首页 > 最新文献

Epigenetics & Chromatin最新文献

英文 中文
The MMP-2 histone H3 N-terminal tail protease is selectively targeted to the transcription start sites of active genes. MMP-2 组蛋白 H3 N 端尾蛋白酶选择性地靶向活性基因的转录起始位点。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-05-10 DOI: 10.1186/s13072-023-00491-w
Benjamin H Weekley, Judd C Rice

Background: Proteolysis of the histone H3 N-terminal tail (H3NT) is an evolutionarily conserved epigenomic feature of nearly all eukaryotes, generating a cleaved H3 product that is retained in ~ 5-10% of the genome. Although H3NT proteolysis within chromatin was first reported over 60 years ago, the genomic sites targeted for H3NT proteolysis and the impact of this histone modification on chromatin structure and function remain largely unknown. The goal of this study was to identify the specific regions targeted for H3NT proteolysis and investigate the consequence of H3NT "clipping" on local histone post-translational modification (PTM) dynamics.

Results: Leveraging recent findings that matrix metalloproteinase 2 (MMP-2) functions as the principal nuclear H3NT protease in the human U2OS osteosarcoma cell line, a ChIP-Seq approach was used to map MMP-2 localization genome wide. The results indicate that MMP-2 is selectively targeted to the transcription start sites (TSSs) of protein coding genes, primarily at the + 1 nucleosome. MMP-2 localization was exclusive to highly expressed genes, further supporting a functional role for H3NT proteolysis in transcriptional regulation. MMP-2 dependent H3NT proteolysis at the TSSs of these genes resulted in a > twofold reduction of activation-associated histone H3 PTMs, including H3K4me3, H3K9ac and H3K18ac. One of genes requiring MMP-2 mediated H3NT proteolysis for proficient expression was the lysosomal cathepsin B protease (CTSB), which we discovered functions as a secondary nuclear H3NT protease in U2OS cells.

Conclusions: This study revealed that the MMP-2 H3NT protease is selectively targeted to the TSSs of active protein coding genes in U2OS cells. The resulting H3NT proteolysis directly alters local histone H3 PTM patterns at TSSs, which likely functions to regulate transcription. MMP-2 mediated H3NT proteolysis directly activates CTSB, a secondary H3NT protease that generates additional cleaved H3 products within chromatin.

背景:组蛋白 H3 N 端尾(H3NT)的蛋白水解是几乎所有真核生物在进化过程中保守的表观基因组特征,其产生的 H3 裂解产物保留在约 5-10% 的基因组中。虽然染色质内的 H3NT 蛋白水解在 60 多年前就被首次报道,但 H3NT 蛋白水解的基因组靶点以及这种组蛋白修饰对染色质结构和功能的影响在很大程度上仍不为人所知。本研究的目的是确定H3NT蛋白水解的特定靶区,并研究H3NT "剪切 "对局部组蛋白翻译后修饰(PTM)动态的影响:最近发现基质金属蛋白酶2(MMP-2)是人类U2OS骨肉瘤细胞系中主要的核H3NT蛋白酶,利用这一发现,我们采用ChIP-Seq方法绘制了MMP-2的全基因组定位图。结果表明,MMP-2 选择性地靶向于蛋白编码基因的转录起始位点(TSS),主要是在 + 1 核小体上。MMP-2 定位于高表达基因,进一步支持了 H3NT 蛋白水解在转录调控中的功能性作用。在这些基因的 TSS 处,依赖于 MMP-2 的 H3NT 蛋白分解导致活化相关的组蛋白 H3 PTMs(包括 H3K4me3、H3K9ac 和 H3K18ac)减少了两倍以上。需要MMP-2介导的H3NT蛋白水解才能熟练表达的基因之一是溶酶体酪蛋白B蛋白酶(CTSB),我们发现该蛋白酶在U2OS细胞中发挥着次级核H3NT蛋白酶的功能:本研究发现,在 U2OS 细胞中,MMP-2 H3NT 蛋白酶选择性地靶向活性蛋白编码基因的 TSSs。由此产生的 H3NT 蛋白水解直接改变了 TSSs 上的局部组蛋白 H3 PTM 模式,从而可能起到调节转录的作用。MMP-2 介导的 H3NT 蛋白水解可直接激活 CTSB,后者是一种次级 H3NT 蛋白酶,可在染色质中产生额外的裂解 H3 产物。
{"title":"The MMP-2 histone H3 N-terminal tail protease is selectively targeted to the transcription start sites of active genes.","authors":"Benjamin H Weekley, Judd C Rice","doi":"10.1186/s13072-023-00491-w","DOIUrl":"10.1186/s13072-023-00491-w","url":null,"abstract":"<p><strong>Background: </strong>Proteolysis of the histone H3 N-terminal tail (H3NT) is an evolutionarily conserved epigenomic feature of nearly all eukaryotes, generating a cleaved H3 product that is retained in ~ 5-10% of the genome. Although H3NT proteolysis within chromatin was first reported over 60 years ago, the genomic sites targeted for H3NT proteolysis and the impact of this histone modification on chromatin structure and function remain largely unknown. The goal of this study was to identify the specific regions targeted for H3NT proteolysis and investigate the consequence of H3NT \"clipping\" on local histone post-translational modification (PTM) dynamics.</p><p><strong>Results: </strong>Leveraging recent findings that matrix metalloproteinase 2 (MMP-2) functions as the principal nuclear H3NT protease in the human U2OS osteosarcoma cell line, a ChIP-Seq approach was used to map MMP-2 localization genome wide. The results indicate that MMP-2 is selectively targeted to the transcription start sites (TSSs) of protein coding genes, primarily at the + 1 nucleosome. MMP-2 localization was exclusive to highly expressed genes, further supporting a functional role for H3NT proteolysis in transcriptional regulation. MMP-2 dependent H3NT proteolysis at the TSSs of these genes resulted in a > twofold reduction of activation-associated histone H3 PTMs, including H3K4me3, H3K9ac and H3K18ac. One of genes requiring MMP-2 mediated H3NT proteolysis for proficient expression was the lysosomal cathepsin B protease (CTSB), which we discovered functions as a secondary nuclear H3NT protease in U2OS cells.</p><p><strong>Conclusions: </strong>This study revealed that the MMP-2 H3NT protease is selectively targeted to the TSSs of active protein coding genes in U2OS cells. The resulting H3NT proteolysis directly alters local histone H3 PTM patterns at TSSs, which likely functions to regulate transcription. MMP-2 mediated H3NT proteolysis directly activates CTSB, a secondary H3NT protease that generates additional cleaved H3 products within chromatin.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"16"},"PeriodicalIF":3.9,"publicationDate":"2023-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10170761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9523494","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Caf1 regulates the histone methyltransferase activity of Ash1 by sensing unmodified histone H3. Caf1通过感应未修饰的组蛋白H3来调节Ash1的组蛋白甲基转移酶活性。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-29 DOI: 10.1186/s13072-023-00487-6
Eojin Yoon, Ji-Joon Song

Histone modifications are one of the many key mechanisms that regulate gene expression. Ash1 is a histone H3K36 methyltransferase and is involved in gene activation. Ash1 forms a large complex with Mrg15 and Caf1/p55/Nurf55/RbAp48 (AMC complex). The Ash1 subunit alone exhibits very low activity due to the autoinhibition, and the binding of Mrg15 releases the autoinhibition. Caf1 is a scaffolding protein commonly found in several chromatin modifying complexes and has two histone binding pockets: one for H3 and the other for H4. Caf1 has the ability to sense unmodified histone H3K4 residues using the H3 binding pocket. However, the role of Caf1 in the AMC complex has not been investigated. Here, we dissected the interaction among the AMC complex subunits, revealing that Caf1 uses the histone H4 binding pocket to interact with Ash1 near the histone binding module cluster. Furthermore, we showed that H3K4 methylation inhibits AMC HMTase activity via Caf1 sensing unmodified histone H3K4 to regulate the activity in an internucleosomal manner, suggesting that crosstalk between H3K4 and H3K36 methylation. Our work revealed a delicate mechanism by which the AMC histone H3K36 methyltransferase complex is regulated.

组蛋白修饰是调控基因表达的众多关键机制之一。Ash1是一种组蛋白H3K36甲基转移酶,参与基因激活。Ash1与Mrg15和Caf1/p55/Nurf55/RbAp48形成一个大复合体(AMC复合体)。由于自身抑制作用,Ash1亚基单独表现出非常低的活性,Mrg15的结合释放了自抑制作用。Caf1是一种支架蛋白,常见于几种染色质修饰复合物中,具有两个组蛋白结合袋:一个用于H3,另一个用于H4。Caf1具有利用H3结合袋感知未修饰组蛋白H3K4残基的能力。然而,Caf1在AMC复合体中的作用尚未被研究。在这里,我们剖析了AMC复合体亚基之间的相互作用,揭示了Caf1使用组蛋白H4结合口袋与组蛋白结合模块簇附近的Ash1相互作用。此外,我们发现H3K4甲基化通过Caf1感知未修饰的组蛋白H3K4以核体间方式调节活性来抑制AMC HMTase活性,这表明H3K4和H3K36甲基化之间存在串扰。我们的工作揭示了一种微妙的机制,通过这种机制,AMC组蛋白H3K36甲基转移酶复合物受到调节。
{"title":"Caf1 regulates the histone methyltransferase activity of Ash1 by sensing unmodified histone H3.","authors":"Eojin Yoon,&nbsp;Ji-Joon Song","doi":"10.1186/s13072-023-00487-6","DOIUrl":"https://doi.org/10.1186/s13072-023-00487-6","url":null,"abstract":"<p><p>Histone modifications are one of the many key mechanisms that regulate gene expression. Ash1 is a histone H3K36 methyltransferase and is involved in gene activation. Ash1 forms a large complex with Mrg15 and Caf1/p55/Nurf55/RbAp48 (AMC complex). The Ash1 subunit alone exhibits very low activity due to the autoinhibition, and the binding of Mrg15 releases the autoinhibition. Caf1 is a scaffolding protein commonly found in several chromatin modifying complexes and has two histone binding pockets: one for H3 and the other for H4. Caf1 has the ability to sense unmodified histone H3K4 residues using the H3 binding pocket. However, the role of Caf1 in the AMC complex has not been investigated. Here, we dissected the interaction among the AMC complex subunits, revealing that Caf1 uses the histone H4 binding pocket to interact with Ash1 near the histone binding module cluster. Furthermore, we showed that H3K4 methylation inhibits AMC HMTase activity via Caf1 sensing unmodified histone H3K4 to regulate the activity in an internucleosomal manner, suggesting that crosstalk between H3K4 and H3K36 methylation. Our work revealed a delicate mechanism by which the AMC histone H3K36 methyltransferase complex is regulated.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"15"},"PeriodicalIF":3.9,"publicationDate":"2023-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10148413/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9455750","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Partial erosion on under-methylated regions and chromatin reprogramming contribute to oncogene activation in IDH mutant gliomas. 在IDH突变型胶质瘤中,低甲基化区域的部分侵蚀和染色质重编程有助于癌基因激活。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-28 DOI: 10.1186/s13072-023-00490-x
Xinyu Wang, Lijun Dai, Yang Liu, Chenghao Li, Dandan Fan, Yue Zhou, Pengcheng Li, Qingran Kong, Jianzhong Su

Background: IDH1/2 hotspot mutations are well known to drive oncogenic mutations in gliomas and are well-defined in the WHO 2021 classification of central nervous system tumors. Specifically, IDH mutations lead to aberrant hypermethylation of under-methylated regions (UMRs) in normal tissues through the disruption of TET enzymes. However, the chromatin reprogramming and transcriptional changes induced by IDH-related hypermethylation in gliomas remain unclear.

Results: Here, we have developed a precise computational framework based on Hidden Markov Model to identify altered methylation states of UMRs at single-base resolution. By applying this framework to whole-genome bisulfite sequencing data from 75 normal brain tissues and 15 IDH mutant glioma tissues, we identified two distinct types of hypermethylated UMRs in IDH mutant gliomas. We named them partially hypermethylated UMRs (phUMRs) and fully hypermethylated UMRs (fhUMRs), respectively. We found that the phUMRs and fhUMRs exhibit distinct genomic features and chromatin states. Genes related to fhUMRs were more likely to be repressed in IDH mutant gliomas. In contrast, genes related to phUMRs were prone to be up-regulated in IDH mutant gliomas. Such activation of phUMR genes is associated with the accumulation of active H3K4me3 and the loss of H3K27me3, as well as H3K36me3 accumulation in gene bodies to maintain gene expression stability. In summary, partial erosion on UMRs was accompanied by locus-specific changes in key chromatin marks, which may contribute to oncogene activation.

Conclusions: Our study provides a computational strategy for precise decoding of methylation encroachment patterns in IDH mutant gliomas, revealing potential mechanistic insights into chromatin reprogramming that contribute to oncogenesis.

背景:IDH1/2热点突变在胶质瘤中驱动致癌突变是众所周知的,并且在WHO 2021中枢神经系统肿瘤分类中被明确定义。具体来说,IDH突变通过TET酶的破坏导致正常组织中低甲基化区域(UMRs)的异常高甲基化。然而,在胶质瘤中由idh相关的高甲基化引起的染色质重编程和转录变化尚不清楚。在这里,我们开发了一个基于隐马尔可夫模型的精确计算框架,用于在单碱基分辨率下识别umr甲基化状态的改变。通过将这一框架应用于来自75个正常脑组织和15个IDH突变胶质瘤组织的全基因组亚硫酸盐测序数据,我们在IDH突变胶质瘤中发现了两种不同类型的高甲基化UMRs。我们将它们分别命名为部分高甲基化UMRs (phUMRs)和完全高甲基化UMRs (fhUMRs)。我们发现phUMRs和fhUMRs表现出不同的基因组特征和染色质状态。与fhUMRs相关的基因在IDH突变型胶质瘤中更有可能被抑制。相反,与phUMRs相关的基因在IDH突变型胶质瘤中容易上调。phUMR基因的这种激活与活性H3K4me3的积累和H3K27me3的丧失以及H3K36me3在基因体内的积累有关,以维持基因表达的稳定性。总之,umr上的部分侵蚀伴随着关键染色质标记的位点特异性变化,这可能有助于癌基因的激活。结论:我们的研究为IDH突变胶质瘤中甲基化侵犯模式的精确解码提供了一种计算策略,揭示了染色质重编程促进肿瘤发生的潜在机制。
{"title":"Partial erosion on under-methylated regions and chromatin reprogramming contribute to oncogene activation in IDH mutant gliomas.","authors":"Xinyu Wang,&nbsp;Lijun Dai,&nbsp;Yang Liu,&nbsp;Chenghao Li,&nbsp;Dandan Fan,&nbsp;Yue Zhou,&nbsp;Pengcheng Li,&nbsp;Qingran Kong,&nbsp;Jianzhong Su","doi":"10.1186/s13072-023-00490-x","DOIUrl":"https://doi.org/10.1186/s13072-023-00490-x","url":null,"abstract":"<p><strong>Background: </strong>IDH1/2 hotspot mutations are well known to drive oncogenic mutations in gliomas and are well-defined in the WHO 2021 classification of central nervous system tumors. Specifically, IDH mutations lead to aberrant hypermethylation of under-methylated regions (UMRs) in normal tissues through the disruption of TET enzymes. However, the chromatin reprogramming and transcriptional changes induced by IDH-related hypermethylation in gliomas remain unclear.</p><p><strong>Results: </strong>Here, we have developed a precise computational framework based on Hidden Markov Model to identify altered methylation states of UMRs at single-base resolution. By applying this framework to whole-genome bisulfite sequencing data from 75 normal brain tissues and 15 IDH mutant glioma tissues, we identified two distinct types of hypermethylated UMRs in IDH mutant gliomas. We named them partially hypermethylated UMRs (phUMRs) and fully hypermethylated UMRs (fhUMRs), respectively. We found that the phUMRs and fhUMRs exhibit distinct genomic features and chromatin states. Genes related to fhUMRs were more likely to be repressed in IDH mutant gliomas. In contrast, genes related to phUMRs were prone to be up-regulated in IDH mutant gliomas. Such activation of phUMR genes is associated with the accumulation of active H3K4me3 and the loss of H3K27me3, as well as H3K36me3 accumulation in gene bodies to maintain gene expression stability. In summary, partial erosion on UMRs was accompanied by locus-specific changes in key chromatin marks, which may contribute to oncogene activation.</p><p><strong>Conclusions: </strong>Our study provides a computational strategy for precise decoding of methylation encroachment patterns in IDH mutant gliomas, revealing potential mechanistic insights into chromatin reprogramming that contribute to oncogenesis.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"13"},"PeriodicalIF":3.9,"publicationDate":"2023-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10142198/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9455749","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An optimized approach for multiplexing single-nuclear ATAC-seq using oligonucleotide-conjugated antibodies. 使用寡核苷酸连接抗体进行单核 ATAC-seq 多路复用的优化方法。
IF 4.2 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-28 DOI: 10.1186/s13072-023-00486-7
Betelehem Solomon Bera, Taylor V Thompson, Eric Sosa, Hiroko Nomaru, David Reynolds, Robert A Dubin, Shahina B Maqbool, Deyou Zheng, Bernice E Morrow, John M Greally, Masako Suzuki

Background: Single-cell technologies to analyze transcription and chromatin structure have been widely used in many research areas to reveal the functions and molecular properties of cells at single-cell resolution. Sample multiplexing techniques are valuable when performing single-cell analysis, reducing technical variation and permitting cost efficiencies. Several commercially available methods have been used in many scRNA-seq studies. On the other hand, while several methods have been published, multiplexing techniques for single nuclear assay for transposase-accessible chromatin (snATAC)-seq assays remain under development. We developed a simple nucleus hashing method using oligonucleotide-conjugated antibodies recognizing nuclear pore complex proteins, NuHash, to perform snATAC-seq library preparations by multiplexing.

Results: We performed multiplexing snATAC-seq analyses on a mixture of human and mouse cell samples (two samples, 2-plex, and four samples, 4-plex) using NuHash. The analyses on nuclei with at least 10,000 read counts showed that the demultiplexing accuracy of NuHash was high, and only ten out of 9144 nuclei (2-plex) and 150 of 12,208 nuclei (4-plex) had discordant classifications between NuHash demultiplexing and discrimination using reference genome alignments. The differential open chromatin region (OCR) analysis between female and male samples revealed that male-specific OCRs were enriched in chromosome Y (four out of nine). We also found that five female-specific OCRs (20 OCRs) were on chromosome X. A comparative analysis between snATAC-seq and deeply sequenced bulk ATAC-seq on the same samples revealed that the bulk ATAC-seq signal intensity was positively correlated with the number of cell clusters detected in snATAC-seq. Moreover, when we categorized snATAC-seq peaks based on the number of cell clusters in which the peak was present, we observed different distributions over different genomic features between the groups. This result suggests that the peak intensities of bulk ATAC-seq can be used to identify different types of functional loci.

Conclusions: Our multiplexing method using oligo-conjugated anti-nuclear pore complex proteins, NuHash, permits high-accuracy demultiplexing of samples. The NuHash protocol is straightforward, works on frozen samples, and requires no modifications for snATAC-seq library preparation.

背景:分析转录和染色质结构的单细胞技术已广泛应用于许多研究领域,以单细胞分辨率揭示细胞的功能和分子特性。在进行单细胞分析时,样品复用技术非常重要,它能减少技术差异并提高成本效益。许多 scRNA-seq 研究都采用了几种市售方法。另一方面,虽然有几种方法已经发表,但用于转座酶可接触染色质(snATAC)-seq 检测的单核检测多路复用技术仍在开发中。我们利用识别核孔复合蛋白的寡核苷酸连接抗体--NuHash--开发了一种简单的核散列方法,通过多路复用进行snATAC-seq文库制备:我们利用NuHash对人类和小鼠的混合细胞样本(2个样本,2-plex;4个样本,4-plex)进行了多重snATAC-seq分析。对至少有 10,000 个读数的细胞核进行的分析表明,NuHash 的解复用准确率很高,在 9144 个细胞核(2-plex)和 12,208 个细胞核(4-plex)中,只有 10 个细胞核(2-plex)和 150 个细胞核(4-plex)的 NuHash 解复用分类与使用参考基因组比对的判别不一致。雌性和雄性样本的开放染色质区(OCR)差异分析表明,男性特异性 OCR 在 Y 染色体中富集(9 个中的 4 个)。对同一样本进行的 snATAC-seq 和深度测序的大量 ATAC-seq 比较分析表明,大量 ATAC-seq 信号强度与 snATAC-seq 中检测到的细胞簇数量呈正相关。此外,当我们根据出现峰值的细胞簇数量对 snATAC-seq 峰值进行分类时,我们观察到不同组间不同基因组特征的分布情况不同。这一结果表明,批量ATAC-seq的峰强度可用于鉴定不同类型的功能位点:结论:我们使用寡聚抗核孔复合蛋白的多路复用方法 NuHash 可以对样本进行高精度的解多路复用。NuHash 方案简单明了,适用于冷冻样本,且无需对 snATAC-seq 文库制备进行修改。
{"title":"An optimized approach for multiplexing single-nuclear ATAC-seq using oligonucleotide-conjugated antibodies.","authors":"Betelehem Solomon Bera, Taylor V Thompson, Eric Sosa, Hiroko Nomaru, David Reynolds, Robert A Dubin, Shahina B Maqbool, Deyou Zheng, Bernice E Morrow, John M Greally, Masako Suzuki","doi":"10.1186/s13072-023-00486-7","DOIUrl":"10.1186/s13072-023-00486-7","url":null,"abstract":"<p><strong>Background: </strong>Single-cell technologies to analyze transcription and chromatin structure have been widely used in many research areas to reveal the functions and molecular properties of cells at single-cell resolution. Sample multiplexing techniques are valuable when performing single-cell analysis, reducing technical variation and permitting cost efficiencies. Several commercially available methods have been used in many scRNA-seq studies. On the other hand, while several methods have been published, multiplexing techniques for single nuclear assay for transposase-accessible chromatin (snATAC)-seq assays remain under development. We developed a simple nucleus hashing method using oligonucleotide-conjugated antibodies recognizing nuclear pore complex proteins, NuHash, to perform snATAC-seq library preparations by multiplexing.</p><p><strong>Results: </strong>We performed multiplexing snATAC-seq analyses on a mixture of human and mouse cell samples (two samples, 2-plex, and four samples, 4-plex) using NuHash. The analyses on nuclei with at least 10,000 read counts showed that the demultiplexing accuracy of NuHash was high, and only ten out of 9144 nuclei (2-plex) and 150 of 12,208 nuclei (4-plex) had discordant classifications between NuHash demultiplexing and discrimination using reference genome alignments. The differential open chromatin region (OCR) analysis between female and male samples revealed that male-specific OCRs were enriched in chromosome Y (four out of nine). We also found that five female-specific OCRs (20 OCRs) were on chromosome X. A comparative analysis between snATAC-seq and deeply sequenced bulk ATAC-seq on the same samples revealed that the bulk ATAC-seq signal intensity was positively correlated with the number of cell clusters detected in snATAC-seq. Moreover, when we categorized snATAC-seq peaks based on the number of cell clusters in which the peak was present, we observed different distributions over different genomic features between the groups. This result suggests that the peak intensities of bulk ATAC-seq can be used to identify different types of functional loci.</p><p><strong>Conclusions: </strong>Our multiplexing method using oligo-conjugated anti-nuclear pore complex proteins, NuHash, permits high-accuracy demultiplexing of samples. The NuHash protocol is straightforward, works on frozen samples, and requires no modifications for snATAC-seq library preparation.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"14"},"PeriodicalIF":4.2,"publicationDate":"2023-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10142415/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9499757","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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 modifier alpha-ketoglutarate modulates aberrant gene body methylation and hydroxymethylation marks in diabetic heart. 表观遗传修饰因子α -酮戊二酸调节糖尿病心脏异常基因体甲基化和羟甲基化标记。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-27 DOI: 10.1186/s13072-023-00489-4
Rohini Dhat, Dattatray Mongad, Sivarupa Raji, Silpa Arkat, Nitish R Mahapatra, Nishant Singhal, Sandhya L Sitasawad

Background: Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Hyperglycemic myocardial microenvironment significantly alters chromatin architecture and the transcriptome, resulting in aberrant activation of signaling pathways in a diabetic heart. Epigenetic marks play vital roles in transcriptional reprogramming during the development of DCM. The current study is aimed to profile genome-wide DNA (hydroxy)methylation patterns in the hearts of control and streptozotocin (STZ)-induced diabetic rats and decipher the effect of modulation of DNA methylation by alpha-ketoglutarate (AKG), a TET enzyme cofactor, on the progression of DCM.

Methods: Diabetes was induced in male adult Wistar rats with an intraperitoneal injection of STZ. Diabetic and vehicle control animals were randomly divided into groups with/without AKG treatment. Cardiac function was monitored by performing cardiac catheterization. Global methylation (5mC) and hydroxymethylation (5hmC) patterns were mapped in the Left ventricular tissue of control and diabetic rats with the help of an enrichment-based (h)MEDIP-sequencing technique by using antibodies specific for 5mC and 5hmC. Sequencing data were validated by performing (h)MEDIP-qPCR analysis at the gene-specific level, and gene expression was analyzed by qPCR. The mRNA and protein expression of enzymes involved in the DNA methylation and demethylation cycle were analyzed by qPCR and western blotting. Global 5mC and 5hmC levels were also assessed in high glucose-treated DNMT3B knockdown H9c2 cells.

Results: We found the increased expression of DNMT3B, MBD2, and MeCP2 with a concomitant accumulation of 5mC and 5hmC, specifically in gene body regions of diabetic rat hearts compared to the control. Calcium signaling was the most significantly affected pathway by cytosine modifications in the diabetic heart. Additionally, hypermethylated gene body regions were associated with Rap1, apelin, and phosphatidyl inositol signaling, while metabolic pathways were most affected by hyperhydroxymethylation. AKG supplementation in diabetic rats reversed aberrant methylation patterns and restored cardiac function. Hyperglycemia also increased 5mC and 5hmC levels in H9c2 cells, which was normalized by DNMT3B knockdown or AKG supplementation.

Conclusion: This study demonstrates that reverting hyperglycemic damage to cardiac tissue might be possible by erasing adverse epigenetic signatures by supplementing epigenetic modulators such as AKG along with an existing antidiabetic treatment regimen.

背景:糖尿病性心肌病(DCM)是糖尿病患者死亡的主要原因。高血糖心肌微环境显著改变染色质结构和转录组,导致糖尿病心脏信号通路异常激活。表观遗传标记在DCM发育过程中的转录重编程中起着至关重要的作用。本研究旨在分析对照组和链脲佐菌素(STZ)诱导的糖尿病大鼠心脏全基因组DNA(羟基)甲基化模式,并破译TET酶辅助因子α -酮戊二酸(AKG)对DCM进展的DNA甲基化调节作用。方法:用STZ腹腔注射诱导雄性成年Wistar大鼠糖尿病。糖尿病动物和对照动物随机分为AKG治疗组和未治疗组。心导管术监测心功能。利用5mC和5hmC特异性抗体,利用基于富集的(h) medip测序技术,在对照和糖尿病大鼠左心室组织中绘制了全球甲基化(5mC)和羟甲基化(5hmC)模式。通过(h)基因特异性水平的MEDIP-qPCR分析验证测序数据,并通过qPCR分析基因表达。采用qPCR和western blotting分析DNA甲基化和去甲基化周期相关酶的mRNA和蛋白表达。在高糖处理的DNMT3B敲低的H9c2细胞中,也评估了总体5mC和5hmC水平。结果:与对照组相比,我们发现DNMT3B、MBD2和MeCP2的表达增加,并伴有5mC和5hmC的积累,特别是在糖尿病大鼠心脏的基因体区域。在糖尿病心脏中,胞嘧啶修饰对钙信号通路的影响最为显著。此外,高甲基化的基因体区域与Rap1、apelin和磷脂酰肌醇信号相关,而代谢途径受高羟甲基化的影响最大。糖尿病大鼠补充AKG可逆转异常甲基化模式并恢复心功能。高血糖也增加了H9c2细胞中的5mC和5hmC水平,通过敲除DNMT3B或补充AKG使其正常化。结论:本研究表明,通过补充表观遗传调节剂(如AKG)和现有的抗糖尿病治疗方案来消除不良的表观遗传特征,可能会恢复高血糖对心脏组织的损害。
{"title":"Epigenetic modifier alpha-ketoglutarate modulates aberrant gene body methylation and hydroxymethylation marks in diabetic heart.","authors":"Rohini Dhat,&nbsp;Dattatray Mongad,&nbsp;Sivarupa Raji,&nbsp;Silpa Arkat,&nbsp;Nitish R Mahapatra,&nbsp;Nishant Singhal,&nbsp;Sandhya L Sitasawad","doi":"10.1186/s13072-023-00489-4","DOIUrl":"https://doi.org/10.1186/s13072-023-00489-4","url":null,"abstract":"<p><strong>Background: </strong>Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Hyperglycemic myocardial microenvironment significantly alters chromatin architecture and the transcriptome, resulting in aberrant activation of signaling pathways in a diabetic heart. Epigenetic marks play vital roles in transcriptional reprogramming during the development of DCM. The current study is aimed to profile genome-wide DNA (hydroxy)methylation patterns in the hearts of control and streptozotocin (STZ)-induced diabetic rats and decipher the effect of modulation of DNA methylation by alpha-ketoglutarate (AKG), a TET enzyme cofactor, on the progression of DCM.</p><p><strong>Methods: </strong>Diabetes was induced in male adult Wistar rats with an intraperitoneal injection of STZ. Diabetic and vehicle control animals were randomly divided into groups with/without AKG treatment. Cardiac function was monitored by performing cardiac catheterization. Global methylation (5mC) and hydroxymethylation (5hmC) patterns were mapped in the Left ventricular tissue of control and diabetic rats with the help of an enrichment-based (h)MEDIP-sequencing technique by using antibodies specific for 5mC and 5hmC. Sequencing data were validated by performing (h)MEDIP-qPCR analysis at the gene-specific level, and gene expression was analyzed by qPCR. The mRNA and protein expression of enzymes involved in the DNA methylation and demethylation cycle were analyzed by qPCR and western blotting. Global 5mC and 5hmC levels were also assessed in high glucose-treated DNMT3B knockdown H9c2 cells.</p><p><strong>Results: </strong>We found the increased expression of DNMT3B, MBD2, and MeCP2 with a concomitant accumulation of 5mC and 5hmC, specifically in gene body regions of diabetic rat hearts compared to the control. Calcium signaling was the most significantly affected pathway by cytosine modifications in the diabetic heart. Additionally, hypermethylated gene body regions were associated with Rap1, apelin, and phosphatidyl inositol signaling, while metabolic pathways were most affected by hyperhydroxymethylation. AKG supplementation in diabetic rats reversed aberrant methylation patterns and restored cardiac function. Hyperglycemia also increased 5mC and 5hmC levels in H9c2 cells, which was normalized by DNMT3B knockdown or AKG supplementation.</p><p><strong>Conclusion: </strong>This study demonstrates that reverting hyperglycemic damage to cardiac tissue might be possible by erasing adverse epigenetic signatures by supplementing epigenetic modulators such as AKG along with an existing antidiabetic treatment regimen.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"12"},"PeriodicalIF":3.9,"publicationDate":"2023-04-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10134649/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9751745","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ovulatory signal-triggered chromatin remodeling in ovarian granulosa cells by HDAC2 phosphorylation activation-mediated histone deacetylation. 通过HDAC2磷酸化激活介导的组蛋白去乙酰化,排卵信号触发卵巢颗粒细胞染色质重塑。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-19 DOI: 10.1186/s13072-023-00485-8
Jiamin Jin, Peipei Ren, Xiang Li, Yinyi Zhang, Weijie Yang, Yerong Ma, Mengru Lai, Chao Yu, Songying Zhang, Yin-Li Zhang

Background: Epigenetic reprogramming is involved in luteinizing hormone (LH)-induced ovulation; however, the underlying mechanisms are largely unknown.

Results: We here observed a rapid histone deacetylation process between two waves of active transcription mediated by the follicle-stimulating hormone (FSH) and the LH congener human chorionic gonadotropin (hCG), respectively. Analysis of the genome-wide H3K27Ac distribution in hCG-treated granulosa cells revealed that a rapid wave of genome-wide histone deacetylation remodels the chromatin, followed by the establishment of specific histone acetylation for ovulation. HDAC2 phosphorylation activation coincides with histone deacetylation in mouse preovulatory follicles. When HDAC2 was silenced or inhibited, histone acetylation was retained, leading to reduced gene transcription, retarded cumulus expansion, and ovulation defect. HDAC2 phosphorylation was associated with CK2α nuclear translocation, and inhibition of CK2α attenuated HDAC2 phosphorylation, retarded H3K27 deacetylation, and inactivated the ERK1/2 signaling cascade.

Conclusions: This study demonstrates that the ovulatory signal erases histone acetylation through activation of CK2α-mediated HDAC2 phosphorylation in granulosa cells, which is an essential prerequisite for subsequent successful ovulation.

背景:表观遗传重编程参与黄体生成素(LH)诱导的排卵;然而,潜在的机制在很大程度上是未知的。结果:我们在这里分别观察到促卵泡激素(FSH)和LH同系物人绒毛膜促性腺激素(hCG)介导的两波活性转录之间的快速组蛋白去乙酰化过程。对hcg处理的颗粒细胞全基因组H3K27Ac分布的分析显示,全基因组组蛋白去乙酰化的快速波重塑了染色质,随后建立了特异性组蛋白乙酰化的排卵。小鼠排卵前卵泡中HDAC2磷酸化激活与组蛋白去乙酰化一致。当HDAC2被沉默或抑制时,组蛋白乙酰化被保留,导致基因转录减少、积云扩张迟缓和排卵缺陷。HDAC2磷酸化与CK2α核易位相关,抑制CK2α可减弱HDAC2磷酸化,延缓H3K27去乙酰化,并使ERK1/2信号级联失活。结论:本研究表明,排卵信号通过激活颗粒细胞中ck2 α-介导的HDAC2磷酸化来消除组蛋白乙酰化,这是随后成功排卵的必要前提。
{"title":"Ovulatory signal-triggered chromatin remodeling in ovarian granulosa cells by HDAC2 phosphorylation activation-mediated histone deacetylation.","authors":"Jiamin Jin,&nbsp;Peipei Ren,&nbsp;Xiang Li,&nbsp;Yinyi Zhang,&nbsp;Weijie Yang,&nbsp;Yerong Ma,&nbsp;Mengru Lai,&nbsp;Chao Yu,&nbsp;Songying Zhang,&nbsp;Yin-Li Zhang","doi":"10.1186/s13072-023-00485-8","DOIUrl":"https://doi.org/10.1186/s13072-023-00485-8","url":null,"abstract":"<p><strong>Background: </strong>Epigenetic reprogramming is involved in luteinizing hormone (LH)-induced ovulation; however, the underlying mechanisms are largely unknown.</p><p><strong>Results: </strong>We here observed a rapid histone deacetylation process between two waves of active transcription mediated by the follicle-stimulating hormone (FSH) and the LH congener human chorionic gonadotropin (hCG), respectively. Analysis of the genome-wide H3K27Ac distribution in hCG-treated granulosa cells revealed that a rapid wave of genome-wide histone deacetylation remodels the chromatin, followed by the establishment of specific histone acetylation for ovulation. HDAC2 phosphorylation activation coincides with histone deacetylation in mouse preovulatory follicles. When HDAC2 was silenced or inhibited, histone acetylation was retained, leading to reduced gene transcription, retarded cumulus expansion, and ovulation defect. HDAC2 phosphorylation was associated with CK2α nuclear translocation, and inhibition of CK2α attenuated HDAC2 phosphorylation, retarded H3K27 deacetylation, and inactivated the ERK1/2 signaling cascade.</p><p><strong>Conclusions: </strong>This study demonstrates that the ovulatory signal erases histone acetylation through activation of CK2α-mediated HDAC2 phosphorylation in granulosa cells, which is an essential prerequisite for subsequent successful ovulation.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"11"},"PeriodicalIF":3.9,"publicationDate":"2023-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10116676/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9420852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Multilevel interrogation of H3.3 reveals a primordial role in transcription regulation. H3.3的多水平询问揭示了其在转录调控中的原始作用。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-04-07 DOI: 10.1186/s13072-023-00484-9
Syed Nabeel-Shah, Jyoti Garg, Kanwal Ashraf, Renu Jeyapala, Hyunmin Lee, Alexandra Petrova, James D Burns, Shuye Pu, Zhaolei Zhang, Jack F Greenblatt, Ronald E Pearlman, Jean-Philippe Lambert, Jeffrey Fillingham

Background: Eukaryotic cells can rapidly adjust their transcriptional profile in response to molecular needs. Such dynamic regulation is, in part, achieved through epigenetic modifications and selective incorporation of histone variants into chromatin. H3.3 is the ancestral H3 variant with key roles in regulating chromatin states and transcription. Although H3.3 has been well studied in metazoans, information regarding the assembly of H3.3 onto chromatin and its possible role in transcription regulation remain poorly documented outside of Opisthokonts.

Results: We used the nuclear dimorphic ciliate protozoan, Tetrahymena thermophila, to investigate the dynamics of H3 variant function in evolutionarily divergent eukaryotes. Functional proteomics and immunofluorescence analyses of H3.1 and H3.3 revealed a highly conserved role for Nrp1 and Asf1 histone chaperones in nuclear influx of histones. Cac2, a putative subunit of H3.1 deposition complex CAF1, is not required for growth, whereas the expression of the putative ortholog of the H3.3-specific chaperone Hir1 is essential in Tetrahymena. Our results indicate that Cac2 and Hir1 have distinct localization patterns during different stages of the Tetrahymena life cycle and suggest that Cac2 might be dispensable for chromatin assembly. ChIP-seq experiments in growing Tetrahymena show H3.3 enrichment over the promoters, gene bodies, and transcription termination sites of highly transcribed genes. H3.3 knockout followed by RNA-seq reveals large-scale transcriptional alterations in functionally important genes.

Conclusion: Our results provide an evolutionary perspective on H3.3's conserved role in maintaining the transcriptional landscape of cells and on the emergence of specialized chromatin assembly pathways.

背景:真核细胞可以根据分子需要快速调整其转录谱。这种动态调节部分是通过表观遗传修饰和选择性地将组蛋白变体结合到染色质中来实现的。H3.3是祖先的H3变异,在调节染色质状态和转录中起关键作用。尽管H3.3在后生动物中已经得到了很好的研究,但关于H3.3在染色质上的组装及其在转录调节中的可能作用的信息在Opisthokonts之外仍然很少有文献记载。结果:我们利用核二态纤毛虫原生动物嗜热四膜虫(Tetrahymena thermomophila)研究了进化分歧真核生物中H3变异功能的动态变化。H3.1和H3.3的功能蛋白质组学和免疫荧光分析显示,Nrp1和Asf1组蛋白伴侣蛋白在组蛋白核内流中的作用高度保守。Cac2是H3.1沉积复合体CAF1的一个假定的亚基,它不是生长所必需的,而在四膜虫中,h3.3特异性伴侣蛋白Hir1的假定同源物的表达是必不可少的。我们的研究结果表明,在四膜虫生命周期的不同阶段,Cac2和Hir1具有不同的定位模式,这表明Cac2可能在染色质组装中是必不可少的。在生长的四膜虫中进行的ChIP-seq实验显示,H3.3在高转录基因的启动子、基因体和转录终止位点富集。H3.3基因敲除后进行RNA-seq分析,揭示了功能重要基因的大规模转录改变。结论:我们的研究结果为H3.3在维持细胞转录景观中的保守作用以及特殊染色质组装途径的出现提供了进化视角。
{"title":"Multilevel interrogation of H3.3 reveals a primordial role in transcription regulation.","authors":"Syed Nabeel-Shah,&nbsp;Jyoti Garg,&nbsp;Kanwal Ashraf,&nbsp;Renu Jeyapala,&nbsp;Hyunmin Lee,&nbsp;Alexandra Petrova,&nbsp;James D Burns,&nbsp;Shuye Pu,&nbsp;Zhaolei Zhang,&nbsp;Jack F Greenblatt,&nbsp;Ronald E Pearlman,&nbsp;Jean-Philippe Lambert,&nbsp;Jeffrey Fillingham","doi":"10.1186/s13072-023-00484-9","DOIUrl":"https://doi.org/10.1186/s13072-023-00484-9","url":null,"abstract":"<p><strong>Background: </strong>Eukaryotic cells can rapidly adjust their transcriptional profile in response to molecular needs. Such dynamic regulation is, in part, achieved through epigenetic modifications and selective incorporation of histone variants into chromatin. H3.3 is the ancestral H3 variant with key roles in regulating chromatin states and transcription. Although H3.3 has been well studied in metazoans, information regarding the assembly of H3.3 onto chromatin and its possible role in transcription regulation remain poorly documented outside of Opisthokonts.</p><p><strong>Results: </strong>We used the nuclear dimorphic ciliate protozoan, Tetrahymena thermophila, to investigate the dynamics of H3 variant function in evolutionarily divergent eukaryotes. Functional proteomics and immunofluorescence analyses of H3.1 and H3.3 revealed a highly conserved role for Nrp1 and Asf1 histone chaperones in nuclear influx of histones. Cac2, a putative subunit of H3.1 deposition complex CAF1, is not required for growth, whereas the expression of the putative ortholog of the H3.3-specific chaperone Hir1 is essential in Tetrahymena. Our results indicate that Cac2 and Hir1 have distinct localization patterns during different stages of the Tetrahymena life cycle and suggest that Cac2 might be dispensable for chromatin assembly. ChIP-seq experiments in growing Tetrahymena show H3.3 enrichment over the promoters, gene bodies, and transcription termination sites of highly transcribed genes. H3.3 knockout followed by RNA-seq reveals large-scale transcriptional alterations in functionally important genes.</p><p><strong>Conclusion: </strong>Our results provide an evolutionary perspective on H3.3's conserved role in maintaining the transcriptional landscape of cells and on the emergence of specialized chromatin assembly pathways.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"10"},"PeriodicalIF":3.9,"publicationDate":"2023-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10080907/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9399099","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PCGF6/MAX/KDM5D facilitates MAZ/CDK4 axis expression and pRCC progression by hypomethylation of the DNA promoter. PCGF6/MAX/KDM5D通过DNA启动子的低甲基化促进MAZ/CDK4轴表达和pRCC进展。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-03-09 DOI: 10.1186/s13072-023-00483-w
Meng Zhu, Ruo-Nan Zhang, Hong Zhang, Chang-Bao Qu, Xiao-Chong Zhang, Li-Xin Ren, Zhan Yang, Jun-Fei Gu

Polycomb group RING finger protein 6 (PCGF6) plays an important role as a regulator of transcription in a variety of cellular processes, including tumorigenesis. However, the function and expression of PCGF6 in papillary RCC (pRCC) remain unclear. In the present study, we found that PCGF6 expression was significantly elevated in pRCC tissues, and high expression of PCGF6 was associated with poor survival of patients with pRCC. The overexpression of PCGF6 promoted while depletion of PCGF6 depressed the proliferation of pRCC cells in vitro. Interestingly, myc-related zinc finger protein (MAZ), a downstream molecular of PCGF6, was upregulated in pRCC with hypomethylation promoter. Mechanically, PCGF6 promoted MAZ expression by interacting with MAX and KDM5D to form a complex, and MAX recruited PCGF6 and KDM5D to the CpG island of the MAZ promoter and facilitated H3K4 histone demethylation. Furthermore, CDK4 was a downstream molecule of MAZ that participated in PCGF6/MAZ-regulated progression of pRCC. These results indicated that the upregulation of PCGF6 facilitated MAZ/CDK4 axis expression and pRCC progression by hypomethylation of the MAZ promoter. The PCGF6/MAZ/CDK4 regulatory axis may be a potential target for the treatment of ccRCC.

Polycomb group RING finger protein 6 (PCGF6)在包括肿瘤发生在内的多种细胞过程中作为转录调节因子发挥着重要作用。然而,PCGF6在乳头状RCC (pRCC)中的功能和表达尚不清楚。在本研究中,我们发现PCGF6在pRCC组织中的表达显著升高,且PCGF6的高表达与pRCC患者的生存不良相关。在体外实验中,PCGF6过表达促进了pRCC细胞的增殖,而PCGF6缺失抑制了pRCC细胞的增殖。有趣的是,PCGF6的下游分子myc相关锌指蛋白(MAZ)在pRCC中通过低甲基化启动子上调。机制上,PCGF6通过与MAX和KDM5D相互作用形成复合物促进MAZ表达,MAX将PCGF6和KDM5D招募到MAZ启动子的CpG岛,促进H3K4组蛋白去甲基化。此外,CDK4是MAZ的下游分子,参与PCGF6/MAZ调控的pRCC进展。这些结果表明,PCGF6的上调通过MAZ启动子的低甲基化促进了MAZ/CDK4轴的表达和pRCC的进展。PCGF6/MAZ/CDK4调控轴可能是治疗ccRCC的潜在靶点。
{"title":"PCGF6/MAX/KDM5D facilitates MAZ/CDK4 axis expression and pRCC progression by hypomethylation of the DNA promoter.","authors":"Meng Zhu,&nbsp;Ruo-Nan Zhang,&nbsp;Hong Zhang,&nbsp;Chang-Bao Qu,&nbsp;Xiao-Chong Zhang,&nbsp;Li-Xin Ren,&nbsp;Zhan Yang,&nbsp;Jun-Fei Gu","doi":"10.1186/s13072-023-00483-w","DOIUrl":"https://doi.org/10.1186/s13072-023-00483-w","url":null,"abstract":"<p><p>Polycomb group RING finger protein 6 (PCGF6) plays an important role as a regulator of transcription in a variety of cellular processes, including tumorigenesis. However, the function and expression of PCGF6 in papillary RCC (pRCC) remain unclear. In the present study, we found that PCGF6 expression was significantly elevated in pRCC tissues, and high expression of PCGF6 was associated with poor survival of patients with pRCC. The overexpression of PCGF6 promoted while depletion of PCGF6 depressed the proliferation of pRCC cells in vitro. Interestingly, myc-related zinc finger protein (MAZ), a downstream molecular of PCGF6, was upregulated in pRCC with hypomethylation promoter. Mechanically, PCGF6 promoted MAZ expression by interacting with MAX and KDM5D to form a complex, and MAX recruited PCGF6 and KDM5D to the CpG island of the MAZ promoter and facilitated H3K4 histone demethylation. Furthermore, CDK4 was a downstream molecule of MAZ that participated in PCGF6/MAZ-regulated progression of pRCC. These results indicated that the upregulation of PCGF6 facilitated MAZ/CDK4 axis expression and pRCC progression by hypomethylation of the MAZ promoter. The PCGF6/MAZ/CDK4 regulatory axis may be a potential target for the treatment of ccRCC.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"9"},"PeriodicalIF":3.9,"publicationDate":"2023-03-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9996882/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9223958","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Proximity labeling reveals a new in vivo network of interactors for the histone demethylase KDM5. 接近标记揭示了组蛋白去甲基化酶KDM5的一个新的体内相互作用物网络。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-02-18 DOI: 10.1186/s13072-023-00481-y
Matanel Yheskel, Simone Sidoli, Julie Secombe

Background: KDM5 family proteins are multi-domain regulators of transcription that when dysregulated contribute to cancer and intellectual disability. KDM5 proteins can regulate transcription through their histone demethylase activity in addition to demethylase-independent gene regulatory functions that remain less characterized. To expand our understanding of the mechanisms that contribute to KDM5-mediated transcription regulation, we used TurboID proximity labeling to identify KDM5-interacting proteins.

Results: Using Drosophila melanogaster, we enriched for biotinylated proteins from KDM5-TurboID-expressing adult heads using a newly generated control for DNA-adjacent background in the form of dCas9:TurboID. Mass spectrometry analyses of biotinylated proteins identified both known and novel candidate KDM5 interactors, including members of the SWI/SNF and NURF chromatin remodeling complexes, the NSL complex, Mediator, and several insulator proteins.

Conclusions: Combined, our data shed new light on potential demethylase-independent activities of KDM5. In the context of KDM5 dysregulation, these interactions may play key roles in the alteration of evolutionarily conserved transcriptional programs implicated in human disorders.

背景:KDM5家族蛋白是转录的多结构域调控因子,当失调时会导致癌症和智力残疾。KDM5蛋白可以通过其组蛋白去甲基化酶活性来调节转录,除了去甲基化酶独立的基因调节功能之外,这些功能尚未得到充分的研究。为了扩大我们对kdm5介导的转录调控机制的理解,我们使用TurboID接近标记来鉴定kdm5相互作用的蛋白。结果:我们使用新生成的dCas9:TurboID dna邻近背景对照,从表达kdm5 -TurboID的果蝇成年头部富集生物素化蛋白。生物素化蛋白的质谱分析鉴定出已知的和新的候选KDM5相互作用物,包括SWI/SNF和NURF染色质重塑复合体的成员,NSL复合体,中介蛋白和一些绝缘体蛋白。综上所述,我们的数据揭示了KDM5潜在的去甲基化酶非依赖性活性。在KDM5失调的背景下,这些相互作用可能在涉及人类疾病的进化保守转录程序的改变中发挥关键作用。
{"title":"Proximity labeling reveals a new in vivo network of interactors for the histone demethylase KDM5.","authors":"Matanel Yheskel,&nbsp;Simone Sidoli,&nbsp;Julie Secombe","doi":"10.1186/s13072-023-00481-y","DOIUrl":"https://doi.org/10.1186/s13072-023-00481-y","url":null,"abstract":"<p><strong>Background: </strong>KDM5 family proteins are multi-domain regulators of transcription that when dysregulated contribute to cancer and intellectual disability. KDM5 proteins can regulate transcription through their histone demethylase activity in addition to demethylase-independent gene regulatory functions that remain less characterized. To expand our understanding of the mechanisms that contribute to KDM5-mediated transcription regulation, we used TurboID proximity labeling to identify KDM5-interacting proteins.</p><p><strong>Results: </strong>Using Drosophila melanogaster, we enriched for biotinylated proteins from KDM5-TurboID-expressing adult heads using a newly generated control for DNA-adjacent background in the form of dCas9:TurboID. Mass spectrometry analyses of biotinylated proteins identified both known and novel candidate KDM5 interactors, including members of the SWI/SNF and NURF chromatin remodeling complexes, the NSL complex, Mediator, and several insulator proteins.</p><p><strong>Conclusions: </strong>Combined, our data shed new light on potential demethylase-independent activities of KDM5. In the context of KDM5 dysregulation, these interactions may play key roles in the alteration of evolutionarily conserved transcriptional programs implicated in human disorders.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"8"},"PeriodicalIF":3.9,"publicationDate":"2023-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9938590/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9453625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
The transgenic IG-DMR sequence of the mouse Dlk1-Dio3 domain acquired imprinted DNA methylation during the post-fertilization period. 小鼠Dlk1-Dio3结构域的转基因IG-DMR序列在受精后获得了印迹DNA甲基化。
IF 3.9 2区 生物学 Q1 GENETICS & HEREDITY Pub Date : 2023-02-17 DOI: 10.1186/s13072-023-00482-x
Hitomi Matsuzaki, Shokichi Sugihara, Keiji Tanimoto

Background: Allele-specific methylation of the imprinting control region (ICR) is the molecular basis for the genomic imprinting phenomenon that is unique to placental mammals. We previously showed that the ICR at the mouse H19 gene locus (H19 ICR) was unexpectedly established after fertilization and not during spermatogenesis in transgenic mice (TgM), and that the same activity was essential for the maintenance of paternal methylation of the H19 ICR at the endogenous locus in pre-implantation embryos. To examine the universality of post-fertilization imprinted methylation across animal species or imprinted loci, we generated TgM with two additional sequences.

Results: The rat H19 ICR, which is very similar in structure to the mouse H19 ICR, unexpectedly did not acquire imprinted methylation even after fertilization, suggesting a lack of essential sequences in the transgene fragment. In contrast, the mouse IG-DMR, the methylation of which is acquired during spermatogenesis at the endogenous locus, did not acquire methylation in the sperm of TgM, yet became highly methylated in blastocysts after fertilization, but only when the transgene was paternally inherited. Since these two sequences were evaluated at the same genomic site by employing the transgene co-placement strategy, it is likely that the phenotype reflects the intrinsic activity of these fragments rather than position-effect variegation.

Conclusions: Our results suggested that post-fertilization imprinted methylation is a versatile mechanism for protecting paternal imprinted methylation from reprogramming during the pre-implantation period.

背景:印迹控制区(ICR)的等位基因特异性甲基化是胎盘哺乳动物特有的基因组印迹现象的分子基础。我们之前的研究表明,在转基因小鼠(TgM)中,小鼠H19基因位点的ICR (H19 ICR)意外地在受精后而不是在精子发生期间建立,并且在着床前胚胎中,相同的活性对于维持内源位点H19 ICR的父本甲基化是必不可少的。为了研究受精后印迹甲基化在动物物种或印迹位点之间的普遍性,我们用两个额外的序列生成了TgM。结果:与小鼠H19 ICR结构非常相似的大鼠H19 ICR,即使在受精后也未获得印迹甲基化,表明转基因片段缺乏必要的序列。相比之下,小鼠IG-DMR的甲基化是在精子发生过程中在内源性位点获得的,在TgM的精子中没有甲基化,但在受精后的囊胚中高度甲基化,但只有当转基因是父本遗传时才会发生甲基化。由于这两个序列是通过采用转基因共放置策略在同一基因组位点进行评估的,因此表型很可能反映了这些片段的内在活性,而不是位置效应变异。结论:我们的研究结果表明,受精后的印迹甲基化是一种保护父本印迹甲基化在植入前不被重编程的通用机制。
{"title":"The transgenic IG-DMR sequence of the mouse Dlk1-Dio3 domain acquired imprinted DNA methylation during the post-fertilization period.","authors":"Hitomi Matsuzaki,&nbsp;Shokichi Sugihara,&nbsp;Keiji Tanimoto","doi":"10.1186/s13072-023-00482-x","DOIUrl":"https://doi.org/10.1186/s13072-023-00482-x","url":null,"abstract":"<p><strong>Background: </strong>Allele-specific methylation of the imprinting control region (ICR) is the molecular basis for the genomic imprinting phenomenon that is unique to placental mammals. We previously showed that the ICR at the mouse H19 gene locus (H19 ICR) was unexpectedly established after fertilization and not during spermatogenesis in transgenic mice (TgM), and that the same activity was essential for the maintenance of paternal methylation of the H19 ICR at the endogenous locus in pre-implantation embryos. To examine the universality of post-fertilization imprinted methylation across animal species or imprinted loci, we generated TgM with two additional sequences.</p><p><strong>Results: </strong>The rat H19 ICR, which is very similar in structure to the mouse H19 ICR, unexpectedly did not acquire imprinted methylation even after fertilization, suggesting a lack of essential sequences in the transgene fragment. In contrast, the mouse IG-DMR, the methylation of which is acquired during spermatogenesis at the endogenous locus, did not acquire methylation in the sperm of TgM, yet became highly methylated in blastocysts after fertilization, but only when the transgene was paternally inherited. Since these two sequences were evaluated at the same genomic site by employing the transgene co-placement strategy, it is likely that the phenotype reflects the intrinsic activity of these fragments rather than position-effect variegation.</p><p><strong>Conclusions: </strong>Our results suggested that post-fertilization imprinted methylation is a versatile mechanism for protecting paternal imprinted methylation from reprogramming during the pre-implantation period.</p>","PeriodicalId":49253,"journal":{"name":"Epigenetics & Chromatin","volume":"16 1","pages":"7"},"PeriodicalIF":3.9,"publicationDate":"2023-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9936741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9522200","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","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
Epigenetics & Chromatin
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