Chem ; 288(12): 8222-8230, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23389033

RESUMEN

BMP4 has been shown to induce C3H10T1/2 pluripotent stem cells to commit to adipocyte lineage. In addition to several proteins identified, microRNAs also play a critical role in the process. In this study, we identified microRNA-140 (miR-140) as a direct downstream component of the BMP4 signaling pathway during the commitment of C3H10T1/2 cells to adipocyte lineage. Overexpression of miR-140 in C3H10T1/2 cells promoted commitment, whereas knockdown of its expression led to impairment. Additional studies indicated that Ostm1 is a bona fide target of miR-140, which is significantly decreased during commitment, and Ostm1 was also demonstrated to function as an anti-adipogenic factor.


Asunto(s)
Adipocitos/metabolismo , Proteínas de la Membrana/genética , MicroARNs/fisiología , Células Madre Pluripotentes/fisiología , Interferencia de ARN , Células 3T3-L1 , Adipogénesis/genética , Animales , Secuencia de Bases , Proteína Morfogenética Ósea 4/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C3H , MicroARNs/genética , MicroARNs/metabolismo , Activación Transcripcional
8.
Mol Endocrinol ; 26(5): 798-808, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22422618

RESUMEN

Runx2, a runt-related transcriptional factor family member, is involved in the regulation of osteoblast differentiation. Interestingly, it is abundant in growth-arrested 3T3-L1 preadipocytes and was dramatically down-regulated during adipocyte differentiation. Knockdown of Runx2 expression promoted 3T3-L1 adipocyte differentiation, whereas overexpression inhibited adipocyte differentiation and promoted the trans-differentiation of 3T3-L1 preadipocytes to bone cells. Runx2 was down-regulated specifically by dexamethasone (DEX). Only type I Runx2 was expressed in 3T3-L1 preadipocytes. Using luciferase assay and chromatin immunoprecipitation-quantitative PCR analysis, it was found that DEX repressed this type of Runx2 at the transcriptional level through direct binding of the glucocorticoid receptor (GR) to a GR-binding element in the Runx2 P2 promoter. Further studies indicated that GR recruited histone deacetylase 1 to the Runx2 P2 promoter which then mediated the deacetylation of histone H4 and down-regulated Runx2 expression. Runx2 might play its repressive role through the induction of p27 expression, which blocked 3T3-L1 adipocyte differentiation by inhibiting mitotic clonal expansion. Taken together, we identified Runx2 as a new downstream target of DEX and explored a new pathway between DEX, Runx2, and p27 which contributed to the mechanism of the 3T3-L1 adipocyte differentiation.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Antiinflamatorios/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Dexametasona/farmacología , Regulación hacia Abajo/efectos de los fármacos , Células 3T3-L1 , Adipocitos/citología , Animales , Transdiferenciación Celular/efectos de los fármacos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/antagonistas & inhibidores , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
Biochem Biophys Res Commun ; 419(3): 573-7, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22374070

RESUMEN

TAZ (transcriptional co-activator with PDZ binding motif) is a transcriptional modulator of mesenchymal stem cell differentiation. We have found that TAZ was expressed in postconfluent 3T3-L1 preadipocytes and downregulated during differentiation. Downregulation of TAZ was specifically mediated by dexamethasone (DEX), one component of induction cocktails routinely used in adipocyte differentiation. DEX repressed the transcription of TAZ by direct binding of the glucocorticoid receptor (GR) to the GR binding element in its promoter. More importantly, overexpression of TAZ inhibited adipogenesis and promoted the trans-differentiation of preadipocytes into osteocytes. This establishes a new functional interaction between DEX and TAZ that contributes to the mechanism of adipogenesis.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Dexametasona/farmacología , Factores de Transcripción/antagonistas & inhibidores , Células 3T3-L1 , Aciltransferasas , Adipocitos/metabolismo , Adipogénesis/genética , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Regulación hacia Abajo , Ratones , Mitosis/efectos de los fármacos , Osteocitos/citología , Transcripción Genética/efectos de los fármacos
10.
Biochem Biophys Res Commun ; 419(3): 550-5, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22369944

RESUMEN

CCAAT/enhancer-binding protein (C/EBP) ß plays an important role in proliferation and differentiation of 3T3-L1 preadipocytes. C/EBPß is sequentially phosphorylated during the 3T3-L1 adipocyte differentiation program, first by MAPK/Cyclin A/cdk2 on Thr(188) and subsequently by GSK3ß on Ser(184) or Thr(179). Dual phosphorylation is critical for the gain of DNA binding activity of C/EBPß. In this manuscript, we found that phosphorylation also contributed to the stability of C/EBPß. Both ex vivo and in vitro experiments showed that phosphorylation by MAPK/Cyclin A/cdk2 and GSK3ß protected C/EBPß from µ-calpain-mediated proteolysis, while phosphorylation on Thr(188) by MAPK/Cyclin A/cdk2 contributed more to the stabilization of C/EBPß, Further studies indicated that phosphorylation mimic C/EBPß was insensitive to both calpain accelerator and calpain inhibitor. Thus, phosphorylation might contribute to the stability as well as the gain of DNA binding activity of C/EBPß.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Calpaína/metabolismo , Proteolisis , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Ciclina A/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Células HEK293 , Humanos , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Estabilidad Proteica
11.
Mol Biol Cell ; 22(13): 2165-74, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21562223

RESUMEN

CCAAT enhancer binding protein ß (C/EBPß) is required for both mitotic clonal expansion (MCE) and terminal differentiation during the 3T3-L1 adipocyte differentiation program. Whereas the mechanism of C/EBPß during terminal differentiation is well understood, the mechanism of C/EBPß in MCE is not. We provide evidence that histone H4, the most conserved cell cycle-related histone, the change of which is strictly correlated with DNA content change during the cell cycle, is transcriptionally activated by C/EBPß during MCE. Expression of histone H4 is increased at 16 h after induction when 3T3-L1 preadipocytes synchronously reenter S phase, which is correlated with the sequential phosphorylation and activation of C/EBPß, and expression was partially suppressed when A-C/EBP (dominant negative for C/EBP protein) was overexpressed. One C/EBP-binding site was identified in one of the histone H4 gene promoters (hist4h4), confirmed by both electrophoretic mobility shift assay and chromatin immunoprecipitation assay. C/EBP-binding sites were also found in 9 of 11 other histone H4 promoters, which can also be transactivated by C/EBPß. Knockdown of C/EBPß by stealth small interfering RNA partially decreased H4 gene expression and arrested cells in G1 phase as indicated by bromodeoxyuridine incorporation and fluorescence-activated cell sorting analysis of DNA content. This study provides new insights into why C/EBPß is required for MCE during 3T3-L1 adipocyte differentiation and why C/EBPß plays important roles in the proliferation of other cell types.


Asunto(s)
Adipocitos/citología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/fisiología , Histonas/genética , Histonas/metabolismo , Activación Transcripcional , Células 3T3-L1 , Adipocitos/metabolismo , Animales , Sitios de Unión , Proteína beta Potenciadora de Unión a CCAAT/antagonistas & inhibidores , Inmunoprecipitación de Cromatina/métodos , Citometría de Flujo/métodos , Fase G1/fisiología , Histonas/antagonistas & inhibidores , Ratones , Mitosis/fisiología , Fosforilación/fisiología , Regiones Promotoras Genéticas/genética , Fase S/fisiología
12.
BMC Dev Biol ; 10: 47, 2010 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-20459638

RESUMEN

BACKGROUND: Adipocyte hyperplasia is associated with obesity and arises due to adipogenic differentiation of resident multipotent stem cells in the vascular stroma of adipose tissue and remote stem cells of other organs. The mechanistic characterization of adipocyte differentiation has been researched in murine pre-adipocyte models (i.e. 3T3-L1 and 3T3-F442A), revealing that growth-arrest pre-adipocytes undergo mitotic clonal expansion and that regulation of the differentiation process relies on the sequential expression of three key transcription factors (C/EBPbeta, C/EBPalpha and PPARgamma). However, the mechanisms underlying adipocyte differentiation from multipotent stem cells, particularly human mesenchymal stem cells (hBMSCs), remain poorly understood. This study investigated cell cycle regulation and the roles of C/EBPbeta, C/EBPalpha and PPARgamma during adipocyte differentiation from hBMSCs. RESULTS: Utilising a BrdU incorporation assay and manual cell counting it was demonstrated that induction of adipocyte differentiation in culture resulted in 3T3-L1 pre-adipocytes but not hBMSCs undergoing mitotic clonal expansion. Knock-down and over-expression assays revealed that C/EBPbeta, C/EBPalpha and PPARgamma were required for adipocyte differentiation from hBMSCs. C/EBPbeta and C/EBPalpha individually induced adipocyte differentiation in the presence of inducers; PPARgamma alone initiated adipocyte differentiation but the cells failed to differentiate fully. Therefore, the roles of these transcription factors during human adipocyte differentiation are different from their respective roles in mouse. CONCLUSIONS: The characteristics of hBMSCs during adipogenic differentiation are different from those of murine cells. These findings could be important in elucidating the mechanisms underlying human obesity further.


Asunto(s)
Adipocitos/citología , Adipogénesis , Células de la Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Adipocitos/metabolismo , Adulto , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Ciclo Celular , Humanos , Masculino , Persona de Mediana Edad , PPAR gamma/metabolismo
13.
J Biol Chem ; 284(29): 19248-54, 2009 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-19478079

RESUMEN

CCAAT enhancer-binding protein (C/EBP)beta is a basic leucine zipper transcription factor family member, and can be phosphorylated, acetylated, and sumoylated. C/EBPbeta undergoes sequential phosphorylation during 3T3-L1 adipocyte differentiation. Phosphorylation on Thr(188) by MAPK or cyclin A/cdk2 primes the phosphorylations on Ser(184)/Thr(179) by GSK3beta, and these phosphorylations are required for the acquisition of DNA binding activity of C/EBPbeta. Here we show that C/EBPbeta is modified by O-GlcNAc, a dynamic single sugar modification found on nucleocytoplasmic proteins. The GlcNAcylation sites are Ser(180) and Ser(181), which are in the regulation domain and are very close to the phosphorylation sites (Thr(188), Ser(184), and Thr(179)) required for the gain of DNA binding activity. Both in vitro and ex vivo experiments demonstrate that GlcNAcylation on Ser(180) and Ser(181) prevents phosphorylation on Thr(188), Ser(184), and Thr(179), as indicated by the decreased relative phosphorylation and DNA binding activity of C/EBPbeta delayed the adipocyte differentiation program. Mutation of both Ser(180) and Ser(181) to Ala significantly increase the transcriptional activity of C/EBPbeta. These data suggest that GlcNAcylation regulates both the phosphorylation and DNA binding activity of C/EBPbeta.


Asunto(s)
Acetilglucosamina/metabolismo , Adipocitos/metabolismo , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , Células 3T3-L1 , Adipocitos/citología , Secuencia de Aminoácidos , Animales , Western Blotting , Proteína beta Potenciadora de Unión a CCAAT/química , Proteína beta Potenciadora de Unión a CCAAT/genética , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Glicosilación , Luciferasas/genética , Luciferasas/metabolismo , Espectrometría de Masas/métodos , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Datos de Secuencia Molecular , Oligonucleótidos/genética , Oligonucleótidos/metabolismo , Fragmentos de Péptidos/química , Fosforilación , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tripsina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...