Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 948
Filter
1.
Skin Res Technol ; 30(7): e13842, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38965799

ABSTRACT

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.


Subject(s)
Adenosine , Melanoma , RNA, Long Noncoding , Skin Neoplasms , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma/mortality , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/mortality , Adenosine/analogs & derivatives , Adenosine/metabolism , Prognosis , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Melanoma, Cutaneous Malignant , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Nomograms
2.
Epigenetics ; 19(1): 2374979, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38970823

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Cerebral Cortex , DNA Methylation , Proto-Oncogene Proteins , Animals , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Cerebral Cortex/metabolism , Mice, Knockout , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , CpG Islands , Mutation
3.
Mol Cancer ; 23(1): 139, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970106

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Cell Cycle Proteins , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins , RNA, Messenger , Radiation Tolerance , Uterine Cervical Neoplasms , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/radiotherapy , Uterine Cervical Neoplasms/pathology , Humans , Female , Radiation Tolerance/genetics , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Cell Line, Tumor , Prognosis , Xenograft Model Antitumor Assays , Methyltransferases/genetics , Methyltransferases/metabolism
4.
Biomolecules ; 14(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38927065

ABSTRACT

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.


Subject(s)
5-Methylcytosine , DNA Methylation , Smoking , Humans , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Male , Female , Smoking/genetics , Smoking/adverse effects , Middle Aged , Aged , Cohort Studies , Genome-Wide Association Study , Epigenesis, Genetic , CpG Islands/genetics , Adult
5.
Genome Biol ; 25(1): 142, 2024 06 03.
Article in English | MEDLINE | ID: mdl-38825692

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Enhancer Elements, Genetic , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Humans , Neural Networks, Computer , Gene Expression Regulation , Epigenesis, Genetic , DNA Methylation
6.
Open Biol ; 14(6): 230449, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38862018

ABSTRACT

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.


Subject(s)
5-Methylcytosine , DNA Methylation , Epigenesis, Genetic , Neural Networks, Computer , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/chemistry , 5-Methylcytosine/metabolism , Nanopore Sequencing/methods , Nanopores , Humans , Markov Chains , DNA/chemistry , DNA/genetics
7.
Cell Death Dis ; 15(6): 387, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824145

ABSTRACT

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.


Subject(s)
Adipose Tissue , Cell Differentiation , Epigenesis, Genetic , Mesenchymal Stem Cells , Mitochondria , Obesity , Humans , Mesenchymal Stem Cells/metabolism , Obesity/metabolism , Obesity/genetics , Obesity/pathology , Mitochondria/metabolism , Adipose Tissue/metabolism , Cell Differentiation/genetics , Female , Male , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Adult , Middle Aged , Cell Proliferation
8.
Medicine (Baltimore) ; 103(25): e38623, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905403

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Prognosis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Biomarkers, Tumor/genetics , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Male , Female , Gene Expression Regulation, Neoplastic , Methylation , Middle Aged
9.
Anal Chem ; 96(24): 9984-9993, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38833588

ABSTRACT

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.


Subject(s)
Cobalt , DNA , Epigenesis, Genetic , Metal-Organic Frameworks , Phenylenediamines , Metal-Organic Frameworks/chemistry , Cobalt/chemistry , DNA/chemistry , Phenylenediamines/chemistry , 5-Methylcytosine/chemistry , 5-Methylcytosine/analysis , 5-Methylcytosine/analogs & derivatives , Cytosine/chemistry , Cytosine/analogs & derivatives , Limit of Detection
10.
Nat Commun ; 15(1): 5136, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879605

ABSTRACT

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.


Subject(s)
Methyl-CpG-Binding Protein 2 , MicroRNAs , Neural Stem Cells , Neurogenesis , RNA, Long Noncoding , MicroRNAs/metabolism , MicroRNAs/genetics , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Neurogenesis/genetics , Animals , Mice , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Brain/metabolism , Brain/embryology , Humans , Cell Differentiation , DNA Methylation , Polypyrimidine Tract-Binding Protein/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Cell Proliferation , Mice, Inbred C57BL , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Male , Exons/genetics , Neurons/metabolism , Ribonuclease III
11.
Genes (Basel) ; 15(5)2024 04 24.
Article in English | MEDLINE | ID: mdl-38790164

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Biomarkers, Tumor , Brain Neoplasms , Cell-Free Nucleic Acids , Glioma , Membrane Glycoproteins , Humans , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Cell-Free Nucleic Acids/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Gene Expression Regulation, Neoplastic , DNA Methylation
12.
J Biochem Mol Toxicol ; 38(6): e23749, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38800929

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Adenocarcinoma , Colonic Neoplasms , Disease Progression , Methyltransferases , Humans , Methyltransferases/metabolism , Methyltransferases/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
13.
Ann Diagn Pathol ; 72: 152332, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38776734

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Biomarkers, Tumor , Melanoma , Skin Neoplasms , Tubulin , Humans , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/pathology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Prognosis , Male , Female , Tubulin/metabolism , Tubulin/analysis , Skin Neoplasms/diagnosis , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Middle Aged , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , 5-Methylcytosine/analysis , Aged , Adult , Immunohistochemistry/methods , Nevus, Pigmented/diagnosis , Nevus, Pigmented/pathology , Nevus, Pigmented/metabolism , Melanoma, Cutaneous Malignant , Aged, 80 and over , Melanocytes/pathology , Melanocytes/metabolism
14.
Nat Commun ; 15(1): 3899, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724548

ABSTRACT

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.


Subject(s)
5-Methylcytosine , Adenosine , Sequence Analysis, RNA , Transcriptome , Adenosine/analogs & derivatives , Adenosine/metabolism , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , Humans , Methylation , Sequence Analysis, RNA/methods , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA/metabolism , RNA/genetics
15.
Biosci Rep ; 44(5)2024 May 29.
Article in English | MEDLINE | ID: mdl-38743016

ABSTRACT

Varicose vein disease (VVD) is a common health problem worldwide. Microfibril-associated protein 5 (MFAP5) is one of the potential key players in its pathogenesis. Our previous microarray analysis revealed the cg06256735 and cg15815843 loci in the regulatory regions of the MFAP5 gene as hypomethylated in varicose veins which correlated with its up-regulation. The aim of this work was to validate preliminary microarray data, estimate the level of 5-hydroxymethylcytosine (5hmC) at these loci, and determine the methylation status of one of them in different layers of the venous wall. For this, methyl- and hydroxymethyl-sensitive restriction techniques were used followed by real-time PCR and droplet digital PCR, correspondingly, as well as bisulfite pyrosequencing of +/- oxidized DNA. Our microarray data on hypomethylation at the cg06256735 and cg15815843 loci in whole varicose vein segments were confirmed and it was also demonstrated that the level of 5hmC at these loci is increased in VVD. Specifically, among other layers of the venous wall, tunica (t.) intima is the main contributor to hypomethylation at the cg06256735 locus in varicose veins. Thus, it was shown that hypomethylation at the cg06256735 and cg15815843 loci takes place in VVD, with evidence to suggest that it happens through their active demethylation leading to up-regulation of the MFAP5 gene, and t. intima is most involved in this biochemical process.


Subject(s)
5-Methylcytosine , DNA Methylation , Varicose Veins , Varicose Veins/genetics , Varicose Veins/metabolism , Humans , Male , Female , Middle Aged , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Adult , Aged , Regulatory Sequences, Nucleic Acid/genetics , Genetic Loci
16.
Gastric Cancer ; 27(4): 735-746, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38584223

ABSTRACT

BACKGROUND: 5-Hydroxymethylcytosine-enriched gene profiles and regions show tissue-specific and tumor specific. There is a potential value to explore cell-free DNA 5-hydroxymethylcytosine feature biomarkers for early gastric cancer detection. METHODS: A matched case‒control study design with 50 gastric cancer patients and 50 controls was performed to sequence the different 5-hydroxymethylcytosine modification features of cell free DNA. Significantly differential 5-hydroxymethylcytosine modification genes were identified to construct a gastric cancer diagnostic model. Data set from GEO was used as an external testing set to test the robustness of the diagnostic model. RESULTS: Accounting for more than 90% of 5-hydroxymethylcytosine peaks were distributed in the gene body in both the gastric cancer and control groups. The diagnostic model was developed based on five different 5-hydroxymethylcytosine modification genes, FBXL7, PDE3A, TPO, SNTG2 and STXBP5. The model could effectively distinguish gastric cancer patients from controls in the training (AUC = 0.95, sensitivity = 88.6%, specificity = 94.3%), validation (AUC = 0.87, sensitivity = 73.3%, specificity = 93.3%) and testing (AUC = 0.90, sensitivity = 81.9%, specificity = 90.2%) sets. The risk scores of the controls from the model were significantly lower than those of gastric cancer patients in both our own data (P < 0.001) and GEO external testing data (P < 0.001), and no significant difference between different TNM stage patients (P = 0.09 and 0.66). Furthermore, there was no significant difference between the healthy control and benign gastric disease patients in the testing set from GEO (P = 0.10). CONCLUSIONS: The characteristics of 5-hydroxymethylcytosine in cell free DNA are specific to gastric cancer patients, and the diagnostic model constructed by five genes' 5-hydroxymethylcytosine features could effectively identify gastric cancer patients.


Subject(s)
5-Methylcytosine , Biomarkers, Tumor , Cell-Free Nucleic Acids , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/pathology , 5-Methylcytosine/analogs & derivatives , Biomarkers, Tumor/genetics , Male , Case-Control Studies , Female , Middle Aged , Cell-Free Nucleic Acids/genetics , Aged , Early Detection of Cancer/methods , DNA Methylation
17.
Cells ; 13(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38667328

ABSTRACT

Immune checkpoint inhibitors (ICIs) drastically improve therapeutic outcomes for lung cancer, but accurately predicting individual patient responses to ICIs remains a challenge. We performed the genome-wide profiling of 5-hydroxymethylcytosine (5hmC) in 85 plasma cell-free DNA (cfDNA) samples from lung cancer patients and developed a 5hmC signature that was significantly associated with progression-free survival (PFS). We built a 5hmC predictive model to quantify the 5hmC level and validated the model in the validation, test, and control sets. Low weighted predictive scores (wp-scores) were significantly associated with a longer PFS compared to high wp-scores in the validation [median 7.6 versus 1.8 months; p = 0.0012; hazard ratio (HR) 0.12; 95% confidence interval (CI), 0.03-0.54] and test (median 14.9 versus 3.3 months; p = 0.00074; HR 0.10; 95% CI, 0.02-0.50) sets. Objective response rates in patients with a low or high wp-score were 75.0% (95% CI, 42.8-94.5%) versus 0.0% (95% CI, 0.0-60.2%) in the validation set (p = 0.019) and 80.0% (95% CI, 44.4-97.5%) versus 0.0% (95% CI, 0.0-36.9%) in the test set (p = 0.0011). The wp-scores were also significantly associated with PFS in patients receiving single-agent ICI treatment (p < 0.05). In addition, the 5hmC predictive signature demonstrated superior predictive capability to tumor programmed death-ligand 1 and specificity to ICI treatment response prediction. Moreover, we identified novel 5hmC-associated genes and signaling pathways integral to ICI treatment response in lung cancer. This study provides proof-of-concept evidence that the cfDNA 5hmC signature is a robust biomarker for predicting ICI treatment response in lung cancer.


Subject(s)
5-Methylcytosine , 5-Methylcytosine/analogs & derivatives , Cell-Free Nucleic Acids , Immunotherapy , Lung Neoplasms , Humans , 5-Methylcytosine/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/blood , Male , Female , Immunotherapy/methods , Aged , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Immune Checkpoint Inhibitors/therapeutic use , Treatment Outcome
18.
Sci Rep ; 14(1): 9116, 2024 04 20.
Article in English | MEDLINE | ID: mdl-38643305

ABSTRACT

RNA modifications are pivotal in the development of newly synthesized structures, showcasing a vast array of alterations across various RNA classes. Among these, 5-hydroxymethylcytosine (5HMC) stands out, playing a crucial role in gene regulation and epigenetic changes, yet its detection through conventional methods proves cumbersome and costly. To address this, we propose Deep5HMC, a robust learning model leveraging machine learning algorithms and discriminative feature extraction techniques for accurate 5HMC sample identification. Our approach integrates seven feature extraction methods and various machine learning algorithms, including Random Forest, Naive Bayes, Decision Tree, and Support Vector Machine. Through K-fold cross-validation, our model achieved a notable 84.07% accuracy rate, surpassing previous models by 7.59%, signifying its potential in early cancer and cardiovascular disease diagnosis. This study underscores the promise of Deep5HMC in offering insights for improved medical assessment and treatment protocols, marking a significant advancement in RNA modification analysis.


Subject(s)
5-Methylcytosine/analogs & derivatives , Algorithms , Neural Networks, Computer , Bayes Theorem , Support Vector Machine , RNA
19.
Dev Cell ; 59(8): 1010-1027.e8, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38569549

ABSTRACT

Ten-eleven translocation (TET) enzymes iteratively oxidize 5-methylcytosine (5mC) to generate 5-hydroxymethylcytosine (5hmC), 5-formylcytosine, and 5-carboxylcytosine to facilitate active genome demethylation. Whether these bases are required to promote replication-coupled dilution or activate base excision repair during mammalian germline reprogramming remains unresolved due to the inability to decouple TET activities. Here, we generated two mouse lines expressing catalytically inactive TET1 (Tet1-HxD) and TET1 that stalls oxidation at 5hmC (Tet1-V). Tet1 knockout and catalytic mutant primordial germ cells (PGCs) fail to erase methylation at select imprinting control regions and promoters of meiosis-associated genes, validating the requirement for the iterative oxidation of 5mC for complete germline reprogramming. TET1V and TET1HxD rescue most hypermethylation of Tet1-/- sperm, suggesting the role of TET1 beyond its oxidative capability. We additionally identify a broader class of hypermethylated regions in Tet1 mutant mouse sperm that depend on TET oxidation for reprogramming. Our study demonstrates the link between TET1-mediated germline reprogramming and sperm methylome patterning.


Subject(s)
5-Methylcytosine , 5-Methylcytosine/analogs & derivatives , DNA Methylation , DNA-Binding Proteins , Genomic Imprinting , Oxidation-Reduction , Proto-Oncogene Proteins , Spermatozoa , Animals , Male , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Spermatozoa/metabolism , 5-Methylcytosine/metabolism , Cellular Reprogramming/genetics , Mice, Knockout , Mice, Inbred C57BL
20.
Proc Natl Acad Sci U S A ; 121(14): e2321611121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38547058

ABSTRACT

Malignant glioma exhibits immune evasion characterized by highly expressing the immune checkpoint CD47. RNA 5-methylcytosine(m5C) modification plays a pivotal role in tumor pathogenesis. However, the mechanism underlying m5C-modified RNA metabolism remains unclear, as does the contribution of m5C-modified RNA to the glioma immune microenvironment. In this study, we demonstrate that the canonical 28SrRNA methyltransferase NSUN5 down-regulates ß-catenin by promoting the degradation of its mRNA, leading to enhanced phagocytosis of tumor-associated macrophages (TAMs). Specifically, the NSUN5-induced suppression of ß-catenin relies on its methyltransferase activity mediated by cysteine 359 (C359) and is not influenced by its localization in the nucleolus. Intriguingly, NSUN5 directly interacts with and deposits m5C on CTNNB1 caRNA (chromatin-associated RNA). NSUN5-induced recruitment of TET2 to chromatin is independent of its methyltransferase activity. The m5C modification on caRNA is subsequently oxidized into 5-hydroxymethylcytosine (5hmC) by TET2, which is dependent on its binding affinity for Fe2+ and α-KG. Furthermore, NSUN5 enhances the chromatin recruitment of RBFOX2 which acts as a 5hmC-specific reader to recognize and facilitate the degradation of 5hmC caRNA. Notably, hmeRIP-seq analysis reveals numerous mRNA substrates of NSUN5 that potentially undergo this mode of metabolism. In addition, NSUN5 is epigenetically suppressed by DNA methylation and is negatively correlated with IDH1-R132H mutation in glioma patients. Importantly, pharmacological blockage of DNA methylation or IDH1-R132H mutant and CD47/SIRPα signaling synergistically enhances TAM-based phagocytosis and glioma elimination in vivo. Our findings unveil a general mechanism by which NSUN5/TET2/RBFOX2 signaling regulates RNA metabolism and highlight NSUN5 targeting as a potential strategy for glioma immune therapy.


Subject(s)
5-Methylcytosine , 5-Methylcytosine/analogs & derivatives , DNA-Binding Proteins , Dioxygenases , Glioma , Muscle Proteins , Humans , 5-Methylcytosine/metabolism , beta Catenin/metabolism , Chromatin , CD47 Antigen/genetics , RNA , Immune Evasion , Glioma/pathology , RNA, Messenger/metabolism , Methyltransferases/metabolism , RNA, Small Nuclear , Tumor Microenvironment , RNA Splicing Factors/genetics , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...