Bacteroidaceae

拟杆菌科
  • 文章类型: Journal Article
    背景和目的:代谢功能障碍相关脂肪性肝炎(MASH)相关肝细胞癌(HCC)的发病率在全球范围内呈上升趋势。伴随着肥胖和代谢综合征的流行。根据关于肝癌和牙周炎的潜在关联的初步报告,本研究旨在分析MASH相关HCC(MASH-HCC)中牙周细菌的参与以及口腔和肠道细菌菌群。材料和方法:41例MASH患者和19例MASH-HCC患者参与了研究,完成调查问卷,正在进行牙周检查,提供唾液样本,漱口水,粪便,和外周血。通过16S核糖体RNA测序分析口腔和粪便微生物组谱。采用贝叶斯网络分析法分析各因素之间的因果关系,包括MASH-HCC,考试,和细菌。结果:与MASH组相比,MASH-HCC组的肠道微生物区系中梭杆菌属的占有率明显更高(p=0.002)。然而,Butyricicocus(p=0.022)和Roseburia(p<0.05)的占有率显着降低。贝叶斯网络分析显示,不存在影响HCC的牙周致病菌和肠道细菌。然而,HCC直接影响牙周细菌牙龈卟啉单胞菌,连翘坦菌,具核梭杆菌,和唾液中中中膜普雷沃氏菌,以及乳杆菌属,罗斯布里亚,梭杆菌,普雷沃氏菌,梭菌属,Ruminococus,锥虫,和肠道中的SMB53。此外,口腔中的牙龈卟啉单胞菌直接影响肠道中的乳杆菌属和链球菌属。结论:MASH-HCC直接影响牙周致病菌和肠道细菌,牙龈卟啉单胞菌可能影响与胃肠道癌症相关的肠道细菌。
    Background and Objectives: The incidence of metabolic dysfunction-associated steatohepatitis (MASH)-related hepatocellular carcinoma (HCC) is increasing worldwide, alongside the epidemic of obesity and metabolic syndrome. Based on preliminary reports regarding the potential association of HCC and periodontitis, this study aimed to analyze the involvement of periodontal bacteria as well as the oral and intestinal bacterial flora in MASH-related HCC (MASH-HCC). Materials and Methods: Forty-one patients with MASH and nineteen with MASH-HCC participated in the study, completing survey questionnaires, undergoing periodontal examinations, and providing samples of saliva, mouth-rinsed water, feces, and peripheral blood. The oral and fecal microbiome profiles were analyzed by 16S ribosomal RNA sequencing. Bayesian network analysis was used to analyze the causation between various factors, including MASH-HCC, examinations, and bacteria. Results: The genus Fusobacterium had a significantly higher occupancy rate (p = 0.002) in the intestinal microflora of the MASH-HCC group compared to the MASH group. However, Butyricicoccus (p = 0.022) and Roseburia (p < 0.05) had significantly lower occupancy rates. The Bayesian network analysis revealed the absence of periodontal pathogenic bacteria and enteric bacteria affecting HCC. However, HCC directly affected the periodontal bacterial species Porphyromonas gingivalis, Tannerella forsythia, Fusobacterium nucleatum, and Prevotella intermedia in the saliva, as well as the genera Lactobacillus, Roseburia, Fusobacterium, Prevotella, Clostridium, Ruminococcus, Trabulsiella, and SMB53 in the intestine. Furthermore, P. gingivalis in the oral cavity directly affected the genera Lactobacillus and Streptococcus in the intestine. Conclusions: MASH-HCC directly affects periodontal pathogenic and intestinal bacteria, and P. gingivalis may affect the intestinal bacteria associated with gastrointestinal cancer.
    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
    Graves病(GD)是甲状腺功能亢进的最常见原因,其发病机制仍未完全阐明。许多研究表明肠道微生物群与甲状腺疾病的发展有关。本研究采用孟德尔随机化分析来调查GD患者的肠道菌群特征。旨在为Graves病的病因和治疗提供新的见解。
    采用双样本孟德尔随机化(MR)分析来评估Graves病与肠道微生物群组成之间的因果关系。肠道菌群数据来自国际财团MiBioGen,而Graves\'疾病数据是从FINNGEN获得的。选择合格的单核苷酸多态性(SNP)作为工具变量。多种分析方法,包括逆方差加权(IVW),MR-Egger回归,加权中位数,加权模式,和MR-RAPS,被利用。敏感性分析采用MR-Egger截距试验进行,Cochran的Q测试,和留一法分析作为质量控制措施。
    在欧洲人群中进行的孟德尔随机研究显示,与拟杆菌科相关的格雷夫斯病的风险降低(赔率(OR)[95%置信区间(CI)]:0.89[0.89〜0.90],调整后的P值:<0.001),拟杆菌(OR:[95%CI]:0.555[0.437~0.706],调整后的P值:<0.001),和Veillonella(OR[95%CI]:0.632[0.492~0.811],调整后的P值:0.016)。没有明显的异质性证据,或检测到水平多效性。此外,初步MR分析确定了13种细菌,包括肠杆菌组和XIII家族AD3011组,表现出与格雷夫斯病发作显著相关,暗示潜在的因果效应。
    肠道微生物群与Graves病之间存在因果关系。拟杆菌科,拟杆菌,和Veillonella作为对抗Graves病发展的保护因素出现。前瞻性补充益生菌可能为将来治疗Graves病提供一种新的辅助治疗途径。
    UNASSIGNED: Graves\' disease (GD) is the most common cause of hyperthyroidism, and its pathogenesis remains incompletely elucidated. Numerous studies have implicated the gut microbiota in the development of thyroid disorders. This study employs Mendelian randomization analysis to investigate the characteristics of gut microbiota in GD patients, aiming to offer novel insights into the etiology and treatment of Graves\' disease.
    UNASSIGNED: Two-sample Mendelian randomization (MR) analysis was employed to assess the causal relationship between Graves\' disease and the gut microbiota composition. Gut microbiota data were sourced from the international consortium MiBioGen, while Graves\' disease data were obtained from FINNGEN. Eligible single nucleotide polymorphisms (SNPs) were selected as instrumental variables. Multiple analysis methods, including inverse variance-weighted (IVW), MR-Egger regression, weighted median, weighted mode, and MR-RAPS, were utilized. Sensitivity analyses were conducted employing MR-Egger intercept test, Cochran\'s Q test, and leave-one-out analysis as quality control measures.
    UNASSIGNED: The Mendelian randomization study conducted in a European population revealed a decreased risk of Graves\' disease associated with Bacteroidaceae (Odds ratio (OR) [95% confidence interval (CI)]: 0.89 [0.89 ~ 0.90], adjusted P value: <0.001), Bacteroides (OR: [95% CI]: 0.555 [0.437 ~ 0.706], adjusted P value: <0.001), and Veillonella (OR [95% CI]: 0.632 [0.492 ~ 0.811], adjusted P value: 0.016). No significant evidence of heterogeneity, or horizontal pleiotropy was detected. Furthermore, the preliminary MR analysis identified 13 bacterial species including Eubacterium brachy group and Family XIII AD3011 group, exhibiting significant associations with Graves\' disease onset, suggesting potential causal effects.
    UNASSIGNED: A causal relationship exists between gut microbiota and Graves\' disease. Bacteroidaceae, Bacteroides, and Veillonella emerge as protective factors against Graves\' disease development. Prospective probiotic supplementation may offer a novel avenue for adjunctive treatment in the management of Graves\' disease in the future.
    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
    溃疡性结肠炎(UC)是一种影响结肠和直肠的慢性炎症性肠道疾病,其发病机理归因于遗传背景,环境因素,和肠道微生物。本研究旨在通过层次分析法探讨肠型在UC中的作用。使用机器学习确定差异细菌,并进行物种共现网络(SCN)分析。收集UC患者的粪便细菌数据,使用QIIME2生物信息学管道进行16SrRNA宏基因组分析。肠型聚类在家庭层面进行,在每种肠型中对UC和健康对照(HC)进行深度神经网络(DNN)分类模型训练。来自11个16SrRNA肠道微生物组数据集的结果揭示了三种肠型:拟杆菌科(ET-B),落叶松科(ET-L),和梭菌科(ET-C)。Ruminococus(R.gnavus)的丰度在ET-B和ET-C的UC受试者中明显高于ET-C。对于ET-B和ET-C受试者,R.gnavus也与UCSCN中的梭菌呈正相关,在ET-C受试者中具有较高的相关性。相反,Odoribacter(O.)内脏和拟杆菌属(B.)均匀性与色氨酸代谢和AMP激活蛋白激酶(AMPK)信号通路呈正相关,而R.gnavus呈负相关。与梭菌的体外共培养实验(C.)艰难梭菌表明,ET-B受试者的粪便微生物群比ET-L受试者具有更高的艰难梭菌丰度。总之,ET-B肠型使个体易患UC,以R.gnavus为潜在危险因素,O.splanchnicus和B.uniphis为保护性细菌,那些患有UC的人可能最终成为ET-C。
    Ulcerative colitis (UC) is a chronic inflammatory intestinal disease affecting the colon and rectum, with its pathogenesis attributed to genetic background, environmental factors, and gut microbes. This study aimed to investigate the role of enterotypes in UC by conducting a hierarchical analysis, determining differential bacteria using machine learning, and performing Species Co-occurrence Network (SCN) analysis. Fecal bacterial data were collected from UC patients, and a 16S rRNA metagenomic analysis was performed using the QIIME2 bioinformatics pipeline. Enterotype clustering was conducted at the family level, and deep neural network (DNN) classification models were trained for UC and healthy controls (HC) in each enterotype. Results from eleven 16S rRNA gut microbiome datasets revealed three enterotypes: Bacteroidaceae (ET-B), Lachnospiraceae (ET-L), and Clostridiaceae (ET-C). Ruminococcus (R. gnavus) abundance was significantly higher in UC subjects with ET-B and ET-C than in those with ET-L. R. gnavus also showed a positive correlation with Clostridia in UC SCN for ET-B and ET-C subjects, with a higher correlation in ET-C subjects. Conversely, Odoribacter (O.) splanchnicus and Bacteroides (B.) uniformis exhibited a positive correlation with tryptophan metabolism and AMP-activated protein kinase (AMPK) signaling pathways, while R. gnavus showed a negative correlation. In vitro co-culture experiments with Clostridium (C.) difficile demonstrated that fecal microbiota from ET-B subjects had a higher abundance of C. difficile than ET-L subjects. In conclusion, the ET-B enterotype predisposes individuals to UC, with R. gnavus as a potential risk factor and O. splanchnicus and B. uniformis as protective bacteria, and those with UC may have ultimately become ET-C.
    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
    Introduction.脆弱拟杆菌是革兰氏阴性厌氧菌,是人类胃肠道微生物群的成员,经常被发现为肠道外机会性病原体。B.fragilis包含两个不同的组-I和II-以基因的存在/不存在为特征[cepA和ccrA(cfiA),分别]通过丝氨酸或金属β-内酰胺酶的产生赋予对β-内酰胺抗生素的抗性。尚未对公开可用的B.fragilis序列数据进行大规模分析,物种的抗性组仍然不明确。假设/差距声明。已提议将I和IIB.fragilis重新分类为两个不同的物种,但还需要其他证据。目标。为了研究GenBank脆弱芽孢杆菌基因组的基因组多样性,并确定公开可用的脆弱芽孢杆菌基因组中I和II划分菌株的患病率,并产生进一步的证据来证明脆弱芽孢杆菌I和II划分菌株代表不同的基因组物种。方法论。在基因库中列为脆弱芽孢杆菌的高质量(n=377)基因组包括在全基因组和功能分析中。基因组数据还使用综合抗生素抗性数据库进行抗性组分析。结果。平均核苷酸同一性和系统发育分析表明,脆弱芽孢杆菌I和II表示不同的物种:脆弱芽孢杆菌(n=275个基因组)和脆弱芽孢杆菌A(n=102个基因组;基因组分类学数据库名称),分别。对脆弱芽孢杆菌和脆弱芽孢杆菌A的pangenome的探索揭示了这两个物种在核心和辅助基因水平上的分离。结论。研究结果表明,脆弱芽孢杆菌A,以前称为IIB.fragilis,是一个单独的物种,与脆弱的B.fragilissensustricto不同。脆弱芽孢杆菌pangenome分析支持以前的基因组,系统发育和抗性组筛选分析共同加强了I和II是两个独立的物种。此外,已证实脆弱芽孢杆菌I和II的辅助基因的差异主要与碳水化合物代谢有关,并表明除抗菌素耐药性以外的差异也可用于区分这两个物种。
    Introduction. Bacteroides fragilis is a Gram-negative anaerobe that is a member of the human gastrointestinal microbiota and is frequently found as an extra-intestinal opportunistic pathogen. B. fragilis comprises two distinct groups - divisions I and II - characterized by the presence/absence of genes [cepA and ccrA (cfiA), respectively] that confer resistance to β-lactam antibiotics by either serine or metallo-β-lactamase production. No large-scale analyses of publicly available B. fragilis sequence data have been undertaken, and the resistome of the species remains poorly defined.Hypothesis/Gap Statement. Reclassification of divisions I and II B. fragilis as two distinct species has been proposed but additional evidence is required.Aims. To investigate the genomic diversity of GenBank B. fragilis genomes and establish the prevalence of division I and II strains among publicly available B. fragilis genomes, and to generate further evidence to demonstrate that B. fragilis division I and II strains represent distinct genomospecies.Methodology. High-quality (n=377) genomes listed as B. fragilis in GenBank were included in pangenome and functional analyses. Genome data were also subject to resistome profiling using The Comprehensive Antibiotic Resistance Database.Results. Average nucleotide identity and phylogenetic analyses showed B. fragilis divisions I and II represent distinct species: B. fragilis sensu stricto (n=275 genomes) and B. fragilis A (n=102 genomes; Genome Taxonomy Database designation), respectively. Exploration of the pangenome of B. fragilis sensu stricto and B. fragilis A revealed separation of the two species at the core and accessory gene levels.Conclusion. The findings indicate that B. fragilis A, previously referred to as division II B. fragilis, is an individual species and distinct from B. fragilis sensu stricto. The B. fragilis pangenome analysis supported previous genomic, phylogenetic and resistome screening analyses collectively reinforcing that divisions I and II are two separate species. In addition, it was confirmed that differences in the accessory genes of B. fragilis divisions I and II are primarily associated with carbohydrate metabolism and suggest that differences other than antimicrobial resistance could also be used to distinguish between these two species.
    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
    已知人类肠道微生物群在某些疾病如肾脏并发症期间直接和间接地改变。拟杆菌被认为是人类肠道菌群中主要和最丰富的门,已被分类为肠型1。本研究旨在评估拟杆菌属的丰度。在终末期肾病(ESRD)和慢性肾病(CKD)患者的粪便菌群中,并将其与健康个体粪便菌群中的拟杆菌组成进行比较。
    从20名CKD/ESRD患者和20名没有任何肾脏并发症的健康个体收集新鲜粪便样品。通过QIAamp粪便迷你试剂盒从粪便样品中提取纯微生物DNA。MiSeq系统用于通过下一代测序方法分析肠道成分。
    从患者和健康组的40个粪便样本中分离并鉴定了651个细菌菌株。生物信息学分析定义了18种不同类型的拟杆菌属物种,占所有菌株的2.76%。统计学分析显示各研究组间差异无统计学意义(P>0.05)。在健康和患者组中,包括B.dorei,B.制服,和B.Ovatus分配了最丰富的自己。最低的丰度与B.eggerthii有关,CKD/ESRD患者和A.furcosa,B.barnesiae,B.coprocola的丰度在健康人中最低。
    这项研究表明,尽管以前有所有证据表明拟杆菌在肠道微生物群中的作用,它在健康人和CKD/ESRD患者之间没有不同的分布。
    UNASSIGNED: Human intestine microbiota are known to be directly and indirectly altered during some diseases such as kidney complications. Bacteroides is considered as the main and the most abundant phylum among human gut microbiota, which has been classified as enterotype 1. This study aimed to assess the abundance of Bacteroides spp. in fecal flora of end-stage renal disease (ESRD) and chronic kidney disease (CKD) patients and compare it with the Bacteroides composition among fecal flora of healthy individual.
    UNASSIGNED: Fresh fecal samples were collected from 20 CKD/ESRD patients and 20 healthy individual without any kidney complications. The pure microbial DNA was extracted by QIAamp Stool Mini Kit from stool samples. MiSeq system was used to analyze the intestinal composition by next generation sequencing method.
    UNASSIGNED: A number of 651 bacterial strains were isolated and identified from 40 fecal samples of both patients and healthy groups. Bioinformatics analysis defined 18 different types of Bacteroides species which included 2.76% of all strains. Statistical analysis showed no significant difference between study groups (P>0.05). In both healthy and patient groups three species including B. dorei, B. uniformis, and B. ovatus have allocated the most abundance to themselves. The lowest abundance was related to B. eggerthii, A. furcosa and B. barnesiae among CKD/ESRD patients and A. furcosa, B. barnesiae, and B. coprocola had the lowest abundance among healthy people.
    UNASSIGNED: This study indicates despite all previous evidence of Bacteroides role in gut microbiota, it had no different distribution between healthy persons and CKD/ESRD patients.
    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
    肠道微生物群与苯诱导的造血毒性的发展有关。通过益生菌补充剂调节肠道稳态已被认为是预防不良健康影响的有效策略。然而,益生菌在苯诱导的造血毒性中的作用和机制尚不清楚。暴露45天后,苯引起小鼠骨髓造血毒性。此外,我们发现苯改变了小鼠的肠道屏障,导致细菌科的丰度增加和全身性炎症的激活。有趣的是,Fe2+积累,脂质过氧化,在苯暴露小鼠的肠组织中观察到铁凋亡蛋白的差异表达。粪便微生物移植后,来自苯暴露小鼠的粪便微生物导致受体小鼠肠道铁凋亡的发展。特别是,口服益生菌可显着逆转细菌科和肠道铁性凋亡的升高,最终改善苯引起的造血损伤。我们进一步使用苯代谢物1,4-BQ治疗人正常结肠上皮细胞(NCM460),并干预铁凋亡抑制剂liproxstatin-1(Lip-1)以验证肠道铁凋亡与炎症之间的关系。结果表明,1,4-BQ处理导致细胞内ROS水平升高,铁凋亡蛋白和炎症因子IL-5和IL-13的异常表达。然而,使用Lip-1显著抑制氧化应激,铁性凋亡,和NCM460细胞的炎症。该结果表明,铁凋亡可能通过介导Th2型全身性炎症参与苯诱导的造血毒性。总的来说,这些发现揭示了拟杆菌科-肠道铁死亡-炎症在苯诱导的造血毒性中的作用,并强调益生菌可能是预防不良血液学结局的有前景的策略.
    The intestinal microbiota is associated with the development of benzene-induced hematopoietic toxicity. Modulation of intestinal homeostasis by probiotic supplementation has been considered an effective strategy to prevent adverse health effects. However, the role and mechanism of probiotics in benzene-induced hematopoietic toxicity are unclear. After 45 days of exposure, benzene caused bone marrow hematopoietic toxicity in mice. Furthermore, we found that benzene altered the intestinal barrier in mice, leading to an increase in the abundance of Bacteroidaceae and the activation of systemic inflammation. Interestingly, Fe2+ accumulation, lipid peroxidation, and differential expression of ferroptosis proteins were observed in the intestinal tissues of benzene-exposed mice. After fecal microbiota transplantation, stool microbes from benzene-exposed mice led to the development of intestinal ferroptosis in recipient mice. In particular, oral probiotics significantly reversed elevated Bacteroidaceae and intestinal ferroptosis, ultimately improving benzene-induced hematopoietic damage. We further used the benzene metabolite 1,4-BQ to treat human normal colonic epithelial cells (NCM460) and intervened with the ferroptosis inhibitor liproxstatin-1 (Lip-1) to validate the relationship between intestinal ferroptosis and inflammation. The results showed that 1,4-BQ treatment resulted in increased intracellular ROS levels and abnormal expression of ferroptosis proteins and the inflammatory factors IL-5 and IL-13. However, the use of Lip-1 significantly inhibited oxidative stress, ferroptosis, and inflammation in NCM460 cells. This result suggested that ferroptosis might be involved in benzene-induced hematopoietic toxicity by mediating Th2-type systemic inflammation. Overall, these findings revealed a role for Bacteroidaceae-intestinal ferroptosis-inflammation in benzene-induced hematopoietic toxicity and highlighted that probiotics could be a promising strategy to prevent adverse hematologic outcomes.
    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
    人类肠道菌群的特点是人际差异大,这不仅与健康和疾病相关,而且决定了营养干预的结果。随着人们对开发靶向肠道微生物群调节剂的兴趣日益浓厚,将胡萝卜衍生的鼠李糖半乳糖醛酸I(cRG-I)的选择性与证明较低(菊粉,IN)和高选择性(黄原胶,XA),在1.5g/d的人类等效剂量(HED)。体外SIFR®技术的高通量,验证以生成临床发现的预测性见解,允许包括24名成年人。在体外肠道微生物群建模的背景下,这种前所未有的大量样品允许覆盖临床相关的肠道微生物群组成和功能的人际差异。一个关键发现是cRG-I补充剂(已经在0.3g/d的HED下)降低了人际组成差异,这是由于在成年人中始终存在的分类群的选择性刺激,包括与Dorei/vulgatus和长双歧杆菌相关的OTU(疑似梯形物种),拟杆菌,青春双歧杆菌和产生丁酸的分类群,例如Blautiasp。,hallii厌氧丁酸,和prausnitzii粪杆菌.相比之下,IN和XA治疗都增加了人际组成差异。对于IN,这是因为其特异性低。对于XA,正是XA发酵的极高选择性导致了15个反应者和9个无反应者之间的巨大差异,由存在/不存在高度特异性的XA发酵类群引起。虽然所有测试化合物都显着增强乙酸,丙酸盐,丁酸盐,和天然气生产,cRG-I导致明显更高的乙酸(+40%),丙酸盐(+22%),然而,与国内相比,天然气产量较低(-44%)。因此,cRG-I可以产生总体上更稳健的有益效果,同时也有更好的耐受性。此外,由于其对微生物组成和代谢产物产生的显着均质化作用,与特异性较低或过度特异性的底物相比,cRG-I可导致更可预测的结果。
    The human gut microbiota is characterized by large interpersonal differences, which are not only linked to health and disease but also determine the outcome of nutritional interventions. In line with the growing interest for developing targeted gut microbiota modulators, the selectivity of a carrot-derived rhamnogalacturonan I (cRG-I) was compared to substrates with demonstrated low (inulin, IN) and high selectivity (xanthan, XA), at a human equivalent dose (HED) of 1.5 g/d. The high throughput of the ex vivo SIFR® technology, validated to generate predictive insights for clinical findings, enabled the inclusion of 24 human adults. Such an unprecedented high number of samples in the context of in vitro gut microbiota modelling allowed a coverage of clinically relevant interpersonal differences in gut microbiota composition and function. A key finding was that cRG-I supplementation (already at an HED of 0.3 g/d) lowered interpersonal compositional differences due to the selective stimulation of taxa that were consistently present among human adults, including OTUs related to Bacteroides dorei/vulgatus and Bifidobacterium longum (suspected keystone species), Bacteroides thetaiotaomicron, Bifidobacterium adolescentis and butyrate-producing taxa such as Blautia sp., Anaerobutyricum hallii, and Faecalibacterium prausnitzii. In contrast, both IN and XA treatments increased interpersonal compositional differences. For IN, this followed from its low specificity. For XA, it was rather the extremely high selectivity of XA fermentation that caused large differences between 15 responders and 9 nonresponders, caused by the presence/absence of highly specific XA-fermenting taxa. While all test compounds significantly enhanced acetate, propionate, butyrate, and gas production, cRG-I resulted in a significantly higher acetate (+40%), propionate (+22%), yet a lower gas production (-44%) compared to IN. cRG-I could thus result in overall more robust beneficial effects, while also being better tolerated. Moreover, owing to its remarkable homogenization effect on microbial composition and metabolite production, cRG-I could lead to more predictable outcomes compared to substrates that are less specific or overly specific.
    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
    拟杆菌和Phocaeicola,拟杆菌科的成员,是最早定植于人类婴儿肠道的微生物之一。虽然已知这些微生物可以从母亲传播给孩子,我们对共享和潜在传播的特定菌株的理解是有限的.在这项研究中,我们旨在调查母亲及其婴儿中的拟杆菌和Phocaeicola的共有菌株。我们分析了从PreventADALL研究中招募的18孕周孕妇的粪便样本,以及早期婴儿的后代样本,包括出生后10分钟内采集的皮肤拭子样本,第一个可用的粪便样本(胎粪),和3个月大的粪便样本。我们筛选了464个胎粪样本的拟杆菌科,随后选择144对母子进行纵向分析,基于拟杆菌科的存在,纵向样本可用性,和交付模式。我们的结果表明,拟杆菌科成员主要在阴道分娩婴儿的样本中检测到。我们发现了高患病率的Phocaeicolavulgatus,Phocaeicoladorei,拟杆菌,和拟杆菌在母亲和阴道出生的婴儿身上。然而,在应变水平,我们观察到只有两种菌株的高流行率:B.caccae菌株和P.vulgatus菌株。值得注意的是,B.caccae菌株被鉴定为母婴共享菌株的新成分,在全世界公开的宏基因组中也观察到其高患病率。我们的研究结果表明,分娩方式可能在塑造婴儿肠道微生物群的早期定植中起作用。特别是拟杆菌科成员的定植。重要性我们的研究提供了证据,表明婴儿出生后10分钟内皮肤上存在拟杆菌科菌株,在胎粪样本中,在3个月大的粪便样本中,阴道分娩的婴儿与母亲分享。使用应变分辨率分析,我们确定了两个菌株,属于拟杆菌属和Phocaeicolavulgatus,在母亲和婴儿之间共享。有趣的是,B.caccae菌株在世界范围内表现出很高的患病率,而P.vulgatus菌株不太常见。我们的发现还表明,阴道分娩与拟杆菌科成员的早期定植有关,而剖宫产分娩与延迟定植有关。鉴于这些微生物可能影响结肠环境,我们的结果表明,在菌株水平上了解细菌与宿主的关系可能对婴儿的健康和以后的发育有影响.
    Bacteroides and Phocaeicola, members of the family Bacteroidaceae, are among the first microbes to colonize the human infant gut. While it is known that these microbes can be transmitted from mother to child, our understanding of the specific strains that are shared and potentially transmitted is limited. In this study, we aimed to investigate the shared strains of Bacteroides and Phocaeicola in mothers and their infants. We analyzed fecal samples from pregnant woman recruited at 18 weeks of gestation from the PreventADALL study, as well as offspring samples from early infancy, including skin swab samples taken within 10 min after birth, the first available fecal sample (meconium), and fecal samples at 3 months of age. We screened 464 meconium samples for Bacteroidaceae, with subsequent selection of 144 mother-child pairs for longitudinal analysis, based on the presence of Bacteroidaceae, longitudinal sample availability, and delivery mode. Our results showed that Bacteroidaceae members were mainly detected in samples from vaginally delivered infants. We identified high prevalences of Phocaeicola vulgatus, Phocaeicola dorei, Bacteroides caccae, and Bacteroides thetaiotaomicron in mothers and vaginally born infants. However, at the strain level, we observed high prevalences of only two strains: a B. caccae strain and a P. vulgatus strain. Notably, the B. caccae strain was identified as a novel component of mother-child shared strains, and its high prevalence was also observed in publicly available metagenomes worldwide. Our findings suggest that mode of delivery may play a role in shaping the early colonization of the infant gut microbiota, in particular the colonization of Bacteroidaceae members. IMPORTANCE Our study provides evidence that Bacteroidaceae strains present on infants\' skin within 10 min after birth, in meconium samples, and in fecal samples at 3 months of age in vaginally delivered infants are shared with their mothers. Using strain resolution analyses, we identified two strains, belonging to Bacteroides caccae and Phocaeicola vulgatus, as shared between mothers and their infants. Interestingly, the B. caccae strain showed a high prevalence worldwide, while the P. vulgatus strain was less common. Our findings also showed that vaginal delivery was associated with early colonization of Bacteroidaceae members, whereas cesarean section delivery was associated with delayed colonization. Given the potential for these microbes to influence the colonic environment, our results suggest that understanding the bacterial-host relationship at the strain level may have implications for infant health and development later in life.
    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
    由于强调病原体铁敏感性的体外研究,因此人们对土著人类肠道共生微生物群对铁的反应知之甚少。在一项对健康人补充铁的研究中,我们发现,一些参与者的微生物群逐渐变化与细菌铁内化相关.为了确定由于分类单元特异性铁敏感性的直接影响,我们使用参与者粪便样本得出不同的体外群落。这些社区的铁补充导致了小的成分变化,模仿体内的那些,而缺铁显著抑制了生长,不可逆转,多样性的累积减少和优势物种的替换。在无菌培养中,单个物种对铁剥夺的敏感性通常预示着社区中铁的依赖性。最后,外源血红素作为无机铁的来源,以防止某些物种的消耗。我们的结果强调了体内和体外研究在理解环境因素如何影响肠道微生物方面的互补性。
    Responses of the indigenous human gut commensal microbiota to iron are poorly understood because of an emphasis on in vitro studies of pathogen iron sensitivity. In a study of iron supplementation in healthy humans, we identified gradual microbiota shifts in some participants correlated with bacterial iron internalization. To identify direct effects due to taxon-specific iron sensitivity, we used participant stool samples to derive diverse in vitro communities. Iron supplementation of these communities caused small compositional shifts, mimicking those in vivo, whereas iron deprivation dramatically inhibited growth with irreversible, cumulative reduction in diversity and replacement of dominant species. Sensitivity of individual species to iron deprivation in axenic culture generally predicted iron dependency in a community. Finally, exogenous heme acted as a source of inorganic iron to prevent depletion of some species. Our results highlight the complementarity of in vivo and in vitro studies in understanding how environmental factors affect gut microbiotas.
    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
    纵向研究发现,人类肠道微生物群随着时间的推移是稳定的,一些主要的细菌谱系甚至菌株持续多年。最近使用宏基因组数据将其扩展到肠道噬菌体。这里,我们专注于培养人类肠道的主要拟杆菌,拟杆菌和Phocaeicola菌株,以及来自两个健康捐献者的噬菌体.确认了拟杆菌和Phocaeicola物种和菌株的持久性。我们分离出28个基因不同的噬菌体,分成7个不同的簇,其中两个是新的。此外,来自几个群体的噬菌体,尽管在基因上相当同质,具有感染从几个采样时间点和不同供体分离的属于不同物种的菌株的能力。我们认为感染几种宿主物种的能力,它们的营养生态位不同,可能会促进优势肠道噬菌体组的长期持续存在。
    The longitudinal studies have found that the human gut microbiota is stable over time with some major bacterial lineages or even strains persisting for years. This was recently extended to gut bacteriophages using the metagenomic data. Here, we focused on cultivation of the major Bacteroidetes of human gut, the Bacteroides and Phocaeicola strains, and their bacteriophages from two healthy donors. The persistence of Bacteroides and Phocaeicola species and strains was confirmed. We isolated 28 genetically different phages grouped into seven distinct clusters, two of these were new. Moreover, the bacteriophages from several groups, although being genetically quite homogeneous, had the ability to infect the strains belonging to different species isolated from several sampling time-points and different donors. We propose that the ability to infect several host species, which differ in their nutritional niches, may promote long-term persistence of dominant gut bacteriophage groups.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号