首页 > 最新文献

Clinical Epigenetics最新文献

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 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
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.
CSLCs(癌症干细胞样细胞)是肿瘤发生的核心,受到表观遗传修饰的内在影响。本研究旨在阐明 DNMT1/miR-152-3p/SOS1 轴调控 LCSLCs(肺癌干细胞样细胞)自我更新和肿瘤生长的内在机制。使用 TargetScan Human 8.0 预测了 miR-152-3p 的靶基因。在悬浮培养的非小细胞肺癌(NSCLC)细胞系 H460 和 A549 细胞衍生的球状细胞中比较了 LCSLC 的自我更新和肿瘤生长。通过体外和体内功能增益实验评估了 DNMT1/miR-152-3p/SOS1 轴的功能效应。此外,我们还采用荧光素酶报告实验来分析 DNMT1、miR-152-3p 和 SOS1 之间的相互作用。我们的研究结果表明,DNMT1 和 miR-152-3p 之间存在负相互作用,导致 miR-152-3p 水平降低。这反过来又会导致 miR-152-3p 对靶基因 SOS1 的抑制作用减弱,最终激活 SOS1,并在 LCSLC 的自我更新和肿瘤生长中发挥重要作用。然而,SOS1 的改变并不影响 DNMT1/miR-152-3p 的调控。因此,有理由推断 DNMT1/miR-152-3p 负反馈环通过 SOS1 关键性地维持了 LCSLC 的自我更新和肿瘤生长。这项研究揭示了NSCLC中CSLC(癌症干细胞)自我更新和肿瘤生长的新机制,并确定了NSCLC治疗的潜在靶点。
{"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 Pub Date : 2024-04-10 DOI: 10.1186/s13148-024-01666-2
Yuxi Cheng, Zhengzheng Song, Xiaodan Fang, Zhangui Tang
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.
多角体群(PcG)由一系列具有表观遗传调控效应的蛋白质组成,在从多能性、发育到癌变等各种生物过程中发挥着至关重要的作用。在这些蛋白中,泽斯特同源增强子 2(EZH2)是多聚酶抑制复合体 2(PRC2)的催化元件,在调节同源(Hox)基因的表达和 x 染色体失活的初始阶段发挥作用。在包括头颈部鳞状细胞癌(HNSCC)在内的多种人类癌症中,EZH2 经常过表达或被激活,并被确定为一种不良预后因素。值得注意的是,EZH2 是参与调控 STAT3/HOTAIR 轴的重要基因,通过调节口腔癌中的相关癌基因,影响 HNSCC 的增殖、分化并促进转移。目前,各种小分子化合物已被开发为特异性靶向 EZH2 的抑制剂,并已获准用于治疗难治性肿瘤。在这篇综述中,我们将深入研究 EZH2/PRC2 在 HNSCC 中介导的表观遗传调控,重点探讨 EZH2 的潜在作用和机制、其对靶向药物治疗的重要贡献以及其与癌症标志物和上皮-间质转化的关联。此外,我们还旨在揭示其作为口腔鳞状细胞癌治疗策略的潜力。
{"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 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
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.
对生物年龄加速的研究可能有助于识别高危人群,减轻与年龄有关的疾病不断增加的全球负担。我们利用DNA甲基化(DNAm)时钟研究了精神分裂症(SCZ)的生物衰老,这种精神疾病与年龄相关的残疾和发病率增加有关。在四个欧洲队列的 1090 例 SCZ 病例和 1206 例对照的全血 DNAm 样本中,我们使用三种 DNAm 时钟(即 Hannum、Horvath 和 Levine)对不同的衰老进行了荟萃分析。为了剖析DNAm衰老如何导致SCZ,我们整合了病程和SCZ多基因风险的信息,并按照生理年龄和生理性别进行了分层分析。我们发现,在 SCZ 中,基于血液的 DNAm 老化与发病以来的病程无关。我们观察到了性别特异性和非线性年龄效应,这些效应在不同的时钟之间存在差异,并指出了在 SCZ 中老化改变可能存在的不同年龄窗口。最值得注意的是,与对照组相比,SCZ 病例的细胞固有年龄(Horvath 时钟)在年轻时减慢,而表型年龄(Levine 时钟)则在成年后加速。表型老化的加速在多基因负担较高的女性 SCZ 患者中最为明显,年龄加速为 + 3.82 岁(CI 2.02-5.61,P = 1.1E-03)。表型老化和SCZ多基因风险对疾病的影响是叠加的,共同解释了高达14.38%的疾病状态变异。我们的研究为越来越多的证据表明SCZ患者的DNAm老化发生改变做出了贡献,并指出SCZ患者在年轻时的内在年龄减速,而在成年后的表型年龄加速。由于表型年龄的增加与全因死亡风险的增加有关,我们的研究结果表明,根据莱文时钟的测量,特定的、可识别的患者群体的死亡风险增加。我们的研究没有发现 DNAm 的衰老可以用患者的病程来解释,但我们确实观察到了年龄和性别特异性效应,这值得进一步研究。最后,我们的研究结果表明,将遗传学和表观遗传学预测因子结合起来可以改善对疾病结果的预测,并有助于精神分裂症的疾病管理。
{"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 Pub Date : 2024-04-05 DOI: 10.1186/s13148-024-01667-1
Xiaozhu Ma, Shuai Mei, Qidamugai Wuyun, Li Zhou, Dating Sun, Jiangtao Yan
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.
糖尿病心肌病(DCM)是一种严重的并发症,对糖尿病患者的健康构成重大威胁。糖尿病心肌病错综复杂的病理机制导致舒张功能障碍,并在晚期损害收缩功能。不断积累的研究发现,DCM 与各种表观遗传调控机制有关,包括 DNA 甲基化、组蛋白修饰、非编码 RNA 和其他表观遗传分子。最近,由于先进的高通量技术的发展,人们对表观遗传学在 DCM 病理生理学中的作用有了更深入的了解,这有助于开发潜在的治疗策略。在这篇综述中,我们简要介绍了表观遗传学调控,并更新了 DCM 的相关进展。我们提出了表观遗传因子和非编码 RNA(ncRNA)在 DCM 诊断和治疗中作为潜在生物标志物和药物的作用,为表观基因组学在 DCM 中的应用提供了新的视角和认识。
{"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 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
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.
硒与癌症之间引人入胜的联系就像一个令人着迷的谜题,让研究人员始终充满好奇。虽然硒已显示出通过补充硒来降低癌症风险的前景,但硒与宫颈癌表观遗传学的相互作用仍然是一个引人入胜、但在很大程度上尚未被探索的领域。揭示硒的作用及其与表观遗传因素相互作用的复杂性,可以为防治这种复杂疾病提供有价值的见解。硒对宫颈癌细胞有不同程度的显著抑制作用。在体外研究中,硒能有效抑制宫颈癌细胞的增殖、迁移和侵袭,同时促进细胞凋亡。硒对人宫颈癌衍生的器官组织也有明显的抑制作用。此外,在一项体内研究中,服用二氧化硒溶液可有效抑制小鼠宫颈癌肿瘤的生长。硒产生抑制作用的机制之一是它能够抑制组蛋白去甲基化酶,特别是 JMJD3 和 UTX。这种抑制作用在体外和体内均可观察到。值得注意的是,用 GSK-J4 抑制 JMJD3 和 UTX 时,在体外和体内模型中都能观察到类似的生物效应,能有效抑制宫颈癌患者的类器官模型。抑制 JMJD3 和 UTX 还能诱导 G2/M 期停滞、促进细胞凋亡并逆转上皮-间质转化(EMT)。ChIP-qPCR 分析证实,抑制 JMJD3 和 UTX 会增加特定组蛋白修饰 H3K27me3 在宫颈癌细胞(HeLa 和 SiHa 细胞)靶基因转录起始位点(TSS)的招募。此外,与邻近的正常宫颈组织相比,JMJD3 和 UTX 在宫颈癌组织中的表达明显升高,这表明它们有可能成为治疗靶点。我们的研究表明,硒对宫颈癌细胞的生长、迁移和侵袭有明显的抑制作用,并能促进细胞凋亡,有望成为一种治疗药物。我们发现组蛋白去甲基化酶 JMJD3 和 UTX 是硒的特异性靶点,对它们的抑制复制了所观察到的对癌细胞行为的影响。这些研究结果表明,JMJD3和UTX可能是以硒为基础治疗宫颈癌的重要靶点。
{"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 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

Background: 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.

Results: 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.

Conclusions: 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.

背景:癌症中的核小体重新定位被认为会导致基因组的组织和基因表达发生许多变化。了解这些变化对于阐明癌症的基本方面非常重要。这对基于无细胞DNA(cfDNA)的医学诊断也很重要,无细胞DNA来自基因组DNA区域,受到核糖体的保护而不被消化:结果:我们利用 MNase 辅助组蛋白 H3 ChIP-seq 技术在同一乳腺癌患者的成对肿瘤组织和正常组织中生成了高分辨率核小体图,并将其与血浆中相应的 cfDNA 进行了比较。这项分析以患者特异性的方式检测到了关键调控区域的单核糖体重新定位,以及不同患者之间常见的癌症特异性模式。与正常组织相比,肿瘤组织中获得的核小体对癌症通路特别有参考价值,CpG岛富集了约20倍,其中很大一部分标志着编码DNA结合蛋白的基因启动子。肿瘤组织的特点是核小体之间的平均距离(核小体重复长度,NRL)减少了 5-10 bp,这与多能细胞和分化细胞之间的差异在性质上相似。这种效应与基因活性、DNA甲基化差异以及连接组蛋白变体H1.4和H1X的局部占有率变化相关:我们的研究为乳腺癌患者的高分辨率核糖体图提供了新的资源,并首次报道了同一患者的成对肿瘤组织与正常乳腺组织中 NRL 系统性下降的影响。我们的研究结果提供了对肿瘤组织中核糖体重新定位的新的机理认识,这对患者的诊断、分层和监测很有价值。
{"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 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
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.
急性淋巴细胞白血病(ALL)是儿童中发病率最高的癌症,尽管在治疗效果方面取得了长足的进步,但复发仍会带来巨大的死亡风险和长期并发症。为了应对这一挑战,我们采用了一种有监督的机器学习技术,特别是随机生存森林,利用北欧国家接受治疗的 763 名儿童 ALL 患者的 DNA 甲基化阵列数据来预测复发和死亡风险。复发风险预测因子(RRP)是基于16个CpG位点构建的,在训练集和测试集中的c指数分别为0.667和0.677。死亡率风险预测因子(MRP)由 53 个 CpG 位点组成,在训练集和测试集中的 c 指数分别为 0.751 和 0.754。为了验证预测因子的预后价值,我们进一步分析了加拿大(42 人)和北欧(384 人)两组独立的 ALL 患者。外部验证证实了我们的研究结果,加拿大队列中的RRP c指数为0.667,北欧独立队列中的RRP和MRP c指数分别为0.529和0.621。在纳入传统的风险组数据后,RRP 和 MRP 模型的精确度有所提高,这凸显了协同整合临床预后因素的潜力。MRP 模型还能定义复发率和死亡率较高的风险组。我们的研究结果证明了DNA甲基化作为预后因素的潜力,以及作为完善小儿ALL风险分层的工具的潜力。这可能会导致基于表观遗传分析的个性化治疗策略。
{"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 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

Background: 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.

Methods: 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.

Results: 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+B220+ 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.

Conclusions: 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.

背景:miR-182启动子高甲基化经常发生在包括急性髓性白血病在内的各种肿瘤中,并导致miR-182的低表达。然而,成人急性淋巴细胞白血病(ALL)细胞是否存在高miR-182启动子甲基化尚未确定:为了评估 miR-182 启动子的甲基化状态,研究人员进行了甲基化和非甲基化特异性 PCR 分析、亚硫酸氢盐测序分析和 MethylTarget™ 检测,以测量 miR-182 启动子的甲基化频率。从 miR-182 基因敲除(182KO)和 182 野生型(182WT)小鼠身上分离骨髓细胞,分别构建 BCR-ABL (P190) 和 Notch 诱导的小鼠 B-ALL 和 T-ALL 模型。结果:由于ALL blasts中miR-182启动子的DNA超甲基化,ALL blasts中miR-182(miR-182-5P)的表达大大低于正常对照组(NCs),而正常对照组(NCs)则没有。在BCR-ABL(P190)诱导的小鼠B-ALL模型中,敲除miR-182(182KO)明显加速了ALL的发展、促进了浸润并缩短了OS。此外,与 182WT ALL 细胞群相比,182KO ALL 细胞群富含更多的白血病启动细胞(CD43+B220+ 细胞,LICs),并具有更高的致白血病活性。此外,在Notch诱导的小鼠T-ALL模型中,去掉miR-182会降低OS,这表明miR-182敲除会加速ALL的发展。从机理上讲,miR-182的过表达通过直接靶向PBX3和BCL2抑制增殖并诱导凋亡,而PBX3和BCL2是两个著名的致癌基因,是miR-182的关键靶点。最重要的是,DAC联合Ven对miR-182启动子高甲基化的ALL细胞有协同作用,但对miR-182启动子低甲基化的ALL细胞没有协同作用:总之,我们发现miR-182是ALL细胞中的肿瘤抑制基因,而miR-182因高甲基化而低表达会促进ALL细胞的恶性表型。DAC+Ven联合治疗可用于miR-182启动子高甲基化ALL患者的临床试验。此外,miR-182启动子的甲基化频率应成为DAC+Ven治疗ALL患者的潜在生物标志物。
{"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 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

Background: 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.

Results: 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.

Conclusions: 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.

背景:原发部位不明的头颈部癌症(hnCUP)的原发组织不明,导致诊断程序繁琐,患者无法选择特异性治疗方案,而且治疗效果可能不佳。最常见的组织学亚型--鳞状细胞癌可能来自不同的肿瘤原发部位,包括口腔、口咽、喉、头颈部皮肤、肺和食道。DNA 甲基化图谱具有高度的组织特异性,已成功用于组织来源的分类。因此,我们开发了一种支持向量机(SVM)分类器,该分类器利用公开的常见颈部转移鳞状细胞癌(n = 1103)DNA甲基化图谱进行训练,以确定我们自己的鳞状细胞hnCUP患者样本群(n = 28)的原发组织。甲基化分析是通过 Illumina 的 Infinium MethylationEPIC v1.0 BeadChip 进行的:结果:在测试的分类器中,SVM 算法的总体准确率最高,达到 87%。鳞状细胞 hnCUP 样本的 DNA 甲基化水平与通常转移到宫颈淋巴结的鳞状细胞癌相似。最常预测的癌症部位是口腔(11 例,占 39%),其次是口咽和喉部(均为 7 例,占 25%)、皮肤(2 例,占 7%)和食道(1 例,占 4%)。这些频率与流行病学研究中预期的淋巴结转移分布一致:结论:在DNA甲基化水平上,hnCUP与常转移至宫颈淋巴结的原发肿瘤组织癌症类型相当。我们基于 SVM 的分类器能准确预测这些癌症的原发组织,可大大降低 hnCUP 诊断的侵入性,并在临床验证后提供更精确的治疗。
{"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 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

Background: 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.

Results: 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, PFDR < 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 × 10-9), 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).

Conclusions: 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.

背景:血液蛋白质水平的表观遗传评分(EpiScores)与疾病预后和大脑健康状况相关联,凸显了其作为临床生物标志物的潜在作用。表观遗传评分通常是通过惩罚性回归得出的,CpG位点的DNA甲基化(DNAm)水平的线性加权和可预测蛋白质水平。在此,我们研究了之前发表的 84 个蛋白质 EpiScores,将其作为三个独立队列中一般认知功能和大脑健康的横断面和纵向测量指标以及痴呆症事件的可能生物标志物:使用 84 个蛋白质 EpiScores 作为候选生物标志物,在三个独立队列中测试了与一般认知功能的关联(横向和纵向):苏格兰一代(GS)以及 1921 年和 1936 年洛锡安出生队列(分别为 LBC1921 和 LBC1936)。对所有三个队列的一般认知功能结果进行荟萃分析,发现了 18 项 EpiScore 关联(荟萃分析标准化估计值的绝对值从 0.03 到 0.14 不等,中位数为 0.04,PFDR 为 -9),在 GS 中与痴呆时间有关(危险比为 1.24,95% 置信区间为 1.08-1.44,P = 0.003),但在 LBC1936 中与痴呆时间无关(危险比为 1.11,P = 0.32):EpiScores可能会对一般认知功能和大脑整体健康状况不佳的风险概况以及痴呆症的风险做出贡献,但这些分数需要在进一步的研究中加以验证。
{"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
期刊
Clinical Epigenetics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
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
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 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 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1