DNA (Cytosine-5-)-Methyltransferase 1

DNA (胞嘧啶 - 5 -) - 甲基转移酶 1
  • 文章类型: Journal Article
    赖氨酸特异性组蛋白去甲基酶1(LSD1),将单甲基化或二甲基化组蛋白H3在赖氨酸4(H3K4me1/2)上,对早期胚胎发生和发育至关重要。在这里,我们表明LSD1对于小鼠胚胎干细胞(ESC)的自我更新是不必要的,但对于小鼠ESC的生长和分化是必需的。重新引入催化受损的LSD1(LSD1MUT)可以恢复小鼠ESC的增殖能力,然而,LSD1的酶活性对于确保适当的分化至关重要。的确,Lsd1敲除(KO)小鼠ESC中H3K4me1的增加不会导致与干性相关的全局基因表达程序的重大变化。然而,LSD1而非LSD1MUT的消融导致DNMT1和UHRF1蛋白减少,并伴随整体低甲基化.我们表明,LSD1和LSD1MUT都通过与HDAC1和泛素特异性肽酶7(USP7)的相互作用来控制UHRF1和DNMT1的蛋白质稳定性,因此,促进DNMT1和UHRF1的去乙酰化和去泛素化。我们的研究阐明了LSD1控制小鼠ESCDNA甲基化的机制,独立于其赖氨酸脱甲基酶活性。
    Lysine-specific histone demethylase 1 (LSD1), which demethylates mono- or di- methylated histone H3 on lysine 4 (H3K4me1/2), is essential for early embryogenesis and development. Here we show that LSD1 is dispensable for mouse embryonic stem cell (ESC) self-renewal but is required for mouse ESC growth and differentiation. Reintroduction of a catalytically-impaired LSD1 (LSD1MUT) recovers the proliferation capability of mouse ESCs, yet the enzymatic activity of LSD1 is essential to ensure proper differentiation. Indeed, increased H3K4me1 in Lsd1 knockout (KO) mouse ESCs does not lead to major changes in global gene expression programs related to stemness. However, ablation of LSD1 but not LSD1MUT results in decreased DNMT1 and UHRF1 proteins coupled to global hypomethylation. We show that both LSD1 and LSD1MUT control protein stability of UHRF1 and DNMT1 through interaction with HDAC1 and the ubiquitin-specific peptidase 7 (USP7), consequently, facilitating the deacetylation and deubiquitination of DNMT1 and UHRF1. Our studies elucidate a mechanism by which LSD1 controls DNA methylation in mouse ESCs, independently of its lysine demethylase activity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:小分子荧光探针在不同宫颈病变中甲基化检测的价值分析。
    方法:(1)4组不同病变组织的灰度值差异显著(p<0.05)。两组之间灰度值的比较显示,CA组明显超过LSIL和宫颈炎组,且HSIL组明显高于LSIL组和宫颈炎组(p<0.05);(2)以55.21为中线计算入组受试者的平均灰度值,>55.21为正,≤55.21为负。
    结果:结果显示,宫颈炎组的阳性率为0.00%,LSIL组67.74%,HSIL组83.33%,和CA组100.00%。四组之间的结果明显不同(p<0.05);(3)DAPI之间的比较,探针,明亮,和合并的宫颈炎图像,LSIL,HSIL,和CA表明不同的宫颈病变有相当不同的染色。
    结论:灰度值,阳性率,不同宫颈病变的染色图像明显不同。该小分子荧光探针对宫颈病变具有较好的鉴别价值,可考虑推广应用。
    BACKGROUND: Value analysis of a small-molecule fluorescent probe for methylation detection in different cervical lesions.
    METHODS: (1) The grayscale values of distinct lesion tissues were remarkably distinct among the four groups (p < 0.05). The comparison of the grayscale value between the two groups showed that the CA group noticeably exceeded the LSIL and cervicitis groups, and the HSIL group was apparently higher than the LSIL and cervicitis groups (p < 0.05); (2) The mean grayscale values of the enrolled subjects were calculated with 55.21 as the midline, with >55.21 as positive and ≤55.21 as negative.
    RESULTS: The results showed that the positive rate of the cervicitis group was 0.00%, the LSIL group 67.74%, the HSIL group 83.33%, and the CA group 100.00%. The results among the four groups were notably distinct (p < 0.05); (3) The comparison among DAPI, probe, bright, and merged images of cervicitis, LSIL, HSIL, and CA indicated that different cervical lesions were with quite various stains.
    CONCLUSIONS: The grayscale value, positive rate, and stained picture of distinct cervical lesions were remarkably different. The small-molecule fluorescent probe has a good value in differentiating cervical lesions and can be considered for popularization and application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DNMT1是一种必需的DNA甲基转移酶,可催化甲基转移到DNA中的CpG岛,并产生突出的表观遗传标记。DNMT1的催化活性依赖于其构象可塑性和将构象从自抑制状态改变为活化状态的能力。这里,我们提出了apoDNMT1和DNTM1的四种低温EM重建:非生产性DNA,DNTM1:H3Ub2-肽,DNTM1:生产性DNA复合物。我们的结构证明了DNMT1的N端调节域在从apo“自动抑制”过渡到DNA结合的“非生产性”以及最终的DNA结合的“生产性”过渡过程中的灵活性。DNMT1的状态。此外,我们讨论了DNMT1选择性小分子抑制剂和泛素化组蛋白H3对DNMT1甲基转移酶活性的调节。我们观察到,尽管存在抑制剂,DNMT1仍以“非生产性”状态结合DNA,并在与双泛素化H3肽类似物的复合物中呈现全长DNMT1的低温EM重建。一起来看,我们的结果提供了DNMT1反应循环的结构见解。
    DNMT1 is an essential DNA methyltransferase that catalyzes the transfer of methyl groups to CpG islands in DNA and generates a prominent epigenetic mark. The catalytic activity of DNMT1 relies on its conformational plasticity and ability to change conformation from an auto-inhibited to an activated state. Here, we present four cryo-EM reconstructions of apo DNMT1 and DNTM1: non-productive DNA, DNTM1: H3Ub2-peptide, DNTM1: productive DNA complexes. Our structures demonstrate the flexibility of DNMT1\'s N-terminal regulatory domains during the transition from an apo \'auto-inhibited\' to a DNA-bound \'non-productive\' and finally a DNA-bound \'productive\' state of DNMT1. Furthermore, we address the regulation of DNMT1\'s methyltransferase activity by a DNMT1-selective small-molecule inhibitor and ubiquitinated histone H3. We observe that DNMT1 binds DNA in a \'non-productive\' state despite the presence of the inhibitor and present the cryo-EM reconstruction of full-length DNMT1 in complex with a di-ubiquitinated H3 peptide analogue. Taken together, our results provide structural insights into the reaction cycle of DNMT1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人类沉默中心(HUSH)复合物与LINE-1逆转录转座子(L1s)和其他基因组重复序列的转录本结合,招募MORC2和其他效应子重塑染色质。HUSH和MORC2如何与DNA甲基化一起运作,重复转录的中心表观遗传调节因子,仍然很大程度上未知。在这里,我们询问了人类神经祖细胞(hNPC)中的这种关系,一种耐受DNA甲基转移酶DNMT1去除的脑发育体细胞模型。当hNPCs中MORC2或HUSH亚基TSOR丢失时,L1通过稳健的启动子甲基化保持沉默。然而,进化年轻的L1s的基因组去甲基化和激活吸引MORC2结合,DNMT1和MORC2的同时消耗导致L1转录物的大量积累。我们在着丝粒α卫星和聚集的原钙粘蛋白基因上确定了相同的机制层次结构,对染色体结构和神经发育重要的重复元件。我们的数据描述了体细胞中重复序列的表观遗传控制,对理解HUSH-MORC2在整个人类发育过程中的低甲基化环境中的重要功能具有重要意义。
    The human silencing hub (HUSH) complex binds to transcripts of LINE-1 retrotransposons (L1s) and other genomic repeats, recruiting MORC2 and other effectors to remodel chromatin. How HUSH and MORC2 operate alongside DNA methylation, a central epigenetic regulator of repeat transcription, remains largely unknown. Here we interrogate this relationship in human neural progenitor cells (hNPCs), a somatic model of brain development that tolerates removal of DNA methyltransferase DNMT1. Upon loss of MORC2 or HUSH subunit TASOR in hNPCs, L1s remain silenced by robust promoter methylation. However, genome demethylation and activation of evolutionarily-young L1s attracts MORC2 binding, and simultaneous depletion of DNMT1 and MORC2 causes massive accumulation of L1 transcripts. We identify the same mechanistic hierarchy at pericentromeric α-satellites and clustered protocadherin genes, repetitive elements important for chromosome structure and neurodevelopment respectively. Our data delineate the epigenetic control of repeats in somatic cells, with implications for understanding the vital functions of HUSH-MORC2 in hypomethylated contexts throughout human development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在这项研究中,在八种不同的绵羊组织中构建miR-148a的表达谱,包括乳腺组织,在六个不同的发育时期。miR-148a对小鼠活力的影响,扩散,并研究了绵羊乳腺上皮细胞(OMEC)的乳脂合成,并验证了miR-148a与两个预测靶基因的靶关系。miR-148a的表达具有明显的组织特异性和时间特异性。miR-148a在研究的所有八种绵羊组织中表达,在乳腺组织中表达水平最高(p<0.05)。此外,miR-148a在研究的六个发育期中的每一个期间在绵羊乳腺组织中表达,在泌乳高峰期最高(p<0.05)。miR-148a的过表达增加了OMEC的活力,Edu标记的阳性OMEC的数量和百分比,和两个细胞增殖标记基因的表达水平。miR-148a还增加了S期OMEC的百分比。相比之下,与miR-148a模拟物相比,用miR-148a抑制剂转染产生相反的效果.这些结果表明miR-148a促进小尾寒羊OMEC的存活和增殖。miR-148a模拟物使OMECs中的甘油三酯含量增加37.78%(p<0.01),并提高三种乳脂合成标记基因的表达水平。然而,miR-148a抑制剂使甘油三酯水平降低了87.11%(p<0.01)。这些结果表明miR-148a促进OMEC中的乳脂合成。双荧光素酶报告基因检测显示miR-148a降低了野生型载体中DNA甲基转移酶1(DNMT1)和过氧化物酶体增殖物激活受体γ辅激活因子1-A(PPARGC1A)的荧光素酶活性,表明它们是miR-148a的靶基因。miR-148a在绵羊乳腺组织中的表达与PPARGC1A呈高度负相关(r=-0.789,p<0.001)。而与DNMT1呈中度负相关(r=-0.515,p=0.029)。这是第一个揭示miR-148a的分子机制的研究,扩散,和绵羊乳脂合成OMECs。
    In this study, the expression profiles of miR-148a were constructed in eight different ovine tissues, including mammary gland tissue, during six different developmental periods. The effect of miR-148a on the viability, proliferation, and milk fat synthesis of ovine mammary epithelial cells (OMECs) was investigated, and the target relationship of miR-148a with two predicted target genes was verified. The expression of miR-148a exhibited obvious tissue-specific and temporal-specific patterns. miR-148a was expressed in all eight ovine tissues investigated, with the highest expression level in mammary gland tissue (p < 0.05). Additionally, miR-148a was expressed in ovine mammary gland tissue during each of the six developmental periods studied, with its highest level at peak lactation (p < 0.05). The overexpression of miR-148a increased the viability of OMECs, the number and percentage of Edu-labeled positive OMECs, and the expression levels of two cell-proliferation marker genes. miR-148a also increased the percentage of OMECs in the S phase. In contrast, transfection with an miR-148a inhibitor produced the opposite effect compared to the miR-148a mimic. These results indicate that miR-148a promotes the viability and proliferation of OMECs in Small-tailed Han sheep. The miR-148a mimic increased the triglyceride content by 37.78% (p < 0.01) and the expression levels of three milk fat synthesis marker genes in OMECs. However, the miR-148a inhibitor reduced the triglyceride level by 87.11% (p < 0.01). These results suggest that miR-148a promotes milk fat synthesis in OMECs. The dual-luciferase reporter assay showed that miR-148a reduced the luciferase activities of DNA methyltransferase 1 (DNMT1) and peroxisome proliferator-activated receptor gamma coactivator 1-A (PPARGC1A) in wild-type vectors, suggesting that they are target genes of miR-148a. The expression of miR-148a was highly negatively correlated with PPARGC1A (r = -0.789, p < 0.001) in ovine mammary gland tissue, while it had a moderate negative correlation with DNMT1 (r = -0.515, p = 0.029). This is the first study to reveal the molecular mechanisms of miR-148a underlying the viability, proliferation, and milk fat synthesis of OMECs in sheep.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    低甲基化药物(HMA)是骨髓增生异常肿瘤(MDS)和急性髓系白血病(AML)的一线疗法。然而,获得性抵抗和治疗失败是司空见惯的。为了解决这个问题,我们在人类MDS来源的细胞系中进行全基因组CRISPR-Cas9筛选,MDS-L,并将TOPORS确定为与HMA协同作用的功能丧失靶标,在异种移植模型中降低白血病负担并提高生存率。我们证明,TOPORS的耗竭通过诱发白血病母细胞对受损的DNA损伤反应(DDR)的介导对HMA处理的TOPORS耗竭细胞中SUMO化DNMT1的积累来介导对HMA的敏感性。HMA与TOPORS靶向的组合不会损害健康的造血。虽然TOPORS的抑制剂不可用,我们表明,用TAK-981抑制蛋白质SUMO化部分地出现HMA敏感性和DDR损害。总的来说,我们的数据表明,HMA联合SUMOylation或TOPORS抑制是高危MDS(HR-MDS)或AML的合理治疗选择.
    Hypomethylating agents (HMAs) are frontline therapies for Myelodysplastic Neoplasms (MDS) and Acute Myeloid Leukemia (AML). However, acquired resistance and treatment failure are commonplace. To address this, we perform a genome-wide CRISPR-Cas9 screen in a human MDS-derived cell line, MDS-L, and identify TOPORS as a loss-of-function target that synergizes with HMAs, reducing leukemic burden and improving survival in xenograft models. We demonstrate that depletion of TOPORS mediates sensitivity to HMAs by predisposing leukemic blasts to an impaired DNA damage response (DDR) accompanied by an accumulation of SUMOylated DNMT1 in HMA-treated TOPORS-depleted cells. The combination of HMAs with targeting of TOPORS does not impair healthy hematopoiesis. While inhibitors of TOPORS are unavailable, we show that inhibition of protein SUMOylation with TAK-981 partially phenocopies HMA-sensitivity and DDR impairment. Overall, our data suggest that the combination of HMAs with inhibition of SUMOylation or TOPORS is a rational treatment option for High-Risk MDS (HR-MDS) or AML.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    DNA低甲基化剂(HMA)用于治疗骨髓性恶性肿瘤,尽管它们的治疗效果并不令人满意。在这里,我们表明CRISPR-Cas9筛选揭示了拓扑异构酶1结合精氨酸/富含丝氨酸的蛋白(TOPORS)的敲除,编码泛素/SUMOE3连接酶,增强HMA对髓系白血病细胞的疗效,对正常造血几乎没有影响,表明TOPORS参与了对HMA的抵抗。将HMA掺入DNA并捕获DNA甲基转移酶-1(DNMT1)以形成DNA-DNMT1交联,进行SUMOylation,其次是蛋白酶体退化。持续交联是细胞毒性的。指针环指域,介导泛素化,负责HMA抗性。在TOPORS敲除细胞中,DNMT1通过HMA处理稳定,由于泛素化效率低下,导致未解决的SUMO化DNMT1的积累。这表明TOPORS泛素化SUMO化的DNMT1,从而促进DNA-DNMT1交联的解析。始终如一,泛素化抑制剂,TAK-243和SUMO化抑制剂,TAK-981显示通过DNMT1稳定与HMA的协同作用。我们的研究提供了一种基于HMA的新型治疗策略,该策略会干扰DNA-DNMT1交联的分辨率。
    DNA hypomethylating agents (HMAs) are used for the treatment of myeloid malignancies, although their therapeutic effects have been unsatisfactory. Here we show that CRISPR-Cas9 screening reveals that knockout of topoisomerase 1-binding arginine/serine-rich protein (TOPORS), which encodes a ubiquitin/SUMO E3 ligase, augments the efficacy of HMAs on myeloid leukemic cells with little effect on normal hematopoiesis, suggesting that TOPORS is involved in resistance to HMAs. HMAs are incorporated into the DNA and trap DNA methyltransferase-1 (DNMT1) to form DNA-DNMT1 crosslinks, which undergo SUMOylation, followed by proteasomal degradation. Persistent crosslinking is cytotoxic. The TOPORS RING finger domain, which mediates ubiquitination, is responsible for HMA resistance. In TOPORS knockout cells, DNMT1 is stabilized by HMA treatment due to inefficient ubiquitination, resulting in the accumulation of unresolved SUMOylated DNMT1. This indicates that TOPORS ubiquitinates SUMOylated DNMT1, thereby promoting the resolution of DNA-DNMT1 crosslinks. Consistently, the ubiquitination inhibitor, TAK-243, and the SUMOylation inhibitor, TAK-981, show synergistic effects with HMAs through DNMT1 stabilization. Our study provides a novel HMA-based therapeutic strategy that interferes with the resolution of DNA-DNMT1 crosslinks.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在哺乳动物中,全局被动去甲基化有助于早期胚胎发育过程中的表观遗传重编程。在这个阶段,大部分DNA甲基转移酶1(DNMT1)蛋白被排除在细胞核之外,这被认为是主要原因。然而,DNMT1的剩余核活性是否受其他机制调控尚不清楚.这里,我们报告说,通过小鼠受精卵中的蛋白酶体降解来调整核DNMT1的丰度。我们确定了一个母性因素,Pramel15,其目标是DNMT1通过Cullin-RINGE3连接酶降解。Pramel15的缺失会升高受精卵原核中的DNMT1水平,损害合子DNA去甲基化,并导致早期胚胎中DNA甲基化的随机增加。因此,Pramel15可以调节合子DNA复制过程中细胞核中DNMT1的残留水平,从而确保早期胚胎中有效的DNA甲基化重编程。
    In mammals, global passive demethylation contributes to epigenetic reprogramming during early embryonic development. At this stage, the majority of DNA-methyltransferase 1 (DNMT1) protein is excluded from nucleus, which is considered the primary cause. However, whether the remaining nuclear activity of DNMT1 is regulated by additional mechanisms is unclear. Here, we report that nuclear DNMT1 abundance is finetuned through proteasomal degradation in mouse zygotes. We identify a maternal factor, Pramel15, which targets DNMT1 for degradation via Cullin-RING E3 ligases. Loss of Pramel15 elevates DNMT1 levels in the zygote pronuclei, impairs zygotic DNA demethylation, and causes a stochastic gain of DNA methylation in early embryos. Thus, Pramel15 can modulate the residual level of DNMT1 in the nucleus during zygotic DNA replication, thereby ensuring efficient DNA methylation reprogramming in early embryos.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    SNF2家族ATPaseHELLS及其激活剂CDCA7的突变导致免疫缺陷,着丝粒不稳定,面部畸形综合症,以异染色质处的DNA低甲基化为特征。尚不清楚为什么CDCA7-HELLS是唯一的核小体重塑复合物,其缺陷消除了DNA甲基化的维持。我们在这里将CDCA7的独特锌指结构域鉴定为进化上保守的半甲基化感应锌指(HMZF)结构域。CDCA7-核小体复合物的冷冻电子显微镜结构分析表明,HMZF结构域可以识别核小体核心颗粒内面向外的DNA大沟中的半甲基化CpG,而维持甲基转移酶DNMT1的关键激活剂UHRF1则不能。CDCA7将HELLS募集到半甲基化染色质,并促进与复制解偶联的维持DNA甲基化相关的UHRF1介导的H3泛素化。我们建议CDCA7-HELLS核小体重塑复合物通过感测UHRF1和DNMT1无法接触的半甲基化CpG来帮助维持染色质上的DNA甲基化。
    Mutations of the SNF2 family ATPase HELLS and its activator CDCA7 cause immunodeficiency, centromeric instability, and facial anomalies syndrome, characterized by DNA hypomethylation at heterochromatin. It remains unclear why CDCA7-HELLS is the sole nucleosome remodeling complex whose deficiency abrogates the maintenance of DNA methylation. We here identify the unique zinc-finger domain of CDCA7 as an evolutionarily conserved hemimethylation-sensing zinc finger (HMZF) domain. Cryo-electron microscopy structural analysis of the CDCA7-nucleosome complex reveals that the HMZF domain can recognize hemimethylated CpG in the outward-facing DNA major groove within the nucleosome core particle, whereas UHRF1, the critical activator of the maintenance methyltransferase DNMT1, cannot. CDCA7 recruits HELLS to hemimethylated chromatin and facilitates UHRF1-mediated H3 ubiquitylation associated with replication-uncoupled maintenance DNA methylation. We propose that the CDCA7-HELLS nucleosome remodeling complex assists the maintenance of DNA methylation on chromatin by sensing hemimethylated CpG that is otherwise inaccessible to UHRF1 and DNMT1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    这项研究探讨了预测的microRNA对NB4细胞系中DNA甲基转移酶(DNMT)和PODXL基因的影响,旨在阐明它们在急性髓系白血病(AML)发病机制中的作用。采用了全面的方法学框架来探索6-姜酚对DNMT的治疗意义。这包括一套用于蛋白质结构预测的生物信息学工具,对接,分子动力学,和ADMET分析,以及miRNA和PODXL表达水平的经验评估。这种多方面的策略有助于深入了解6-姜辣素在DNMT调制中的潜在功效。这些发现表明了一个微妙的相互作用,其中6-姜辣素给药调节miRNA表达水平,NB4细胞中DNMT1和DNMT3A表达降低。这种改变间接影响PODXL表达,有助于致癌表型的表现。DNMT1和DNMT3A在NB4细胞中的过表达可能有助于AML,似乎可通过miR-193a和miR-200c等microRNA进行调节。用6-姜酚后处理,观察到DNMT1和DNMT3A表达改变,最终导致miR-193a和miR-200c的上调。这种级联效应导致癌细胞中肿瘤抑制基因的失调,包括PODXL的下调,以及癌变特征的出现。这些见解强调了6-姜辣素在AML背景下靶向DNMT和microRNA的治疗前景。
    This investigation delves into the influence of predicted microRNAs on DNA methyltransferases (DNMTs) and the PODXL gene within the NB4 cell line, aiming to elucidate their roles in the pathogenesis of acute myeloid leukemia (AML). A comprehensive methodological framework was adopted to explore the therapeutic implications of 6-gingerol on DNMTs. This encompassed a suite of bioinformatics tools for protein structure prediction, docking, molecular dynamics, and ADMET profiling, alongside empirical assessments of miRNA and PODXL expression levels. Such a multifaceted strategy facilitated an in-depth understanding of 6-gingerol\'s potential efficacy in DNMT modulation. The findings indicate a nuanced interplay where 6-gingerol administration modulated miRNA expression levels, decreasing in DNMT1 and DNMT3A expression in NB4 cells. This alteration indirectly influenced PODXL expression, contributing to the manifestation of oncogenic phenotypes. The overexpression of DNMT1 and DNMT3A in NB4 cells may contribute to AML, which appears modulable via microRNAs such as miR-193a and miR-200c. Post-treatment with 6-gingerol, DNMT1 and DNMT3A expression alterations were observed, culminating in the upregulation of miR-193a and miR-200c. This cascade effect led to the dysregulation of tumor suppressor genes in cancer cells, including downregulation of PODXL, and the emergence of cancerous traits. These insights underscore the therapeutic promise of 6-gingerol in targeting DNMTs and microRNAs within the AML context.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号