Hematopoietic stem cell

造血干细胞
  • 文章类型: Journal Article
    目的:母亲肥胖越来越普遍,并对后代健康产生负面影响。肥胖母亲的孩子患造血系统异常和代谢相关疾病的风险更高,如2型糖尿病。有趣的是,疾病风险通常取决于后代的性别,提示母亲肥胖对后代造血干细胞和祖细胞(HSPC)功能的性别特异性重编程作用。然而,母亲肥胖暴露对后代HSPC功能的影响,HSPC调节后代代谢健康的能力在很大程度上被研究不足。本研究旨在验证肥胖小鼠的后代在HSPC功能上表现出性别差异,从而影响后代的代谢健康这一假设。
    方法:我们首先使用出生后第21天(P21)和8周龄的C57BL/6J小鼠评估骨髓造血干细胞和祖细胞表型。我们还对HSPC(线-,Sca1+,cKit+细胞)来自P21小鼠,用于竞争性初次和二次移植,以及转录组学分析。体重,肥胖,在初次和二次移植受体动物中进行胰岛素耐量试验和葡萄糖耐量试验.
    结果:我们发现了后代HSPC功能对母亲肥胖暴露的反应的性别差异,肥胖水坝(MatOb)的雄性后代显示出减少的HSPC数量和植入,而雌性MatOb后代基本上未受影响。RNA-seq揭示了雌性MatOb后代的免疫刺激途径。最后,只有男性MatOb后代HSPC的接受者表现出葡萄糖耐受不良。
    结论:这项研究证明了母亲肥胖暴露对后代HSPC功能的持久影响,并暗示HSPC参与了代谢调节。
    OBJECTIVE: Maternal obesity is increasingly common and negatively impacts offspring health. Children of mothers with obesity are at higher risk of developing diseases linked to hematopoietic system abnormalitiesand metabolism such as type 2 diabetes. Interestingly, disease risks are often dependent on the offspring\'s sex, suggesting sex-specific reprogramming effect of maternal obesity on offspring hematopoietic stem and progenitor cell (HSPC) function. However, the impact of maternal obesity exposure on offspring HSPC function, and the capability of HSPC to regulate offspring metabolic health is largely understudied. This study aims to test the hypothesis that offspring of obese mice exhibit sex-differences in HSPC function that affect offspring\'s metabolic health.
    METHODS: We first assessed bone marrow hematopoietic stem and progenitor cell phenotype using postnatal day 21 (P21) and 8-week-old C57BL/6J mice born to control and diet-induced obese dams. We also sorted HSPC (Lineage-, Sca1+, cKit+ cells) from P21 mice for competitive primary and secondary transplant, as well as transcriptomic analysis. Body weight, adiposity, insulin tolerance test and glucose tolerance tests were performed in primary and secondary transplant recipient animals.
    RESULTS: We discovered sex-differences in offspring HSPC function in response to maternal obesity exposure, where male offspring of obese dams (MatOb) showed decreased HSPC numbers and engraftment, while female MatOb offspring remained largely unaffected. RNA-seq revealed immune stimulatory pathways in female MatOb offspring. Finally, only recipients of male MatOb offspring HSPC exhibited glucose intolerance.
    CONCLUSIONS: This study demonstrated the lasting effect of maternal obesity exposure on offspring HSPC function and implicates HSPC in metabolic regulation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    The number of somatic mutations among all tissues increases along with age. This process was well-studied in hematopoietic stem cells (HSCs). Some mutations lead to a proliferative advantage and expansion of HSCs to form a dominant clone. Clonal hematopoiesis is general in the elderly population. Clonal hematopoiesis of indeterminate potential (CHIP) is a more common phenomenon in the elderly and is defined as somatic mutations in clonal blood cells without any other hematological malignancies. The development of CHIP is an independent risk factor for hematological malignancies, cardiovascular diseases, and reduced overall survival. CHIP is frequently associated with mutations in DNMT3A and TET2 genes involved in DNA methylation. The epigenetic human body clocks have been developed based on the age-related changes in methylation, making it possible to detect epigenetic aging. The combination of epigenetic aging and CHUP is associated with adverse health outcomes. Further research will reveal the significance of clonal hematopoiesis and CHIP in aging, acquiring various diseases, and determining the feasibility of influencing the mutagenic potential of clones.
    С возрастом во всех тканях увеличивается количество соматических мутаций. Лучше всего этот процесс изучен в стволовых кроветворных клетках. Некоторые мутации могут привести к пролиферативному преимуществу и экспансии стволовых кроветворных клеток с образованием клона. Клональное кроветворение широко распространено у пожилых людей. Клональный гемопоэз неопределенного потенциала (КГНП) — феномен, который чаще встречается в пожилом возрасте и характеризуется соматическими мутациями в клетках-предшественницах гемопоэза с формированием нескольких минорных клонов, экспансия которых способна постепенно вытеснить нормальный гемопоэз. Развитие КГНП является независимым фактором риска опухолей системы крови, сердечно-сосудистых заболеваний и общей летальности. При КГНП чаще всего мутируют гены DNMT3A и TET2, которые участвуют в метилировании ДНК. На основании возрастного изменения метилирования разработаны эпигенетические часы организма человека, позволяющие выявить эпигенетическое старение. Сочетание последнего и КГНП связано с неблагоприятными исходами для здоровья. Дальнейшее исследования позволят понять значение клонального гемопоэза и КГНП в процессе старения и развитии различных заболеваний, определить возможности целенаправленного воздействия на мутировавшие клоны.
    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
    大多数造血干细胞移植用于神经系统自身免疫性疾病,使用患者的造血干细胞(HSC)。获得HSC接枝是该方法的第一步。这通常涉及使用高剂量环磷酰胺和非格司亭将骨髓HSC动员到循环中,一种基于粒细胞集落刺激因子的药物。与造血干细胞移植期间经历的那些相比,这些药物的毒性通常是可控的,并且不良事件较不严重和较不频繁。动员后,通过白细胞去除术从循环中收集HSC。一些中心使用离体免疫磁性选择程序耗尽淋巴细胞的移植物。将HSC移植物冷冻保存直到干细胞移植需要。移植物的质量测试确保了无菌性,并且其含有足够的HSC和造血祖细胞。HSC移植物动员的临床和实验室方面,收藏,和存储必须符合国家监管机构制定的标准,并得到国际专业标准组织的认可。经验丰富的干细胞移植团队对于最大程度地减少程序毒性和增强成功收集非常重要。
    Most hematopoietic stem cell transplants performed for an autoimmune disease of the nervous system, use the patient\'s hematopoietic stem cells (HSCs). Obtaining an HSC graft is the first step of the process. This typically involves mobilization of bone marrow HSCs into the circulation using high-dose cyclophosphamide followed by filgrastim, a drug based on granulocyte colony-stimulating factor. Toxicity from these agents is usually manageable and adverse events are less severe and less frequent than those experienced during the hematopoietic stem cell transplant. Following mobilization, HSCs are collected from the circulation by leukapheresis. Some centers deplete the graft of lymphocytes using an ex vivo immunomagnetic selection procedure. HSC grafts are cryopreserved until required for the stem cell transplant. Quality testing of the graft ensures sterility and it contains sufficient HSCs and hematopoietic progenitors. The clinical and laboratory aspects of HSC graft mobilization, collection, and storage must meet standards set by national regulatory bodies and accredited by international professional standards organizations. Experienced stem cell transplant teams are important for minimizing procedural toxicity and enhancing successful collection.
    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
    血细胞的产生来自造血干细胞(HSC)的活性,由它们的自我更新能力和产生所有成熟血细胞类型的能力来定义。小鼠仍然是血液学研究中研究最多的物种之一,和用于定义和分离小鼠HSC的标记是良好建立的。鉴于骨髓中HSC的频率非常低,通过红细胞裂解和磁性细胞分离的干细胞预富集通常作为分离过程的一部分进行以减少分选时间。有几种富集前策略可用,不同的速度,丰富程度,最终细胞产量和成本。在目前的研究中,我们进行了并排比较,并提供了决策树,以帮助研究人员根据其下游应用选择用于小鼠HSC分离的预富集策略.然后,我们将不同的预富集技术与HSC的代谢组学分析相结合,在那里速度,预富集过程中的产量和温度是关键因素,并且发现预富集策略的选择显着影响HSC中检测到的代谢物的数量和单个代谢物的水平。
    Blood cell production arises from the activity of hematopoietic stem cells (HSCs), defined by their self-renewal capacity and ability to give rise to all mature blood cell types. The mouse remains one of the most studied species in hematological research, and markers to define and isolate mouse HSCs are well-established. Given the very low frequency of HSCs in the bone marrow, stem cell pre-enrichment by red blood cell lysis and magnetic cell separation is often performed as part of the isolation process to reduce sorting times. Several pre-enrichment strategies are available, differing in their speed, degree of enrichment, final cell yield and cost. In the current study, we performed a side-by-side comparison and provide a decision tree to help researchers select a pre-enrichment strategy for mouse HSC isolation depending on their downstream application. We then compared different pre-enrichment techniques in combination with metabolomics analysis of HSCs, where speed, yield and temperature during pre-enrichment are crucial factors, and found that the choice of pre-enrichment strategy significantly impacts the number of metabolites detected and levels of individual metabolites in HSCs.
    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
    染色质引发促进细胞类型特异性基因表达,谱系分化,和发展。染色质引发的机制尚未完全了解。这里,我们报道,缺乏BAF(BRG1/BRM相关因子)染色质重塑复合物的Baf155亚基的小鼠造血干细胞和祖细胞(HSPCs)产生的成熟血细胞数量显着减少,导致移植后造血再生失败和5-氟尿嘧啶(5-FU)损伤。Baf155缺陷型HSPC产生的中性粒细胞特别少,B细胞,和体内稳态的CD8+T细胞,支持更具免疫抑制性的肿瘤微环境并增强肿瘤生长。单核多组学分析显示,Baf155缺陷型HSPC无法在富含推定增强子和造血谱系转录因子结合基序的选定区域中建立可接近的染色质。我们的研究提供了对Baf155在造血谱系染色质引发中的作用以及Baf155缺乏在再生和肿瘤免疫中的功能后果的基本机制理解。
    Chromatin priming promotes cell-type-specific gene expression, lineage differentiation, and development. The mechanism of chromatin priming has not been fully understood. Here, we report that mouse hematopoietic stem and progenitor cells (HSPCs) lacking the Baf155 subunit of the BAF (BRG1/BRM-associated factor) chromatin remodeling complex produce a significantly reduced number of mature blood cells, leading to a failure of hematopoietic regeneration upon transplantation and 5-fluorouracil (5-FU) injury. Baf155-deficient HSPCs generate particularly fewer neutrophils, B cells, and CD8+ T cells at homeostasis, supporting a more immune-suppressive tumor microenvironment and enhanced tumor growth. Single-nucleus multiomics analysis reveals that Baf155-deficient HSPCs fail to establish accessible chromatin in selected regions that are enriched for putative enhancers and binding motifs of hematopoietic lineage transcription factors. Our study provides a fundamental mechanistic understanding of the role of Baf155 in hematopoietic lineage chromatin priming and the functional consequences of Baf155 deficiency in regeneration and tumor immunity.
    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
    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
    成人血细胞是由造血干细胞(HSC)在骨髓中产生的,其起源可以追溯到胎儿发育阶段。的确,在小鼠发育过程中,在妊娠的第10-11天,主动脉-性腺-中肾(AGM)区域是HSC产生的主要部位,在背主动脉中观察到与干细胞发生相关的特征性细胞簇。在其他部位如卵黄囊和胎盘中也观察到与造血相关的类似簇。在这次审查中,我概述了这些集群的形成和功能,重点关注特征良好的主动脉内造血簇(IAHC)。
    Adult blood cells are produced in the bone marrow by hematopoietic stem cells (HSCs), the origin of which can be traced back to fetal developmental stages. Indeed, during mouse development, at days 10-11 of gestation, the aorta-gonad-mesonephros (AGM) region is a primary site of HSC production, with characteristic cell clusters related to stem cell genesis observed in the dorsal aorta. Similar clusters linked with hematopoiesis are also observed in the other sites such as the yolk sac and placenta. In this review, I outline the formation and function of these clusters, focusing on the well-characterized intra-aortic hematopoietic clusters (IAHCs).
    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
    血管内移植的骨髓细胞,包括骨髓单个核细胞(BM-MNC)和间充质干细胞,通过缝隙连接将水溶性分子转移到脑内皮细胞。移植BM-MNC后,这促进了海马神经发生和神经元功能的增强。在这里,我们报道了移植的BM-MNC对脑中神经干细胞(NSC)的影响。令人惊讶的是,细胞移植后10分钟,观察到水溶性分子从移植的BM-MNC和外周单核细胞直接转移到海马中的NSC,并且还观察到从BM-MNC向GFAP阳性皮质星形胶质细胞的转移。体外研究表明,BM-MNC消除了星形胶质细胞中HIF1α的表达。我们认为,循环中的细胞和大脑中的NSC之间的水溶性分子的瞬时和直接转移可能是脑功能修复的生物学机制之一。
    Intravascularly transplanted bone marrow cells, including bone marrow mononuclear cells (BM-MNC) and mesenchymal stem cells, transfer water-soluble molecules to cerebral endothelial cells via gap junctions. After transplantation of BM-MNC, this fosters hippocampal neurogenesis and enhancement of neuronal function. Herein, we report the impact of transplanted BM-MNC on neural stem cells (NSC) in the brain. Surprisingly, direct transfer of water-soluble molecules from transplanted BM-MNC and peripheral mononuclear cells to NSC in the hippocampus was observed already 10 min after cell transplantation, and transfer from BM-MNC to GFAP-positive cortical astrocytes was also observed. In vitro investigations revealed that BM-MNC abolish the expression of hypoxia-inducible factor-1α in astrocytes. We suggest that the transient and direct transfer of water-soluble molecules between cells in circulation and NSC in the brain may be one of the biological mechanisms underlying the repair of brain function.
    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
    在应激造血下,以前的研究表明造血干细胞(HSC)从骨髓(BM)迁移到髓外部位,如脾脏。然而,几乎没有直接证据表明HSC从BM迁移到脾脏。这里,我们通过5-氟尿嘧啶(5-FU)诱导骨髓消融,并显示了造血再生过程中HSC通过光转化荧光团从BM迁移到脾脏的直接证据。此外,在稳态期间,HSC优先迁移到BM而不是脾脏,但是在造血再生过程中,HSC更喜欢脾脏作为同等或更大的迁移位点。此外,在早期阶段,通过减弱的HSC保留从BM中排出的HSC。然而,晚期HSC获得了显著增强的细胞自主运动,这与趋化性无关。总的来说,在造血再生过程中,在迁移到脾脏之前,从BM动员的HSC从被动事件动态变化为主动事件。
    Under stress hematopoiesis, previous studies have suggested the migration of hematopoietic stem cells (HSCs) from bone marrow (BM) to extramedullary sites such as the spleen. However, there is little direct evidence of HSC migration from the BM to the spleen. Here, we induced myeloablation via 5-fluorouracil (5-FU) and showed direct evidence of HSC migration from BM to spleen during hematopoietic regeneration via a photoconvertible fluorophore. Moreover, during steady state, HSCs preferentially migrated to BM rather than spleen, but during hematopoietic regeneration, HSCs preferred to spleen as a migration site equivalently or greater. Furthermore, in the early phase, HSCs egressed from BM through the attenuated HSC retention. However, HSCs in the late phase gained significantly enhanced cell-autonomous motility, which was independent of chemotaxis. Collectively, HSC mobilization from BM before the migration to the spleen was dynamically changed from passive to active events during hematopoietic regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号