Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65.607
Filter
1.
Zhonghua Bing Li Xue Za Zhi ; 53(7): 702-708, 2024 Jul 08.
Article in Chinese | MEDLINE | ID: mdl-38955702

ABSTRACT

Objective: To investigate the clinicopathological characteristics and prognostic factors of sporadic mismatch repair deficient (dMMR) colorectal cancer. Methods: A total of 120 cases of sporadic dMMR colorectal cancer from July 2015 to April 2021 were retrospectively collected in Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. Patients with Lynch syndrome; synchronous multiple colorectal cancers; preoperative anti-tumor treatments such as chemotherapy and radiotherapy; and those with incomplete follow-up information were excluded based on family history and next-generation sequencing (NGS) test results. Immunohistochemical stains were used to detect the expression of mismatch repair proteins, methylation-specific PCR for methylation testing, and fluorescent PCR for BRAF V600E gene mutation detection. The clinical and pathological data, and gene mutation status were analyzed. Follow-up was done to assess survival and prognosis including progression-free survival and overall survival rate. Results: Sporadic dMMR colorectal cancer occurred more frequently in the right side of the colon, in females, and in the elderly. Morphologically, it was mostly moderately-differentiated, and most patients had low-grade tumor budding. In terms of immunohistochemical expression, MLH1 and PMS2 loss were dominant, and there were age and location-specificities in protein expression. MLH1 methylation was commonly detected in elderly female patients and rare in young male patients; while MLH1 and PMS2 deficiency, and BRAF V600E mutation occurred more often on the right side (P<0.05). The 3-year and 5-year progression-free survival rates were 90.7% and 88.7% respectively, and the 3-year and 5-year overall survival rates were 92.8% and 90.7% respectively. Tumor budding status was an independent risk factor affecting patient recurrence (hazard ratio=3.375, 95% confidence interval: 1.060-10.741, P=0.039), patients with low-grade tumor budding had better prognosis, and those with medium or high-grade tumor budding had poor prognosis. Conclusion: For dMMR colorectal cancer patients, tumor budding status is an independent risk factor for recurrence.


Subject(s)
Colorectal Neoplasms , DNA Mismatch Repair , Proto-Oncogene Proteins B-raf , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Male , Female , Prognosis , Retrospective Studies , Proto-Oncogene Proteins B-raf/genetics , MutL Protein Homolog 1/genetics , MutL Protein Homolog 1/metabolism , Mutation , Survival Rate , Middle Aged , Aged , DNA Methylation , Adult , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Colorectal Neoplasms, Hereditary Nonpolyposis/metabolism
2.
Oncol Nurs Forum ; 51(4): 349-360, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38950092

ABSTRACT

OBJECTIVES: To determine associations among DNA methylation of brain-derived neurotrophic factor (BDNF) and RAS p21 protein activator 2 (RASA2) genes with processing speed and perceived cognitive function. SAMPLE & SETTING: This was a cross-sectional, secondary analysis of baseline data from a randomized controlled trial, the Exercise Program in Cancer and Cognition Study. METHODS & VARIABLES: Data included M values for DNA methylation of the BDNF and RASA2 genes; processing speed, objectively measured using the Grooved Pegboard and Digit Vigilance Test scores; and perceived cognitive function, self-reported using the Patient Assessment of Own Functioning Inventory. Regression analysis was conducted. RESULTS: Greater methylation of cg21291635 of the BDNF gene (p = 0.01) and cg20247102 of the RASA2 gene (p = 0.013) were associated with poorer processing speed, whereas greater methylation of cg20108357 of the BDNF gene (p < 0.001) and cg00567892 of the RASA2 gene (p = 0.019) were associated with better perceived cognitive function. IMPLICATIONS FOR NURSING: Gene methylation variations were demonstrated, suggesting the genes' potential roles and two possible distinct mechanisms of cognitive function in cancer. .


Subject(s)
Brain-Derived Neurotrophic Factor , Breast Neoplasms , Cognition , DNA Methylation , Postmenopause , Humans , Brain-Derived Neurotrophic Factor/genetics , Female , Middle Aged , Cross-Sectional Studies , Breast Neoplasms/genetics , Breast Neoplasms/psychology , Aged , Postmenopause/psychology , Postmenopause/genetics
3.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 46(3): 329-333, 2024 Jun.
Article in Chinese | MEDLINE | ID: mdl-38953256

ABSTRACT

Objective To evaluate the value of SOX1 and PAX1 gene methylation detection in the secondary triage of high-grade cervical lesions.Methods Exfoliated cervical cells were collected from 122 patients tested positive for human papilloma virus (HPV) and subjected to thin-prep cytologic test (TCT) and SOX1/PAX1 gene methylation tests.Results The HPV test combined with TCT showed the sensitivity of 95.24% and the specificity of 23.75% for detecting cervical intraepithelial neoplasia (CIN) grade 2 and above (CIN2+).After the addition of the SOX1/PAX1 gene methylation detection in secondary triage,the sensitivity for detecting CIN2+ was 83.33%,which had no statistically significant difference from the sensitivity of TCT combined with HPV test (P=0.078).However,the specificity reached 77.50%,which was significantly higher than that of HPV test combined with TCT (P<0.001).The SOX1/PAX1 gene methylation level in the CIN2+ group was higher than those in the normal cervical tissue and the CIN1 group(P<0.001).The cut-off values of SOX1 and PAX1 gene methylation for CIN2+ detection were -11.81 and -11.98,respectively.Conclusion Adding the detection of SOX1/PAX1 gene methylation in secondary triage significantly improves the efficiency and accuracy of CIN2+ detection.


Subject(s)
DNA Methylation , Paired Box Transcription Factors , SOXB1 Transcription Factors , Uterine Cervical Dysplasia , Uterine Cervical Neoplasms , Humans , Female , Paired Box Transcription Factors/genetics , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/diagnosis , Uterine Cervical Dysplasia/virology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/virology , SOXB1 Transcription Factors/genetics , Adult , Middle Aged , Sensitivity and Specificity , Young Adult
4.
Addict Biol ; 29(7): e13422, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38949208

ABSTRACT

Opioid use disorder (OUD) is a multifaceted condition influenced by sex, genetic and environmental factors that could be linked with epigenetic changes. Understanding how these factors interact is crucial to understand and address the development and progression of this disorder. Our aim was to elucidate different potential epigenetic and genetic mechanisms between women and men that correlate with OUD under real-world pain unit conditions. Associations between analgesic response and the DNA methylation level of the opioid mu receptor (OPRM1) gene (CpG sites 1-5 selected in the promoter region) were evaluated in 345 long opioid-treated chronic non cancer pain: cases with OUD (n = 67) and controls (without OUD, n = 278). Cases showed younger ages, low employment status and quality of life, but higher morphine equivalent daily dose and psychotropic use, compared to the controls. The patients with OUD showed a significant decrease in OPRM1 DNA methylation, which correlated with clinical outcomes like pain relief, depression and different adverse events. Significant differences were found at the five CpG sites studied for men, and exclusively in women for CpG site 3, in relation to OUD diagnosis. These findings support the importance of epigenetics and sex as biological variables to be considered toward efficient OUD understanding and therapy development.


Subject(s)
Chronic Pain , DNA Methylation , Epigenesis, Genetic , Opioid-Related Disorders , Receptors, Opioid, mu , Humans , Receptors, Opioid, mu/genetics , DNA Methylation/genetics , Male , Female , Chronic Pain/genetics , Chronic Pain/drug therapy , Opioid-Related Disorders/genetics , Middle Aged , Adult , Sex Factors , Analgesics, Opioid/therapeutic use , Case-Control Studies , CpG Islands/genetics , Quality of Life
5.
Adv Protein Chem Struct Biol ; 141: 223-253, 2024.
Article in English | MEDLINE | ID: mdl-38960475

ABSTRACT

Recent advances in genome-wide studies have revealed numerous epigenetic regulations brought about by genes involved in cellular metabolism. Isocitrate dehydrogenase (IDH), an essential enzyme, that converts isocitrate into -ketoglutarate (KG) predominantly in the tricarboxylic acid (TCA) cycle, has gained particular importance due to its cardinal role in the metabolic pathway in cells. IDH1, IDH2, and IDH3 are the three isomeric IDH enzymes that have been shown to regulate cellular metabolism. Of particular importance, IDH2 genes are associated with several cancers, including gliomas, oligodendroglioma, and astrocytomas. These mutations lead to the production of oncometabolite D-2-hydroxyglutarate (D-2-HG), which accumulates in cells promoting tumor growth. The enhanced levels of D-2-HG competitively inhibit α-KG dependent enzymes, inhibiting cell TCA cycle, upregulating the cell growth and survival relevant HIF-1α pathway, promoting DNA hypermethylation related epigenetic activity, all of which synergistically contribute to carcinogenesis. The present review discusses epigenetic mechanisms inIDH2 regulation in cells and further its clinical implications.


Subject(s)
Epigenesis, Genetic , Isocitrate Dehydrogenase , Neoplasms , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Humans , Neoplasms/genetics , Neoplasms/metabolism , DNA Methylation
6.
J Biochem Mol Toxicol ; 38(7): e23764, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963172

ABSTRACT

Obesity is an established risk factor for numerous malignancies, although it remains uncertain whether the disease itself or weight-loss drugs are responsible for a greater predisposition to cancer. The objective of the current study was to determine the impact of dulaglutide on genetic and epigenetic DNA damage caused by obesity, which is a crucial factor in the development of cancer. Mice were administered a low-fat or high-fat diet for 12 weeks, followed by a 5-week treatment with dulaglutide. Following that, modifications of the DNA bases were examined using the comet assay. To clarify the underlying molecular mechanisms, oxidized and methylated DNA bases, changes in the redox status, levels of inflammatory cytokines, and the expression levels of some DNA repair genes were evaluated. Animals fed a high-fat diet exhibited increased body weights, elevated DNA damage, oxidation of DNA bases, and DNA hypermethylation. In addition, obese mice showed altered inflammatory responses, redox imbalances, and repair gene expressions. The findings demonstrated that dulaglutide does not exhibit genotoxicity in the investigated conditions. Following dulaglutide administration, animals fed a high-fat diet demonstrated low DNA damage, less oxidation and methylation of DNA bases, restored redox balance, and improved inflammatory responses. In addition, dulaglutide treatment restored the upregulated DNMT1, Ogg1, and p53 gene expression. Overall, dulaglutide effectively maintains DNA integrity in obese animals. It reduces oxidative DNA damage and hypermethylation by restoring redox balance, modulating inflammatory responses, and recovering altered gene expressions. These findings demonstrate dulaglutide's expediency in treating obesity and its associated complications.


Subject(s)
DNA Damage , DNA Methylation , DNA Repair , Diet, High-Fat , Glucagon-Like Peptides , Immunoglobulin Fc Fragments , Oxidation-Reduction , Recombinant Fusion Proteins , Animals , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , DNA Methylation/drug effects , Immunoglobulin Fc Fragments/pharmacology , DNA Damage/drug effects , Mice , DNA Repair/drug effects , Diet, High-Fat/adverse effects , Recombinant Fusion Proteins/pharmacology , Male , Oxidation-Reduction/drug effects , Inflammation/metabolism , Inflammation/genetics , Oxidative Stress/drug effects , Obesity/metabolism , Obesity/drug therapy , Obesity/genetics , Gene Expression Regulation/drug effects , Mice, Inbred C57BL
7.
J Transl Med ; 22(1): 608, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956589

ABSTRACT

BACKGROUND: Urothelial carcinoma (UC) is the second most common urological malignancy. Despite numerous molecular markers have been evaluated during the past decades, no urothelial markers for diagnosis and recurrence monitoring have shown consistent clinical utility. METHODS: The methylation level of tissue samples from public database and clinical collected were analyzed. Patients with UC and benign diseases of the urinary system (BUD) were enrolled to establish TAGMe (TAG of Methylation) assessment in a training cohort (n = 567) using restriction enzyme-based bisulfite-free qPCR. The performance of TAGMe assessment was further verified in the validation cohort (n = 198). Urine samples from 57 UC patients undergoing postoperative surveillance were collected monthly for six months after surgery to assess the TAGMe methylation. RESULTS: We identified TAGMe as a potentially novel Universal-Cancer-Only Methylation (UCOM) marker was hypermethylated in multi-type cancers and investigated its application in UC. Restriction enzyme-based bisulfite-free qPCR was used for detection, and the results of which were consistent with gold standard pyrosequencing. Importantly, hypermethylated TAGMe showed excellent sensitivity of 88.9% (95% CI: 81.4-94.1%) and specificity of 90.0% (95% CI: 81.9-95.3%) in efficiently distinguishing UC from BUD patients in urine and also performed well in different clinical scenarios of UC. Moreover, the abnormality of TAGMe as an indicator of recurrence might precede clinical recurrence by three months to one year, which provided an invaluable time window for timely and effective intervention to prevent UC upstaging. CONCLUSION: TAGMe assessment based on a novel single target in urine is effective and easy to perform in UC diagnosis and recurrence monitoring, which may reduce the burden of cystoscopy. Trial registration ChiCTR2100052507. Registered on 30 October 2021.


Subject(s)
Biomarkers, Tumor , DNA Methylation , Neoplasm Recurrence, Local , Humans , DNA Methylation/genetics , Male , Female , Biomarkers, Tumor/urine , Biomarkers, Tumor/genetics , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/diagnosis , Aged , Urothelium/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/urine , Cohort Studies , Urologic Neoplasms/genetics , Urologic Neoplasms/diagnosis , Urologic Neoplasms/urine , Reproducibility of Results , Membrane Proteins , Neoplasm Proteins
8.
Hum Genomics ; 18(1): 75, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956648

ABSTRACT

BACKGROUND: Aging represents a significant risk factor for the occurrence of cerebral small vessel disease, associated with white matter (WM) lesions, and to age-related cognitive alterations, though the precise mechanisms remain largely unknown. This study aimed to investigate the impact of polygenic risk scores (PRS) for WM integrity, together with age-related DNA methylation, and gene expression alterations, on cognitive aging in a cross-sectional healthy aging cohort. The PRSs were calculated using genome-wide association study (GWAS) summary statistics for magnetic resonance imaging (MRI) markers of WM integrity, including WM hyperintensities, fractional anisotropy (FA), and mean diffusivity (MD). These scores were utilized to predict age-related cognitive changes and evaluate their correlation with structural brain changes, which distinguish individuals with higher and lower cognitive scores. To reduce the dimensionality of the data and identify age-related DNA methylation and transcriptomic alterations, Sparse Partial Least Squares-Discriminant Analysis (sPLS-DA) was used. Subsequently, a canonical correlation algorithm was used to integrate the three types of omics data (PRS, DNA methylation, and gene expression data) and identify an individual "omics" signature that distinguishes subjects with varying cognitive profiles. RESULTS: We found a positive association between MD-PRS and long-term memory, as well as a correlation between MD-PRS and structural brain changes, effectively discriminating between individuals with lower and higher memory scores. Furthermore, we observed an enrichment of polygenic signals in genes related to both vascular and non-vascular factors. Age-related alterations in DNA methylation and gene expression indicated dysregulation of critical molecular features and signaling pathways involved in aging and lifespan regulation. The integration of multi-omics data underscored the involvement of synaptic dysfunction, axonal degeneration, microtubule organization, and glycosylation in the process of cognitive aging. CONCLUSIONS: These findings provide valuable insights into the biological mechanisms underlying the association between WM coherence and cognitive aging. Additionally, they highlight how age-associated DNA methylation and gene expression changes contribute to cognitive aging.


Subject(s)
Cognitive Aging , DNA Methylation , Genome-Wide Association Study , Multifactorial Inheritance , Humans , DNA Methylation/genetics , Female , Male , Multifactorial Inheritance/genetics , Aged , Middle Aged , Cross-Sectional Studies , White Matter/diagnostic imaging , White Matter/pathology , Risk Factors , Magnetic Resonance Imaging , Aging/genetics , Aging/pathology , Brain/diagnostic imaging , Brain/metabolism , Brain/pathology , Genetic Risk Score
9.
J Transl Med ; 22(1): 618, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961476

ABSTRACT

BACKGROUND: Cell free DNA (cfDNA)-based assays hold great potential in detecting early cancer signals yet determining the tissue-of-origin (TOO) for cancer signals remains a challenging task. Here, we investigated the contribution of a methylation atlas to TOO detection in low depth cfDNA samples. METHODS: We constructed a tumor-specific methylation atlas (TSMA) using whole-genome bisulfite sequencing (WGBS) data from five types of tumor tissues (breast, colorectal, gastric, liver and lung cancer) and paired white blood cells (WBC). TSMA was used with a non-negative least square matrix factorization (NNLS) deconvolution algorithm to identify the abundance of tumor tissue types in a WGBS sample. We showed that TSMA worked well with tumor tissue but struggled with cfDNA samples due to the overwhelming amount of WBC-derived DNA. To construct a model for TOO, we adopted the multi-modal strategy and used as inputs the combination of deconvolution scores from TSMA with other features of cfDNA. RESULTS: Our final model comprised of a graph convolutional neural network using deconvolution scores and genome-wide methylation density features, which achieved an accuracy of 69% in a held-out validation dataset of 239 low-depth cfDNA samples. CONCLUSIONS: In conclusion, we have demonstrated that our TSMA in combination with other cfDNA features can improve TOO detection in low-depth cfDNA samples.


Subject(s)
DNA Methylation , Genome, Human , Neoplasms , Neural Networks, Computer , Humans , DNA Methylation/genetics , Neoplasms/genetics , Neoplasms/blood , Neoplasms/diagnosis , Cell-Free Nucleic Acids/blood , Cell-Free Nucleic Acids/genetics , Organ Specificity/genetics , Algorithms
10.
Genome Biol ; 25(1): 175, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961490

ABSTRACT

BACKGROUND: Transposable elements play a critical role in maintaining genome architecture during neurodevelopment. Short Interspersed Nuclear Elements (SINEs), a major subtype of transposable elements, are known to harbor binding sites for the CCCTC-binding factor (CTCF) and pivotal in orchestrating chromatin organization. However, the regulatory mechanisms controlling the activity of SINEs in the developing brain remains elusive. RESULTS: In our study, we conduct a comprehensive genome-wide epigenetic analysis in mouse neural precursor cells using ATAC-seq, ChIP-seq, whole genome bisulfite sequencing, in situ Hi-C, and RNA-seq. Our findings reveal that the SET domain bifurcated histone lysine methyltransferase 1 (SETDB1)-mediated H3K9me3, in conjunction with DNA methylation, restricts chromatin accessibility on a selective subset of SINEs in neural precursor cells. Mechanistically, loss of Setdb1 increases CTCF access to these SINE elements and contributes to chromatin loop reorganization. Moreover, de novo loop formation contributes to differential gene expression, including the dysregulation of genes enriched in mitotic pathways. This leads to the disruptions of cell proliferation in the embryonic brain after genetic ablation of Setdb1 both in vitro and in vivo. CONCLUSIONS: In summary, our study sheds light on the epigenetic regulation of SINEs in mouse neural precursor cells, suggesting their role in maintaining chromatin organization and cell proliferation during neurodevelopment.


Subject(s)
Chromatin , Histone-Lysine N-Methyltransferase , Neural Stem Cells , Short Interspersed Nucleotide Elements , Animals , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Mice , Chromatin/metabolism , DNA Methylation , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Epigenesis, Genetic , Histones/metabolism , Brain/metabolism , Brain/cytology
11.
Cancer Med ; 13(13): e7470, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38963018

ABSTRACT

INTRODUCTION: Identifying reliable biomarkers that reflect cancer survivorship symptoms remains a challenge for researchers. DNA methylation (DNAm) measurements reflecting epigenetic changes caused by anti-cancer therapy may provide needed insights. Given lack of consensus describing utilization of DNAm data to predict survivorship issues, a review evaluating the current landscape is warranted. OBJECTIVE: Provide an overview of current studies examining associations of DNAm with survivorship burdens in cancer survivors. METHODS: A literature review was conducted including studies if they focused on cohorts of cancer survivors, utilized peripheral blood cell DNAm data, and evaluated the associations of DNAm and survivorship issues. RESULTS: A total of 22 studies were identified, with majority focused on breast (n = 7) or childhood cancer (n = 9) survivors, and half studies included less than 100 patients (n = 11). Survivorship issues evaluated included those related to neurocognition (n = 5), psychiatric health (n = 3), general wellness (n = 9), chronic conditions (n = 5), and treatment specific toxicities (n = 4). Studies evaluated epigenetic age metrics (n = 10) and DNAm levels at individual CpG sites or regions (n = 12) for their associations with survivorship issues in cancer survivors along with relevant confounding factors. Significant associations of measured DNAm in the peripheral blood samples of cancer survivors and survivorship issues were identified. DISCUSSION/CONCLUSION: Studies utilizing epigenetic age metrics and differential methylation analysis demonstrated significant associations of DNAm measurements with survivorship burdens. Associations were observed encompassing diverse survivorship outcomes and timeframes relative to anti-cancer therapy initiation. These findings underscore the potential of these measurements as useful biomarkers in survivorship care and research.


Subject(s)
Cancer Survivors , DNA Methylation , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/blood , Epigenesis, Genetic , Survivorship , Biomarkers, Tumor/genetics , Female
12.
BMC Plant Biol ; 24(1): 631, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38965467

ABSTRACT

BACKGROUND: DNA methylation contributes to the epigenetic regulation of nuclear gene expression, and is associated with plant growth, development, and stress responses. Compelling evidence has emerged that long non-coding RNA (lncRNA) regulates DNA methylation. Previous genetic and physiological evidence indicates that lncRNA-CRIR1 plays a positive role in the responses of cassava plants to cold stress. However, it is unclear whether global DNA methylation changes with CRIR1-promoted cold tolerance. RESULTS: In this study, a comprehensive comparative analysis of DNA methylation and transcriptome profiles was performed to reveal the gene expression and epigenetic dynamics after CRIR1 overexpression. Compared with the wild-type plants, CRIR1-overexpressing plants present gained DNA methylation in over 37,000 genomic regions and lost DNA methylation in about 16,000 genomic regions, indicating a global decrease in DNA methylation after CRIR1 overexpression. Declining DNA methylation is not correlated with decreased/increased expression of the DNA methylase/demethylase genes, but is associated with increased transcripts of a few transcription factors, chlorophyll metabolism and photosynthesis-related genes, which could contribute to the CRIR1-promoted cold tolerance. CONCLUSIONS: In summary, a first set of transcriptome and epigenome data was integrated in this study to reveal the gene expression and epigenetic dynamics after CRIR1 overexpression, with the identification of several TFs, chlorophyll metabolism and photosynthesis-related genes that may be involved in CRIR1-promoted cold tolerance. Therefore, our study has provided valuable data for the systematic study of molecular insights for plant cold stress response.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Plant , Transcriptome , RNA, Long Noncoding/genetics , Epigenome , Cold-Shock Response/genetics , Cold Temperature
13.
BMC Biol ; 22(1): 147, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38965555

ABSTRACT

BACKGROUND: Physical activity is well known for its multiple health benefits and although the knowledge of the underlying molecular mechanisms is increasing, our understanding of the role of epigenetics in long-term training adaptation remains incomplete. In this intervention study, we included individuals with a history of > 15 years of regular endurance or resistance training compared to age-matched untrained controls performing endurance or resistance exercise. We examined skeletal muscle DNA methylation of genes involved in key adaptation processes, including myogenesis, gene regulation, angiogenesis and metabolism. RESULTS: A greater number of differentially methylated regions and differentially expressed genes were identified when comparing the endurance group with the control group than in the comparison between the strength group and the control group at baseline. Although the cellular composition of skeletal muscle samples was generally consistent across groups, variations were observed in the distribution of muscle fiber types. Slow-twitch fiber type genes MYH7 and MYL3 exhibited lower promoter methylation and elevated expression in endurance-trained athletes, while the same group showed higher methylation in transcription factors such as FOXO3, CREB5, and PGC-1α. The baseline DNA methylation state of those genes was associated with the transcriptional response to an acute bout of exercise. Acute exercise altered very few of the investigated CpG sites. CONCLUSIONS: Endurance- compared to resistance-trained athletes and untrained individuals demonstrated a different DNA methylation signature of selected skeletal muscle genes, which may influence transcriptional dynamics following a bout of acute exercise. Skeletal muscle fiber type distribution is associated with methylation of fiber type specific genes. Our results suggest that the baseline DNA methylation landscape in skeletal muscle influences the transcription of regulatory genes in response to an acute exercise bout.


Subject(s)
DNA Methylation , Exercise , Muscle, Skeletal , Resistance Training , Humans , Male , Exercise/physiology , Adult , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Epigenesis, Genetic , Physical Endurance/genetics
14.
Cell Mol Life Sci ; 81(1): 284, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967794

ABSTRACT

Hepatocellular carcinoma (HCC) is a malignancy that occurs worldwide and is generally associated with poor prognosis. The development of resistance to targeted therapies such as sorafenib is a major challenge in clinical cancer treatment. In the present study, Ten-eleven translocation protein 1 (TET1) was found to be highly expressed in sorafenib-resistant HCC cells and knockdown of TET1 can substantially improve the therapeutic effect of sorafenib on HCC, indicating the potential important roles of TET1 in sorafenib resistance in HCC. Mechanistic studies determined that TET1 and Yes-associated protein 1 (YAP1) synergistically regulate the promoter methylation and gene expression of DNA repair-related genes in sorafenib-resistant HCC cells. RNA sequencing indicated the activation of DNA damage repair signaling was extensively suppressed by the TET1 inhibitor Bobcat339. We also identified TET1 as a direct transcriptional target of YAP1 by promoter analysis and chromatin-immunoprecipitation assays in sorafenib-resistant HCC cells. Furthermore, we showed that Bobcat339 can overcome sorafenib resistance and synergized with sorafenib to induce tumor eradication in HCC cells and mouse models. Finally, immunostaining showed a positive correlation between TET1 and YAP1 in clinical samples. Our findings have identified a previously unrecognized molecular pathway underlying HCC sorafenib resistance, thus revealing a promising strategy for cancer therapy.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinoma, Hepatocellular , DNA Repair , Drug Resistance, Neoplasm , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Liver Neoplasms , Proto-Oncogene Proteins , Sorafenib , Transcription Factors , YAP-Signaling Proteins , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic/drug effects , Animals , DNA Repair/drug effects , DNA Repair/genetics , YAP-Signaling Proteins/metabolism , Mice , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice, Nude , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Hippo Signaling Pathway , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Mice, Inbred BALB C , DNA Methylation/drug effects
15.
Nat Commun ; 15(1): 5580, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961062

ABSTRACT

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.


Subject(s)
5-Methylcytosine , CpG Islands , DNA Methylation , Nanopore Sequencing , 5-Methylcytosine/metabolism , 5-Methylcytosine/chemistry , Nanopore Sequencing/methods , Animals , Mice , Humans , CpG Islands/genetics , Deep Learning , Algorithms , Sequence Analysis, DNA/methods , Whole Genome Sequencing/methods , Sulfites/chemistry
16.
Epigenetics ; 19(1): 2375022, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38967555

ABSTRACT

Infinium Methylation BeadChip arrays remain one of the most popular platforms for epigenome-wide association studies, but tools for downstream pathway analysis have their limitations. Functional class scoring (FCS) is a group of pathway enrichment techniques that involve the ranking of genes and evaluation of their collective regulation in biological systems, but the implementations described for Infinium methylation array data do not retain direction information, which is important for mechanistic understanding of genomic regulation. Here, we evaluate several candidate FCS methods that retain directional information. According to simulation results, the best-performing method involves the mean aggregation of probe limma t-statistics by gene followed by a rank-ANOVA enrichment test using the mitch package. This method, which we call 'LAM,' outperformed an existing over-representation analysis method in simulations, and showed higher sensitivity and robustness in an analysis of real lung tumour-normal paired datasets. Using matched RNA-seq data, we examine the relationship of methylation differences at promoters and gene bodies with RNA expression at the level of pathways in lung cancer. To demonstrate the utility of our approach, we apply it to three other contexts where public data were available. First, we examine the differential pathway methylation associated with chronological age. Second, we investigate pathway methylation differences in infants conceived with in vitro fertilization. Lastly, we analyse differential pathway methylation in 19 disease states, identifying hundreds of novel associations. These results show LAM is a powerful method for the detection of differential pathway methylation complementing existing methods. A reproducible vignette is provided to illustrate how to implement this method.


Subject(s)
DNA Methylation , Lung Neoplasms , Humans , Lung Neoplasms/genetics , Promoter Regions, Genetic , Female , Genome-Wide Association Study/methods , Epigenesis, Genetic
17.
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
18.
Nat Commun ; 15(1): 5693, 2024 Jul 07.
Article in English | MEDLINE | ID: mdl-38972954

ABSTRACT

Leukemias with ambiguous lineage comprise several loosely defined entities, often without a clear mechanistic basis. Here, we extensively profile the epigenome and transcriptome of a subgroup of such leukemias with CpG Island Methylator Phenotype. These leukemias exhibit comparable hybrid myeloid/lymphoid epigenetic landscapes, yet heterogeneous genetic alterations, suggesting they are defined by their shared epigenetic profile rather than common genetic lesions. Gene expression enrichment reveals similarity with early T-cell precursor acute lymphoblastic leukemia and a lymphoid progenitor cell of origin. In line with this, integration of differential DNA methylation and gene expression shows widespread silencing of myeloid transcription factors. Moreover, binding sites for hematopoietic transcription factors, including CEBPA, SPI1 and LEF1, are uniquely inaccessible in these leukemias. Hypermethylation also results in loss of CTCF binding, accompanied by changes in chromatin interactions involving key transcription factors. In conclusion, epigenetic dysregulation, and not genetic lesions, explains the mixed phenotype of this group of leukemias with ambiguous lineage. The data collected here constitute a useful and comprehensive epigenomic reference for subsequent studies of acute myeloid leukemias, T-cell acute lymphoblastic leukemias and mixed-phenotype leukemias.


Subject(s)
CpG Islands , DNA Methylation , Epigenesis, Genetic , Gene Regulatory Networks , Humans , DNA Methylation/genetics , CpG Islands/genetics , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , CCCTC-Binding Factor/metabolism , CCCTC-Binding Factor/genetics , Gene Expression Regulation, Leukemic , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin/metabolism , Chromatin/genetics , Male , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Female , Hematopoiesis/genetics , Child , Transcriptome , Proto-Oncogene Proteins , Trans-Activators
19.
Cell Mol Biol Lett ; 29(1): 99, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38978023

ABSTRACT

Skeletal muscular atrophy is a complex disease involving a large number of gene expression regulatory networks and various biological processes. Despite extensive research on this topic, its underlying mechanisms remain elusive, and effective therapeutic approaches are yet to be established. Recent studies have shown that epigenetics play an important role in regulating skeletal muscle atrophy, influencing the expression of numerous genes associated with this condition through the addition or removal of certain chemical modifications at the molecular level. This review article comprehensively summarizes the different types of modifications to DNA, histones, RNA, and their known regulators. We also discuss how epigenetic modifications change during the process of skeletal muscle atrophy, the molecular mechanisms by which epigenetic regulatory proteins control skeletal muscle atrophy, and assess their translational potential. The role of epigenetics on muscle stem cells is also highlighted. In addition, we propose that alternative splicing interacts with epigenetic mechanisms to regulate skeletal muscle mass, offering a novel perspective that enhances our understanding of epigenetic inheritance's role and the regulatory network governing skeletal muscle atrophy. Collectively, advancements in the understanding of epigenetic mechanisms provide invaluable insights into the study of skeletal muscle atrophy. Moreover, this knowledge paves the way for identifying new avenues for the development of more effective therapeutic strategies and pharmaceutical interventions.


Subject(s)
Epigenesis, Genetic , Muscle, Skeletal , Muscular Atrophy , Humans , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Animals , Histones/metabolism , Histones/genetics , DNA Methylation/genetics , Alternative Splicing/genetics
20.
Neurosciences (Riyadh) ; 29(3): 168-176, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38981632

ABSTRACT

OBJECTIVES: To elucidate the relationship between DNA methylation profiling (DMP) and pathological diagnosis (PD) in pediatric glial and glioneuronal tumors with B-Raf proto-oncogene, serine/threonine kinase (BRAF) mutations, addressing their diagnostic challenges. METHODS: This retrospective study, conducted in Saudi Arabia, analyzed 47 cases from the Children's Brain Tumor Network online database using scanned images, next-generation sequencing data, and methylation profiles processed using the Heidelberg methylation brain tumor classifiers v12.5 and v12.8. The data was last access on 10 November 2023. RESULTS: The highest prevalence of BRAF mutations was observed in pilocytic astrocytoma and ganglioglioma. The DMP was consistent with PD in 23 cases, but discrepancies emerged in others, including diagnostic changes in diffuse leptomeningeal glioneuronal tumor and polymorphous low-grade neuroepithelial tumor of the young. A key inconsistency appeared between a pilocytic astrocytoma MC and a glioneuronal tumor PD. Two high-grade astrocytomas were misclassified as pleomorphic xanthoastrocytomas. Additionally, low variant allelic frequency in gangliogliomas likely contributed to misclassifications as control in 5 cases. CONCLUSION: This study emphasized the importance of integrating DMP with PD in diagnosing pediatric glial and glioneuronal tumors with BRAF mutations. Although DMP offers significant diagnostic insights, its limitations, particularly in cases with low tumor content, necessitate cautious interpretation, as well as its use as a complementary diagnostic tool, rather than a definitive method.


Subject(s)
Brain Neoplasms , DNA Methylation , Mutation , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/diagnostic imaging , Child , Male , Female , DNA Methylation/genetics , Retrospective Studies , Child, Preschool , Ganglioglioma/genetics , Ganglioglioma/pathology , Ganglioglioma/diagnostic imaging , Adolescent , Glioma/genetics , Glioma/pathology , Glioma/diagnosis , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/diagnostic imaging , Astrocytoma/diagnosis , Infant , Saudi Arabia
SELECTION OF CITATIONS
SEARCH DETAIL
...