Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 61(4): 520-534, 2016 Feb 18.
Article in English | MEDLINE | ID: mdl-26853146

ABSTRACT

Altered energy metabolism is a cancer hallmark as malignant cells tailor their metabolic pathways to meet their energy requirements. Glucose and glutamine are the major nutrients that fuel cellular metabolism, and the pathways utilizing these nutrients are often altered in cancer. Here, we show that the long ncRNA CCAT2, located at the 8q24 amplicon on cancer risk-associated rs6983267 SNP, regulates cancer metabolism in vitro and in vivo in an allele-specific manner by binding the Cleavage Factor I (CFIm) complex with distinct affinities for the two subunits (CFIm25 and CFIm68). The CCAT2 interaction with the CFIm complex fine-tunes the alternative splicing of Glutaminase (GLS) by selecting the poly(A) site in intron 14 of the precursor mRNA. These findings uncover a complex, allele-specific regulatory mechanism of cancer metabolism orchestrated by the two alleles of a long ncRNA.


Subject(s)
Glutaminase/genetics , Neoplasms/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Alleles , Alternative Splicing , Energy Metabolism , HCT116 Cells , Humans , Neoplasms/genetics , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA, Messenger/metabolism
2.
Br J Haematol ; 201(4): 718-724, 2023 05.
Article in English | MEDLINE | ID: mdl-36786170

ABSTRACT

Despite the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway being frequently altered in T-ALL/LBL, no specific therapy has been approved for T-ALL/LBL patients with constitutive signalling by JAK/STAT, so there is an urgent need to identify pathway members that may be potential therapeutic targets. In the present study, we searched for JAK/STAT pathway members potentially modulated through aberrant methylation and identified SOCS3 hypermethylation as a recurrent event in T-ALL/LBL. Additionally, we explored the implications of SOCS3 deregulation in T-ALL/LBL and demonstrated that SOCS3 counteracts the constitutive activation of the JAK/STAT pathway through different molecular mechanisms. Therefore, SOCS3 emerges as a potential therapeutic target in T-ALL/LBL.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Janus Kinases/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , STAT Transcription Factors/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocytes/metabolism
3.
Cardiovasc Diabetol ; 22(1): 44, 2023 03 04.
Article in English | MEDLINE | ID: mdl-36870961

ABSTRACT

BACKGROUND: Obesity is a negative chronic metabolic health condition that represents an additional risk for the development of multiple pathologies. Epidemiological studies have shown how maternal obesity or gestational diabetes mellitus during pregnancy constitute serious risk factors in relation to the appearance of cardiometabolic diseases in the offspring. Furthermore, epigenetic remodelling may help explain the molecular mechanisms that underlie these epidemiological findings. Thus, in this study we explored the DNA methylation landscape of children born to mothers with obesity and gestational diabetes during their first year of life. METHODS: We used Illumina Infinium MethylationEPIC BeadChip arrays to profile more than 770,000 genome-wide CpG sites in blood samples from a paediatric longitudinal cohort consisting of 26 children born to mothers who suffered from obesity or obesity with gestational diabetes mellitus during pregnancy and 13 healthy controls (measurements taken at 0, 6 and 12 month; total N = 90). We carried out cross-sectional and longitudinal analyses to derive DNA methylation alterations associated with developmental and pathology-related epigenomics. RESULTS: We identified abundant DNA methylation changes during child development from birth to 6 months and, to a lesser extent, up to 12 months of age. Using cross-sectional analyses, we discovered DNA methylation biomarkers maintained across the first year of life that could discriminate children born to mothers who suffered from obesity or obesity with gestational diabetes. Importantly, enrichment analyses suggested that these alterations constitute epigenetic signatures that affect genes and pathways involved in the metabolism of fatty acids, postnatal developmental processes and mitochondrial bioenergetics, such as CPT1B, SLC38A4, SLC35F3 and FN3K. Finally, we observed evidence of an interaction between developmental DNA methylation changes and maternal metabolic condition alterations. CONCLUSIONS: Our observations highlight the first six months of development as being the most crucial for epigenetic remodelling. Furthermore, our results support the existence of systemic intrauterine foetal programming linked to obesity and gestational diabetes that affects the childhood methylome beyond birth, which involves alterations related to metabolic pathways, and which may interact with ordinary postnatal development programmes.


Subject(s)
Diabetes, Gestational , Obesity, Maternal , Pregnancy , Humans , Female , Child , Epigenome , Cross-Sectional Studies , Epigenomics , Obesity , Epigenesis, Genetic
5.
Nature ; 523(7559): 177-82, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26106858

ABSTRACT

Exosomes are lipid-bilayer-enclosed extracellular vesicles that contain proteins and nucleic acids. They are secreted by all cells and circulate in the blood. Specific detection and isolation of cancer-cell-derived exosomes in the circulation is currently lacking. Using mass spectrometry analyses, we identify a cell surface proteoglycan, glypican-1 (GPC1), specifically enriched on cancer-cell-derived exosomes. GPC1(+) circulating exosomes (crExos) were monitored and isolated using flow cytometry from the serum of patients and mice with cancer. GPC1(+) crExos were detected in the serum of patients with pancreatic cancer with absolute specificity and sensitivity, distinguishing healthy subjects and patients with a benign pancreatic disease from patients with early- and late-stage pancreatic cancer. Levels of GPC1(+) crExos correlate with tumour burden and the survival of pre- and post-surgical patients. GPC1(+) crExos from patients and from mice with spontaneous pancreatic tumours carry specific KRAS mutations, and reliably detect pancreatic intraepithelial lesions in mice despite negative signals by magnetic resonance imaging. GPC1(+) crExos may serve as a potential non-invasive diagnostic and screening tool to detect early stages of pancreatic cancer to facilitate possible curative surgical therapy.


Subject(s)
Exosomes/metabolism , Glypicans , Pancreatic Neoplasms/diagnosis , Animals , Biomarkers/blood , Cell Line, Tumor , Exosomes/genetics , Female , Glypicans/blood , Glypicans/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Male , Mice , NIH 3T3 Cells , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/metabolism
6.
Int J Cancer ; 146(2): 373-387, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31211412

ABSTRACT

Loss of 5-hydroxymethylcytosine (5hmC) has been associated with mutations of the ten-eleven translocation (TET) enzymes in several types of cancer. However, tumors with wild-type TET genes can also display low 5hmC levels, suggesting that other mechanisms involved in gene regulation might be implicated in the decline of this epigenetic mark. Here we show that DNA hypermethylation and loss of DNA hydroxymethylation, as well as a marked reduction of activating histone marks in the TET3 gene, impair TET3 expression and lead to a genome-wide reduction in 5hmC levels in glioma samples and cancer cell lines. Epigenetic drugs increased expression of TET3 in glioblastoma cells and ectopic overexpression of TET3 impaired in vitro cell growth and markedly reduced tumor formation in immunodeficient mice models. TET3 overexpression partially restored the genome-wide patterns of 5hmC characteristic of control brain samples in glioblastoma cell lines, while elevated TET3 mRNA levels were correlated with better prognosis in glioma samples. Our results suggest that epigenetic repression of TET3 might promote glioblastoma tumorigenesis through the genome-wide alteration of 5hmC.


Subject(s)
Brain Neoplasms/genetics , Carcinogenesis/genetics , Dioxygenases/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Animals , Biopsy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Methylation , Down-Regulation , Glioblastoma/mortality , Glioblastoma/pathology , Histone Code/genetics , Humans , Mice , Prognosis , RNA, Messenger/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
7.
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
8.
J Transl Med ; 17(1): 15, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626398

ABSTRACT

BACKGROUND: Early life is a period of drastic epigenetic remodeling in which the epigenome is especially sensitive to extrinsic and intrinsic influence. However, the epigenome-wide dynamics of the DNA methylation changes that occur during this period have not been sufficiently characterized in longitudinal studies. METHODS: To this end, we studied the DNA methylation status of more than 750,000 CpG sites using Illumina MethylationEPIC arrays on 33 paired blood samples from 11 subjects at birth and at 5 and 10 years of age, then characterized the chromatin context associated with these loci by integrating our data with histone, chromatin-state and enhancer-element external datasets, and, finally, validated our results through bisulfite pyrosequencing in two independent longitudinal cohorts of 18 additional subjects. RESULTS: We found abundant DNA methylation changes (110,726 CpG sites) during the first lustrum of life, while far fewer alterations were observed in the subsequent 5 years (460 CpG sites). However, our analysis revealed persistent DNA methylation changes at 240 CpG sites, indicating that there are genomic locations of considerable epigenetic change beyond immediate birth. The chromatin context of hypermethylation changes was associated with repressive genomic locations and genes with developmental and cell signaling functions, while hypomethylation changes were linked to enhancer regions and genes with immunological and mRNA and protein metabolism functions. Significantly, our results show that genes that suffer simultaneous hyper- and hypomethylation are functionally distinct from exclusively hyper- or hypomethylated genes, and that enhancer-associated methylation is different in hyper- and hypomethylation scenarios, with hypomethylation being more associated to epigenetic changes at blood tissue-specific enhancer elements. CONCLUSIONS: These data show that epigenetic remodeling is dramatically reduced after the first 5 years of life. However, there are certain loci which continue to manifest DNA methylation changes, pointing towards a possible functionality beyond early development. Furthermore, our results deepen the understanding of the genomic context associated to hyper- or hypomethylation alterations during time, suggesting that hypomethylation of blood tissue-specific enhancer elements could be of importance in the establishment of functional states in blood tissue during early-life.


Subject(s)
DNA Methylation/genetics , Genome, Human , Child , Child, Preschool , Chromatin/metabolism , CpG Islands/genetics , Female , Humans , Infant, Newborn , Longitudinal Studies , Male , Reproducibility of Results
9.
Acta Neuropathol ; 138(6): 1053-1074, 2019 12.
Article in English | MEDLINE | ID: mdl-31428936

ABSTRACT

Tumors have aberrant proteomes that often do not match their corresponding transcriptome profiles. One possible cause of this discrepancy is the existence of aberrant RNA modification landscapes in the so-called epitranscriptome. Here, we report that human glioma cells undergo DNA methylation-associated epigenetic silencing of NSUN5, a candidate RNA methyltransferase for 5-methylcytosine. In this setting, NSUN5 exhibits tumor-suppressor characteristics in vivo glioma models. We also found that NSUN5 loss generates an unmethylated status at the C3782 position of 28S rRNA that drives an overall depletion of protein synthesis, and leads to the emergence of an adaptive translational program for survival under conditions of cellular stress. Interestingly, NSUN5 epigenetic inactivation also renders these gliomas sensitive to bioactivatable substrates of the stress-related enzyme NQO1. Most importantly, NSUN5 epigenetic inactivation is a hallmark of glioma patients with long-term survival for this otherwise devastating disease.


Subject(s)
Brain Neoplasms/metabolism , Epigenesis, Genetic , Glioma/metabolism , Methyltransferases/metabolism , Muscle Proteins/metabolism , Protein Biosynthesis/physiology , Ribosomes/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , DNA Methylation , Humans , Methyltransferases/genetics , Mice, Nude , Muscle Proteins/genetics , Neoplasm Transplantation , RNA, Ribosomal, 28S
11.
Proc Natl Acad Sci U S A ; 113(3): E328-37, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26729869

ABSTRACT

Nuclear corepressor 1 (NCoR) associates with nuclear receptors and other transcription factors leading to transcriptional repression. We show here that NCoR depletion enhances cancer cell invasion and increases tumor growth and metastatic potential in nude mice. These changes are related to repressed transcription of genes associated with increased metastasis and poor prognosis in patients. Strikingly, transient NCoR silencing leads to heterochromatinization and stable silencing of the NCoR gene, suggesting that NCoR loss can be propagated, contributing to tumor progression even in the absence of NCoR gene mutations. Down-regulation of the thyroid hormone receptor ß1 (TRß) appears to be associated with cancer onset and progression. We found that expression of TRß increases NCoR levels and that this induction is essential in mediating inhibition of tumor growth and metastasis by this receptor. Moreover, NCoR is down-regulated in human hepatocarcinomas and in the more aggressive breast cancer tumors, and its expression correlates positively with that of TRß. These data provide a molecular basis for the anticancer actions of this corepressor and identify NCoR as a potential molecular target for development of novel cancer therapies.


Subject(s)
Homeostasis , Nuclear Receptor Co-Repressor 1/genetics , Aged , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation , DNA Methylation/genetics , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Heterochromatin/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 2/metabolism , Promoter Regions, Genetic/genetics , RNA, Small Interfering/metabolism , Thyroid Hormone Receptors beta , Xenograft Model Antitumor Assays
12.
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
13.
Blood ; 137(7): 994-999, 2021 02 18.
Article in English | MEDLINE | ID: mdl-32915956
15.
J Transl Med ; 14(1): 160, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27259700

ABSTRACT

BACKGROUND: Early life epigenetic programming influences adult health outcomes. Moreover, DNA methylation levels have been found to change more rapidly during the first years of life. Our aim was the identification and characterization of the CpG sites that are modified with time during the first years of life. We hypothesize that these DNA methylation changes would lead to the detection of genes that might be epigenetically modulated by environmental factors during early childhood and which, if disturbed, might contribute to susceptibility to diseases later in life. METHODS: The study of the DNA methylation pattern of 485577 CpG sites was performed on 30 blood samples from 15 subjects, collected both at birth and at 5 years old, using Illumina(®) Infinium 450 k array. To identify differentially methylated CpG (dmCpG) sites, the methylation status of each probe was examined using linear models and the Empirical Bayes Moderated t test implemented in the limma package of R/Bioconductor. Surogate variable analysis was used to account for batch effects. RESULTS: DNA methylation levels significantly changed from birth to 5 years of age in 6641 CpG sites. Of these, 36.79 % were hypermethylated and were associated with genes related mainly to developmental ontology terms, while 63.21 % were hypomethylated probes and associated with genes related to immune function. CONCLUSIONS: Our results suggest that DNA methylation alterations with age during the first years of life might play a significant role in development and the regulation of leukocyte-specific functions. This supports the idea that blood leukocytes experience genome remodeling related to their interaction with environmental factors, underlining the importance of environmental exposures during the first years of life and suggesting that new strategies should be take into consideration for disease prevention.


Subject(s)
DNA Methylation/genetics , Centromere/metabolism , Child, Preschool , Cluster Analysis , CpG Islands/genetics , Gene Ontology , Humans , Infant , Infant, Newborn , Longitudinal Studies , Telomere/metabolism
16.
J Transl Med ; 14(1): 207, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27393146

ABSTRACT

BACKGROUND: Age-associated changes in genomic DNA methylation have been primarily attributed to 5-methylcytosine (5mC). However, the recent discovery of 5-hydroxymethylcytosine (5hmC) suggests that this epigenetic mark might also play a role in the process. METHODS: Here, we analyzed the genome-wide profile of 5hmc in mesenchymal stem cells (MSCs) obtained from bone-marrow donors, aged 2-89 years. RESULTS: We identified 10,685 frequently hydroxymethylated CpG sites in MSCs that were, as in other cell types, significantly associated with low density CpG regions, introns, the histone posttranslational modification H3k4me1 and enhancers. Study of the age-associated changes to 5hmC identified 785 hyper- and 846 hypo-hydroxymethylated CpG sites in the MSCs obtained from older individuals. CONCLUSIONS: DNA hyper-hydroxymethylation in the advanced-age group was associated with loss of 5mC, which suggests that, at specific CpG sites, this epigenetic modification might play a role in DNA methylation changes during lifetime. Since bone-marrow MSCs have many clinical applications, and the fact that the epigenomic alterations in this cell type associated with aging identified in this study could have associated functional effects, the age of donors should be taken into account in clinical settings.


Subject(s)
5-Methylcytosine/analogs & derivatives , Aging/genetics , Bone Marrow Cells/cytology , DNA Methylation/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , 5-Methylcytosine/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Chromatin/metabolism , CpG Islands/genetics , Genome, Human , Genomics , Humans , Middle Aged , Young Adult
17.
Blood ; 134(11): 900-905, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31221673
18.
Trends Genet ; 28(10): 474-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22704242

ABSTRACT

Aberrant promoter DNA hypermethylation of tumor suppressor genes is a hallmark of cancer. This alteration is largely dependent on the action of de novo DNA methyltransferases (DNMTs) early during tumor progression, which supports the oncogenic role for these enzymes. However, recent research has identified several inactivating mutations of de novo DNMTs in various types of tumor. In addition, it has been shown that loss of de novo DNA methylation activity at advanced tumor stages leads to the promoter DNA demethylation-dependent expression of specific oncogenes. These new data support the notion that de novo DNMTs also have an important role in the maintenance of DNA methylation and suggest that, in addition to acting as oncogenes, they also behave as tumor suppressors. This potential dual role might have clinical implications, as DNMTs are currently considered bona fide targets in cancer therapy.


Subject(s)
DNA Modification Methylases/metabolism , Oncogenes , Tumor Suppressor Proteins/metabolism , Animals , Epigenesis, Genetic , Humans , Neoplasms/enzymology , Tumor Suppressor Proteins/genetics
19.
Genome Res ; 22(2): 407-19, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21613409

ABSTRACT

Most of the studies characterizing DNA methylation patterns have been restricted to particular genomic loci in a limited number of human samples and pathological conditions. Herein, we present a compromise between an extremely comprehensive study of a human sample population with an intermediate level of resolution of CpGs at the genomic level. We obtained a DNA methylation fingerprint of 1628 human samples in which we interrogated 1505 CpG sites. The DNA methylation patterns revealed show this epigenetic mark to be critical in tissue-type definition and stemness, particularly around transcription start sites that are not within a CpG island. For disease, the generated DNA methylation fingerprints show that, during tumorigenesis, human cancer cells underwent a progressive gain of promoter CpG-island hypermethylation and a loss of CpG methylation in non-CpG-island promoters. Although transformed cells are those in which DNA methylation disruption is more obvious, we observed that other common human diseases, such as neurological and autoimmune disorders, had their own distinct DNA methylation profiles. Most importantly, we provide proof of principle that the DNA methylation fingerprints obtained might be useful for translational purposes by showing that we are able to identify the tumor type origin of cancers of unknown primary origin (CUPs). Thus, the DNA methylation patterns identified across the largest spectrum of samples, tissues, and diseases reported to date constitute a baseline for developing higher-resolution DNA methylation maps and provide important clues concerning the contribution of CpG methylation to tissue identity and its changes in the most prevalent human diseases.


Subject(s)
DNA Methylation , Cell Line , Cell Transformation, Neoplastic/genetics , Cluster Analysis , CpG Islands , Epigenomics/methods , Gene Expression Profiling , Gene Expression Regulation , Humans , Neoplasms/genetics
20.
J Transl Med ; 13: 25, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25623364

ABSTRACT

BACKGROUND: Maternal smoking during pregnancy is a major risk factor for adverse health outcomes. The main objective of the study was to assess the impact of in utero tobacco exposure on DNA methylation in children born at term with appropriate weight at birth. METHODS: Twenty mother-newborn dyads, after uncomplicated pregnancies, in the absence of perinatal illness were included. All mothers were healthy with no cardiovascular risk factors, except for the associated risks among those mothers who smoked. Umbilical cord blood and maternal peripheral venous blood were collected and an epigenome-wide association study was performed using a 450 K epigenome-wide scan (Illumina Infinium HumanMethylation 450BeadChip) with adjustment to normalize the DNA methylation for data cell variability in whole blood. RESULTS: The maternal plasmatic cotinine levels ranged from 10.70-115.40 ng/ml in the exposed group to 0-0.59 ng/ml in the non-exposed group. After adjusting for multiple comparisons in 427102 probes, statistically significant differences for 31 CpG sites, associated to 25 genes were observed. There was a greater than expected proportion of statistically-significant loci located in CpG islands (Fisher's exact test, p = 0.029) and of those CpG islands, 90.3% exhibit higher methylation levels in the exposed group. The most striking and significant CpG site, cg05727225, is located in the chromosome 11p15.4, within the adrenomedullin gene. CONCLUSIONS: In utero tobacco exposure, even in the absence of fetal growth restriction, may alter the epigenome, contributing to global DNA hypomethylation. Therefore, DNA status can be used as a biomarker of prenatal insults. Considering the possibility to reverse epigenetic modifications, a window of opportunity exists to change the programmed chronic disease.


Subject(s)
DNA Methylation , Maternal Exposure , Tobacco Smoke Pollution , Adrenomedullin/chemistry , Adult , Cluster Analysis , CpG Islands , Epigenesis, Genetic , Epigenomics , Female , Fetal Blood , Gene Expression Regulation , Genome-Wide Association Study , Humans , Immunoassay , Infant, Newborn , Mothers , Pregnancy , Risk Factors , Nicotiana/adverse effects , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL