Intestinal stem cells

肠干细胞
  • 文章类型: Editorial
    肠干细胞(ISC)在炎症状态下取代受损的肠粘膜上皮细胞的增殖和分化是改善肠道炎症的关键步骤。然而,当这种无序的扩散继续时,它诱导ISC进入癌变状态。肠粘膜屏障的自由表面上的肠微生物群能够持续地与ISC相互作用。微生物代谢产物能够通过转录因子调节肠道干细胞和祖细胞的增殖,当处于稳定状态时,分化的结肠细胞能够分解这些代谢物,从而保护肠道隐窝的干细胞。在未来,肠道菌群及其代谢产物介导ISC分化的调节将是一种潜在的肠道疾病治疗方法。
    Proliferation and differentiation of intestinal stem cell (ISC) to replace damaged gut mucosal epithelial cells in inflammatory states is a critical step in ameliorating gut inflammation. However, when this disordered proliferation continues, it induces the ISC to enter a cancerous state. The gut microbiota on the free surface of the gut mucosal barrier is able to interact with ISC on a sustained basis. Microbiota metabolites are able to regulate the proliferation of gut stem and progenitor cells through transcription factors, while in steady state, differentiated colonocytes are able to break down such metabolites, thereby protecting stem cells at the gut crypt. In the future, the gut flora and its metabolites mediating the regulation of ISC differentiation will be a potential treatment for enteropathies.
    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
    肠道充满了复杂而动态的各种微生物,统称为肠道微生物群,与宿主相互作用并有助于其代谢和生理。饮食被认为是肠道微生物群的关键调节剂,当摄入的营养物质与常驻微生物群组成相互作用并形成时。此外,最近的研究强调了饮食和微生物来源的营养素之间的相互作用,直接影响肠道干细胞调节其周转,以确保健康的肠道屏障。尽管先进的测序方法允许人类肠道微生物组的表征,评估饮食-微生物群-宿主相互作用的机制研究取决于遗传易处理模型的使用,如果蝇。在这次审查中,我们首先讨论了人类和苍蝇肠道之间的相似性,然后我们关注饮食和微生物群对控制肠道干细胞自我更新和分化的营养传感信号级联的影响,以及疾病。最后,我们强调使用果蝇模型评估微生物群在肠道相关病理中的作用,以及了解介导肠道功能障碍的不同全身表现的机制.
    The intestine is populated by a complex and dynamic assortment of microbes, collectively called gut microbiota, that interact with the host and contribute to its metabolism and physiology. Diet is considered a key regulator of intestinal microbiota, as ingested nutrients interact with and shape the resident microbiota composition. Furthermore, recent studies underscore the interplay of dietary and microbiota-derived nutrients, which directly impinge on intestinal stem cells regulating their turnover to ensure a healthy gut barrier. Although advanced sequencing methodologies have allowed the characterization of the human gut microbiome, mechanistic studies assessing diet-microbiota-host interactions depend on the use of genetically tractable models, such as Drosophila melanogaster. In this review, we first discuss the similarities between the human and fly intestines and then we focus on the effects of diet and microbiota on nutrient-sensing signaling cascades controlling intestinal stem cell self-renewal and differentiation, as well as disease. Finally, we underline the use of the Drosophila model in assessing the role of microbiota in gut-related pathologies and in understanding the mechanisms that mediate different whole-body manifestations of gut dysfunction.
    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
    目前接受的肠上皮细胞组织模型提出Lgr5+隐窝基柱状(CBC)细胞代表唯一的肠干细胞(ISC)隔室。然而,先前的研究表明,Lgr5+细胞对肠道再生是可有可无的,导致两个主要假设:一个赞成静止储备ISC的存在,另一个呼吁分化的细胞可塑性。为了调查这些可能性,我们通过高分辨率单细胞谱分析以无偏方式研究了隐窝上皮细胞.这些研究,结合体内谱系追踪,表明Lgr5不是特定的ISC标记,并且存在于隐窝碱基之外并位于峡部区域,其中未分化细胞参与照射(IR)损伤后的肠道稳态和再生。我们的结果提供了肠上皮细胞组织的替代模型,表明干细胞潜能不限于CBC细胞,去分化和储备ISC都不是肠再生的驱动因素。
    The currently accepted intestinal epithelial cell organization model proposes that Lgr5+ crypt-base columnar (CBC) cells represent the sole intestinal stem cell (ISC) compartment. However, previous studies have indicated that Lgr5+ cells are dispensable for intestinal regeneration, leading to two major hypotheses: one favoring the presence of a quiescent reserve ISC and the other calling for differentiated cell plasticity. To investigate these possibilities, we studied crypt epithelial cells in an unbiased fashion via high-resolution single-cell profiling. These studies, combined with in vivo lineage tracing, show that Lgr5 is not a specific ISC marker and that stemness potential exists beyond the crypt base and resides in the isthmus region, where undifferentiated cells participate in intestinal homeostasis and regeneration following irradiation (IR) injury. Our results provide an alternative model of intestinal epithelial cell organization, suggesting that stemness potential is not restricted to CBC cells, and neither de-differentiation nor reserve ISC are drivers of intestinal regeneration.
    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
    在流行的模型中,Lgr5细胞是唯一的肠干细胞(ISC),通过难以捉摸的上隐窝转运扩增(TA)中间体通过子代的向上迁移来维持稳态上皮再生。这里,我们在推定的TA细胞的位置确定了一个由Fgfbp1标记的增殖性上隐窝群体,在转录上与Lgr5+细胞不同。使用动力学报告子进行时间分辨的命运映射和Fgfbp1-CreERT2谱系追踪,我们确定Fgfbp1+细胞是多能的,并产生Lgr5+细胞,与它们的ISC功能一致。Fgfbp1+细胞也在Lgr5+细胞耗尽后维持上皮再生。我们证明了由上隐窝细胞产生的FGFBP1,是隐窝增殖和上皮稳态的重要因素。我们的发现支持一种模型,其中组织再生起源于上隐窝Fgfbp1细胞,该细胞产生沿着隐窝-绒毛轴双向传播的后代,并作为隐窝基底中Lgr5细胞的来源。
    In the prevailing model, Lgr5+ cells are the only intestinal stem cells (ISCs) that sustain homeostatic epithelial regeneration by upward migration of progeny through elusive upper crypt transit-amplifying (TA) intermediates. Here, we identify a proliferative upper crypt population marked by Fgfbp1, in the location of putative TA cells, that is transcriptionally distinct from Lgr5+ cells. Using a kinetic reporter for time-resolved fate mapping and Fgfbp1-CreERT2 lineage tracing, we establish that Fgfbp1+ cells are multi-potent and give rise to Lgr5+ cells, consistent with their ISC function. Fgfbp1+ cells also sustain epithelial regeneration following Lgr5+ cell depletion. We demonstrate that FGFBP1, produced by the upper crypt cells, is an essential factor for crypt proliferation and epithelial homeostasis. Our findings support a model in which tissue regeneration originates from upper crypt Fgfbp1+ cells that generate progeny propagating bi-directionally along the crypt-villus axis and serve as a source of Lgr5+ cells in the crypt base.
    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
    结直肠癌是全球关注的健康问题,发病率和死亡率都很高。传统的治疗方法如手术,化疗,和放射治疗在提高患者生存率方面有局限性。最近的研究强调了肠道菌群和肠道干细胞在维持肠道健康中的作用及其在结直肠癌治疗中的潜在治疗应用。肠道微生物群和干细胞之间的相互作用影响上皮自我更新和整体肠道稳态。新的治疗方法,包括免疫疗法,靶向治疗,使用干细胞的再生医学,和肠道微生物群的调节,正在探索改善治疗结果。因此,本章概述了肠道菌群和肠道干细胞在结直肠癌治疗中的潜在治疗应用。
    Colorectal cancer is a global health concern with high incidence and mortality rates. Conventional treatments like surgery, chemotherapy, and radiation therapy have limitations in improving patient survival rates. Recent research highlights the role of gut microbiota and intestinal stem cells in maintaining intestinal health and their potential therapeutic applications in colorectal cancer treatment. The interaction between gut microbiota and stem cells influences epithelial self-renewal and overall intestinal homeostasis. Novel therapeutic approaches, including immunotherapy, targeted therapy, regenerative medicine using stem cells, and modulation of gut microbiota, are being explored to improve treatment outcomes. Accordingly, this chapter provides an overview of the potential therapeutic applications of gut microbiota and intestinal stem cells in treating colorectal cancer.
    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
    炎症性肠病(IBD)的特征是肠道微生物群的菌群失调和肠干细胞(ISC)的功能障碍。然而,尚未描述IBD微生物因子与ISC之间的直接相互作用。这里,我们将α2A-肾上腺素能受体(ADRA2A)鉴定为在ISC中高表达的GPCR。通过PRESTO-Tango筛选,我们证明了酪胺,主要由肠球菌通过酪氨酸脱羧酶(tyrDC)产生,作为ADRA2A的微生物配体。使用工程化的tyrDC缺陷型粪肠球菌菌株和肠上皮细胞特异性Adra2a敲除小鼠,我们显示肠球菌衍生的酪胺抑制ISC增殖,从而通过ADRA2A损害上皮再生并加剧DSS诱导的结肠炎。重要的是,用ADRA2A拮抗剂阻断轴,育亨宾,破坏酪胺介导的对ISC的抑制并缓解结肠炎。我们的发现强调了ISC中的微生物配体-GPCR对,揭示ISC的微生物调节与结肠炎恶化之间的因果关系,并产生靶向治疗方法以恢复结肠炎中的ISC功能。
    Inflammatory bowel disease (IBD) is characterized by dysbiosis of the gut microbiota and dysfunction of intestinal stem cells (ISCs). However, the direct interactions between IBD microbial factors and ISCs are undescribed. Here, we identify α2A-adrenergic receptor (ADRA2A) as a highly expressed GPCR in ISCs. Through PRESTO-Tango screening, we demonstrate that tyramine, primarily produced by Enterococcus via tyrosine decarboxylase (tyrDC), serves as a microbial ligand for ADRA2A. Using an engineered tyrDC-deficient Enterococcus faecalis strain and intestinal epithelial cell-specific Adra2a knockout mice, we show that Enterococcus-derived tyramine suppresses ISC proliferation, thereby impairing epithelial regeneration and exacerbating DSS-induced colitis through ADRA2A. Importantly, blocking the axis with an ADRA2A antagonist, yohimbine, disrupts tyramine-mediated suppression on ISCs and alleviates colitis. Our findings highlight a microbial ligand-GPCR pair in ISCs, revealing a causal link between microbial regulation of ISCs and colitis exacerbation and yielding a targeted therapeutic approach to restore ISC function in colitis.
    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
    肠干细胞(ISC)通过精确调节增殖和分化来控制肠上皮的更新,在肠道生理学中起着关键作用。肠道微生物群通过多种作用与上皮紧密相互作用,包括免疫和代谢相互作用,这转化为微生物活性和ISC功能之间的紧密联系。鉴于肠道微生物群在影响大量营养素和微量营养素代谢方面的不同功能,膳食营养素对宿主-微生物群相互作用产生显著影响,因此,ISC的命运。因此,了解在调节ISC稳态中复杂的宿主-微生物群相互作用对于改善肠道健康至关重要。这里,我们回顾了在理解形成ISC功能的宿主-微生物群免疫和代谢相互作用方面的最新进展,例如模式识别受体和微生物代谢产物的作用,包括乳酸和吲哚代谢产物。此外,微生物群对膳食营养素的不同调节作用,包括蛋白质,碳水化合物,维生素,和矿物质(如铁和锌),对它们对ISC的影响进行了彻底的探讨。因此,我们强调了在ISC稳态中控制宿主-微生物群相互作用的多方面机制。从这篇综述中获得的见解为制定基于饮食或微生物群的干预措施以促进肠道健康提供了策略。
    Intestinal stem cells (ISCs) play a pivotal role in gut physiology by governing intestinal epithelium renewal through the precise regulation of proliferation and differentiation. The gut microbiota interacts closely with the epithelium through myriad of actions, including immune and metabolic interactions, which translate into tight connections between microbial activity and ISC function. Given the diverse functions of the gut microbiota in affecting the metabolism of macronutrients and micronutrients, dietary nutrients exert pronounced effects on host-microbiota interactions and, consequently, the ISC fate. Therefore, understanding the intricate host-microbiota interaction in regulating ISC homeostasis is imperative for improving gut health. Here, we review recent advances in understanding host-microbiota immune and metabolic interactions that shape ISC function, such as the role of pattern-recognition receptors and microbial metabolites, including lactate and indole metabolites. Additionally, the diverse regulatory effects of the microbiota on dietary nutrients, including proteins, carbohydrates, vitamins, and minerals (e.g. iron and zinc), are thoroughly explored in relation to their impact on ISCs. Thus, we highlight the multifaceted mechanisms governing host-microbiota interactions in ISC homeostasis. Insights gained from this review provide strategies for the development of dietary or microbiota-based interventions to foster gut health.
    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
    肠内分泌细胞(EEC)分泌5-羟色胺(肠嗜铬细胞[EC]细胞)或特定的肽激素(非EC细胞),它们具有重要的代谢功能。终端EEC多样性的基础仍然模糊。通过在人肠干细胞的2D培养物中强制转录因子(TF)NEUROG3的活性,我们复制了生理EEC分化,并检查了最终形成离散细胞类型的转录和顺式调节动力学。丰富的EEC前体表达阶段特异性基因和TF。在表达前末端NEUROD1之前,有丝分裂后前体在转录上不同的ASCL1+和HES6hi细胞状态之间振荡。任一因子的缺失都大大加速了EEC的分化并破坏了EEC的个性;ASCL1或NEUROD1缺乏对EC和非EC细胞特征产生相反的后果。这些TF主要结合未分化干细胞中可获得的顺式元件,并且他们定制了作为离散EEC身份基础的TF组合的后续表达。因此,早期的TF振荡会延迟EEC的成熟,从而在医学上重要的细胞系中实现准确的多样性。
    Enteroendocrine cells (EECs) secrete serotonin (enterochromaffin [EC] cells) or specific peptide hormones (non-EC cells) that serve vital metabolic functions. The basis for terminal EEC diversity remains obscure. By forcing activity of the transcription factor (TF) NEUROG3 in 2D cultures of human intestinal stem cells, we replicated physiologic EEC differentiation and examined transcriptional and cis-regulatory dynamics that culminate in discrete cell types. Abundant EEC precursors expressed stage-specific genes and TFs. Before expressing pre-terminal NEUROD1, post-mitotic precursors oscillated between transcriptionally distinct ASCL1+ and HES6hi cell states. Loss of either factor accelerated EEC differentiation substantially and disrupted EEC individuality; ASCL1 or NEUROD1 deficiency had opposing consequences on EC and non-EC cell features. These TFs mainly bind cis-elements that are accessible in undifferentiated stem cells, and they tailor subsequent expression of TF combinations that underlie discrete EEC identities. Thus, early TF oscillations retard EEC maturation to enable accurate diversity within a medically important cell lineage.
    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
    在接受盆腔肿瘤放射治疗的患者中,肠道容易受到辐射损伤。然而,目前尚无可用于预防或治疗放射性肠病(RIE)的有效药物.在这项研究中,我们旨在研究源自肠道微生物群的吲哚-3-甲醛(I3A)对RIE的影响。分离并培养肠道类器官以筛选辐射防护色氨酸代谢物。在雄性C57BL/6J小鼠中使用13Gy全腹照射建立RIE模型。口服I3A后,通过对存活率的观察来评估其辐射防护能力,临床评分,和病理分析。通过免疫荧光和免疫组织化学观察肠干细胞存活和肠屏障的变化。随后,通过16SrRNA和转录组测序研究了I3A的辐射防护机制,分别。最后,培养人结肠癌细胞和类器官以评估I3A对肿瘤放疗的影响。I3A对肠类器官表现出最有效的辐射防护作用。口服I3A治疗显著提高了受照射小鼠的存活率,改善临床和组织学评分,减轻粘膜损伤,增强Lgr5+肠干细胞的增殖和分化,维持肠屏障完整性。此外,I3A增强了益生菌的丰度,并激活AhR/IL-10/Wnt信号通路促进肠上皮增殖。作为一种重要的色氨酸代谢产物,I3A通过AhR/IL-10/Wnt信号通路促进肠上皮细胞增殖,并上调益生菌的丰度以治疗RIE。微生物来源的I3A对RIE的治疗具有潜在的临床应用价值。
    The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5+ intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.
    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:干细胞和微生物稳态的破坏加速了衰老过程。因此,保持这些平衡有效地延缓衰老并减轻与年龄有关的疾病的症状。最近的研究表明,靶向内质网(ER)应激和免疫缺陷(IMD)信号可能在维持衰老的肠干细胞(ISC)和微生物平衡的稳态中起积极作用。先前的研究表明,来自人参C.A.Meyer的总人参皂苷(TG)可能通过减轻ER应激和介导IMD途径而表现出潜在的抗衰老特性。然而,目前尚不清楚TG是否通过调节ER应激和IMD途径促进健康衰老来改善ISC和微生物稳态.
    目的:为了阐明TG是否促进果蝇的健康及其潜在的分子机制,重点关注其在调节ER应激和IMD途径中的作用,以维持ISC和肠道菌群稳态。
    方法:采用高效液相色谱法检测TG中的主要皂苷单体。存活率,肠长,屏障功能,和摄食/排泄行为测定用于评估TG对果蝇寿命和肠道健康的影响。在干细胞水平,“esg-荧光素酶”报告系统,esg-GFP/delta干细胞荧光标记,和磷酸组蛋白H3有丝分裂活性测定用于确定TG是否可以预防果蝇的自然衰老或与氧化应激相关的ISC过度增殖。免疫荧光染色检测TG对衰老过程中内质网应激的影响。利用基因编辑技术操纵ER应激靶基因及其相关c-JunN末端激酶(JNK)基因的过表达或干扰,以验证TG维持与年龄相关的ISC增殖稳态的分子机制。分子对接和等温滴定量热法验证了TG和ER应激靶基因之间的直接相互作用。此外,在肠道菌群水平,16SrDNA测序用于分析TG对果蝇肠道菌群多样性和丰度的影响以及可能涉及的功能途径。进行RT-qPCR以确定TG是否介导IMD途径中靶基因的表达。进行了显性细菌物种特异性单关联分析,以验证TG对IMD靶基因和ISC增殖的影响是否取决于显性细菌物种的直接控制。
    结果:我们的结果表明,服用TG可以延缓衰老果蝇的肠道形态和功能下降。TG通过抑制ER应激IRE1介导的JNK信号传导来防止年龄相关的ISC过度增殖。此外,口服TG通过重塑肠道微生物群和抑制醋杆菌介导的IMD靶基因激活来预防衰老相关的ISC和肠道微生物群菌群失调。
    结论:TG通过抑制ISC的过度增殖和减轻肠道微生物失衡促进健康衰老,从而为抗衰老TG产品的研发提供新的见解。
    BACKGROUND: Disruption of stem cell and microbial homeostasis accelerates the aging process. Hence, maintaining these balances effectively delays aging and alleviates the symptoms of age-related diseases. Recent research indicates that targeting endoplasmic reticulum (ER) stress and immune deficiency (IMD) signalling may play a positive role in maintaining homeostasis in aging intestinal stem cells (ISC) and microbial equilibrium. Previous research has suggested that total ginsenosides (TG) derived from Panax ginseng C. A. Meyer may exhibit potential anti-aging properties by mitigating ER stress and mediating the IMD pathway. Nevertheless, it remains unclear whether TG improve ISC and microbial homeostasis by modulating ER stress and the IMD pathway to promote healthy aging.
    OBJECTIVE: To elucidate whether TG promotes healthspan in Drosophila and its underlying molecular mechanisms, focusing on its role in regulating ER stress and the IMD pathway to maintain ISC and intestinal microbiota homeostasis.
    METHODS: High performance liquid chromatography was performed to detect the main saponin monomer in TG. Survival rate, gut length, barrier function, and feeding/excretion behaviour assays were used to evaluate the effects of TG on the lifespan and gut health of Drosophila. At the stem cell level, \"esg-luciferase\" reporter system, esg-GFP/delta stem cell fluorescent labelling, and phospho-histone H3+ mitotic activity assays were employed to determine whether TG prevented natural aging or oxidative stress-associated ISC over-proliferation in Drosophila. Immunofluorescence staining was used to detect the effects of TG on ER stress during aging. Overexpression or interference of ER stress target genes and their related c-Jun N-terminal kinase (JNK) gene was manipulated using gene editing technology to verify the molecular mechanism by which TG maintains age-related ISC proliferation homeostasis. Molecular docking and isothermal titration calorimetry were used to verify the direct interactions between TG and ER stress target genes. In addition, at the intestinal flora level, 16S rDNA sequencing was used to analyse the effect of TG on the diversity and abundance of Drosophila intestinal flora and the possible functional pathways involved. RT-qPCR was performed to determine whether TG mediated the expression of target genes in the IMD pathway. A dominant bacterial species-specific mono-association analysis were performed to verify whether the effects of TG on IMD target genes and ISC proliferation depended on the direct control of the dominant bacterial species.
    RESULTS: Our results suggest that administration of TG delays the decline in gut morphology and function in aging Drosophila. TG prevents age-associated ISC hyperproliferation by inhibiting ER stress IRE1-mediated JNK signaling. Furthermore, oral TG prevented aging-associated ISC and gut microbiota dysbiosis by remodelling the gut microbiota and inhibiting Acetobacter-mediated activation of IMD target genes.
    CONCLUSIONS: TG promotes healthy aging by inhibiting the excessive proliferation of ISC and alleviating intestinal microbial imbalance, thereby providing new insights for the research and development of anti-aging TG products.
    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