首页 > 最新文献

NAR Cancer最新文献

英文 中文
Correction to 'Tumor-selective, antigen-independent delivery of a pH sensitive peptide-topoisomerase inhibitor conjugate suppresses tumor growth without systemic toxicity'. 更正“肿瘤选择性,抗原不依赖于pH敏感肽-拓扑异构酶抑制剂偶联物的递送抑制肿瘤生长而无全身毒性”。
Pub Date : 2021-12-07 eCollection Date: 2021-12-01 DOI: 10.1093/narcan/zcab047
Sophia Gayle, Robert Aiello, Nalin Leelatian, Jason M Beckta, Jane Bechtold, Patricia Bourassa, Johanna Csengery, Robert J Maguire, Dan Marshall, Ranjini K Sundaram, Jinny Van Doorn, Kelli Jones, Hunter Moore, Lori Lopresti-Morrow, Timothy Paradis, Laurie Tylaska, Qing Zhang, Hannah Visca, Yana K Reshetnyak, Oleg A Andreev, Donald M Engelman, Peter M Glazer, Ranjit S Bindra, Vishwas M Paralkar

[This corrects the article DOI: 10.1093/narcan/zcab021.].

[这更正了文章DOI: 10.1093/narcan/zcab021.]。
{"title":"Correction to 'Tumor-selective, antigen-independent delivery of a pH sensitive peptide-topoisomerase inhibitor conjugate suppresses tumor growth without systemic toxicity'.","authors":"Sophia Gayle,&nbsp;Robert Aiello,&nbsp;Nalin Leelatian,&nbsp;Jason M Beckta,&nbsp;Jane Bechtold,&nbsp;Patricia Bourassa,&nbsp;Johanna Csengery,&nbsp;Robert J Maguire,&nbsp;Dan Marshall,&nbsp;Ranjini K Sundaram,&nbsp;Jinny Van Doorn,&nbsp;Kelli Jones,&nbsp;Hunter Moore,&nbsp;Lori Lopresti-Morrow,&nbsp;Timothy Paradis,&nbsp;Laurie Tylaska,&nbsp;Qing Zhang,&nbsp;Hannah Visca,&nbsp;Yana K Reshetnyak,&nbsp;Oleg A Andreev,&nbsp;Donald M Engelman,&nbsp;Peter M Glazer,&nbsp;Ranjit S Bindra,&nbsp;Vishwas M Paralkar","doi":"10.1093/narcan/zcab047","DOIUrl":"https://doi.org/10.1093/narcan/zcab047","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1093/narcan/zcab021.].</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8651162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39710316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Polycomb group proteins in cancer: multifaceted functions and strategies for modulation. 肿瘤中的多梳蛋白:多面功能和调节策略。
Pub Date : 2021-12-01 DOI: 10.1093/narcan/zcab039
Sijie Wang, Sandra C Ordonez-Rubiano, Alisha Dhiman, Guanming Jiao, Brayden P Strohmier, Casey J Krusemark, Emily C Dykhuizen

Polycomb repressive complexes (PRCs) are a heterogenous collection of dozens, if not hundreds, of protein complexes composed of various combinations of subunits. PRCs are transcriptional repressors important for cell-type specificity during development, and as such, are commonly mis-regulated in cancer. PRCs are broadly characterized as PRC1 with histone ubiquitin ligase activity, or PRC2 with histone methyltransferase activity; however, the mechanism by which individual PRCs, particularly the highly diverse set of PRC1s, alter gene expression has not always been clear. Here we review the current understanding of how PRCs act, both individually and together, to establish and maintain gene repression, the biochemical contribution of individual PRC subunits, the mis-regulation of PRC function in different cancers, and the current strategies for modulating PRC activity. Increased mechanistic understanding of PRC function, as well as cancer-specific roles for individual PRC subunits, will uncover better targets and strategies for cancer therapies.

多梳抑制复合物(PRCs)是由不同亚基组合组成的数十种(如果不是数百种)蛋白质复合物的异质集合。prc是发育过程中对细胞类型特异性很重要的转录抑制因子,因此,在癌症中通常被错误调节。PRCs一般分为具有组蛋白泛素连接酶活性的PRC1和具有组蛋白甲基转移酶活性的PRC2;然而,个体PRCs,特别是高度多样化的PRC1s,改变基因表达的机制并不总是很清楚。在这里,我们回顾了目前对PRC如何单独或共同作用以建立和维持基因抑制的理解,单个PRC亚基的生化贡献,不同癌症中PRC功能的错误调节,以及当前调节PRC活性的策略。增加对PRC功能的机制理解,以及单个PRC亚基的癌症特异性作用,将发现更好的癌症治疗靶点和策略。
{"title":"Polycomb group proteins in cancer: multifaceted functions and strategies for modulation.","authors":"Sijie Wang,&nbsp;Sandra C Ordonez-Rubiano,&nbsp;Alisha Dhiman,&nbsp;Guanming Jiao,&nbsp;Brayden P Strohmier,&nbsp;Casey J Krusemark,&nbsp;Emily C Dykhuizen","doi":"10.1093/narcan/zcab039","DOIUrl":"https://doi.org/10.1093/narcan/zcab039","url":null,"abstract":"<p><p>Polycomb repressive complexes (PRCs) are a heterogenous collection of dozens, if not hundreds, of protein complexes composed of various combinations of subunits. PRCs are transcriptional repressors important for cell-type specificity during development, and as such, are commonly mis-regulated in cancer. PRCs are broadly characterized as PRC1 with histone ubiquitin ligase activity, or PRC2 with histone methyltransferase activity; however, the mechanism by which individual PRCs, particularly the highly diverse set of PRC1s, alter gene expression has not always been clear. Here we review the current understanding of how PRCs act, both individually and together, to establish and maintain gene repression, the biochemical contribution of individual PRC subunits, the mis-regulation of PRC function in different cancers, and the current strategies for modulating PRC activity. Increased mechanistic understanding of PRC function, as well as cancer-specific roles for individual PRC subunits, will uncover better targets and strategies for cancer therapies.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8489530/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10058377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 10
NAD+ bioavailability mediates PARG inhibition-induced replication arrest, intra S-phase checkpoint and apoptosis in glioma stem cells. NAD+生物利用度介导PARG抑制诱导的胶质瘤干细胞复制阻滞、s期检查点和凋亡。
Pub Date : 2021-12-01 DOI: 10.1093/narcan/zcab044
Jianfeng Li, Kate M Saville, Md Ibrahim, Xuemei Zeng, Steve McClellan, Anusha Angajala, Alison Beiser, Joel F Andrews, Mai Sun, Christopher A Koczor, Jennifer Clark, Faisal Hayat, Mikhail V Makarov, Anna Wilk, Nathan A Yates, Marie E Migaud, Robert W Sobol

Elevated expression of the DNA damage response proteins PARP1 and poly(ADP-ribose) glycohydrolase (PARG) in glioma stem cells (GSCs) suggests that glioma may be a unique target for PARG inhibitors (PARGi). While PARGi-induced cell death is achieved when combined with ionizing radiation, as a single agent PARG inhibitors appear to be mostly cytostatic. Supplementation with the NAD+ precursor dihydronicotinamide riboside (NRH) rapidly increased NAD+ levels in GSCs and glioma cells, inducing PARP1 activation and mild suppression of replication fork progression. Administration of NRH+PARGi triggers hyperaccumulation of poly(ADP-ribose) (PAR), intra S-phase arrest and apoptosis in GSCs but minimal PAR induction or cytotoxicity in normal astrocytes. PAR accumulation is regulated by select PARP1- and PAR-interacting proteins. The involvement of XRCC1 highlights the base excision repair pathway in responding to replication stress while enhanced interaction of PARP1 with PCNA, RPA and ORC2 upon PAR accumulation implicates replication associated PARP1 activation and assembly with pre-replication complex proteins upon initiation of replication arrest, the intra S-phase checkpoint and the onset of apoptosis.

DNA损伤反应蛋白PARP1和聚(adp -核糖)糖水解酶(PARG)在胶质瘤干细胞(GSCs)中的表达升高表明胶质瘤可能是PARG抑制剂(PARGi)的独特靶点。虽然PARG诱导的细胞死亡是在与电离辐射联合使用时实现的,但作为单一药物,PARG抑制剂似乎主要是细胞抑制剂。补充NAD+前体二氢烟碱酰胺核苷(NRH)可迅速增加GSCs和胶质瘤细胞中的NAD+水平,诱导PARP1激活并轻度抑制复制叉的进展。NRH+PARGi在GSCs中引发多聚adp核糖(PAR)的过度积累、s期阻滞和凋亡,但在正常星形胶质细胞中PAR的诱导或细胞毒性很小。PAR的积累受PARP1-和PAR-相互作用蛋白的调控。XRCC1的参与强调了碱基切除修复途径在应对复制应激中的作用,而PARP1在PAR积累时与PCNA、RPA和ORC2的相互作用增强,暗示了复制相关的PARP1激活,并在复制停止、s期内checkpoint和凋亡开始时与复制前复合体蛋白组装。
{"title":"NAD<sup>+</sup> bioavailability mediates PARG inhibition-induced replication arrest, intra S-phase checkpoint and apoptosis in glioma stem cells.","authors":"Jianfeng Li,&nbsp;Kate M Saville,&nbsp;Md Ibrahim,&nbsp;Xuemei Zeng,&nbsp;Steve McClellan,&nbsp;Anusha Angajala,&nbsp;Alison Beiser,&nbsp;Joel F Andrews,&nbsp;Mai Sun,&nbsp;Christopher A Koczor,&nbsp;Jennifer Clark,&nbsp;Faisal Hayat,&nbsp;Mikhail V Makarov,&nbsp;Anna Wilk,&nbsp;Nathan A Yates,&nbsp;Marie E Migaud,&nbsp;Robert W Sobol","doi":"10.1093/narcan/zcab044","DOIUrl":"https://doi.org/10.1093/narcan/zcab044","url":null,"abstract":"<p><p>Elevated expression of the DNA damage response proteins PARP1 and poly(ADP-ribose) glycohydrolase (PARG) in glioma stem cells (GSCs) suggests that glioma may be a unique target for PARG inhibitors (PARGi). While PARGi-induced cell death is achieved when combined with ionizing radiation, as a single agent PARG inhibitors appear to be mostly cytostatic. Supplementation with the NAD<sup>+</sup> precursor dihydronicotinamide riboside (NRH) rapidly increased NAD<sup>+</sup> levels in GSCs and glioma cells, inducing PARP1 activation and mild suppression of replication fork progression. Administration of NRH+PARGi triggers hyperaccumulation of poly(ADP-ribose) (PAR), intra S-phase arrest and apoptosis in GSCs but minimal PAR induction or cytotoxicity in normal astrocytes. PAR accumulation is regulated by select PARP1- and PAR-interacting proteins. The involvement of XRCC1 highlights the base excision repair pathway in responding to replication stress while enhanced interaction of PARP1 with PCNA, RPA and ORC2 upon PAR accumulation implicates replication associated PARP1 activation and assembly with pre-replication complex proteins upon initiation of replication arrest, the intra S-phase checkpoint and the onset of apoptosis.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8600031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10636562","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Misregulation of the expression and activity of DNA methyltransferases in cancer. 肿瘤中DNA甲基转移酶表达和活性的错误调控。
Pub Date : 2021-12-01 DOI: 10.1093/narcan/zcab045
Isaiah K Mensah, Allison B Norvil, Lama AlAbdi, Sarah McGovern, Christopher J Petell, Ming He, Humaira Gowher

In mammals, DNA methyltransferases DNMT1 and DNMT3's (A, B and L) deposit and maintain DNA methylation in dividing and nondividing cells. Although these enzymes have an unremarkable DNA sequence specificity (CpG), their regional specificity is regulated by interactions with various protein factors, chromatin modifiers, and post-translational modifications of histones. Changes in the DNMT expression or interacting partners affect DNA methylation patterns. Consequently, the acquired gene expression may increase the proliferative potential of cells, often concomitant with loss of cell identity as found in cancer. Aberrant DNA methylation, including hypermethylation and hypomethylation at various genomic regions, therefore, is a hallmark of most cancers. Additionally, somatic mutations in DNMTs that affect catalytic activity were mapped in Acute Myeloid Leukemia cancer cells. Despite being very effective in some cancers, the clinically approved DNMT inhibitors lack specificity, which could result in a wide range of deleterious effects. Elucidating distinct molecular mechanisms of DNMTs will facilitate the discovery of alternative cancer therapeutic targets. This review is focused on: (i) the structure and characteristics of DNMTs, (ii) the prevalence of mutations and abnormal expression of DNMTs in cancer, (iii) factors that mediate their abnormal expression and (iv) the effect of anomalous DNMT-complexes in cancer.

在哺乳动物中,DNA甲基转移酶DNMT1和DNMT3 (A、B和L)在分裂和非分裂细胞中沉积并维持DNA甲基化。尽管这些酶具有不显著的DNA序列特异性(CpG),但它们的区域特异性受到与各种蛋白质因子、染色质修饰剂和组蛋白翻译后修饰的相互作用的调节。DNMT表达或相互作用伙伴的变化影响DNA甲基化模式。因此,获得性基因表达可能增加细胞的增殖潜能,通常伴随着癌症中发现的细胞特性的丧失。因此,异常的DNA甲基化,包括不同基因组区域的高甲基化和低甲基化,是大多数癌症的标志。此外,在急性髓系白血病癌细胞中发现了影响催化活性的dnmt体细胞突变。尽管在某些癌症中非常有效,但临床批准的DNMT抑制剂缺乏特异性,这可能导致广泛的有害影响。阐明DNMTs的不同分子机制将有助于发现替代的癌症治疗靶点。本文综述的重点是:(i) dnmt的结构和特征,(ii)癌症中dnmt的突变和异常表达的流行,(iii)介导其异常表达的因素,以及(iv)异常dnmt复合物在癌症中的作用。
{"title":"Misregulation of the expression and activity of DNA methyltransferases in cancer.","authors":"Isaiah K Mensah,&nbsp;Allison B Norvil,&nbsp;Lama AlAbdi,&nbsp;Sarah McGovern,&nbsp;Christopher J Petell,&nbsp;Ming He,&nbsp;Humaira Gowher","doi":"10.1093/narcan/zcab045","DOIUrl":"https://doi.org/10.1093/narcan/zcab045","url":null,"abstract":"<p><p>In mammals, DNA methyltransferases DNMT1 and DNMT3's (A, B and L) deposit and maintain DNA methylation in dividing and nondividing cells. Although these enzymes have an unremarkable DNA sequence specificity (CpG), their regional specificity is regulated by interactions with various protein factors, chromatin modifiers, and post-translational modifications of histones. Changes in the DNMT expression or interacting partners affect DNA methylation patterns. Consequently, the acquired gene expression may increase the proliferative potential of cells, often concomitant with loss of cell identity as found in cancer. Aberrant DNA methylation, including hypermethylation and hypomethylation at various genomic regions, therefore, is a hallmark of most cancers. Additionally, somatic mutations in DNMTs that affect catalytic activity were mapped in Acute Myeloid Leukemia cancer cells. Despite being very effective in some cancers, the clinically approved DNMT inhibitors lack specificity, which could result in a wide range of deleterious effects. Elucidating distinct molecular mechanisms of DNMTs will facilitate the discovery of alternative cancer therapeutic targets. This review is focused on: (i) the structure and characteristics of DNMTs, (ii) the prevalence of mutations and abnormal expression of DNMTs in cancer, (iii) factors that mediate their abnormal expression and (iv) the effect of anomalous DNMT-complexes in cancer.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/29/06/zcab045.PMC8634572.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39696531","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 9
Modulating gene expression in breast cancer via DNA secondary structure and the CRISPR toolbox. 通过DNA二级结构和CRISPR工具箱调节乳腺癌中的基因表达。
Pub Date : 2021-12-01 DOI: 10.1093/narcan/zcab048
Jessica A Kretzmann, Kelly L Irving, Nicole M Smith, Cameron W Evans

Breast cancer is the most commonly diagnosed malignancy in women, and while the survival prognosis of patients with early-stage, non-metastatic disease is ∼75%, recurrence poses a significant risk and advanced and/or metastatic breast cancer is incurable. A distinctive feature of advanced breast cancer is an unstable genome and altered gene expression patterns that result in disease heterogeneity. Transcription factors represent a unique therapeutic opportunity in breast cancer, since they are known regulators of gene expression, including gene expression involved in differentiation and cell death, which are themselves often mutated or dysregulated in cancer. While transcription factors have traditionally been viewed as 'undruggable', progress has been made in the development of small-molecule therapeutics to target relevant protein-protein, protein-DNA and enzymatic active sites, with varying levels of success. However, non-traditional approaches such as epigenetic editing, transcriptional control via CRISPR/dCas9 systems, and gene regulation through non-canonical nucleic acid secondary structures represent new directions yet to be fully explored. Here, we discuss these new approaches and current limitations in light of new therapeutic opportunities for breast cancers.

乳腺癌是女性中最常见的恶性肿瘤,虽然早期非转移性疾病患者的生存预后约为75%,但复发的风险很大,晚期和/或转移性乳腺癌是无法治愈的。晚期乳腺癌的一个显著特征是不稳定的基因组和改变的基因表达模式,导致疾病异质性。转录因子代表了乳腺癌的独特治疗机会,因为它们是已知的基因表达调节剂,包括参与分化和细胞死亡的基因表达,而这些基因表达本身在癌症中经常发生突变或失调。虽然转录因子传统上被认为是“不可药物的”,但针对相关蛋白质-蛋白质、蛋白质- dna和酶活性位点的小分子疗法的开发已经取得了进展,并取得了不同程度的成功。然而,表观遗传编辑、通过CRISPR/dCas9系统进行转录调控、通过非典型核酸二级结构进行基因调控等非传统途径则是有待充分探索的新方向。在这里,我们讨论这些新的方法和目前的局限性,在新的治疗机会乳腺癌。
{"title":"Modulating gene expression in breast cancer via DNA secondary structure and the CRISPR toolbox.","authors":"Jessica A Kretzmann,&nbsp;Kelly L Irving,&nbsp;Nicole M Smith,&nbsp;Cameron W Evans","doi":"10.1093/narcan/zcab048","DOIUrl":"https://doi.org/10.1093/narcan/zcab048","url":null,"abstract":"<p><p>Breast cancer is the most commonly diagnosed malignancy in women, and while the survival prognosis of patients with early-stage, non-metastatic disease is ∼75%, recurrence poses a significant risk and advanced and/or metastatic breast cancer is incurable. A distinctive feature of advanced breast cancer is an unstable genome and altered gene expression patterns that result in disease heterogeneity. Transcription factors represent a unique therapeutic opportunity in breast cancer, since they are known regulators of gene expression, including gene expression involved in differentiation and cell death, which are themselves often mutated or dysregulated in cancer. While transcription factors have traditionally been viewed as 'undruggable', progress has been made in the development of small-molecule therapeutics to target relevant protein-protein, protein-DNA and enzymatic active sites, with varying levels of success. However, non-traditional approaches such as epigenetic editing, transcriptional control via CRISPR/dCas9 systems, and gene regulation through non-canonical nucleic acid secondary structures represent new directions yet to be fully explored. Here, we discuss these new approaches and current limitations in light of new therapeutic opportunities for breast cancers.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8693572/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9693217","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
LncRNA Hmrhl regulates expression of cancer related genes in chronic myelogenous leukemia through chromatin association. LncRNA Hmrhl通过染色质关联调控慢性髓性白血病肿瘤相关基因的表达。
Pub Date : 2021-11-01 eCollection Date: 2021-12-01 DOI: 10.1093/narcan/zcab042
Subhendu Roy Choudhury, Sangeeta Dutta, Utsa Bhaduri, Manchanahalli R Satyanarayana Rao

Long non-coding RNA has emerged as a key regulator of myriad gene functions. One such lncRNA mrhl, reported by our group, was found to have important role in spermatogenesis and embryonic development in mouse. Recently, its human homolog, Hmrhl was shown to have differential expression in several type of cancers. In the present study, we further characterize molecular features of Hmrhl and gain insight into its functional role in leukemia by gene silencing and transcriptome-based studies. Results indicate its high expression in CML patient samples as well as in K562 cell line. Silencing experiments suggest role of Hmrhl in cell proliferation, migration & invasion. RNA-seq and ChiRP-seq data analysis further revealed its association with important biological processes, including perturbed expression of crucial TFs and cancer-related genes. Among them ZIC1, PDGRFβ and TP53 were identified as regulatory targets, with high possibility of triplex formation by Hmrhl at their promoter site. Further, overexpression of PDGRFβ in Hmrhl silenced cells resulted in rescue effect of cancer associated cellular phenotypes. In addition, we also found TAL-1 to be a potential regulator of Hmrhl expression in K562 cells. Thus, we hypothesize that Hmrhl lncRNA may play a significant role in the pathobiology of CML.

长链非编码RNA已成为无数基因功能的关键调控因子。本课题组报道的其中一个lncRNA mrhl在小鼠精子发生和胚胎发育中发挥重要作用。最近,它的人类同源物Hmrhl被证明在几种类型的癌症中有不同的表达。在本研究中,我们进一步表征了Hmrhl的分子特征,并通过基因沉默和转录组研究深入了解其在白血病中的功能作用。结果表明其在CML患者和K562细胞系中均有高表达。沉默实验提示Hmrhl在细胞增殖、迁移和侵袭中起作用。RNA-seq和ChiRP-seq数据分析进一步揭示了其与重要生物学过程的关联,包括关键tf和癌症相关基因的表达紊乱。其中ZIC1, PDGRFβ和TP53被确定为调控靶点,Hmrhl在其启动子位点形成三联体的可能性很高。此外,在Hmrhl沉默细胞中,PDGRFβ的过表达导致癌症相关细胞表型的拯救作用。此外,我们还发现TAL-1是K562细胞中Hmrhl表达的潜在调节因子。因此,我们推测Hmrhl lncRNA可能在CML的病理生物学中发挥重要作用。
{"title":"LncRNA Hmrhl regulates expression of cancer related genes in chronic myelogenous leukemia through chromatin association.","authors":"Subhendu Roy Choudhury,&nbsp;Sangeeta Dutta,&nbsp;Utsa Bhaduri,&nbsp;Manchanahalli R Satyanarayana Rao","doi":"10.1093/narcan/zcab042","DOIUrl":"https://doi.org/10.1093/narcan/zcab042","url":null,"abstract":"<p><p>Long non-coding RNA has emerged as a key regulator of myriad gene functions. One such lncRNA mrhl, reported by our group, was found to have important role in spermatogenesis and embryonic development in mouse. Recently, its human homolog, Hmrhl was shown to have differential expression in several type of cancers. In the present study, we further characterize molecular features of Hmrhl and gain insight into its functional role in leukemia by gene silencing and transcriptome-based studies. Results indicate its high expression in CML patient samples as well as in K562 cell line. Silencing experiments suggest role of Hmrhl in cell proliferation, migration & invasion. RNA-seq and ChiRP-seq data analysis further revealed its association with important biological processes, including perturbed expression of crucial TFs and cancer-related genes. Among them <i>ZIC1</i>, <i>PDGRFβ</i> and <i>TP53</i> were identified as regulatory targets, with high possibility of triplex formation by Hmrhl at their promoter site. Further, overexpression of <i>PDGRFβ</i> in Hmrhl silenced cells resulted in rescue effect of cancer associated cellular phenotypes. In addition, we also found TAL-1 to be a potential regulator of Hmrhl expression in K562 cells. Thus, we hypothesize that Hmrhl lncRNA may play a significant role in the pathobiology of CML.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/f7/95/zcab042.PMC8559160.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39677769","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
Can the epigenome contribute to risk stratification for cancer onset? 表观基因组是否有助于癌症发病的风险分层?
Pub Date : 2021-11-01 eCollection Date: 2021-12-01 DOI: 10.1093/narcan/zcab043
Sophie A Lelièvre

The increasing burden of cancer requires identifying and protecting individuals at highest risk. The epigenome provides an indispensable complement to genetic alterations for a risk stratification approach for the following reasons: gene transcription necessary for cancer onset is directed by epigenetic modifications and many risk factors studied so far have been associated with alterations related to the epigenome. The risk level depends on the plasticity of the epigenome during phases of life particularly sensitive to environmental and dietary impacts. Modifications in the activity of DNA regulatory regions and altered chromatin compaction may accumulate, hence leading to the increase of cancer risk. Moreover, tissue architecture directs the unique organization of the epigenome for each tissue and cell type, which allows the epigenome to control cancer risk in specific organs. Investigations of epigenetic signatures of risk should help identify a continuum of alterations leading to a threshold beyond which the epigenome cannot maintain homeostasis. We propose that this threshold may be similar in the population for a given tissue, but the pace to reach this threshold will depend on the combination of germline inheritance and the risk and protective factors encountered, particularly during windows of epigenetic susceptibility, by individuals.

日益增加的癌症负担要求识别和保护高危人群。由于以下原因,表观基因组为风险分层方法的遗传改变提供了不可或缺的补充:癌症发病所需的基因转录是由表观遗传修饰指导的,迄今为止研究的许多危险因素都与表观基因组相关的改变有关。风险水平取决于对环境和饮食影响特别敏感的生命阶段表观基因组的可塑性。DNA调控区活性的改变和染色质压实的改变可能会累积,从而导致癌症风险的增加。此外,组织结构指导着每一种组织和细胞类型的表观基因组的独特组织,这使得表观基因组能够控制特定器官的癌症风险。对风险的表观遗传特征的研究应该有助于确定导致表观基因组无法维持稳态的阈值的连续变化。我们认为这个阈值可能在群体中与给定组织相似,但达到这个阈值的速度将取决于种系遗传和遇到的风险和保护因素的组合,特别是在个体的表观遗传易感性窗口期间。
{"title":"Can the epigenome contribute to risk stratification for cancer onset?","authors":"Sophie A Lelièvre","doi":"10.1093/narcan/zcab043","DOIUrl":"https://doi.org/10.1093/narcan/zcab043","url":null,"abstract":"<p><p>The increasing burden of cancer requires identifying and protecting individuals at highest risk. The epigenome provides an indispensable complement to genetic alterations for a risk stratification approach for the following reasons: gene transcription necessary for cancer onset is directed by epigenetic modifications and many risk factors studied so far have been associated with alterations related to the epigenome. The risk level depends on the plasticity of the epigenome during phases of life particularly sensitive to environmental and dietary impacts. Modifications in the activity of DNA regulatory regions and altered chromatin compaction may accumulate, hence leading to the increase of cancer risk. Moreover, tissue architecture directs the unique organization of the epigenome for each tissue and cell type, which allows the epigenome to control cancer risk in specific organs. Investigations of epigenetic signatures of risk should help identify a continuum of alterations leading to a threshold beyond which the epigenome cannot maintain homeostasis. We propose that this threshold may be similar in the population for a given tissue, but the pace to reach this threshold will depend on the combination of germline inheritance and the risk and protective factors encountered, particularly during windows of epigenetic susceptibility, by individuals.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/f6/d4/zcab043.PMC8559165.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39677770","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
UNMASC: tumor-only variant calling with unmatched normal controls. UNMASC:只有肿瘤的变种呼叫与不匹配的正常对照。
Pub Date : 2021-10-06 eCollection Date: 2021-12-01 DOI: 10.1093/narcan/zcab040
Paul Little, Heejoon Jo, Alan Hoyle, Angela Mazul, Xiaobei Zhao, Ashley H Salazar, Douglas Farquhar, Siddharth Sheth, Maheer Masood, Michele C Hayward, Joel S Parker, Katherine A Hoadley, Jose Zevallos, D Neil Hayes

Despite years of progress, mutation detection in cancer samples continues to require significant manual review as a final step. Expert review is particularly challenging in cases where tumors are sequenced without matched normal control DNA. Attempts have been made to call somatic point mutations without a matched normal sample by removing well-known germline variants, utilizing unmatched normal controls, and constructing decision rules to classify sequencing errors and private germline variants. With budgetary constraints related to computational and sequencing costs, finding the appropriate number of controls is a crucial step to identifying somatic variants. Our approach utilizes public databases for canonical somatic variants as well as germline variants and leverages information gathered about nearby positions in the normal controls. Drawing from our cohort of targeted capture panel sequencing of tumor and normal samples with varying tumortypes and demographics, these served as a benchmark for our tumor-only variant calling pipeline to observe the relationship between our ability to correctly classify variants against a number of unmatched normals. With our benchmarked samples, approximately ten normal controls were needed to maintain 94% sensitivity, 99% specificity and 76% positive predictive value, far outperforming comparable methods. Our approach, called UNMASC, also serves as a supplement to traditional tumor with matched normal variant calling workflows and can potentially extend to other concerns arising from analyzing next generation sequencing data.

尽管取得了多年的进展,但癌症样本中的突变检测仍然需要大量的人工审查作为最后一步。在没有匹配的正常对照DNA进行肿瘤测序的情况下,专家审查尤其具有挑战性。通过去除已知的种系变异,利用不匹配的正常对照,以及构建决策规则来分类测序错误和私人种系变异,已经尝试在没有匹配正常样本的情况下调用体细胞点突变。由于与计算和测序成本相关的预算限制,找到适当数量的控制是识别体细胞变异的关键一步。我们的方法利用了典型体细胞变异和种系变异的公共数据库,并利用了在正常对照中收集的有关附近位置的信息。从我们针对不同肿瘤类型和人口统计学的肿瘤和正常样本的靶向捕获面板测序队列中提取,这些作为我们仅肿瘤变体调用管道的基准,以观察我们正确分类变体与许多不匹配的正常之间的关系。对于我们的基准样本,大约需要10个正常对照才能保持94%的灵敏度,99%的特异性和76%的阳性预测值,远远优于同类方法。我们的方法,称为UNMASC,也可以作为传统肿瘤的补充,具有匹配的正常变异调用工作流程,并且可以潜在地扩展到分析下一代测序数据所引起的其他问题。
{"title":"UNMASC: tumor-only variant calling with unmatched normal controls.","authors":"Paul Little,&nbsp;Heejoon Jo,&nbsp;Alan Hoyle,&nbsp;Angela Mazul,&nbsp;Xiaobei Zhao,&nbsp;Ashley H Salazar,&nbsp;Douglas Farquhar,&nbsp;Siddharth Sheth,&nbsp;Maheer Masood,&nbsp;Michele C Hayward,&nbsp;Joel S Parker,&nbsp;Katherine A Hoadley,&nbsp;Jose Zevallos,&nbsp;D Neil Hayes","doi":"10.1093/narcan/zcab040","DOIUrl":"https://doi.org/10.1093/narcan/zcab040","url":null,"abstract":"<p><p>Despite years of progress, mutation detection in cancer samples continues to require significant manual review as a final step. Expert review is particularly challenging in cases where tumors are sequenced without matched normal control DNA. Attempts have been made to call somatic point mutations without a matched normal sample by removing well-known germline variants, utilizing unmatched normal controls, and constructing decision rules to classify sequencing errors and private germline variants. With budgetary constraints related to computational and sequencing costs, finding the appropriate number of controls is a crucial step to identifying somatic variants. Our approach utilizes public databases for canonical somatic variants as well as germline variants and leverages information gathered about nearby positions in the normal controls. Drawing from our cohort of targeted capture panel sequencing of tumor and normal samples with varying tumortypes and demographics, these served as a benchmark for our tumor-only variant calling pipeline to observe the relationship between our ability to correctly classify variants against a number of unmatched normals. With our benchmarked samples, approximately ten normal controls were needed to maintain 94% sensitivity, 99% specificity and 76% positive predictive value, far outperforming comparable methods. Our approach, called UNMASC, also serves as a supplement to traditional tumor with matched normal variant calling workflows and can potentially extend to other concerns arising from analyzing next generation sequencing data.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-10-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8494212/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39506841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 4
Correction to 'Genome instability and pressure on non-homologous end joining drives chemotherapy resistance via a DNA repair crisis switch in triple negative breast cancer'. 更正“基因组不稳定和非同源末端连接的压力通过三阴性乳腺癌的DNA修复危机开关驱动化疗耐药性”。
Pub Date : 2021-09-22 eCollection Date: 2021-09-01 DOI: 10.1093/narcan/zcab041
Adrian P Wiegmans, Ambber Ward, Ekaterina Ivanova, Pascal H G Duijf, Mark N Adams, Idris Mohd Najib, Romy Van Oosterhout, Martin C Sadowski, Greg Kelly, Scott W Morrical, Ken O'Byrne, Jason S Lee, Derek J Richard

[This corrects the article DOI: 10.1093/nar/zcab022.].

[这更正了文章DOI: 10.1093/nar/zcab022.]。
{"title":"Correction to 'Genome instability and pressure on non-homologous end joining drives chemotherapy resistance via a DNA repair crisis switch in triple negative breast cancer'.","authors":"Adrian P Wiegmans,&nbsp;Ambber Ward,&nbsp;Ekaterina Ivanova,&nbsp;Pascal H G Duijf,&nbsp;Mark N Adams,&nbsp;Idris Mohd Najib,&nbsp;Romy Van Oosterhout,&nbsp;Martin C Sadowski,&nbsp;Greg Kelly,&nbsp;Scott W Morrical,&nbsp;Ken O'Byrne,&nbsp;Jason S Lee,&nbsp;Derek J Richard","doi":"10.1093/narcan/zcab041","DOIUrl":"https://doi.org/10.1093/narcan/zcab041","url":null,"abstract":"<p><p>[This corrects the article DOI: 10.1093/nar/zcab022.].</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8457359/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39478379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SCISSOR™: a single-cell inferred site-specific omics resource for tumor microenvironment association study. SCISSOR™:用于肿瘤微环境关联研究的单细胞特异性位点组学资源。
Pub Date : 2021-09-09 eCollection Date: 2021-09-01 DOI: 10.1093/narcan/zcab037
Xiang Cui, Fei Qin, Xuanxuan Yu, Feifei Xiao, Guoshuai Cai

Tumor tissues are heterogeneous with different cell types in tumor microenvironment, which play an important role in tumorigenesis and tumor progression. Several computational algorithms and tools have been developed to infer the cell composition from bulk transcriptome profiles. However, they ignore the tissue specificity and thus a new resource for tissue-specific cell transcriptomic reference is needed for inferring cell composition in tumor microenvironment and exploring their association with clinical outcomes and tumor omics. In this study, we developed SCISSOR™ (https://thecailab.com/scissor/), an online open resource to fulfill that demand by integrating five orthogonal omics data of >6031 large-scale bulk samples, patient clinical outcomes and 451 917 high-granularity tissue-specific single-cell transcriptomic profiles of 16 cancer types. SCISSOR™ provides five major analysis modules that enable flexible modeling with adjustable parameters and dynamic visualization approaches. SCISSOR™ is valuable as a new resource for promoting tumor heterogeneity and tumor-tumor microenvironment cell interaction research, by delineating cells in the tissue-specific tumor microenvironment and characterizing their associations with tumor omics and clinical outcomes.

肿瘤微环境中肿瘤组织具有异质性,细胞类型不同,在肿瘤发生和发展中起着重要作用。已经开发了几种计算算法和工具来从大量转录组谱中推断细胞组成。然而,它们忽略了组织特异性,因此需要一种新的组织特异性细胞转录组参考资源来推断肿瘤微环境中的细胞组成,并探索它们与临床结果和肿瘤组学的关联。在这项研究中,我们开发了在线开放资源SCISSOR™(https://thecailab.com/scissor/),通过整合5个正交组学数据,包括6031个大规模样本、患者临床结果和451 917个16种癌症类型的高粒度组织特异性单细胞转录组学图谱,来满足这一需求。SCISSOR™提供了五个主要的分析模块,使灵活的建模与可调参数和动态可视化方法。通过描绘组织特异性肿瘤微环境中的细胞并表征其与肿瘤组学和临床结果的关联,SCISSOR™作为促进肿瘤异质性和肿瘤-肿瘤微环境细胞相互作用研究的新资源是有价值的。
{"title":"SCISSOR™: a single-cell inferred site-specific omics resource for tumor microenvironment association study.","authors":"Xiang Cui,&nbsp;Fei Qin,&nbsp;Xuanxuan Yu,&nbsp;Feifei Xiao,&nbsp;Guoshuai Cai","doi":"10.1093/narcan/zcab037","DOIUrl":"https://doi.org/10.1093/narcan/zcab037","url":null,"abstract":"<p><p>Tumor tissues are heterogeneous with different cell types in tumor microenvironment, which play an important role in tumorigenesis and tumor progression. Several computational algorithms and tools have been developed to infer the cell composition from bulk transcriptome profiles. However, they ignore the tissue specificity and thus a new resource for tissue-specific cell transcriptomic reference is needed for inferring cell composition in tumor microenvironment and exploring their association with clinical outcomes and tumor omics. In this study, we developed SCISSOR™ (https://thecailab.com/scissor/), an online open resource to fulfill that demand by integrating five orthogonal omics data of >6031 large-scale bulk samples, patient clinical outcomes and 451 917 high-granularity tissue-specific single-cell transcriptomic profiles of 16 cancer types. SCISSOR™ provides five major analysis modules that enable flexible modeling with adjustable parameters and dynamic visualization approaches. SCISSOR™ is valuable as a new resource for promoting tumor heterogeneity and tumor-tumor microenvironment cell interaction research, by delineating cells in the tissue-specific tumor microenvironment and characterizing their associations with tumor omics and clinical outcomes.</p>","PeriodicalId":18879,"journal":{"name":"NAR Cancer","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2021-09-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/f9/5d/zcab037.PMC8428296.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39410244","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
期刊
NAR Cancer
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