Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Immunol ; 191(9): 4619-27, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24068664

RESUMEN

FOXP3(+) regulatory T cells (Tregs) are critical regulators of self-tolerance and immune homeostasis. In mice and humans, two subsets of FOXP3(+) Tregs have been defined based on their differential expression of Helios, a transcription factor of the Ikaros family. Whereas the origin, specificity, and differential function of the two subsets are as yet a source of controversy, their characterization thus far has been limited by the absence of surface markers to distinguish them. In this article, we show that human memory Helios(+) and Helios(-) Tregs are phenotypically distinct and can be separated ex vivo based on their differential expression of IL-1RI, which is restricted to Helios(-) Tregs, in combination with CCR7. The two populations isolated using this strategy are distinct with respect to the expression of other Ikaros family members. Namely, whereas Eos, which has been reported to mediate FOXP3-dependent gene silencing, is expressed in Helios(+) Tregs, Aiolos, which is involved in the differentiation of TH17 and induced Tregs, is instead expressed in Helios(-) Tregs. In addition, whereas both subsets are suppressive ex vivo, Helios(-) Tregs display increased suppressive capacity in comparison to Helios(+) Tregs, but respond to IL-1ß by downregulating their suppressive activity. Together, these data support the concept that human Helios(-) memory Tregs encompass induced Tregs that can readily respond to changes in the environment by modulating their suppressive capacity.


Asunto(s)
Factor de Transcripción Ikaros/metabolismo , Interleucina-1beta/metabolismo , Receptores Tipo I de Interleucina-1/metabolismo , Linfocitos T Reguladores/metabolismo , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Humanos , Memoria Inmunológica , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Receptores CCR7/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología
2.
Eur J Immunol ; 43(6): 1667-75, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23519784

RESUMEN

Patrolling Ly6C(-) monocytes are blood-circulating cells that play a role in inflammation and in the defense against pathogens. Here, we show that similar to natural killer (NK) cells, patrolling monocytes express high levels of S1PR5, a G-coupled receptor for sphingosine-1 phosphate. We found that S1pr5(-/-) mice lack peripheral Ly6C(-) monocytes but have a normal number of these cells in the bone marrow (BM). Various lines of evidence exclude a direct contribution of S1PR5 in the survival of Ly6C(-) monocytes at the periphery. Rather, our data support a role for S1PR5 in the egress of Ly6C(-) monocytes from the BM. In particular, we observed a reduced frequency of patrolling monocytes in BM sinusoids of S1PR5 KO mice. Unexpectedly, S1P was not a chemoattractant for patrolling monocytes and had no significant effect on their viability in vitro. Moreover, the disruption of S1P gradients in vivo did not alter Ly6C(-) monocyte trafficking and viability. These data suggest that S1PR5 regulates the trafficking of monocytes via a mechanism independent of S1P gradients.


Asunto(s)
Antígenos Ly/metabolismo , Médula Ósea/inmunología , Monocitos/inmunología , Receptores de Lisoesfingolípidos/metabolismo , Animales , Circulación Sanguínea , Movimiento Celular/inmunología , Supervivencia Celular , Células Cultivadas , Femenino , Homeostasis , Vigilancia Inmunológica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Lisoesfingolípidos/genética
3.
Clin Cancer Res ; 15(10): 3519-29, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19451595

RESUMEN

PURPOSE: We have investigated whether the folate supplementation could be used to limit the aggressiveness of glioma through the DNA remethylation because (a) the cancer genome is characterized by a low level of DNA methylation (or 5-methylcytosine, 5 mC); and (b) folate is the main generator of S-adenosyl-methionine, the methyl donor molecule in the DNA methylation reaction catalyzed by the DNA methyltranferases. EXPERIMENTAL DESIGN: The effects of folate supplementations were analyzed on the global DNA methylation status, the methylation status of DNA repeat element, the sensitivity of temozolomide-induced apoptosis, and the proliferation index of glioma cells. Finally, we analyzed whether the DNA methylation level could be used as a prognostic factor and/or a biomarker in an antiglioma therapy using folate supplementation as an adjuvant. RESULTS: Our data show that gliomagenesis is accompanied by a reduction in 5 mC levels and that this low level of 5 mC is a poor prognostic factor in Glioblastoma Multiforme patients. We also show that folate supplementation enhanced the DNA remethylation through the Sp1/Sp3-mediated transcriptional up-regulation of genes coding for Dnmt3a and Dnmt3b proteins, two de novo methyltranferases. Finally, we show that the folate-induced DNA methylation limits proliferation and increases the sensitivity to temozolomide-induced apoptosis in glioma cells through methylation of the genes implicated in these processes (PDGF-B, MGMT, survivin, and bcl-w). CONCLUSION: This study suggests that folate supplementation could be a promising adjuvant for the future design of antiglioma therapies in preclinical and/or clinical studies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proliferación Celular , Metilación de ADN , Ácido Fólico/administración & dosificación , Glioma/genética , Anciano , Apoptosis/efectos de los fármacos , Apoptosis/genética , Becaplermina , Línea Celular Tumoral , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Suplementos Dietéticos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Proteínas Inhibidoras de la Apoptosis , Proteínas Asociadas a Microtúbulos/genética , Persona de Mediana Edad , Invasividad Neoplásica , Factor de Crecimiento Derivado de Plaquetas/genética , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis , Secuencias Repetitivas de Ácidos Nucleicos/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp3/metabolismo , Análisis de Supervivencia , Survivin , Temozolomida , Proteínas Supresoras de Tumor/genética , ADN Metiltransferasa 3B
4.
Int J Cancer ; 123(2): 288-295, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18412241

RESUMEN

Our study explored the influence of diet on gliomagenesis and associated systemic effects (SE) in rats. The experimental diet contained various ingredients supposed to interfere with carcinogenesis, mainly phytochemicals (PtcD for phytochemical diet) and its effects were compared to those of the same diet without the phytochemicals (BD for basal diet). Glioma was induced by ethylnitrosourea to pregnant females fed the diets from the start of gestation until the moment of sacrifice of the offpsrings. In male rats fed the PtcD or the BD the incidence of gliomas was markedly reduced compared to rats fed a standard diet (StD). In females this effect was weaker and was limited to the PtcD. A significant proportion of rats with brain tumors and fed the StD exhibited SE evidenced by weight loss, a shorter survival, reduction in liver weight and an increased proportion of liver mitochondria, effects that were not observed in their counterpart fed PtcD. Comparison of the expression of genes involved in the balance proliferation/apoptosis and in the response to oxidative stress in male brain tumors showed that the prevention of SE was associated with an increase in bcl-2 and catalase and a decrease in ki-67, sod-1 and sod-2 transcripts. These results show that the degree of agressiveness of gliomas can be modulated by dietary interventions and suggest that some phytochemicals with antioxidant properties could participate to the mechanism.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis , Neoplasias Encefálicas/dietoterapia , Neoplasias Encefálicas/prevención & control , Glioma/dietoterapia , Glioma/prevención & control , Estrés Oxidativo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/análisis , Neoplasias Encefálicas/inducido químicamente , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Etilnitrosourea , Femenino , Frutas , Glioma/inducido químicamente , Glioma/metabolismo , Glioma/patología , Inmunohistoquímica , Masculino , Mitocondrias Hepáticas/metabolismo , Reacción en Cadena de la Polimerasa , Embarazo , ARN Neoplásico/análisis , Ratas , Verduras
5.
BMC Cancer ; 6: 234, 2006 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17018136

RESUMEN

BACKGROUND: Accumulating data suggest that liver is a major target organ of systemic effects observed in the presence of a cancer. In this study, we investigated the consequences of the presence of chemically induced brain tumors in rats on biophysical parameters accounting for the dynamics of water in liver mitochondria. METHODS: Tumors of the central nervous system were induced by intraveinous administration of ethylnitrosourea (ENU) to pregnant females on the 19th day of gestation. The mitochondrial crude fraction was isolated from the liver of each animal and the dynamic parameters of total water and its macromolecule-associated fraction (structured water, H2Ost) were calculated from Nuclear Magnetic Resonance (NMR) measurements. RESULTS: The presence of a malignant brain tumor induced a loss of water structural order that implicated changes in the physical properties of the hydration shells of liver mitochondria macromolecules. This feature was linked to an increase in the membrane cholesterol content, a way to limit water penetration into the bilayer and then to reduce membrane permeability. As expected, these alterations in mitochondrial plasticity affected ionic exchanges and led to abnormal features of mitochondrial biogenesis and caspase activation. CONCLUSION: This study enlightens the sensitivity of the structured water phase in the liver mitochondria machinery to external conditions such as tumor development at a distant site. The profound metabolic and functional changes led to abnormal features of ion transport, mitochondrial biogenesis and caspase activation.


Asunto(s)
Neoplasias Encefálicas/química , Mitocondrias Hepáticas/química , Agua/química , Animales , Neoplasias Encefálicas/enzimología , Caspasas/metabolismo , Colesterol/análisis , Ácidos Grasos Insaturados/análisis , Femenino , Iones/análisis , Hígado/enzimología , Resonancia Magnética Nuclear Biomolecular , Ratas
6.
J Exp Med ; 212(12): 2015-25, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26503444

RESUMEN

Natural killer (NK) cell maturation is a tightly controlled process that endows NK cells with functional competence and the capacity to recognize target cells. Here, we found that the transcription factor (TF) Zeb2 was the most highly induced TF during NK cell maturation. Zeb2 is known to control epithelial to mesenchymal transition, but its role in immune cells is mostly undefined. Targeted deletion of Zeb2 resulted in impaired NK cell maturation, survival, and exit from the bone marrow. NK cell function was preserved, but mice lacking Zeb2 in NK cells were more susceptible to B16 melanoma lung metastases. Reciprocally, ectopic expression of Zeb2 resulted in a higher frequency of mature NK cells in all organs. Moreover, the immature phenotype of Zeb2(-/-) NK cells closely resembled that of Tbx21(-/-) NK cells. This was caused by both a dependence of Zeb2 expression on T-bet and a probable cooperation of these factors in gene regulation. Transgenic expression of Zeb2 in Tbx21(-/-) NK cells partially restored a normal maturation, establishing that timely induction of Zeb2 by T-bet is an essential event during NK cell differentiation. Finally, this novel transcriptional cascade could also operate in human as T-bet and Zeb2 are similarly regulated in mouse and human NK cells.


Asunto(s)
Proteínas de Homeodominio/inmunología , Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , Proteínas Represoras/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Asesinas Naturales/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc
7.
J Exp Med ; 211(3): 563-77, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24516120

RESUMEN

Trail(+)DX5(-)Eomes(-) natural killer (NK) cells arise in the mouse fetal liver and persist in the adult liver. Their relationships with Trail(-)DX5(+) NK cells remain controversial. We generated a novel Eomes-GFP reporter murine model to address this question. We found that Eomes(-) NK cells are not precursors of classical Eomes(+) NK cells but rather constitute a distinct lineage of innate lymphoid cells. Eomes(-) NK cells are strictly dependent on both T-bet and IL-15, similarly to NKT cells. We observed that, in the liver, expression of T-bet in progenitors represses Eomes expression and the development of Eomes(+) NK cells. Reciprocally, the bone marrow (BM) microenvironment restricts T-bet expression in developing NK cells. Ectopic expression of T-bet forces the development of Eomes(-) NK cells, demonstrating that repression of T-bet is essential for the development of Eomes(+) NK cells. Gene profile analyses show that Eomes(-) NK cells share part of their transcriptional program with NKT cells, including genes involved in liver homing and NK cell receptors. Moreover, Eomes(-) NK cells produce a broad range of cytokines, including IL-2 and TNF in vitro and in vivo, during immune responses against vaccinia virus. Thus, mutually exclusive expression of T-bet and Eomes drives the development of different NK cell lineages with complementary functions.


Asunto(s)
Médula Ósea/metabolismo , Linaje de la Célula/inmunología , Células Asesinas Naturales/inmunología , Hígado/metabolismo , Nicho de Células Madre/inmunología , Proteínas de Dominio T Box/metabolismo , Traslado Adoptivo , Animales , Diferenciación Celular/inmunología , Cartilla de ADN/genética , Citometría de Flujo , Técnicas de Sustitución del Gen , Células Asesinas Naturales/citología , Ratones , Análisis por Micromatrices , Modelos Animales , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Dominio T Box/genética
8.
Eur J Cancer ; 48(15): 2431-41, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22325970

RESUMEN

A hallmark of cancer is the paradoxical co-presence, in the same tumour, of local and global DNA hypomethylation together with the regional hypermethylation of certain genes. Due to the oncogenic role of these different DNA methylation alterations, two therapeutic strategies are possible: the use of DNA methylating agents (DMA, such as folate) to inhibit global or local DNA hypomethylation or the use of DNA hypomethylating agents (DHA, such as 5-aza-2-deoxycytidine) to abrogate the accumulation of hypermethylated genes. Here we explored the use of folate to treat gliomas in a mouse model, using tumours induced by either PDGF-B or Ras/Akt overexpression, or by ethylnitrosourea (ENU) treatment. Under all conditions the volume of tumours were significantly less in folate treated mice than in untreated mice. Quantitative methylated DNA immunoprecipitation (qMeDIP) and quantitative methylated specific PCR (qMSP) analysis of methylation status showed that folate treatment, increased the methylation level of DNA repeat elements in tumour and in colorectal tissue and that of MGMT and specific oncogenes (PDGF-B or survivin) in tumours (but not in colorectal tissue), but had no effect on the expression of tumour suppressor genes (p53, PTENorbax) in tumours or in colorectal tissue. This suggests that folate has anti-neoplastic effects in gliomas and that no preneoplastic or neoplastic alterations were observed in unaffected colorectal tissue in response to the potential tumourigenic effects of folate. Collectively, our data support the proposal to include folate as a promising adjuvant in the design of anti-glioma therapeutic protocols in clinical studies.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Ácido Fólico/farmacología , Glioma/tratamiento farmacológico , Animales , Transformación Celular Neoplásica/genética , Metilación de ADN/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Glioma/genética , Humanos , Ratones , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Clin Epigenetics ; 2(2): 369-381, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21949549

RESUMEN

Elevated activation of the platelet-derived growth factor (PDGF) pathway, apoptosis evasion phenotype, and global DNA hypomethylation are hallmarks frequently observed in cancers, such as in low-grade glioma (LGG). However, the orchestration of these malignant functions is not fully elucidated in LGG. Our study reveals that the co-presence of these hallmarks in the same LGG is frequent and confers poor prognosis in patients with LGG. Our data also indicate that the apoptosis evasion phenotype of these cells harboring a hypomethylation-induced activation of the PDGF pathway is associated with a hypomethylation of the bcl-xl and bcl-w genes and the phosphorylation and/or downregulation of three major pro-apoptotic BH3-only proteins: PUMA, Bad, and Bim. Consistent with this, we demonstrate that the use of folate, a DNA-methylating agent, promotes the reprogramming of the sensitivity of glioma cells to ABT-737/etoposide-induced apoptosis and reduces the dose of ABT-737 required to promote etoposide-induced apoptosis. This work supports the idea that the inclusion of folate and/or ABT-737 could be a promising adjuvant in the design of anti-glioma therapeutic protocols in clinical studies. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s13148-011-0035-5) contains supplementary material, which is available to authorized users.

10.
PLoS One ; 5(6): e11333, 2010 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-20613874

RESUMEN

Global DNA hypomethylation is a hallmark of cancer cells, but its molecular mechanisms have not been elucidated. Here, we show that the disruption of Dnmt1/PCNA/UHRF1 interactions promotes a global DNA hypomethylation in human gliomas. We then demonstrate that the Dnmt1 phosphorylations by Akt and/or PKC abrogate the interactions of Dnmt1 with PCNA and UHRF1 in cellular and acellular studies including mass spectrometric analyses and the use of primary cultured patient-derived glioma. By using methylated DNA immunoprecipitation, methylation and CGH arrays, we show that global DNA hypomethylation is associated with genes hypomethylation, hypomethylation of DNA repeat element and chromosomal instability. Our results reveal that the disruption of Dnmt1/PCNA/UHRF1 interactions acts as an oncogenic event and that one of its signatures (i.e. the low level of mMTase activity) is a molecular biomarker associated with a poor prognosis in GBM patients. We identify the genetic and epigenetic alterations which collectively promote the acquisition of tumor/glioma traits by human astrocytes and glial progenitor cells as that promoting high proliferation and apoptosis evasion.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Transformación Celular Neoplásica/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Neuroglía/patología , Proteínas Nucleares/genética , Antígeno Nuclear de Célula en Proliferación/genética , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , Metilación de ADN , Humanos , Espectrometría de Masas , Ratones , Fosforilación , Pronóstico , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA