Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Stem Cells ; 32(5): 1136-48, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24446210

RESUMEN

The cellular microenvironment plays a relevant role in cancer development. We have reported that mesenchymal stromal/stem cells (MSCs) deficient for p53 alone or together with RB (p53(-/-)RB(-/-)) originate leiomyosarcoma after subcutaneous (s.c.) inoculation. Here, we show that intrabone or periosteal inoculation of p53(-/-) or p53(-/-)RB(-/-) bone marrow- or adipose tissue-derived MSCs originated metastatic osteoblastic osteosarcoma (OS). To assess the contribution of bone environment factors to OS development, we analyzed the effect of the osteoinductive factor bone morphogenetic protein-2 (BMP-2) and calcified substrates on p53(-/-)RB(-/-) MSCs. We show that BMP-2 upregulates the expression of osteogenic markers in a WNT signaling-dependent manner. In addition, the s.c. coinfusion of p53(-/-)RB(-/-) MSCs together with BMP-2 resulted in appearance of tumoral osteoid areas. Likewise, when p53(-/-)RB(-/-) MSCs were inoculated embedded in a calcified ceramic scaffold composed of hydroxyapatite and tricalciumphosphate (HA/TCP), tumoral bone formation was observed in the surroundings of the HA/TCP scaffold. Moreover, the addition of BMP-2 to the ceramic/MSC implants further increased the tumoral osteoid matrix. Together, these data indicate that bone microenvironment signals are essential to drive OS development.


Asunto(s)
Neoplasias Óseas/patología , Huesos/patología , Microambiente Celular , Células Madre Mesenquimatosas/patología , Osteosarcoma/patología , Animales , Western Blotting , Proteína Morfogenética Ósea 2/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Huesos/efectos de los fármacos , Huesos/metabolismo , Fosfatos de Calcio/química , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Cerámica/química , Durapatita/química , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Andamios del Tejido/química , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
2.
Stem Cells ; 31(10): 2061-72, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23836491

RESUMEN

Increasing evidence supports that mesenchymal stromal/stem cells (MSCs) may represent the target cell for sarcoma development. Although different sarcomas have been modeled in mice upon expression of fusion oncogenes in MSCs, sarcomagenesis has not been successfully modeled in human MSCs (hMSCs). We report that FUS-CHOP, a hallmark fusion gene in mixoid liposarcoma (MLS), has an instructive role in lineage commitment, and its expression in hMSC sequentially immortalized/transformed with up to five oncogenic hits (p53 and Rb deficiency, hTERT over-expression, c-myc stabilization, and H-RAS(v12) mutation) drives the formation of serially transplantable MLS. This is the first model of sarcoma based on the expression of a sarcoma-associated fusion protein in hMSC, and allowed us to unravel the differentiation processes and signaling pathways altered in the MLS-initiating cells. This study will contribute to test novel therapeutic approaches and constitutes a proof-of-concept to use hMSCs as target cell for modeling other fusion gene-associated human sarcomas.


Asunto(s)
Liposarcoma Mixoide/metabolismo , Células Madre Mesenquimatosas/patología , Proteínas de Fusión Oncogénica/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Factor de Transcripción CHOP/metabolismo , Adipogénesis , Animales , Carcinogénesis/metabolismo , Línea Celular Transformada , Expresión Génica , Humanos , Liposarcoma Mixoide/patología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Proteínas de Fusión Oncogénica/genética , Proteína FUS de Unión a ARN/genética , Transducción de Señal , Factor de Transcripción CHOP/genética , Transcriptoma
3.
Stem Cells ; 29(2): 179-92, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21732477

RESUMEN

Human sarcomas have been modeled in mice by expression of specific fusion genes in mesenchymal stem cells (MSCs). However, sarcoma models based on human MSCs are still missing. We attempted to develop a model of liposarcoma by expressing FUS (FUsed in Sarcoma; also termed TLS, Translocated in LipoSarcoma)-CHOP (C/EBP HOmologous Protein; also termed DDIT3, DNA Damage-Inducible Transcript 3), a hallmark mixoid liposarcoma-associated fusion oncogene, in wild-type and p53-deficient mouse and human adipose-derived mesenchymal stem/stromal cells (ASCs). FUS-CHOP induced liposarcoma-like tumors when expressed in p53(-/-) but not in wild-type (wt) mouse ASCs (mASCs). In the absence of FUS-CHOP, p53(-/-) mASCs forms leiomyosarcoma, indicating that the expression of FUS-CHOP redirects the tumor genesis/phenotype. FUS-CHOP expression in wt mASCs does not initiate sarcomagenesis, indicating that p53 deficiency is required to induce FUS-CHOP-mediated liposarcoma in fat-derived mASCs. In a human setting, p53-deficient human ASCs (hASCs) displayed a higher in vitro growth rate and a more extended lifespan than wt hASCs. However, FUS-CHOP expression did not induce further changes in culture homeostasis nor initiated liposarcoma in either wt or p53-depleted hASCs. These results indicate that FUS-CHOP expression in a p53-deficient background is sufficient to initiate liposarcoma in mouse but not in hASCs, suggesting the need of additional cooperating mutations in hASCs. A microarray gene expression profiling has shed light into the potential deregulated pathways in liposarcoma formation from p53-deficient mASCs expressing FUS-CHOP, which might also function as potential cooperating mutations in the transformation process from hASCs.


Asunto(s)
Adipocitos/metabolismo , Liposarcoma/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Factor de Transcripción CHOP/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Adipocitos/citología , Animales , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Liposarcoma/genética , Liposarcoma/patología , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína FUS de Unión a ARN/genética , Proteínas Recombinantes de Fusión/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción CHOP/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
4.
Stem Cells ; 29(2): 251-62, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21732483

RESUMEN

Human ESCs provide access to the earliest stages of human development and may serve as an unlimited source of functional cells for future cell therapies. The optimization of methods directing the differentiation of human embryonic stem cells (hESCs) into tissue-specific precursors becomes crucial. We report an efficient enrichment of mesenchymal stem cells (MSCs) from hESCs through specific inhibition of SMAD-2/3 signaling. Human ESC-derived MSCs (hESC-MSCs) emerged as a population of fibroblastoid cells expressing a MSC phenotype: CD73+ CD90+ CD105+ CD44+ CD166+ CD45- CD34- CD14- CD19- human leucocyte antigen-DR (HLA-DR)-. After 28 days of SMAD-2/3 inhibition, hESC cultures were enriched (>42%) in multipotent MSCs. CD73+CD90+ hESC-MSCs were fluorescence activated cell sorting (FACS)-isolated and long-term cultures were established and maintained for many passages displaying a faster growth than somatic tissue-derived MSCs while maintaining MSC morphology and phenotype. They displayed osteogenic, adipogenic, and chondrocytic differentiation potential and exhibited potent immunosuppressive and anti-inflammatory properties in vitro and in vivo, where hESC-MSCs were capable of protecting against an experimental model of inflammatory bowel disease. Interestingly, the efficient enrichment of hESCs into MSCs through inhibition of SMAD-2/3 signaling was not reproducible with distinct induced pluripotent stem cell lines. Our findings provide mechanistic insights into the differentiation of hESCs into immunosuppressive and anti-inflammatory multipotent MSCs with potential future clinical applications.


Asunto(s)
Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Enfermedades Inflamatorias del Intestino/prevención & control , Células Madre Multipotentes/inmunología , Células Madre Multipotentes/metabolismo , Proteína Smad2/antagonistas & inhibidores , Proteína smad3/antagonistas & inhibidores , Antígenos CD , Benzamidas/farmacología , Diferenciación Celular/fisiología , Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Dioxoles/farmacología , Células Madre Embrionarias/citología , Citometría de Flujo , Humanos , Terapia de Inmunosupresión , Enfermedades Inflamatorias del Intestino/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/citología , Transducción de Señal
5.
Environ Sci Pollut Res Int ; 26(12): 11605-11617, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30484048

RESUMEN

Pyrolysis of waste materials to produce biochar is an excellent and suitable alternative supporting a circular bio-based economy. One of the properties attributed to biochar is the capacity for sorbing organic contaminants, which is determined by its composition and physicochemical characteristics. In this study, the capacity of waste-derived biochar to retain volatile fuel organic compounds (benzene, toluene, ethylbenzene and xylene (BTEX) and fuel oxygenates (FO)) from artificially contaminated water was assessed using batch-based sorption experiments. Additionally, the sorption isotherms were established. The results showed significant differences between BTEX and FO sorption on biochar, being the most hydrophobic and non-polar contaminants those showing the highest retention. Furthermore, the sorption process reflected a multilayer behaviour and a relatively high sorption capacity of the biochar materials. Langmuir and Freundlich models were adequate to describe the experimental results and to detect general differences in the sorption behaviour of volatile fuel organic compounds. It was also observed that the feedstock material and biochar pyrolysis conditions had a significant influence in the sorption process. The highest sorption capacity was found in biochars produced at high temperature (> 400 °C) and thus rich in aromatic C, such as eucalyptus and corn cob biochars. Overall, waste-derived biochar offers a viable alternative to be used in the remediation of volatile fuel organic compounds from water due to its high sorption capacity.


Asunto(s)
Restauración y Remediación Ambiental/métodos , Compuestos Orgánicos Volátiles/química , Contaminantes Químicos del Agua/química , Adsorción , Carbón Orgánico , Calor , Compuestos Orgánicos Volátiles/análisis , Contaminantes Químicos del Agua/análisis
6.
Cell Res ; 22(1): 62-77, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21931359

RESUMEN

Because of their unique properties, multipotent mesenchymal stem cells (MSCs) represent one of the most promising adult stem cells being used worldwide in a wide array of clinical applications. Overall, compelling evidence supports the long-term safety of ex vivo expanded human MSCs, which do not seem to transform spontaneously. However, experimental data reveal a link between MSCs and cancer, and MSCs have been reported to inhibit or promote tumor growth depending on yet undefined conditions. Interestingly, solid evidence based on transgenic mice and genetic intervention of MSCs has placed these cells as the most likely cell of origin for certain sarcomas. This research area is being increasingly explored to develop accurate MSC-based models of sarcomagenesis, which will be undoubtedly valuable in providing a better understanding about the etiology and pathogenesis of mesenchymal cancer, eventually leading to the development of more specific therapies directed against the sarcoma-initiating cell. Unfortunately, still little is known about the mechanisms underlying MSC transformation and further studies are required to develop bona fide sarcoma models based on human MSCs. Here, we comprehensively review the existing MSC-based models of sarcoma and discuss the most common mechanisms leading to tumoral transformation of MSCs and sarcomagenesis.


Asunto(s)
Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica , Células Madre Mesenquimatosas/patología , Células Madre Multipotentes/patología , Sarcoma Experimental/patología , Animales , Puntos de Control del Ciclo Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Fusión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Células Madre Multipotentes/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Sarcoma Experimental/metabolismo , Transducción de Señal
7.
Cancer Res ; 70(10): 4185-94, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20442289

RESUMEN

Sarcomas have been modeled in mice by the expression of specific fusion genes in mesenchymal stem cells (MSC), supporting the concept that MSCs might be the target initiating cell in sarcoma. In this study, we evaluated the potential oncogenic effects of p53 and/or retinoblastoma (Rb) deficiency in MSC transformation and sarcomagenesis. We derived wild-type, p53(-/-), Rb(-/-), and p53(-/-)Rb(-/-) MSC cultures and fully characterized their in vitro growth properties and in vivo tumorigenesis capabilities. In contrast with wild-type MSCs, Rb(-/-), p53(-/-), and p53(-/-)Rb(-/-) MSCs underwent in vitro transformation and showed severe alterations in culture homeostasis. More importantly, p53(-/-) and p53(-/-)Rb(-/-) MSCs, but not Rb(-/-) MSCs, were capable of tumor development in vivo after injection into immunodeficient mice. p53(-/-) or p53(-/-)Rb(-/-) MSCs originated leiomyosarcoma-like tumors, linking this type of smooth muscle sarcoma to p53 deficiency in fat tissue-derived MSCs. Sca1+ and Sca1 low/- cell populations isolated from ex vivo-established, transformed MSC lines from p53(-/-)Rb(-/-) tumors showed identical sarcomagenesis potential, with 100% tumor penetrance and identical latency, tumor weight, and histologic profile. Our findings define the differential roles of p53 and Rb in MSC transformation and offer proof-of-principle that MSCs could provide useful tools to dissect the sarcoma pathogenesis.


Asunto(s)
Transformación Celular Neoplásica , Leiomiosarcoma/patología , Células Madre Mesenquimatosas/patología , Proteína de Retinoblastoma/fisiología , Sarcoma Experimental/patología , Proteína p53 Supresora de Tumor/fisiología , Animales , Western Blotting , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Técnica del Anticuerpo Fluorescente , Técnicas In Vitro , Integrasas/metabolismo , Leiomiosarcoma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoma Experimental/metabolismo
8.
Cell Res ; 19(6): 698-709, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19308090

RESUMEN

A paracrine regulation was recently proposed in human embryonic stem cells (hESCs) grown in mouse embryonic fibroblast (MEF)-conditioned media (MEF-CM), where hESCs spontaneously differentiate into autologous fibroblast-like cells to maintain culture homeostasis by producing TGF-beta and insulin-like growth factor-II (IGF-II) in response to basic fibroblast growth factor (bFGF). Although the importance of TGF-beta family members in the maintenance of pluripotency of hESCs is widely established, very little is known about the role of IGF-II. In order to ease hESC culture conditions and to reduce xenogenic components, we sought (i) to determine whether hESCs can be maintained stable and pluripotent using CM from human foreskin fibroblasts (HFFs) and human mesenchymal stem cells (hMSCs) rather than MEF-CM, and (ii) to analyze whether the cooperation of bFGF with TGF-beta and IGF-II to maintain hESCs in MEF-CM may be extrapolated to hESCs maintained in allogeneic mesenchymal stem cell (MSC)-CM and HFF-CM. We found that MSCs and HFFs express all FGF receptors (FGFR1-4) and specifically produce TGF-beta in response to bFGF. However, HFFs but not MSCs secrete IGF-II. Despite the absence of IGF-II in MSC-CM, hESC pluripotency and culture homeostasis were successfully maintained in MSC-CM for over 37 passages. Human ESCs derived on MSCs and hESCs maintained in MSC-CM retained hESC morphology, euploidy, expression of surface markers and transcription factors linked to pluripotency and displayed in vitro and in vivo multilineage developmental potential, suggesting that IGF-II may be dispensable for hESC pluripotency. In fact, IGF-II blocking had no effect on the homeostasis of hESC cultures maintained either on HFF-CM or on MSC-CM. These data indicate that hESCs are successfully maintained feeder-free with IGF-II-lacking MSC-CM, and that the previously proposed paracrine mechanism by which bFGF cooperates with TGF-beta and IGF-II in the maintenance of hESCs in MEF-CM may not be fully extrapolated to hESCs maintained in CM from human MSCs.


Asunto(s)
Células Madre Embrionarias/citología , Factor II del Crecimiento Similar a la Insulina/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Medios de Cultivo Condicionados , Células Madre Embrionarias/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Mesenquimatosas/citología
9.
Neoplasia ; 11(4): 397-407, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19308294

RESUMEN

There is growing evidence about the role of mesenchymal stem cells (MSCs) as cancer stem cells in many sarcomas. Nevertheless, little is still known about the cellular and molecular mechanisms underlying MSCs transformation. We aimed at investigating the role of p53 and p21, two important regulators of the cell cycle progression and apoptosis normally involved in protection against tumorigenesis. Mesenchymal stem cells from wild-type, p21(-/-)p53(+/+), and p21(-/-)p53(+/-) mice were cultured in vitro and analyzed for the appearance of tumoral transformation properties after low, medium, and high number of passages both in vitro and in vivo. Wild-type or p21(-/-)p53(+/+) MSCs did not show any sign of tumoral transformation. Indeed, after short-term in vitro culture, wild-type MSCs became senescent, and p21(-/-)p53(+/+) MSCs showed an elevated spontaneous apoptosis rate. Conversely, MSCs carrying a mutation in one allele of the p53 gene (p21(-/-)p53(+/-) MSCs) completely lost p53 expression after in vitro long-term culture. Loss of p53 was accompanied by a significant increase in the growth rate, gain of karyotypic instability, loss of p16 expression, and lack of senescence response. Finally, these cells were able to form fibrosarcomas partially differentiated into different mesenchymal lineages when injected in immunodeficient mice both after subcutaneous and intrafemoral injection. These findings show that MSCs are very sensitive to mutations in genes involved in cell cycle control and that these deficiencies can be at the origin of some mesodermic tumors.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Células Madre Mesenquimatosas/patología , Células Madre Neoplásicas/patología , Proteína p53 Supresora de Tumor/genética , Animales , Western Blotting , Transformación Celular Neoplásica/metabolismo , Senescencia Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Citometría de Flujo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Mutantes , Mutación , Células Madre Neoplásicas/metabolismo
11.
Development ; 135(19): 3197-207, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18715948

RESUMEN

The Iroquois (Irx) genes encode evolutionary conserved homeoproteins. We report that Xenopus genes Irx1 and Irx3 are expressed and required during different stages of Xenopus pronephros development. They are initially expressed during mid-neurulation in domains extending over most of the prospective pronephric territory. Expression onset takes place after kidney anlage specification, but before pronephric organogenesis occurs. Later, during nephron segmentation, expression becomes restricted to the intermediate tubule region of the proximal-distal axis. Loss- and gain-of-function analyses, performed with specific morpholinos and inducible wild-type and dominant-negative constructs, reveal a dual requirement for Irx1 and Irx3 during pronephros development. During neurula stages, these genes maintain the specification of the pronephric territory and define its size. This seems to occur, at least in part, through positive regulation of Bmp signalling. Subsequently, Irx genes are required for proper formation of the intermediate tubule. Finally, we find that retinoic acid signalling activates both Irx1 and Irx3 genes in the pronephros.


Asunto(s)
Genes Homeobox , Riñón/embriología , Riñón/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Animales , Secuencia de Bases , Tipificación del Cuerpo/efectos de los fármacos , Tipificación del Cuerpo/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes Homeobox/efectos de los fármacos , Proteínas de Homeodominio/antagonistas & inhibidores , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Tretinoina/farmacología , Proteínas de Xenopus/antagonistas & inhibidores , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
12.
PLoS One ; 3(9): e3294, 2008 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-18820729

RESUMEN

Developmental genes are silenced in embryonic stem cells by a bivalent histone-based chromatin mark. It has been proposed that this mark also confers a predisposition to aberrant DNA promoter hypermethylation of tumor suppressor genes (TSGs) in cancer. We report here that silencing of a significant proportion of these TSGs in human embryonic and adult stem cells is associated with promoter DNA hypermethylation. Our results indicate a role for DNA methylation in the control of gene expression in human stem cells and suggest that, for genes repressed by promoter hypermethylation in stem cells in vivo, the aberrant process in cancer could be understood as a defect in establishing an unmethylated promoter during differentiation, rather than as an anomalous process of de novo hypermethylation.


Asunto(s)
Metilación de ADN , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Neoplasias/genética , Neoplasias/metabolismo , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Silenciador del Gen , Genes Relacionados con las Neoplasias , Células HL-60 , Células HeLa , Humanos , Regiones Promotoras Genéticas , Factores de Tiempo , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA