enteric neuropathies

肠神经病
  • 文章类型: Journal Article
    这篇综述讨论了肠神经系统(ENS)中DNA损伤和修复的探索较少的领域,通常被称为“第二大脑”。“虽然中枢神经系统的DNA修复机制和相关的神经病理学已经得到了广泛的研究,ENS,可以自主协调胃肠功能,经历了与其基因组完整性相关的独特挑战和脆弱性。ENS对DNA损伤的敏感性因其有限的保护屏障而加剧,不仅导致内源性基因毒性暴露,如氧化应激,但也有外来威胁,如摄入的环境污染物,局部炎症反应,和肠道生态失调。这里,我们讨论了肠神经病中DNA修复缺陷的证据,最值得注意的是,据报道,RAD21和LIG3的遗传突变与慢性肠假性梗阻和线粒体胃肠道脑肌病之间的关系,分别。我们还介绍了DNA修复综合征中较少认识的胃肠道并发症,包括像Cockayne综合征这样的病症.该综述最后指出了DNA修复缺陷不仅在先天性疾病中而且在与衰老相关的肠道功能障碍中的潜在作用。以及迫切需要进一步研究以建立DNA损伤积累与ENS特异性病理表型之间的直接因果关系。
    This review discusses the less-explored realm of DNA damage and repair within the enteric nervous system (ENS), often referred to as the \"second brain.\" While the central nervous system has been extensively studied for its DNA repair mechanisms and associated neuropathologies, the ENS, which can autonomously coordinate gastrointestinal function, experiences unique challenges and vulnerabilities related to its genome integrity. The susceptibility of the ENS to DNA damage is exacerbated by its limited protective barriers, resulting in not only endogenous genotoxic exposures, such as oxidative stress, but also exogenous threats, such as ingested environmental contaminants, local inflammatory responses, and gut dysbiosis. Here, we discuss the evidence for DNA repair defects in enteric neuropathies, most notably, the reported relationship between inherited mutations in RAD21 and LIG3 with chronic intestinal pseudo-obstruction and mitochondrial gastrointestinal encephalomyopathy disorders, respectively. We also introduce the lesser-recognized gastrointestinal complications in DNA repair syndromes, including conditions like Cockayne syndrome. The review concludes by pointing out the potential role of DNA repair defects in not only congenital disorders but also aging-related gut dysfunction, as well as the crucial need for further research to establish direct causal links between DNA damage accumulation and ENS-specific pathologic phenotypes.
    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
    肠神经病的特征是肠道神经支配异常,包括肠神经系统,在其他功能障碍中诱导严重的肠道运动障碍。大部分胃肠道受迷走神经支配,其传出分支与肠神经系统有密切的联系,其传入分布在消化壁的不同层。迷走神经是自主神经系统的重要组成部分,参与应激反应,在微生物群-肠道-大脑轴的界面,具有抗炎和促动力特性,调节肠道通透性,具有显著的可塑性和再生能力。针对迷走神经的这些特性,迷走神经刺激(或非刺激/药理学方法),可能对肠神经病的治疗管理感兴趣。
    Enteric neuropathies are characterized by abnormalities of gut innervation, which includes the enteric nervous system, inducing severe gut dysmotility among other dysfunctions. Most of the gastrointestinal tract is innervated by the vagus nerve, the efferent branches of which have close interconnections with the enteric nervous system and whose afferents are distributed throughout the different layers of the digestive wall. The vagus nerve is a key element of the autonomic nervous system, involved in the stress response, at the interface of the microbiota-gut-brain axis, has anti-inflammatory and prokinetic properties, modulates intestinal permeability, and has a significant capacity of plasticity and regeneration. Targeting these properties of the vagus nerve, with vagus nerve stimulation (or non-stimulation/ pharmacological methods), could be of interest in the therapeutic management of enteric neuropathies.
    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
    肠神经系统(ENS)是胃肠道(GI)壁内神经元和神经胶质的广泛网络,可调节许多基本的GI功能。因此,由于发育缺陷导致的ENS疾病,炎症,感染,或年龄相关的神经变性导致严重的神经肠道疾病。尽管这些疾病的患病率和严重性,缺乏有效的治疗方法,因为它们不能直接解决潜在的病理学问题。神经元干细胞疗法代表了一种有希望的方法,通过替换缺失或受损的神经元来治疗ENS疾病,通过消除对免疫抑制的需要,自体干细胞来源将是最佳的。我们利用猪模型来解决有关细胞分离的关键问题,delivery,雕刻,以及与人类治疗相关的大型动物的命运。我们成功地从1个月大的猪切除的一段小肠中分离出神经干细胞。将肠神经元干细胞(ENSC)扩增为在低氧(5%)培养条件下最佳生长的神经球。通过慢病毒绿色荧光蛋白(GFP)转导标记肠神经元干细胞,然后移植到收获它们的同一只猪中。然后利用内窥镜超声将ENSC(每只动物10,000-30,000个神经球)递送到直肠壁中。注射后10天和28天,自体来源的ENSC被发现移植在直肠壁内,神经胶质分化,没有异位扩散的证据。这些发现强烈支持使用临床有用和微创技术分离和递送自体细胞的可行性,使我们更接近首次在人类ENSC治疗神经肠道疾病。
    The enteric nervous system (ENS) is an extensive network of neurons and glia within the wall of the gastrointestinal (GI) tract that regulates many essential GI functions. Consequently, disorders of the ENS due to developmental defects, inflammation, infection, or age-associated neurodegeneration lead to serious neurointestinal diseases. Despite the prevalence and severity of these diseases, effective treatments are lacking as they fail to directly address the underlying pathology. Neuronal stem cell therapy represents a promising approach to treating diseases of the ENS by replacing the absent or injured neurons, and an autologous source of stem cells would be optimal by obviating the need for immunosuppression. We utilized the swine model to address key questions concerning cell isolation, delivery, engraftment, and fate in a large animal relevant to human therapy. We successfully isolated neural stem cells from a segment of small intestine resected from 1-month-old swine. Enteric neuronal stem cells (ENSCs) were expanded as neurospheres that grew optimally in low-oxygen (5%) culture conditions. Enteric neuronal stem cells were labeled by lentiviral green fluorescent protein (GFP) transduction, then transplanted into the same swine from which they had been harvested. Endoscopic ultrasound was then utilized to deliver the ENSCs (10,000-30,000 neurospheres per animal) into the rectal wall. At 10 and 28 days following injection, autologously derived ENSCs were found to have engrafted within rectal wall, with neuroglial differentiation and no evidence of ectopic spreading. These findings strongly support the feasibility of autologous cell isolation and delivery using a clinically useful and minimally invasive technique, bringing us closer to first-in-human ENSC therapy for neurointestinal 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
    背景:肠神经病,由肠神经系统异常引起的,与显著的发病率和高昂的医疗费用有关,但目前的治疗方法并不令人满意。基于细胞的疗法提供了一种创新的方法来替代缺失或异常的肠神经元,从而恢复肠道功能。
    方法:从Wnt1-Cre;R26td番茄小鼠的胃肠道中分离肠神经元干细胞(ENSC)并产生神经球(NS)。NS移植是通过注射到nNOS-/-小鼠的中结肠间质进行的,结肠运动障碍的模型,使用1次(n=12)或3次(n=12)注射(每次注射30NS),纵向间隔1-2mm。6周后使用肌电图(EMG)评估功能结果,电场刺激(EFS),光遗传学,通过测量结肠直肠的运动。
    结果:移植的ENSC在nNOS-/-受体结肠中形成了硝能神经元。与单独单次注射相比,多次注射ENSC导致明显更大的覆盖面积,并且与结肠功能的显着改善有关。通过(1)通过肌电图记录增加结肠肌肉活动,(2)更快的直肠珠排出,和(3)增加体内粪便颗粒产量。器官浴研究揭示了通过光遗传学刺激表达通道视紫红质的ENSC和响应EFS而恢复平滑肌松弛的直接神经肌肉通讯。
    结论:这些结果表明,移植的ENSC可以在结肠运动障碍模型中形成有效的神经肌肉连接并改善结肠运动功能,并另外揭示了多个细胞递送位点导致改善的反应,为优化临床试验设计铺平了道路。
    Enteric neuropathies, which result from abnormalities of the enteric nervous system, are associated with significant morbidity and high health-care costs, but current treatments are unsatisfactory. Cell-based therapy offers an innovative approach to replace the absent or abnormal enteric neurons and thereby restore gut function.
    Enteric neuronal stem cells (ENSCs) were isolated from the gastrointestinal tract of Wnt1-Cre;R26tdTomato mice and generated neurospheres (NS). NS transplants were performed via injection into the mid-colon mesenchyme of nNOS-/- mouse, a model of colonic dysmotility, using either 1 (n = 12) or 3 (n = 12) injections (30 NS per injection) targeted longitudinally 1-2 mm apart. Functional outcomes were assessed up to 6 weeks later using electromyography (EMG), electrical field stimulation (EFS), optogenetics, and by measuring colorectal motility.
    Transplanted ENSCs formed nitrergic neurons in the nNOS-/- recipient colon. Multiple injections of ENSCs resulted in a significantly larger area of coverage compared to single injection alone and were associated with a marked improvement in colonic function, demonstrated by (1) increased colonic muscle activity by EMG recording, (2) faster rectal bead expulsion, and (3) increased fecal pellet output in vivo. Organ bath studies revealed direct neuromuscular communication by optogenetic stimulation of channelrhodopsin-expressing ENSCs and restoration of smooth muscle relaxation in response to EFS.
    These results demonstrate that transplanted ENSCs can form effective neuromuscular connections and improve colonic motor function in a model of colonic dysmotility, and additionally reveal that multiple sites of cell delivery led to an improved response, paving the way for optimized clinical trial design.
    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
    严重的肠动力障碍的特征在于肠内容物的无效推进。因此,患者出现致残/痛苦的症状,如恶心和呕吐以及排便习惯的改变,直至放射学上可证明的肠道亚阻塞性发作。慢性假性肠梗阻(CIPO)是严重肠动力障碍的典型临床表型。这种综合征是由于改变了内在(肠)神经支配和外在神经供应的形态功能完整性(因此是神经病变)而发生的。Cajal间质细胞(ICC)(间细胞病),和平滑肌细胞(肌病)。在过去的几年里,已在CIPO患者的不同亚群中鉴定出几种基因。本综述的重点是涵盖与CIPO相关的肠动力障碍的最新更新,突出显示(a)具有主要的潜在神经病变的形式,(b)以肌病为主的形式,和(c)线粒体疾病,具有明显的肠道功能障碍作为其临床表型的一部分。我们将对通过最近的证据证明的基因进行彻底的描述,这些基因会导致导致CIPO中异常的肠道收缩模式的神经(ICC)肌病。这种严重疾病的易感基因的发现可能为开发CIPO和其他形式的肠动力障碍的肠神经(ICC)肌病的目标疗法铺平道路。
    Severe gut motility disorders are characterized by the ineffective propulsion of intestinal contents. As a result, the patients develop disabling/distressful symptoms, such as nausea and vomiting along with altered bowel habits up to radiologically demonstrable intestinal sub-obstructive episodes. Chronic intestinal pseudo-obstruction (CIPO) is a typical clinical phenotype of severe gut dysmotility. This syndrome occurs due to changes altering the morpho-functional integrity of the intrinsic (enteric) innervation and extrinsic nerve supply (hence neuropathy), the interstitial cells of Cajal (ICC) (mesenchymopathy), and smooth muscle cells (myopathy). In the last years, several genes have been identified in different subsets of CIPO patients. The focus of this review is to cover the most recent update on enteric dysmotility related to CIPO, highlighting (a) forms with predominant underlying neuropathy, (b) forms with predominant myopathy, and (c) mitochondrial disorders with a clear gut dysfunction as part of their clinical phenotype. We will provide a thorough description of the genes that have been proven through recent evidence to cause neuro-(ICC)-myopathies leading to abnormal gut contractility patterns in CIPO. The discovery of susceptibility genes for this severe condition may pave the way for developing target therapies for enteric neuro-(ICC)-myopathies underlying CIPO and other forms of gut dysmotility.
    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
    促进肠神经系统(ENS)中的成人神经发生可能是治愈肠神经病的潜在治疗方法。肠神经胶质细胞(EGCs)是ENS中最丰富的神经胶质细胞。越来越多的证据表明,在ENS的成年神经发生过程中,EGC可能是提供新神经元的补充来源。在大脑中,已经对星形胶质细胞的神经元转换特性进行了深入的研究,和小分子已经成功地用于诱导星形胶质细胞到神经元的转变。然而,关于ENS中胶质细胞到神经元转换的研究仍然缺乏。在这项研究中,我们使用GFAP-Cre:Rosa-tdTomato小鼠在体内和体外追踪ENS中的神经胶质向神经元的转分化。我们表明,GFAP启动子驱动的tdTomato仅标记了EGC,并且是在成年小鼠ENS中追踪EGC及其后代细胞的合适标记。有趣的是,我们发现RepSox或其他ALK5抑制剂单独在体外诱导EGC有效转分化为神经元.ALK5的敲低进一步证实TGFβR-1/ALK5信号通路在EGC向神经元的转变中起着重要作用。RepSox诱导的神经元为Calbindin和nNOS阳性,并表现出典型的神经元电生理特性。最后,我们表明,施用RepSox(3,10mg·kg-1·d-1,i.g.)2周可显着促进ENS中EGC向神经元的转化,并影响成年小鼠的胃肠道运动。这项研究提供了一种通过小分子化合物将成年小鼠EGCs有效转化为神经元的方法,这可能是胃肠道神经病的一种有希望的治疗策略。
    Promoting adult neurogenesis in the enteric nervous system (ENS) may be a potential therapeutic approach to cure enteric neuropathies. Enteric glial cells (EGCs) are the most abundant glial cells in the ENS. Accumulating evidence suggests that EGCs can be a complementary source to supply new neurons during adult neurogenesis in the ENS. In the brain, astrocytes have been intensively studied for their neuronal conversion properties, and small molecules have been successfully used to induce the astrocyte-to-neuron transition. However, research on glia-to-neuron conversion in the ENS is still lacking. In this study, we used GFAP-Cre:Rosa-tdTomato mice to trace glia-to-neuron transdifferentiation in the ENS in vivo and in vitro. We showed that GFAP promoter-driven tdTomato exclusively labelled EGCs and was a suitable marker to trace EGCs and their progeny cells in the ENS of adult mice. Interestingly, we discovered that RepSox or other ALK5 inhibitors alone induced efficient transdifferentiation of EGCs into neurons in vitro. Knockdown of ALK5 further confirmed that the TGFβR-1/ALK5 signalling pathway played an essential role in the transition of EGCs to neurons. RepSox-induced neurons were Calbindin- and nNOS-positive and displayed typical neuronal electrophysiological properties. Finally, we showed that administration of RepSox (3, 10 mg· kg-1 ·d-1, i.g.) for 2 weeks significantly promoted the conversion of EGCs to neurons in the ENS and influenced gastrointestinal motility in adult mice. This study provides a method for efficiently converting adult mouse EGCs into neurons by small-molecule compounds, which might be a promising therapeutic strategy for gastrointestinal neuropathy.
    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
    The bewildering complexity of the enteric nervous system makes it susceptible to develop a wide array of motility disorders, collectively called enteric neuropathies. These gastrointestinal conditions are among the most challenging to manage, mainly given poor characterization of their etiopathophysiology and outcomes. Not surprisingly, therefore, targeted or curative therapies for enteric neuropathies are lacking and management is largely symptomatic. Nonetheless, recent advances in neurogastroenterology have witnessed improvements in established strategies, such as intestinal transplantation and the emergence of new treatments including novel drugs, electrical pacing, and manipulation of fecal microbiota, as well as stem cell and gene therapy.
    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
    单纯疱疹病毒1型(HSV-1),一种普遍存在于人群中的嗜神经病原体,感染人类和啮齿动物的肠神经系统(ENS),并导致大鼠肠神经肌肉功能障碍。尽管炎性细胞在肌间神经丛浸润和肠神经变性是功能性肠道疾病患者的共同特征,与HSV-1有致病联系的证据仍未确定,主要是因为其潜在机制在很大程度上是未知的.在这项研究中,我们证明了在胃内给药后,HSV-1会感染肌间丛内的神经元,从而导致ENS的功能和结构改变。通过用HSV-1复制缺陷菌株感染小鼠,我们发现胃肠道神经肌肉异常与病毒复制无关。的确,暴露于紫外线灭活的HSV-1的肠神经元产生单核细胞趋化蛋白1(MCP-1/CCL2),以招募纵向肌肉肌间神经丛中的活化巨噬细胞。浸润的巨噬细胞产生活性氧和氮,并直接损害肠神经元,导致胃肠动力障碍。在HSV-1感染的小鼠中,通过体内施用(i)含有二氯亚甲基双膦酸(氯膦酸盐)的脂质体以消耗组织巨噬细胞,可以改善肠神经肌肉功能障碍。(ii)CCR2趋化因子受体拮抗剂RS504393阻断CCL2/CCR2通路,(iii)Nω-硝基-L-精氨酸甲酯盐酸盐(L-NAME)和AR-C102222以淬灭通过iNOS产生的氮反应性物种的产生。总之,这些数据表明HSV-1感染通过产生特定的化学引诱因子使肠神经元募集巨噬细胞。所产生的炎症反应对于肠动力障碍是强制性的。这些发现提供了对HSV-1感染后ENS中发生的神经免疫通讯的见解,并允许识别胃肠道疾病的原始病理生理机制以及识别新的治疗靶标。
    Herpes Simplex Virus type 1 (HSV-1), a neurotropic pathogen widespread in human population, infects the enteric nervous system (ENS) in humans and rodents and causes intestinal neuromuscular dysfunction in rats. Although infiltration of inflammatory cells in the myenteric plexus and neurodegeneration of enteric nerves are common features of patients suffering from functional intestinal disorders, the proof of a pathogenic link with HSV-1 is still unsettled mainly because the underlying mechanisms are largely unknown. In this study we demonstrated that following intragastrical administration HSV-1 infects neurons within the myenteric plexus resulting in functional and structural alterations of the ENS. By infecting mice with HSV-1 replication-defective strain we revealed that gastrointestinal neuromuscular anomalies were however independent of viral replication. Indeed, enteric neurons exposed to UV-inactivated HSV-1 produced monocyte chemoattractant protein-1 (MCP-1/CCL2) to recruit activated macrophages in the longitudinal muscle myenteric plexus. Infiltrating macrophages produced reactive oxygen and nitrogen species and directly harmed enteric neurons resulting in gastrointestinal dysmotility. In HSV-1 infected mice intestinal neuromuscular dysfunctions were ameliorated by in vivo administration of (i) liposomes containing dichloromethylene bisphosphonic acid (clodronate) to deplete tissue macrophages, (ii) CCR2 chemokine receptor antagonist RS504393 to block the CCL2/CCR2 pathway, (iii) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) and AR-C 102222 to quench production of nitrogen reactive species produced via iNOS. Overall these data demonstrate that HSV-1 infection makes enteric neurons recruit macrophages via production of a specific chemoattractant factor. The resulting inflammatory reaction is mandatory for intestinal dysmotility. These findings provide insights into the neuro-immune communication that occurs in the ENS following HSV-1 infection and allow recognition of an original pathophysiologic mechanism underlying gastrointestinal diseases as well as identification of novel therapeutic targets.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The enteric nervous system (ENS) is the largest part of the peripheral nervous system and is entirely neural crest-derived. It provides the intrinsic innervation of the gut, controlling different aspects of gut function, such as motility. In this review, we will discuss key points of Zebrafish ENS development, genes, and signaling pathways regulating ENS development, as well as contributions of the Zebrafish model system to better understand ENS disorders. During their migration, enteric progenitor cells (EPCs) display a gradient of developmental states based on their proliferative and migratory characteristics, and show spatiotemporal heterogeneity based on gene expression patterns. Many genes and signaling pathways that regulate the migration and proliferation of EPCs have been identified, but later stages of ENS development, especially steps of neuronal and glial differentiation, remain poorly understood. In recent years, Zebrafish have become increasingly important to test candidate genes for ENS disorders (e.g., from genome-wide association studies), to identify environmental influences on ENS development (e.g., through large-scale drug screens), and to investigate the role the gut microbiota play in ENS development and disease. With its unique advantages as a model organism, Zebrafish will continue to contribute to a better understanding of ENS development, function, and disease. Developmental Dynamics 247:268-278, 2018. © 2017 Wiley Periodicals, Inc.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    目前开发干细胞疗法作为神经肠疾病的新治疗方法的努力受到无法获得用于研究人肠中细胞移植的模型系统的限制。我们建议,当皮下或腹内移植到小鼠中时,异种移植模型支持胎儿人类肠道中的肠神经系统(ENS)发育。
    将胎儿人小肠和大肠移植到小肠系膜和免疫缺陷小鼠的皮下组织中长达4个月。使用免疫组织化学研究了肠道细胞结构和ENS发育。
    在腹部和皮下移植中,肠道发育正常,形成成熟的上皮和间充质层。ENS在两个含有肠神经元和神经胶质的神经节丛中形成图案,包括胆碱能和硝能神经元亚型。肠壁中存在Cajal的c-Kit免疫反应性间质细胞。
    腹部异种移植物代表了支持胎儿人类肠的生长和发育的新型模型。这种体内方法将是研究ENS成熟的有用方法,神经肠道疾病的病理生理学,以及用于治疗肠神经病变的神经元干细胞的长期存活和功能分化。
    Current efforts to develop stem cell therapy as a novel treatment for neurointestinal diseases are limited by the unavailability of a model system to study cell transplantation in the human intestine. We propose that xenograft models support enteric nervous system (ENS) development in the fetal human intestine when transplanted into mice subcutaneously or intra-abdominally.
    Fetal human small and large intestine were grafted onto the small intestinal mesentery and into the subcutaneous tissue of immunodeficient mice for up to 4 months. Intestinal cytoarchitecture and ENS development were studied using immunohistochemistry.
    In both abdominal and subcutaneous grafts, the intestine developed normally with formation of mature epithelial and mesenchymal layers. The ENS was patterned in two ganglionated plexuses containing enteric neurons and glia, including cholinergic and nitrergic neuronal subtypes. c-Kit-immunoreactive interstitial cells of Cajal were present in the gut wall.
    Abdominal xenografts represent a novel model that supports the growth and development of fetal human intestine. This in vivo approach will be a useful method to study maturation of the ENS, the pathophysiology of neurointestinal diseases, and the long-term survival and functional differentiation of neuronal stem cells for the treatment of enteric neuropathies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号