HSCs

HSC
  • 文章类型: Journal Article
    转录因子GATA2在造血中具有关键作用。杂合种系GATA2突变导致以免疫缺陷为特征的综合征,骨髓衰竭和易患骨髓增生异常综合征(MDS)和急性髓系白血病。这些患者的临床症状多种多样,驱动GATA2相关表型的机制在很大程度上是未知的。探讨GATA2单倍体功能不全对造血功能的影响,我们建立了一个斑马鱼模型,带有gata2b杂合突变(gata2b+/-),GATA2的直系同源物。形态学分析显示gata2b+/-肾骨髓中的骨髓和红系发育不良。因为Gata2b在谱系分化过程中会影响转录和染色质可及性,这通过单细胞(sc)RNA-seq和单核(sn)ATAC-seq进行评估。Sn-ATAC-seq表明,与野生型相比,gata2b+/-斑马鱼HSPC中转录起始位点(TSS)和-3.5-4.1kb推定增强子之间的共同可及性更强,增加gata2b表达并导致在HSPC中更高的全基因组Gata2b基序使用。由于gata2b基因座的可及性增加,gata2b+/-染色质在谱系分化过程中也更容易获得。scRNA-seq数据显示髓样分化缺陷,也就是说,细胞周期进程受损,cebpa和cebpb的表达降低,核糖体生物发生的特征增加。这些数据还揭示了红系祖细胞的分化延迟,Gata1a的异常增殖特征和下调,红细胞生成的主要调节因子,随着年龄的增长而恶化。这些发现表明,细胞内在的补偿机制,需要在杂合HSPCs中获得正常水平的Gata2b以维持其完整性,导致谱系分化异常,从而代表了对MDS倾向的关键步骤。
    The transcription factor GATA2 has a pivotal role in haematopoiesis. Heterozygous germline GATA2 mutations result in a syndrome characterized by immunodeficiency, bone marrow failure and predispositions to myelodysplastic syndrome (MDS) and acute myeloid leukaemia. Clinical symptoms in these patients are diverse and mechanisms driving GATA2-related phenotypes are largely unknown. To explore the impact of GATA2 haploinsufficiency on haematopoiesis, we generated a zebrafish model carrying a heterozygous mutation of gata2b (gata2b+/-), an orthologue of GATA2. Morphological analysis revealed myeloid and erythroid dysplasia in gata2b+/- kidney marrow. Because Gata2b could affect both transcription and chromatin accessibility during lineage differentiation, this was assessed by single-cell (sc) RNA-seq and single-nucleus (sn) ATAC-seq. Sn-ATAC-seq showed that the co-accessibility between the transcription start site (TSS) and a -3.5-4.1 kb putative enhancer was more robust in gata2b+/- zebrafish HSPCs compared to wild type, increasing gata2b expression and resulting in higher genome-wide Gata2b motif use in HSPCs. As a result of increased accessibility of the gata2b locus, gata2b+/- chromatin was also more accessible during lineage differentiation. scRNA-seq data revealed myeloid differentiation defects, that is, impaired cell cycle progression, reduced expression of cebpa and cebpb and increased signatures of ribosome biogenesis. These data also revealed a differentiation delay in erythroid progenitors, aberrant proliferative signatures and down-regulation of Gata1a, a master regulator of erythropoiesis, which worsened with age. These findings suggest that cell-intrinsic compensatory mechanisms, needed to obtain normal levels of Gata2b in heterozygous HSPCs to maintain their integrity, result in aberrant lineage differentiation, thereby representing a critical step in the predisposition to MDS.
    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:肝癌以复杂的炎性肿瘤微环境为代表,其中一系列细胞因子和基质细胞协调了一个显著影响肿瘤发生的环境。白细胞介素-17A(IL-17A),一种主要由Th17细胞分泌的关键促炎细胞因子,已知在肝癌的病因和进展中起重要作用。然而,IL-17A与肝星状细胞(HSC)结合促进肝细胞癌(HCC)发展的确切机制仍有待完全阐明。这项研究旨在揭示IL-17A和HSC在HCC的背景下的相互作用。
    方法:使用二乙基亚硝胺在雄性Sprague-Dawley大鼠中建立HCC模型,以探讨IL-17A和HSCs在HCC发病中的作用。使用腺相关病毒实现了Il17a的体内过表达。一套分子技术,包括RT-qPCR,酶联免疫吸附测定,西方印迹,细胞计数试剂盒-8测定和集落形成测定,用于体外分析。
    结果:研究结果表明,IL-17A是肝癌促进的关键介质,主要通过激活肝祖细胞(HPCs)。这种促肿瘤发生的影响似乎是由HSC介导的,而不是通过对HPCs的直接影响。值得注意的是,IL-17A诱导的成纤维细胞激活蛋白(FAP)在HSC中的表达成为HCC进展的关键因素。在IL-17A刺激的HSC中沉默Fap被观察到逆转HSC的HCC促进作用。
    结论:来自本研究的集体证据提示HSC内的IL-17A/FAP信号轴通过增强HPC激活而促进HCC发展。这些发现支持IL-17A作为HCC的诊断和预防靶标的潜力。为治疗干预提供新的途径。
    BACKGROUND: Liver cancer is typified by a complex inflammatory tumor microenvironment, where an array of cytokines and stromal cells orchestrate a milieu that significantly influences tumorigenesis. Interleukin-17A (IL-17A), a pivotal pro-inflammatory cytokine predominantly secreted by Th17 cells, is known to play a substantial role in the etiology and progression of liver cancer. However, the precise mechanism by which IL-17A engages with hepatic stellate cells (HSCs) to facilitate the development of hepatocellular carcinoma (HCC) remains to be fully elucidated. This investigation seeks to unravel the interplay between IL-17A and HSCs in the context of HCC.
    METHODS: An HCC model was established in male Sprague-Dawley rats using diethylnitrosamine to explore the roles of IL-17A and HSCs in HCC pathogenesis. In vivo overexpression of Il17a was achieved using adeno-associated virus. A suite of molecular techniques, including RT-qPCR, enzyme-linked immunosorbent assays, Western blotting, cell counting kit-8 assays and colony formation assays, was employed for in vitro analyses.
    RESULTS: The study findings indicate that IL-17A is a key mediator in HCC promotion, primarily through the activation of hepatic progenitor cells (HPCs). This pro-tumorigenic influence appears to be mediated by HSCs, rather than through a direct effect on HPCs. Notably, IL-17A-induced expression of fibroblast activation protein (FAP) in HSCs emerged as a critical factor in HCC progression. Silencing Fap in IL-17A-stimulated HSCs was observed to reverse the HCC-promoting effects of HSCs.
    CONCLUSIONS: The collective evidence from this study implicates the IL-17A/FAP signaling axis within HSCs as a contributor to HCC development by enhancing HPC activation. These findings bolster the potential of IL-17A as a diagnostic and preventative target for HCC, offering new avenues for therapeutic intervention.
    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
    肝星状细胞(HSC)激活和肝纤维化介导的胆道闭锁(BA)发展,但是对潜在的分子机制知之甚少。本研究旨在探讨circRNAhsa_circ_0009096在HSC增殖和肝纤维化中的作用。
    用转化生长因子β(TGF-β1)处理LX-2细胞建立肝纤维化模型。进行RNaseR和放线菌素D测定以检测hsa_circ_0009096稳定性。hsa_circ_0009096,miR-370-3p,并使用逆转录-qPCR检测目标基因。使用双荧光素酶报告基因测定验证hsa_circ_0009096与miR-370-3p的直接结合。使用流式细胞术评估LX-2细胞的细胞周期进程和凋亡。α-平滑肌肌动蛋白(α-SMA),胶原蛋白1A1(COL1A1),使用免疫细胞化学和蛋白质印迹分析LX-2细胞中的TGFβ受体2(TGFBR2)蛋白水平。
    Hsa_circ_0009096对RNaseR和放线菌D消化的抗性高于UTRNmRNA。在TGF-β1处理的LX-2细胞中Hsa_circ_0009096表达显著增加,伴随着α-SMA和COL1A1表达升高。Hsa_circ_0009096siRNA在LX-2细胞中有效促进miR-370-3p并抑制TGFBR2表达,通过hsa_circ_0009096与miR-370-3p的直接缔合介导。Hsa_circ_0009096siRNA干扰细胞周期进程,促进细胞凋亡,TGF-β1处理的LX-2细胞中α-SMA和COL1A1的表达降低。MiR-370-3p抑制剂减轻了细胞周期进程的改变,凋亡,和α-SMA,由hsa_circ_0009096siRNA引起的LX-2细胞中的COL1A1和TGFBR2表达。总之,hsa_circ_0009096通过spongingmiR-370-3p加速TGFBR2表达促进BA发病过程中HSC增殖和肝纤维化。
    UNASSIGNED: Hepatic stellate cell (HSC) activation and hepatic fibrosis mediated biliary atresia (BA) development, but the underlying molecular mechanisms are poorly understood. This study aimed to investigate the roles of circRNA hsa_circ_0009096 in the regulation of HSC proliferation and hepatic fibrosis.
    UNASSIGNED: A cellular hepatic fibrosis model was established by treating LX-2 cells with transforming growth factor β (TGF-β1). RNaseR and actinomycin D assays were performed to detect hsa_circ_0009096 stability. Expression of hsa_circ_0009096, miR-370-3p, and target genes was detected using reverse transcription-qPCR. Direct binding of hsa_circ_0009096 to miR-370-3p was validated using dual luciferase reporter assay. Cell cycle progression and apoptosis of LX-2 cells were assessed using flow cytometry. The alpha-smooth muscle actin (α-SMA), collagen 1A1 (COL1A1), and TGF beta receptor 2 (TGFBR2) protein levels in LX-2 cells were analyzed using immunocytochemistry and western blotting.
    UNASSIGNED: Hsa_circ_0009096 exhibited more resistance to RNase R and actinomycinD digestion than UTRN mRNA. Hsa_circ_0009096 expression increased significantly in LX-2 cells treated with TGF-β1, accompanied by elevated α-SMA and COL1A1 expression. Hsa_circ_0009096 siRNAs effectively promoted miR-370-3p and suppressed TGFBR2 expression in LX-2 cells, mediated by direct association of hsa_circ_0009096 with miR-370-3p. Hsa_circ_0009096 siRNA interfered with the cell cycle progression, promoted apoptosis, and reduced α-SMA and COL1A1 expression in LX-2 cells treated with TGF-β1. MiR-370-3p inhibitors mitigated the alterations in cell cycle progression, apoptosis, and α-SMA, COL1A1, and TGFBR2 expression in LX-2 cells caused by hsa_circ_0009096 siRNA. In conclusion, hsa_circ_0009096 promoted HSC proliferation and hepatic fibrosis during BA pathogenesis by accelerating TGFBR2 expression by sponging miR-370-3p.
    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:环状RNA(circularRNA,circRNA)是一种新型的功能性非编码RNA(ncRNA),在多种人类肝病的发生和发展中起作用,包括肝纤维化(LF)。LF是肝损伤后的可逆修复反应,肝星状细胞(HSC)的激活是核心事件。然而,circRNAs诱导LF中HSC活化的调控机制仍知之甚少。在这项研究中,研究了介导炎症反应并导致HSC活化的circAno6/miR-296-3p/toll样受体4(TLR4)信号轴。
    方法:首先,将circAno6过表达质粒和小干扰RNA转染到细胞中以确定circAno6是否可以影响HSC的功能。第二,实时定量聚合酶链反应(RT-qPCR),酶联免疫吸附测定(ELISA),免疫印迹(WB)和免疫荧光(IF)检测circAno6质粒/siRNA转染对HSC活化指标的影响,炎症标志物和circAno6/miR-296-3p/TLR4信号轴。然后通过核质分离和荧光原位杂交(FISH)检查circAno6的亚细胞位置。最后,荧光素酶报告基因检测用于鉴定circAno6和miR-296-3p之间的关系,以及miR-296-3p和TLR4之间的关系.
    结果:CircAno6在HSC中显著上调,并与细胞增殖和α-平滑肌肌动蛋白(α-SMA)呈正相关,胶原蛋白I,NOD-类受体热蛋白吲哚相关蛋白3(NLRP3),白细胞介素-1β(IL-1β)和白细胞介素-18(IL-18)的表达。circAno6的过表达增加了炎症反应并诱导了HSC活化,而干扰导致相反的效果。FISH实验揭示了circAno6在细胞质中的定位。然后,双荧光素酶报告基因试验证实miR-296-3p显著抑制了circAno6-WT和TLR4-WT组的荧光素酶活性.
    结论:这项研究表明circAno6和miR-296-3p/TLR4可能形成调节轴并调节炎症反应,这又诱导HSC激活。靶向cirAno6可能是治疗LF的潜在治疗策略。
    BACKGROUND: Circular RNA (circRNA) is a novel functional non-coding RNA(ncRNA) that plays a role in the occurrence and development of multiple human liver diseases, including liver fibrosis (LF). LF is a reversible repair response after liver injury, and the activation of hepatic stellate cells (HSCs) is the core event. However, the regulatory mechanisms by which circRNAs induce the activation of HSCs in LF are still poorly understood. The circAno6/miR-296-3p/toll-like receptor 4 (TLR4) signaling axis that mediates the inflammatory response and causes the activation of HSCs was investigated in this study.
    METHODS: First, a circAno6 overexpression plasmid and small interfering RNA were transfected into cells to determine whether circAno6 can affect the function of HSCs. Second, real-time quantitative polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA), western blotting (WB) and immunofluorescence (IF) were used to detect the effects of circAno6 plasmid/siRNA transfection on HSC activation indices, inflammatory markers and the circAno6/miR-296-3p/TLR4 signaling axis. The subcellular position of circAno6 was then examined by nucleo-cytoplasmic separation and fluorescence in situ hybridization (FISH). Finally, a luciferase reporter gene assay was used to identify the relationship between circAno6 and miR-296-3p as well as the relationship between miR-296-3p and TLR4.
    RESULTS: CircAno6 was considerably upregulated in HSCs and positively correlated with cell proliferation and alpha-smooth muscle actin (α-SMA), collagen I, NOD-likereceptorthermalproteindomainassociatedprotein 3 (NLRP3), interleukin-1β (IL-1β) and interleukin-18 (IL-18) expression. Overexpression of circAno6 increased the inflammatory response and induced HSC activation, whereas interference resulted in the opposite effects. FISH experiments revealed the localization of circAno6 in the cytoplasm. Then, a double luciferase reporter assay confirmed that miR-296-3p significantly inhibited luciferase activity in the circAno6-WT and TLR4-WT groups.
    CONCLUSIONS: This study suggests that circAno6 and miR-296-3p/TLR4 may form a regulatory axis and regulate the inflammatory response, which in turn induces HSC activation. Targeting circAno6 may be a potential therapeutic strategy to treat LF.
    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
    抑制活化的肝星状细胞(HSC)是治疗肝纤维化的一个有前途的方法,铁性凋亡已经成为实现这种抑制的关键机制。柚皮素的作用,一种具有抗炎特性的类黄酮,没有彻底检查肝纤维化。因此,采用胆汁淤积模型研究柚皮素对肝纤维化的影响。我们的研究结果表明,肝组织损伤和纤维化的小鼠进行胆管结扎(BDL)的显着恶化,伴随着纤维发生相关基因表达的大幅上调。值得注意的是,柚皮素给药可显着减轻这些小鼠的肝损伤和纤维化。此外,柚皮素对HSCs的激活具有抑制作用,同时诱导铁性凋亡。重要的是,柚皮素显著增加HSC的自噬活性。这种作用被自噬抑制剂3-MA的共同给药抵消,导致柚皮素诱导的HSC铁凋亡显着降低。在BDL模型小鼠中,柚皮素对肝纤维化有缓解作用,提示与柚皮素诱导的HSC铁性凋亡潜在相关。这些结果为柚皮素诱导的铁凋亡的分子机制提供了新的见解,并强调自噬依赖性铁凋亡作为肝纤维化的有希望的治疗策略。
    Inhibition of activated hepatic stellate cells (HSCs) is a promising approach for treating liver fibrosis, and the ferroptosis has emerged as a pivotal mechanism to achieve this inhibition. The effects of naringenin, a flavonoid with anti-inflammatory properties, have not been thoroughly examined in liver fibrosis. Therefore, we used cholestasis model to study the effect of naringenin on liver fibrosis. Our findings demonstrated a significant exacerbation of liver tissue damage and fibrosis in mice subjected to bile duct ligation (BDL), accompanied by a substantial upregulation of fibrogenesis-related gene expression. Notably, naringenin administration markedly alleviated liver injury and fibrosis in these mice. Furthermore, naringenin exhibited inhibitory effects on the activation of HSCs, concurrently inducing ferroptosis. Importantly, naringenin significantly increased autophagic activity in HSCs. This effect was counteracted by co-administration of the autophagy inhibitor 3-MA, leading to a notable reduction in naringenin-induced HSC ferroptosis. In BDL model mice, naringenin demonstrated a mitigating effect on liver fibrosis, suggesting a potential correlation with naringenin-induced ferroptosis of HSCs. These results provide novel insights into the molecular mechanisms of naringenin-induced ferroptosis and highlight autophagy-dependent ferroptosis as a promising therapeutic strategy for liver fibrosis.
    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/祖细胞(HSPC)的碱基编辑是治疗免疫血液病的有吸引力的策略。然而,使用腺嘌呤碱基编辑的HSPC治疗X连锁严重联合免疫缺陷(SCID-X1)的可行性,剂量反应关系对免疫细胞生成的影响,潜在风险尚未在体内得到证实。这里,建立了人源化SCID-X1小鼠模型,86.67%±2.52%(n=3)的小鼠造血干细胞(HSC)致病突变得到纠正,没有检测到单向导RNA(sgRNA)依赖性脱靶效应。对具有不同免疫缺陷背景的小鼠移植后16周的外周血的分析揭示了在移植不同量的修饰的HSC后有效的免疫细胞生成。因此,在安全范围内大规模输注基因校正的HSC可以实现快速,稳定,和持久的免疫细胞再生。组织切片染色进一步证明了免疫器官组织结构的恢复,在多个器官中没有肿瘤形成。总的来说,这些数据表明,碱基编辑的HSC是SCID-X1的潜在治疗方法,免疫细胞再生需要阈值输注剂量的基因校正细胞.本研究为碱基编辑的HSCs治疗SCID-X1的临床应用奠定了理论基础。
    Base editing of hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematologic diseases. However, the feasibility of using adenine-base-edited HSPCs for treating X-linked severe combined immunodeficiency (SCID-X1), the influence of dose-response relationships on immune cell generation, and the potential risks have not been demonstrated in vivo. Here, a humanized SCID-X1 mouse model was established, and 86.67% ± 2.52% (n = 3) of mouse hematopoietic stem cell (HSC) pathogenic mutations were corrected, with no single-guide-RNA (sgRNA)-dependent off-target effects detected. Analysis of peripheral blood over 16 weeks post-transplantation in mice with different immunodeficiency backgrounds revealed efficient immune cell generation following transplantation of different amounts of modified HSCs. Therefore, a large-scale infusion of gene-corrected HSCs within a safe range can achieve rapid, stable, and durable immune cell regeneration. Tissue-section staining further demonstrated the restoration of immune organ tissue structures, with no tumor formation in multiple organs. Collectively, these data suggest that base-edited HSCs are a potential therapeutic approach for SCID-X1 and that a threshold infusion dose of gene-corrected cells is required for immune cell regeneration. This study lays a theoretical foundation for the clinical application of base-edited HSCs in treating SCID-X1.
    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:芍药苷对肝纤维化的缓解作用已经确立;然而,这种效应的分子机制和特定靶标仍然难以捉摸。
    目的:本研究旨在探讨Pae对肝纤维化中肝星状细胞(HSCs)活化的调控作用的分子机制。特别关注Pae在调节组蛋白甲基化修饰中的作用。
    方法:通过建立四氯化碳(CCl4)诱导的小鼠体内和体外模型和转化生长因子β1(TGF-β1)诱导的LX-2细胞,评价Pae的治疗效果。分别。分子对接,表面等离子体共振(SPR),采用染色质免疫沉淀-实时定量PCR(ChIP-qPCR)等分子生物学方法阐明Pae调控HSCs活化的分子机制。
    结果:我们的研究发现Pae抑制CCl4诱导的小鼠和LX-2细胞肝脏中的HSCs活化和组蛋白三甲基化修饰。我们证明了Pae对HSC活化的抑制作用取决于过氧化物酶体增殖物激活受体γ(PPARγ)的表达和zeste同源物2(EZH2)的增强子。机械上,Pae直接与EZH2结合以有效抑制其酶活性。这种衰减导致PPARγ启动子区组蛋白H3K27三甲基化的抑制,诱导PPARγ表达上调。
    结论:这项研究不仅揭示了Pae诱导的肝纤维化缓解的精确目标,而且强调了EZH2介导的H3K27三甲基化在驱动肝纤维化发病机制中的重要意义。
    BACKGROUND: The alleviating effect of paeoniflorin (Pae) on liver fibrosis has been established; however, the molecular mechanism and specific target(s) underlying this effect remain elusive.
    OBJECTIVE: This study was to investigate the molecular mechanism underlying the regulatory effect of Pae on hepatic stellate cells (HSCs) activation in liver fibrosis, with a specific focus on the role of Pae in modulating histone methylation modifications.
    METHODS: The therapeutic effect of Pae was evaluated by establishing in vivo and in vitro models of carbon tetrachloride (CCl4)-induced mice and transforming growth factor β1 (TGF-β1)-induced LX-2 cells, respectively. Molecular docking, surface plasmon resonance (SPR), chromatin immunoprecipitation-quantitative real time PCR (ChIP-qPCR) and other molecular biological methods were used to clarify the molecular mechanism of Pae regulating HSCs activation.
    RESULTS: Our study found that Pae inhibited HSCs activation and histone trimethylation modification in liver of CCl4-induced mice and LX-2 cells. We demonstrated that the inhibitory effect of Pae on the activation of HSCs was dependent on peroxisome proliferator-activated receptor γ (PPARγ) expression and enhancer of zeste homolog 2 (EZH2). Mechanistically, Pae directly binded to EZH2 to effectively suppress its enzymatic activity. This attenuation leaded to the suppression of histone H3K27 trimethylation in the PPARγ promoter region, which induced upregulation of PPARγ expression.
    CONCLUSIONS: This investigative not only sheds new light on the precise targets that underlie the remission of hepatic fibrogenesis induced by Pae but also emphasizes the critical significance of EZH2-mediated H3K27 trimethylation in driving the pathogenesis of liver fibrosis.
    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:间充质干细胞/基质细胞(MSC)由于其在维持造血干/祖细胞(HSPC)功能中的作用而具有重要的治疗价值。源自人多能干细胞的MSC由于其无限的供应而代表了理想的替代方案。然而,来自HPSC的具有神经c来源的MSCs对HSPCs维持的作用尚未见报道。
    方法:流式细胞术分析,应用RNA测序和分化能力检测来自3D人脑类器官的基质细胞的特征。人脐带血CD34+(UCB-CD34+)细胞在不同的共培养条件下培养,所述共培养条件由基质细胞和脐带MSC(UC-MSC)组成,有或没有细胞因子混合物。通过LTC-IC测定法在体外测试基质细胞的造血基质能力,并通过将脐带血有核细胞和基质细胞共移植到免疫缺陷小鼠体内进行体内测试。RNA和蛋白质组测序用于检测MSCs对HSPCs的作用。
    结果:基质细胞,来自H1-hESCs和人类诱导多能干细胞前脑器官,能够分化成经典的间充质衍生细胞(成骨细胞,软骨细胞,和脂肪细胞)。这些细胞表达MSC标记,因此被称为多能干细胞来源的MSCs(pMSCs)。pMSCs早期表现为神经cast起源,CD271表达。当人UCB-CD34+HSPCs在UC-MSCs或pMSCs上共培养时,后者导致UCB-CD34+HSPCs在长期培养和有效维持其可移植性方面的强劲扩增。通过RNA测序的比较表明,人UCB-CD34+HSPC与pMSC的共培养为HSC维持提供了改善的微环境。pMSC高度表达Wnt信号传导抑制剂SFRP1和SFRP2,表明它们可能有助于通过拮抗Wnt激活来调节细胞周期以促进UCB-CD34HSPC的维持。
    结论:报道了一种在无血清培养条件下从3D人脑类器官中收获具有神经c来源的MSCs的新方法。我们证明了pMSC在长期培养中支持人UCB-HSPC体外扩增并维持其可移植能力。RNA和蛋白质组测序表明,pMSC通过涉及细胞-细胞接触和分泌因子以及抑制Wnt信号传导的机制为HSC维持提供了改善的微环境。这代表了大规模生产神经c来源的MSC的新方法,并为开发用于治疗造血不良的人类造血基质细胞疗法提供了潜在的方法。
    BACKGROUND: Mesenchymal stem/stromal cells (MSCs) are of great therapeutic value due to their role in maintaining the function of hematopoietic stem/progenitor cells (HSPCs). MSCs derived from human pluripotent stem cells represent an ideal alternative because of their unlimited supply. However, the role of MSCs with neural crest origin derived from HPSCs on the maintenance of HSPCs has not been reported.
    METHODS: Flow cytometric analysis, RNA sequencing and differentiation ability were applied to detect the characteristics of stromal cells from 3D human brain organoids. Human umbilical cord blood CD34+ (UCB-CD34+) cells were cultured in different coculture conditions composed of stromal cells and umbilical cord MSCs (UC-MSCs) with or without a cytokine cocktail. The hematopoietic stroma capacity of stromal cells was tested in vitro with the LTC-IC assay and in vivo by cotransplantation of cord blood nucleated cells and stroma cells into immunodeficient mice. RNA and proteomic sequencing were used to detect the role of MSCs on HSPCs.
    RESULTS: The stromal cells, derived from both H1-hESCs and human induced pluripotent stem cells forebrain organoids, were capable of differentiating into the classical mesenchymal-derived cells (osteoblasts, chondrocytes, and adipocytes). These cells expressed MSC markers, thus named pluripotent stem cell-derived MSCs (pMSCs). The pMSCs showed neural crest origin with CD271 expression in the early stage. When human UCB-CD34+ HSPCs were cocultured on UC-MSCs or pMSCs, the latter resulted in robust expansion of UCB-CD34+ HSPCs in long-term culture and efficient maintenance of their transplantability. Comparison by RNA sequencing indicated that coculture of human UCB-CD34+ HSPCs with pMSCs provided an improved microenvironment for HSC maintenance. The pMSCs highly expressed the Wnt signaling inhibitors SFRP1 and SFRP2, indicating that they may help to modulate the cell cycle to promote the maintenance of UCB-CD34+ HSPCs by antagonizing Wnt activation.
    CONCLUSIONS: A novel method for harvesting MSCs with neural crest origin from 3D human brain organoids under serum-free culture conditions was reported. We demonstrate that the pMSCs support human UCB-HSPC expansion in vitro in a long-term culture and the maintenance of their transplantable ability. RNA and proteomic sequencing indicated that pMSCs provided an improved microenvironment for HSC maintenance via mechanisms involving cell-cell contact and secreted factors and suppression of Wnt signaling. This represents a novel method for large-scale production of MSCs of neural crest origin and provides a potential approach for development of human hematopoietic stromal cell therapy for treatment of dyshematopoiesis.
    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
    胞质磺基转移酶(SULTs)是II期缀合酶,它们在肝脏中广泛表达,主要介导许多外源性物质和内源性化合物的硫酸化。然而,各种SULTs基因在肝细胞癌(HCC)中的作用尚未报道。本研究旨在分析SULTs基因在HCC中的表达和潜在的功能作用,并确定SULT2A1在HCC干性中的作用以及可能的机制。我们发现所有12个SULTs基因在HCC中差异表达。此外,还研究了临床病理特征和生存率。多因素回归分析显示SULT2A1和SULT1C2可作为HCC的独立预后因素。SULT1C4、SULT1E1和SULT2A1与免疫浸润显著相干。肝癌中SULT2A1缺乏促进化疗耐药和干性维持。机械上,SULT2A1的沉默激活了AKT信号通路,一方面,促进下游干性基因c-Myc的表达,另一方面,促进NRF2表达以减少ROS的积累,并共同增加HCC的干性。此外,敲低NR1I3参与了精干维持中SULT2A1的转录调控。此外,SULT2A1敲低肝癌细胞促进肝星状细胞(HSC)的增殖和活化,从而发挥潜在的基质重塑作用。我们的研究揭示了SULTs基因在HCC中的表达和作用,并确定了SULT2A1对HCC的发生和发展的贡献。
    The cytosolic sulfotransferases (SULTs) are phase II conjugating enzymes, which are widely expressed in the liver and mainly mediate the sulfation of numerous xenobiotics and endogenous compounds. However, the role of various SULTs genes has not been reported in hepatocellular carcinoma (HCC). This study aims to analyze the expression and potential functional roles of SULTs genes in HCC and to identify the role of SULT2A1 in HCC stemness as well as the possible mechanism. We found that all of the 12 SULTs genes were differentially expressed in HCC. Moreover, clinicopathological features and survival rates were also investigated. Multivariate regression analysis showed that SULT2A1 and SULT1C2 could be used as independent prognostic factors in HCC. SULT1C4, SULT1E1, and SULT2A1 were significantly associated with immune infiltration. SULT2A1 deficiency in HCC promoted chemotherapy resistance and stemness maintenance. Mechanistically, silencing of SULT2A1 activated the AKT signaling pathway, on the one hand, promoted the expression of downstream stemness gene c-Myc, on the other hand, facilitated the NRF2 expression to reduce the accumulation of ROS, and jointly increased HCC stemness. Moreover, knockdown NR1I3 was involved in the transcriptional regulation of SULT2A1 in stemness maintenance. In addition, SULT2A1 knockdown HCC cells promoted the proliferation and activation of hepatic stellate cells (HSCs), thereby exerting a potential stroma remodeling effect. Our study revealed the expression and role of SULTs genes in HCC and identified the contribution of SULT2A1 to the initiation and progression of HCC.
    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:同种异体肝细胞移植是治疗急性肝缺损的一种新兴方法。然而,移植细胞的持久移植仍然是一项艰巨的任务,因为它们被接受者的免疫系统积极清除。因此,详细了解先天或适应性免疫细胞对同种异体移植肝细胞的反应是合理化基于细胞的治疗的关键.
    方法:这里,我们通过应用冷冻损伤(CI)在肝脏表面诱导了急性炎症再生小生境(3-96h),以系统地评估在持续微炎症环境中针对移植的同种异体肝祖细胞的先天免疫反应。
    结果:所得到的数据突出表明,受损部位显著地重新填充了数量交替的先天免疫细胞,包括中性粒细胞,单核细胞和枯否细胞(KCs),从3到96小时。移植的allo-HP,受伤后6小时移植,在移植后24小时内被先天免疫反应共同消除。KC的选择性消耗表明从第2天到第6天单核细胞的延迟募集。此外,移植后54小时肝祖细胞的脾内移植被KC拆除,而在缺乏KCs的样品中可以观察到移植细胞的时间依赖性更好的存活和易位到损伤部位。
    结论:总体而言,这项研究提供了证据,表明KCs消融能够更好地存活和整合allo-HP在持续的肝脏炎症环境中,对合理化基于细胞的治疗干预对肝脏缺陷的影响。
    BACKGROUND: Allogeneic hepatocyte transplantation is an emerging approach to treat acute liver defects. However, durable engraftment of the transplanted cells remains a daunting task, as they are actively cleared by the recipient\'s immune system. Therefore, a detailed understanding of the innate or adaptive immune cells-derived responses against allogeneic transplanted hepatic cells is the key to rationalize cell-based therapies.
    METHODS: Here, we induced an acute inflammatory regenerative niche (3-96 h) on the surface of the liver by the application of cryo-injury (CI) to systematically evaluate the innate immune response against transplanted allogeneic hepatic progenitors in a sustained micro-inflammatory environment.
    RESULTS: The resulting data highlighted that the injured site was significantly repopulated by alternating numbers of innate immune cells, including neutrophils, monocytes and Kupffer cells (KCs), from 3 to 96 h. The transplanted allo-HPs, engrafted 6 h post-injury, were collectively eliminated by the innate immune response within 24 h of transplantation. Selective depletion of the KCs demonstrated a delayed recruitment of monocytes from day 2 to day 6. In addition, the intrasplenic engraftment of the hepatic progenitors 54 h post-transplantation was dismantled by KCs, while a time-dependent better survival and translocation of the transplanted cells into the injured site could be observed in samples devoid of KCs.
    CONCLUSIONS: Overall, this study provides evidence that KCs ablation enables a better survival and integration of allo-HPs in a sustained liver inflammatory environment, having implications for rationalizing the cell-based therapeutic interventions against liver defects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号