Hematopoietic stem cell

造血干细胞
  • 文章类型: Journal Article
    单基因血液病是全世界最常见的遗传疾病之一。这些疾病导致显著的儿童和成人发病率,有些人可能会在出生前死亡。新型离体造血干细胞(HSC)基因编辑疗法在改变治疗前景方面具有巨大的希望,但并非没有潜在的限制。体内基因编辑疗法为这些疾病提供了潜在的更安全和更容易获得的治疗,但由于缺乏靶向HSC的递送载体而受到阻碍。它位于难以接近的骨髓生态位。这里,我们认为,这种生物屏障可以通过利用HSC在胎儿发育过程中容易进入的肝脏中的优势来克服。为了便于将基因编辑货物运送到胎儿HSC,我们开发了一种可电离的脂质纳米颗粒(LNP)平台,靶向HSC表面的CD45受体。在验证靶向LNP在体外通过CD45特异性机制改善了信使核糖核酸(mRNA)向造血谱系细胞的递送后,我们证明了这个平台介导的安全,强力,以及在多种小鼠模型中体内HSC的长期基因调节。我们在体外进一步优化了该LNP平台以封装和递送基于CRISPR的核酸货物。最后,我们发现,经过优化和靶向的LNPs增强了子宫内单次静脉注射后胎儿HSC概念验证基因座处的基因编辑.通过在胎儿发育过程中体内靶向HSC,我们的系统优化目标编辑机械(STEM)LNP可能提供一种可翻译的策略来治疗出生前的单基因血液病.
    Monogenic blood diseases are among the most common genetic disorders worldwide. These diseases result in significant pediatric and adult morbidity, and some can result in death prior to birth. Novel ex vivo hematopoietic stem cell (HSC) gene editing therapies hold tremendous promise to alter the therapeutic landscape but are not without potential limitations. In vivo gene editing therapies offer a potentially safer and more accessible treatment for these diseases but are hindered by a lack of delivery vectors targeting HSCs, which reside in the difficult-to-access bone marrow niche. Here, we propose that this biological barrier can be overcome by taking advantage of HSC residence in the easily accessible liver during fetal development. To facilitate the delivery of gene editing cargo to fetal HSCs, we developed an ionizable lipid nanoparticle (LNP) platform targeting the CD45 receptor on the surface of HSCs. After validating that targeted LNPs improved messenger ribonucleic acid (mRNA) delivery to hematopoietic lineage cells via a CD45-specific mechanism in vitro, we demonstrated that this platform mediated safe, potent, and long-term gene modulation of HSCs in vivo in multiple mouse models. We further optimized this LNP platform in vitro to encapsulate and deliver CRISPR-based nucleic acid cargos. Finally, we showed that optimized and targeted LNPs enhanced gene editing at a proof-of-concept locus in fetal HSCs after a single in utero intravenous injection. By targeting HSCs in vivo during fetal development, our Systematically optimized Targeted Editing Machinery (STEM) LNPs may provide a translatable strategy to treat monogenic blood diseases before birth.
    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
    使用CRISPR/Cas系统作为有效的基因组编辑工具对造血干细胞(HSC)进行基因操作,对于解决血液系统疾病具有巨大的希望。推进这种治疗的一个重要障碍在于有效地将CRISPR/Cas递送至HSC。虽然存在各种交付格式,核糖核蛋白复合物(RNP)是一种特别有效的选择。RNP复合物提供增强的基因编辑能力,没有病毒载体,具有快速的活动和最小化的脱靶效应。然而,新的递送方法,如基于微流体的技术,filtroporation,纳米粒子,和细胞穿透肽不断进化。本研究旨在对这些方法以及RNP复合物向HSC的递送方法的最新研究进行全面综述。
    Gene manipulation of hematopoietic stem cells (HSCs) using the CRISPR/Cas system as a potent genome editing tool holds immense promise for addressing hematologic disorders. An essential hurdle in advancing this treatment lies in effectively delivering CRISPR/Cas to HSCs. While various delivery formats exist, Ribonucleoprotein complex (RNP) emerges as a particularly efficient option. RNP complexes offer enhanced gene editing capabilities, devoid of viral vectors, with rapid activity and minimized off-target effects. Nevertheless, novel delivery methods such as microfluidic-based techniques, filtroporation, nanoparticles, and cell-penetrating peptides are continually evolving. This study aims to provide a comprehensive review of these methods and the recent research on delivery approaches of RNP complexes to HSCs.
    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:确定性造血过程中的内皮到造血转化(EHT)过程在脊椎动物中高度保守。在斑马鱼EHT期间已检测到转座因子(TE)的阶段特异性表达,并且可能通过激活炎症信号传导来促进造血干细胞(HSC)形成。然而,人们对TEs如何在人类和小鼠中对EHT过程做出贡献知之甚少。
    结果:我们重建了人和小鼠的单细胞EHT轨迹,并解析了TEs在EHT过程中的动态表达模式。大多数TE沿着保守的EHT轨迹呈现瞬时共同上调模式,与表观遗传沉默系统的时间松弛相吻合。TE产品可以被多个模式识别受体感知,触发炎症信号以促进HSC的出现。有趣的是,我们观察到缺氧相关信号在TE表达较高的细胞中富集。此外,我们构建了可获得的TE的造血顺式调节网络,并鉴定了可能促进特定EHT标记基因表达的潜在TE衍生增强子.
    结论:我们的研究提供了一个系统的视野,说明如何通过转录和顺式调控网络动态控制TE以促进造血命运决定。并预先训练新生造血干细胞的免疫力。
    BACKGROUND: The endothelial-to-hematopoietic transition (EHT) process during definitive hematopoiesis is highly conserved in vertebrates. Stage-specific expression of transposable elements (TEs) has been detected during zebrafish EHT and may promote hematopoietic stem cell (HSC) formation by activating inflammatory signaling. However, little is known about how TEs contribute to the EHT process in human and mouse.
    RESULTS: We reconstructed the single-cell EHT trajectories of human and mouse and resolved the dynamic expression patterns of TEs during EHT. Most TEs presented a transient co-upregulation pattern along the conserved EHT trajectories, coinciding with the temporal relaxation of epigenetic silencing systems. TE products can be sensed by multiple pattern recognition receptors, triggering inflammatory signaling to facilitate HSC emergence. Interestingly, we observed that hypoxia-related signals were enriched in cells with higher TE expression. Furthermore, we constructed the hematopoietic cis-regulatory network of accessible TEs and identified potential TE-derived enhancers that may boost the expression of specific EHT marker genes.
    CONCLUSIONS: Our study provides a systematic vision of how TEs are dynamically controlled to promote the hematopoietic fate decisions through transcriptional and cis-regulatory networks, and pre-train the immunity of nascent HSCs.
    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
    白血病干细胞(LSCs)与白血病发展之间的关联已在遗传改变的背景下得到广泛确立。表观遗传途径,和信号通路调控。造血干细胞位于骨髓层次结构的顶部,可以自我更新并逐渐产生血液和免疫细胞。微环境,小生境细胞,以及调节它们的复杂信号通路会由于衰老而获得基因突变和表观遗传改变,慢性炎症环境,压力,和癌症,导致造血干细胞失调以及异常血液和免疫细胞的产生,导致血液恶性肿瘤和血癌。获得这些突变的细胞以比其他细胞更快的速率生长并诱导克隆扩增。过度生长导致血癌的发展。标准疗法靶向原始细胞,迅速扩散;然而,可以诱导疾病复发的LSCs在治疗后仍然存在,导致复发和预后不良。为了克服这些限制,研究人员集中在LSCs的特征和信号系统以及靶向它们阻断LSCs的疗法上.这篇综述旨在全面了解造血系统恶性肿瘤的类型,导致它们的白血病干细胞的特征,这些细胞获得化疗抗性的机制,以及针对这些机制的疗法。
    The association between leukemic stem cells (LSCs) and leukemia development has been widely established in the context of genetic alterations, epigenetic pathways, and signaling pathway regulation. Hematopoietic stem cells are at the top of the bone marrow hierarchy and can self-renew and progressively generate blood and immune cells. The microenvironment, niche cells, and complex signaling pathways that regulate them acquire genetic mutations and epigenetic alterations due to aging, a chronic inflammatory environment, stress, and cancer, resulting in hematopoietic stem cell dysregulation and the production of abnormal blood and immune cells, leading to hematological malignancies and blood cancer. Cells that acquire these mutations grow at a faster rate than other cells and induce clone expansion. Excessive growth leads to the development of blood cancers. Standard therapy targets blast cells, which proliferate rapidly; however, LSCs that can induce disease recurrence remain after treatment, leading to recurrence and poor prognosis. To overcome these limitations, researchers have focused on the characteristics and signaling systems of LSCs and therapies that target them to block LSCs. This review aims to provide a comprehensive understanding of the types of hematopoietic malignancies, the characteristics of leukemic stem cells that cause them, the mechanisms by which these cells acquire chemotherapy resistance, and the therapies targeting these mechanisms.
    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
    脐带血造血干细胞(UCB-HSC)基于其自我更新和效力特征在疾病的治疗中具有重要作用。了解细胞周期每个步骤中涉及的基因谱和信号通路可以改善HSC的治疗方法。这项研究的目的是预测HSCsG0,G1和分化阶段涉及的基因谱和信号通路。
    干预(n=8)和非干预(n=3)数据集从基因表达综合(GEO)数据库获得,并进行杂交和分析以确定与HSCs的G0,G1和分化阶段中的每一个相关的高表达和低表达基因。然后,将STRING的分数注释到基因数据中。使用Cytoscape软件构建基因网络,并丰富了KEGG和GO数据库。
    由于介入和非介入数据的内部和内部交叉,确定了高表达和低表达基因。非介入数据用于构建基因网络(n=6),并使用介入数据改进了节点。在G0,G1和分化阶段的每个阶段都提出了几种重要的信号传导途径。
    数据显示,不同的信号通路在G0,G1和分化阶段的每个阶段都被激活,因此它们的基因可能被靶向以改善HSC治疗。
    UNASSIGNED: Umbilical cord blood hematopoietic stem cells (UCB-HSCs) have important roles in the treatment of illnesses based on their self-renewal and potency characteristics. Knowing the gene profiles and signaling pathways involved in each step of the cell cycle could improve the therapeutic approaches of HSCs. The aim of this study was to predict the gene profiles and signaling pathways involved in the G0, G1, and differentiation stages of HSCs.
    UNASSIGNED: Interventional (n = 8) and non-interventional (n = 3) datasets were obtained from the Gene Expression Omnibus (GEO) database, and were crossed and analyzed to determine the high- and low-express genes related to each of the G0, G1, and differentiation stages of HSCs. Then, the scores of STRING were annotated to the gene data. The gene networks were constructed using Cytoscape software, and enriched with the KEGG and GO databases.
    UNASSIGNED: The high- and low-express genes were determined due to inter and intra intersections of the interventional and non-interventional data. The non-interventional data were applied to construct the gene networks (n = 6) with the nodes improved using the interventional data. Several important signaling pathways were suggested in each of the G0, G1, and differentiation stages.
    UNASSIGNED: The data revealed that the different signaling pathways are activated in each of the G0, G1, and differentiation stages so that their genes may be targeted to improve the HSC therapy.
    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甲基化在造血分化中起关键作用。Epimutation是DNA甲基化的随机变异,可诱导表观遗传异质性。然而,对正常造血和造血疾病的影响尚不清楚。在这项研究中,我们开发了一个名为EpiMut的Julia软件包,可以快速准确地量化表象。EpiMut用于评估并提供稳态造血分化中的表象景观,涉及从造血干/祖细胞到成熟细胞的13种血细胞。我们表明,大量基因组区域表现出表观遗传变异,而不是骨髓和淋巴谱系之间DNA甲基化水平的显着差异。在每个谱系的分化过程中观察到了进化的逐步动力学。重要的是,我们发现,表象显着丰富与谱系分化相关的信号。此外,各种来源的造血干细胞(HSCs)和急性髓系白血病的表观表达与HSCs的功能和恶性细胞疾病有关。一起来看,我们的研究全面记录了一张表观图谱,揭示了它在人类造血中的重要作用,从而提供对造血调控的见解。
    DNA methylation plays a critical role in hematopoietic differentiation. Epimutation is a stochastic variation in DNA methylation that induces epigenetic heterogeneity. However, the effects of epimutations on normal hematopoiesis and hematopoietic diseases remain unclear. In this study, we developed a Julia package called EpiMut that enabled rapid and accurate quantification of epimutations. EpiMut was used to evaluate and provide an epimutation landscape in steady-state hematopoietic differentiation involving 13 types of blood cells ranging from hematopoietic stem/progenitor cells to mature cells. We showed that substantial genomic regions exhibited epigenetic variations rather than significant differences in DNA methylation levels between the myeloid and lymphoid lineages. Stepwise dynamics of epimutations were observed during the differentiation of each lineage. Importantly, we found that epimutation significantly enriched signals associated with lineage differentiation. Furthermore, epimutations in hematopoietic stem cells (HSCs) derived from various sources and acute myeloid leukemia were related to the function of HSCs and malignant cell disorders. Taken together, our study comprehensively documented an epimutation map and uncovered its important roles in human hematopoiesis, thereby offering insights into hematopoietic regulation.
    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
    原发性造血干细胞和祖细胞(HSPC)中的精确基因编辑将促进单基因疾病的治愈性治疗以及疾病建模。即使使用CRISPR/Cas系统,也能实现精确的效率,然而,保持有限。通过引导RNA递送的优化,供体设计,和添加剂,我们现在已经获得了初级脐带血HSCPs的平均精确编辑效率>90%,且毒性最小,且未观察到脱靶编辑.实现这种高效率所需的主要方案修改是添加DNA-PK抑制剂AZD7648,以及除了破坏PAM序列的突变之外,在供体中包含间隔区破坏沉默突变。严重的,编辑甚至跨越了祖先阶层,在集落形成细胞测定或高自我更新潜力的长期培养起始细胞的频率中,不会实质上扭曲层次结构或影响谱系输出。由于许多疾病的建模需要杂合性,我们还证明,通过添加确定的突变体和野生型供体的混合物,可以调整整体编辑和接合性.有了这些优化,编辑在接近完美的效率现在可以直接在人类HSPC完成。这将为治疗策略和疾病建模开辟新的途径。
    Precision gene editing in primary hematopoietic stem and progenitor cells (HSPCs) would facilitate both curative treatments for monogenic disorders as well as disease modelling. Precise efficiencies even with the CRISPR/Cas system, however, remain limited. Through an optimization of guide RNA delivery, donor design, and additives, we have now obtained mean precise editing efficiencies >90% on primary cord blood HSCPs with minimal toxicity and without observed off-target editing. The main protocol modifications needed to achieve such high efficiencies were the addition of the DNA-PK inhibitor AZD7648, and the inclusion of spacer-breaking silent mutations in the donor in addition to mutations disrupting the PAM sequence. Critically, editing was even across the progenitor hierarchy, did not substantially distort the hierarchy or affect lineage outputs in colony-forming cell assays or the frequency of high self-renewal potential long-term culture initiating cells. As modelling of many diseases requires heterozygosity, we also demonstrated that the overall editing and zygosity can be tuned by adding in defined mixtures of mutant and wild-type donors. With these optimizations, editing at near-perfect efficiency can now be accomplished directly in human HSPCs. This will open new avenues in both therapeutic strategies and disease modelling.
    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:造血干细胞(HSC)再生是骨髓抑制造血恢复的基础,这是细胞毒性的威胁生命的副作用。骨髓抑制损伤后,HSC利基被严重破坏,虽然人们对生态位是否以及如何重塑和调节HSC再生知之甚少。
    方法:通过将小鼠暴露于亚致死剂量的电离辐射,建立了辐射损伤诱导的骨髓抑制的小鼠模型。数量的动态变化,通过流式细胞术确定HSCs和巨核细胞的分布和功能,免疫荧光,集落测定和骨髓移植,结合转录组学分析。使用共培养系统和过继转移确定HSC和巨核细胞之间的通讯。在体内和体外研究了信号机制,并使用巨核细胞特异性敲除小鼠和转基因小鼠进行巩固。
    结果:放射损伤后,巨核细胞成为HSC生态位的主要成分,并定位在更接近HSC的位置。同时,短暂性胰岛素样生长因子1(IGF1)的高分泌主要在辐射损伤后的巨核细胞中引起,而造血干细胞再生平行巨核细胞IGF1高分泌。机械上,HSC对巨核细胞IGF1分泌过多特别敏感,和IGF1信号下游的mTOR不仅促进HSC的激活,包括增殖和线粒体氧化代谢,但也抑制铁素吞噬限制HSC铁凋亡。因此,扩散之间的微妙协调,线粒体氧化代谢和铁凋亡确保了辐射损伤后功能性HSC的扩增。重要的是,在辐射损伤后,及时给予IGF1同时促进HSC再生和造血恢复,代表骨髓抑制的一种优越的治疗方法。
    结论:我们的研究确定巨核细胞是骨髓抑制性损伤的最后一道防线,巨核细胞IGF1是保护HSC再生的新生态位信号。
    BACKGROUND: Hematopoietic stem cell (HSC) regeneration underlies hematopoietic recovery from myelosuppression, which is a life-threatening side effect of cytotoxicity. HSC niche is profoundly disrupted after myelosuppressive injury, while if and how the niche is reshaped and regulates HSC regeneration are poorly understood.
    METHODS: A mouse model of radiation injury-induced myelosuppression was built by exposing mice to a sublethal dose of ionizing radiation. The dynamic changes in the number, distribution and functionality of HSCs and megakaryocytes were determined by flow cytometry, immunofluorescence, colony assay and bone marrow transplantation, in combination with transcriptomic analysis. The communication between HSCs and megakaryocytes was determined using a coculture system and adoptive transfer. The signaling mechanism was investigated both in vivo and in vitro, and was consolidated using megakaryocyte-specific knockout mice and transgenic mice.
    RESULTS: Megakaryocytes become a predominant component of HSC niche and localize closer to HSCs after radiation injury. Meanwhile, transient insulin-like growth factor 1 (IGF1) hypersecretion is predominantly provoked in megakaryocytes after radiation injury, whereas HSCs regenerate paralleling megakaryocytic IGF1 hypersecretion. Mechanistically, HSCs are particularly susceptible to megakaryocytic IGF1 hypersecretion, and mTOR downstream of IGF1 signaling not only promotes activation including proliferation and mitochondrial oxidative metabolism of HSCs, but also inhibits ferritinophagy to restrict HSC ferroptosis. Consequently, the delicate coordination between proliferation, mitochondrial oxidative metabolism and ferroptosis ensures functional HSC expansion after radiation injury. Importantly, punctual IGF1 administration simultaneously promotes HSC regeneration and hematopoietic recovery after radiation injury, representing a superior therapeutic approach for myelosuppression.
    CONCLUSIONS: Our study identifies megakaryocytes as a last line of defense against myelosuppressive injury and megakaryocytic IGF1 as a novel niche signal safeguarding HSC regeneration.
    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
    急性髓性白血病(AML)的特征是骨髓和外周血中未成熟骨髓细胞的积累。近一半的AML患者在标准诱导治疗后复发,迫切需要新的治疗形式。由于缺乏疗效和安全性,嵌合抗原受体(CAR)T疗法迄今在AML中尚未成功。的确,最有吸引力的抗原靶标是干细胞标志物,如CD33或CD123。我们证明了CD37,一种成熟的B细胞标志物,在AML样本中表达,其存在与欧洲白血病网(ELN)2017年风险分层相关。我们将抗淋巴瘤CD37CAR重新用于治疗AML,并显示CD37CART细胞特异性杀死AML细胞,分泌促炎细胞因子,并在体内控制癌症进展。重要的是,CD37CART细胞对造血干细胞没有毒性。因此,CD37是一种有前途且安全的CAR-T细胞AML靶标。
    Acute myeloid leukemia (AML) is characterized by the accumulation of immature myeloid cells in the bone marrow and the peripheral blood. Nearly half of the AML patients relapse after standard induction therapy, and new forms of therapy are urgently needed. Chimeric antigen receptor (CAR) T therapy has so far not been successful in AML due to lack of efficacy and safety. Indeed, the most attractive antigen targets are stem cell markers such as CD33 or CD123. We demonstrate that CD37, a mature B cell marker, is expressed in AML samples, and its presence correlates with the European LeukemiaNet (ELN) 2017 risk stratification. We repurpose the anti-lymphoma CD37CAR for the treatment of AML and show that CD37CAR T cells specifically kill AML cells, secrete proinflammatory cytokines, and control cancer progression in vivo. Importantly, CD37CAR T cells display no toxicity toward hematopoietic stem cells. Thus, CD37 is a promising and safe CAR T cell AML target.
    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
    造血在整个生命中持续产生来自造血干细胞(HSC)的所有类型的血细胞。代谢状态是HSC自我更新和分化的已知调节因子,但是代谢传感器是否以及如何进行O-GlcNAcylation,可以通过抑制其循环酶O-GlcNAcase(OGA)和O-GlcNAc转移酶(OGT)来调节,对造血的贡献在很大程度上仍然未知。在这里,等基因,OGA耗尽(OGAi)和OGT耗尽(OGTi)人类诱导多能干细胞(hiPSCs)的单细胞克隆成功地从主hiPSC系MUSIi012-A,从包含表观遗传记忆的CD34造血干/祖细胞(HSPC)重新编程。已建立的OGAi和OGTihiPSC表现出细胞O-GlcNAcylation的增加或减少,伴随着OGA和OGT的丧失,分别,表型和核型正常,并保留了多能性,尽管它们可能倾向于向某些细菌谱系分化。在通过中胚层诱导和内皮到造血转变进行造血分化时,我们发现OGA抑制加速了hiPSC对HSPCs的承诺,而O-GlcNAc稳态的破坏会影响其对红系谱系的承诺.来自所有组的分化的HSPC能够产生所有造血祖细胞,从而证实了它们的功能特征。总之,已建立的OGTi和OGAihiPSC的单细胞克隆为进一步剖析O-GlcNAcylation在血细胞发育中不同阶段和谱系的作用提供了有价值的平台.这些hiPSC中OGA和OGT的不完全敲除使它们容易受到额外的操纵,即,通过小分子,允许O-GlcNAcylation的分子动力学研究。
    Hematopoiesis continues throughout life to produce all types of blood cells from hematopoietic stem cells (HSCs). Metabolic state is a known regulator of HSC self-renewal and differentiation, but whether and how metabolic sensor O-GlcNAcylation, which can be modulated via an inhibition of its cycling enzymes O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), contributes to hematopoiesis remains largely unknown. Herein, isogenic, single-cell clones of OGA-depleted (OGAi) and OGT-depleted (OGTi) human induced pluripotent stem cells (hiPSCs) were successfully generated from the master hiPSC line MUSIi012-A, which were reprogrammed from CD34+ hematopoietic stem/progenitor cells (HSPCs) containing epigenetic memory. The established OGAi and OGTi hiPSCs exhibiting an increase or decrease in cellular O-GlcNAcylation concomitant with their loss of OGA and OGT, respectively, appeared normal in phenotype and karyotype, and retained pluripotency, although they may favor differentiation toward certain germ lineages. Upon hematopoietic differentiation through mesoderm induction and endothelial-to-hematopoietic transition, we found that OGA inhibition accelerates hiPSC commitment toward HSPCs and that disruption of O-GlcNAc homeostasis affects their commitment toward erythroid lineage. The differentiated HSPCs from all groups were capable of giving rise to all hematopoietic progenitors, thus confirming their functional characteristics. Altogether, the established single-cell clones of OGTi and OGAi hiPSCs represent a valuable platform for further dissecting the roles of O-GlcNAcylation in blood cell development at various stages and lineages of blood cells. The incomplete knockout of OGA and OGT in these hiPSCs makes them susceptible to additional manipulation, i.e., by small molecules, allowing the molecular dynamics studies of O-GlcNAcylation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号