CP: stem cell research

CP: 干细胞研究
  • 文章类型: Journal Article
    繁殖对动物至关重要。为了成功,社会行为的协调,生理学,配子的生产是必要的。社会线索是如何感知的,它们是如何影响生理和配子生成的?而女性,从昆虫到哺乳动物,提供了关于这种协调的多种见解,它的存在在男性中仍然未知。这里,通过使用果蝇雄性作为模型,我们描述了一种现象,通过这种现象,潜在的交配伙伴的可用性触发了睾丸干细胞群的激活状态,促进精子发生.我们揭示了它对信息素通信的依赖,即使在没有交配或与雌性的其他互动的情况下。最后,我们确定了器官间通讯信号网络负责肌肉分泌的肿瘤坏死因子α(TNF-α)/Eiger和神经分泌的章鱼胺触发剂,分别,Jun氨基末端激酶(JNK)途径和囊肿干细胞中钙动力学的变化。因此,生殖系干细胞增加其增殖。
    Reproduction is paramount to animals. For it to be successful, a coordination of social behavior, physiology, and gamete production is necessary. How are social cues perceived and how do they affect physiology and gametogenesis? While females, ranging from insects to mammals, have provided multiple insights about this coordination, its existence remains largely unknown in males. Here, by using the Drosophila male as a model, we describe a phenomenon by which the availability of potential mating partners triggers an activation state on the stem cell populations of the testis, boosting spermatogenesis. We reveal its reliance on pheromonal communication, even in the absence of mating or other interactions with females. Finally, we identify the interorgan communication signaling network responsible-muscle-secreted tumor necrosis factor alpha (TNF-α)/Eiger and neuronally secreted octopamine trigger, respectively, the Jun N-terminal kinase (JNK) pathway and a change in calcium dynamics in the cyst stem cells. As a consequence, germ line stem cells increase their proliferation.
    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
    在老鼠身上,第一批肝脏驻留的巨噬细胞,被称为库普弗细胞(KCs),被认为来源于卵黄囊(YS)造血祖细胞,其在造血干细胞(HSC)出现之前被指定。为了研究人类KC的发育,我们从人类多能干细胞(hPSC)和移植衍生巨噬细胞祖细胞到先前用hPSC-肝窦内皮细胞(LSECs)人源化的NSG小鼠中进行了YS样造血。我们证明hPSC-LSEC促进稳定的hPSC-YS-巨噬细胞植入至少7周。移植的YS-巨噬细胞的单细胞RNA测序(scRNA-seq)揭示了均匀的MARCO表达KC基因签名和单核细胞样巨噬细胞基因的低表达。相比之下,人脐带血(CB)衍生的巨噬细胞祖细胞产生的移植物除KC外还包含多个造血谱系。功能分析显示移植的KC在体内主动进行吞噬作用和红细胞吞噬作用。一起来看,这些发现表明,有可能从hPSC衍生的人KC,YS样祖细胞。
    In mice, the first liver-resident macrophages, known as Kupffer cells (KCs), are thought to derive from yolk sac (YS) hematopoietic progenitors that are specified prior to the emergence of the hematopoietic stem cell (HSC). To investigate human KC development, we recapitulated YS-like hematopoiesis from human pluripotent stem cells (hPSCs) and transplanted derivative macrophage progenitors into NSG mice previously humanized with hPSC-liver sinusoidal endothelial cells (LSECs). We demonstrate that hPSC-LSECs facilitate stable hPSC-YS-macrophage engraftment for at least 7 weeks. Single-cell RNA sequencing (scRNA-seq) of engrafted YS-macrophages revealed a homogeneous MARCO-expressing KC gene signature and low expression of monocyte-like macrophage genes. In contrast, human cord blood (CB)-derived macrophage progenitors generated grafts that contain multiple hematopoietic lineages in addition to KCs. Functional analyses showed that the engrafted KCs actively perform phagocytosis and erythrophagocytosis in vivo. Taken together, these findings demonstrate that it is possible to generate human KCs from hPSC-derived, YS-like progenitors.
    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
    男性不育是一种公认的放化疗副作用。现有精原干细胞(SSC)可以作为任何后续回收的起源。然而,哪种类型的SSC,它们生存和抵抗毒性的机制,它们如何重新开始精子发生仍然是未知的。这里,我们鉴定了在小鼠生精小管中以相对休眠状态出现的少量表达含有Set结构域的蛋白4(Sed4)的SSC。超越高剂量放化疗,然后这些细胞激活以恢复精子发生。删除Setd4+SSC时恢复失败。确认是胎儿起源,这些Sett4+SSC被证明有助于早期睾丸发育,也有助于成年期稳态精子发生。激活后,染色质重塑增加了它们的全基因组可及性,使Notch1和极光激活与p21和p53相应的沉默。这里,Settd4SSC被认为是放化疗诱导的不育症中睾丸发育和生精恢复的起源。
    Male infertility is a recognized side effect of chemoradiotherapy. Extant spermatogonial stem cells (SSCs) may act as originators for any subsequent recovery. However, which type of SSCs, the mechanism by which they survive and resist toxicity, and how they act to restart spermatogenesis remain largely unknown. Here, we identify a small population of Set domain-containing protein 4 (Setd4)-expressing SSCs that occur in a relatively dormant state in the mouse seminiferous tubule. Extant beyond high-dose chemoradiotherapy, these cells then activate to recover spermatogenesis. Recovery fails when Setd4+ SSCs are deleted. Confirmed to be of fetal origin, these Setd4+ SSCs are shown to facilitate early testicular development and also contribute to steady-state spermatogenesis in adulthood. Upon activation, chromatin remodeling increases their genome-wide accessibility, enabling Notch1 and Aurora activation with corresponding silencing of p21 and p53. Here, Setd4+ SSCs are presented as the originators of both testicular development and spermatogenesis recovery in chemoradiotherapy-induced infertility.
    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
    响应急性损伤的伤口愈合是由实质的协调和短暂激活介导的,基质,和解决稳态的免疫细胞。Environmental,遗传,与炎症和衰老相关的表观遗传因素可导致微环境持续激活和纤维化。这里,我们确定了白细胞介素-4(IL-4)细胞因子信号在间质巨噬细胞和II型肺泡上皮细胞(ATII)中的相反作用.我们表明,在博来霉素诱导的肺损伤后,巨噬细胞中的IL4Ra信号促进肺泡上皮的再生。使用类器官和小鼠模型,我们显示IL-4直接作用于ATII的一部分,诱导转录因子SOX9的表达,并将其重新编程为具有气道和肺泡谱系潜能的祖细胞样状态.在衰老和博来霉素诱导的肺损伤的背景下,这导致上皮细胞异常分化和支气管扩张,与间质性肺病中观察到的细胞和组织学变化一致。
    Wound healing in response to acute injury is mediated by the coordinated and transient activation of parenchymal, stromal, and immune cells that resolves to homeostasis. Environmental, genetic, and epigenetic factors associated with inflammation and aging can lead to persistent activation of the microenvironment and fibrosis. Here, we identify opposing roles of interleukin-4 (IL-4) cytokine signaling in interstitial macrophages and type II alveolar epithelial cells (ATIIs). We show that IL4Ra signaling in macrophages promotes regeneration of the alveolar epithelium after bleomycin-induced lung injury. Using organoids and mouse models, we show that IL-4 directly acts on a subset of ATIIs to induce the expression of the transcription factor SOX9 and reprograms them toward a progenitor-like state with both airway and alveolar lineage potential. In the contexts of aging and bleomycin-induced lung injury, this leads to aberrant epithelial cell differentiation and bronchiolization, consistent with cellular and histological changes observed in interstitial lung disease.
    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
    粒细胞集落刺激因子(G-CSF)广泛用于增强化疗后的骨髓恢复和动员造血干细胞(HSC)进行移植。不幸的是,在化疗过程中,癌症患者可以获得引起治疗相关骨髓增生异常综合征(MDS)或急性髓细胞性白血病(AML)的白血病性突变.这提出了一个问题,即治疗性G-CSF是否可以通过不成比例地刺激突变HSC和其他骨髓祖细胞来增强与治疗相关的MDS/AML。治疗相关的MDS/AML中的常见突变涉及染色体7缺失,使许多肿瘤抑制基因失活。包括KMT2C。这里,我们显示Kmt2c缺失使小鼠HSC和骨髓祖细胞对G-CSF过敏,HSC动员增加,粒细胞-单核细胞祖细胞(GMPs)的粒细胞产生增加。此外,Kmt2c独立于其SET甲基转移酶功能减弱G-CSF应答。总之,这些数据引起了人们的担忧,即单体7可以使祖细胞对G-CSF过敏,因此,临床使用G-CSF可能会增加治疗相关MDS/AML的风险。
    Granulocyte colony-stimulating factor (G-CSF) is widely used to enhance myeloid recovery after chemotherapy and to mobilize hematopoietic stem cells (HSCs) for transplantation. Unfortunately, through the course of chemotherapy, cancer patients can acquire leukemogenic mutations that cause therapy-related myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). This raises the question of whether therapeutic G-CSF might potentiate therapy-related MDS/AML by disproportionately stimulating mutant HSCs and other myeloid progenitors. A common mutation in therapy-related MDS/AML involves chromosome 7 deletions that inactivate many tumor suppressor genes, including KMT2C. Here, we show that Kmt2c deletions hypersensitize murine HSCs and myeloid progenitors to G-CSF, as evidenced by increased HSC mobilization and enhanced granulocyte production from granulocyte-monocyte progenitors (GMPs). Furthermore, Kmt2c attenuates the G-CSF response independently from its SET methyltransferase function. Altogether, the data raise concerns that monosomy 7 can hypersensitize progenitors to G-CSF, such that clinical use of G-CSF may amplify the risk of therapy-related MDS/AML.
    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
    子宫内膜蛋白(EMCN)目前代表鼠和人HSC表达的唯一造血干细胞(HSC)标记。这里,我们报道,与EMCN-LT-HSCs相比,EMCN+长期再增殖HSCs(LT-HSCs;CD150+CD48-LSK)具有更高的长期多谱系再增殖能力.细胞周期分析和转录谱分析表明EMCN+LT-HSC与EMCN-LT-HSC相比更静止。Emcn-/-和Emcn+/+小鼠表现出相当的稳态造血,以及频率,转录程序,和他们的LT-HSC的长期多谱系再繁殖能力。补充功能分析进一步显示,用5-氟尿嘧啶处理后细胞周期进入增加,粒细胞集落刺激因子(GCSF)动员Emcn-/-LT-HSC减少,证明LT-HSC的EMCN表达与响应造血应激的静止相关,对于有效的LT-HSC动员是必不可少的。将野生型骨髓细胞移植到Emcn-/-或Emcn/受体中表明,EMCN对于LT-HSC池的内皮细胞依赖性维持/自我更新和移植后持续的血细胞产生至关重要。
    Endomucin (EMCN) currently represents the only hematopoietic stem cell (HSC) marker expressed by both murine and human HSCs. Here, we report that EMCN+ long-term repopulating HSCs (LT-HSCs; CD150+CD48-LSK) have a higher long-term multi-lineage repopulating capacity compared to EMCN- LT-HSCs. Cell cycle analyses and transcriptional profiling demonstrated that EMCN+ LT-HSCs were more quiescent compared to EMCN- LT-HSCs. Emcn-/- and Emcn+/+ mice displayed comparable steady-state hematopoiesis, as well as frequencies, transcriptional programs, and long-term multi-lineage repopulating capacity of their LT-HSCs. Complementary functional analyses further revealed increased cell cycle entry upon treatment with 5-fluorouracil and reduced granulocyte colony-stimulating factor (GCSF) mobilization of Emcn-/- LT-HSCs, demonstrating that EMCN expression by LT-HSCs associates with quiescence in response to hematopoietic stress and is indispensable for effective LT-HSC mobilization. Transplantation of wild-type bone marrow cells into Emcn-/- or Emcn+/+ recipients demonstrated that EMCN is essential for endothelial cell-dependent maintenance/self-renewal of the LT-HSC pool and sustained blood cell production post-transplant.
    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
    与大多数造血谱系相反,巨核细胞(MK)可以快速直接从造血干细胞(HSC)中获得。潜在的机制尚不清楚,however.这里,我们显示DNA损伤诱导HSC中的MK标记和G2阻滞,DNA损伤反应不可或缺的一部分,在HSC中MK引发随后不可逆的MK分化就足够了,但不是在祖细胞中。我们还表明,复制应激导致HSC中的DNA损伤,并且至少部分是由于尿嘧啶在体外和体内的错误掺入。与这个概念一致,胸苷减弱DNA损伤,改善HSC维护,并减少CD41+MK定向HSC的产生。因此,复制胁迫和伴随的MK分化是HSC维持的障碍之一。DNA损伤诱导的MK引发可能允许快速生成对立即生物体存活至关重要的谱系,同时还从HSC池中移除受损的细胞。
    In contrast to most hematopoietic lineages, megakaryocytes (MKs) can derive rapidly and directly from hematopoietic stem cells (HSCs). The underlying mechanism is unclear, however. Here, we show that DNA damage induces MK markers in HSCs and that G2 arrest, an integral part of the DNA damage response, suffices for MK priming followed by irreversible MK differentiation in HSCs, but not in progenitors. We also show that replication stress causes DNA damage in HSCs and is at least in part due to uracil misincorporation in vitro and in vivo. Consistent with this notion, thymidine attenuated DNA damage, improved HSC maintenance, and reduced the generation of CD41+ MK-committed HSCs. Replication stress and concomitant MK differentiation is therefore one of the barriers to HSC maintenance. DNA damage-induced MK priming may allow rapid generation of a lineage essential to immediate organismal survival, while also removing damaged cells from the HSC pool.
    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
    人类诱导多能干细胞(hiPSC)来源的肠道类器官是研究发育生物学和个性化治疗的有价值的工具,但是它们的封闭拓扑和相对不成熟的状态限制了应用。这里,我们使用芯片上器官技术在更生理的体外微环境中开发出具有顶端和基底外侧通路的hiPSC衍生肠屏障。为了沿着隐窝-绒毛轴复制生长因子梯度,我们将细胞局部暴露于扩增和分化培养基中。在这些条件下,肠上皮细胞自组织成具有生理屏障完整性的绒毛样褶皱,肌成纤维细胞和神经元在底部通道中出现并形成上皮下组织。生长因子梯度有效地平衡分裂和成熟细胞类型,并诱导肠上皮成分,包括吸收和分泌谱系,类似于人类小肠的成分。这种特征良好的hiPSC衍生的芯片肠系统可以促进对人类小肠中的生理过程和治疗开发的个性化研究。
    Human induced pluripotent stem cell (hiPSC)-derived intestinal organoids are valuable tools for researching developmental biology and personalized therapies, but their closed topology and relative immature state limit applications. Here, we use organ-on-chip technology to develop a hiPSC-derived intestinal barrier with apical and basolateral access in a more physiological in vitro microenvironment. To replicate growth factor gradients along the crypt-villus axis, we locally expose the cells to expansion and differentiation media. In these conditions, intestinal epithelial cells self-organize into villus-like folds with physiological barrier integrity, and myofibroblasts and neurons emerge and form a subepithelial tissue in the bottom channel. The growth factor gradients efficiently balance dividing and mature cell types and induce an intestinal epithelial composition, including absorptive and secretory lineages, resembling the composition of the human small intestine. This well-characterized hiPSC-derived intestine-on-chip system can facilitate personalized studies on physiological processes and therapy development in the human small intestine.
    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
    新的证据强调了配对样(PRD样)同源盒转录因子(TF)在胚胎基因组激活(EGA)中的调节作用。然而,大多数PRD样基因在啮齿动物中丢失,从而促使对其他哺乳动物的PRD样TFs进行研究。这里,我们表明,PRD样TFs在人的EGA过程中短暂表达,猴子,和猪受精的胚胎,然而他们在克隆的胚胎中表现出表达不足。这项研究,以猪为研究模型,通过基因组分析确定LEUTX是猪EGA的关键PRD样激活剂,并发现LEUTX过表达恢复了EGA失败并改善了猪克隆胚胎的植入前发育和克隆效率。机械上,LEUTX通过募集乙酰转移酶p300和KAT2A打开EGA相关基因组区域并建立组蛋白乙酰化。这些发现揭示了LEUTX对猪EGA的调控机制,这可能为其他非啮齿动物哺乳动物的早期胚胎发育研究提供有价值的见解。
    Emerging evidence highlights the regulatory role of paired-like (PRD-like) homeobox transcription factors (TFs) in embryonic genome activation (EGA). However, the majority of PRD-like genes are lost in rodents, thus prompting an investigation into PRD-like TFs in other mammals. Here, we showed that PRD-like TFs were transiently expressed during EGA in human, monkey, and porcine fertilized embryos, yet they exhibited inadequate expression in their cloned embryos. This study, using pig as the research model, identified LEUTX as a key PRD-like activator of porcine EGA through genomic profiling and found that LEUTX overexpression restored EGA failure and improved preimplantation development and cloning efficiency in porcine cloned embryos. Mechanistically, LEUTX opened EGA-related genomic regions and established histone acetylation via recruiting acetyltransferases p300 and KAT2A. These findings reveal the regulatory mechanism of LEUTX to govern EGA in pigs, which may provide valuable insights into the study of early embryo development for other non-rodent mammals.
    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