microbiome modulation

  • 文章类型: Journal Article
    肠道微生物群对人类健康的重要性已日益得到认可。从这个角度来看,微生物组调节,有针对性地改变微生物组成,获得了兴趣。噬菌体溶素,噬菌体编码的肽聚糖降解酶,是目前正在临床开发中用于治疗细菌感染的有前途的新型抗生素。由于它们的高特异性,溶素被认为是微生物组友好的。这篇综述探讨了使用溶素作为微生物组调节剂的机遇和挑战。首先,内溶素的高特异性,可以使用蛋白质工程或靶向递送方法进一步调节,正在讨论。接下来,考虑了评估溶素的微生物组友好性的障碍和可能的解决方案。最后,讨论了溶素向肠道的递送,包括可能的递送方法,如基于颗粒和益生菌的载体。绘制发展溶素作为微生物组调节剂的障碍,并确定克服这些障碍的可能方法可以帮助它们的发展。这样,这些创新抗菌剂的应用可以扩大,从而充分利用他们的特点。
    The importance of the microbiota in the intestinal tract for human health has been increasingly recognized. In this perspective, microbiome modulation, a targeted alteration of the microbial composition, has gained interest. Phage lysins, peptidoglycan-degrading enzymes encoded by bacteriophages, are a promising new class of antibiotics currently under clinical development for treating bacterial infections. Due to their high specificity, lysins are considered microbiome-friendly. This review explores the opportunities and challenges of using lysins as microbiome modulators. First, the high specificity of endolysins, which can be further modulated using protein engineering or targeted delivery methods, is discussed. Next, obstacles and possible solutions to assess the microbiome-friendliness of lysins are considered. Finally, lysin delivery to the intestinal tract is discussed, including possible delivery methods such as particle-based and probiotic vehicles. Mapping the hurdles to developing lysins as microbiome modulators and identifying possible ways to overcome these hurdles can help in their development. In this way, the application of these innovative antimicrobial agents can be expanded, thereby taking full advantage of their characteristics.
    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
    目的:益生菌在炎症性肠病(IBD)中显示出希望,然而,知识差距仍然存在。我们对评估益生菌对克罗恩病(CD)和溃疡性结肠炎(UC)影响的随机对照试验(RCT)进行了系统综述和最新的Meta分析。
    方法:MEDLINE,WebofScience,并在2023年9月之前搜索了Cochrane中央控制试验登记册。主要结果是临床缓解和复发;次要结果包括内镜反应和缓解。和不良事件。我们使用R中的随机效应模型计算比值比(OR)。使用AMSTAR-2评估系统评价的质量;使用Cochrane协作工具评估试验偏倚风险。使用等级框架对证据确定性进行评级。
    结果:在2613个结果中,67项研究(22项系统评价和45项RCT)符合资格标准。在更新的荟萃分析中,UC和CD临床缓解的OR分别为2.00(95%CI1.28-3.11)和1.61(95%CI0.21-12.50),分别。亚组分析显示,5-ASA和益生菌联合应用可能有利于诱导轻度至中度UC缓解(OR2.35,95%CI1.29-4.28)。益生菌降低了复发性囊炎的复发几率(OR0.03,95%CI0.00-0.25),并倾向于降低非活动性UC的临床复发(OR0.65,95%CI0.42-1.01)。对于CD没有鉴定出针对复发的保护作用。多应变制剂在实现缓解和预防UC复发方面表现优异。益生菌的使用与更好的内镜结果无关。不良事件与对照相似。然而,证据的总体确定性较低.
    结论:益生菌,特别是多应变配方,对于UC患者以及复发性囊炎的临床缓解和预防复发似乎是有效的。尽管如此,对CD没有发现显著影响.还强调了益生菌的有利安全性。
    OBJECTIVE: Probiotics show promise in inflammatory bowel disease (IBD), yet knowledge gaps persist. We performed an overview of systematic reviews and an updated metanalysis of randomized controlled trials (RCT) assessing the effect of probiotics on Crohn\'s disease (CD) and ulcerative colitis (UC).
    METHODS: MEDLINE, Web of Science, and the Cochrane Central Register of Controlled Trials were searched up to September 2023. Primary outcomes were clinical remission and recurrence; secondary outcomes included endoscopic response and remission, and adverse events. We calculated odds ratios (OR) using a random-effects model in R. The quality of systematic reviews was assessed using the AMSTAR-2; the trials\' risk of bias was evaluated using the Cochrane Collaboration tool. Evidence certainty was rated using the GRADE framework.
    RESULTS: Out of 2613 results, 67 studies (22 systematic reviews and 45 RCTs) met the eligibility criteria. In the updated meta-analysis, the OR for clinical remission in UC and CD was 2.00 (95% CI 1.28-3.11) and 1.61 (95% CI 0.21-12.50), respectively. The subgroup analysis suggested that combining 5-ASA and probiotics may be beneficial for inducing remission in mild-to-moderate UC (OR 2.35, 95% CI 1.29-4.28). Probiotics decreased the odds of recurrence in relapsing pouchitis (OR 0.03, 95% CI 0.00-0.25) and trended toward reducing clinical recurrence in inactive UC (OR 0.65, 95% CI 0.42-1.01). No protective effect against recurrence was identified for CD. Multi-strain formulations appear superior in achieving remission and preventing recurrence in UC. The use of probiotics was not associated with better endoscopic outcomes. Adverse events were similar to control. However, the overall certainty of evidence was low.
    CONCLUSIONS: Probiotics, particularly multi-strain formulations, appear efficacious for the induction of clinical remission and the prevention of relapse in UC patients as well as for relapsing pouchitis. Notwithstanding, no significant effect was identified for CD. The favorable safety profile of probiotics was also highlighted.
    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
    该研究的目的是评估五种常用甜味剂(葡萄糖,菊粉,异麦芽酮糖,塔格糖,海藻糖)含有口腔微生物组上的漱口水。
    单中心,双盲,平行随机临床试验与健康,18-55岁的志愿者(N=65),他每天用10%的一种甜味剂溶液冲洗三次,持续两周。通过V4高变区(IlluminaMiSeq)的16SRNA基因扩增子测序分析了牙龈上牙菌斑和舌背涂层的微生物组成。作为次要结果,测量牙菌斑红色荧光和唾液pH。
    两组的牙菌斑菌群发生了显着变化:菊粉(F=2.0239,p=0.0006PERMANOVA,Aitchison距离)和异麦芽酮糖(F=0.67,p=0.0305)。对于舌头微生物群,异麦芽酮糖(F=0.8382,p=0.0452)和海藻糖(F=1.0119,p=0.0098)均有显著变化。在牌匾上,菊粉组有13种显著变化,而舌苔,海藻糖组改变了三个物种(ALDEx2,p<0.1)。次要结果没有观察到显著变化。
    对口腔微生物群的影响是甜味剂依赖性的,对斑块微生物群的影响最显著。菊粉在所测试的甜味剂中表现出最强的微生物调节潜力。需要进一步的全面临床研究。
    UNASSIGNED: The aim of the study was to evaluate the modulating effects of five commonly used sweetener (glucose, inulin, isomaltulose, tagatose, trehalose) containing mouth rinses on the oral microbiome.
    UNASSIGNED: A single-centre, double-blind, parallel randomized clinical trial was performed with healthy, 18-55-year-old volunteers (N = 65), who rinsed thrice-daily for two weeks with a 10% solution of one of the allocated sweeteners. Microbiota composition of supragingival dental plaque and the tongue dorsum coating was analysed by 16S RNA gene amplicon sequencing of the V4 hypervariable region (Illumina MiSeq). As secondary outcomes, dental plaque red fluorescence and salivary pH were measured.
    UNASSIGNED: Dental plaque microbiota changed significantly for two groups: inulin (F = 2.0239, p = 0.0006 PERMANOVA, Aitchison distance) and isomaltulose (F = 0.67, p = 0.0305). For the tongue microbiota, significant changes were observed for isomaltulose (F = 0.8382, p = 0.0452) and trehalose (F = 1.0119, p = 0.0098). In plaque, 13 species changed significantly for the inulin group, while for tongue coating, three species changed for the trehalose group (ALDEx2, p < 0.1). No significant changes were observed for the secondary outcomes.
    UNASSIGNED: The effects on the oral microbiota were sweetener dependant with the most pronounced effect on plaque microbiota. Inulin exhibited the strongest microbial modulating potential of the sweeteners tested. Further full-scale clinical studies are required.
    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
    已知肠道细菌产生细菌素以抑制其他细菌的生长。因此,细菌素作为潜在的微生物组编辑工具已经引起了越来越多的关注。在这项研究中,我们检查了75种II类细菌素对48种代表性肠道微生物群的抑制谱。细菌素在大肠杆菌中异源表达,并在体外进行评估。离体和体内。体外测定显示22种细菌素抑制至少一种物种,并显示出对某些病症和疾病相关物种的选择性抑制模式。选择三种细菌素用于对小鼠粪便的离体评估。基于培养的粪便的16SrRNA测序,我们表明两种细菌素:Actifencin(#13)和BacteroidotocinA(#22)选择性地抑制乳杆菌和拟杆菌的生长,分别。益生菌:大肠杆菌Nissle1917被工程化以在小鼠中表达这两种细菌素。然而,在体内无法观察到在体外和离体实验中发现的选择性抑制模式。我们的研究描述了异源高通量细菌素表达和筛选的方法,并阐明了II类细菌素对肠道微生物群的抑制模式。
    Gut bacteria are known to produce bacteriocins to inhibit the growth of other bacteria. Consequently, bacteriocins have attracted increased attention as potential microbiome-editing tools. In this study we examine the inhibitory spectrum of 75 class II bacteriocins against 48 representative gut microbiota species. The bacteriocins were heterologously expressed in Escherichia coli and evaluated in vitro, ex vivo and in vivo. In vitro assays revealed 22 bacteriocins to inhibit at least one species and showed selective inhibition patterns against species implicated in certain disorders and diseases. Three bacteriocins were selected for ex vivo assessment on mouse feces. Based on 16S rRNA sequencing of the cultivated feces we showed that the two bacteriocins: Actifencin (#13) and Bacteroidetocin A (#22) selectively inhibited the growth of Lactobacillus and Bacteroides, respectively. The probiotic: E. coli Nissle 1917 was engineered to express these two bacteriocins in mice. However, the selective inhibitory patterns found in the in vitro and ex vivo experiments could not be observed in vivo. Our study describes a methodology for heterologous high throughput bacteriocin expression and screening and elucidates the inhibitory patterns of class II bacteriocins on the gut microbiota.
    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
    在炎症性肠病(IBD)治疗的进展中,现有的治疗方法存在局限性;它们不能完全治愈IBD,并可能引发不良副作用.因此,新疗法和多方面治疗策略的探索为患者提供了更广泛的选择.在IBD的框架内,肠道菌群通过多种机制在疾病发作中起着关键作用。噬菌体,作为天然微生物调节剂,通过准确识别和消除特定病原体来证明其显著的特异性,因此持有治疗的希望。虽然临床试验肯定了噬菌体疗法的安全性,其功效在储存和运输过程中容易受到外部影响,这可能会影响其在微生物群中的感染性和调节作用。提高噬菌体的稳定性和精确的剂量控制-确保储存和运输的稳健性,一致的剂量,和有针对性的交付到感染部位-是至关重要的。这篇综述深入地探讨了IBD治疗的最新进展及其固有的挑战,关注微生物群和噬菌体之间的相互作用。它强调了噬菌体作为IBD治疗中微生物组调节剂的潜力,提供对噬菌体包封和靶向递送机制研究的详细见解。特别注意各种载体系统的功能,特别是关于它们的保护特性和结肠特异性递送的能力。本综述旨在为噬菌体作为微生物组调节剂在IBD治疗中的应用提供理论基础。为增强肠道微生物群的调节铺平了道路。
    In the advancement of Inflammatory Bowel Disease (IBD) treatment, existing therapeutic methods exhibit limitations; they do not offer a complete cure for IBD and can trigger adverse side effects. Consequently, the exploration of novel therapies and multifaceted treatment strategies provides patients with a broader range of options. Within the framework of IBD, gut microbiota plays a pivotal role in disease onset through diverse mechanisms. Bacteriophages, as natural microbial regulators, demonstrate remarkable specificity by accurately identifying and eliminating specific pathogens, thus holding therapeutic promise. Although clinical trials have affirmed the safety of phage therapy, its efficacy is prone to external influences during storage and transport, which may affect its infectivity and regulatory roles within the microbiota. Improving the stability and precise dosage control of bacteriophages-ensuring robustness in storage and transport, consistent dosing, and targeted delivery to infection sites-is crucial. This review thoroughly explores the latest developments in IBD treatment and its inherent challenges, focusing on the interaction between the microbiota and bacteriophages. It highlights bacteriophages\' potential as microbiome modulators in IBD treatment, offering detailed insights into research on bacteriophage encapsulation and targeted delivery mechanisms. Particular attention is paid to the functionality of various carrier systems, especially regarding their protective properties and ability for colon-specific delivery. This review aims to provide a theoretical foundation for using bacteriophages as microbiome modulators in IBD treatment, paving the way for enhanced regulation of the intestinal microbiota.
    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
    保持宿主健康和体内平衡在很大程度上取决于人类肠道微生物组,生活在胃肠道中的各种细菌种群不断变化。本文旨在探索肠道微生物组的多方面功能,并阐明研究草药对肠道微生物组的组成和功能的影响的不断发展的研究领域。通过全面的概述,我们的目标是提供洞察草药和肠道微生物组之间的复杂关系,促进更好地了解它们对人类健康的潜在影响。肠道菌群由数万亿的微生物组成,主要是细菌,还有病毒,真菌,和古细菌。它作为一个复杂的生态系统,以各种方式与宿主交互。它有助于营养代谢,调节免疫系统,提供对病原体的保护,并影响宿主生理学。此外,它与一系列健康结果有关,包括消化,代谢健康,甚至精神健康。最近的研究揭示了草药调节肠道微生物组的潜力。草药,来源于植物,常用于传统医学系统,含有各种各样的植物化学物质,可以直接或间接影响肠道微生物组成。这些植物化学物质可以充当益生元,促进有益菌的生长,或具有抗菌特性,针对有害病原体。一些研究已经证明了特定草药对肠道微生物组的影响。例如,草药提取物已被证明可以增强有益细菌的丰度,如双歧杆菌和乳酸菌,同时减少潜在的有害微生物。此外,草药对某些致病菌表现出有希望的抗菌作用。草药对肠道微生物组的调节具有潜在的治疗意义。研究表明,可以利用草药干预来缓解胃肠道疾病,支持免疫功能,甚至影响代谢健康。然而,重要的是要注意,由于遗传因素,个体对草药治疗的反应可能会有所不同,饮食,和基线微生物组组成。总之,肠道微生物群是维持人类健康的关键角色,草药对其调制是一个新兴的研究领域。了解草药化合物与肠道微生物群之间的复杂相互作用将为个性化医疗保健的创新方法以及旨在促进肠道健康和整体健康的草药疗法的开发铺平道路。
    Preserving host health and homeostasis is largely dependent on the human gut microbiome, a varied and ever-changing population of bacteria living in the gastrointestinal tract. This article aims to explore the multifaceted functions of the gut microbiome and shed light on the evolving field of research investigating the impact of herbal medicines on both the composition and functionality of the gut microbiome. Through a comprehensive overview, we aim to provide insights into the intricate relationship between herbal remedies and the gut microbiome, fostering a better understanding of their potential implications for human health.The gut microbiota is composed of trillions of microorganisms, predominantly bacteria, but also viruses, fungi, and archaea. It functions as a complex ecosystem that interacts with the host in various ways. It aids in nutrient metabolism, modulates the immune system, provides protection against pathogens, and influences host physiology. Moreover, it has been linked to a range of health outcomes, including digestion, metabolic health, and even mental well-being. Recent research has shed light on the potential of herbal medicines to modulate the gut microbiome. Herbal medicines, derived from plants and often used in traditional medicine systems, contain a diverse array of phytochemicals, which can directly or indirectly impact gut microbial composition. These phytochemicals can either act as prebiotics, promoting the growth of beneficial bacteria, or possess antimicrobial properties, targeting harmful pathogens. Several studies have demonstrated the effects of specific herbal medicines on the gut microbiome. For example, extracts from herbs have been shown to enhance the abundance of beneficial bacteria, such as Bifidobacterium and Lactobacillus, while reducing potentially harmful microbes. Moreover, herbal medicines have exhibited promising antimicrobial effects against certain pathogenic bacteria. The modulation of the gut microbiome by herbal medicines has potential therapeutic implications. Research suggests herbal interventions could be harnessed to alleviate gastrointestinal disorders, support immune function, and even impact metabolic health. However, it is important to note that individual responses to herbal treatments can vary due to genetics, diet, and baseline microbiome composition. In conclusion, the gut microbiome is a critical player in maintaining human health, and its modulation by herbal medicines is a burgeoning area of research. Understanding the complex interactions between herbal compounds and gut microbiota will pave the way for innovative approaches to personalized healthcare and the development of herbal-based therapeutics aimed at promoting gut health and overall well-being.
    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
    囊胚病是人类和动物中最常见的胃肠道原生生物。尽管囊胚病的临床意义尚不清楚,越来越多的生物体被视为肠道微生物组的共生成员。然而,它对微生物组的影响仍在争论中。目前尚不清楚囊胚是否直接促进健康的肠道和微生物组,或者它是否更有可能在健康的肠道环境中定植并持续存在。在健康的人中,囊胚病通常与增加的细菌多样性和肠道微生物组的显着差异有关。根据现有知识,无法确定肠道微生物组的差异是否是囊胚细胞定植的原因或结果.尽管这种真核生物在肠道微生物组中的作用可能在某些方面仍然未知,并且其影响各不相同,可能是由于亚型和亚型内变异和免疫调节,需要更多的研究来更详细地描述这些机制。这篇综述涵盖了囊胚在肠道微生物组和免疫调节中的作用的最新发现,它对自身免疫性疾病中微生物组的影响,胚泡是否像细菌一样在肠-脑轴中发挥作用,以及它与益生菌的关系。
    Blastocystis is the most common gastrointestinal protist found in humans and animals. Although the clinical significance of Blastocystis remains unclear, the organism is increasingly being viewed as a commensal member of the gut microbiome. However, its impact on the microbiome is still being debated. It is unclear whether Blastocystis promotes a healthy gut and microbiome directly or whether it is more likely to colonize and persist in a healthy gut environment. In healthy people, Blastocystis is frequently associated with increased bacterial diversity and significant differences in the gut microbiome. Based on current knowledge, it is not possible to determine whether differences in the gut microbiome are the cause or result of Blastocystis colonization. Although it is possible that some aspects of this eukaryote\'s role in the intestinal microbiome remain unknown and that its effects vary, possibly due to subtype and intra-subtype variations and immune modulation, more research is needed to characterize these mechanisms in greater detail. This review covers recent findings on the effects of Blastocystis in the gut microbiome and immune modulation, its impact on the microbiome in autoimmune diseases, whether Blastocystis has a role like bacteria in the gut-brain axis, and its relationship with probiotics.
    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
    早期瘤胃微生物群的调节有望促进小牛生长,健康,和反刍动物的长期生产。然而,瘤胃微生物群调节对后肠微生物群建立的影响一直受到关注。在这项研究中,在12个月的同卵双生小牛中检查粪便微生物群的发育。治疗组(T组)在断奶前接受成年母牛新鲜瘤胃液接种,而对照组没有(C组)。评估了接种物对小牛肠道健康的影响,并将其作为微生物接种途径进入后肠。早期瘤胃调节对年龄相关的粪便微生物群发育没有影响。粪便细菌群落随着饮食变化而逐渐演变,并分为断奶前和断奶后群落。细菌丰富度随着年龄的增长而增加,并在第9个月时趋于稳定,而断奶后样品中的样品间差异减少。第4个月后,粪便样本中的古细菌负荷增加,而两组的古细菌丰富度均增加并稳定在第9个月。在断奶前,样本间相似性较高,从第4个月开始差异增加。第4个月在粪便中检测到厌氧真菌,丰富度在第7个月达到峰值。在第6个月之前,真菌群落组成与成熟群落明显不同。初乳时,小牛瘤胃,和供体接种物被评估为后肠定植的种子来源,小牛自身的瘤胃被确定为粪便细菌和真菌的主要播种源。初乳是粪便中检测到的几种细菌的来源,但是这些在断奶之前是暂时重要的。供体接种物对肠道健康的影响有限,因为T组和C组之间的腹泻率相似。总之,早期微生物群的调节显示出反刍动物发育的潜力。然而,对于适应后肠环境的细菌,更有针对性的方法可能是有效调节后肠的必要。这项研究有助于我们理解肠道微生物群与小腿健康和生长之间的复杂关系。
    Early-life modulation of rumen microbiota holds promise for enhancing calf growth, health, and long-term production in ruminants. However, limited attention has been given to the impact of rumen microbiota modulation on the establishment of hindgut microbiota. In this study, fecal microbiota development was examined in identical twin calves for 12 months. The treatment group (T-group) received adult cow fresh rumen liquid inoculum during the pre-weaning period, while the control group did not (C-group). The effects of inoculum were assessed on calf gut health and as microbial seeding route into the hindgut. The early rumen modulation had no effect on age-related fecal microbiota development. The fecal bacterial community evolved gradually following dietary changes and categorized into pre-weaning and post-weaning communities. Bacterial richness increased with age and stabilized at month 9, while between-sample variation reduced in post-weaning samples. Archaeal load in fecal samples increased after month 4, while archaeal richness increased and stabilized in both groups by month 9. Between-sample similarity was higher during the pre-weaning period, with increased dissimilarity from month 4 onward. Anaerobic fungi were detected in feces at month 4, with richness peaking at month 7. Before month 6, fungal community composition distinctly differed from mature communities. When colostrum, calf rumen, and donor inoculum were evaluated as seeding sources for hindgut colonization, the calf\'s own rumen was identified as the primary seeding source for fecal bacteria and fungi. Colostrum was a source for several bacteria detected in feces, but these were of temporary importance until weaning. The donor inoculum had limited impact on gut health as diarrhea rates were similar between the T-group and C-group. In conclusion, early-life microbiota modulation shows potential in ruminant development. However, a more targeted approach with bacteria adapted to the hindgut environment may be necessary to modulate hindgut effectively. This research contributes to our understanding of the complex relationship between gut microbiota and calf health and growth.
    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
    肠道微生物组的精确工程有望成为与该微生物群落破坏相关的疾病的有效治疗方法。将活的生物治疗产品(LBP)植入可预测的,可控的方式是这种方法持续成功的关键,对于大多数正在开发的LBP来说仍然是一个挑战。我们最近证明了长双歧杆菌亚种的高水平植入。婴儿(B.婴儿)在成人中,当与特定的益生元共同给药时,人乳寡糖(HMO)。这里,我们提出了一种细胞动力学-药效学方法,类似于基于小分子和生物药物的药代动力学-药效学分析,为了确定HMO如何控制扩张,丰度,和婴儿芽孢杆菌在基于人类微生物群的模型中的代谢输出。我们的数据表明,HMO剂量控制了微生物组中婴儿芽孢杆菌的稳态丰度,婴儿芽孢杆菌与HMO一起影响肠道代谢物水平,依赖HMO的方式。我们还发现HMO为婴儿芽孢杆菌在5-log范围的细菌接种物中的扩张创造了特权生态位。这些结果表明,使用这种共生方法,婴儿芽孢杆菌的水平和微生物群落代谢输出都得到了显着控制。并为精确设计所需的微生物和代谢物以治疗一系列疾病铺平道路。
    Precision engineering of the gut microbiome holds promise as an effective therapeutic approach for diseases associated with a disruption in this microbial community. Engrafting a live biotherapeutic product (LBP) in a predictable, controllable manner is key to the consistent success of this approach and has remained a challenge for most LBPs under development. We recently demonstrated high-level engraftment of Bifidobacterium longum subsp. infantis (B. infantis) in adults when co-dosed with a specific prebiotic, human milk oligosaccharides (HMO). Here, we present a cellular kinetic-pharmacodynamic approach, analogous to pharmacokinetic-pharmacodynamic-based analyses of small molecule- and biologic-based drugs, to establish how HMO controls expansion, abundance, and metabolic output of B. infantis in a human microbiota-based model in gnotobiotic mice. Our data demonstrate that the HMO dose controls steady-state abundance of B. infantis in the microbiome, and that B. infantis together with HMO impacts gut metabolite levels in a targeted, HMO-dependent manner. We also found that HMO creates a privileged niche for B. infantis expansion across a 5-log range of bacterial inocula. These results demonstrate remarkable control of both B. infantis levels and the microbiome community metabolic outputs using this synbiotic approach, and pave the way for precision engineering of desirable microbes and metabolites to treat a range of 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
    目的:近年来,肠道菌群-脑通讯信号已经成为一种新的干预靶点,有可能改善一些与中枢神经系统相关的疾病。因此,有能力产生神经递质的益生菌,例如,已经成为治疗与神经递质失衡相关的疾病的有吸引力的替代品。在这里,我们进一步深入研究了产生γ-氨基丁酸(GABA)的双歧杆菌菌株的作用,以前证明有助于降低血清谷氨酸水平,小鼠模型中的肠道微生物组组成和代谢活性。我们的结果表明,产生GABA的菌株给药导致肠道微生物群调节的特定模式,与在接受非产GABA菌株的动物中观察到的不同。这开辟了新的途径来描绘IPLA60004施用有助于降低血清谷氨酸水平的具体机制,并确定这种作用是否可以在与高谷氨酸血清浓度相关的疾病的患者中发挥健康益处。
    OBJECTIVE: The gut microbiome-brain communication signaling has emerged in recent years as a novel target for intervention with the potential to ameliorate some conditions associated with the central nervous system. Hence, probiotics with capacity to produce neurotransmitters, for instance, have come up as appealing alternatives to treat disorders associated with disbalanced neurotransmitters. Herein, we further deep into the effects of administering a gamma-aminobutyric acid (GABA)-producing Bifidobacterium strain, previously demonstrated to contribute to reduce serum glutamate levels, in the gut microbiome composition and metabolic activity in a mouse model. Our results demonstrate that the GABA-producing strain administration results in a specific pattern of gut microbiota modulation, different from the one observed in animals receiving non-GABA-producing strains. This opens new avenues to delineate the specific mechanisms by which IPLA60004 administration contributes to reducing serum glutamate levels and to ascertain whether this effect could exert health benefits in patients of diseases associated with high-glutamate serum concentrations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号