intestinal stem cells

肠干细胞
  • 文章类型: Journal Article
    肠衰竭相关性肝病(IFALD)是短肠综合征(SBS)患者长期肠外营养的严重并发症,是SBS患者死亡的主要原因。预防IFALD是SBS治疗的主要挑战之一。肠屏障功能受损是引发IFALD的关键因素,因此促进肠道修复尤为重要。肠道修复主要依赖于肠干细胞(ISC)的功能,这需要强大的线粒体脂肪酸氧化(FAO)进行自我更新。在这里,我们报告说,IFALD中LGR5+ISC功能异常可能归因于法尼醇X受体(FXR)信号传导受损,由类固醇和胆汁酸激活的转录因子。在手术活检和患者来源的类器官(PDO)中,具有IFALD的SBS患者代表较低的LGR5+细胞群体和降低的FXR表达。此外,用T-βMCA治疗PDO(FXR的拮抗剂)剂量依赖性地减少LGR5细胞的数量和肠细胞的增殖率,伴随着粮农组织涉及的关键基因减少,包括CPT1a。有趣的是,然而,在PDOs(FXR的激动剂)中使用Tropifexor治疗只会提高粮农组织的能力,在ISC功能和肠细胞增殖方面没有改善。总之,这些发现表明,FXR受损可能会通过破坏粮农组织进程加速LGR5+ISC人口的枯竭,这可能成为SBS患者针对IFALD的预防性干预的新的潜在目标。
    Intestinal failure-associated liver disease (IFALD) is a serious complication of long-term parenteral nutrition in patients with short bowel syndrome (SBS), and is the main cause of death in SBS patients. Prevention of IFALD is one of the major challenges in the treatment of SBS. Impairment of intestinal barrier function is a key factor in triggering IFALD, therefore promoting intestinal repair is particularly important. Intestinal repair mainly relies on the function of intestinal stem cells (ISC), which require robust mitochondrial fatty acid oxidation (FAO) for self-renewal. Herein, we report that aberrant LGR5+ ISC function in IFALD may be attributed to impaired farnesoid X receptor (FXR) signaling, a transcriptional factor activated by steroids and bile acids. In both surgical biopsies and patient-derived organoids (PDOs), SBS patients with IFALD represented lower population of LGR5+ cells and decreased FXR expression. Moreover, treatment with T-βMCA in PDOs (an antagonist for FXR) dose-dependently reduced the population of LGR5+ cells and the proliferation rate of enterocytes, concomitant with decreased key genes involved in FAO including CPT1a. Interestingly, however, treatment with Tropifexor in PDOs (an agonist for FXR) only enhanced FAO capacity, without improvement in ISC function and enterocyte proliferation. In conclusion, these findings suggested that impaired FXR may accelerate the depletion of LGR5 + ISC population through disrupted FAO processes, which may serve as a new potential target of preventive interventions against IFALD for SBS patients.
    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:在涡虫和刺虫中观察到的显着再生能力与成体干细胞的活跃增殖及其后代的精确分化密切相关,在低再生动物的衰老过程中,这两种情况通常都会恶化。虽然在高度再生的生物体中保守的再生特异性基因可以赋予再生能力和长期维持组织稳态,目前尚不清楚将这些再生基因引入低再生动物是否可以改善其再生和衰老过程。
    结果:这里,我们在果蝇中异位表达高度再生的物种特异性JmjC结构域编码基因(HRJDs),一种广泛使用的低再生模式生物。令人惊讶的是,HRJD表达阻碍了正在发育的翼盘中的组织再生,但在非再生条件下在成体中肠的肠干细胞谱系中表达时延长了生物体的寿命。值得注意的是,HRJD增强肠干细胞的增殖活性,同时保持其分化保真度,改善与年龄相关的肠道屏障功能下降。
    结论:这些发现共同表明,在衰老动物中表达时,引入高度再生的物种特异性基因可以改善干细胞功能并促进健康的寿命。
    BACKGROUND: The remarkable regenerative abilities observed in planarians and cnidarians are closely linked to the active proliferation of adult stem cells and the precise differentiation of their progeny, both of which typically deteriorate during aging in low regenerative animals. While regeneration-specific genes conserved in highly regenerative organisms may confer regenerative abilities and long-term maintenance of tissue homeostasis, it remains unclear whether introducing these regenerative genes into low regenerative animals can improve their regeneration and aging processes.
    RESULTS: Here, we ectopically express highly regenerative species-specific JmjC domain-encoding genes (HRJDs) in Drosophila, a widely used low regenerative model organism. Surprisingly, HRJD expression impedes tissue regeneration in the developing wing disc but extends organismal lifespan when expressed in the intestinal stem cell lineages of the adult midgut under non-regenerative conditions. Notably, HRJDs enhance the proliferative activity of intestinal stem cells while maintaining their differentiation fidelity, ameliorating age-related decline in gut barrier functions.
    CONCLUSIONS: These findings together suggest that the introduction of highly regenerative species-specific genes can improve stem cell functions and promote a healthy lifespan when expressed in aging animals.
    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
    肠干细胞(ISC)是维持肠道更新所必需的。这里,研究发现,高原大麦β-葡聚糖(HBG)可减轻结肠炎小鼠的病理症状,促进肠干细胞的增殖。值得注意的是,代谢组学研究表明,HBG治疗可显著增加二十二碳六烯酸(DHA).DHA是过氧化物酶体增殖物激活受体α(PPARα)的配体,这可以促进ISC的增殖。期望,HBG促进结肠炎小鼠肠道PPARα的表达和ISC的增殖。进一步的实验证实,DHA显著促进PPARα的表达和ISC在肠道类器官中的增殖。有趣的是,PPARα抑制剂可逆转DHA对ISC增殖的影响。一起,我们的数据表明HBG可能通过DHA加速PPARα介导的ISC增殖。这为多糖在维持肠道稳态中的有效应用提供了新的见解。
    Intestinal stem cells (ISCs) are necessary to maintain intestinal renewal. Here, we found that the highland barley β-glucan (HBG) alleviated pathological symptoms and promoted the proliferation of intestinal stem cells in colitis mice. Notably, metabolomics studies showed that docosahexaenoic acid (DHA) was significantly increased by the HBG treatment. DHA is a ligand for peroxisome proliferator-activated receptor α (PPARα), which can promote ISC proliferation. Expectedly, HBG facilitated the expression of intestinal PPARα and the proliferation of ISCs in colitis mice. Further experiments verified that DHA significantly facilitated the expression of PPARα and the proliferation of ISCs in intestinal organoids. Intriguingly, the effect of DHA on ISC proliferation was reversed by the PPARα inhibitor. Together, our data indicate that HBG might accelerate PPARα-mediated ISC proliferation through DHA. This provides new insights into the effective application of polysaccharides in maintaining intestinal homeostasis.
    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
    去泛素酶(DUB)对于维持蛋白质稳态和将蛋白质组装成功能复合物至关重要。尽管人们对DUBs的生物学功能越来越感兴趣,DUB在调节肠干细胞(ISC)和肠道稳态中的作用仍然未知。这里,我们通过诱导敲低成人中肠ISC和肠母细胞(EB)中DUBs的表达进行体内RNAi筛选,以鉴定果蝇中肠道稳态的DUB调节因子。我们筛选了43个DUB,并确定了ISC稳态所需的8个DUB。usp1,CG7857,usp5,rpn8,usp10和csn5的敲除减少了ISC/EB的数量,而CG4968和usp8的敲低增加了ISC/EB的数量。此外,ISC/EBs中usp1,CG4968,CG7857或rpn8的敲低破坏了肠屏障的完整性并缩短了寿命,表明这些DUB对维持肠道稳态的要求。此外,我们提供证据表明Usp1通过调节Notch信号活性介导ISC谱系分化。我们的研究表明,第一次,维持果蝇肠道稳态所需的去泛素酶,并为DUB和肠道稳态之间的功能联系提供新的见解。
    Deubiquitinases (DUBs) are essential for the maintenance of protein homeostasis and assembly of proteins into functional complexes. Despite growing interest in DUBs biological functions, the roles of DUBs in regulating intestinal stem cells (ISCs) and gut homeostasis remain largely unknown. Here, we perform an in vivo RNAi screen through induced knock-down of DUBs expression in adult midgut ISCs and enteroblasts (EBs) to identify DUB regulators of intestinal homeostasis in Drosophila. We screen 43 DUBs and identify 8 DUBs that are required for ISCs homeostasis. Knocking-down of usp1, CG7857, usp5, rpn8, usp10 and csn5 decreases the number of ISCs/EBs, while knocking-down of CG4968 and usp8 increases the number of ISCs/EBs. Moreover, knock-down of usp1, CG4968, CG7857, or rpn8 in ISCs/EBs disrupts the intestinal barrier integrity and shortens the lifespan, indicating the requirement of these DUBs for the maintenance of gut homeostasis. Furthermore, we provide evidences that USP1 mediates ISC lineage differentiation via modulating the Notch signaling activity. Our study identifies, for the first time, the deubiquitinases required for the maintenance of intestinal homeostasis in Drosophila, and provide new insights into the functional links between the DUBs and intestinal homeostasis.
    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
    全氟辛酸(PFOA),一种持久性有机污染物,以其化学稳定性而闻名,广泛分散在环境中,通过各种暴露途径,如摄入,对哺乳动物构成重大健康风险,吸入,和皮肤接触。在这项研究中,小鼠通过饮用水暴露于PFOA(0、0.2、2mg/L)180天,以研究其对结肠的毒性作用。我们通过RNA测序鉴定了差异表达的基因,并验证了PFOA对这些基因在结肠组织中表达的影响。我们的发现表明,长期暴露于PFOA会对小鼠结肠造成炎症性肠病(IBD)样损害。我们发现PFOA可以引起肠屏障的损伤。PFOA暴露后Wnt信号通路的抑制导致小鼠结肠中干细胞功能受损。此外,PFOA激活PPAR信号通路,破坏结肠组织中的细胞脂质代谢。此外,PFOA诱导结肠组织炎症反应,促进NLR家族,含pyrin域3(NLRP3)的炎症体激活与细胞凋亡。这项研究提供了对长期暴露于PFOA导致小鼠结肠组织损伤的机制的透彻理解。
    Perfluorooctanoic acid (PFOA), a persistent organic pollutant known for its chemical stability, is widely dispersed in the environment, posing significant health risks to mammals through various exposure routes such as ingestion, inhalation, and dermal contact. In this study, mice were exposed to PFOA (0, 0.2, 2 mg/L) through drinking water for 180 days to investigate its toxic effects on the colon. We identified differentially expressed genes through RNA sequencing and validated the impact of PFOA on the expression of these genes in colon tissue. Our findings revealed that long-term exposure to PFOA caused inflammatory bowel disease (IBD)-like damage to the mouse colon. We found PFOA could induce damage to the intestinal barrier. Inhibition of the Wnt signaling pathway following PFOA exposure results in impaired stem cell function in the colon of mice. Furthermore, PFOA activated the PPAR signaling pathway, disrupting cellular lipid metabolism in colon tissues. Additionally, PFOA induced inflammatory responses in colon tissue, facilitating NLR family, pyrin domain containing 3 (NLRP3) inflammasome activation and cell apoptosis. This study offers a thorough understanding of the mechanisms responsible for the damage to mouse colon tissue resulting from long-term exposure to PFOA.
    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: 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

公众号