Germ-free mice

无菌小鼠
  • 文章类型: Journal Article
    肠上皮的更新严格依赖于细胞增殖之间的精细调节,分化,和凋亡。虽然小鼠肠道微生物群已经被证明可以改变一些上皮细胞动力学参数,人们对人体肠道微生物群的作用知之甚少。这里,我们调查了与人类菌群相关的C3H/HeN无菌小鼠的肠细胞增殖速率(HFA,n=8),在无菌(n=15)和全毒小鼠(n=16)中。牺牲前一小时,所有小鼠腹膜内接种5-溴脱氧尿苷(BrdU),和BrdU阳性细胞数/总细胞数(标记指数,LI),在空肠和结肠,通过免疫组织化学进行评估。还通过扫描电子显微镜(SEM)观察样品。此外,将HFA小鼠大肠中的微生物群组成与人类供体粪便样本的组成进行了比较。在HFA的小肠中没有发现LI的差异,Holoxenic,和无菌老鼠.相反,HFA小鼠大肠的LI显着高于无菌和全氧对应物(分别为p=0.017和p=0.048)。在全氧和HFA小鼠中,SEM分析揭示了不同类型的细菌与肠上皮紧密接触。最后,在优势种群方面,HFA小鼠的结肠微生物群组成与人类供体的组成广泛重叠,虽然双歧杆菌和乳杆菌消失了。尽管本研究分析的样本量很小,这些初步发现表明,人类肠道菌群可能促进结肠粘膜的高增殖率。鉴于不受控制的增殖在结直肠癌发生中的众所周知的作用,这些结果可能值得在更大的人群研究中进一步研究.
    Intestinal epithelium renewal strictly depends on fine regulation between cell proliferation, differentiation, and apoptosis. While murine intestinal microbiota has been shown to modify some epithelial cell kinetics parameters, less is known about the role of the human intestinal microbiota. Here, we investigated the rate of intestinal cell proliferation in C3H/HeN germ-free mice associated with human flora (HFA, n = 8), and in germ-free (n = 15) and holoxenic mice (n = 16). One hour before sacrifice, all mice were intraperitoneally inoculated with 5-bromodeoxyuridine (BrdU), and the number of BrdU-positive cells/total cells (labelling index, LI), both in the jejunum and the colon, was evaluated by immunohistochemistry. Samples were also observed by scanning electron microscopy (SEM). Moreover, the microbiota composition in the large bowel of the HFA mice was compared to that of of human donor\'s fecal sample. No differences in LI were found in the small bowels of the HFA, holoxenic, and germ-free mice. Conversely, the LI in the large bowel of the HFA mice was significantly higher than that in the germ-free and holoxenic counterparts (p = 0.017 and p = 0.048, respectively). In the holoxenic and HFA mice, the SEM analysis disclosed different types of bacteria in close contact with the intestinal epithelium. Finally, the colonic microbiota composition of the HFA mice widely overlapped with that of the human donor in terms of dominant populations, although Bifidobacteria and Lactobacilli disappeared. Despite the small sample size analyzed in this study, these preliminary findings suggest that human intestinal microbiota may promote a high proliferation rate of colonic mucosa. In light of the well-known role of uncontrolled proliferation in colorectal carcinogenesis, these results may deserve further investigation in a larger population study.
    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
    肠道微生物群负责人类健康的基本功能。肠道微生物群和其他器官之间通过神经的几个交流轴,内分泌,和免疫途径已经被描述,和肠道微生物群组成的扰动与许多新出现的疾病的发病和进展有关。这里,我们分析了周围神经,背根神经节(DRG),和具有以下肠道微生物群状态的新生和年轻成年小鼠的骨骼肌:a)无菌(GF),b)侏儒,选择性定植12种特定的肠道细菌菌株(Oligo-Mouse-Microbiota,OMM12),或c)天然复杂肠道微生物群(CGM)。体视学和形态学分析显示,肠道微生物群的缺乏会损害体细胞正中神经的发育,导致更小的直径和髓鞘过多的轴突,以及较小的无髓鞘纤维。因此,DRG和坐骨神经转录组学分析强调了一组差异表达的发育和髓鞘形成基因。有趣的是,神经调节蛋白1(NRG1)的III型同工型,已知是雪旺氏细胞髓鞘形成所必需的神经元信号,在年轻的成年GF小鼠中过表达,随后过度表达转录因子早期生长反应2(Egr2),一种由施万细胞在髓鞘形成开始时表达的基本基因。最后,GF状态导致组织学萎缩的骨骼肌,神经肌肉接头形成受损,和相关基因的表达失调。总之,我们首次证明了肠道微生物群对躯体周围神经系统的正常发育及其与骨骼肌的功能连接的调节作用,因此表明存在一种新的肠道微生物群-周围神经系统轴。\'
    Gut microbiota is responsible for essential functions in human health. Several communication axes between gut microbiota and other organs via neural, endocrine, and immune pathways have been described, and perturbation of gut microbiota composition has been implicated in the onset and progression of an emerging number of diseases. Here, we analyzed peripheral nerves, dorsal root ganglia (DRG), and skeletal muscles of neonatal and young adult mice with the following gut microbiota status: a) germ-free (GF), b) gnotobiotic, selectively colonized with 12 specific gut bacterial strains (Oligo-Mouse-Microbiota, OMM12), or c) natural complex gut microbiota (CGM). Stereological and morphometric analyses revealed that the absence of gut microbiota impairs the development of somatic median nerves, resulting in smaller diameter and hypermyelinated axons, as well as in smaller unmyelinated fibers. Accordingly, DRG and sciatic nerve transcriptomic analyses highlighted a panel of differentially expressed developmental and myelination genes. Interestingly, the type III isoform of Neuregulin1 (NRG1), known to be a neuronal signal essential for Schwann cell myelination, was overexpressed in young adult GF mice, with consequent overexpression of the transcription factor Early Growth Response 2 (Egr2), a fundamental gene expressed by Schwann cells at the onset of myelination. Finally, GF status resulted in histologically atrophic skeletal muscles, impaired formation of neuromuscular junctions, and deregulated expression of related genes. In conclusion, we demonstrate for the first time a gut microbiota regulatory impact on proper development of the somatic peripheral nervous system and its functional connection to skeletal muscles, thus suggesting the existence of a novel \'Gut Microbiota-Peripheral Nervous System-axis.\'
    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
    蓝氏贾第鞭毛虫是世界范围内腹泻病的重要原虫,低收入和中等收入国家儿童的发育迟缓和认知障碍,以及发达地区长期感染后综合征。G.lamblia位于近端小肠的管腔和上皮表面,但不是粘膜侵入性的。原生动物寄生虫具有遗传多样性,在菌株和组合之间具有显着的基因组差异。动物模型,尤其是鼠类模型,在确定宿主防御G.Lamblia的机制方面发挥了重要作用,但是小鼠不容易感染大多数人类致病菌株。抗生素预处理可以增加易感性,表明正常的微生物群在控制小鼠的蓝氏酵母菌感染中起作用,但对不同菌株易感性的更广泛影响尚不清楚。这里,我们已经使用侏儒小鼠来证明,对于遗传组合A和B的广泛的人类致病性菌株,可以实现强大的肠道感染。在滋养体攻击后,侏儒小鼠能够以与常规小鼠相似的动力学根除感染。无菌小鼠也可以通过粘膜途径用保护性抗原有效免疫,α1-giardin,以依赖于CD4T细胞的方式。这些结果表明,侏儒症小鼠模型对于研究贾第鞭毛虫病中的获得性宿主防御是强大的,因为小鼠广泛易受不同的G.lamblia菌株的影响,但在控制和根除这种内腔病原体所需的粘膜免疫方面没有明显的缺陷。
    Giardia lamblia is an important protozoan cause of diarrheal disease worldwide, delayed development and cognitive impairment in children in low- and middle-income countries, and protracted post-infectious syndromes in developed regions. G. lamblia resides in the lumen and at the epithelial surface of the proximal small intestine but is not mucosa invasive. The protozoan parasite is genetically diverse with significant genome differences across strains and assemblages. Animal models, particularly murine models, have been instrumental in defining mechanisms of host defense against G. lamblia, but mice cannot be readily infected with most human pathogenic strains. Antibiotic pretreatment can increase susceptibility, suggesting that the normal microbiota plays a role in controlling G. lamblia infection in mice, but the broader implications on susceptibility to diverse strains are not known. Here, we have used gnotobiotic mice to demonstrate that robust intestinal infection can be achieved for a broad set of human-pathogenic strains of the genetic assemblages A and B. Furthermore, gnotobiotic mice were able to eradicate infection with a similar kinetics to conventional mice after trophozoite challenge. Germ-free mice could also be effectively immunized by the mucosal route with a protective antigen, α1-giardin, in a manner dependent on CD4 T cells. These results indicate that the gnotobiotic mouse model is powerful for investigating acquired host defenses in giardiasis, as the mice are broadly susceptible to diverse G. lamblia strains yet display no apparent defects in mucosal immunity needed for controlling and eradicating this lumen-dwelling pathogen.
    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
    虽然肠道菌群在炎症性肠病(IBD)发病机制中的作用是无可争议的。验证IBD相关菌群失调的致病作用的尝试仅限于在遗传易感小鼠或化学诱导的结肠炎中促进该疾病的报道。我们旨在进一步测试宿主对来自克罗恩病患者的粪便微生物组移植(FMT)对无菌(xGF)小鼠粘膜稳态的反应。我们对健康患者和明确的克罗恩回肠结肠炎(CD_L3)患者的粪便微生物群进行了表征,并将其转移到无菌小鼠中,并通过16S谱分析分析了由此产生的微生物和粘膜稳态。鸟枪宏基因组学,组织学,免疫荧光(IF)和RNAseq分析。我们观察到与健康对照微生物群相比,CD_L3微生物群的植入显著减少。来自CD_L3患者的FMT没有导致回肠炎,但导致具有与CD一致的特征的结肠炎:结肠炎的中断模式,更近端结肠定位,孤立的淋巴滤泡扩大和/或三级淋巴器官新生,转录组模式与上皮重编程和近端结肠中Paneth细胞样特征的促进以及CD的免疫失调特征一致。所观察到的炎症反应与持续增加的gnavusruminococcus丰度有关,Erespelatoclostridiumramosum,Faecalimonas脐带,Blautiahominis,丁酸梭菌,和副营养梭菌和产毒艰难梭菌的意外生长,低于用于接种的社区的检测水平。我们的研究提供了第一个证据,表明CD患者的生态失调群落的转移可以导致xGF小鼠结肠中的自发性炎症变化,并鉴定了能够促进致病性和条件致病性细菌定植的特征微生物群落。
    Although the role of the intestinal microbiota in the pathogenesis of inflammatory bowel disease (IBD) is beyond debate, attempts to verify the causative role of IBD-associated dysbiosis have been limited to reports of promoting the disease in genetically susceptible mice or in chemically induced colitis. We aimed to further test the host response to fecal microbiome transplantation (FMT) from Crohn\'s disease patients on mucosal homeostasis in ex-germ-free (xGF) mice. We characterized and transferred fecal microbiota from healthy patients and patients with defined Crohn\'s ileocolitis (CD_L3) to germ-free mice and analyzed the resulting microbial and mucosal homeostasis by 16S profiling, shotgun metagenomics, histology, immunofluorescence (IF) and RNAseq analysis. We observed a markedly reduced engraftment of CD_L3 microbiome compared to healthy control microbiota. FMT from CD_L3 patients did not lead to ileitis but resulted in colitis with features consistent with CD: a discontinued pattern of colitis, more proximal colonic localization, enlarged isolated lymphoid follicles and/or tertiary lymphoid organ neogenesis, and a transcriptomic pattern consistent with epithelial reprograming and promotion of the Paneth cell-like signature in the proximal colon and immune dysregulation characteristic of CD. The observed inflammatory response was associated with persistently increased abundance of Ruminococcus gnavus, Erysipelatoclostridium ramosum, Faecalimonas umbilicate, Blautia hominis, Clostridium butyricum, and C. paraputrificum and unexpected growth of toxigenic C. difficile, which was below the detection level in the community used for inoculation. Our study provides the first evidence that the transfer of a dysbiotic community from CD patients can lead to spontaneous inflammatory changes in the colon of xGF mice and identifies a signature microbial community capable of promoting colonization of pathogenic and conditionally pathogenic bacteria.
    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
    嗜酸性粒细胞和微生物之间的联系越来越受到关注。我们先前表明,从传统奶牛场收集的粉尘中包含的无菌微生物产品的气道管理实际上消除了过敏原致敏的特异性无病原体(SPF)小鼠的支气管肺泡灌洗(BAL)嗜酸性粒细胞增多和其他主要哮喘表型。有趣的是,在给予无菌的BAL后,发现了对过敏原诱导的BAL嗜酸性粒细胞的抑制和对气道屏障完整性的促进,药理学级细菌裂解物,OM-85,对过敏原致敏的SPF小鼠的气道室。这里,我们询问气道递送微生物产物的内在特性是否足以抑制过敏性肺部炎症,或者这些作用是否由宿主微生物群的重编程介导.我们比较了无菌(GF)小鼠和与健康小鼠肠道微生物群相关并在SPF条件下维持多代的GF小鼠的后代(Ex-GF小鼠)。用OM-85鼻内治疗这些小鼠,并在主要关注BAL嗜酸性粒细胞增多的过敏性哮喘的卵清蛋白和链格孢菌模型中进行评估。变应原诱导的BAL嗜酸性粒细胞增多的水平在GF和常规Ex-GF小鼠中相当。OM-85细菌裂解物的气道给药足以抑制Ex-GF和GF小鼠中过敏原诱导的肺嗜酸性粒细胞增多,这表明在这些模型中,细菌产物的保护作用不需要宿主微生物群,局部气道暴露于微生物产物是有效的保护来源。对GF小鼠BAL嗜酸性粒细胞增多的OM-85依赖性抑制伴随着肺2型细胞因子和嗜酸性粒细胞吸引趋化因子的抑制,提示OM-85可能至少通过减少嗜酸性粒细胞肺募集来起作用.
    The nexus between eosinophils and microbes is attracting increasing attention. We previously showed that airway administration of sterile microbial products contained in dust collected from traditional dairy farms virtually abrogated bronchoalveolar lavage (BAL) eosinophilia and other cardinal asthma phenotypes in allergen-sensitized specific pathogen-free (SPF) mice. Interestingly, comparable inhibition of allergen-induced BAL eosinophilia and promotion of airway barrier integrity were found upon administration of a sterile, pharmacological-grade bacterial lysate, OM-85, to the airway compartment of allergen-sensitized SPF mice. Here, we asked whether intrinsic properties of airway-delivered microbial products were sufficient to inhibit allergic lung inflammation or whether these effects were mediated by reprogramming of the host microbiota. We compared germ-free (GF) mice and offspring of GF mice associated with healthy mouse gut microbiota and maintained under SPF conditions for multiple generations (Ex-GF mice). These mice were treated intranasally with OM-85 and evaluated in the ovalbumin and Alternaria models of allergic asthma focusing primarily on BAL eosinophilia. Levels of allergen-induced BAL eosinophilia were comparable in GF and conventionalized Ex-GF mice. Airway administration of the OM-85 bacterial lysate was sufficient to inhibit allergen-induced lung eosinophilia in both Ex-GF and GF mice, suggesting that host microbiota are not required for the protective effects of bacterial products in these models and local airway exposure to microbial products is an effective source of protection. OM-85-dependent inhibition of BAL eosinophilia in GF mice was accompanied by suppression of lung type 2 cytokines and eosinophil-attracting chemokines, suggesting that OM-85 may work at least by decreasing eosinophil lung recruitment.
    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
    通过比较特发性身材矮小(ISS)儿童与正常身高兄弟姐妹的肠道菌群(GM)在ISS发病机理中的作用。
    本病例对照研究,在施耐德儿童医学中心内分泌和糖尿病研究所进行4/2018-11/2020,涉及30对3-10岁健康的青春期前兄弟姐妹,每个人都包括一个拥有国际空间站的兄弟姐妹和一个具有正常身高的兄弟姐妹。来自两个兄弟姐妹的粪便分析的结果测量包括通过16SrRNA测序分析的GM组成,粪便代谢组学,并监测粪便移植后无菌(GF)小鼠的生长。
    对ISS儿童的粪便分析确定了更高的预测水平的编码嘧啶酶的基因,嘌呤,黄素,辅酶B,和硫胺素生物合成,较低水平的几种氨基酸,与正常身高的兄弟姐妹相比,欧洲古细菌门的患病率明显更高(p<0.001)。ISS甲氧西林杆菌水平较高的儿童,古细菌肠道群落中的优势物种,在身材上明显短于那些水平较低的人(p=0.022)。接受ISS儿童粪便移植的小鼠没有发育迟缓,可能是由于在粪便收集过程中暴露于氧气引起的甲氧杆菌的根除。
    我们的研究结果表明,GM的不同特征可能解释线性增长的变化。ISS组中显示的不同水平的Methanobrevibacter反映了ISS的多因素性质以及GM部分解释生长变化的潜在能力。特定微生物群的靶向可以提供个性化治疗,以改善ISS儿童的生长。
    To investigate the role of gut microbiota (GM) in pathogenesis of idiopathic short stature (ISS) by comparing GM of ISS children to their normal-height siblings.
    This case-control study, conducted at the Schneider Children\'s Medical Center\'s Institute for Endocrinology and Diabetes between 4/2018-11/2020, involved 30 pairs of healthy pre-pubertal siblings aged 3-10 years, each comprising one sibling with ISS and one with normal height. Outcome measures from fecal analysis of both siblings included GM composition analyzed by 16S rRNA sequencing, fecal metabolomics, and monitoring the growth of germ-free (GF) mice after fecal transplantation.
    Fecal analysis of ISS children identified higher predicted levels of genes encoding enzymes for pyrimidine, purine, flavin, coenzyme B, and thiamine biosynthesis, lower levels of several amino acids, and a significantly higher prevalence of the phylum Euryarchaeota compared to their normal-height siblings (p<0.001). ISS children with higher levels of Methanobrevibacter, the dominant species in the archaeal gut community, were significantly shorter in stature than those with lower levels (p=0.022). Mice receiving fecal transplants from ISS children did not experience stunted growth, probably due to the eradication of Methanobrevibacter caused by exposure to oxygen during fecal collection.
    Our findings suggest that different characteristics in the GM may explain variations in linear growth. The varying levels of Methanobrevibacter demonstrated within the ISS group reflect the multifactorial nature of ISS and the potential ability of the GM to partially explain growth variations. The targeting of specific microbiota could provide personalized therapies to improve growth in children with ISS.
    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
    家猪(Susscrofa)是用于肉类生产的主要陆生动物。肠道菌群显著影响宿主营养,新陈代谢,和豁免权。因此,对肠道微生物结构和功能的表征将提高我们对肠道微生物资源和宿主-微生物相互作用机制的理解。这里,我们使用宏基因组学和16SrRNA基因扩增子测序研究了7个猪品种的肠道微生物组。我们建立了一个扩展的肠道微生物参考目录,包括17020160个基因,并鉴定了4910个宏基因组组装的基因组(MAG)。我们还分析了肠道耐药性,以概述猪的抗微生物耐药性基因的概况。通过分析微生物的相对丰度,我们确定了三种核心优势的肠道微生物(顺丁菌,CopriPrevotella,和本研究中使用的猪中的镰刀杆菌属糖蛋白)。口服三种核心优势肠道微生物显着增加器官指数(包括心脏,脾,脾和胸腺),但减少了无菌(GF)小鼠的胃肠道长度。3种核心微生物显著增强肠上皮屏障功能,改变肠黏膜形态,从十二指肠和回肠隐窝深度的增加可以明显看出。此外,这三种核心微生物显著影响了几种代谢途径(如“类固醇激素生物合成”,“初级胆汁酸生物合成”,“苯丙氨酸,酪氨酸和色氨酸生物合成和“苯丙氨酸代谢”)在GF小鼠中。这些发现提供了猪肠道微生物组的全景视图,并深入了解了核心优势肠道微生物对宿主的功能贡献。
    Domestic pigs (Sus scrofa) are the leading terrestrial animals used for meat production. The gut microbiota significantly affect host nutrition, metabolism, and immunity. Hence, characterization of the gut microbial structure and function will improve our understanding of gut microbial resources and the mechanisms underlying host-microbe interactions. Here, we investigated the gut microbiomes of seven pig breeds using metagenomics and 16S rRNA gene amplicon sequencing. We established an expanded gut microbial reference catalog comprising 17 020 160 genes and identified 4910 metagenome-assembled genomes. We also analyzed the gut resistome to provide an overview of the profiles of the antimicrobial resistance genes in pigs. By analyzing the relative abundances of microbes, we identified three core-predominant gut microbes (Phascolarctobacterium succinatutens, Prevotella copri, and Oscillibacter valericigenes) in pigs used in this study. Oral administration of the three core-predominant gut microbes significantly increased the organ indexes (including the heart, spleen, and thymus), but decreased the gastrointestinal lengths in germ-free mice. The three core microbes significantly enhanced intestinal epithelial barrier function and altered the intestinal mucosal morphology, as was evident from the increase in crypt depths in the duodenum and ileum. Furthermore, the three core microbes significantly affected several metabolic pathways (such as \"steroid hormone biosynthesis,\" \"primary bile acid biosynthesis,\" \"phenylalanine, tyrosine and tryptophan biosynthesis,\" and \"phenylalanine metabolism\") in germ-free mice. These findings provide a panoramic view of the pig gut microbiome and insights into the functional contributions of the core-predominant gut microbes to the host.
    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
    硬化链球菌(S。anginosus)在胃癌(GC)患者的胃粘膜中富集。这里,我们显示,在小鼠的胃中定植了安吉氏杆菌并诱发了急性胃炎。血管炎链球菌感染自发诱导的进行性慢性胃炎,壁细胞萎缩,粘液化生,和常规小鼠的发育不良,这一发现在无菌小鼠中得到了证实。此外,在致癌物诱导的胃肿瘤发生和YTN16GC细胞同种异体移植中,安神链球菌加速GC进展。始终如一,安吉氏杆菌破坏了胃屏障功能,促进细胞增殖,抑制细胞凋亡。机械上,我们鉴定出了一种多孔链球菌表面蛋白,TMPC,与胃上皮细胞上的膜联蛋白A2(ANXA2)受体相互作用。TMPC与ANXA2介导的脊柱葡萄球菌的附着和定植以及诱导的丝裂原活化蛋白激酶(MAPK)活化的相互作用。ANXA2基因敲除取消了脊柱血吸虫对MAPK的诱导。因此,这项研究揭示了在TMPC-ANXA2-MAPK轴中通过与胃上皮细胞的直接相互作用作为促进胃癌发生的病原体。
    Streptococcus anginosus (S. anginosus) was enriched in the gastric mucosa of patients with gastric cancer (GC). Here, we show that S. anginosus colonized the mouse stomach and induced acute gastritis. S. anginosus infection spontaneously induced progressive chronic gastritis, parietal cell atrophy, mucinous metaplasia, and dysplasia in conventional mice, and the findings were confirmed in germ-free mice. In addition, S. anginosus accelerated GC progression in carcinogen-induced gastric tumorigenesis and YTN16 GC cell allografts. Consistently, S. anginosus disrupted gastric barrier function, promoted cell proliferation, and inhibited apoptosis. Mechanistically, we identified an S. anginosus surface protein, TMPC, that interacts with Annexin A2 (ANXA2) receptor on gastric epithelial cells. Interaction of TMPC with ANXA2 mediated attachment and colonization of S. anginosus and induced mitogen-activated protein kinase (MAPK) activation. ANXA2 knockout abrogated the induction of MAPK by S. anginosus. Thus, this study reveals S. anginosus as a pathogen that promotes gastric tumorigenesis via direct interactions with gastric epithelial cells in the TMPC-ANXA2-MAPK axis.
    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
    已在几个物种中报道了母系行为的母系传播。研究,主要是老鼠,已经提出了产后经验的重要性以及表观遗传机制在介导这些传播中的参与。这项研究旨在确定母系行为的母系传播是否发生在小鼠中以及是否涉及微生物群。我们首先观察到早期断奶(EW)雌性小鼠表现出较低水平的母性行为,特别是舔/梳理自己的小狗(LG),比正常断奶(NW)雌性小鼠。在第二代中也观察到了母亲行为特征的这种差异,即使所有老鼠都正常断奶。在随后的交叉培养实验中,LG的水平受到养育母亲的影响,而不是亲生母亲的影响。最后,我们将EW或NW小鼠的粪便微生物群移植到培养幼崽的无菌(GF)小鼠中。分析了幼崽生长后对自己后代表现出的母性行为,与来自NW小鼠的微生物群定植的GF小鼠相比,在来自EW小鼠的微生物群定植的GF小鼠中的LG水平较低。这些结果清楚地表明,在母亲的行为特征中,LG在小鼠中代际传播,表明微生物群的垂直传播参与了这一过程。这项研究证明了母亲行为特征代际传播的普遍性,并为微生物群的作用提供了新的见解。
    The matrilineal transmission of maternal behavior has been reported in several species. Studies, primarily on rats, have suggested the importance of postnatal experience and the involvement of epigenetic mechanisms in mediating these transmissions. This study aims to determine whether the matrilineal transmission of maternal behavior occurs in mice and whether the microbiota is involved. We first observed that early weaned (EW) female mice showed lower levels of maternal behavior, particularly licking/grooming (LG) of their own pups, than normally weaned (NW) female mice. This difference in maternal behavioral traits was also observed in the second generation, even though all mice were weaned normally. In the subsequent cross-fostering experiment, the levels of LG were influenced by the nurturing mother but not the biological mother. Finally, we transplanted the fecal microbiota from EW or NW mice into germ-free (GF) mice raising pups. The maternal behaviors that the pups exhibited toward their own offspring after growth were analyzed, and the levels of LG in GF mice colonized with microbiota from EW mice were lower than those in GF mice colonized with microbiota from NW mice. These results clearly indicate that, among maternal behavioral traits, LG is intergenerationally transmitted in mice and suggest that the vertical transmission of microbiota is involved in this process. This study demonstrates the universality of the intergenerational transmission of maternal behavioral traits and provides new insights into the role of 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
    目的:剧烈的饮食干预已被证明可以促进肠道菌群的快速和显著的组成变化,但是适度饮食变化的影响尚不清楚。这里,我们旨在阐明人类习惯性饮食范围内的适度饮食变化对肠道微生物群组成的影响.
    方法:我们进行了一项试点饮食干预,其中五名健康志愿者食用素食即食食品三天,以标准化饮食摄入量,然后改用肉类即食西餐和高糖饮料两天。我们从每天的粪便采样中进行16SrRNA测序,以评估由干预饮食引起的肠道微生物群变化。此外,我们使用自愿者的粪便样本对接受相同灭菌饮食的无菌小鼠进行定植,以研究在个体差异减少的情况下,适度饮食干预对肠道微生物群的影响.
    结果:在人为干预中,我们发现,无论饮食如何,个体之间和个体内部的粪便微生物群组成都不同.然而,当我们给研究参与者的粪便定植的小鼠喂食相同的饮食时,我们观察到重要的,通常是特定于捐赠者的,这种适度饮食干预后小鼠微生物群的变化。
    结论:人类习惯性饮食的适度变化有可能改变肠道微生物群。饲喂人源化小鼠人类饮食可能有助于我们理解个体人类肠道微生物对适度饮食变化的反应,并有助于改善个性化干预措施。
    Drastic diet interventions have been shown to promote rapid and significant compositional changes of the gut microbiota, but the impact of moderate diet variations is less clear. Here, we aimed to clarify the impact of moderate diet variations that remain within the spectrum of the habitual human diet on gut microbiota composition.
    We performed a pilot diet intervention where five healthy volunteers consumed a vegetarian ready-made meal for three days to standardize dietary intake before switching to a meat-based ready-made western-style meal and high sugar drink for two days. We performed 16S rRNA sequencing from daily fecal sampling to assess gut microbiota changes caused by the intervention diet. Furthermore, we used the volunteers\' fecal samples to colonize germ-free mice that were fed the same sterilized diets to study the effect of a moderate diet intervention on the gut microbiota in a setting of reduced interindividual variation.
    In the human intervention, we found that fecal microbiota composition varied between and within individuals regardless of diet. However, when we fed the same diets to mice colonized with the study participants\' feces, we observed significant, often donor-specific, changes in the mouse microbiota following this moderate diet intervention.
    Moderate variations in the habitual human diet have the potential to alter the gut microbiota. Feeding humanized mice human diets may facilitate our understanding of individual human gut microbiota responses to moderate dietary changes and help improve individualized interventions.
    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