Clinical Epigenetics最新文献

筛选
英文 中文
DNMT1/miR-152-3p/SOS1 signaling axis promotes self-renewal and tumor growth of cancer stem-like cells derived from non-small cell lung cancer DNMT1/miR-152-3p/SOS1 信号轴促进非小细胞肺癌干细胞的自我更新和肿瘤生长
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-15 DOI: 10.1186/s13148-024-01663-5
Qing Yuan, Rubo Wang, Xiang Li, Fei Sun, Jiazhi Lin, Zhimin Fu, Jiansong Zhang
{"title":"DNMT1/miR-152-3p/SOS1 signaling axis promotes self-renewal and tumor growth of cancer stem-like cells derived from non-small cell lung cancer","authors":"Qing Yuan, Rubo Wang, Xiang Li, Fei Sun, Jiazhi Lin, Zhimin Fu, Jiansong Zhang","doi":"10.1186/s13148-024-01663-5","DOIUrl":"https://doi.org/10.1186/s13148-024-01663-5","url":null,"abstract":"CSLCs(Cancer stem cell-like cells), which are central to tumorigenesis, are intrinsically influenced by epigenetic modifications. This study aimed to elucidate the underlying mechanism involving the DNMT1/miR-152-3p/SOS1 axis in regulating the self-renewal and tumor growth of LCSLCs (lung cancer stem-like cells). Target genes of miR-152-3p were predicted using TargetScan Human 8.0. Self-renewal and tumor growth of LCSLC were compared in suspension-cultured non-small cell lung cancer (NSCLC) cell lines H460 and A549 cell-derived globe cells. Functional effects of the DNMT1/miR-152-3p/SOS1 axis were assessed through gain-of-function experiments in vitro and in vivo. Additionally, luciferase reporter assays were employed to analyze the interaction among DNMT1, miR-152-3p, and SOS1. Our findings highlight a negative interaction between DNMT1 and miR-152-3p, resulting in reduced miR-152-3p level. This, in turn, leads to the alleviation of the inhibitory effect of miR-152-3p on the target gene SOS1, ultimately activating SOS1 and playing an essential role in self-renewal and tumor growth of LCSLC. However, the alteration of SOS1 does not affect DNMT1/miR-152-3p regulation. Therefore, it is reasonable to infer that the DNMT1/miR-152-3p negative feedback loop critically sustains self-renewal and tumor growth of LCSLC through SOS1. This study reveals a novel mechanism underpinning self-renewal and tumor growth of CSLC (cancer stem cell) in NSCLC and identifies potential therapeutic targets for NSCLC treatment.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140591585","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Polycomb repressive complex 2 and its core component EZH2: potential targeted therapeutic strategies for head and neck squamous cell carcinoma 多聚胞抑制复合体 2 及其核心成分 EZH2:头颈部鳞状细胞癌的潜在靶向治疗策略
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-10 DOI: 10.1186/s13148-024-01666-2
Yuxi Cheng, Zhengzheng Song, Xiaodan Fang, Zhangui Tang
{"title":"Polycomb repressive complex 2 and its core component EZH2: potential targeted therapeutic strategies for head and neck squamous cell carcinoma","authors":"Yuxi Cheng, Zhengzheng Song, Xiaodan Fang, Zhangui Tang","doi":"10.1186/s13148-024-01666-2","DOIUrl":"https://doi.org/10.1186/s13148-024-01666-2","url":null,"abstract":"The polycomb group (PcG) comprises a set of proteins that exert epigenetic regulatory effects and play crucial roles in diverse biological processes, ranging from pluripotency and development to carcinogenesis. Among these proteins, enhancer of zeste homolog 2 (EZH2) stands out as a catalytic component of polycomb repressive complex 2 (PRC2), which plays a role in regulating the expression of homologous (Hox) genes and initial stages of x chromosome inactivation. In numerous human cancers, including head and neck squamous cell carcinoma (HNSCC), EZH2 is frequently overexpressed or activated and has been identified as a negative prognostic factor. Notably, EZH2 emerges as a significant gene involved in regulating the STAT3/HOTAIR axis, influencing HNSCC proliferation, differentiation, and promoting metastasis by modulating related oncogenes in oral cancer. Currently, various small molecule compounds have been developed as inhibitors specifically targeting EZH2 and have gained approval for treating refractory tumors. In this review, we delve into the epigenetic regulation mediated by EZH2/PRC2 in HNSCC, with a specific focus on exploring the potential roles and mechanisms of EZH2, its crucial contribution to targeted drug therapy, and its association with cancer markers and epithelial–mesenchymal transition. Furthermore, we aim to unravel its potential as a therapeutic strategy for oral squamous cell carcinoma.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140591312","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Meta-analysis of epigenetic aging in schizophrenia reveals multifaceted relationships with age, sex, illness duration, and polygenic risk 对精神分裂症表观遗传老化的元分析揭示了与年龄、性别、病程和多基因风险的多方面关系
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-08 DOI: 10.1186/s13148-024-01660-8
Anil P. S. Ori, Loes M. Olde Loohuis, Jerry Guintivano, Eilis Hannon, Emma Dempster, David St. Clair, Nick J. Bass, Andrew McQuillin, Jonathan Mill, Patrick F. Sullivan, Rene S. Kahn, Steve Horvath, Roel A. Ophoff
{"title":"Meta-analysis of epigenetic aging in schizophrenia reveals multifaceted relationships with age, sex, illness duration, and polygenic risk","authors":"Anil P. S. Ori, Loes M. Olde Loohuis, Jerry Guintivano, Eilis Hannon, Emma Dempster, David St. Clair, Nick J. Bass, Andrew McQuillin, Jonathan Mill, Patrick F. Sullivan, Rene S. Kahn, Steve Horvath, Roel A. Ophoff","doi":"10.1186/s13148-024-01660-8","DOIUrl":"https://doi.org/10.1186/s13148-024-01660-8","url":null,"abstract":"The study of biological age acceleration may help identify at-risk individuals and reduce the rising global burden of age-related diseases. Using DNA methylation (DNAm) clocks, we investigated biological aging in schizophrenia (SCZ), a mental illness that is associated with an increased prevalence of age-related disabilities and morbidities. In a whole blood DNAm sample of 1090 SCZ cases and 1206 controls across four European cohorts, we performed a meta-analysis of differential aging using three DNAm clocks (i.e., Hannum, Horvath, and Levine). To dissect how DNAm aging contributes to SCZ, we integrated information on duration of illness and SCZ polygenic risk, as well as stratified our analyses by chronological age and biological sex. We found that blood-based DNAm aging is significantly altered in SCZ independent from duration of the illness since onset. We observed sex-specific and nonlinear age effects that differed between clocks and point to possible distinct age windows of altered aging in SCZ. Most notably, intrinsic cellular age (Horvath clock) is decelerated in SCZ cases in young adulthood, while phenotypic age (Levine clock) is accelerated in later adulthood compared to controls. Accelerated phenotypic aging was most pronounced in women with SCZ carrying a high polygenic burden with an age acceleration of + 3.82 years (CI 2.02–5.61, P = 1.1E−03). Phenotypic aging and SCZ polygenic risk contributed additively to the illness and together explained up to 14.38% of the variance in disease status. Our study contributes to the growing body of evidence of altered DNAm aging in SCZ and points to intrinsic age deceleration in younger adulthood and phenotypic age acceleration in later adulthood in SCZ. Since increased phenotypic age is associated with increased risk of all-cause mortality, our findings indicate that specific and identifiable patient groups are at increased mortality risk as measured by the Levine clock. Our study did not find that DNAm aging could be explained by the duration of illness of patients, but we did observe age- and sex-specific effects that warrant further investigation. Finally, our results show that combining genetic and epigenetic predictors can improve predictions of disease outcomes and may help with disease management in schizophrenia.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140591402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetics in diabetic cardiomyopathy 糖尿病心肌病的表观遗传学
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-05 DOI: 10.1186/s13148-024-01667-1
Xiaozhu Ma, Shuai Mei, Qidamugai Wuyun, Li Zhou, Dating Sun, Jiangtao Yan
{"title":"Epigenetics in diabetic cardiomyopathy","authors":"Xiaozhu Ma, Shuai Mei, Qidamugai Wuyun, Li Zhou, Dating Sun, Jiangtao Yan","doi":"10.1186/s13148-024-01667-1","DOIUrl":"https://doi.org/10.1186/s13148-024-01667-1","url":null,"abstract":"Diabetic cardiomyopathy (DCM) is a critical complication that poses a significant threat to the health of patients with diabetes. The intricate pathological mechanisms of DCM cause diastolic dysfunction, followed by impaired systolic function in the late stages. Accumulating researches have revealed the association between DCM and various epigenetic regulatory mechanisms, including DNA methylation, histone modifications, non-coding RNAs, and other epigenetic molecules. Recently, a profound understanding of epigenetics in the pathophysiology of DCM has been broadened owing to advanced high-throughput technologies, which assist in developing potential therapeutic strategies. In this review, we briefly introduce the epigenetics regulation and update the relevant progress in DCM. We propose the role of epigenetic factors and non-coding RNAs (ncRNAs) as potential biomarkers and drugs in DCM diagnosis and treatment, providing a new perspective and understanding of epigenomics in DCM.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140591306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting histone demethylases JMJD3 and UTX: selenium as a potential therapeutic agent for cervical cancer 以组蛋白去甲基化酶 JMJD3 和 UTX 为靶点:硒作为宫颈癌的潜在治疗剂
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-04 DOI: 10.1186/s13148-024-01665-3
Dezhi Chen, Bo Cai, Yingying Zhu, Yimin Ma, Xiaoting Yu, Jieqi Xiong, Jiaying Shen, Weiwei Tie, Yisheng Zhang, Fei Guo
{"title":"Targeting histone demethylases JMJD3 and UTX: selenium as a potential therapeutic agent for cervical cancer","authors":"Dezhi Chen, Bo Cai, Yingying Zhu, Yimin Ma, Xiaoting Yu, Jieqi Xiong, Jiaying Shen, Weiwei Tie, Yisheng Zhang, Fei Guo","doi":"10.1186/s13148-024-01665-3","DOIUrl":"https://doi.org/10.1186/s13148-024-01665-3","url":null,"abstract":"The intriguing connection between selenium and cancer resembles a captivating puzzle that keeps researchers engaged and curious. While selenium has shown promise in reducing cancer risks through supplementation, its interaction with epigenetics in cervical cancer remains a fascinating yet largely unexplored realm. Unraveling the intricacies of selenium's role and its interaction with epigenetic factors could unlock valuable insights in the battle against this complex disease. Selenium has shown remarkable inhibitory effects on cervical cancer cells in various ways. In in vitro studies, it effectively inhibits the proliferation, migration, and invasion of cervical cancer cells, while promoting apoptosis. Selenium also demonstrates significant inhibitory effects on human cervical cancer-derived organoids. Furthermore, in an in vivo study, the administration of selenium dioxide solution effectively suppresses the growth of cervical cancer tumors in mice. One of the mechanisms behind selenium's inhibitory effects is its ability to inhibit histone demethylases, specifically JMJD3 and UTX. This inhibition is observed both in vitro and in vivo. Notably, when JMJD3 and UTX are inhibited with GSK-J4, similar biological effects are observed in both in vitro and in vivo models, effectively inhibiting organoid models derived from cervical cancer patients. Inhibiting JMJD3 and UTX also induces G2/M phase arrest, promotes cellular apoptosis, and reverses epithelial-mesenchymal transition (EMT). ChIP-qPCR analysis confirms that JMJD3 and UTX inhibition increases the recruitment of a specific histone modification, H3K27me3, to the transcription start sites (TSS) of target genes in cervical cancer cells (HeLa and SiHa cells). Furthermore, the expressions of JMJD3 and UTX are found to be significantly higher in cervical cancer tissues compared to adjacent normal cervical tissues, suggesting their potential as therapeutic targets. Our study highlights the significant inhibitory effects of selenium on the growth, migration, and invasion of cervical cancer cells, promoting apoptosis and displaying promising potential as a therapeutic agent. We identified the histone demethylases JMJD3 and UTX as specific targets of selenium, and their inhibition replicates the observed effects on cancer cell behavior. These findings suggest that JMJD3 and UTX could be valuable targets for selenium-based treatments of cervical cancer.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140591497","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nucleosome reorganisation in breast cancer tissues. 乳腺癌组织中的核小体重组
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-04-01 DOI: 10.1186/s13148-024-01656-4
Divya R Jacob, Wilfried M Guiblet, Hulkar Mamayusupova, Mariya Shtumpf, Isabella Ciuta, Luminita Ruje, Svetlana Gretton, Milena Bikova, Clark Correa, Emily Dellow, Shivam P Agrawal, Navid Shafiei, Anastasija Drobysevskaja, Chris M Armstrong, Jonathan D G Lam, Yevhen Vainshtein, Christopher T Clarkson, Graeme J Thorn, Kai Sohn, Madapura M Pradeepa, Sankaran Chandrasekharan, Greg N Brooke, Elena Klenova, Victor B Zhurkin, Vladimir B Teif
{"title":"Nucleosome reorganisation in breast cancer tissues.","authors":"Divya R Jacob, Wilfried M Guiblet, Hulkar Mamayusupova, Mariya Shtumpf, Isabella Ciuta, Luminita Ruje, Svetlana Gretton, Milena Bikova, Clark Correa, Emily Dellow, Shivam P Agrawal, Navid Shafiei, Anastasija Drobysevskaja, Chris M Armstrong, Jonathan D G Lam, Yevhen Vainshtein, Christopher T Clarkson, Graeme J Thorn, Kai Sohn, Madapura M Pradeepa, Sankaran Chandrasekharan, Greg N Brooke, Elena Klenova, Victor B Zhurkin, Vladimir B Teif","doi":"10.1186/s13148-024-01656-4","DOIUrl":"10.1186/s13148-024-01656-4","url":null,"abstract":"<p><strong>Background: </strong>Nucleosome repositioning in cancer is believed to cause many changes in genome organisation and gene expression. Understanding these changes is important to elucidate fundamental aspects of cancer. It is also important for medical diagnostics based on cell-free DNA (cfDNA), which originates from genomic DNA regions protected from digestion by nucleosomes.</p><p><strong>Results: </strong>We have generated high-resolution nucleosome maps in paired tumour and normal tissues from the same breast cancer patients using MNase-assisted histone H3 ChIP-seq and compared them with the corresponding cfDNA from blood plasma. This analysis has detected single-nucleosome repositioning at key regulatory regions in a patient-specific manner and common cancer-specific patterns across patients. The nucleosomes gained in tumour versus normal tissue were particularly informative of cancer pathways, with ~ 20-fold enrichment at CpG islands, a large fraction of which marked promoters of genes encoding DNA-binding proteins. The tumour tissues were characterised by a 5-10 bp decrease in the average distance between nucleosomes (nucleosome repeat length, NRL), which is qualitatively similar to the differences between pluripotent and differentiated cells. This effect was correlated with gene activity, differential DNA methylation and changes in local occupancy of linker histone variants H1.4 and H1X.</p><p><strong>Conclusions: </strong>Our study offers a novel resource of high-resolution nucleosome maps in breast cancer patients and reports for the first time the effect of systematic decrease of NRL in paired tumour versus normal breast tissues from the same patient. Our findings provide a new mechanistic understanding of nucleosome repositioning in tumour tissues that can be valuable for patient diagnostics, stratification and monitoring.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10986098/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Refining risk prediction in pediatric acute lymphoblastic leukemia through DNA methylation profiling 通过DNA甲基化图谱完善小儿急性淋巴细胞白血病的风险预测
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-03-28 DOI: 10.1186/s13148-024-01662-6
Adrián Mosquera Orgueira, Olga Krali, Carlos Pérez Míguez, Andrés Peleteiro Raíndo, José Ángel Díaz Arias, Marta Sonia González Pérez, Manuel Mateo Pérez Encinas, Manuel Fernández Sanmartín, Daniel Sinnet, Mats Heyman, Gudmar Lönnerholm, Ulrika Norén-Nyström, Kjeld Schmiegelow, Jessica Nordlund
{"title":"Refining risk prediction in pediatric acute lymphoblastic leukemia through DNA methylation profiling","authors":"Adrián Mosquera Orgueira, Olga Krali, Carlos Pérez Míguez, Andrés Peleteiro Raíndo, José Ángel Díaz Arias, Marta Sonia González Pérez, Manuel Mateo Pérez Encinas, Manuel Fernández Sanmartín, Daniel Sinnet, Mats Heyman, Gudmar Lönnerholm, Ulrika Norén-Nyström, Kjeld Schmiegelow, Jessica Nordlund","doi":"10.1186/s13148-024-01662-6","DOIUrl":"https://doi.org/10.1186/s13148-024-01662-6","url":null,"abstract":"Acute lymphoblastic leukemia (ALL) is the most prevalent cancer in children, and despite considerable progress in treatment outcomes, relapses still pose significant risks of mortality and long-term complications. To address this challenge, we employed a supervised machine learning technique, specifically random survival forests, to predict the risk of relapse and mortality using array-based DNA methylation data from a cohort of 763 pediatric ALL patients treated in Nordic countries. The relapse risk predictor (RRP) was constructed based on 16 CpG sites, demonstrating c-indexes of 0.667 and 0.677 in the training and test sets, respectively. The mortality risk predictor (MRP), comprising 53 CpG sites, exhibited c-indexes of 0.751 and 0.754 in the training and test sets, respectively. To validate the prognostic value of the predictors, we further analyzed two independent cohorts of Canadian (n = 42) and Nordic (n = 384) ALL patients. The external validation confirmed our findings, with the RRP achieving a c-index of 0.667 in the Canadian cohort, and the RRP and MRP achieving c-indexes of 0.529 and 0.621, respectively, in an independent Nordic cohort. The precision of the RRP and MRP models improved when incorporating traditional risk group data, underscoring the potential for synergistic integration of clinical prognostic factors. The MRP model also enabled the definition of a risk group with high rates of relapse and mortality. Our results demonstrate the potential of DNA methylation as a prognostic factor and a tool to refine risk stratification in pediatric ALL. This may lead to personalized treatment strategies based on epigenetic profiling.","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140314270","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Low expression of miR-182 caused by DNA hypermethylation accelerates acute lymphocyte leukemia development by targeting PBX3 and BCL2: miR-182 promoter methylation is a predictive marker for hypomethylation agents + BCL2 inhibitor venetoclax. DNA高甲基化导致的miR-182低表达通过靶向PBX3和BCL2加速急性淋巴细胞白血病的发展:miR-182启动子甲基化是低甲基化药物+BCL2抑制剂venetoclax的预测标记。
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-03-26 DOI: 10.1186/s13148-024-01658-2
Danyang Li, Yigang Yuan, Chen Meng, Zihan Lin, Min Zhao, Liuzhi Shi, Min Li, Daijiao Ye, Yue Cai, Xiaofei He, Haige Ye, Shujuan Zhou, Haixia Zhou, Shenmeng Gao
{"title":"Low expression of miR-182 caused by DNA hypermethylation accelerates acute lymphocyte leukemia development by targeting PBX3 and BCL2: miR-182 promoter methylation is a predictive marker for hypomethylation agents + BCL2 inhibitor venetoclax.","authors":"Danyang Li, Yigang Yuan, Chen Meng, Zihan Lin, Min Zhao, Liuzhi Shi, Min Li, Daijiao Ye, Yue Cai, Xiaofei He, Haige Ye, Shujuan Zhou, Haixia Zhou, Shenmeng Gao","doi":"10.1186/s13148-024-01658-2","DOIUrl":"10.1186/s13148-024-01658-2","url":null,"abstract":"<p><strong>Background: </strong>miR-182 promoter hypermethylation frequently occurs in various tumors, including acute myeloid leukemia, and leads to low expression of miR-182. However, whether adult acute lymphocyte leukemia (ALL) cells have high miR-182 promoter methylation has not been determined.</p><p><strong>Methods: </strong>To assess the methylation status of the miR-182 promoter, methylation and unmethylation-specific PCR analysis, bisulfite-sequencing analysis, and MethylTarget™ assays were performed to measure the frequency of methylation at the miR-182 promoter. Bone marrow cells were isolated from miR-182 knockout (182KO) and 182 wild type (182WT) mice to construct BCR-ABL (P190) and Notch-induced murine B-ALL and T-ALL models, respectively. Primary ALL samples were performed to investigate synergistic effects of the hypomethylation agents (HMAs) and the BCL2 inhibitor venetoclax (Ven) in vitro.</p><p><strong>Results: </strong>miR-182 (miR-182-5P) expression was substantially lower in ALL blasts than in normal controls (NCs) because of DNA hypermethylation at the miR-182 promoter in ALL blasts but not in normal controls (NCs). Knockout of miR-182 (182KO) markedly accelerated ALL development, facilitated the infiltration, and shortened the OS in a BCR-ABL (P190)-induced murine B-ALL model. Furthermore, the 182KO ALL cell population was enriched with more leukemia-initiating cells (CD43<sup>+</sup>B220<sup>+</sup> cells, LICs) and presented higher leukemogenic activity than the 182WT ALL population. Furthermore, depletion of miR-182 reduced the OS in a Notch-induced murine T-ALL model, suggesting that miR-182 knockout accelerates ALL development. Mechanistically, overexpression of miR-182 inhibited proliferation and induced apoptosis by directly targeting PBX3 and BCL2, two well-known oncogenes, that are key targets of miR-182. Most importantly, DAC in combination with Ven had synergistic effects on ALL cells with miR-182 promoter hypermethylation, but not on ALL cells with miR-182 promoter hypomethylation.</p><p><strong>Conclusions: </strong>Collectively, we identified miR-182 as a tumor suppressor gene in ALL cells and low expression of miR-182 because of hypermethylation facilitates the malignant phenotype of ALL cells. DAC + Ven cotreatment might has been applied in the clinical try for ALL patients with miR-182 promoter hypermethylation. Furthermore, the methylation frequency at the miR-182 promoter should be a potential biomarker for DAC + Ven treatment in ALL patients.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-03-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10964616/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Head and neck cancer of unknown primary: unveiling primary tumor sites through machine learning on DNA methylation profiles. 原发灶不明的头颈部癌症:通过对 DNA 甲基化图谱的机器学习揭示原发肿瘤部位。
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-03-25 DOI: 10.1186/s13148-024-01657-3
Leonhard Stark, Atsuko Kasajima, Fabian Stögbauer, Benedikt Schmidl, Jakob Rinecker, Katharina Holzmann, Sarah Färber, Nicole Pfarr, Katja Steiger, Barbara Wollenberg, Jürgen Ruland, Christof Winter, Markus Wirth
{"title":"Head and neck cancer of unknown primary: unveiling primary tumor sites through machine learning on DNA methylation profiles.","authors":"Leonhard Stark, Atsuko Kasajima, Fabian Stögbauer, Benedikt Schmidl, Jakob Rinecker, Katharina Holzmann, Sarah Färber, Nicole Pfarr, Katja Steiger, Barbara Wollenberg, Jürgen Ruland, Christof Winter, Markus Wirth","doi":"10.1186/s13148-024-01657-3","DOIUrl":"10.1186/s13148-024-01657-3","url":null,"abstract":"<p><strong>Background: </strong>The unknown tissue of origin in head and neck cancer of unknown primary (hnCUP) leads to invasive diagnostic procedures and unspecific and potentially inefficient treatment options for patients. The most common histologic subtype, squamous cell carcinoma, can stem from various tumor primary sites, including the oral cavity, oropharynx, larynx, head and neck skin, lungs, and esophagus. DNA methylation profiles are highly tissue-specific and have been successfully used to classify tissue origin. We therefore developed a support vector machine (SVM) classifier trained with publicly available DNA methylation profiles of commonly cervically metastasizing squamous cell carcinomas (n = 1103) in order to identify the primary tissue of origin of our own cohort of squamous cell hnCUP patient's samples (n = 28). Methylation analysis was performed with Infinium MethylationEPIC v1.0 BeadChip by Illumina.</p><p><strong>Results: </strong>The SVM algorithm achieved the highest overall accuracy of tested classifiers, with 87%. Squamous cell hnCUP samples on DNA methylation level resembled squamous cell carcinomas commonly metastasizing into cervical lymph nodes. The most frequently predicted cancer localization was the oral cavity in 11 cases (39%), followed by the oropharynx and larynx (both 7, 25%), skin (2, 7%), and esophagus (1, 4%). These frequencies concord with the expected distribution of lymph node metastases in epidemiological studies.</p><p><strong>Conclusions: </strong>On DNA methylation level, hnCUP is comparable to primary tumor tissue cancer types that commonly metastasize to cervical lymph nodes. Our SVM-based classifier can accurately predict these cancers' tissues of origin and could significantly reduce the invasiveness of hnCUP diagnostics and enable a more precise therapy after clinical validation.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10964705/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287130","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic scores of blood-based proteins as biomarkers of general cognitive function and brain health. 作为一般认知功能和大脑健康生物标志物的血液蛋白质表观遗传评分。
IF 5.7 2区 医学
Clinical Epigenetics Pub Date : 2024-03-25 DOI: 10.1186/s13148-024-01661-7
Hannah M Smith, Joanna E Moodie, Karla Monterrubio-Gómez, Danni A Gadd, Robert F Hillary, Aleksandra D Chybowska, Daniel L McCartney, Archie Campbell, Paul Redmond, Danielle Page, Adele Taylor, Janie Corley, Sarah E Harris, Maria Valdés Hernández, Susana Muñoz Maniega, Mark E Bastin, Joanna M Wardlaw, Ian J Deary, James P Boardman, Donncha S Mullin, Tom C Russ, Simon R Cox, Riccardo E Marioni
{"title":"Epigenetic scores of blood-based proteins as biomarkers of general cognitive function and brain health.","authors":"Hannah M Smith, Joanna E Moodie, Karla Monterrubio-Gómez, Danni A Gadd, Robert F Hillary, Aleksandra D Chybowska, Daniel L McCartney, Archie Campbell, Paul Redmond, Danielle Page, Adele Taylor, Janie Corley, Sarah E Harris, Maria Valdés Hernández, Susana Muñoz Maniega, Mark E Bastin, Joanna M Wardlaw, Ian J Deary, James P Boardman, Donncha S Mullin, Tom C Russ, Simon R Cox, Riccardo E Marioni","doi":"10.1186/s13148-024-01661-7","DOIUrl":"10.1186/s13148-024-01661-7","url":null,"abstract":"<p><strong>Background: </strong>Epigenetic Scores (EpiScores) for blood protein levels have been associated with disease outcomes and measures of brain health, highlighting their potential usefulness as clinical biomarkers. They are typically derived via penalised regression, whereby a linear weighted sum of DNA methylation (DNAm) levels at CpG sites are predictive of protein levels. Here, we examine 84 previously published protein EpiScores as possible biomarkers of cross-sectional and longitudinal measures of general cognitive function and brain health, and incident dementia across three independent cohorts.</p><p><strong>Results: </strong>Using 84 protein EpiScores as candidate biomarkers, associations with general cognitive function (both cross-sectionally and longitudinally) were tested in three independent cohorts: Generation Scotland (GS), and the Lothian Birth Cohorts of 1921 and 1936 (LBC1921 and LBC1936, respectively). A meta-analysis of general cognitive functioning results in all three cohorts identified 18 EpiScore associations (absolute meta-analytic standardised estimates ranged from 0.03 to 0.14, median of 0.04, P<sub>FDR</sub> < 0.05). Several associations were also observed between EpiScores and global brain volumetric measures in the LBC1936. An EpiScore for the S100A9 protein (a known Alzheimer disease biomarker) was associated with general cognitive functioning (meta-analytic standardised beta: - 0.06, P = 1.3 × 10<sup>-9</sup>), and with time-to-dementia in GS (Hazard ratio 1.24, 95% confidence interval 1.08-1.44, P = 0.003), but not in LBC1936 (Hazard ratio 1.11, P = 0.32).</p><p><strong>Conclusions: </strong>EpiScores might make a contribution to the risk profile of poor general cognitive function and global brain health, and risk of dementia, however these scores require replication in further studies.</p>","PeriodicalId":10366,"journal":{"name":"Clinical Epigenetics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-03-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10962132/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140287129","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信