Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Metab ; 54: 101398, 2021 12.
Article in English | MEDLINE | ID: mdl-34801767

ABSTRACT

OBJECTIVE: To analyze the genome-wide epigenomic and transcriptomic changes induced by long term resistance or endurance training in the hippocampus of wild-type mice. METHODS: We performed whole-genome bisulfite sequencing (WGBS) and RNA sequencing (RNA-seq) of mice hippocampus after 4 weeks of specific training. In addition, we used a novel object recognition test before and after the intervention to determine whether the exercise led to an improvement in cognitive function. RESULTS: Although the majority of DNA methylation changes identified in this study were training-model specific, most were associated with hypomethylation and were enriched in similar histone marks, chromatin states, and transcription factor biding sites. It is worth highlighting the significant association found between the loss of DNA methylation in Tet1 binding sites and gene expression changes, indicating the importance of these epigenomic changes in transcriptional regulation. However, endurance and resistance training activate different gene pathways, those being associated with neuroplasticity in the case of endurance exercise, and interferon response pathways in the case of resistance exercise, which also appears to be associated with improved learning and memory functions. CONCLUSIONS: Our results help both understand the molecular mechanisms by which different exercise models exert beneficial effects for brain health and provide new potential therapeutic targets for future research.


Subject(s)
Brain/metabolism , Epigenome/genetics , Exercise Test , Physical Conditioning, Animal , Animals , Male , Mice , Mice, Inbred C57BL
3.
Nucleic Acids Res ; 47(10): 5016-5037, 2019 06 04.
Article in English | MEDLINE | ID: mdl-30923829

ABSTRACT

Histone H4 acetylation at Lysine 16 (H4K16ac) is a key epigenetic mark involved in gene regulation, DNA repair and chromatin remodeling, and though it is known to be essential for embryonic development, its role during adult life is still poorly understood. Here we show that this lysine is massively hyperacetylated in peripheral neutrophils. Genome-wide mapping of H4K16ac in terminally differentiated blood cells, along with functional experiments, supported a role for this histone post-translational modification in the regulation of cell differentiation and apoptosis in the hematopoietic system. Furthermore, in neutrophils, H4K16ac was enriched at specific DNA repeats. These DNA regions presented an accessible chromatin conformation and were associated with the cleavage sites that generate the 50 kb DNA fragments during the first stages of programmed cell death. Our results thus suggest that H4K16ac plays a dual role in myeloid cells as it not only regulates differentiation and apoptosis, but it also exhibits a non-canonical structural role in poising chromatin for cleavage at an early stage of neutrophil cell death.


Subject(s)
Apoptosis , Cell Differentiation , Chromatin/metabolism , Histones/metabolism , Lysine/metabolism , Myeloid Cells/metabolism , Acetylation , Animals , Cells, Cultured , Chromatin/genetics , Epigenesis, Genetic , Humans , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Protein Processing, Post-Translational , Transcription, Genetic
4.
Hum Mol Genet ; 27(17): 3046-3059, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29878202

ABSTRACT

Aberrant DNA hypermethylation is a hallmark of cancer although the underlying molecular mechanisms are still poorly understood. To study the possible role of 5-hydroxymethylcytosine (5hmC) in this process we analyzed the global and locus-specific genome-wide levels of 5hmC and 5-methylcytosine (5mC) in human primary samples from 12 non-tumoral brains and 53 gliomas. We found that the levels of 5hmC identified in non-tumoral samples were significantly reduced in gliomas. Strikingly, hypo-hydroxymethylation at 4627 (9.3%) CpG sites was associated with aberrant DNA hypermethylation and was strongly enriched in CpG island shores. The DNA regions containing these CpG sites were enriched in H3K4me2 and presented a different genuine chromatin signature to that characteristic of the genes classically aberrantly hypermethylated in cancer. As this 5mC gain is inversely correlated with loss of 5hmC and has not been identified with classical sodium bisulfite-based technologies, we conclude that our data identifies a novel 5hmC-dependent type of aberrant DNA hypermethylation in glioma.


Subject(s)
5-Methylcytosine/analogs & derivatives , Biomarkers, Tumor/genetics , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Genome, Human , Glioma/pathology , 5-Methylcytosine/metabolism , Case-Control Studies , CpG Islands , Glioma/genetics , Glioma/metabolism , Humans
5.
Genome Res ; 25(1): 27-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25271306

ABSTRACT

In differentiated cells, aging is associated with hypermethylation of DNA regions enriched in repressive histone post-translational modifications. However, the chromatin marks associated with changes in DNA methylation in adult stem cells during lifetime are still largely unknown. Here, DNA methylation profiling of mesenchymal stem cells (MSCs) obtained from individuals aged 2 to 92 yr identified 18,735 hypermethylated and 45,407 hypomethylated CpG sites associated with aging. As in differentiated cells, hypermethylated sequences were enriched in chromatin repressive marks. Most importantly, hypomethylated CpG sites were strongly enriched in the active chromatin mark H3K4me1 in stem and differentiated cells, suggesting this is a cell type-independent chromatin signature of DNA hypomethylation during aging. Analysis of scedasticity showed that interindividual variability of DNA methylation increased during aging in MSCs and differentiated cells, providing a new avenue for the identification of DNA methylation changes over time. DNA methylation profiling of genetically identical individuals showed that both the tendency of DNA methylation changes and scedasticity depended on nongenetic as well as genetic factors. Our results indicate that the dynamics of DNA methylation during aging depend on a complex mixture of factors that include the DNA sequence, cell type, and chromatin context involved and that, depending on the locus, the changes can be modulated by genetic and/or external factors.


Subject(s)
Aging/genetics , DNA Methylation , DNA/genetics , Stem Cells/cytology , Adolescent , Aged , Aged, 80 and over , Cell Differentiation , Cells, Cultured , Child , Child, Preschool , Chromatin/genetics , Epigenesis, Genetic , Histones/genetics , Humans , Microarray Analysis , Middle Aged , Promoter Regions, Genetic , Protein Processing, Post-Translational , Sequence Analysis, DNA , Twins, Monozygotic , Young Adult
6.
J Clin Endocrinol Metab ; 98(7): 2811-21, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23666970

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the global patterns of aberrant DNA methylation in thyroid cancer. RESEARCH DESIGN AND METHODS: We have used DNA methylation arrays to determine, for the first time, the genome-wide promoter methylation status of papillary, follicular, medullary, and anaplastic thyroid tumors. RESULTS: We identified 262 and 352 hypermethylated and 13 and 21 hypomethylated genes in differentiated papillary and follicular tumors, respectively. Interestingly, the other tumor types analyzed displayed more hypomethylated genes (280 in anaplastic and 393 in medullary tumors) than aberrantly hypermethylated genes (86 in anaplastic and 131 in medullary tumors). Among the genes indentified, we show that 4 potential tumor suppressor genes (ADAMTS8, HOXB4, ZIC1, and KISS1R) and 4 potential oncogenes (INSL4, DPPA2, TCL1B, and NOTCH4) are frequently regulated by aberrant methylation in primary thyroid tumors. In addition, we show that aberrant promoter hypomethylation-associated overexpression of MAP17 might promote tumor growth in thyroid cancer. CONCLUSIONS: Thyroid cancer subtypes present differential promoter methylation signatures, and nondifferentiated subtypes are characterized by aberrant promoter hypomethylation rather than hypermethylation. Additional studies are needed to determine the potential clinical interest of the tumor subtype-specific DNA methylation signatures described herein and the role of aberrant promoter hypomethylation in nondifferentiated thyroid tumors.


Subject(s)
DNA Methylation , Down-Regulation , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Up-Regulation , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/metabolism , Adenocarcinoma, Follicular/pathology , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Carcinoma, Neuroendocrine , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Line, Tumor , Cohort Studies , Genome-Wide Association Study , Humans , Neoplasm Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Thyroid Cancer, Papillary , Thyroid Carcinoma, Anaplastic , Thyroid Gland/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Tissue Banks , Tumor Cells, Cultured
7.
J Mol Med (Berl) ; 91(8): 939-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23475283

ABSTRACT

The basic mechanisms underlying promoter DNA hypermethylation in cancer are still largely unknown. It has been proposed that the levels of the methyl donor group in DNA methylation reactions, S-adenosylmethionine (SAMe), might be involved. SAMe levels depend on the glycine-N-methyltransferase (GNMT), a one-carbon group methyltransferase, which catalyzes the conversion of SAMe to S-adenosylhomocysteine in hepatic cells. GNMT has been proposed to display tumor suppressor activity and to be frequently repressed in hepatocellular carcinoma (HCC). In this study, we show that GNMT shows aberrant DNA hypermethylation in some HCC cell lines and primary tumors (20 %). GNMT hypermethylation could contribute to gene repression and its restoration in cell lines displaying hypermethylation-reduced tumor growth in vitro. In agreement, human primary tumors expressing GNMT were of smaller size than tumors showing GNMT hypermethylation. Genome-wide analyses of gene promoter methylation identified 277 genes whose aberrant methylation in HCC was associated with GNMT methylation/expression. The findings in this manuscript indicate that DNA hypermethylation plays an important role in the repression of GNMT in HCC and that loss of GNMT in human HCC could promote the establishment of aberrant DNA methylation patterns at specific gene promoters.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Methylation , Glycine N-Methyltransferase/genetics , Liver Neoplasms/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival , Epigenetic Repression , Glycine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Liver Neoplasms/metabolism , RNA, Messenger/metabolism , S-Adenosylmethionine/metabolism
8.
Cancer Res ; 73(1): 395-405, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23108143

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF/CSF2) is a cytokine produced in the hematologic compartment that may enhance antitumor immune responses, mainly by activation of dendritic cells. Here, we show that more than one-third of human colorectal tumors exhibit aberrant DNA demethylation of the GM-CSF promoter and overexpress the cytokine. Mouse engraftment experiments with autologous and homologous colon tumors engineered to repress the ectopic secretion of GM-CSF revealed the tumor-secreted GM-CSF to have an immune-associated antitumor effect. Unexpectedly, an immune-independent antitumor effect was observed that depended on the ectopic expression of GM-CSF receptor subunits by tumors. Cancer cells expressing GM-CSF and its receptor did not develop into tumors when autografted into immunocompetent mice. Similarly, 100% of the patients with human colon tumors that overexpressed GM-CSF and its receptor subunits survived at least 5 years after diagnosis. These data suggest that expression of GM-CSF and its receptor subunits by colon tumors may be a useful marker for prognosis as well as for patient stratification in cancer immunotherapy.


Subject(s)
Colorectal Neoplasms/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Animals , Biomarkers, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , DNA Methylation , Enzyme-Linked Immunosorbent Assay , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Prognosis , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Tissue Array Analysis , Transfection
9.
Hepatology ; 56(6): 2363-74, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22653837

ABSTRACT

UNLABELLED: Mutations in polycystins are a cause of polycystic liver disease. In polycystin-2 (PC2)-defective mice, cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA)-dependent activation of the Rat Sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen signal-regulated kinase-extracellular signal-regulated kinase (ERK) 1/2 pathway stimulates the growth of liver cysts. To test the hypothesis that sorafenib, a Raf inhibitor used for the treatment of liver and kidney cancers, inhibits liver cyst growth in PC2-defective mice, we treated PC2 (i.e., Pkd2(flox/-) :pCxCreER(TM) [Pkd2cKO]) mice with sorafenib-tosylate for 8 weeks (20-60 mg/kg/day). Sorafenib caused an unexpected increase in liver cyst area, cell proliferation (Ki67), and expression of phosphorylated ERK (pERK) compared with Pkd2cKO mice treated with vehicle. When given to epithelial cells isolated from liver cysts of Pkd2cKO mice (Pkd2cKO-cells), sorafenib progressively stimulated pERK1/2 and cell proliferation [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium and bromodeoxyuridine assay (MTS)] at doses between 0.001 and 1 µM; however, both pERK1/2 and cell proliferation significantly decreased at the dose of 10 µM. Raf kinase activity assay showed that whereas B-Raf is inhibited by sorafenib in both wild-type (WT) and Pkd2cKO cells, Raf-1 is inhibited in WT cells but is significantly stimulated in Pkd2cKO cells. In Pkd2cKO cells pretreated with the PKA inhibitor 14-22 amide, myristolated (1 µM) and in mice treated with octreotide in combination with sorafenib, the paradoxical activation of Raf/ERK1/2 was abolished, and cyst growth was inhibited. CONCLUSION: In PC2-defective cells, sorafenib inhibits B-Raf but paradoxically activates Raf-1, resulting in increased ERK1/2 phosphorylation, cell proliferation, and cyst growth in vivo. These effects are consistent with the ability of Raf inhibitors to transactivate Raf-1 when a PKA-activated Ras promotes Raf-1/B-Raf heterodimerization, and are inhibited by interfering with cAMP/PKA signaling both in vitro and in vivo, as shown by the reduction of liver cysts in mice treated with combined octreotide and sorafenib.


Subject(s)
Benzenesulfonates/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , Cysts/enzymology , Liver Diseases/enzymology , MAP Kinase Signaling System/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , TRPP Cation Channels/deficiency , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Benzenesulfonates/therapeutic use , Bile Ducts/cytology , Bile Ducts/metabolism , Caspase 3/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Cysts/drug therapy , Cysts/pathology , Drug Therapy, Combination , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Ki-67 Antigen/metabolism , Liver Diseases/drug therapy , Liver Diseases/pathology , Mice , Mice, Knockout , Niacinamide/analogs & derivatives , Octreotide/pharmacology , Octreotide/therapeutic use , Phenylurea Compounds , Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Pyridines/therapeutic use , Sorafenib , TRPP Cation Channels/genetics
10.
Eur J Cancer ; 48(14): 2270-81, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22244828

ABSTRACT

Altered promoter DNA methylation, one of the most important molecular alterations in cancer, is proposed to correlate with deregulation of DNA methyltransferases, although the molecular mechanisms implicated are still poorly understood. Here we show that the de novo DNA methyltransferase DNMT3B is frequently repressed in human colorectal cancer cell lines (CCL) and primary tumours by aberrant DNA hypermethylation of its distal promoter. At the epigenome level, DNMT3B promoter hypermethylation was associated with the hypomethylation of gene promoters usually hypermethylated in the healthy colon. Forced DNMT3B overexpression in cancer cells restored the methylation levels of these promoters in the healthy colon. Our results show a new molecular mechanism of aberrant DNMT3B regulation in colon cancer and suggest that its expression is associated with the methylation of constitutively hypermethylated promoters in the healthy colon.


Subject(s)
Colorectal Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Promoter Regions, Genetic , Adult , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Colorectal Neoplasms/enzymology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA Methylation/drug effects , Decitabine , Down-Regulation , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HCT116 Cells , HT29 Cells , Humans , Male , Middle Aged , Promoter Regions, Genetic/drug effects , Transfection , DNA Methyltransferase 3B
11.
Nucleic Acids Res ; 40(1): 116-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21911366

ABSTRACT

Global mechanisms defining the gene expression programs specific for hematopoiesis are still not fully understood. Here, we show that promoter DNA demethylation is associated with the activation of hematopoietic-specific genes. Using genome-wide promoter methylation arrays, we identified 694 hematopoietic-specific genes repressed by promoter DNA methylation in human embryonic stem cells and whose loss of methylation in hematopoietic can be associated with gene expression. The association between promoter methylation and gene expression was studied for many hematopoietic-specific genes including CD45, CD34, CD28, CD19, the T cell receptor (TCR), the MHC class II gene HLA-DR, perforin 1 and the phosphoinositide 3-kinase (PI3K) and results indicated that DNA demethylation was not always sufficient for gene activation. Promoter demethylation occurred either early during embryonic development or later on during hematopoietic differentiation. Analysis of the genome-wide promoter methylation status of induced pluripotent stem cells (iPSCs) generated from somatic CD34(+) HSPCs and differentiated derivatives from CD34(+) HSPCs confirmed the role of DNA methylation in regulating the expression of genes of the hemato-immune system, and indicated that promoter methylation of these genes may be associated to stemness. Together, these data suggest that promoter DNA demethylation might play a role in the tissue/cell-specific genome-wide gene regulation within the hematopoietic compartment.


Subject(s)
DNA Methylation , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Dedifferentiation , Cell Line , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Infant, Newborn , Mice , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...