Self-renewal

自我更新
  • 文章类型: Journal Article
    干细胞生态位对于调节干细胞的行为至关重要。果蝇神经干细胞(神经母细胞,NBs)被胶质细胞紧密包裹,但目前尚不清楚神经胶质细胞是否能调节NBs的自我更新和分化。这里,我们发现由胶质细胞产生的铁蛋白,与Zip13合作将铁运输到NB中用于能源生产,这对NB的自我更新和增殖至关重要。神经胶质铁蛋白编码基因的敲除通过下调乌头酸酶活性和NAD+水平导致NBs能量短缺,这导致Prospero进入细胞核介导的NBs的低增殖和过早分化。更重要的是,铁蛋白是肿瘤抑制的潜在靶点。此外,胶质铁蛋白的产生水平受NBs状态的影响,建立双细胞铁稳态。在这项研究中,我们证明神经胶质细胞对维持NBs的自我更新是必不可少的,揭示了NB胶质细胞在大脑发育过程中的新作用。
    铁是几乎所有生物体的必需营养素。例如,铁有助于DNA的复制,细胞内能量的产生,以及氧气在身体周围的运输。缺铁是所有营养缺乏中最常见的,影响全世界超过40%的儿童。这会导致贫血,也会损害大脑和神经系统的发育,可能导致长期的认知损伤,即使在缺乏治疗之后。人们对铁如何促进大脑和神经系统的发育知之甚少。特别是,它是否以及如何支持神经干细胞(或简称NSC),这些神经干细胞在成熟的大脑中产生各种神经类型。为了调查,Maetal.通过实验降低了果蝇幼虫发育中大脑中铁蛋白(一种储存铁的蛋白质)的水平。铁蛋白的这种减少导致NSC的数量减少和大脑变小。出乎意料的是,当支持神经干细胞并向神经干细胞发送信号的神经胶质细胞中铁蛋白水平降低时,这种作用最大,而不是干细胞本身。Maetal.然后用荧光显微镜证实胶质细胞制造并含有大量的铁蛋白,这些铁蛋白可以转运到神经干细胞。在缺乏铁蛋白的果蝇的饮食中添加铁补充剂不会导致发育中的果蝇大脑中的干细胞数量正常,而添加减少铁含量的化合物导致干细胞数量减少。一起,这表明铁蛋白将铁从神经胶质细胞转运至神经干细胞。没有铁蛋白和铁,NSC无法产生足够的能量来分裂和制造新的干细胞。这导致神经干细胞失去干细胞的特征,并过早地转变为其他类型的神经元或神经胶质细胞。一起,这些发现表明,当铁不能从神经胶质细胞转移到神经干细胞时,这将导致大脑发育的缺陷。未来的实验将必须测试在哺乳动物发育中的大脑中是否也发生类似的铁从支持细胞到神经干细胞的运输。以及这种机制是否适用于身体其他部位的干细胞。
    Stem cell niche is critical for regulating the behavior of stem cells. Drosophila neural stem cells (Neuroblasts, NBs) are encased by glial niche cells closely, but it still remains unclear whether glial niche cells can regulate the self-renewal and differentiation of NBs. Here, we show that ferritin produced by glia, cooperates with Zip13 to transport iron into NBs for the energy production, which is essential to the self-renewal and proliferation of NBs. The knockdown of glial ferritin encoding genes causes energy shortage in NBs via downregulating aconitase activity and NAD+ level, which leads to the low proliferation and premature differentiation of NBs mediated by Prospero entering nuclei. More importantly, ferritin is a potential target for tumor suppression. In addition, the level of glial ferritin production is affected by the status of NBs, establishing a bicellular iron homeostasis. In this study, we demonstrate that glial cells are indispensable to maintain the self-renewal of NBs, unveiling a novel role of the NB glial niche during brain development.
    Iron is an essential nutrient for almost all living organisms. For example, iron contributes to the replication of DNA, the generation of energy inside cells, and the transport of oxygen around the body. Iron deficiency is the most common of all nutrient deficiencies, affecting over 40% of children worldwide. This can lead to anemia and also impair how the brain and nervous system develop, potentially resulting in long-lasting cognitive damage, even after the deficiency has been treated. It is poorly understood how iron contributes to the development of the brain and nervous system. In particular, whether and how it supports nerve stem cells (or NSCs for short) which give rise to the various neural types in the mature brain. To investigate, Ma et al. experimentally reduced the levels of ferritin (a protein which stores iron) in the developing brains of fruit fly larvae. This reduction in ferritin led to lower numbers of NSCs and a smaller brain. Unexpectedly, this effect was largest when ferritin levels were reduced in glial cells which support and send signals to NSCs, rather than in the stem cells themselves. Ma et al. then used fluorescence microscopy to confirm that glial cells make and contain a lot of ferritin which can be transported to NSCs. Adding iron supplements to the diet of flies lacking ferritin did not lead to normal numbers of stem cells in the brains of the developing fruit flies, whereas adding compounds that reduce the amount of iron led to lower numbers of stem cells. Together, this suggests that ferritin transports iron from glial cells to the NSCs. Without ferritin and iron, the NSCs could not produce enough energy to divide and make new stem cells. This caused the NSCs to lose the characteristics of stem cells and prematurely turn into other types of neurons or glial cells. Together, these findings show that when iron cannot move from glial cells to NSCs this leads to defects in brain development. Future experiments will have to test whether a similar transport of iron from supporting cells to NSCs also occurs in the developing brains of mammals, and whether this mechanism applies to stem cells in other parts of the body.
    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
    人精原干细胞(SSCs)具有良好的可塑性,在生殖医学和再生医学中具有重要的应用价值。然而,长链非编码RNA(LncRNA)在调节人类SSC命运决定中的功能和机制仍然未知。在这里,我们已经证明LncRNAACVR2B-as1(激活素A受体2B型反义RNA1)通过与ALDOA相互作用通过糖酵解活性控制人SSC的自我更新和凋亡。LncRNAACVR2B-as1在人SSC中高度表达。LncRNAACVR2B-as1沉默抑制人SSCs的增殖和DNA合成并增强其凋亡。机械上,我们的ChIRP-MS和RIP分析显示,ACVR2B-as1与人SSC中的ALDOA相互作用。ACVR2B-as1的高表达增强了细胞增殖,DNA合成,和人SSC的糖酵解,但通过上调ALDOA抑制其凋亡。重要的是,ALDOA的过表达抵消了ACVR2B-as1敲低对上述生物过程的影响。总的来说,这些结果表明,ACVR2B-as1与ALDOA相互作用,通过增强糖酵解活性来控制人SSC的自我更新和凋亡。这项研究具有重要意义,因为它为人类SSC命运决定的分子机制提供了新的见解,并且可能为解决男性不育的病因提供创新的方法。
    Human spermatogonial stem cells (SSCs) have significant applications in reproductive medicine and regenerative medicine because of their great plasticity. Nevertheless, it remains unknown about the functions and mechanisms of long non-coding RNA (LncRNA) in regulating the fate determinations of human SSCs. Here we have demonstrated that LncRNA ACVR2B-as1 (activin A receptor type 2B antisense RNA 1) controls the self-renewal and apoptosis of human SSCs by interaction with ALDOA via glycolysis activity. LncRNA ACVR2B-as1 is highly expressed in human SSCs. LncRNA ACVR2B-as1 silencing suppresses the proliferation and DNA synthesis and enhances the apoptosis of human SSCs. Mechanistically, our ChIRP-MS and RIP assays revealed that ACVR2B-as1 interacted with ALDOA in human SSCs. High expression of ACVR2B-as1 enhanced the proliferation, DNA synthesis, and glycolysis of human SSCs but inhibited their apoptosis through up-regulation of ALDOA. Importantly, overexpression of ALDOA counteracted the effect of ACVR2B-as1 knockdown on the aforementioned biological processes. Collectively, these results indicate that ACVR2B-as1 interacts with ALDOA to control the self-renewal and apoptosis of human SSCs by enhancing glycolysis activity. This study is of great significance because it sheds a novel insight into molecular mechanisms underlying the fate decisions of human SSCs and it may offer innovative approaches to address the etiology of male 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
    肝细胞癌(HCC)的进展受代谢过程中断的影响,在预后结果方面提出了挑战。肝细胞癌(HCC),癌症相关死亡的主要原因,与肝癌干细胞(LCSC)的代谢重编程和干细胞样特性密切相关。这项研究探索了tLyP-1修饰的细胞外囊泡(EV)递送CTCFshRNA(tLyp-1-EV-shCTCF)调节线粒体DNA甲基化诱导的糖酵解代谢重编程和LCSC自我更新的潜在分子机制。通过一系列的方法,包括蛋白质印迹,纳米粒子跟踪分析,和免疫荧光,我们证明了tLyp-1-EV在HCC细胞中的成功递送和内化。我们的结果确定SALL3是在HCC和LCSC中表达不足的关键因素,而CTCF过度表达。SALL3过表达通过阻断CTCF-DNMT3A相互作用抑制LCSC自我更新和免疫逃避,从而抑制DNMT3A甲基转移酶活性和随后的线粒体DNA甲基化介导的糖酵解代谢重编程。体内实验进一步支持了这些发现,显示tLyp-1-EV-shCTCF治疗通过上调SALL3表达显著降低肿瘤生长,从而抑制糖酵解代谢重编程并增强针对HCC细胞的免疫应答。这项研究为SALL3和线粒体DNA甲基化在HCC进展中的作用提供了新的见解,为对抗HCC及其干细胞样特性提供潜在的治疗靶点。
    The progression of hepatocellular carcinoma (HCC) is influenced by disrupted metabolic processes, presenting challenges in prognostic outcomes. Hepatocellular carcinoma (HCC), a leading cause of cancer-related mortality, is closely associated with metabolic reprogramming and stem cell-like properties in liver cancer stem cells (LCSCs). This study explored the potential molecular mechanisms by which tLyP-1-modified extracellular vesicles (EVs) delivering CTCF shRNA (tLyp-1-EV-shCTCF) regulate mitochondrial DNA methylation-induced glycolytic metabolic reprogramming and LCSC self-renewal. Through a series of methods, including Western blot, nanoparticle tracking analysis, and immunofluorescence, we demonstrated the successful delivery and internalization of tLyp-1-EV in HCC cells. Our results identified SALL3 as a critical factor underexpressed in HCC and LCSCs, while CTCF was overexpressed. Overexpression of SALL3 inhibited LCSC self-renewal and immune evasion by blocking the CTCF-DNMT3A interaction, thus repressing DNMT3A methyltransferase activity and subsequent mitochondrial DNA methylation-mediated glycolytic metabolic reprogramming. In vivo experiments further supported these findings, showing that tLyp-1-EV-shCTCF treatment significantly reduced tumor growth by upregulating SALL3 expression, thereby inhibiting glycolytic metabolic reprogramming and enhancing the immune response against HCC cells. This study provides novel insights into the role of SALL3 and mitochondrial DNA methylation in HCC progression, offering potential therapeutic targets for combating HCC and its stem cell-like properties.
    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
    背景:这项研究调查了CXXC5在骨髓微环境中造血干细胞(HSC)自我更新和分化中的作用,利用先进的方法,如单细胞RNA测序(scRNA-seq),CRISPR-Cas9和蛋白质组学分析。
    方法:我们采用流式细胞术从骨髓样本中分离HSC,然后使用10x基因组学平台进行scRNA-seq分析,以检查细胞聚类和CXXC5表达模式。CRISPR-Cas9和慢病毒载体促进HSC中CXXC5的敲除和过表达。通过qRT-PCR评估对HSC的影响,蛋白质印迹,CCK-8,CFU,和LTC-IC分析,与流式细胞术一起测量细胞凋亡和细胞比例。还使用小鼠模型来评估CXXC5操作对HSC植入和存活率的影响。
    结果:我们的发现强调了细胞聚集的多样性以及CXXC5在HSC调节中的重要作用。基因敲除实验显示增殖减少,分化加速,而过度表达导致增殖增强和分化延迟。蛋白质组学分析确定了受CXXC5影响的关键生物过程,包括细胞增殖,分化,和凋亡。体内结果表明,CXXC5沉默会损害骨髓移植模型中的HSC移植。
    结论:CXXC5对于调节骨髓微环境中的HSC自我更新和分化至关重要。它的操作提供了一种增强HSC功能的新方法,并为血液病提供了潜在的治疗靶标。
    BACKGROUND: This study investigates the role of CXXC5 in the self-renewal and differentiation of hematopoietic stem cells (HSCs) within the bone marrow microenvironment, utilizing advanced methodologies such as single-cell RNA sequencing (scRNA-seq), CRISPR-Cas9, and proteomic analysis.
    METHODS: We employed flow cytometry to isolate HSCs from bone marrow samples, followed by scRNA-seq analysis using the 10x Genomics platform to examine cell clustering and CXXC5 expression patterns. CRISPR-Cas9 and lentiviral vectors facilitated the knockout and overexpression of CXXC5 in HSCs. The impact on HSCs was assessed through qRT-PCR, Western blot, CCK-8, CFU, and LTC-IC assays, alongside flow cytometry to measure apoptosis and cell proportions. A mouse model was also used to evaluate the effects of CXXC5 manipulation on HSC engraftment and survival rates.
    RESULTS: Our findings highlight the diversity of cell clustering and the significant role of CXXC5 in HSC regulation. Knockout experiments showed reduced proliferation and accelerated differentiation, whereas overexpression led to enhanced proliferation and delayed differentiation. Proteomic analysis identified key biological processes influenced by CXXC5, including cell proliferation, differentiation, and apoptosis. In vivo results demonstrated that CXXC5 silencing impaired HSC engraftment in a bone marrow transplantation model.
    CONCLUSIONS: CXXC5 is crucial for the regulation of HSC self-renewal and differentiation in the bone marrow microenvironment. Its manipulation presents a novel approach for enhancing HSC function and provides a potential therapeutic target for hematological diseases.
    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
    生殖系干细胞是一种至关重要的干细胞类型,可以稳定地将遗传信息传递给下一代,为生物的繁殖和生存提供必要的基础。雄性哺乳动物生殖系干细胞是独特的细胞类型,包括原始生殖细胞和精原干细胞。它们可以分化成生殖细胞,比如精子和卵子,从而促进后代繁殖。此外,它们通过自我更新机制不断产生干细胞,以支持生殖系统的正常功能。自噬涉及使用溶酶体降解由相关基因调节的蛋白质和细胞器。该过程在维持生殖干细胞的稳态和合成中起着重要作用。降解,和生殖干细胞产品的回收。最近,生殖干细胞的发育调控机制得到进一步阐明,和自噬已被证明参与调节生殖系干细胞的自我更新和分化。在这次审查中,我们引入自噬伴随生殖干细胞的发育,重点研究了伴随男性精原干细胞发育的自噬过程以及相关基因和蛋白的作用。我们还简要概述了自噬功能障碍对生殖干细胞和生殖的影响。
    Germline stem cells are a crucial type of stem cell that can stably pass on genetic information to the next generation, providing the necessary foundation for the reproduction and survival of organisms. Male mammalian germline stem cells are unique cell types that include primordial germ cells and spermatogonial stem cells. They can differentiate into germ cells, such as sperm and eggs, thereby facilitating offspring reproduction. In addition, they continuously generate stem cells through self-renewal mechanisms to support the normal function of the reproductive system. Autophagy involves the use of lysosomes to degrade proteins and organelles that are regulated by relevant genes. This process plays an important role in maintaining the homeostasis of germline stem cells and the synthesis, degradation, and recycling of germline stem cell products. Recently, the developmental regulatory mechanism of germline stem cells has been further elucidated, and autophagy has been shown to be involved in the regulation of self-renewal and differentiation of germline stem cells. In this review, we introduce autophagy accompanying the development of germline stem cells, focusing on the autophagy process accompanying the development of male spermatogonial stem cells and the roles of related genes and proteins. We also briefly outline the effects of autophagy dysfunction on germline stem cells and reproduction.
    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
    克隆造血(CH),突变克隆的相对扩增,来自造血干细胞(HSC),具有获得性体细胞或细胞遗传学改变,可改善细胞适应性。患有CH的个体患血液病和非血液病的风险较高,比如心血管疾病,总体死亡率较高。最初被认为仅限于一小部分老年人,单细胞测序和生物信息学的最新进展表明,具有多个扩展突变克隆的CH在老年人群中普遍存在。就在几年前,人类生命周期的系统发育重建和新的敏感测序技术表明,CH可以在生命早期开始,几十年前,它被认为是可能的。这些研究还表明,通过异常炎症起作用的环境因素可能是促进克隆扩展和疾病进展的共同主题。然而,这种现象的许多方面仍有待阐明,确切的机制,特定于上下文的驱动程序,和克隆扩增的途径仍有待建立。这里,我们回顾了我们目前对驱动CH的细胞机制的理解,并特别关注促炎因子如何影响正常和突变的HSC命运以促进克隆选择.
    Clonal hematopoiesis (CH), the relative expansion of mutant clones, is derived from hematopoietic stem cells (HSCs) with acquired somatic or cytogenetic alterations that improve cellular fitness. Individuals with CH have a higher risk for hematological and non-hematological diseases, such as cardiovascular disease, and have an overall higher mortality rate. Originally thought to be restricted to a small fraction of elderly people, recent advances in single-cell sequencing and bioinformatics have revealed that CH with multiple expanded mutant clones is universal in the elderly population. Just a few years ago, phylogenetic reconstruction across the human lifespan and novel sensitive sequencing techniques showed that CH can start earlier in life, decades before it was thought possible. These studies also suggest that environmental factors acting through aberrant inflammation might be a common theme promoting clonal expansion and disease progression. However, numerous aspects of this phenomenon remain to be elucidated and the precise mechanisms, context-specific drivers, and pathways of clonal expansion remain to be established. Here, we review our current understanding of the cellular mechanisms driving CH and specifically focus on how pro-inflammatory factors affect normal and mutant HSC fates to promote clonal selection.
    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:建立稳定的猪胚胎干细胞(pESCs)有助于基础和生物医学研究,包括比较发育生物学,以及评估干细胞疗法的安全性。尽管有这些优势,从体外囊胚获得的大多数pESCs需要复杂的培养基和饲养层,常规使用,基因改造,和分化为特定的细胞类型困难。我们旨在建立具有单细胞传代能力的pESCs,高增殖潜能,并且使用简化的无血清培养基从体外来源的胚泡长期培养中稳定。
    方法:我们使用各种基础培养基(DMEM/F10(1:1),DMEM/F12和a-MEM)和因子(FGF2,IWR-1,CHIR99021和WH-4-023)。在饲养或无饲养条件下分析建立的pESC的多能性和自我更新能力。最终,我们在无血清条件下开发了由FGF2,IWR-1和WH-4-023组成的简化培养基(FIW)。
    结果:pESC-FIW细胞系能够以短细胞倍增时间进行单细胞传代,并表达多能性标记POU5F1,SOX2和NANOG,以及细胞表面标记SSEA1、SSEA4和TRA-1-60。pESC-FIW显示稳定的增殖速率和正常的核型,即使经过50个通道。转录组分析显示,pESC-FIW与报道的在复杂培养基中维持的pESC相似,并显示出胃泌素上胚细胞特征。使用mTeSR™在纤连蛋白包被的平板上在无饲养条件下维持pESC-FIW多次传代,用于无饲养培养的商业培养基,表现出与在饲养条件下观察到的特征相似的特征。
    结论:这些结果表明,WNT和SRC的抑制足以建立能够在无血清条件下进行单细胞传代和无饲养细胞扩增的pESC。pESCs易于维护,有利于其在农业和生物医学基因编辑技术中的应用。以及血统承诺研究。
    BACKGROUND: The establishment of stable porcine embryonic stem cells (pESCs) can contribute to basic and biomedical research, including comparative developmental biology, as well as assessing the safety of stem cell-based therapies. Despite these advantages, most pESCs obtained from in vitro blastocysts require complex media and feeder layers, making routine use, genetic modification, and differentiation into specific cell types difficult. We aimed to establish pESCs with a single cell-passage ability, high proliferative potency, and stable in long-term culture from in vitro-derived blastocysts using a simplified serum-free medium.
    METHODS: We evaluated the establishment efficiency of pESCs from in vitro blastocysts using various basal media (DMEM/F10 (1:1), DMEM/F12, and a-MEM) and factors (FGF2, IWR-1, CHIR99021, and WH-4-023). The pluripotency and self-renewal capacity of the established pESCs were analyzed under feeder or feeder-free conditions. Ultimately, we developed a simplified culture medium (FIW) composed of FGF2, IWR-1, and WH-4-023 under serum-free conditions.
    RESULTS: The pESC-FIW lines were capable of single-cell passaging with short cell doubling times and expressed the pluripotency markers POU5F1, SOX2, and NANOG, as well as cell surface markers SSEA1, SSEA4, and TRA-1-60. pESC-FIW showed a stable proliferation rate and normal karyotype, even after 50 passages. Transcriptome analysis revealed that pESC-FIW were similar to reported pESC maintained in complex media and showed gastrulating epiblast cell characteristics. pESC-FIW were maintained for multiple passages under feeder-free conditions on fibronectin-coated plates using mTeSR™, a commercial medium used for feeder-free culture, exhibiting characteristics similar to those observed under feeder conditions.
    CONCLUSIONS: These results indicated that inhibition of WNT and SRC was sufficient to establish pESCs capable of single-cell passaging and feeder-free expansion under serum-free conditions. The easy maintenance of pESCs facilitates their application in gene editing technology for agriculture and biomedicine, as well as lineage commitment studies.
    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)的功能受HSC内部信号通路及其微环境的调节。趋化因子和趋化因子配体在调节HSC功能中起重要作用。然而,它们在HSC中的功能尚未完全了解。
    方法:我们建立了Cxcr3和Cxcl10敲除小鼠模型(Cxcr3-/-和Cxcl10-/-),以分析Cxcr3或Cxcl10在调节HSC功能中的作用。通过流式细胞术评估LT-HSC的细胞周期分布。Cxcr3-/-和Cxcl10-/-干/祖细胞显示降低的自我更新能力,如在连续移植测定中所测量的。为了研究缺乏Cxcr3或Cxcl10的骨髓微环境的影响,我们将CD45.1供体细胞移植到Cxcr3-/-或Cxcl10-/-受体小鼠(CD45.2)中,并检查了供体贡献的造血功能。
    结果:缺乏Cxcl10及其受体Cxcr3导致小鼠BM细胞减少,LT-HSC比例显著增加。Cxcl10-/-干/祖细胞在二次移植测定中显示降低的自我更新能力。值得注意的是,Cxcl10-/-供体来源的细胞优先分化为B淋巴细胞,有髓样分化能力的偏斜。同时,Cxcr3缺陷型HSC在二次移植中表现出重建缺点,但谱系偏差不显著。有趣的是,骨髓微环境中不存在Cxcl10或Cxcr3并不影响HSC功能.
    结论:Cxcl10和Cxcr3调节HSC的功能,包括自我更新和分化,增加了对趋化因子在调节HSC功能中的作用的理解。
    BACKGROUND: The function of hematopoietic stem cells (HSC) is regulated by HSC internal signaling pathways and their microenvironment. Chemokines and chemokine ligands play important roles in the regulation of HSC function. Yet, their functions in HSC are not fully understood.
    METHODS: We established Cxcr3 and Cxcl10 knockout mouse models (Cxcr3-/- and Cxcl10-/-) to analyze the roles of Cxcr3 or Cxcl10 in regulating HSC function. The cell cycle distribution of LT-HSC was assessed via flow cytometry. Cxcr3-/- and Cxcl10-/- stem/progenitor cells showed reduced self-renewal capacity as measured in serial transplantation assays. To study the effects of Cxcr3 or Cxcl10 deficient bone marrow microenvironment, we transplanted CD45.1 donor cells into Cxcr3-/-or Cxcl10-/- recipient mice (CD45.2) and examined donor-contributed hematopoiesis.
    RESULTS: Deficiency of Cxcl10 and its receptor Cxcr3 led to decreased BM cellularity in mice, with a significantly increased proportion of LT-HSC. Cxcl10-/- stem/progenitor cells showed reduced self-renewal capacity in the secondary transplantation assay. Notably, Cxcl10-/- donor-derived cells preferentially differentiated into B lymphocytes, with skewed myeloid differentiation ability. Meanwhile, Cxcr3-deficient HSCs demonstrated a reconstitution disadvantage in secondary transplantation, but the lineage bias was not significant. Interestingly, the absence of Cxcl10 or Cxcr3 in bone marrow microenvironment did not affect HSC function.
    CONCLUSIONS: The Cxcl10 and Cxcr3 regulate the function of HSC, including self-renewal and differentiation, adding to the understanding of the roles of chemokines in the regulation of HSC function.
    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
    简介:小鼠胚胎干细胞(ESC)的自我更新可以通过GSK3和MEK激酶的双重抑制来维持。MEK有两个高度同源的下游激酶,细胞外信号调节激酶1和2(ERK1/2)。然而,ERK1/2在小鼠ESC自我更新和分化中的确切作用尚不清楚.方法:我们使用遗传和化学遗传方法结合小分子抑制剂选择性删除或抑制ERK1,ERK2或两者。然后评估ERK旁系物特异性抑制对小鼠ESC自我更新和分化的影响。结果:发现ERK1/2对于小鼠ESC存活和自我更新是不必要的。两种ERK旁系同源物的抑制,结合GSK3抑制,足以维持小鼠ESC的自我更新。相比之下,仅选择性缺失或抑制一个ERK类似物并不能模拟MEK抑制在促进小鼠ESC自我更新中的作用。关于ESC差异化,抑制ERK1/2阻止了中内胚层分化。此外,选择性抑制ERK1而非ERK2促进了中内胚层分化。讨论:这些发现表明ERK1和ERK2在调节ESC自我更新和分化中具有重叠和不同的作用。这项研究为ERK1/2在控制ESC维持和谱系承诺方面的分子机制提供了新的见解。可能为未来在研究和治疗应用中控制干细胞命运的策略提供信息。
    Introduction: Mouse embryonic stem cell (ESC) self-renewal can be maintained through dual inhibition of GSK3 and MEK kinases. MEK has two highly homologous downstream kinases, extracellular signal-regulated kinase 1 and 2 (ERK1/2). However, the exact roles of ERK1/2 in mouse ESC self-renewal and differentiation remain unclear. Methods: We selectively deleted or inhibited ERK1, ERK2, or both using genetic and chemical genetic approaches combined with small molecule inhibitors. The effects of ERK paralog-specific inhibition on mouse ESC self-renewal and differentiation were then assessed. Results: ERK1/2 were found to be dispensable for mouse ESC survival and self-renewal. The inhibition of both ERK paralogs, in conjunction with GSK3 inhibition, was sufficient to maintain mouse ESC self-renewal. In contrast, selective deletion or inhibition of only one ERK paralog did not mimic the effect of MEK inhibition in promoting mouse ESC self-renewal. Regarding ESC differentiation, inhibition of ERK1/2 prevented mesendoderm differentiation. Additionally, selective inhibition of ERK1, but not ERK2, promoted mesendoderm differentiation. Discussion: These findings suggest that ERK1 and ERK2 have both overlapping and distinct roles in regulating ESC self-renewal and differentiation. This study provides new insights into the molecular mechanisms of ERK1/2 in governing ESC maintenance and lineage commitment, potentially informing future strategies for controlling stem cell fate in research and therapeutic applications.
    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
    急性髓系白血病(AML)是一种致命的血液系统疾病,其特征是由造血中的各种突变形成的不同组织中未成熟的髓样母细胞的无控制增殖。尽管有强烈的化疗方案,患者经常经历不良的结果,导致缓解率不达标。近年来,长链非编码RNA(lncRNAs)越来越成为重要的预后和治疗热点,由于它们对许多功能表观遗传失调的贡献,转录,以及导致细胞表达改变的翻译后机制,导致白血病细胞化疗耐药增加和凋亡减少。通过这次审查,我强调并讨论了了解lncRNAs赋予AML治疗抗性的主要机制的最新进展。此外,我还提供了当前靶向lncRNA表达策略的观点。更好地了解lncRNAs在控制AML治疗结果中的关键作用将有助于改善现有药物并设计新的药物。
    Acute Myeloid Leukemia (AML) is a fatal hematological disease characterized by the unchecked proliferation of immature myeloid blasts in different tissues developed by various mutations in hematopoiesis. Despite intense chemotherapeutic regimens, patients often experience poor outcomes, leading to substandard remission rates. In recent years, long non-coding RNAs (lncRNAs) have increasingly become important prognostic and therapeutic hotspots, due to their contributions to dysregulating many functional epigenetic, transcriptional, and post-translational mechanisms leading to alterations in cell expressions, resulting in increased chemoresistance and reduced apoptosis in leukemic cells. Through this review, I highlight and discuss the latest advances in understanding the major mechanisms through which lncRNAs confer therapy resistance in AML. In addition, I also provide perspective on the current strategies to target lncRNA expressions. A better knowledge of the critical role that lncRNAs play in controlling treatment outcomes in AML will help improve existing medications and devise new ones.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号