Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Exp Med ; 212(12): 2015-25, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26503444

ABSTRACT

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.


Subject(s)
Homeodomain Proteins/immunology , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Repressor Proteins/immunology , T-Box Domain Proteins/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Flow Cytometry , Gene Expression/immunology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Killer Cells, Natural/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Zinc Finger E-box Binding Homeobox 2
2.
J Exp Med ; 211(3): 563-77, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24516120

ABSTRACT

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.


Subject(s)
Bone Marrow/metabolism , Cell Lineage/immunology , Killer Cells, Natural/immunology , Liver/metabolism , Stem Cell Niche/immunology , T-Box Domain Proteins/metabolism , Adoptive Transfer , Animals , Cell Differentiation/immunology , DNA Primers/genetics , Flow Cytometry , Gene Knock-In Techniques , Killer Cells, Natural/cytology , Mice , Microarray Analysis , Models, Animal , Real-Time Polymerase Chain Reaction , T-Box Domain Proteins/genetics
3.
J Immunol ; 191(9): 4619-27, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068664

ABSTRACT

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.


Subject(s)
Ikaros Transcription Factor/metabolism , Interleukin-1beta/metabolism , Receptors, Interleukin-1 Type I/metabolism , T-Lymphocytes, Regulatory/metabolism , Down-Regulation , Forkhead Transcription Factors/metabolism , Humans , Immunologic Memory , Interleukin-10/metabolism , Interleukin-17/metabolism , Receptors, CCR7/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
4.
Eur J Immunol ; 43(6): 1667-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23519784

ABSTRACT

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.


Subject(s)
Antigens, Ly/metabolism , Bone Marrow/immunology , Monocytes/immunology , Receptors, Lysosphingolipid/metabolism , Animals , Blood Circulation , Cell Movement/immunology , Cell Survival , Cells, Cultured , Female , Homeostasis , Immunologic Surveillance , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lysosphingolipid/genetics
5.
Eur J Cancer ; 48(15): 2431-41, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22325970

ABSTRACT

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.


Subject(s)
Cell Transformation, Neoplastic/drug effects , Folic Acid/pharmacology , Glioma/drug therapy , Animals , Cell Transformation, Neoplastic/genetics , DNA Methylation/drug effects , Dietary Supplements , Disease Models, Animal , Glioma/genetics , Humans , Mice , Survival Analysis , Xenograft Model Antitumor Assays
6.
Clin Epigenetics ; 2(2): 369-381, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21949549

ABSTRACT

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.

7.
PLoS One ; 5(6): e11333, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20613874

ABSTRACT

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.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Cell Transformation, Neoplastic/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Neuroglia/pathology , Nuclear Proteins/genetics , Proliferating Cell Nuclear Antigen/genetics , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , Humans , Mass Spectrometry , Mice , Phosphorylation , Prognosis , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases
8.
Clin Cancer Res ; 15(10): 3519-29, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19451595

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cell Proliferation , DNA Methylation , Folic Acid/administration & dosage , Glioma/genetics , Aged , Apoptosis/drug effects , Apoptosis/genetics , Becaplermin , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dietary Supplements , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/pathology , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Invasiveness , Platelet-Derived Growth Factor/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-sis , Repetitive Sequences, Nucleic Acid/genetics , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Survival Analysis , Survivin , Temozolomide , Tumor Suppressor Proteins/genetics , DNA Methyltransferase 3B
9.
Int J Cancer ; 123(2): 288-295, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18412241

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis , Brain Neoplasms/diet therapy , Brain Neoplasms/prevention & control , Glioma/diet therapy , Glioma/prevention & control , Oxidative Stress , Animals , Apoptosis/drug effects , Biomarkers, Tumor/analysis , Brain Neoplasms/chemically induced , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Ethylnitrosourea , Female , Fruit , Glioma/chemically induced , Glioma/metabolism , Glioma/pathology , Immunohistochemistry , Male , Mitochondria, Liver/metabolism , Polymerase Chain Reaction , Pregnancy , RNA, Neoplasm/analysis , Rats , Vegetables
10.
BMC Cancer ; 6: 234, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17018136

ABSTRACT

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.


Subject(s)
Brain Neoplasms/chemistry , Mitochondria, Liver/chemistry , Water/chemistry , Animals , Brain Neoplasms/enzymology , Caspases/metabolism , Cholesterol/analysis , Fatty Acids, Unsaturated/analysis , Female , Ions/analysis , Liver/enzymology , Nuclear Magnetic Resonance, Biomolecular , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...