Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Biol Cell ; 33(9): ar78, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35704469

RESUMEN

Cellular differentiation is characterized by changes in cell morphology that are largely determined by actin dynamics. We previously showed that depolymerization of the actin cytoskeleton triggers the differentiation of preadipocytes into mature adipocytes as a result of inhibition of the transcriptional coactivator activity of megakaryoblastic leukemia 1 (MKL1). The extracellular matrix (ECM) influences cell morphology via interaction with integrins, and reorganization of the ECM is associated with cell differentiation. Here we show that interaction between actin dynamics and ECM rearrangement plays a key role in adipocyte differentiation. We found that depolymerization of the actin cytoskeleton precedes disruption and degradation of fibrillar fibronectin (FN) structures at the cell surface after the induction of adipogenesis in cultured preadipocytes. A FN matrix suppressed both reorganization of the actin cytoskeleton into the pattern characteristic of adipocytes and terminal adipocyte differentiation, and these inhibitory effects were overcome by knockdown of integrin α5 (ITGα5). Peroxisome proliferator-activated receptor γ was required for down-regulation of FN during adipocyte differentiation, and MKL1 was necessary for the expression of ITGα5. Our findings suggest that cell-autonomous down-regulation of FN-ITGα5 interaction contributes to reorganization of the actin cytoskeleton and completion of adipocyte differentiation.


Asunto(s)
Adipogénesis , Fibronectinas , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Diferenciación Celular , Fibronectinas/metabolismo , Integrina alfa5/metabolismo
2.
Genes Cells ; 27(1): 5-13, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34695306

RESUMEN

Here, we established dedifferentiated fat (DFAT) cells from mature bovine adipocytes and then examined the effects of volatile fatty acids on the differentiation of these DFAT cells into adipocytes in vitro. When mature adipocytes were isolated from bovine adipose tissue and cultured using the ceiling culture method, they were dedifferentiated into fibroblast-like cells without lipid droplets. These fibroblast-like cells, termed bovine DFAT (b-DFAT) cells, actively proliferated. After adipogenic induction, increased expression of adipocyte-specific genes occurred in b-DFAT cells and they redifferentiated into adipocytes with an accumulation of lipid droplets in their cytoplasm. The effects of volatile fatty acids on adipocyte differentiation in b-DFAT cells were also examined. Specifically, acetate, butyrate, and propionate added to adipogenic induction medium significantly enhanced the adipogenesis of b-DFAT cells compared with that observed in control cells; the addition of 10-3  mol of acetate enhanced adipogenesis of b-DFAT cells to the greatest extent. These results suggest that b-DFAT cells derived from bovine mature adipocytes are appropriate for the study of bovine adipocyte differentiation and that the optimum concentration treatment of acetate, a major energy source for ruminants, promotes adipogenesis of b-DFAT cells in vitro.


Asunto(s)
Tejido Adiposo , Desdiferenciación Celular , Adipocitos , Animales , Bovinos , Diferenciación Celular , Ácidos Grasos Volátiles
3.
Genes Cells ; 25(12): 811-824, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33064855

RESUMEN

Mature adipocyte-derived dedifferentiated fat (DFAT) cells have been identified to possess similar multipotency to mesenchymal stem cells, but a method for converting DFAT cells into hepatocytes was previously unknown. Here, using comprehensive analysis of gene expression profiles, we have extracted three transcription factors, namely Foxa2, Hnf4a and Sall1 (FHS), that can convert DFAT cells into hepatocytes. Hepatogenic induction has converted FHS-infected DFAT cells into an epithelial-like morphological state and promoted the expression of hepatocyte-specific features. Furthermore, the DFAT-derived hepatocyte-like (D-Hep) cells catalyzed the detoxification of several compounds. These results indicate that the transduction of DFAT cells with three genes, which were extracted by comprehensive gene expression analysis, efficiently generated D-Hep cells with detoxification abilities similar to those of primary hepatocytes. Thus, D-Hep cells may be useful as a new cell source for surrogate hepatocytes and may be applied to drug discovery studies, such as hepatotoxicity screening and drug metabolism tests.


Asunto(s)
Tejido Adiposo/citología , Transdiferenciación Celular , Técnicas de Reprogramación Celular/métodos , Hepatocitos/citología , Tejido Adiposo/metabolismo , Animales , Células Cultivadas , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transducción Genética/métodos
4.
Genes Cells ; 25(3): 165-174, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31925986

RESUMEN

Adipocyte differentiation is accompanied by a pronounced change in the actin cytoskeleton characterized by the reorganization of filamentous (F)-actin stress fibers into cortical F-actin structures. We previously showed that depolymerization of F-actin stress fibers induced by inactivation of RhoA-ROCK (Rho-associated kinase) signaling acts as a trigger for adipocyte differentiation. The relevance and underlying mechanism of the formation of cortical F-actin structures from depolymerized actin during adipocyte differentiation have remained unclear, however. We have now examined the mechanistic relation between actin dynamics and adipogenic induction. Transient exposure to the actin-depolymerizing agent latrunculin A (LatA) supported the formation of adipocyte-associated cortical actin structures and the completion of terminal adipocyte differentiation in the presence of insulin, whereas long-term exposure to LatA prevented such actin reorganization as well as terminal adipogenesis. Moreover, these effects of insulin were prevented by inhibition of phosphatidylinositol 3-kinase (PI3K)-Rac1 signaling and the actin-related protein 2/3 (Arp2/3) complex which is a critical component of the cortical actin networks. Our findings thus suggest that the insulin-PI3K-Rac1 axis leads to the formation of adipocyte-associated cortical actin structures which is essential for the completion of adipocyte differentiation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Adipocitos/metabolismo , Insulina/metabolismo , Neuropéptidos/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Ratones
5.
J Oral Sci ; 60(1): 14-23, 2018 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-29479028

RESUMEN

Dedifferentiated fat (DFAT) cells were isolated from mature adipocytes using the ceiling culture method. Recently, we successfully isolated DFAT cells from adipocytes with a relatively small size (<40 µm). DFAT cells have a higher osteogenic potential than that of medium adipocytes. Therefore, the objective of this study was to determine the optimal concentration of collagenase solution for isolating small adipocytes from human buccal fat pads (BFPs). Four concentrations of collagenase solution (0.01%, 0.02%, 0.1%, and 0.5%) were used, and their effectiveness was assessed by the number of small adipocytes and DFAT cells isolated. The total number of floating adipocytes that dissociated with 0.02% collagenase was 2.5 times of that dissociated with 0.1% collagenase. The number of floating adipocytes with a diameter of ≤29 µm that dissociated with 0.02% collagenase was thrice of those dissociated with 0.1% and 0.5% collagenase. The number of DFAT cells that dissociated with 0.02% collagenase was 1.5 times of that dissociated with 0.1% collagenase. In addition, DFAT cells that dissociated with 0.02% collagenase had a higher osteogenic differentiation potential than those that dissociated with 0.1% collagenase. These results suggest that 0.02% is the optimal collagenase concentration for isolating small adipocytes from BFPs.


Asunto(s)
Adipocitos/citología , Mejilla , Colagenasas/metabolismo , Adipocitos/enzimología , Medios de Cultivo , Humanos
6.
Tissue Eng Part C Methods ; 22(3): 250-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26651216

RESUMEN

Dedifferentiated fat (DFAT) cells derived from mature adipocytes have mesenchymal stem cells' (MSCs) characteristics. Generally, mature adipocytes are 60-110 µm in diameter; however, association between adipocyte size and dedifferentiation efficiency is still unknown. This study, therefore, investigated the dedifferentiation efficiency of adipocytes based on cell diameter. Buccal fat pad was harvested from five human donors and dissociated by collagenase digestion. After exclusion of unwanted stromal cells by centrifugation, floating adipocytes were collected and their size distribution was analyzed. The floating adipocytes were then separated into two groups depending on cell size using 40- and 100-µm nylon mesh filters: cell diameters less than 40 µm (small adipocytes: S-adipocytes) and cell diameters of 40-100 µm (large adipocytes: L-adipocytes). Finally, we evaluated the efficiency of adipocyte dedifferentiation and then characterized the resultant DFAT cells. The S-adipocytes showed a higher capacity to dedifferentiate into DFAT cells (S-DFAT cells) compared to the L-adipocytes (L-DFAT cells). The S-DFAT cells also showed a relatively higher proportion of CD146-positive cells than L-DFAT cells, and exhibited more osteogenic differentiation ability based on the alkaline phosphatase activity and amount of calcium deposition. These results suggested that the S- and L-DFAT cells had distinct characteristics, and that the higher dedifferentiation potential of S-adipocytes compared to L-adipocytes gives the former group an advantage in yielding DFAT cells.


Asunto(s)
Tejido Adiposo/citología , Diferenciación Celular , Boca/citología , Osteogénesis , Adipocitos/citología , Adipogénesis , Adulto , Desdiferenciación Celular , Proliferación Celular , Separación Celular , Forma de la Célula , Tamaño de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Fibroblastos/citología , Citometría de Flujo , Fluorescencia , Perfilación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Humanos , Masculino , Reacción en Cadena en Tiempo Real de la Polimerasa , Adulto Joven
7.
Biochem Biophys Res Commun ; 454(2): 341-6, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25451251

RESUMEN

Mature adipocyte-derived dedifferentiated fat cells (DFAT) have a potential to be useful as new cell-source for cell-based therapy for spinal cord injury (SCI), but the mechanisms remain unclear. The objective of this study was to examine whether DFAT-induced functional recovery is achieved through remyelination and/or glial scar reduction in a mice model of SCI. To accomplish this we subjected adult female mice (n=22) to SCI. On the 8th day post-injury locomotor tests were performed, and the mice were randomly divided into two groups (control and DFAT). The DFAT group received stereotaxic injection of DFAT, while the controls received DMEM medium. Functional tests were conducted at repeated intervals, until the 36th day, and immunohistochemistry or staining was performed on the spinal cord sections. DFAT transplantation significantly improved locomotor function of their hindlimbs, and promoted remyelination and glial scar reduction, when compared to the controls. There were significant and positive correlations between promotion of remyelination or/and reduction of glial scar, and recovery of locomotor function. Furthermore, transplanted DFAT expressed markers for neuron, astrocyte, and oligodendrocyte, along with neurotrophic factors, within the injured spinal cord. In conclusion, DFAT-induced functional recovery in mice after SCI is probably mediated by both cell-autonomous and cell-non-autonomous effects on remyelination of the injured spinal cord.


Asunto(s)
Adipocitos/trasplante , Vaina de Mielina/patología , Recuperación de la Función , Traumatismos de la Médula Espinal/terapia , Médula Espinal/fisiopatología , Adipocitos/citología , Animales , Desdiferenciación Celular , Diferenciación Celular , Cicatriz/fisiopatología , Cicatriz/terapia , Femenino , Locomoción , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Factores de Crecimiento Nervioso/análisis , Neurogénesis , Neuronas/citología , Médula Espinal/citología , Médula Espinal/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Regeneración de la Medula Espinal
8.
Nat Commun ; 5: 3368, 2014 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-24569594

RESUMEN

Cellular differentiation is regulated through activation and repression of defined transcription factors. A hallmark of differentiation is a pronounced change in cell shape, which is determined by dynamics of the actin cytoskeleton. Here we show that regulation of the transcriptional coactivator MKL1 (megakaryoblastic leukemia 1) by actin cytoskeleton dynamics drives adipocyte differentiation mediated by peroxisome proliferator-activated receptor γ (PPARγ), a master transcriptional regulator of adipogenesis. Induction of adipocyte differentiation results in disruption of actin stress fibres through downregulation of RhoA-ROCK signalling. The consequent rapid increase in monomeric G-actin leads to the interaction of G-actin with MKL1, which prevents nuclear translocation of MKL1 and allows expression of PPARγ followed by adipogenic differentiation. Moreover, we found that MKL1 and PPARγ act in a mutually antagonistic manner in the adipocytic differentiation programme. Our findings thus provide new mechanistic insight into the relation between the dynamics of cell shape and transcriptional regulation during cellular differentiation.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Adipocitos/metabolismo , Diferenciación Celular , Transactivadores/metabolismo , Células 3T3-L1 , Actinas/genética , Actinas/metabolismo , Adipocitos/citología , Animales , Línea Celular , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Immunoblotting , Cinética , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Análisis de Secuencia por Matrices de Oligonucleótidos , PPAR gamma/genética , PPAR gamma/metabolismo , Unión Proteica , Transporte de Proteínas , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Imagen de Lapso de Tiempo , Transactivadores/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
9.
Biochem J ; 447(2): 239-48, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22839299

RESUMEN

Transdifferentiation is the conversion of cells from one differentiated cell type into another. How functionally differentiated cells already committed to a specific cell lineage can transdifferentiate into other cell types is a key question in cell biology and regenerative medicine. In the present study we show that porcine ovarian follicular GCs (granulosa cells) can transdifferentiate into osteoblasts in vitro and in vivo. Pure GCs isolated and cultured in Dulbecco's modified Eagle's medium supplemented with 20% FBS (fetal bovine serum) proliferated and dedifferentiated into fibroblast-like cells. We referred to these cells as DFOG (dedifferentiated follicular granulosa) cells. Microarray analysis showed that DFOG cells lost expression of GC-specific marker genes, but gained the expression of osteogenic marker genes during dedifferentiation. After osteogenic induction, DFOG cells underwent terminal osteoblast differentiation and matrix mineralization in vitro. Furthermore, when DFOG cells were transplanted subcutaneously into SCID mice, these cells formed ectopic osteoid tissue. These results indicate that DFOG cells derived from GCs can differentiate into osteoblasts in vitro and in vivo. We suggest that GCs provide a useful model for studying the mechanisms of transdifferentiation into other cell lineages in functionally differentiated cells.


Asunto(s)
Transdiferenciación Celular , Células de la Granulosa/citología , Osteoblastos/citología , Ovario/citología , Animales , Huesos/fisiología , Femenino , Ratones , Ratones SCID , Osteoblastos/trasplante , Sus scrofa
10.
Biochem Biophys Res Commun ; 407(3): 562-7, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21419102

RESUMEN

Cellular dedifferentiation signifies the withdrawal of cells from a specific differentiated state to a stem cell-like undifferentiated state. However, the mechanism of dedifferentiation remains obscure. Here we performed comparative transcriptome analyses during dedifferentiation in mature adipocytes (MAs) to identify the transcriptional signatures of multipotent dedifferentiated fat (DFAT) cells derived from MAs. Using microarray systems, we explored similarly expressed as well as significantly differentially expressed genes in MAs during dedifferentiation. This analysis revealed significant changes in gene expression during this process, including a significant reduction in expression of genes for lipid metabolism concomitantly with a significant increase in expression of genes for cell movement, cell migration, tissue developmental processes, cell growth, cell proliferation, cell morphogenesis, altered cell shape, and cell differentiation. Our observations indicate that the transcriptional signatures of DFAT cells derived from MAs are summarized in terms of a significant decrease in functional phenotype-related genes and a parallel increase in cell proliferation, altered cell morphology, and regulation of the differentiation of related genes. A better understanding of the mechanisms involved in dedifferentiation may enable scientists to control and possibly alter the plasticity of the differentiated state, which may lead to benefits not only in stem cell research but also in regenerative medicine.


Asunto(s)
Adipocitos/citología , Desdiferenciación Celular/genética , Células Madre Multipotentes/metabolismo , Animales , Movimiento Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Morfogénesis/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Porcinos
11.
Cell Struct Funct ; 33(2): 211-22, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19088398

RESUMEN

We investigated whether de-differentiated fat (DFAT) cells, a mature adipocyte-derived preadipocyte cell line, can be induced to trans-differentiate into osteoblasts in vitro and in vivo. All-trans retinoic acid (RA) induced expression of osteoblast-specific mRNAs encoding Cbfa1/Runx2, osterix, alkaline phosphatase, osteopontin, parathyroid hormone receptor, and osteocalcin in the DFAT cells, but did not induce the expression of adipocyte-specific mRNAs encoding PPARgamma2, C/EBPalpha, and GLUT4. Moreover, alkaline phosphatase activity was expressed in DFAT cells and the cells underwent mineralization of the bone matrix in vitro. Furthermore, when DFAT cells were transplanted subcutaneously into C57BL/6N mice in diffusion chambers, these cells formed ectopic osteoid tissue without any host cell-invasion of the chambers. These results indicate that DFAT cells derived from mature adipocytes can be converted into fully differentiated osteoblasts in vitro and in vivo using RA. DFAT cells provide a unique model for studying the lineage commitment of the adipocytes and osteoblasts derived from mesenchymal stem cells. Identification of the pathways that regulate these processes could lead to the development of new therapeutic strategies for control of unwarranted growth of bone and adipose tissue.


Asunto(s)
Adipocitos/citología , Adipocitos/efectos de los fármacos , Desdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Tretinoina/farmacología , Adipogénesis/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Animales , Biomarcadores/metabolismo , Trasplante Óseo , Huesos/efectos de los fármacos , Huesos/metabolismo , Línea Celular , Trasplante de Células , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Subcutáneas , Ratones , Minerales/metabolismo , Osteogénesis/efectos de los fármacos , Cavidad Peritoneal/citología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Tretinoina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...