hematopoietic stem and progenitor cell

造血干细胞和祖细胞
  • 文章类型: Journal Article
    在胚胎发生期间,人类造血干细胞(HSC)首先通过将特化的造血内皮(HE)细胞转化为早熟的HSC前体而出现在主动脉-性腺-中肾(AGM)区域。这个过程被称为内皮到造血转换(EHT),其中HE细胞经历剧烈的功能和形态变化,支抗依赖性内皮细胞对自由漂浮的圆形造血细胞。尽管它在人类HSC发展中发挥了重要作用,EHT的分子机制在很大程度上是未知的。这是由于缺乏与小鼠和其他模型生物相反的实时可视化人类HSC前体出现的方法。在这项研究中,通过在无饲养层的单层培养物中诱导人多能干细胞的HE,我们实现了对体外人类EHT的实时观察。通过对培养物的连续观察和单细胞追踪,可以可视化单个内皮细胞产生造血细胞并随后形成造血细胞簇的过程。分子标记表达中的剧烈HE到HSC转换也证实了EHT。值得注意的是,HSC前体的出现与不对称细胞分裂无关,而造血细胞团是通过EHT后漂浮细胞的增殖和组装形成的。这些结果揭示了人类EHT中未被重视的动态,我们预计我们的体外人类EHT模型将提供一个机会,以提高我们对人类HSC发育的理解。
    During embryogenesis, human hematopoietic stem cells (HSCs) first emerge in the aorta-gonad-mesonephros (AGM) region via transformation of specialized hemogenic endothelial (HE) cells into premature HSC precursors. This process is termed endothelial-to-hematopoietic transition (EHT), in which the HE cells undergo drastic functional and morphological changes from flat, anchorage-dependent endothelial cells to free-floating round hematopoietic cells. Despite its essential role in human HSC development, molecular mechanisms underlying the EHT are largely unknown. This is due to lack of methods to visualize the emergence of human HSC precursors in real time in contrast to mouse and other model organisms. In this study, by inducing HE from human pluripotent stem cells in feeder-free monolayer cultures, we achieved real-time observation of the human EHT in vitro. By continuous observation and single-cell tracking in the culture, it was possible to visualize a process that a single endothelial cell gives rise to a hematopoietic cell and subsequently form a hematopoietic-cell cluster. The EHT was also confirmed by a drastic HE-to-HSC switching in molecular marker expressions. Notably, HSC precursor emergence was not linked to asymmetric cell division, whereas the hematopoietic cell cluster was formed through proliferation and assembling of the floating cells after the EHT. These results reveal unappreciated dynamics in the human EHT, and we anticipate that our human EHT model in vitro will provide an opportunity to improve our understanding of the human HSC 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
    造血干细胞(HSC)具有在生物体的整个生命周期中维持所有血细胞类型的连续生产的能力。虽然主要位于成年人的骨髓中,HSC起源于胚胎发育。HSC出生的可视化,他们的发展轨迹,与它们连续的壁龛的特定相互作用极大地促进了我们对控制HSC形成和扩展的生物学和力学的理解。应用于活胚胎或非固定样品的体内技术已经显著提供了对HSC的细胞和解剖起源的宝贵见解。这些成像技术还揭示了HSC与周围微环境或小生境中相邻细胞类型之间的动态相互作用。如内皮细胞或巨噬细胞。这篇综述深入探讨了在理解起源方面取得的进步,生产,和HSC的细胞相互作用,特别是在小鼠和斑马鱼的胚胎发育过程中,专注于采用(实时)成像分析的研究。
    Hematopoietic stem cells (HSCs) possess the ability to sustain the continuous production of all blood cell types throughout an organism\'s lifespan. Although primarily located in the bone marrow of adults, HSCs originate during embryonic development. Visualization of the birth of HSCs, their developmental trajectory, and the specific interactions with their successive niches have significantly contributed to our understanding of the biology and mechanics governing HSC formation and expansion. Intravital techniques applied to live embryos or non-fixed samples have remarkably provided invaluable insights into the cellular and anatomical origins of HSCs. These imaging technologies have also shed light on the dynamic interactions between HSCs and neighboring cell types within the surrounding microenvironment or niche, such as endothelial cells or macrophages. This review delves into the advancements made in understanding the origin, production, and cellular interactions of HSCs, particularly during the embryonic development of mice and zebrafish, focusing on studies employing (live) imaging analysis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    β-地中海贫血是由缺陷性β-珠蛋白(HBB[血红蛋白亚基β])形成引起的,在严重的情况下,需要定期输血和铁螯合才能生存。造血干细胞的基因组编辑允许纠正潜在的突变作为治愈性治疗。作为基于双链断裂的编辑器的潜在更安全的替代品,碱基编辑器(BE)催化碱基转换,以精确编辑DNA靶位点,促使我们重新克隆和评估两个最近发表的腺嘌呤BE(ABEs;SpRY和SpG),其具有宽松的原型间隔区相邻基序要求,以纠正常见的HBBIVSI-110(G>A)剪接突变。ABE组分作为RNA的核转染到患者来源的CD34+细胞中,实现了对异常剪接至关重要的上游序列元件的高达90%的编辑,允许完整表征每个ABE的靶碱基编辑谱,并检测靶插入和缺失的差异。此外,这项研究确定了对两个相邻上下文碱基的剪接校正的相反影响,建立编辑窗口中多个BE编辑事件的频率分布,并显示了HBBIVSI-110(G>A)对我们的ABE的高效功能校正,包括RNA的水平,蛋白质,和红系分化。
    β-Thalassemia is brought about by defective β-globin (HBB [hemoglobin subunit β]) formation and, in severe cases, requires regular blood transfusion and iron chelation for survival. Genome editing of hematopoietic stem cells allows correction of underlying mutations as curative therapy. As potentially safer alternatives to double-strand-break-based editors, base editors (BEs) catalyze base transitions for precision editing of DNA target sites, prompting us to reclone and evaluate two recently published adenine BEs (ABEs; SpRY and SpG) with relaxed protospacer adjacent motif requirements for their ability to correct the common HBBIVSI-110(G>A) splice mutation. Nucleofection of ABE components as RNA into patient-derived CD34+ cells achieved up to 90% editing of upstream sequence elements critical for aberrant splicing, allowing full characterization of the on-target base-editing profile of each ABE and the detection of differences in on-target insertions and deletions. In addition, this study identifies opposing effects on splice correction for two neighboring context bases, establishes the frequency distribution of multiple BE editing events in the editing window, and shows high-efficiency functional correction of HBBIVSI-110(G>A) for our ABEs, including at the levels of RNA, protein, and erythroid differentiation.
    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
    在脊椎动物中,最早的造血干细胞和祖细胞(HSPCs)来源于一个专门的内皮细胞亚群,血源性内皮细胞,在主动脉-性腺-中肾区域通过内皮-造血过渡。HSPC的产生受到各种因素和信号的有效和准确的调节;然而,对这些信号的精确控制仍未完全理解。转录后调控对基因表达至关重要,因为转录本通常被RNA结合蛋白(RBP)结合以调节RNA代谢。这里,我们报告说,RBP蛋白Csde1介导的翻译控制对于斑马鱼早期发育过程中HSPC的产生至关重要。遗传突变体和形态突变体表明,csde1的消耗会损害斑马鱼胚胎中HSPC的产生。机械上,Csde1通过调节Wnt/β-catenin信号活性来调节HSPC的产生。我们证明Csde1与ctnnb1mRNA(编码β-catenin,Wnt信号的效应子)并调节翻译,但不调节ctnnb1mRNA的稳定性,进一步增强β-catenin蛋白水平和Wnt信号转导活性。一起,我们将Csde1确定为重要的转录后调节因子,并提供了有关Wnt/β-catenin信号如何在转录后水平上精确调节的新见解。
    In vertebrates, the earliest hematopoietic stem and progenitor cells (HSPCs) are derived from a subset of specialized endothelial cells, hemogenic endothelial cells, in the aorta-gonad-mesonephros region through endothelial-to-hematopoietic transition. HSPC generation is efficiently and accurately regulated by a variety of factors and signals; however, the precise control of these signals remains incompletely understood. Post-transcriptional regulation is crucial for gene expression, as the transcripts are usually bound by RNA-binding proteins (RBPs) to regulate RNA metabolism. Here, we report that the RBP protein Csde1-mediated translational control is essential for HSPC generation during zebrafish early development. Genetic mutants and morphants demonstrated that depletion of csde1 impaired HSPC production in zebrafish embryos. Mechanistically, Csde1 regulates HSPC generation through modulating Wnt/β-catenin signaling activity. We demonstrate that Csde1 binds to ctnnb1 mRNAs (encoding β-catenin, an effector of Wnt signaling) and regulates translation but not stability of ctnnb1 mRNA, which further enhances β-catenin protein level and Wnt signal transduction activities. Together, we identify Csde1 as an important post-transcriptional regulator and provide new insights into how Wnt/β-catenin signaling is precisely regulated at the post-transcriptional level.
    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
    细菌感染可以激活和动员造血干细胞和祖细胞(HSPCs)从骨髓(BM)到脾脏,称为髓外造血(EMH)的过程。最近的研究表明,共生细菌不仅调节宿主免疫系统,而且调节造血稳态。然而,肠道微生物对造血病理的影响尚不清楚.这里,我们发现全身单次注射Akkermansia黏蛋白(A.Mi.),一种粘蛋白降解细菌,快速激活BM骨髓生成和缓慢但持久的肝脾肿大,以功能性HSPCs的扩增和分化为特征,我们称之为延迟EMH。机械上,由A.m.触发的延迟EMH完全由MYD88/TRIF先天免疫信号通路介导,持续刺激脾骨髓细胞分泌白细胞介素(IL)-1α,反过来,激活表达IL-1受体(IL-1R)的脾HSPC。Toll样受体-2和-4(TLR2/4)或IL-1α的遗传缺失部分地减少A.m.-诱导延迟EMH,而两条途径的抑制可减轻脾肿大和EMH。我们的结果表明,协同IL-1R和TLR介导的信号调节共生细菌驱动的EMH,这可能与某些自身免疫性疾病有关。
    Bacterial infections can activate and mobilize hematopoietic stem and progenitor cells (HSPCs) from the bone marrow (BM) to the spleen, a process termed extramedullary hematopoiesis (EMH). Recent studies suggest that commensal bacteria regulate not only the host immune system but also hematopoietic homeostasis. However, the impact of gut microbes on hematopoietic pathology remains unclear. Here, we find that systemic single injections of Akkermansia muciniphila (A. m.), a mucin-degrading bacterium, rapidly activate BM myelopoiesis and slow but long-lasting hepato-splenomegaly, characterized by the expansion and differentiation of functional HSPCs, which we term delayed EMH. Mechanistically, delayed EMH triggered by A. m. is mediated entirely by the MYD88/TRIF innate immune signaling pathway, which persistently stimulates splenic myeloid cells to secrete interleukin (IL)-1α, and in turn, activates IL-1 receptor (IL-1R)-expressing splenic HSPCs. Genetic deletion of Toll-like receptor-2 and -4 (TLR2/4) or IL-1α partially diminishes A. m.-induced delayed EMH, while inhibition of both pathways alleviates splenomegaly and EMH. Our results demonstrate that cooperative IL-1R- and TLR-mediated signals regulate commensal bacteria-driven EMH, which might be relevant for certain autoimmune disorders.
    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
    由造血干细胞和祖细胞(HSPC)及其分化谱系组成的造血系统是揭示细胞命运转变一般原理的理想模型。从原肠胚形成开始,相继出现原始造血(产生专门的造血细胞),促确定造血(产生谱系限制的祖细胞),和确定的造血(产生多能HSPCs)。这些新生谱系在几个短暂的造血位点发育,并最终定植到终身造血位点。造血谱系的发展和维持是由细胞内在基因调控网络和细胞外在微环境线索协调的。由于渐进的方法(例如,高通量谱系追踪和单细胞功能和组学分析),我们对造血谱系的发育起源和某些造血器官的功能特性的认识得到了更新;同时,表征稀有细胞类型的新范例,细胞异质性及其原因,并提供了全面的监管景观。这里,我们回顾了发育和出生后造血过程中HSPC生物学的演变观点。此外,我们讨论了HSPCs体外诱导和扩增的最新进展,重点是对临床应用的影响。
    The hematopoietic system composed of hematopoietic stem and progenitor cells (HSPCs) and their differentiated lineages serves as an ideal model to uncover generic principles of cell fate transitions. From gastrulation onwards, there successively emerge primitive hematopoiesis (that produces specialized hematopoietic cells), pro-definitive hematopoiesis (that produces lineage-restricted progenitor cells), and definitive hematopoiesis (that produces multipotent HSPCs). These nascent lineages develop in several transient hematopoietic sites and finally colonize into lifelong hematopoietic sites. The development and maintenance of hematopoietic lineages are orchestrated by cell-intrinsic gene regulatory networks and cell-extrinsic microenvironmental cues. Owing to the progressive methodology (e.g., high-throughput lineage tracing and single-cell functional and omics analyses), our understanding of the developmental origin of hematopoietic lineages and functional properties of certain hematopoietic organs has been updated; meanwhile, new paradigms to characterize rare cell types, cell heterogeneity and its causes, and comprehensive regulatory landscapes have been provided. Here, we review the evolving views of HSPC biology during developmental and postnatal hematopoiesis. Moreover, we discuss recent advances in the in vitro induction and expansion of HSPCs, with a focus on the implications for clinical applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Preprint
    造血干细胞和祖细胞(HSPC)移植是血液学疾病的基本治疗方法,但是对具有相关功能的人类HSPC子集的更精细定义可以使移植物更好地调整和更常规,低风险应用。对于HSPCs的深层表型,经过328种抗原的筛选,我们量化了数百万CD34+和CD34-细胞上的41种表面蛋白和功能调节剂,跨越四个主要的人类造血组织:骨髓,动员的外周血,脐带血,还有胎儿肝脏.我们提出了更精细的HSPC子集定义,并提供了新的,详细的分化轨迹红系和髓系。我们修订的人类造血模型的这些方面通过相应的表观遗传分析和体外克隆分化测定进行了验证。总的来说,我们证明了使用分子调节剂作为细胞身份和功能潜力的替代品的实用性,提供描述的框架,预期隔离,和人类HSPCs的跨组织比较。
    Hematopoietic stem and progenitor cell (HSPC) transplantation is an essential therapy for hematological conditions, but finer definitions of human HSPC subsets with associated function could enable better tuning of grafts and more routine, lower-risk application. To deeply phenotype HSPCs, following a screen of 328 antigens, we quantified 41 surface proteins and functional regulators on millions of CD34+ and CD34- cells, spanning four primary human hematopoietic tissues: bone marrow, mobilized peripheral blood, cord blood, and fetal liver. We propose more granular definitions of HSPC subsets and provide new, detailed differentiation trajectories of erythroid and myeloid lineages. These aspects of our revised human hematopoietic model were validated with corresponding epigenetic analysis and in vitro clonal differentiation assays. Overall, we demonstrate the utility of using molecular regulators as surrogates for cellular identity and functional potential, providing a framework for description, prospective isolation, and cross-tissue comparison of HSPCs in humans.
    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
    在自体造血干/祖细胞(HSPC)中使用CRISPR-Cas9和重组腺相关病毒血清型6(rAAV6)进行离体基因校正以治疗镰状细胞病(SCD)现已进入早期临床研究阶段。为促进CRISPR-Cas9/rAAV6基因组编辑技术的进展,我们分析了人类HSPCs基因组编辑过程中和之后关键试剂的分子变化和细胞反应.我们证明了rAAV6作为供体DNA模板的高稳定性。我们评估了更长HSPC预刺激在编辑细胞数量增加方面的益处。我们观察到p53通路被瞬时激活,在6小时达到峰值,并随着时间的推移而解决。值得注意的是,我们揭示了p21mRNA水平与细胞中rAAV6基因组数量之间的强相关性,以及用siRNA瞬时抑制p53对基因组编辑的有益作用,细胞增殖,细胞存活。就潜在的免疫原性而言,我们发现rAAV6衣壳蛋白是检测不到的,而在基因组编辑后48小时仍检测到痕量的残留Cas9蛋白。我们相信这些信息将为HSPC中基因校正方案的未来改进提供重要的见解。
    Ex vivo gene correction with CRISPR-Cas9 and a recombinant adeno-associated virus serotype 6 (rAAV6) in autologous hematopoietic stem/progenitor cells (HSPCs) to treat sickle cell disease (SCD) has now entered early-phase clinical investigation. To facilitate the progress of CRISPR-Cas9/rAAV6 genome editing technology, we analyzed the molecular changes in key reagents and cellular responses during and after the genome editing procedure in human HSPCs. We demonstrated the high stability of rAAV6 to serve as the donor DNA template. We assessed the benefit of longer HSPC pre-stimulation in terms of increased numbers of edited cells. We observed that the p53 pathway was transiently activated, peaking at 6 h, and resolved over time. Notably, we revealed a strong correlation between p21 mRNA level and rAAV6 genome number in cells and beneficial effects of transient inhibition of p53 with siRNA on genome editing, cell proliferation, and cell survival. In terms of potential immunogenicity, we found that rAAV6 capsid protein was not detectable, while a trace amount of residual Cas9 protein was still detected at 48 h post-genome editing. We believe this information will provide important insights for future improvements of gene correction protocols in HSPCs.
    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
    造血干细胞(HSC)通过利用其自我更新和多能能力,在整个生命中维持造血至关重要。Ferroptosis是一种细胞死亡,其特征是脂质过氧化物的铁依赖性积累,它涉及多种生理和病理条件。最近的研究强调了铁凋亡在HSCs功能维持中的重要作用。这里,我们描述了我们目前在体内和体外获得造血干细胞和祖细胞(HSPC)中的铁细胞凋亡的方案。我们介绍了测量HSPC中总活性氧(ROS)和脂质ROS的程序,以及分析细胞数量,细胞活力,和细胞周期概况。该协议提供了一种有用的方法,用于表征HSPCs中的铁凋亡及其相关参数的状态,用于研究铁死亡对造血的影响。
    Hematopoietic stem cells (HSCs) are critical for maintaining hematopoiesis throughout life by utilizing their self-renewing and multipotent capabilities. Ferroptosis is a type of cell death characterized by the iron-dependent accumulation of lipid peroxides, and it is involved in multiple physiological and pathological conditions. Recent studies have highlighted the important role of ferroptosis in the functional maintenance of HSCs. Here, we describe our current protocols for accessing ferroptosis in hematopoietic stem and progenitor cells (HSPCs) both in vivo and in vitro. We introduce procedures for measuring total reactive oxygen species (ROS) and lipid ROS in HSPCs, as well as analyzing cell number, cell viability, and cell cycle profiles. This protocol provides a useful approach for characterizing the status of ferroptosis and its related parameters in HSPCs and more broadly, for studying the outcomes of ferroptosis on hematopoiesis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    STING是有效的先天和适应性抗肿瘤免疫的关键介质;然而,STING激动剂的瘤内给药在临床试验中显示出有限的治疗益处。在癌症中静脉内递送STING激动剂的全身作用尚未明确。这里,我们证明了STING激动剂的全身给药抑制黑色素瘤生长,改善炎症效应细胞浸润,诱导骨髓动员和髓外造血,引起外周血中免疫成分的广泛变化。全身给药的STING激动剂促进了HSC扩增并影响了谱系命运的承诺,这表现为HSPCs的分化偏向骨髓细胞,以B细胞淋巴生成和红细胞生成为代价。转录组分析显示髓系分化相关基因和I型干扰素相关基因上调。这种有髓样的分化促进了骨髓细胞向活化表型的产生和成熟。此外,Gr-1+骨髓细胞的消耗减弱了STING激动剂的抗肿瘤免疫力。我们的发现揭示了全身施用STING激动剂的抗肿瘤机制,该机制涉及调节HSPC分化和促进骨髓细胞成熟。我们的研究可能有助于解释瘤内施用STING激动剂的有限临床活性。
    STING is a pivotal mediator of effective innate and adaptive anti-tumor immunity; however, intratumoral administration of STING agonists have shown limited therapeutic benefit in clinical trials. The systemic effect of the intravenous delivery of STING agonists in cancer is not well-defined. Here, we demonstrated that systemic administration of STING agonist inhibited melanoma growth, improved inflammatory effector cell infiltration, and induced bone marrow mobilization and extramedullary hematopoiesis, causing widespread changes in immune components in the peripheral blood. The systemically administered STING agonist promoted HSC expansion and influenced lineage fate commitment, which was manifested as the differentiation of HSPCs was skewed toward myeloid cells at the expense of B-cell lymphopoiesis and erythropoiesis. Transcriptome analysis revealed upregulation of myeloid lineage differentiation-related and type I interferon-related genes. This myeloid-biased differentiation promoted the production and maturation of myeloid cells toward an activated phenotype. Furthermore, depletion of Gr-1+ myeloid cells attenuated the anti-tumor immunity of STING agonist. Our findings reveal the anti-tumor mechanism of systemic administration of STING agonist that involves modulating HSPC differentiation and promoting myeloid cells maturation. Our study may help explain the limited clinical activity of STING agonists administered intratumorally.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号