Intestinal colonization

肠道定植
  • 文章类型: Systematic Review
    背景:多重耐药生物(MDRO)的肠道定植通常在重症监护病房(ICU)的患者中发生感染之前,虽然殖民的动力学还没有完全理解。我们对ICU研究进行了系统评价和荟萃分析,这些研究描述了MDRO肠道获取的累积发生率和发生率。
    方法:我们系统地搜索了PubMed,Embase,和WebofScience于2010年至2023年发表的研究报告,报道了ICU中MDRO的肠道获取。MDRO被定义为多重耐药的非假单胞菌革兰氏阴性菌(NP-GN),假单胞菌属。,和耐万古霉素肠球菌(VRE)。我们纳入了观察性研究,这些研究在ICU入院时(48小时内)以及随后的一个或多个时间点获得了肛周或直肠拭子。我们的主要结果是MDRO的肠道获得性发生率,定义为ICU入院后新检测到的任何MDRO(即,基线时不存在),适用于所有有风险的患者时间。这项研究在PROSPERO注册,CRD42023481569。
    结果:在最初确定的482项研究中,14项研究,37,305名患者符合纳入标准。ICU住院期间肠道获取MDRO的合并发生率为5%(范围:1-43%),合并发生率为12.2(95%CI8.1-18.6)/1000患者天。ICU入院后的中位时间为4至26天。NP-GN和假单胞菌属的结果相似。,没有足够的数据来评估VRE。在提供足够数据进行曲线拟合的六项研究中,肠道MDRO定植呈每天1.41%的准线性增加,在ICU住院30天后保持稳定(R2=0.50,p<0.01)。
    结论:获得肠道MDRO在ICU中很常见,并且随着30天随访在ICU中的天数增加。这些数据可能会指导未来的干预措施,以防止ICU中肠道获得MDRO。
    BACKGROUND: Gut colonization with multidrug-resistant organisms (MDRO) frequently precedes infection among patients in the intensive care unit (ICU), although the dynamics of colonization are not completely understood. We performed a systematic review and meta-analysis of ICU studies which described the cumulative incidence and rates of MDRO gut acquisition.
    METHODS: We systematically searched PubMed, Embase, and Web of Science for studies published from 2010 to 2023 reporting on gut acquisition of MDRO in the ICU. MDRO were defined as multidrug resistant non-Pseudomonas Gram-negative bacteria (NP-GN), Pseudomonas spp., and vancomycin-resistant Enterococcus (VRE). We included observational studies which obtained perianal or rectal swabs at ICU admission (within 48 h) and at one or more subsequent timepoints. Our primary outcome was the incidence rate of gut acquisition of MDRO, defined as any MDRO newly detected after ICU admission (i.e., not present at baseline) for all patient-time at risk. The study was registered with PROSPERO, CRD42023481569.
    RESULTS: Of 482 studies initially identified, 14 studies with 37,305 patients met criteria for inclusion. The pooled incidence of gut acquisition of MDRO during ICU hospitalization was 5% (range: 1-43%) with a pooled incidence rate of 12.2 (95% CI 8.1-18.6) per 1000 patient-days. Median time to acquisition ranged from 4 to 26 days after ICU admission. Results were similar for NP-GN and Pseudomonas spp., with insufficient data to assess VRE. Among six studies which provided sufficient data to perform curve fitting, there was a quasi-linear increase in gut MDRO colonization of 1.41% per day which was stable through 30 days of ICU hospitalization (R2 = 0.50, p < 0.01).
    CONCLUSIONS: Acquisition of gut MDRO was common in the ICU and increases with days spent in ICU through 30 days of follow-up. These data may guide future interventions seeking to prevent gut acquisition of MDRO in the ICU.
    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
    肠外大肠杆菌感染对公众健康的威胁越来越大,然而,目前的研究往往忽视了重要的因素,如感染的时间模式,系统发育和克隆背景,或宿主肠道大肠杆菌种群,尽管它们可能的意义。
    在这项研究中,我们分析了明尼阿波利斯退伍军人事务医疗保健系统(2012-2019)患者的7000种临床大肠杆菌分离株,和来自未感染退伍军人的粪便大肠杆菌。我们评估了系统发育组的分布,选定序列类型(ST)的成员资格,及其子集-包括大流行,电阻相关的ST131-H30R,和ST1193谱系-和菌株类型,如脉冲场凝胶电泳所定义。然后,我们分析了这些特征以及单个宿主中感染的时间模式。
    H30R谱系成为主要谱系,无论是整体还是氟喹诺酮耐药分离株,在氟喹诺酮耐药分离株中使用ST1193。反复发作很常见,发生在31%的受试者和41%的发作中,通常是多次和延迟/延长(每个受试者最多23次发作;索引后高达2655d)。值得注意的是,这些复发通常涉及受试者的指数应变(63%的复发),即使影响尿外部位。ST131,H30R,ST1193和氟喹诺酮耐药菌株通常比其他菌株引起更多的复发,尽管复发间隔相似。ST131菌株类型在研究期间发生了显著变化。在感染发作期间以外的时间,通常可以在宿主粪便中检测到引起感染的菌株;检测的可能性随监测强度和与感染的接近程度而变化。H30R和ST1193是粪便-临床克隆重叠的主要原因。
    这些发现为退伍军人中大肠杆菌感染的时间和克隆特征提供了新的见解,并支持开发抗定植干预措施的努力。
    UNASSIGNED: Extraintestinal Escherichia coli infections represent a growing public health threat, However, current studies often overlook important factors such as temporal patterns of infection, phylogenetic and clonal background, or the host gut E. coli population, despite their likely significance.
    UNASSIGNED: In this study, we analyzed >7000 clinical E. coli isolates from patients at the Minneapolis Veterans Affairs Health Care System (2012-2019), and concurrent fecal E. coli from uninfected veterans. We assessed phylogenetic group distribution, membership in selected sequence types (STs), and subsets thereof-including the pandemic, resistance-associated ST131-H30R, and ST1193 lineages-and strain type, as defined by pulsed-field gel electrophoresis. We then analyzed these features alongside the temporal patterns of infection in individual hosts.
    UNASSIGNED: The H30R lineage emerged as the leading lineage, both overall and among fluoroquinolone-resistant isolates, with ST1193 following among fluoroquinolone-resistant isolates. Recurrences were common, occurring in 31% of subjects and 41% of episodes, and often multiple and delayed/prolonged (up to 23 episodes per subject; up to 2655d post-index). Remarkably, these recurrences typically involved the subject\'s index strain (63% of recurrences), even when affecting extra-urinary sites. ST131, H30R, ST1193, and fluoroquinolone-resistant strains generally caused significantly more recurrences than did other strains, despite similar recurrence intervals. ST131 strain types shifted significantly over the study period. Infection-causing strains were commonly detectable in host feces at times other than during an infection episode; the likelihood of detection varied with surveillance intensity and proximity to the infection. H30R and ST1193 were prominent causes of fecal-clinical clonal overlap.
    UNASSIGNED: These findings provide novel insights into the temporal and clonal characteristics of E. coli infections in veterans and support efforts to develop anti-colonization interventions.
    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
    在水产养殖领域,加强动物健康和疾病预防正在逐步解决使用抗生素的替代品,包括疫苗和益生菌。本研究旨在评估重组嗜甲基芽孢杆菌的潜力,工程表达嗜水气单胞菌AH3的外膜通道蛋白TolC和绿色荧光蛋白GFP,作为口服疫苗。最初,将编码tolC和GFP的基因克隆到原核表达系统中,并产生抗TolC小鼠抗血清。随后,将tolC基因亚克隆到修饰的pMDGFP质粒中,将其转化为甲基营养芽孢杆菌WM-1用于蛋白质表达。然后通过共同饲喂将重组的甲基营养芽孢杆菌BmT施用于草鱼,并评估了其作为口服疫苗的功效。我们的发现证明了55kDaTolC和28kDaGFP蛋白的成功表达,制备了具有高特异性的多克隆抗体。BmT表现出GFP-tolC融合蛋白的稳定表达和优异的遗传稳定性。口服免疫后,血清特异性IgM水平和酸性磷酸酶(ACP)活性显着升高,碱性磷酸酶(AKP),超氧化物歧化酶(SOD),草鱼中的溶菌酶(LZM)。同时,免疫相关基因的显著上调,包括IFN-I,IL-10,IL-1β,TNF-α,还有IgT,在肠道中被注意到,头肾,和草鱼的脾脏。定植测试进一步显示,即使在7天的禁食期后,BmT仍在免疫鱼的肠道中持续存在。值得注意的是,口服BmT可提高嗜水菌感染后草鱼的存活率。这些结果表明,本研究中开发的口服BmT疫苗有望在水产养殖中应用。
    In the field of aquaculture, the enhancement of animal health and disease prevention is progressively being tackled using alternatives to antibiotics, including vaccines and probiotics. This study was designed to evaluate the potential of a recombinant Bacillus methylotrophicus, engineered to express the outer membrane channel protein TolC of Aeromonas hydrophila AH3 and the green fluorescent protein GFP, as an oral vaccine. Initially, the genes encoding tolC and GFP were cloned into a prokaryotic expression system, and anti-TolC mouse antiserum was generated. Subsequently, the tolC gene was subcloned into a modified pMDGFP plasmid, which was transformed into B. methylotrophicus WM-1 for protein expression. The recombinant B. methylotrophicus BmT was then administered to grass carp via co-feeding, and its efficacy as an oral vaccine was assessed. Our findings demonstrated successful expression of the 55 kDa TolC and 28 kDa GFP proteins, and the preparation of polyclonal antibodies with high specificity. The BmT exhibited stable expression of the GFP-TolC fusion protein and excellent genetic stability. Following oral immunization, significant elevations were observed in serum-specific IgM levels and the activities of acid phosphatase (ACP), alkaline phosphatase (AKP), superoxide dismutase (SOD), and lysozyme (LZM) in grass carp. Concurrently, significant upregulation of immune-related genes, including IFN-I, IL-10, IL-1β, TNF-α, and IgT, was noted in the intestines, head kidney, and spleen of the grass carp. Colonization tests further revealed that the BmT persisted in the gut of immunized fish even after a fasting period of 7 days. Notably, oral administration of BmT enhanced the survival rate of grass carp following A. hydrophila infection. These results suggest that the oral BmT vaccine developed in this study holds promise for future applications in aquaculture.
    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
    患有血液病的患者被认为具有耐碳青霉烯的革兰氏阴性细菌(CR-GNB)的肠道定植的风险很高。然而,在中国,有关该人群中肠道定植的CR-GNB分离株的危险因素和分子特征的流行病学数据不足。进行了一项多中心病例对照研究,涉及来自中国92家医院的4,641名成人血液病患者。收集的直肠拭子培养后,进行了质谱和抗菌药物敏感性试验以鉴定GNB物种和CR表型。通过回顾性临床资料评估危险因素。全基因组测序用于分析CR-GNB分离株的分子特征。该试验在ClinicalTrials.gov注册为NCT05002582。我们的结果表明,在4,641名成年患者中,10.8%的人通过CR-GNB进行肠道定植。其中,8.1%被耐碳青霉烯类肠杆菌(CRE)定植,2.6%被耐碳青霉烯类铜绿假单胞菌(CRPA)定植,耐碳青霉烯鲍曼不动杆菌(CRAB)定植了0.3%。CR-GNB定植的危险因素包括男性,急性白血病,造血干细胞移植,β-内酰胺抗生素的使用,1周内出现非肛周感染。与CRPA定植患者相比,使用碳青霉烯类抗生素的患者更有可能被CRE定植.NDM是定植CRE中主要的碳青霉烯酶。这项研究揭示了在中国成人血液病患者中CR-GNB肠道定植率较高,CRE是主要的。值得注意的是,相当比例的CRE表现出金属β-内酰胺酶的产生,表明了一个令人担忧的趋势。这些发现强调了积极筛查血液疾病患者CR-GNB定植的重要性。耐IMPORTANCECarbapenem的革兰氏阴性菌(CR-GNB)已成为对公共卫生的重大威胁。血液病患者由于其免疫抑制状态而处于CR-GNB感染的高风险中。CR-GNB定植是后续感染的独立危险因素。了解CR-GNB与血液病患者肠道定植相关的危险因素和分子特征,对经验性治疗至关重要。特别是发热性中性粒细胞减少症患者。然而,流行病学数据仍然不足,我们的研究旨在确定CR-GNB的肠道定植率,确定定殖风险因素,并分析CR-GNB分离株的分子特征。
    Patients with hematological diseases are considered to be at high risk for intestinal colonization by carbapenem-resistant Gram-negative bacteria (CR-GNB). However, the epidemiological data regarding risk factors and molecular characteristics of intestinal colonized CR-GNB isolates in this population are insufficient in China. A multicenter case‒control study involving 4,641 adult patients with hematological diseases from 92 hospitals across China was conducted. Following culture of collected rectal swabs, mass spectrometry and antimicrobial susceptibility tests were performed to identify GNB species and CR phenotype. Risk factors were assessed through retrospective clinical information. Whole-genome sequencing was used to analyze the molecular characteristics of CR-GNB isolates. This trial is registered with ClinicalTrials.gov as NCT05002582. Our results demonstrated that among 4,641 adult patients, 10.8% had intestinal colonization by CR-GNB. Of these, 8.1% were colonized by carbapenem-resistant Enterobacterales (CRE), 2.6% were colonized by carbapenem-resistant Pseudomonas aeruginosa (CRPA), and 0.3% were colonized by carbapenem-resistant Acinetobacter baumannii (CRAB). The risk factors for CR-GNB colonization include male gender, acute leukemia, hematopoietic stem cell transplantation, β-lactam antibiotic usage, and the presence of non-perianal infections within 1 week. Compared with CRPA-colonized patients, patients using carbapenems were more likely to be colonized with CRE. NDM was the predominant carbapenemase in colonized CRE. This study revealed a high CR-GNB intestinal colonization rate among adult patients with hematological diseases in China, with CRE being the predominant one. Notably, a significant proportion of CRE exhibited metallo-β-lactamase production, indicating a concerning trend. These findings emphasize the importance of active screening for CR-GNB colonization in patients with hematological diseases.IMPORTANCECarbapenem-resistant Gram-negative bacteria (CR-GNB) has emerged as a significant threat to public health. Patients with hematological diseases are at high risk of CR-GNB infections due to their immunosuppressed state. CR-GNB colonization is an independent risk factor for subsequent infection. Understanding the risk factors and molecular characteristics of CR-GNB associated with intestinal colonization in patients with hematological diseases is crucial for empirical treatment, particularly in patients with febrile neutropenia. However, the epidemiology data are still insufficient, and our study aims to determine the intestinal colonization rate of CR-GNB, identify colonization risk factors, and analyze the molecular characteristics of colonized CR-GNB isolates.
    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
    避免病原体是一种至关重要的进化保守行为,可以通过预防不同物种的感染来提高存活率。包括秀丽隐杆线虫(C.线虫)。这种行为依赖于暴露于外部环境的带有纤毛的多个化学感应神经元。然而,神经元纤毛在避免病原体中的具体作用尚未完全阐明。在这里,我们发现osm-3(p802)突变体,缺乏化学感觉神经元纤毛,表现出对病原体铜绿假单胞菌PA14的较慢回避,但不表现出对大肠杆菌OP50的回避。当osm-3(p802)突变体暴露于铜绿假单胞菌PAO1时,该观察结果是一致的。在与PA14相遇后,抽水,颠簸,osm-3突变体的排便行为与野生型相当。然而,osm-3突变体显示PA14的肠道定植减少,表明它们具有更强的肠道清除能力。我们进行了RNA-seq以鉴定由于osm-3和PA14感染的丢失而差异表达的对外部刺激作出反应的基因。使用RNAi,我们证明了这些基因中的三个是正常避免病原体所必需的。总之,我们的研究结果表明,化学感应神经元纤毛的丧失减少了秀丽隐杆线虫的病原体回避,同时延缓了肠道定植和扩张。
    Pathogen avoidance is a crucial and evolutionarily conserved behavior that enhances survival by preventing infection in diverse species, including Caenorhabditis elegans (C. elegans). This behavior relies on multiple chemosensory neurons equipped with cilia that are exposed to the external environment. However, the specific role of neuronal cilia in pathogen avoidance has not been completely elucidated. Herein, we discovered that osm-3(p802) mutants, which lack chemosensory neuronal cilia, exhibit slower avoidance of the pathogen Pseudomonas aeruginosa PA14, but not Escherichia coli OP50. This observation was consistent when osm-3(p802) mutants were exposed to P. aeruginosa PAO1. Following an encounter with PA14, the pumping, thrashing, and defecation behaviors of osm-3 mutants were comparable to those of the wild-type. However, the osm-3 mutants demonstrated reduced intestinal colonization of PA14, suggesting that they have stronger intestinal clearance ability. We conducted RNA-seq to identify genes responding to external stimuli that were differentially expressed owing to the loss of osm-3 and PA14 infection. Using RNAi, we demonstrated that three of these genes were essential for normal pathogen avoidance. In conclusion, our findings demonstrate that the loss of chemosensory neuronal cilia reduces pathogen avoidance in C. elegans while delaying intestinal colonization.
    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
    菊粉,日益被研究的膳食纤维,改变肠道微生物群。这项研究的目的是评估菊粉是否减少多药耐药大肠杆菌的肠道定植,并研究其潜在的作用机制。用产生超广谱β-内酰胺酶的大肠杆菌(ESBL-E.大肠杆菌)。菊粉和泮托拉唑(IP)的组合显着降低ESBL-E。大肠杆菌粪便滴度,而泮托拉唑单药则没有,菊粉的作用延迟且有限。使用鸟枪宏基因组测序和qPCR评估粪便微生物组。通过增加74个分类单元来预测IP的功效,包括两种Adlercreutzia。A.caecimuris或A.muris的预防性治疗也降低了ESBL-E。大肠杆菌粪便滴度.通过IP有效治疗的小鼠的粪便菌群富含参与菊粉分解代谢的基因,丙酸的生产和β-内酰胺酶的表达。它们还具有增加的β-内酰胺酶活性和降低的阿莫西林浓度。这些结果表明IP通过微生物群产生丙酸盐和降解阿莫西林起作用。泮托拉唑和菊粉的组合是多药耐药大肠杆菌肠道定植的潜在治疗方法。益生元促进产生丙酸和/或β-内酰胺酶的细菌的能力可用作筛选工具,以鉴定多药耐药肠杆菌的肠道定植的潜在治疗。
    Inulin, an increasingly studied dietary fiber, alters intestinal microbiota. The aim of this study was to assess whether inulin decreases intestinal colonization by multidrug resistant E. coli and to investigate its potential mechanisms of action. Mice with amoxicillin-induced intestinal dysbiosis mice were inoculated with extended spectrum beta-lactamase producing E. coli (ESBL-E. coli). The combination of inulin and pantoprazole (IP) significantly reduced ESBL-E. coli fecal titers, whereas pantoprazole alone did not and inulin had a delayed and limited effect. Fecal microbiome was assessed using shotgun metagenomic sequencing and qPCR. The efficacy of IP was predicted by increased abundance of 74 taxa, including two species of Adlercreutzia. Preventive treatments with A. caecimuris or A. muris also reduced ESBL-E. coli fecal titers. Fecal microbiota of mice effectively treated by IP was enriched in genes involved in inulin catabolism, production of propionate and expression of beta-lactamases. They also had increased beta-lactamase activity and decreased amoxicillin concentration. These results suggest that IP act through production of propionate and degradation of amoxicillin by the microbiota. The combination of pantoprazole and inulin is a potential treatment of intestinal colonization by multidrug-resistant E. coli. The ability of prebiotics to promote propionate and/or beta-lactamase producing bacteria may be used as a screening tool to identify potential treatments of intestinal colonization by multidrug resistant Enterobacterales.
    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
    细菌感染对全球健康构成重大威胁,估计有770万人死亡。由多重耐药病原体驱动的医院疫情,特别是肺炎克雷伯菌(K.肺炎),令人严重关切。这种机会性病原体会导致肺炎,尿路感染,和菌血症,特别是在免疫受损的个体中。高毒力肺炎克雷伯菌的出现增加了复杂性,因为它越来越多地感染健康的个体。最近的流行病学数据表明,无症状的胃肠道运输是同一个体感染的宿主,并允许通过粪便-口腔途径在宿主之间传播。本文对肺炎克雷伯菌的胃肠道定植,深入研究流行病学证据,目前的动物模型,分子定植机制,以及常驻肠道微生物群的保护作用。此外,本综述阐明了体内高通量方法,这些方法对于鉴定肠道定植中的肺炎克雷伯菌因子至关重要.这项全面的探索旨在提高我们对肺炎克雷伯菌肠道发病机制的认识,指导未来的干预和预防策略。
    Bacterial infections pose a significant global health threat, accounting for an estimated 7.7 million deaths. Hospital outbreaks driven by multi-drug-resistant pathogens, notably Klebsiella pneumoniae (K. pneumoniae), are of grave concern. This opportunistic pathogen causes pneumonia, urinary tract infections, and bacteremia, particularly in immunocompromised individuals. The rise of hypervirulent K. pneumoniae adds complexity, as it increasingly infects healthy individuals. Recent epidemiological data suggest that asymptomatic gastrointestinal carriage serves as a reservoir for infections in the same individual and allows for host-to-host transmission via the fecal-oral route. This review focuses on K. pneumoniae\'s gastrointestinal colonization, delving into epidemiological evidence, current animal models, molecular colonization mechanisms, and the protective role of the resident gut microbiota. Moreover, the review sheds light on in vivo high-throughput approaches that have been crucial for identifying K. pneumoniae factors in gut colonization. This comprehensive exploration aims to enhance our understanding of K. pneumoniae gut pathogenesis, guiding future intervention and prevention strategies.
    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
    病原体定植微塑料(MPs)能够促进病原体传播的前提已得到广泛认可,然而,它们在宿主肠道病原体定植中的作用尚未完全阐明。这里,我们研究了聚苯乙烯MPs(PS-MPs)对Veronii气单胞菌定植水平的影响,一种典型的水生病原体,在泥炭泥(泥炭泥)肠中。观察到多种类型的MPs促进维氏A的肠道定植,其中PS-MPs表现出最显著的刺激作用(Veronii定植增加67.18%)。PS-MPs对泥沙的肠道造成严重损害,并引起肠道微生物群失调。某些具有抗Veronii定植抗性的肠道细菌的丰度降低,与乳球菌sp.表现出最强的定植抗性(维龙杆菌定植下降73.64%)。进行粪便微生物移植,这表明PS-MPs诱导的肠道微生物群菌群失调是维氏A在肠道定植增加的原因。已确定PS-MPs重塑了肠道微生物群落,以减弱对A.veronii定植的定植抗性,导致维龙杆菌的肠道定植水平升高。
    The premise that pathogen colonized microplastics (MPs) can promote the spread of pathogens has been widely recognized, however, their role in the colonization of pathogens in a host intestine has not been fully elucidated. Here, we investigated the effect of polystyrene MPs (PS-MPs) on the colonization levels of Aeromonas veronii, a typical aquatic pathogen, in the loach (Misgurnus anguillicaudatus) intestine. Multiple types of MPs were observed to promote the intestinal colonization of A. veronii, among which PS-MPs exhibited the most significant stimulating effect (67.18% increase in A. veronii colonization). PS-MPs inflicted serious damage to the intestinal tracts of loaches and induced intestinal microbiota dysbiosis. The abundance of certain intestinal bacteria with resistance against A. veronii colonization decreased, with Lactococcus sp. showing the strongest colonization resistance (73.64% decline in A. veronii colonization). Fecal microbiota transplantation was performed, which revealed that PS-MPs induced intestinal microbiota dysbiosis was responsible for the increased colonization of A. veronii in the intestine. It was determined that PS-MPs reshaped the intestinal microbiota community to attenuate the colonization resistance against A. veronii colonization, resulting in an elevated intestinal colonization levels of A. veronii.
    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: Case Reports
    由香型假单胞菌引起的人类感染,一种环境细菌,很少见,近年来仅报道了2例与罕见尿路感染和菌血症有关的病例。所有这些病例通常发生在免疫功能受损或降低的老年患者中。同时,由侵入性活检程序或胃肠道疾病如胃肠炎引起的上皮屏障破坏为香型假单胞菌渗透生物体提供了途径。在这项研究中,我们提供了一例病例的首例报告,其中从无潜在疾病的患者发炎的阑尾中分离出了香型假单胞菌和大肠杆菌。与大肠杆菌相比,在阑尾炎患者中从未分离出香茅假单胞菌。我们使用MALDI-TOFMS和基因测序鉴定了该物种。根据我们的发现,我们强调了香型假单胞菌可以在健康个体的肠道中定植,并可能引发阑尾炎等感染的观点.
    Human infections caused by Pseudomonas citronellolis, an environmental bacterium, are infrequent, with only two cases related to uncommon urinary tract infections and bacteremia reported in recent years. All these cases typically occurred in elderly patients with compromised or decreased immune function. Simultaneously, the epithelial barrier disruption induced by invasive biopsy procedures or gastrointestinal disorders such as gastroenteritis provided a pathway for Pseudomonas citronellolis to infiltrate the organism. In this study, we present the first report of a case where Pseudomonas citronellolis and Escherichia coli were isolated from the inflamed appendix of a patient without underlying conditions. Compared to the Escherichia coli, Pseudomonas citronellolis has never been isolated in patients with appendicitis. We identified the species using MALDI-TOF MS and genetic sequencing. Based on our findings, we highlight the perspective that Pseudomonas citronellolis can colonize the intestines of healthy individuals and may trigger infections like appendicitis.
    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
    半乳糖-N-二糖(GNB)是胃肠道(GI)粘膜中粘蛋白糖蛋白的聚糖的重要核心结构。因为某些有益细菌居住在胃肠道中,例如双歧杆菌和乳酸菌,拥有高度专业化的GNB代谢能力,GNB被认为是一种有前途的益生元,用于滋养和操纵胃肠道中的有益细菌。然而,GNB与有益细菌之间的精确相互作用及其伴随的健康促进作用仍然难以捉摸。首先,我们使用肠道细菌菌株评估了有益菌的增殖趋势及其产生的有益菌代谢产物。通过比较GNB的使用,葡萄糖,和菊粉作为碳源,我们发现GNB提高了干酪乳杆菌中乙酸的产量,鼠李糖乳杆菌,gasseri乳杆菌,和约翰森氏乳杆菌.GNB促进乙酸盐产生的能力也通过RNA-seq分析得到证实,这表明催化N-乙酰半乳糖胺-6P脱乙酰并从丙酮酸生物合成乙酰辅酶A的基因簇上调,这两者都导致乙酸盐的产生。探讨GNB在体内促进乙酸产生的作用,用抗生素治疗的BALB/cA小鼠给予GNB和鼠李糖乳杆菌,导致粪便乙酸盐含量比仅施用鼠李糖乳杆菌的小鼠高2.7倍。此外,上次给药后2天,在给予有鼠李糖乳杆菌的GNB的粪便中检测到比仅给予鼠李糖乳杆菌高3.7倍的鼠李糖乳杆菌量。这些发现强烈表明GNB在增强鼠李糖乳杆菌定植和将鼠李糖乳杆菌转化为胃肠道中更高的乙酸生产者方面的益生元潜力。重要乳酸菌的特定成员,它们通常用作益生菌,具有通过产生免疫调节代谢物,如胞外多糖,对人体健康增强至关重要的治疗特性,短链脂肪酸,和细菌素.益生菌在胃肠道中的长停留时间延长了它们的有益健康效果。此外,定殖特性对于在粘膜疫苗接种中应用益生菌以引起局部免疫应答也是期望的。在这项研究中,我们发现GNB可以增强居住在人类胃肠道中的肠道乳酸菌的有益特性,刺激乙酸盐的产生和促进肠道定植。我们的发现为在基于乳酸菌的功能食品中添加GNB提供了理论基础。这也导致了更合理的益生元和益生菌选择支持的治疗方法的发展,改善人类的健康生活方式。
    Galacto-N-biose (GNB) is an important core structure of glycan of mucin glycoproteins in the gastrointestinal (GI) mucosa. Because certain beneficial bacteria inhabiting the GI tract, such as bifidobacteria and lactic acid bacteria, harbor highly specialized GNB metabolic capabilities, GNB is considered a promising prebiotic for nourishing and manipulating beneficial bacteria in the GI tract. However, the precise interactions between GNB and beneficial bacteria and their accompanying health-promoting effects remain elusive. First, we evaluated the proliferative tendency of beneficial bacteria and their production of beneficial metabolites using gut bacterial strains. By comparing the use of GNB, glucose, and inulin as carbon sources, we found that GNB enhanced acetate production in Lacticaseibacillus casei, Lacticaseibacillus rhamnosus, Lactobacillus gasseri, and Lactobacillus johnsonii. The ability of GNB to promote acetate production was also confirmed by RNA-seq analysis, which indicated the upregulation of gene clusters that catalyze the deacetylation of N-acetylgalactosamine-6P and biosynthesize acetyl-CoA from pyruvate, both of which result in acetate production. To explore the in vivo effect of GNB in promoting acetate production, antibiotic-treated BALB/cA mice were administered with GNB with L. rhamnosus, resulting in a fecal acetate content that was 2.7-fold higher than that in mice administered with only L. rhamnosus. Moreover, 2 days after the last administration, a 3.7-fold higher amount of L. rhamnosus was detected in feces administered with GNB with L. rhamnosus than in feces administered with only L. rhamnosus. These findings strongly suggest the prebiotic potential of GNB in enhancing L. rhamnosus colonization and converting L. rhamnosus into higher acetate producers in the GI tract.
    OBJECTIVE: Specific members of lactic acid bacteria, which are commonly used as probiotics, possess therapeutic properties that are vital for human health enhancement by producing immunomodulatory metabolites such as exopolysaccharides, short-chain fatty acids, and bacteriocins. The long residence time of probiotic lactic acid bacteria in the GI tract prolongs their beneficial health effects. Moreover, the colonization property is also desirable for the application of probiotics in mucosal vaccination to provoke a local immune response. In this study, we found that GNB could enhance the beneficial properties of intestinal lactic acid bacteria that inhabit the human GI tract, stimulating acetate production and promoting intestinal colonization. Our findings provide a rationale for the addition of GNB to lactic acid bacteria-based functional foods. This has also led to the development of therapeutics supported by more rational prebiotic and probiotic selection, leading to an improved healthy lifestyle for humans.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号