Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.955
Filtrer
1.
Skin Res Technol ; 30(7): e13842, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38965799

RÉSUMÉ

BACKGROUND: As the most important modifications on the RNA level, N6-methyladenosine (m6A-) and 5-methylcytosine (m5C-) modification could have a direct influence on the RNAs. Long non-coding RNAs (lncRNAs) could also be modified by methylcytosine modification. Compared with mRNAs, the function of lncRNAs could be more potent to some extent in biological processes like tumorigenesis. Until now, rare reports have been done associated with cutaneous melanoma. Herein, we wonder if the m6A- and m5C- modified lncRNAs could influence the immune landscape and prognosis in melanoma, and we also want to find some lncRNAs which could directly affect the malignant behaviors of melanoma. METHODS: Systematically, we explored the expression pattern of m6A- and m5C- modified lncRNAs in melanoma from datasets including UCSC Xena and NCBI GEO, and the prognostic lncRNAs were selected. Then, according to the expression pattern of lncRNAs, melanoma samples from these datasets were divided into several subtypes. Prognostic model, nomogram survival model, drug sensitivity, GO, and KEGG pathway analysis were performed. Furthermore, among several selected lncRNAs, we identified one lncRNA named LINC00893 and investigated its expression pattern and its biological function in melanoma cell lines. RESULTS: We identified 27 m6A- and m5C- related lncRNAs which were significantly associated with survival, and we made a subtype analysis of melanoma samples based on these 27 lncRNAs. Among the two subtypes, we found differences of immune cells infiltration between these two subtypes. Then, LASSO algorithm was used to screen the optimized lncRNAs combination including ZNF252P-AS1, MIAT, FAM13A-AS1, LINC-PINT, LINC00893, AGAP2-AS1, OIP5-AS1, and SEMA6A-AS1. We also found that there was a significant correlation between the different risk groups predicted based on RS model and the actual prognosis. The nomogram survival model based on independent survival prognostic factors was also constructed. Besides, sensitivity to chemotherapeutic agents, GO and KEGG analysis were performed. In different risk groups, a total of 14 drug molecules with different distributions were obtained, which included AZD6482, AZD7762, AZD8055, camptothecin, dasatinib, erlotinib, gefitinib, gemcitabine, GSK269962A, nilotinib, rapamycin, and sorafenib. A total of 55 significantly related biological processes and 17 KEGG signaling pathways were screened. At last, we noticed that LINC00893 had a relatively lower expression in melanoma tissue and cell lines compared with adjacent tissues and epidermal melanocyte, and down-regulation of LINC00893 could promote the malignant behavior of melanoma cells in A875 and MV3. In these two melanoma cell lines, down-regulation of m6A-related molecules like YTHDF3 and METTL3 could promote the expression of LINC00893. CONCLUSION: We made an analysis of m6A- and m5C- related lncRNAs in melanoma samples and a prediction of these lncRNAs' role in prognosis, tumor microenvironment, immune infiltration, and clinicopathological features. We also found that LINC00893, which is potentially regulated by m6A modification, could serve as a tumor-suppressor in melanoma and play an inhibitory role in melanoma metastasis.


Sujet(s)
Adénosine , Mélanome , ARN long non codant , Tumeurs cutanées , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , Mélanome/génétique , Mélanome/anatomopathologie , Mélanome/mortalité , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/mortalité , Adénosine/analogues et dérivés , Adénosine/métabolisme , Pronostic , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Nomogrammes
2.
Epigenetics ; 19(1): 2374979, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38970823

RÉSUMÉ

TET1/2/3 dioxygenases iteratively demethylate 5-methylcytosine, beginning with the formation of 5-hydroxymethylcytosine (5hmC). The post-mitotic brain maintains higher levels of 5hmC than most peripheral tissues, and TET1 ablation studies have underscored the critical role of TET1 in brain physiology. However, deletion of Tet1 precludes the disentangling of the catalytic and non-catalytic functions of TET1. Here, we dissect these functions of TET1 by comparing adult cortex of Tet1 wildtype (Tet1 WT), a novel Tet1 catalytically dead mutant (Tet1 HxD), and Tet1 knockout (Tet1 KO) mice. Using DNA methylation array, we uncover that Tet1 HxD and KO mutations perturb the methylation status of distinct subsets of CpG sites. Gene ontology (GO) analysis on specific differential 5hmC regions indicates that TET1's catalytic activity is linked to neuronal-specific functions. RNA-Seq further shows that Tet1 mutations predominantly impact the genes that are associated with alternative splicing. Lastly, we performed High-performance Liquid Chromatography Mass-Spectrometry lipidomics on WT and mutant cortices and uncover accumulation of lysophospholipids lysophosphatidylethanolamine and lysophosphatidylcholine in Tet1 HxD cortex. In summary, we show that Tet1 HxD does not completely phenocopy Tet1 KO, providing evidence that TET1 modulates distinct cortical functions through its catalytic and non-catalytic roles.


Sujet(s)
5-Méthyl-cytosine , Cortex cérébral , Méthylation de l'ADN , Protéines proto-oncogènes , Animaux , Souris , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Cortex cérébral/métabolisme , Souris knockout , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Ilots CpG , Mutation
3.
Nat Commun ; 15(1): 5580, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38961062

RÉSUMÉ

DNA methylation plays an important role in various biological processes, including cell differentiation, ageing, and cancer development. The most important methylation in mammals is 5-methylcytosine mostly occurring in the context of CpG dinucleotides. Sequencing methods such as whole-genome bisulfite sequencing successfully detect 5-methylcytosine DNA modifications. However, they suffer from the serious drawbacks of short read lengths and might introduce an amplification bias. Here we present Rockfish, a deep learning algorithm that significantly improves read-level 5-methylcytosine detection by using Nanopore sequencing. Rockfish is compared with other methods based on Nanopore sequencing on R9.4.1 and R10.4.1 datasets. There is an increase in the single-base accuracy and the F1 measure of up to 5 percentage points on R.9.4.1 datasets, and up to 0.82 percentage points on R10.4.1 datasets. Moreover, Rockfish shows a high correlation with whole-genome bisulfite sequencing, requires lower read depth, and achieves higher confidence in biologically important regions such as CpG-rich promoters while being computationally efficient. Its superior performance in human and mouse samples highlights its versatility for studying 5-methylcytosine methylation across varied organisms and diseases. Finally, its adaptable architecture ensures compatibility with new versions of pores and chemistry as well as modification types.


Sujet(s)
5-Méthyl-cytosine , Ilots CpG , Méthylation de l'ADN , Séquençage par nanopores , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/composition chimique , Séquençage par nanopores/méthodes , Animaux , Souris , Humains , Ilots CpG/génétique , Apprentissage profond , Algorithmes , Analyse de séquence d'ADN/méthodes , Séquençage du génome entier/méthodes , Sulfites/composition chimique
4.
Mol Cancer ; 23(1): 139, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970106

RÉSUMÉ

BACKGROUND: Radioresistance is the leading cause of death in advanced cervical cancer (CC). Dysregulation of RNA modification has recently emerged as a regulatory mechanism in radiation and drug resistance. We aimed to explore the biological function and clinical significance of 5-methylcytosine (m5C) in cervical cancer radiosensitivity. METHODS: The abundance of RNA modification in radiotherapy-resistant and sensitive CC specimens was quantified by liquid chromatography-tandem mass spectrometry. The essential RNA modification-related genes involved in CC radiosensitivity were screened via RNA sequencing. The effect of NSUN6 on radiosensitivity was verified in CC cell lines, cell-derived xenograft (CDX), and 3D bioprinted patient-derived organoid (PDO). The mechanisms of NSUN6 in regulating CC radiosensitivity were investigated by integrative m5C sequencing, mRNA sequencing, and RNA immunoprecipitation. RESULTS: We found a higher abundance of m5C modification in resistant CC samples, and NSUN6 was the essential m5C-regulating gene concerning radiosensitivity. NSUN6 overexpression was clinically correlated with radioresistance and poor prognosis in cervical cancer. Functionally, higher NSUN6 expression was associated with radioresistance in the 3D PDO model of cervical cancer. Moreover, silencing NSUN6 increased CC radiosensitivity in vivo and in vitro. Mechanistically, NDRG1 was one of the downstream target genes of NSUN6 identified by integrated m5C-seq, mRNA-seq, and functional validation. NSUN6 promoted the m5C modification of NDRG1 mRNA, and the m5C reader ALYREF bound explicitly to the m5C-labeled NDRG1 mRNA and enhanced NDRG1 mRNA stability. NDRG1 overexpression promoted homologous recombination-mediated DNA repair, which in turn led to radioresistance in cervical cancer. CONCLUSIONS: Aberrant m5C hypermethylation and NSUN6 overexpression drive resistance to radiotherapy in cervical cancer. Elevated NSUN6 expression promotes radioresistance in cervical cancer by activating the NSUN6/ALYREF-m5C-NDRG1 pathway. The low expression of NSUN6 in cervical cancer indicates sensitivity to radiotherapy and a better prognosis.


Sujet(s)
5-Méthyl-cytosine , Protéines du cycle cellulaire , Régulation de l'expression des gènes tumoraux , Protéines et peptides de signalisation intracellulaire , ARN messager , Radiotolérance , Tumeurs du col de l'utérus , Tumeurs du col de l'utérus/génétique , Tumeurs du col de l'utérus/métabolisme , Tumeurs du col de l'utérus/radiothérapie , Tumeurs du col de l'utérus/anatomopathologie , Humains , Femelle , Radiotolérance/génétique , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Animaux , Souris , ARN messager/génétique , ARN messager/métabolisme , Lignée cellulaire tumorale , Pronostic , Tests d'activité antitumorale sur modèle de xénogreffe , Methyltransferases/génétique , Methyltransferases/métabolisme
5.
Biomolecules ; 14(6)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38927065

RÉSUMÉ

Despite extensive research on 5-methylcytosine (5mC) in relation to smoking, there has been limited exploration into the interaction between smoking and 5-hydroxymethylcytosine (5hmC). In this study, total DNA methylation (5mC+5hmC), true DNA methylation (5mC) and hydroxymethylation (5hmC) levels were profiled utilizing conventional bisulphite (BS) and oxidative bisulphite (oxBS) treatment, measured with the Illumina Infinium Methylation EPIC BeadChip. An epigenome-wide association study (EWAS) of 5mC+5hmC methylation revealed a total of 38,575 differentially methylated positions (DMPs) and 2023 differentially methylated regions (DMRs) associated with current smoking, along with 82 DMPs and 76 DMRs associated with former smoking (FDR-adjusted p < 0.05). Additionally, a focused examination of 5mC identified 33 DMPs linked to current smoking and 1 DMP associated with former smoking (FDR-adjusted p < 0.05). In the 5hmC category, eight DMPs related to current smoking and two DMPs tied to former smoking were identified, each meeting a suggestive threshold (p < 1 × 10-5). The substantial number of recognized DMPs, including 5mC+5hmC (7069/38,575, 2/82), 5mC (0/33, 1/1), and 5hmC (2/8, 0/2), have not been previously reported. Our findings corroborated previously established methylation positions and revealed novel candidates linked to tobacco smoking. Moreover, the identification of hydroxymethylated CpG sites with suggestive links provides avenues for future research.


Sujet(s)
5-Méthyl-cytosine , Méthylation de l'ADN , Fumer , Humains , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Mâle , Femelle , Fumer/génétique , Fumer/effets indésirables , Adulte d'âge moyen , Sujet âgé , Études de cohortes , Étude d'association pangénomique , Épigenèse génétique , Ilots CpG/génétique , Adulte
6.
Syst Biol Reprod Med ; 70(1): 164-173, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38913941

RÉSUMÉ

With ∼50% recurrent pregnancy loss cases being termed idiopathic (iRPL), understanding of contribution of male factors to iRPL is still lacking. Higher prevalence of sperm DNA fragmentation index (DFI) and lower sperm 5-methylcytosine (5-mC) levels have been previously reported in male partners of iRPL couples and shed light on importance of the male gamete in maintenance of a successful pregnancy. The present study aimed to determine the serum sex steroid hormone levels, sperm DFI and 5-mC and correlation between them in male partners of fertile and iRPL couples. Further, correlation between sperm DFI and 5-mC with semen parameters and paternal age in both groups were determined. 36 male partners of fertile couples and 45 male partners of women experiencing iRPL were enrolled for this study and semen and blood samples were collected. Serum testosterone and estradiol levels were measured by ELISA; sperm DFI and global 5-mC were determined by TUNEL assay and ELISA respectively. Significantly higher serum testosterone levels were noted in the iRPL group (p = 0.028). Incidence of sperm DNA fragmentation was found to be higher in the iRPL study group but with no significance difference. No significant differences in sperm 5-mC values were noted. Upon correlation analysis within both groups, strong significant negative correlation of sperm DFI % and 5-mC % was observed in the control group (p < 0.001) but not the iRPL group (p = 0.249). Hence, we infer that with lower 5-mC levels in sperm genome, there is a higher incidence of sperm DFI in fertile men. However, this trend is not noted in men of iRPL group which could possibly be due to other underlying epigenetic alterations in genomic regions probably unsusceptible to fragmentation. On the other hand, no significant correlations of semen parameters, testosterone, estradiol and paternal age with sperm DFI and 5-mC were noted in both groups.


Sujet(s)
Avortements à répétition , Fragmentation de l'ADN , Méthylation de l'ADN , Spermatozoïdes , Humains , Mâle , Avortements à répétition/génétique , Avortements à répétition/sang , Spermatozoïdes/métabolisme , Adulte , Femelle , Oestradiol/sang , Testostérone/sang , Grossesse , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/sang , Analyse du sperme , Âge paternel
7.
Genome Biol ; 25(1): 142, 2024 06 03.
Article de Anglais | MEDLINE | ID: mdl-38825692

RÉSUMÉ

BACKGROUND: Like its parent base 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC) is a direct epigenetic modification of cytosines in the context of CpG dinucleotides. 5hmC is the most abundant oxidized form of 5mC, generated through the action of TET dioxygenases at gene bodies of actively-transcribed genes and at active or lineage-specific enhancers. Although such enrichments are reported for 5hmC, to date, predictive models of gene expression state or putative regulatory regions for genes using 5hmC have not been developed. RESULTS: Here, by using only 5hmC enrichment in genic regions and their vicinity, we develop neural network models that predict gene expression state across 49 cell types. We show that our deep neural network models distinguish high vs low expression state utilizing only 5hmC levels and these predictive models generalize to unseen cell types. Further, in order to leverage 5hmC signal in distal enhancers for expression prediction, we employ an Activity-by-Contact model and also develop a graph convolutional neural network model with both utilizing Hi-C data and 5hmC enrichment to prioritize enhancer-promoter links. These approaches identify known and novel putative enhancers for key genes in multiple immune cell subsets. CONCLUSIONS: Our work highlights the importance of 5hmC in gene regulation through proximal and distal mechanisms and provides a framework to link it to genome function. With the recent advances in 6-letter DNA sequencing by short and long-read techniques, profiling of 5mC and 5hmC may be done routinely in the near future, hence, providing a broad range of applications for the methods developed here.


Sujet(s)
5-Méthyl-cytosine , Éléments activateurs (génétique) , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Humains , , Régulation de l'expression des gènes , Épigenèse génétique , Méthylation de l'ADN
8.
Open Biol ; 14(6): 230449, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862018

RÉSUMÉ

Nanopore sequencing platforms combined with supervised machine learning (ML) have been effective at detecting base modifications in DNA such as 5-methylcytosine (5mC) and N6-methyladenine (6mA). These ML-based nanopore callers have typically been trained on data that span all modifications on all possible DNA [Formula: see text]-mer backgrounds-a complete training dataset. However, as nanopore technology is pushed to more and more epigenetic modifications, such complete training data will not be feasible to obtain. Nanopore calling has historically been performed with hidden Markov models (HMMs) that cannot make successful calls for [Formula: see text]-mer contexts not seen during training because of their independent emission distributions. However, deep neural networks (DNNs), which share parameters across contexts, are increasingly being used as callers, often outperforming their HMM cousins. It stands to reason that a DNN approach should be able to better generalize to unseen [Formula: see text]-mer contexts. Indeed, herein we demonstrate that a common DNN approach (DeepSignal) outperforms a common HMM approach (Nanopolish) in the incomplete data setting. Furthermore, we propose a novel hybrid HMM-DNN approach, amortized-HMM, that outperforms both the pure HMM and DNN approaches on 5mC calling when the training data are incomplete. This type of approach is expected to be useful for calling other base modifications such as 5-hydroxymethylcytosine and for the simultaneous calling of different modifications, settings in which complete training data are not likely to be available.


Sujet(s)
5-Méthyl-cytosine , Méthylation de l'ADN , Épigenèse génétique , , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/composition chimique , 5-Méthyl-cytosine/métabolisme , Séquençage par nanopores/méthodes , Nanopores , Humains , Chaines de Markov , ADN/composition chimique , ADN/génétique
9.
Cell Death Dis ; 15(6): 387, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38824145

RÉSUMÉ

Obesity exacerbates tissue degeneration and compromises the integrity and reparative potential of mesenchymal stem/stromal cells (MSCs), but the underlying mechanisms have not been sufficiently elucidated. Mitochondria modulate the viability, plasticity, proliferative capacity, and differentiation potential of MSCs. We hypothesized that alterations in the 5-hydroxymethylcytosine (5hmC) profile of mitochondria-related genes may mediate obesity-driven dysfunction of human adipose-derived MSCs. MSCs were harvested from abdominal subcutaneous fat of obese and age/sex-matched non-obese subjects (n = 5 each). The 5hmC profile and expression of nuclear-encoded mitochondrial genes were examined by hydroxymethylated DNA immunoprecipitation sequencing (h MeDIP-seq) and mRNA-seq, respectively. MSC mitochondrial structure (electron microscopy) and function, metabolomics, proliferation, and neurogenic differentiation were evaluated in vitro, before and after epigenetic modulation. hMeDIP-seq identified 99 peaks of hyper-hydroxymethylation and 150 peaks of hypo-hydroxymethylation in nuclear-encoded mitochondrial genes from Obese- versus Non-obese-MSCs. Integrated hMeDIP-seq/mRNA-seq analysis identified a select group of overlapping (altered levels of both 5hmC and mRNA) nuclear-encoded mitochondrial genes involved in ATP production, redox activity, cell proliferation, migration, fatty acid metabolism, and neuronal development. Furthermore, Obese-MSCs exhibited decreased mitochondrial matrix density, membrane potential, and levels of fatty acid metabolites, increased superoxide production, and impaired neuronal differentiation, which improved with epigenetic modulation. Obesity elicits epigenetic changes in mitochondria-related genes in human adipose-derived MSCs, accompanied by structural and functional changes in their mitochondria and impaired fatty acid metabolism and neurogenic differentiation capacity. These observations may assist in developing novel therapies to preserve the potential of MSCs for tissue repair and regeneration in obese individuals.


Sujet(s)
Tissu adipeux , Différenciation cellulaire , Épigenèse génétique , Cellules souches mésenchymateuses , Mitochondries , Obésité , Humains , Cellules souches mésenchymateuses/métabolisme , Obésité/métabolisme , Obésité/génétique , Obésité/anatomopathologie , Mitochondries/métabolisme , Tissu adipeux/métabolisme , Différenciation cellulaire/génétique , Femelle , Mâle , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Adulte , Adulte d'âge moyen , Prolifération cellulaire
10.
Medicine (Baltimore) ; 103(25): e38623, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38905403

RÉSUMÉ

The incidence of papillary thyroid carcinoma (PTC) has increased significantly in recent years, and for patients with metastatic and recurrent PTC, the options for treatment currently available are insufficient. To date, the exact molecular mechanism underlying PTC is still not fully understood. 5-Methylcytosine (m5C) RNA methylation is associated with the prognosis of a variety of tumors. However, the molecular mechanisms and biomarkers associated with m5C in the diagnosis, treatment, and prognosis of this disease have not been fully elucidated. Ten m5C regulators with significantly different expression levels were included in this study. Immune infiltration analysis revealed significant negative correlations between most of these regulators and regulatory T cells. TRDMT1, NSUN5, and NSUN6 had high weights and strong correlations in the protein-protein interaction network. Using gene ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis, 1489 differentially expressed genes were screened from The Cancer Genome Atlas messenger RNA matrix, indicating that these differentially expressed genes were significantly enriched in various pathways and functions related to cancers. Four m5C regulators, NSUN2, NSUN4, NSUN6, and DNMT3B, were screened as prognostic markers by least absolute shrinkage and selection operator regression analysis, and NSUN2 and NSUN6 were identified as risk factors for poor prognosis. We found that the prognostic prediction model constructed using the m5C regulators NSUN2, NSUN4, NSUN6, and DNMT3B showed good prognostic prediction ability and diagnostic ability. This model was applied to predict the survival probability of patients with PTC, the prediction ability of 5-year survival was the best. The multi-factor prognostic prediction model combined with the tumor node metastasis stage and risk score grouping showed better prognostic predictive power.


Sujet(s)
Marqueurs biologiques tumoraux , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/mortalité , Cancer papillaire de la thyroïde/anatomopathologie , Pronostic , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/mortalité , Tumeurs de la thyroïde/anatomopathologie , Marqueurs biologiques tumoraux/génétique , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Mâle , Femelle , Régulation de l'expression des gènes tumoraux , Méthylation , Adulte d'âge moyen
11.
Anal Chem ; 96(24): 9984-9993, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38833588

RÉSUMÉ

Metal-organic frameworks (MOFs) show unique advantages in simulating the dynamics and fidelity of natural coordination. Inspired by zinc finger protein, a second linker was introduced to affect the homogeneous MOF system and thus facilitate the emergence of diverse functionalities. Under the systematic identification of 12 MOF species (i.e., metal ions, linkers) and 6 second linkers (trigger), a dissipative system consisting of Co-BDC-NO2 and o-phenylenediamine (oPD) was screened out, which can rapidly and in situ generate a high photothermal complex (η = 36.9%). Meanwhile, both the carboxylation of epigenetic modifications and metal ion (Fe3+, Ni2+, Cu2+, Zn2+, Co2+ and Mn2+) screening were utilized to improve the local coordination environment so that the adaptable Co-MOF growth on the DNA strand was realized. Thus, epigenetic modification information on DNA was converted to an amplified metal ion signal, and then oPD was further introduced to generate bimodal dissipative signals by which a simple, high-sensitivity detection strategy of 5-hydroxymethylcytosine (LOD = 0.02%) and 5-formylcytosine (LOD = 0.025‰) was developed. The strategy provides one low-cost method (< 0.01 $/sample) for quantifying global epigenetic modifications, which greatly promotes epigenetic modification-based early disease diagnosis. This work also proposes a general heterocoordination design concept for molecular recognition and signal transduction, opening a new MOF-based sensing paradigm.


Sujet(s)
Cobalt , ADN , Épigenèse génétique , Réseaux organométalliques , Phénylènediamines , Réseaux organométalliques/composition chimique , Cobalt/composition chimique , ADN/composition chimique , Phénylènediamines/composition chimique , 5-Méthyl-cytosine/composition chimique , 5-Méthyl-cytosine/analyse , 5-Méthyl-cytosine/analogues et dérivés , Cytosine/composition chimique , Cytosine/analogues et dérivés , Limite de détection
12.
Nat Commun ; 15(1): 5136, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879605

RÉSUMÉ

Coordination of neuronal differentiation with expansion of the neuroepithelial/neural progenitor cell (NEPC/NPC) pool is essential in early brain development. Our in vitro and in vivo studies identify independent and opposing roles for two neural-specific and differentially expressed non-coding RNAs derived from the same locus: the evolutionarily conserved lncRNA Rncr3 and the embedded microRNA miR124a-1. Rncr3 regulates NEPC/NPC proliferation and controls the biogenesis of miR124a, which determines neuronal differentiation. Rncr3 conserved exons 2/3 are cytosine methylated and bound by methyl-CpG binding protein MeCP2, which restricts expression of miR124a embedded in exon 4 to prevent premature neuronal differentiation, and to orchestrate proper brain growth. MeCP2 directly binds cytosine-methylated Rncr3 through previously unrecognized lysine residues and suppresses miR124a processing by recruiting PTBP1 to block access of DROSHA-DGCR8. Thus, miRNA processing is controlled by lncRNA m5C methylation along with the defined m5C epitranscriptomic RNA reader protein MeCP2 to coordinate brain development.


Sujet(s)
Protéine-2 de liaison au CpG méthylé , microARN , Cellules souches neurales , Neurogenèse , ARN long non codant , microARN/métabolisme , microARN/génétique , Protéine-2 de liaison au CpG méthylé/métabolisme , Protéine-2 de liaison au CpG méthylé/génétique , Neurogenèse/génétique , Animaux , Souris , ARN long non codant/métabolisme , ARN long non codant/génétique , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Encéphale/métabolisme , Encéphale/embryologie , Humains , Différenciation cellulaire , Méthylation de l'ADN , Protéine PTB/métabolisme , Protéine PTB/génétique , Prolifération cellulaire , Souris de lignée C57BL , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Mâle , Exons/génétique , Neurones/métabolisme , Ribonuclease III
13.
Epigenetics ; 19(1): 2368995, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38900099

RÉSUMÉ

Machado-Joseph disease (MJD) is an autosomal dominant spinocerebellar ataxia (SCA) caused by a polyglutamine expansion in the ataxin-3 protein, which initiates a cascade of pathogenic events, including transcriptional dysregulation. Genotype-phenotype correlations in MJD are incomplete, suggesting an influence of additional factors, such as epigenetic modifications, underlying the MJD pathogenesis. DNA methylation is known to impact the pathophysiology of neurodegenerative disorders through gene expression regulation and increased methylation has been reported for other SCAs. In this work we aimed to analyse global methylation in MJD carriers. Global 5-mC levels were quantified in blood samples of 33 MJD mutation carriers (patients and preclinical subjects) and 33 healthy controls, matched by age, sex, and smoking status. For a subset of 16 MJD subjects, a pilot follow-up analysis with two time points was also conducted. No differences were found in median global 5-mC levels between MJD mutation carriers and controls and no correlations between methylation levels and clinical or genetic variables were detected. Also, no alterations in global 5-mC levels were observed over time. Our findings do not support an increase in global blood methylation levels associated with MJD.


Sujet(s)
Méthylation de l'ADN , Hétérozygote , Maladie de Machado-Joseph , Mutation , Humains , Maladie de Machado-Joseph/génétique , Maladie de Machado-Joseph/sang , Mâle , Femelle , Adulte , Adulte d'âge moyen , Études cas-témoins , Ataxine-3/génétique , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/sang , Sujet âgé , Épigenèse génétique
14.
J Clin Immunol ; 44(6): 133, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38780872

RÉSUMÉ

PURPOSE: A large proportion of Common variable immunodeficiency (CVID) patients has duodenal inflammation with increased intraepithelial lymphocytes (IEL) of unknown aetiology. The histologic similarities to celiac disease, lead to confusion regarding treatment (gluten-free diet) of these patients. We aimed to elucidate the role of epigenetic DNA methylation in the aetiology of duodenal inflammation in CVID and differentiate it from true celiac disease. METHODS: DNA was isolated from snap-frozen pieces of duodenal biopsies and analysed for differences in genome-wide epigenetic DNA methylation between CVID patients with increased IEL (CVID_IEL; n = 5) without IEL (CVID_N; n = 3), celiac disease (n = 3) and healthy controls (n = 3). RESULTS: The DNA methylation data of 5-methylcytosine in CpG sites separated CVID and celiac diseases from healthy controls. Differential methylation in promoters of genes were identified as potential novel mediators in CVID and celiac disease. There was limited overlap of methylation associated genes between CVID_IEL and Celiac disease. High frequency of differentially methylated CpG sites was detected in over 100 genes nearby transcription start site (TSS) in both CVID_IEL and celiac disease, compared to healthy controls. Differential methylation of genes involved in regulation of TNF/cytokine production were enriched in CVID_IEL, compared to healthy controls. CONCLUSION: This is the first study to reveal a role of epigenetic DNA methylation in the etiology of duodenal inflammation of CVID patients, distinguishing CVID_IEL from celiac disease. We identified potential biomarkers and therapeutic targets within gene promotors and in high-frequency differentially methylated CpG regions proximal to TSS in both CVID_IEL and celiac disease.


Sujet(s)
Maladie coeliaque , Déficit immunitaire commun variable , Ilots CpG , Méthylation de l'ADN , Duodénum , Épigenèse génétique , Humains , Déficit immunitaire commun variable/génétique , Duodénum/métabolisme , Duodénum/anatomopathologie , Maladie coeliaque/génétique , Femelle , Mâle , Adulte , Adulte d'âge moyen , Ilots CpG/génétique , Régions promotrices (génétique)/génétique , Lymphocytes intra-épithéliaux/immunologie , Jeune adulte , Étude d'association pangénomique , 5-Méthyl-cytosine/métabolisme
15.
Genes (Basel) ; 15(5)2024 04 24.
Article de Anglais | MEDLINE | ID: mdl-38790164

RÉSUMÉ

Cell-free DNA (cfDNA) has recently emerged as a promising minimally invasive diagnostic biomarker for various cancers. In this study, our aim was to identify cfDNA biomarkers by investigating genes that displayed significant differences between glioma patients and their corresponding controls. To accomplish this, we utilized publicly available data from the Gene Expression Omnibus, focusing on 5-hydroxymethylcytosine (5hmC) profiles in both cfDNA and genomic DNA (gDNA) from glioma patients and healthy individuals. The intersection of gene lists derived from these comparative analyses unveiled LRIG1 and ZNF703 as the two genes with elevated 5hmC levels in both the cfDNA of glioma patients and gDNA of glioma tissue compared to their respective controls. The gene expression data revealed both genes were upregulated in glioma tissue compared to normal brain tissue. Integration of 5hmC data revealed a strong positive correlation in the glioma tissue group between 5hmC and the gene expression of the LRIG1 gene. Furthermore, exploration using the AmiCa web tool indicated that LRIG1 gene expression was elevated compared to 17 other cancers included in the database, emphasizing its potential as a distinctive biomarker across multiple cancer types.


Sujet(s)
5-Méthyl-cytosine , Marqueurs biologiques tumoraux , Tumeurs du cerveau , Acides nucléiques acellulaires , Gliome , Glycoprotéines membranaires , Humains , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , Gliome/génétique , Gliome/métabolisme , Gliome/anatomopathologie , Acides nucléiques acellulaires/génétique , Marqueurs biologiques tumoraux/génétique , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Régulation de l'expression des gènes tumoraux , Méthylation de l'ADN
16.
J Biochem Mol Toxicol ; 38(6): e23749, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38800929

RÉSUMÉ

Colon adenocarcinoma (COAD) is a common and fatal malignant tumor of digestive system with complex etiology. 5-Methylcytosine (m5C) modification of RNA by the NSUN gene family (NSUN1-NSUN7) and DNMT2 reshape cell biology and regulate tumor development. However, the expression profile, prognostic significance and function of these m5C modifiers in COAD remain largely unclear. By mining multiple integrated tumor databases, we found that NSUN1, NSUN2, NSUN5, and NSUN6 were overexpressed in COAD tumor samples relative to normal samples. Clinically, high expression of NSUN6 was significantly associated with shorter survival (including both disease-free survival and overall survival) in COAD patients. NSUN6 was further confirmed to be upregulated at both tissue and cellular levels of COAD, suggesting that NSUN6 plays a critical role in disease progression. Through comprehensive gene enrichment analysis and cell-based functional validation, it was revealed that NSUN6 promoted the cell cycle progression and cell proliferation of COAD. Mechanistically, NSUN6 upregulates the expression of oncogenic METTL3 and catalyzes its m5C modification in COAD cells. Overexpression of METTL3 significantly relieved the cell cycle inhibition of COAD caused by NSUN6 deficiency. Furthermore, NSUN6 was negatively associated with the abundance of infiltrating immune cells in COAD tumors, such as activated B cells, natural killer cells, effector memory CD8 T cells, and regulatory T cells. Importantly, pan-cancer analysis further uncovered that NSUN6 was dysregulated and heterogeneous in various tumors. Thus our findings extend the role of m5C transferase in COAD and suggest that NSUN6 is a potential biomarker and target for this malignancy.


Sujet(s)
5-Méthyl-cytosine , Adénocarcinome , Tumeurs du côlon , Évolution de la maladie , Methyltransferases , Humains , Methyltransferases/métabolisme , Methyltransferases/génétique , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/génétique , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Adénocarcinome/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux
17.
Ann Diagn Pathol ; 72: 152332, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38776734

RÉSUMÉ

Tubulin ß-3 staining pattern and staining intensity of 5-hydroxymethyl cytosine (5-hmC) are potential diagnostic and prognostic markers in melanocytic lesions that need further evaluation. Melanocytic nevi and primary cutaneous melanomas were immunohistochemically stained for tubulin-ß-3 and 5-hmC. Immunoreactivity and staining patterns were correlated with Breslow-thickness, clinical and pathological characteristics, and progression-free survival. Melanocytes showed positive tubulin ß-3 staining. However, in most nevi, tubulin ß-3 staining appeared as a gradient with intense cytoplasmic staining in cells of the superficial part of the lesion that faded to weak staining in the deep dermal part, while no gradient was found in deep penetrating nevi and melanomas. In 53 % of the melanomas, areas with loss of tubulin ß-3 staining were found. 5-hmC staining intensity was significantly higher in melanocytic nevi compared to melanomas. Breslow thickness in combination with low 5-hmC score and loss of tubulin-ß-3 staining was predictive for poor prognosis. As single markers, tubulin-ß-3 and 5-hmC can be useful to distinguish between melanocytic nevi and melanoma, but staining variability limits the use of 5-hmC. In melanomas measuring >1.5 mm, combination of low 5-hmC score and loss of tubulin-ß-3 staining may have prognostic value.


Sujet(s)
5-Méthyl-cytosine , Marqueurs biologiques tumoraux , Mélanome , Tumeurs cutanées , Tubuline , Humains , Mélanome/diagnostic , Mélanome/métabolisme , Mélanome/anatomopathologie , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Pronostic , Mâle , Femelle , Tubuline/métabolisme , Tubuline/analyse , Tumeurs cutanées/diagnostic , Tumeurs cutanées/anatomopathologie , Tumeurs cutanées/métabolisme , Adulte d'âge moyen , 5-Méthyl-cytosine/analogues et dérivés , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analyse , Sujet âgé , Adulte , Immunohistochimie/méthodes , Naevus pigmentaire/diagnostic , Naevus pigmentaire/anatomopathologie , Naevus pigmentaire/métabolisme , , Sujet âgé de 80 ans ou plus , Mélanocytes/anatomopathologie , Mélanocytes/métabolisme
18.
Front Immunol ; 15: 1362159, 2024.
Article de Anglais | MEDLINE | ID: mdl-38807595

RÉSUMÉ

RNA 5-methylcytosine (m5C) methylation plays a crucial role in hepatocellular carcinoma (HCC). As reported, aberrant m5C methylation is closely associated with the progression, therapeutic efficacy, and prognosis of HCC. The innate immune system functions as the primary defense mechanism in the body against pathogenic infections and tumors since it can activate innate immune pathways through pattern recognition receptors to exert anti-infection and anti-tumor effects. Recently, m5C methylation has been demonstrated to affect the activation of innate immune pathways including TLR, cGAS-STING, and RIG-I pathways by modulating RNA function, unveiling new mechanisms underlying the regulation of innate immune responses by tumor cells. However, research on m5C methylation and its interplay with innate immune pathways is still in its infancy. Therefore, this review details the biological significance of RNA m5C methylation in HCC and discusses its potential regulatory relationship with TLR, cGAS-STING, and RIG-I pathways, thereby providing fresh insights into the role of RNA methylation in the innate immune mechanisms and treatment of HCC.


Sujet(s)
Carcinome hépatocellulaire , Immunité innée , Tumeurs du foie , , Animaux , Humains , 5-Méthyl-cytosine/métabolisme , Carcinome hépatocellulaire/immunologie , Carcinome hépatocellulaire/génétique , Tumeurs du foie/immunologie , Tumeurs du foie/génétique , Protéines membranaires/génétique , Protéines membranaires/immunologie , Nucleotidyltransferases/génétique , Nucleotidyltransferases/métabolisme , ARN/génétique , Transduction du signal/immunologie , /immunologie
19.
Nat Commun ; 15(1): 3899, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38724548

RÉSUMÉ

The epitranscriptome embodies many new and largely unexplored functions of RNA. A significant roadblock hindering progress in epitranscriptomics is the identification of more than one modification in individual transcript molecules. We address this with CHEUI (CH3 (methylation) Estimation Using Ionic current). CHEUI predicts N6-methyladenosine (m6A) and 5-methylcytosine (m5C) in individual molecules from the same sample, the stoichiometry at transcript reference sites, and differential methylation between any two conditions. CHEUI processes observed and expected nanopore direct RNA sequencing signals to achieve high single-molecule, transcript-site, and stoichiometry accuracies in multiple tests using synthetic RNA standards and cell line data. CHEUI's capability to identify two modification types in the same sample reveals a co-occurrence of m6A and m5C in individual mRNAs in cell line and tissue transcriptomes. CHEUI provides new avenues to discover and study the function of the epitranscriptome.


Sujet(s)
5-Méthyl-cytosine , Adénosine , Analyse de séquence d'ARN , Transcriptome , Adénosine/analogues et dérivés , Adénosine/métabolisme , 5-Méthyl-cytosine/métabolisme , 5-Méthyl-cytosine/analogues et dérivés , Humains , Méthylation , Analyse de séquence d'ARN/méthodes , Maturation post-transcriptionnelle des ARN , ARN messager/métabolisme , ARN messager/génétique , ARN/métabolisme , ARN/génétique
20.
Front Immunol ; 15: 1380697, 2024.
Article de Anglais | MEDLINE | ID: mdl-38715608

RÉSUMÉ

The Corona Virus Disease (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), has quickly spread worldwide and resulted in significant morbidity and mortality. Although most infections are mild, some patients can also develop severe and fatal myocarditis. In eukaryotic RNAs, 5-methylcytosine (m5C) is a common kind of post-transcriptional modification, which is involved in regulating various biological processes (such as RNA export, translation, and stability maintenance). With the rapid development of m5C modification detection technology, studies related to viral m5C modification are ever-increasing. These studies have revealed that m5C modification plays an important role in various stages of viral replication, including transcription and translation. According to recent studies, m5C methylation modification can regulate SARS-CoV-2 infection by modulating innate immune signaling pathways. However, the specific role of m5C modification in SARS-CoV-2-induced myocarditis remains unclear. Therefore, this review aims to provide insights into the molecular mechanisms of m5C methylation in SARS-CoV-2 infection. Moreover, the regulatory role of NSUN2 in viral infection and host innate immune response was also highlighted. This review may provide new directions for developing therapeutic strategies for SARS-CoV-2-associated myocarditis.


Sujet(s)
COVID-19 , Myocardite , SARS-CoV-2 , Myocardite/virologie , Myocardite/immunologie , Myocardite/thérapie , Myocardite/génétique , Humains , COVID-19/immunologie , COVID-19/génétique , COVID-19/thérapie , SARS-CoV-2/physiologie , Méthylation , 5-Méthyl-cytosine/métabolisme , Immunité innée , Traitements médicamenteux de la COVID-19 , Animaux , ARN viral/génétique , ARN viral/métabolisme , Maturation post-transcriptionnelle des ARN
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...