enteric neuron

  • 文章类型: Journal Article
    这里,通过证明有丝分裂后肠神经元(EN)的神经支配潜力,我们确定可塑性存在于出生后肠神经系统中。使用BAF53b-Cre小鼠进行选择性神经元示踪,在多个模型系统中显示了成熟的产后EN的神经支配能力。分离的ENs在体外再生神经突,神经突复杂性和方向受与肠胶质细胞(EGC)接触的影响。来自移植EN的神经纤维仅与固有肌层内的EGC连接并沿着EGC传播。居民EGCs在Cre依赖性ENs消融后持续存在,并控制肠肌间神经丛的结构以恢复ENs的神经,如神经纤维投影追踪所示。体内移植和光遗传学实验突出了有丝分裂后神经元的快速神经支配潜力,导致2周内恢复肠道肌肉收缩活动。这些研究说明了有丝分裂后ENs的结构和功能神经支配能力以及EGC在引导和图案化其轨迹中的关键作用。
    Here, we establish that plasticity exists within the postnatal enteric nervous system by demonstrating the reinnervation potential of post-mitotic enteric neurons (ENs). Employing BAF53b-Cre mice for selective neuronal tracing, the reinnervation capabilities of mature postnatal ENs are shown across multiple model systems. Isolated ENs regenerate neurites in vitro, with neurite complexity and direction influenced by contact with enteric glial cells (EGCs). Nerve fibers from transplanted ENs exclusively interface and travel along EGCs within the muscularis propria. Resident EGCs persist after Cre-dependent ablation of ENs and govern the architecture of the myenteric plexus for reinnervating ENs, as shown by nerve fiber projection tracing. Transplantation and optogenetic experiments in vivo highlight the rapid reinnervation potential of post-mitotic neurons, leading to restored gut muscle contractile activity within 2 weeks. These studies illustrate the structural and functional reinnervation capacity of post-mitotic ENs and the critical role of EGCs in guiding and patterning their trajectories.
    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
    背景:腹部手术引起的肠道炎症是术后肠梗阻(POI)发展的重要因素。ST36电针(EA)已被证明可以缓解肠道炎症并恢复POI的胃肠道动力障碍。本研究旨在阐明参与POI中EA抗炎特性的神经免疫途径。
    方法:进行肠道操作(IM)诱导POI后,IM后24小时评估肠道炎症和运动,通过评估胃肠道运输(GIT),细胞因子表达,和白细胞浸润。实验手术,药理干预,基因敲除小鼠用于阐明EA的神经免疫机制。
    结果:在小鼠IM后,ST36处的EA显着改善了GIT并降低了促炎细胞因子的表达和肠肌层中白细胞的浸润。膈下迷走神经切断术消除了EA治疗的抗炎作用,而脾切除术并不妨碍EA治疗的抗炎益处。氯化六甲铵(HEX)的给药有助于显着降低EA抑制炎症和增强运动功能障碍的能力,EA在α7烟碱乙酰胆碱受体(α7nAChR)敲除小鼠中无效。
    结论:ST36处的EA通过需要迷走神经支配但独立于脾脏的神经回路预防肠道炎症和运动障碍。进一步的发现表明,该过程涉及肠神经元介导迷走神经信号,并需要α7nAChR的存在。这些发现表明,在ST36处利用EA可能代表POI和其他免疫相关胃肠道疾病的可能治疗方法。
    BACKGROUND: The induction of intestinal inflammation as a result of abdominal surgery is an essential factor in postoperative ileus (POI) development. Electroacupuncture (EA) at ST36 has been demonstrated to relieve intestinal inflammation and restore gastrointestinal dysmotility in POI. This study aims to elucidate the neuroimmune pathway involved in the anti-inflammatory properties of EA in POI.
    METHODS: After intestinal manipulation (IM) was performed to induce POI, intestinal inflammation and motility were assessed 24 h post-IM, by evaluating gastrointestinal transit (GIT), cytokines expression, and leukocyte infiltration. Experimental surgery, pharmacological intervention, and genetic knockout mice were used to elucidate the neuroimmune mechanisms of EA.
    RESULTS: EA at ST36 significantly improved GIT and reduced the expression of pro-inflammatory cytokines and leukocyte infiltration in the intestinal muscularis following IM in mice. The anti-inflammatory effectiveness of EA treatment was abolished by sub-diaphragmatic vagotomy, whereas splenectomy did not hinder the anti-inflammatory benefits of EA treatment. The hexamethonium chloride (HEX) administration contributes to a notable reduction in the EA capacity to suppress inflammation and enhance motility dysfunction, and EA is ineffective in α7 nicotinic acetylcholine receptor (α7nAChR) knockout mice.
    CONCLUSIONS: EA at ST36 prevents intestinal inflammation and dysmotility through a neural circuit that requires vagal innervation but is independent of the spleen. Further findings revealed that the process involves enteric neurons mediating the vagal signal and requires the presence of α7nAChR. These findings suggest that utilizing EA at ST36 may represent a possible therapeutic approach for POI and other immune-related gastrointestinal 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: Preprint
    脊椎动物肠神经系统(ENS)是驻留在整个胃肠道(GI)内的肠神经元和神经胶质的关键网络。监督基本的胃肠道功能,如肠道运动和水分平衡,ENS是肠-脑轴的关键双向链接。在早期开发中,ENS主要来源于肠神经嵴细胞(ENCCs)。ENCC开发中断,如在先天性巨结肠病(HSCR)等疾病中看到的,导致GI中没有ENS,特别是在结肠。在这项研究中,使用斑马鱼,我们设计了一种基于CRISPR的体内F0屏幕,快速管道整合单细胞RNA测序,CRISPR反向遗传学,和高含量的成像。我们的发现揭示了各种基因,包括那些编码阿片受体的,作为ENS建立的可能的监管机构。此外,我们提供的证据表明阿片受体参与幼虫ENS的神经化学编码。总之,我们的作品呈现了一部小说,针对ENS开发的高效CRISPR屏幕,促进以前未知基因的发现,增加神经系统建设的知识。
    The vertebrate enteric nervous system (ENS) is a crucial network of enteric neurons and glia resident within the entire gastrointestinal tract (GI). Overseeing essential GI functions such as gut motility and water balance, the ENS serves as a pivotal bidirectional link in the gut-brain axis. During early development, the ENS is primarily derived from enteric neural crest cells (ENCCs). Disruptions to ENCC development, as seen in conditions like Hirschsprung disease (HSCR), lead to absence of ENS in the GI, particularly in the colon. In this study, using zebrafish, we devised an in vivo F0 CRISPR-based screen employing a robust, rapid pipeline integrating single-cell RNA sequencing, CRISPR reverse genetics, and high-content imaging. Our findings unveil various genes, including those encoding for opioid receptors, as possible regulators of ENS establishment. In addition, we present evidence that suggests opioid receptor involvement in neurochemical coding of the larval ENS. In summary, our work presents a novel, efficient CRISPR screen targeting ENS development, facilitating the discovery of previously unknown genes, and increasing knowledge of nervous system construction.
    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
    巨噬细胞在妊娠早期填充胚胎,但是他们在发展中的作用并不明确。特别是,巨噬细胞在肠道发育中的规格和功能仍鲜有探索。为了在人类发展背景下研究这一事件,我们从多能干细胞中提取并结合了人类肠道类器官和巨噬细胞。巨噬细胞迁移到类器官中,增殖,并占据了上皮隐窝和神经节的新兴微解剖壁。它们还获得了类似于胎儿肠巨噬细胞的转录组学谱,并显示组织巨噬细胞行为,例如招募组织损伤。使用这个模型,我们显示巨噬细胞减少间充质细胞的糖酵解并限制组织生长而不影响组织结构,与肠神经元的促生长作用相反。总之,我们设计了一个充满巨噬细胞的肠组织模型,我们认为常驻巨噬细胞有助于调节发育中的肠道的代谢和生长。
    Macrophages populate the embryo early in gestation, but their role in development is not well defined. In particular, specification and function of macrophages in intestinal development remain little explored. To study this event in the human developmental context, we derived and combined human intestinal organoid and macrophages from pluripotent stem cells. Macrophages migrate into the organoid, proliferate, and occupy the emerging microanatomical niches of epithelial crypts and ganglia. They also acquire a transcriptomic profile similar to that of fetal intestinal macrophages and display tissue macrophage behaviors, such as recruitment to tissue injury. Using this model, we show that macrophages reduce glycolysis in mesenchymal cells and limit tissue growth without affecting tissue architecture, in contrast to the pro-growth effect of enteric neurons. In short, we engineered an intestinal tissue model populated with macrophages, and we suggest that resident macrophages contribute to the regulation of metabolism and growth of the developing 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
    氧化应激越来越被认为是一系列胃肠道(GI)疾病的主要参与者,以及治疗干预引起的并发症。本文概述了胃肠道疾病中氧化应激的机制,并强调了氧化损伤和肠神经系统(ENS)破坏之间的联系。控制GI功能。ENS的功能障碍是一系列疾病的特征,包括神经肠道疾病和条件,如炎症性肠病(IBD),糖尿病性胃轻瘫,和化疗引起的胃肠道副作用。ENS中的神经元,虽然对正常的肠道功能至关重要,显得特别容易受到氧化损伤。机械上,肠神经元的氧化应激可由内在亚硝基损伤引起,线粒体功能障碍,或炎症相关途径。尽管基于抗氧化剂的疗法显示出有限的疗效,认识到氧化应激在胃肠道疾病中的多方面作用为未来的干预提供了有希望的途径。这篇全面的综述总结了迄今为止有关氧化应激在胃肠道疾病的病理生理学中起关键作用的文献。重点关注其在ENS损伤和功能障碍中的作用,并强调了开发针对这些疾病的靶向疗法的机会。
    Oxidative stress is increasingly recognized as a central player in a range of gastrointestinal (GI) disorders, as well as complications stemming from therapeutic interventions. This article presents an overview of the mechanisms of oxidative stress in GI conditions and highlights a link between oxidative insult and disruption to the enteric nervous system (ENS), which controls GI functions. The dysfunction of the ENS is characteristic of a spectrum of disorders, including neurointestinal diseases and conditions such as inflammatory bowel disease (IBD), diabetic gastroparesis, and chemotherapy-induced GI side effects. Neurons in the ENS, while essential for normal gut function, appear particularly vulnerable to oxidative damage. Mechanistically, oxidative stress in enteric neurons can result from intrinsic nitrosative injury, mitochondrial dysfunction, or inflammation-related pathways. Although antioxidant-based therapies have shown limited efficacy, recognizing the multifaceted role of oxidative stress in GI diseases offers a promising avenue for future interventions. This comprehensive review summarizes the literature to date implicating oxidative stress as a critical player in the pathophysiology of GI disorders, with a focus on its role in ENS injury and dysfunction, and highlights opportunities for the development of targeted therapeutics for these diseases.
    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
    由于缺乏有效的治疗方法,对炎性肠病(IBD)患者的腹痛管理仍然存在问题。乙酰L-肉碱(ALCAR)已被证明可用于治疗不同类型的慢性疼痛,具有出色的耐受性。本工作旨在评估ALCAR在与2,4-二硝基苯磺酸(DNBS)注射诱导的结肠炎相关的持续性内脏痛模型中的抗痛觉过敏功效。应用两种不同的方案。在预防方案中,在递送DNBS前14天至24小时开始每天施用ALCAR。在干预协议中,ALCAR在DNBS注射的同一天开始每日给药,治疗持续14天。在这两种情况下,ALCAR显著降低DNBS处理的动物内脏痛觉过敏的建立,尽管干预方案比预防性方案显示出更大的疗效。干预方案部分减少大鼠结肠损伤,对抗结肠炎引起的肠胶质细胞和脊髓星形胶质细胞活化,通过免疫荧光分析。另一方面,预防方案有效地保护肠神经元免受炎性损伤。这些发现表明ALCAR作为IBD患者的食品补充剂的推定有用性。
    The management of abdominal pain in patients affected by inflammatory bowel diseases (IBDs) still represents a problem because of the lack of effective treatments. Acetyl L-carnitine (ALCAR) has proved useful in the treatment of different types of chronic pain with excellent tolerability. The present work aimed at evaluating the anti-hyperalgesic efficacy of ALCAR in a model of persistent visceral pain associated with colitis induced by 2,4-dinitrobenzene sulfonic acid (DNBS) injection. Two different protocols were applied. In the preventive protocol, ALCAR was administered daily starting 14 days to 24 h before the delivery of DNBS. In the interventive protocol, ALCAR was daily administered starting the same day of DNBS injection, and the treatment was continued for 14 days. In both cases, ALCAR significantly reduced the establishment of visceral hyperalgesia in DNBS-treated animals, though the interventive protocol showed a greater efficacy than the preventive one. The interventive protocol partially reduced colon damage in rats, counteracting enteric glia and spinal astrocyte activation resulting from colitis, as analyzed by immunofluorescence. On the other hand, the preventive protocol effectively protected enteric neurons from the inflammatory insult. These findings suggest the putative usefulness of ALCAR as a food supplement for patients suffering from IBDs.
    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
    神经系统对肠道稳态和功能至关重要,但是关于它对肠道免疫防御的影响仍然存在疑问。通过筛选秀丽隐杆线虫的主要神经递质,我们发现γ-氨基丁酸(GABA)缺乏可增强致病性铜绿假单胞菌PA14感染的敏感性。肠神经元和肠平滑肌之间的GABA能信号促进PMK-1/p38依赖性肠防御,但IIS/DAF-16-和DBL-1/TGF-β非依赖性,通路。转录组学分析显示神经肽,FLP-6,作用于肠道GABA能信号的下游。进一步的数据确定FLP-6由肠平滑肌细胞表达和分泌,并在肠上皮上充当旁分泌分子。FLP-6抑制转录因子ZIP-10和KLF-1,这些转录因子在肠上皮细胞中并行起作用并收敛于PMK-1/p38途径,以进行先天免疫和肠道防御。总的来说,这些发现揭示了一个肠神经元-肌肉-上皮轴,该轴在高等生物中可能在进化上是保守的。
    The nervous system is critical for intestinal homeostasis and function, but questions remain regarding its impact on gut immune defense. By screening the major neurotransmitters of C. elegans, we found that γ-aminobutyric acid (GABA) deficiency enhanced susceptibility to pathogenic Pseudomonas aeruginosa PA14 infection. GABAergic signaling between enteric neurons and intestinal smooth muscle promoted gut defense in a PMK-1/p38-dependent, but IIS/DAF-16- and DBL-1/TGF-β-independent, pathway. Transcriptomic profiling revealed that the neuropeptide, FLP-6, acted downstream of enteric GABAergic signaling. Further data determined that FLP-6 was expressed and secreted by intestinal smooth muscle cells and functioned as a paracrine molecule on the intestinal epithelium. FLP-6 suppressed the transcription factors ZIP-10 and KLF-1 that worked in parallel and converged to the PMK-1/p38 pathway in the intestinal epithelia for innate immunity and gut defense. Collectively, these findings uncover an enteric neuron-muscle-epithelium axis that may be evolutionarily conserved in higher organisms.
    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: Review
    肠神经系统是位于胃肠道中的肠神经元和神经胶质的密集网络。该系统负责通过多种信号传导过程(包括与免疫系统相互作用产生的信号传导过程)执行几种功能,使消化以及维持肠道稳态。肠神经元和肠神经胶质细胞之间的双向通信在肠神经系统功能中起着至关重要的作用而受到越来越多的关注。诸如神经递质的神经元介质刺激肠神经胶质细胞,并且随后的神经胶质传递过程在肠运动控制期间改善神经元信号传导。在这个小型审查中,我们介绍并讨论了肠神经系统中神经元和神经胶质细胞间信号传导的基础,以及这些相互作用与肠道功能的相关性。
    The enteric nervous system is a dense network of enteric neurons and glia housed in the gastrointestinal tract. This system is responsible for performing several functions that enable digestion as well as maintaining gut homeostasis through diverse signaling processes including those that arise from interactions with the immune system. Bidirectional communication between enteric neurons and enteric glia has gained increased attention for playing essential roles in enteric nervous system function. Neuronal mediators such as neurotransmitters stimulate enteric glia and subsequent gliotransmission processes refine neuronal signaling during intestinal motor control. In this mini-review, we present and discuss the basis of intercellular signaling between neurons and glia in the enteric nervous system and the relevance of these interactions to gut 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
    肠神经系统(ENS)是支配整个消化道并调节主要消化功能的内在神经系统。最近的证据表明,ENS的功能严重依赖于肠神经元的连接;然而,破译潜在机制的实验模型是有限的。与中枢神经系统相比,纯神经元培养物已经发展了几十年,被认为是神经科学领域的参考,缺乏肠神经元的等效模型。在这项研究中,我们开发了一种新型的高纯度大鼠胚胎肠神经元模型,具有密集和功能性的突触网络。该方法简单且相对快速。我们使用免疫组织化学表征肠神经元,形态学,和电生理学方法。特别是,我们证明了该培养模型在多电极阵列技术中的适用性,该技术是监测肠神经元网络活动的新方法。这种高纯度肠神经元的体外模型代表了一种有价值的新工具,可以更好地理解建立和维持肠神经元突触连接和功能网络的机制。
    The enteric nervous system (ENS) is the intrinsic nervous system that innervates the entire digestive tract and regulates major digestive functions. Recent evidence has shown that functions of the ENS critically rely on enteric neuronal connectivity; however, experimental models to decipher the underlying mechanisms are limited. Compared to the central nervous system, for which pure neuronal cultures have been developed for decades and are recognized as a reference in the field of neuroscience, an equivalent model for enteric neurons is lacking. In this study, we developed a novel model of highly pure rat embryonic enteric neurons with dense and functional synaptic networks. The methodology is simple and relatively fast. We characterized enteric neurons using immunohistochemical, morphological, and electrophysiological approaches. In particular, we demonstrated the applicability of this culture model to multi-electrode array technology as a new approach for monitoring enteric neuronal network activity. This in vitro model of highly pure enteric neurons represents a valuable new tool for better understanding the mechanisms involved in the establishment and maintenance of enteric neuron synaptic connectivity and functional networks.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号