Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 12(1): 6925, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35484167

ABSTRACT

Evidence about the involvement of genetic factors in the development of gambling disorder (GD) has been assessed. Among studies assessing heritability and biological vulnerability for GD, neurotrophin (NTF) genes have emerged as promising targets, since a growing literature showed a possible link between NTF and addiction-related disorders. Thus, we aimed to explore the role of NTF genes and GD with the hypothesis that some NTF gene polymorphisms could constitute biological risk factors. The sample included 166 patients with GD and 191 healthy controls. 36 single nucleotide polymorphisms (SNPs) from NTFs (NGF, NGFR, NTRK1, BDNF, NTRK2, NTF3, NTRK3, NTF4, CNTF and CNTFR) were selected and genotyped. Linkage disequilibrium (LD) and haplotype constructions were analyzed, in relationship with the presence of GD. Finally, regulatory elements overlapping the identified SNPs variants associated with GD were searched. The between groups comparisons of allele frequencies indicated that 6 SNPs were potentially associated with GD. Single and multiple-marker analyses showed a strong association between both NTF3 and NTRK2 genes, and GD. The present study supports the involvement of the NTF family in the aetiopathogenesis of GD. An altered cross-regulation of different NTF members signalling pathways might be considered as a biological vulnerability factor for GD.


Subject(s)
Gambling , Gambling/genetics , Gene Frequency , Haplotypes , Humans , Nerve Growth Factors/genetics , Polymorphism, Single Nucleotide
2.
Methods ; 187: 68-76, 2021 03.
Article in English | MEDLINE | ID: mdl-32360441

ABSTRACT

Super resolution microscopy has changed our capability to visualize and understand spatial arrangements of RNA- and protein-containing domains in individual cells. In a previous study, we described a novel lncRNA, Tumor-associated NBL2 transcript (TNBL), which originates from a primate specific macrosatellite repeat. We aimed to describe several aspects of TNBL lncRNA, with one focus being pinpointing its precise location in the nucleus, as well as visualizing its interactions with proteins to deduce its functionality. Using a combination of STimulated Emission Depletion (STED) super resolution microscopy, single molecule RNA (smRNA) FISH against TNBL, and immunofluorescence against SAM68 perinucleolar body, we resolved the spatial complexity of the interaction between TNBL aggregates and SAM68 bodies at the perinucleolar region. Here, we describe protocols for a step-by-step optimized smRNA FISH/IF and STED imaging, detailing parameter settings, and three-dimensional data analysis of spatial positioning of subnuclear structures. These protocols can be employed for single-cell imaging of complex nuclear RNA-protein structures.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , Epigenomics/methods , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , Single Molecule Imaging/methods , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , In Situ Hybridization, Fluorescence/methods , Microscopy, Fluorescence/methods , RNA, Long Noncoding/analysis , Spatio-Temporal Analysis
4.
Nucleic Acids Res ; 46(11): 5504-5524, 2018 06 20.
Article in English | MEDLINE | ID: mdl-29912433

ABSTRACT

Primate-specific NBL2 macrosatellite is hypomethylated in several types of tumors, yet the consequences of this DNA hypomethylation remain unknown. We show that NBL2 conserved repeats are close to the centromeres of most acrocentric chromosomes. NBL2 associates with the perinucleolar region and undergoes severe demethylation in a subset of colorectal cancer (CRC). Upon DNA hypomethylation and histone acetylation, NBL2 repeats are transcribed in tumor cell lines and primary CRCs. NBL2 monomers exhibit promoter activity, and are contained within novel, non-polyA antisense lncRNAs, which we designated TNBL (Tumor-associated NBL2 transcript). TNBL is stable throughout the mitotic cycle, and in interphase nuclei preferentially forms a perinucleolar aggregate in the proximity of a subset of NBL2 loci. TNBL aggregates interact with the SAM68 perinucleolar body in a mirror-image cancer specific perinucleolar structure. TNBL binds with high affinity to several proteins involved in nuclear functions and RNA metabolism, such as CELF1 and NPM1. Our data unveil novel DNA and RNA structural features of a non-coding macrosatellite frequently altered in cancer.


Subject(s)
Colonic Neoplasms/genetics , DNA Methylation/genetics , DNA, Satellite/genetics , RNA, Long Noncoding/genetics , Acetylation , Breast Neoplasms/genetics , CELF1 Protein/metabolism , Caco-2 Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Female , HCT116 Cells , Histones/metabolism , Humans , Mitosis/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Ovarian Neoplasms/genetics
5.
Epigenetics ; 12(7): 515-526, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28426282

ABSTRACT

Abundant repetitive DNA sequences are an enigmatic part of the human genome. Despite increasing evidence on the functionality of DNA repeats, their biologic role is still elusive and under frequent debate. Macrosatellites are the largest of the tandem DNA repeats, located on one or multiple chromosomes. The contribution of macrosatellites to genome regulation and human health was demonstrated for the D4Z4 macrosatellite repeat array on chromosome 4q35. Reduced copy number of D4Z4 repeats is associated with local euchromatinization and the onset of facioscapulohumeral muscular dystrophy. Although the role other macrosatellite families may play remains rather obscure, their diverse functionalities within the genome are being gradually revealed. In this review, we will outline structural and functional features of coding and noncoding macrosatellite repeats, and highlight recent findings that bring these sequences into the spotlight of genome organization and disease development.


Subject(s)
DNA, Satellite , Muscular Dystrophy, Facioscapulohumeral/genetics , Chromosomes, Human, Pair 4/genetics , Epigenesis, Genetic , Genome, Human , Humans
6.
Epigenetics ; 12(3): 238-245, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-28121228

ABSTRACT

The Barcelona Conference on Epigenetics and Cancer (BCEC) entitled "Beyond Cancer Genomes" took place October 13th and 14th 2016 in Barcelona. The 2016 BCEC was the fourth edition of a series of annual conferences coordinated by Marcus Buschbeck and subsequently organized by leading research centers in Barcelona together with B•DEBATE, a joint initiative of BIOCAT and "La Caixa" Foundation. Salvador Aznar-Benitah, Eduard Batlle, and Raúl Méndez from the Institute for Research in Biomedicine in Barcelona selected the 2016 BCEC panel of speakers. As the title indicates, this year's conference expanded the epigenetic focus to include additional cancer-relevant topics, such as tumor heterogeneity and RNA regulation. Methods to develop therapeutic approaches on the basis of novel insights have been discussed in great detail. The conference has attracted 217 participants from 11 countries.


Subject(s)
Epigenesis, Genetic , Neoplasms/genetics , Humans
7.
Epigenomes ; 1(1)2017 Jun.
Article in English | MEDLINE | ID: mdl-31867127

ABSTRACT

DNA hypomethylation at repetitive elements accounts for the genome-wide DNA hypomethylation common in cancer, including colorectal cancer (CRC). We identified a pericentromeric repeat element called SST1 frequently hypomethylated (>5% demethylation compared with matched normal tissue) in several cancers, including 28 of 128 (22%) CRCs. SST1 somatic demethylation associated with genome damage, especially in tumors with wild-type TP53. Seven percent of the 128 CRCs exhibited a higher ("severe") level of demethylation (≥10%) that co-occurred with TP53 mutations. SST1 demethylation correlated with distinct histone marks in CRC cell lines and primary tumors: demethylated SST1 associated with high levels of the repressive histone 3 lysine 27 trimethylation (H3K27me3) mark and lower levels of histone 3 lysine 9 trimethylation (H3K9me3). Furthermore, induced demethylation of SST1 by 5-aza-dC led to increased H3K27me3 and reduced H3K9me3. Thus, in some CRCs, SST1 demethylation reflects an epigenetic reprogramming associated with changes in chromatin structure that may affect chromosomal integrity. The chromatin remodeler factor, the helicase lymphoid-specific (HELLS) enzyme, called the "epigenetic guardian of repetitive elements", interacted with SST1 as shown by chromatin immunoprecipitation, and down-regulation of HELLS by shRNA resulted in demethylation of SST1 in vitro. Altogether these results suggest that HELLS contributes to SST1 methylation maintenance. Alterations in HELLS recruitment and function could contribute to the somatic demethylation of SST1 repeat elements undergone before and/or during CRC pathogenesis.

8.
Front Aging Neurosci ; 7: 123, 2015.
Article in English | MEDLINE | ID: mdl-26217219

ABSTRACT

Regenerative capacity of skeletal muscles resides in satellite cells, a self-renewing population of muscle cells. Several studies are investigating epigenetic mechanisms that control myogenic proliferation and differentiation to find new approaches that could boost regeneration of endogenous myogenic progenitor populations. In recent years, a lot of effort has been applied to purify, expand and manipulate adult stem cells from muscle tissue. However, this population of endogenous myogenic progenitors in adults is limited and their access is difficult and invasive. Therefore, other sources of stem cells with potential to regenerate muscles need to be examined. An excellent candidate could be a population of adult stromal cells within fat characterized by mesenchymal properties, which have been termed adipose-derived stem cells (ASCs). These progenitor adult stem cells have been successfully differentiated in vitro to osteogenic, chondrogenic, neurogenic and myogenic lineages. Autologous ASCs are multipotent and can be harvested with low morbidity; thus, they hold promise for a range of therapeutic applications. This review will summarize the use of ASCs in muscle regenerative approaches.

9.
Epigenetics ; 10(4): 352-9, 2015.
Article in English | MEDLINE | ID: mdl-25849957

ABSTRACT

The Second German-Catalan Workshop on Epigenetics and Cancer was held in Barcelona on November 19-21, 2014. The workshop brought together, for the second time, scientists from 2 German and 2 Catalan research institutions: the DKFZ, from Heidelberg, the CRCME, from Freiburg, and the IMPPC and PEBC/IDIBELL, both from Barcelona. The German-Catalan Workshops are intended to establish the framework for building a Research School to foster collaborations between researchers from the different institutions. Exchange programs for graduate students are among the activities of the future School. The topics presented and discussed in 33 talks were diverse and included work on DNA methylation, histone modifications, chromatin biology, characterization of imprinted regions in human tissues, non-coding RNAs, and epigenetic drug discovery. Among novel developments from the previous Workshop are the report of the epigenetics angle of the Warburg effect and the long-range trans-acting interaction of DNA methylation and of nucleosome remodeling. A shift in the view on DNA methylation became apparent by the realization of the intertwined interplay between hyper- and hypo-methylation in differentiation and cancer.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Neoplasms/genetics , Chromatin Assembly and Disassembly , Drug Discovery , Humans
10.
EMBO J ; 31(2): 301-16, 2012 Jan 18.
Article in English | MEDLINE | ID: mdl-22068056

ABSTRACT

Tissue-specific transcriptional activators initiate differentiation towards specialized cell types by inducing chromatin modifications permissive for transcription at target loci, through the recruitment of SWItch/Sucrose NonFermentable (SWI/SNF) chromatin-remodelling complex. However, the molecular mechanism that regulates SWI/SNF nuclear distribution in response to differentiation signals is unknown. We show that the muscle determination factor MyoD and the SWI/SNF subunit BAF60c interact on the regulatory elements of MyoD-target genes in myoblasts, prior to activation of transcription. BAF60c facilitates MyoD binding to target genes and marks the chromatin for signal-dependent recruitment of the SWI/SNF core to muscle genes. BAF60c phosphorylation on a conserved threonine by differentiation-activated p38α kinase is the signal that promotes incorporation of MyoD-BAF60c into a Brg1-based SWI/SNF complex, which remodels the chromatin and activates transcription of MyoD-target genes. Our data support an unprecedented two-step model by which pre-assembled BAF60c-MyoD complex directs recruitment of SWI/SNF to muscle loci in response to differentiation cues.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , MAP Kinase Signaling System , Muscle Development/physiology , Muscle Proteins/physiology , MyoD Protein/physiology , Transcription Factors/physiology , Animals , Cell Line , Chromatin/genetics , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/physiology , Fibroblasts/metabolism , Gene Expression Regulation/genetics , HeLa Cells/metabolism , Humans , Mice , Multiprotein Complexes , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/chemistry , Muscle Proteins/genetics , Myoblasts/metabolism , Nuclear Proteins/physiology , Phosphorylation , Phosphothreonine/analysis , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemistry , Transcription Factors/genetics , Two-Hybrid System Techniques , p38 Mitogen-Activated Protein Kinases/physiology
11.
Cell Cycle ; 10(2): 191-8, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21220942

ABSTRACT

Muscle regeneration relies on adult muscle stem (satellite) cells. Inflammatory cues released within the regenerative microenvironment, such as TNFα, instruct different components of the satellite cell niche toward specialized tasks by regulating specific subsets of genes in each individual cell type. However, how regeneration cues are deciphered and interpreted by the multitude of cell types within the regenerative environment is unknown. We have recently identified an inflammation-activated signaling, consisting of p38α-mediated recruitment of polycomb repressive complex 2 (PRC2) to the Pax7 promoter, in satellite cells. Here we show that p38α-PRC2 regulation of Pax7 expression is restricted to a discrete stage of satellite cell-mediated regeneration. In activated satellite cells, Pax7 locus shows a "bivalent" chromatin signature, with co-existence of H3-K27(3me) and H3-K4(3me), that appears to confer responsiveness to p38α-PRC2 signaling. p38α activation resolves bivalence to H3-K27(3me) which results in Pax7 repression, while p38α blockade promotes Pax7 expression by preventing PRC2-mediated H3-K27(3me) and leading to relative increase in H3-K4(3me). Interestingly, in satellite cell-derived myotubes Pax7 expression cannot be re-induced by p38α blockade, revealing a post-mitotic resistance of Pax7 gene to inflammatory cues. Likewise, in other cell types, such as muscle-derived fibroblasts, Pax7 locus is constitutively repressed by PRC2 and is unresponsive to p38α signaling. Finally, we show that Pax7 repression in embryonic stem cells is not directed by p38α signaling, although it is mediated by PRC2. This evidence indicates a cell type- and differentiation-stage specific control of Pax7 transcription by the p38α-PRC2.


Subject(s)
Mitogen-Activated Protein Kinase 14/metabolism , PAX7 Transcription Factor/metabolism , Repressor Proteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Differentiation , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/metabolism , Mice , PAX7 Transcription Factor/genetics , Polycomb-Group Proteins , Satellite Cells, Skeletal Muscle/cytology , Signal Transduction
12.
Cell Stem Cell ; 7(4): 455-69, 2010 Oct 08.
Article in English | MEDLINE | ID: mdl-20887952

ABSTRACT

How regeneration cues are converted into the epigenetic information that controls gene expression in adult stem cells is currently unknown. We identified an inflammation-activated signaling in muscle stem (satellite) cells, by which the polycomb repressive complex 2 (PRC2) represses Pax7 expression during muscle regeneration. TNF-activated p38α kinase promotes the interaction between YY1 and PRC2, via threonine 372 phosphorylation of EZH2, the enzymatic subunit of the complex, leading to the formation of repressive chromatin on Pax7 promoter. TNF-α antibodies stimulate satellite cell proliferation in regenerating muscles of dystrophic or normal mice. Genetic knockdown or pharmacological inhibition of the enzymatic components of the p38/PRC2 signaling--p38α and EZH2--invariably promote Pax7 expression and expansion of satellite cells that retain their differentiation potential upon signaling resumption. Genetic knockdown of Pax7 impaired satellite cell proliferation in response to p38 inhibition, thereby establishing the biological link between p38/PRC2 signaling to Pax7 and satellite cell decision to proliferate or differentiate.


Subject(s)
PAX7 Transcription Factor/metabolism , Quadriceps Muscle/physiology , Regeneration , Repressor Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Epigenesis, Genetic , Fluorescent Antibody Technique , Gene Knockdown Techniques , Inflammation/genetics , Mice , Mice, Inbred C57BL , PAX7 Transcription Factor/genetics , Polycomb-Group Proteins , Promoter Regions, Genetic , Quadriceps Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism
13.
Genes Dev ; 22(15): 2125-38, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18676817

ABSTRACT

Myogenin is the dominant transcriptional regulator of embryonic and fetal muscle differentiation and during maturation is profoundly down-regulated. We show that a highly conserved 17-bp DNA cis-acting sequence element located upstream of the myogenin promoter (myogHCE) is essential for postnatal repression of myogenin in transgenic animals. We present multiple lines of evidence supporting the idea that repression is mediated by the Y-box protein MSY-3. Electroporation in vivo shows that myogHCE and MSY-3 are required for postnatal repression. We further show that, in the C2C12 cell culture system, ectopic MSY-3 can repress differentiation, while reduced MSY-3 promotes premature differentiation. MSY-3 binds myogHCE simultaneously with the homeodomain protein Pbx in postnatal innervated muscle. We therefore propose a model in which the myogHCE motif operates as a switch by specifying opposing functions; one that was shown previously is regulated by MyoD and Pbx and it specifies a chromatin opening, gene-activating function at the time myoblasts begin to differentiate; the other includes MYS-3 and Pbx, and it specifies a repression function that operates during and after postnatal muscle maturation in vivo and in myoblasts before they begin to differentiate.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation/physiology , Muscle, Skeletal/growth & development , Myogenin/genetics , RNA-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Electroporation , Genetic Vectors , Homeodomain Proteins/metabolism , Lentivirus/genetics , Mice , MyoD Protein/genetics , MyoD Protein/physiology , Myoblasts/physiology , Myogenin/physiology , Pre-B-Cell Leukemia Transcription Factor 1 , Transcription Factors/metabolism
14.
Mol Cell ; 28(2): 200-13, 2007 Oct 26.
Article in English | MEDLINE | ID: mdl-17964260

ABSTRACT

During muscle regeneration, the mechanism integrating environmental cues at the chromatin of muscle progenitors is unknown. We show that inflammation-activated MKK6-p38 and insulin growth factor 1 (IGF1)-induced PI3K/AKT pathways converge on the chromatin of muscle genes to target distinct components of the muscle transcriptosome. p38 alpha/beta kinases recruit the SWI/SNF chromatin-remodeling complex; AKT1 and 2 promote the association of MyoD with p300 and PCAF acetyltransferases, via direct phosphorylation of p300. Pharmacological or genetic interference with either pathway led to partial assembly of discrete chromatin-bound complexes, which reflected two reversible and distinct cellular phenotypes. Remarkably, PI3K/AKT blockade was permissive for chromatin recruitment of MEF2-SWI/SNF complex, whose remodeling activity was compromised in the absence of MyoD and acetyltransferases. The functional interdependence between p38 and IGF1/PI3K/AKT pathways was further established by the evidence that blockade of AKT chromatin targets was sufficient to prevent the activation of the myogenic program triggered by deliberate activation of p38 signaling.


Subject(s)
Chromatin/metabolism , Insulin-Like Growth Factor I/metabolism , MAP Kinase Kinase 6/metabolism , Muscle Development , Myoblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylation , Animals , Cell Line , Cell Shape , Chromones/pharmacology , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation, Developmental , Imidazoles/pharmacology , Insulin-Like Growth Factor I/genetics , MAP Kinase Kinase 6/genetics , Mice , Morpholines/pharmacology , Muscle Development/drug effects , Muscle Development/genetics , MyoD Protein/metabolism , Myoblasts/drug effects , Myoblasts/enzymology , Myogenic Regulatory Factors/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic , Transfection , p300-CBP Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...