首页 > 最新文献

Human Microbiome Journal最新文献

英文 中文
Comparison of the prokaryotic and eukaryotic microbial communities in peripheral blood from amyotrophic lateral sclerosis, multiple sclerosis, and control populations 肌萎缩性侧索硬化症、多发性硬化症和对照人群外周血原核和真核微生物群落的比较
Q1 Medicine Pub Date : 2019-08-01 DOI: 10.1016/j.humic.2019.100060
Jeremy E. Ellis , Dara S. Missan , Matthew Shabilla , Constantine Moschonas , David Saperstein , Delyn Martinez , Christian V. Becker , Stephen E. Fry

Neurodegenerative diseases are estimated to afflict hundreds of thousands of Americans with vastly more worldwide. The etiologies of amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) have yet to be established. Previous studies have suggested an association of these diseases with viruses, bacteria, and eukaryotic microbes, no new therapies have been forthcoming. High-throughput DNA sequencing has enabled the comprehensive analysis of microbial DNA profiles in diseased populations. To date, no amplicon-based next-generation DNA sequencing prokaryotic and eukaryotic community profiling studies have been completed for these diseases. Analysis of peripheral blood samples from control participants as well as ALS and MS participants was used to characterize the hematologic population of microbial DNA. Categorical and multivariate analysis with control for multiple comparisons and aged matched controls revealed differences in microbial DNA contribution in ALS patients compared to others. Notably, sequences that belonging to Ochrophyta were enriched in ALS patient samples. Mechanisms underlying this association, the role of microbial DNA sequences, and the development or progression of ALS may become a fertile subject of inquiry.

据估计,神经退行性疾病折磨着成千上万的美国人,而全世界的人数则多得多。肌萎缩性侧索硬化症(ALS)和多发性硬化症(MS)的病因尚未确定。以前的研究表明这些疾病与病毒、细菌和真核微生物有关,但没有新的治疗方法。高通量DNA测序使患病人群微生物DNA谱的全面分析成为可能。迄今为止,还没有基于扩增子的下一代DNA测序原核和真核生物群落分析研究完成这些疾病。分析来自对照参与者以及ALS和MS参与者的外周血样本用于表征微生物DNA的血液学种群。多组对照和年龄匹配对照的分类和多变量分析显示,ALS患者的微生物DNA贡献与其他患者相比存在差异。值得注意的是,属于Ochrophyta的序列在ALS患者样本中富集。这种关联的机制,微生物DNA序列的作用,以及ALS的发展或进展可能成为探究的肥沃主题。
{"title":"Comparison of the prokaryotic and eukaryotic microbial communities in peripheral blood from amyotrophic lateral sclerosis, multiple sclerosis, and control populations","authors":"Jeremy E. Ellis ,&nbsp;Dara S. Missan ,&nbsp;Matthew Shabilla ,&nbsp;Constantine Moschonas ,&nbsp;David Saperstein ,&nbsp;Delyn Martinez ,&nbsp;Christian V. Becker ,&nbsp;Stephen E. Fry","doi":"10.1016/j.humic.2019.100060","DOIUrl":"10.1016/j.humic.2019.100060","url":null,"abstract":"<div><p>Neurodegenerative diseases are estimated to afflict hundreds of thousands of Americans with vastly more worldwide. The etiologies of amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS) have yet to be established. Previous studies have suggested an association of these diseases with viruses, bacteria, and eukaryotic microbes, no new therapies have been forthcoming. High-throughput DNA sequencing has enabled the comprehensive analysis of microbial DNA profiles in diseased populations. To date, no amplicon-based next-generation DNA sequencing prokaryotic and eukaryotic community profiling studies have been completed for these diseases. Analysis of peripheral blood samples from control participants as well as ALS and MS participants was used to characterize the hematologic population of microbial DNA. Categorical and multivariate analysis with control for multiple comparisons and aged matched controls revealed differences in microbial DNA contribution in ALS patients compared to others. Notably, sequences that belonging to <em>Ochrophyta</em> were enriched in ALS patient samples. Mechanisms underlying this association, the role of microbial DNA sequences, and the development or progression of ALS may become a fertile subject of inquiry.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2019.100060","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"42192592","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Digestive tract mycobiota and microbiota and the effects on the immune system 消化道菌群和微生物群及其对免疫系统的影响
Q1 Medicine Pub Date : 2019-06-01 DOI: 10.1016/j.humic.2019.100056
Nina Gouba , Yeri Esther Hien , Marie Laure Guissou , Maxime Descartes Mbogning Fonkou , Yves Traoré , Zekiba Tarnagda

The human gastrointestinal tract exists as a complex ecosystem and contains a mycobiome and a microbiome that play central roles in host health, disease and immune system regulation. Here, we reviewed the traditional culture-dependent methods, the culturomics methods and the molecular methods used to study the gut mycobiome and microbiome. With the development of next-generation sequencing techniques, these last two methods have greatly broadened the understanding of the roles of gut bacteria in health and disease. Thus, dysbiosis of the gut microbiota and mycobiota has been found to be associated with some diseases; disruptions to the fungal and bacterial commensal communities influence the immune response and impact disease status.

人类胃肠道作为一个复杂的生态系统存在,包含一个真菌组和一个微生物组,在宿主健康、疾病和免疫系统调节中发挥核心作用。本文综述了传统的培养依赖性方法、培养组学方法和分子方法在肠道菌群和微生物组研究中的应用。随着新一代测序技术的发展,后两种方法极大地拓宽了对肠道细菌在健康和疾病中的作用的理解。因此,已发现肠道微生物群和真菌群的生态失调与某些疾病有关;真菌和细菌共生群落的破坏会影响免疫反应并影响疾病状态。
{"title":"Digestive tract mycobiota and microbiota and the effects on the immune system","authors":"Nina Gouba ,&nbsp;Yeri Esther Hien ,&nbsp;Marie Laure Guissou ,&nbsp;Maxime Descartes Mbogning Fonkou ,&nbsp;Yves Traoré ,&nbsp;Zekiba Tarnagda","doi":"10.1016/j.humic.2019.100056","DOIUrl":"10.1016/j.humic.2019.100056","url":null,"abstract":"<div><p>The human gastrointestinal tract exists as a complex ecosystem and contains a mycobiome and a microbiome that play central roles in host health, disease and immune system regulation. Here, we reviewed the traditional culture-dependent methods, the culturomics methods and the molecular methods used to study the gut mycobiome and microbiome. With the development of next-generation sequencing techniques, these last two methods have greatly broadened the understanding of the roles of gut bacteria in health and disease. Thus, dysbiosis of the gut microbiota and mycobiota has been found to be associated with some diseases; disruptions to the fungal and bacterial commensal communities influence the immune response and impact disease status.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2019.100056","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48059246","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 10
Whole genome metagenomic analysis of the gut microbiome of differently fed infants identifies differences in microbial composition and functional genes, including an absent CRISPR/Cas9 gene in the formula-fed cohort 对不同喂养方式婴儿肠道微生物组的全基因组宏基因组分析发现了微生物组成和功能基因的差异,包括配方奶喂养队列中缺失的CRISPR/Cas9基因
Q1 Medicine Pub Date : 2019-06-01 DOI: 10.1016/j.humic.2019.100057
Matthew D. Di Guglielmo , Karl Franke , Courtney Cox , Erin L. Crowgey

Background

Advancements in sequencing capabilities have enhanced the study of the human microbiome. There are limited studies focused on the gastro-intestinal (gut) microbiome of infants, particularly the impact of diet between breast-fed (BF) versus formula-fed (FF). It is unclear what effect, if any, early feeding has on short-term or long-term composition and function of the gut microbiome.

Results

Using a shotgun metagenomics approach, differences in the gut microbiome between BF (n = 10) and FF (n = 5) infants were detected. A Jaccard distance principle coordinate analysis was able to cluster BF versus FF infants based on the presence or absence of species identified in their gut microbiome. Thirty-two genera were identified as statistically different in the gut microbiome sequenced between BF and FF infants. Furthermore, the computational workflow identified 371 bacterial genes that were statistically different between the BF and FF cohorts in abundance. Only seven genes were lower in abundance (or absent) in the FF cohort compared to the BF cohort, including CRISPR/Cas9; whereas, the remaining candidates, including autotransporter adhesins, were higher in abundance in the FF cohort compared to BF cohort.

Conclusions

These studies demonstrated that FF infants have, at an early age, a significantly different gut microbiome with potential implications for function of the fecal microbiota. Interactions between the fecal microbiota and host hinted at here have been linked to numerous diseases. Determining whether these non-abundant or more abundant genes have biological consequence related to infant feeding may aid in understanding the adult gut microbiome, and the pathogenesis of obesity.

测序能力的进步促进了对人类微生物组的研究。关于婴儿胃肠道(肠道)微生物组的研究有限,特别是母乳喂养(BF)和配方奶粉喂养(FF)之间饮食的影响。目前尚不清楚早期喂养对肠道微生物组的短期或长期组成和功能有什么影响,如果有的话。结果采用散弹枪宏基因组学方法,检测BF (n = 10)和FF (n = 5)婴儿肠道微生物组的差异。Jaccard距离原理坐标分析能够根据肠道微生物组中确定的物种的存在与否对BF和FF婴儿进行聚类。在BF和FF婴儿之间的肠道微生物组测序中,鉴定出32个属具有统计学差异。此外,计算工作流确定了371个细菌基因,这些基因在BF和FF队列中有统计学差异。与BF组相比,FF组中只有7个基因丰度较低(或缺失),包括CRISPR/Cas9;而其余候选蛋白,包括自体转运蛋白粘连素,在FF组中的丰度高于BF组。这些研究表明,FF婴儿在早期具有显著不同的肠道微生物群,这对粪便微生物群的功能有潜在的影响。这里暗示的粪便微生物群和宿主之间的相互作用与许多疾病有关。确定这些不丰富或更丰富的基因是否与婴儿喂养有关,可能有助于了解成人肠道微生物组和肥胖的发病机制。
{"title":"Whole genome metagenomic analysis of the gut microbiome of differently fed infants identifies differences in microbial composition and functional genes, including an absent CRISPR/Cas9 gene in the formula-fed cohort","authors":"Matthew D. Di Guglielmo ,&nbsp;Karl Franke ,&nbsp;Courtney Cox ,&nbsp;Erin L. Crowgey","doi":"10.1016/j.humic.2019.100057","DOIUrl":"10.1016/j.humic.2019.100057","url":null,"abstract":"<div><h3>Background</h3><p>Advancements in sequencing capabilities have enhanced the study of the human microbiome. There are limited studies focused on the gastro-intestinal (gut) microbiome of infants, particularly the impact of diet between breast-fed (BF) versus formula-fed (FF). It is unclear what effect, if any, early feeding has on short-term or long-term composition and function of the gut microbiome.</p></div><div><h3>Results</h3><p>Using a shotgun metagenomics approach, differences in the gut microbiome between BF (n = 10) and FF (n = 5) infants were detected. A Jaccard distance principle coordinate analysis was able to cluster BF versus FF infants based on the presence or absence of species identified in their gut microbiome. Thirty-two genera were identified as statistically different in the gut microbiome sequenced between BF and FF infants. Furthermore, the computational workflow identified 371 bacterial genes that were statistically different between the BF and FF cohorts in abundance. Only seven genes were lower in abundance (or absent) in the FF cohort compared to the BF cohort, including CRISPR/Cas9; whereas, the remaining candidates, including autotransporter adhesins, were higher in abundance in the FF cohort compared to BF cohort.</p></div><div><h3>Conclusions</h3><p>These studies demonstrated that FF infants have, at an early age, a significantly different gut microbiome with potential implications for function of the fecal microbiota. Interactions between the fecal microbiota and host hinted at here have been linked to numerous diseases. Determining whether these non-abundant or more abundant genes have biological consequence related to infant feeding may aid in understanding the adult gut microbiome, and the pathogenesis of obesity.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2019.100057","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"39197388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
Analysis of blood and fecal microbiome profile in patients with celiac disease 乳糜泻患者血液和粪便微生物组分析
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.12.001
Gloria Serena , Camron Davies , Murat Cetinbas , Ruslan I. Sadreyev , Alessio Fasano

Celiac disease is a multifactorial autoimmune enteropathy triggered by ingestion of gluten in genetically predisposed individuals. The increase of incidence in celiac disease suggests that additional environmental factors other than gluten may contribute to its onset and development. While intestinal dysbiosis has already been associated with celiac disease, the role that the blood microbiome plays in the loss of tolerance to gluten is unknown. In this study we aimed at evaluating weather celiac patients are characterized by alterations in the blood microbiome and how these changes may relate to the intestinal microbiome composition and, ultimately, to the loss of tolerance to gluten. Our data highlight alterations in the blood microbiome composition and taxonomic diversity in celiac patients as compared to healthy subjects. Although preliminary, these findings suggest that changes in the blood microbiome may contribute to the pathogenesis of celiac disease and open the possibility of new therapeutic and diagnostic tools for celiac patients.

乳糜泻是一种多因素的自身免疫性肠病,由遗传易感个体摄入麸质引发。乳糜泻发病率的增加表明除谷蛋白外的其他环境因素可能有助于其发病和发展。虽然肠道生态失调已经与乳糜泻有关,但血液微生物群在失去对麸质耐受性方面所起的作用尚不清楚。在这项研究中,我们旨在评估乳糜泻患者是否以血液微生物组的改变为特征,以及这些变化如何与肠道微生物组组成以及最终与对麸质耐受性的丧失有关。我们的数据强调了与健康受试者相比,乳糜泻患者血液微生物组组成和分类多样性的变化。虽然是初步的,但这些发现表明血液微生物组的变化可能与乳糜泻的发病机制有关,并为乳糜泻患者提供新的治疗和诊断工具。
{"title":"Analysis of blood and fecal microbiome profile in patients with celiac disease","authors":"Gloria Serena ,&nbsp;Camron Davies ,&nbsp;Murat Cetinbas ,&nbsp;Ruslan I. Sadreyev ,&nbsp;Alessio Fasano","doi":"10.1016/j.humic.2018.12.001","DOIUrl":"10.1016/j.humic.2018.12.001","url":null,"abstract":"<div><p>Celiac disease is a multifactorial autoimmune enteropathy triggered by ingestion of gluten in genetically predisposed individuals. The increase of incidence in celiac disease suggests that additional environmental factors other than gluten may contribute to its onset and development. While intestinal dysbiosis has already been associated with celiac disease, the role that the blood microbiome plays in the loss of tolerance to gluten is unknown. In this study we aimed at evaluating weather celiac patients are characterized by alterations in the blood microbiome and how these changes may relate to the intestinal microbiome composition and, ultimately, to the loss of tolerance to gluten. Our data highlight alterations in the blood microbiome composition and taxonomic diversity in celiac patients as compared to healthy subjects. Although preliminary, these findings suggest that changes in the blood microbiome may contribute to the pathogenesis of celiac disease and open the possibility of new therapeutic and diagnostic tools for celiac patients.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.12.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41959819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 20
Virus and microbiota relationships in humans and other mammals: An evolutionary view 人类和其他哺乳动物中病毒和微生物群的关系:一种进化观点
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.11.001
Maurício Teixeira Lima, Ana Cláudia dos Santos Pereira Andrade, Graziele Pereira Oliveira, Jacques Robert Nicoli, Flaviano dos Santos Martins, Erna Geessien Kroon, Jônatas Santos Abrahão

In the last decades, studies have revealed multiple and strong correlations between the host and its commensal microbiota consisting of bacteria, protozoa, fungi and viruses. This associated microbiota can positively or negatively influence the course of a wide range of infections. Here, we review the interactions between the host and its viral microbiota and discuss new paradigms from an evolutionary perspective. The viral adaptation to a microbial environment in a co-evolutionary approach is highlighted, as well as viral cross transmission in the context of the barriers imposed by the indigenous microbiota. In addition to reviewing the host-microbiota-virus relationships, we focus the discussion on microbiota-virus interactions that could be applied to preventive and therapeutic treatments.

在过去的几十年里,研究已经揭示了宿主与其共生微生物群(包括细菌、原生动物、真菌和病毒)之间的多重和强相关性。这种相关的微生物群可以对多种感染的过程产生积极或消极的影响。在这里,我们回顾了宿主与病毒微生物群之间的相互作用,并从进化的角度讨论了新的范式。强调了在共同进化方法中病毒对微生物环境的适应,以及在本地微生物群施加障碍的背景下病毒的交叉传播。除了回顾宿主-微生物-病毒之间的关系外,我们还重点讨论了微生物-病毒之间的相互作用,这些相互作用可以应用于预防和治疗治疗。
{"title":"Virus and microbiota relationships in humans and other mammals: An evolutionary view","authors":"Maurício Teixeira Lima,&nbsp;Ana Cláudia dos Santos Pereira Andrade,&nbsp;Graziele Pereira Oliveira,&nbsp;Jacques Robert Nicoli,&nbsp;Flaviano dos Santos Martins,&nbsp;Erna Geessien Kroon,&nbsp;Jônatas Santos Abrahão","doi":"10.1016/j.humic.2018.11.001","DOIUrl":"10.1016/j.humic.2018.11.001","url":null,"abstract":"<div><p>In the last decades, studies have revealed multiple and strong correlations between the host and its commensal microbiota consisting of bacteria, protozoa, fungi and viruses. This associated microbiota can positively or negatively influence the course of a wide range of infections. Here, we review the interactions between the host and its viral microbiota and discuss new paradigms from an evolutionary perspective. The viral adaptation to a microbial environment in a co-evolutionary approach is highlighted, as well as viral cross transmission in the context of the barriers imposed by the indigenous microbiota. In addition to reviewing the host-microbiota-virus relationships, we focus the discussion on microbiota-virus interactions that could be applied to preventive and therapeutic treatments.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.11.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43423719","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 11
Current and future targets for faecal microbiota transplantation 粪便微生物群移植的当前和未来目标
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.08.004
James Roger Mcilroy , Jonathan P. Segal , Benjamin H. Mullish , Mohammed Nabil Quraishi , Antonio Gasbarrini , Giovanni Cammarota , Gianluca Ianiro

The human gastrointestinal tract is home to the most diverse microbial ecosystem in the human body and is made up of bacteria, viruses and eukarya. Collectively known as the gut microbiota, our knowledge of these microbial communities has historically been restricted by the relative limitations of culturing techniques. However, the recent development and utilisation of next-generation sequencing techniques has enhanced our understanding of its structure, diversity and function.

There is emerging evidence that the gut microbiota plays a pivotal role in both health and disease. Perturbations to the structure and function of the gut microbiota are known to be associated with certain disease states. Therefore, manipulating the gut microbiota in an attempt to restore structure and function represents a promising therapeutic strategy. Recently, there has been a surge in clinical and scientific interest in manipulating the gut microbiota using a method called faecal microbiota transplantation. This increase in interest has gathered after it was shown in randomised controlled trials to be highly effective in treating recurrent Clostridium difficile infection.

Despite success in treating recurrent Clostridium difficile, there remain many unknowns about how best to optimise its preparation, regulation, mode of delivery and safety. This review aims to summarise the literature surrounding the current knowledge regarding faecal microbiota transplantation and explore potential future research avenues that aim to enhance the safety, efficacy and utilisation of faecal microbiota transplantation.

人体胃肠道是人体微生物生态系统最多样化的家园,由细菌、病毒和真核生物组成。我们对这些微生物群落的认识被统称为肠道微生物群,历史上一直受到培养技术的相对限制。然而,近年来新一代测序技术的发展和利用提高了我们对其结构、多样性和功能的认识。越来越多的证据表明,肠道微生物群在健康和疾病中都起着关键作用。肠道菌群结构和功能的扰动已知与某些疾病状态有关。因此,通过操纵肠道菌群来恢复结构和功能是一种很有前景的治疗策略。最近,临床和科学对使用一种称为粪便微生物群移植的方法来操纵肠道微生物群的兴趣激增。在随机对照试验中显示它对治疗复发性艰难梭菌感染非常有效后,人们对它的兴趣增加了。尽管在治疗复发性艰难梭菌方面取得了成功,但关于如何最好地优化其制备、监管、给药方式和安全性,仍存在许多未知因素。本文综述了目前有关粪便微生物群移植的文献,并探讨了未来可能的研究途径,旨在提高粪便微生物群移植的安全性、有效性和利用率。
{"title":"Current and future targets for faecal microbiota transplantation","authors":"James Roger Mcilroy ,&nbsp;Jonathan P. Segal ,&nbsp;Benjamin H. Mullish ,&nbsp;Mohammed Nabil Quraishi ,&nbsp;Antonio Gasbarrini ,&nbsp;Giovanni Cammarota ,&nbsp;Gianluca Ianiro","doi":"10.1016/j.humic.2018.08.004","DOIUrl":"10.1016/j.humic.2018.08.004","url":null,"abstract":"<div><p>The human gastrointestinal tract is home to the most diverse microbial ecosystem in the human body and is made up of bacteria, viruses and eukarya. Collectively known as the gut microbiota, our knowledge of these microbial communities has historically been restricted by the relative limitations of culturing techniques. However, the recent development and utilisation of next-generation sequencing techniques has enhanced our understanding of its structure, diversity and function.</p><p>There is emerging evidence that the gut microbiota plays a pivotal role in both health and disease. Perturbations to the structure and function of the gut microbiota are known to be associated with certain disease states. Therefore, manipulating the gut microbiota in an attempt to restore structure and function represents a promising therapeutic strategy. Recently, there has been a surge in clinical and scientific interest in manipulating the gut microbiota using a method called faecal microbiota transplantation. This increase in interest has gathered after it was shown in randomised controlled trials to be highly effective in treating recurrent <em>Clostridium difficile</em> infection.</p><p>Despite success in treating recurrent <em>Clostridium difficile,</em> there remain many unknowns about how best to optimise its preparation, regulation, mode of delivery and safety. This review aims to summarise the literature surrounding the current knowledge regarding faecal microbiota transplantation and explore potential future research avenues that aim to enhance the safety, efficacy and utilisation of faecal microbiota transplantation.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.08.004","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"49327775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 7
Association of gut dysbiosis with intestinal metabolites in response to antibiotic treatment 肠道生态失调与肠道代谢物对抗生素治疗反应的关联
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.11.004
Tariq Jamal Khan , Mohammed Nihal Hasan , Esam I. Azhar , Muhammad Yasir

Gut microbiota (GM) is associated with metabolism, provides energy-harvesting efficiency and protection against opportunistic pathogens through competitive exclusion to the host. Previous studies highlighted the temporary as well as permanent alteration to GM resulting from different antibiotics treatment. The diverse class of antibiotics may damage the metabolic homeostasis and can alter the level of intestinal metabolites [including amino acids, bile acids (BAs), glucose, short chain fatty acids (SCFAs)] through alteration in abundance of metabolically active bacteria. The antibiotics administration causes the disturbed profile of related microbial metabolites, especially that of BAs, primary and secondary BAs (conjugated or unconjugated BAs). The antibiotics intake causes the reduced bacterial diversity that makes the individuals susceptible towards diseases. To a large extent, we tried to clarify the adverse effects of classes of antibiotics on the GM composition, and the consequent impacts of dysbiosis on the BAs feedback loop between liver and gut, which involves the farnesoid-X-receptor-fibroblast growth factor (FXR-FGF) pathway. The current review discusses the antibiotics-GM-BAs nexus during Clostridium difficile infection (CDI) and the recommended therapy includes faecal microbial transplant (FMT) in countering the exposure of harmful antibiotic and bacteriotherapy as an alternative therapeutic intervention in treating the recurrent CDI.

肠道微生物群(GM)与代谢有关,提供能量收集效率,并通过对宿主的竞争排斥来保护机会性病原体。以前的研究强调了不同抗生素治疗对转基因造成的暂时和永久的改变。不同种类的抗生素可能会破坏代谢稳态,并通过改变代谢活性细菌的丰度来改变肠道代谢物(包括氨基酸、胆胆酸(BAs)、葡萄糖、短链脂肪酸(SCFAs))的水平。抗生素的使用会引起相关微生物代谢产物的紊乱,尤其是BAs,初级和次级BAs(偶联或未偶联的BAs)。抗生素的摄入导致细菌多样性的减少,使个体容易感染疾病。在很大程度上,我们试图阐明各类抗生素对转基因成分的不良影响,以及随之而来的生态失调对肝脏和肠道之间的BAs反馈回路的影响,其中涉及法氏体- x受体-成纤维细胞生长因子(FXR-FGF)途径。目前的综述讨论了艰难梭菌感染(CDI)期间抗生素- gm - bas的关系,推荐的治疗方法包括粪便微生物移植(FMT),以对抗有害抗生素和细菌治疗的暴露,作为治疗复发性CDI的替代治疗干预措施。
{"title":"Association of gut dysbiosis with intestinal metabolites in response to antibiotic treatment","authors":"Tariq Jamal Khan ,&nbsp;Mohammed Nihal Hasan ,&nbsp;Esam I. Azhar ,&nbsp;Muhammad Yasir","doi":"10.1016/j.humic.2018.11.004","DOIUrl":"10.1016/j.humic.2018.11.004","url":null,"abstract":"<div><p>Gut microbiota (GM) is associated with metabolism, provides energy-harvesting efficiency and protection against opportunistic pathogens through competitive exclusion to the host. Previous studies highlighted the temporary as well as permanent alteration to GM resulting from different antibiotics treatment. The diverse class of antibiotics may damage the metabolic homeostasis and can alter the level of intestinal metabolites [including amino acids, bile acids (BAs), glucose, short chain fatty acids (SCFAs)] through alteration in abundance of metabolically active bacteria. The antibiotics administration causes the disturbed profile of related microbial metabolites, especially that of BAs, primary and secondary BAs (conjugated or unconjugated BAs). The antibiotics intake causes the reduced bacterial diversity that makes the individuals susceptible towards diseases. To a large extent, we tried to clarify the adverse effects of classes of antibiotics on the GM composition, and the consequent impacts of dysbiosis on the BAs feedback loop between liver and gut, which involves the farnesoid-X-receptor-fibroblast growth factor (FXR-FGF) pathway. The current review discusses the antibiotics-GM-BAs nexus during <em>Clostridium difficile</em> infection (CDI) and the recommended therapy includes faecal microbial transplant (FMT) in countering the exposure of harmful antibiotic and bacteriotherapy as an alternative therapeutic intervention in treating the recurrent CDI.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.11.004","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"45166471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 15
Exhaustive repertoire of human vaginal microbiota 人类阴道微生物群的详尽曲目
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.11.002
Khoudia Diop , Jean-Charles Dufour , Anthony Levasseur , Florence Fenollar

Bacteria that colonize the vaginal microbiota of women play an important role in health and homeostasis. Disruption of the proportion of bacteria predisposes to dysbiosis like bacterial vaginosis or severe gynecological conditions such as preterm birth, pelvic inflammatory disease and also sexually transmitted diseases. Knowledge about normal and abnormal vaginal microbiota has become a little clearer in recent years. Culture techniques have made it possible to isolate and describe many bacterial species, whereas molecular methods have highlighted the limits of culture by showing that the vagina was a complex ecosystem containing a wide range of non-cultured or difficult-to-identify bacteria. Based on an exhaustive review of the scientific literature, we built the repertoire of all the bacteria found using culture-based and/or independent methods on the human vagina. So, whether they are valid or not, we inventoried 581 bacteria identified in the human vagina distributed into 10 taxa, mainly in the phyla of Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria with 206 distinct genera classified in 96 different families. This repertoire is essential for microbiologists and clinicians and represents the starting point for a Vaginal Microbiome Project such a project aimed to map the human vaginal microbiota, to better understand the dysbioses or infections caused by its imbalance in order to offer more appropriate treatments.

定植于女性阴道微生物群的细菌在健康和体内平衡中起着重要作用。细菌比例的破坏容易导致生态失调,如细菌性阴道病或严重的妇科疾病,如早产、盆腔炎和性传播疾病。近年来,关于正常和异常阴道微生物群的知识已经变得更加清晰。培养技术使分离和描述许多细菌物种成为可能,而分子方法通过显示阴道是一个复杂的生态系统,包含广泛的非培养或难以识别的细菌,突出了培养的局限性。基于对科学文献的详尽审查,我们建立了使用基于培养和/或独立方法在人类阴道上发现的所有细菌的库。因此,无论其是否有效,我们将在人类阴道中鉴定出的581种细菌分为10个分类群,主要分布在放线菌门、拟杆菌门、厚壁菌门和变形菌门,分别属于96个科206个属。这对微生物学家和临床医生来说是必不可少的,并且代表了阴道微生物组计划的起点,该计划旨在绘制人类阴道微生物群,以更好地了解由其不平衡引起的生态失调或感染,以便提供更合适的治疗。
{"title":"Exhaustive repertoire of human vaginal microbiota","authors":"Khoudia Diop ,&nbsp;Jean-Charles Dufour ,&nbsp;Anthony Levasseur ,&nbsp;Florence Fenollar","doi":"10.1016/j.humic.2018.11.002","DOIUrl":"10.1016/j.humic.2018.11.002","url":null,"abstract":"<div><p>Bacteria that colonize the vaginal microbiota of women play an important role in health and homeostasis. Disruption of the proportion of bacteria predisposes to dysbiosis like bacterial vaginosis or severe gynecological conditions such as preterm birth, pelvic inflammatory disease and also sexually transmitted diseases. Knowledge about normal and abnormal vaginal microbiota has become a little clearer in recent years. Culture techniques have made it possible to isolate and describe many bacterial species, whereas molecular methods have highlighted the limits of culture by showing that the vagina was a complex ecosystem containing a wide range of non-cultured or difficult-to-identify bacteria. Based on an exhaustive review of the scientific literature, we built the repertoire of all the bacteria found using culture-based and/or independent methods on the human vagina. So, whether they are valid or not, we inventoried 581 bacteria identified in the human vagina distributed into 10 taxa, mainly in the phyla of <em>Actinobacteria</em>, <em>Bacteroidetes</em>, <em>Firmicutes</em>, and <em>Proteobacteria</em> with 206 distinct genera classified in 96 different families. This repertoire is essential for microbiologists and clinicians and represents the starting point for a Vaginal Microbiome Project such a project aimed to map the human vaginal microbiota, to better understand the dysbioses or infections caused by its imbalance in order to offer more appropriate treatments.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.11.002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"43249064","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 49
Increasing and eliminating the (Fecal coliforms, thermotolerant coliform and fecal streptococcus) bacteria by resin of the poly (para carboxy acid phenol-d-Glucose) to clean up waste water 利用聚对羧酸苯酚-葡萄糖树脂净化废水,增加和消除粪便大肠菌群、耐热大肠菌群和粪便链球菌
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2018.11.003
Safia Miloudi , Messaoud Chaib , Moulkheir Ayat , Abdelkader Rahmouni

In this research paper we represent a novel synthetic antibacterial phenolic polymer (pAP-DG) containing carboxylic acid group in para position. The study of poly (p-carboxylic acid phenol d-Glucose) as an antibacterial resin was focused on the development of antimicrobial polymers to clean up the water of pathogenic bacteria. The polycondensation reaction was realized between p-carboxy acid phenol (pAP) and d-Glucose (DG) as monomers; using sulfuric acid as homogeneous catalyst.

The obtained polymer was used via fecal coliform (TC), thermotolerant coliforms (CTT) and fecal streptococci (ST) bacteria to evaluate the water quality. The antibacterial activity of the synthesized resin was carried out by the contact method so the capacity of this resin was confirmed. In general, the polymer has a good antimicrobial activity against the microorganisms tested; the resulting polymer was characterized by various spectroscopy methods such as: 1H NMR, 13C NMR, DSC and FTIR and a mechanism has been proposed for this polycondensation reaction.

本文研究了一种新型的合成抗菌酚醛聚合物(pAP-DG),其对位上含有羧酸基团。聚对羧酸苯酚- d-葡萄糖作为抗菌树脂的研究重点是开发用于清除水中致病菌的抗菌聚合物。以对羧酸苯酚(pAP)和d-葡萄糖(DG)为单体进行缩聚反应;以硫酸为均相催化剂。所得聚合物通过粪便大肠菌群(TC)、耐热大肠菌群(CTT)和粪便链球菌(ST)细菌对水质进行评价。采用接触法对合成的树脂进行了抗菌活性测试,从而证实了该树脂的抗菌性能。总的来说,该聚合物对所测微生物具有良好的抗菌活性;通过1H NMR、13C NMR、DSC和FTIR等多种光谱方法对所得聚合物进行了表征,并提出了缩聚反应的机理。
{"title":"Increasing and eliminating the (Fecal coliforms, thermotolerant coliform and fecal streptococcus) bacteria by resin of the poly (para carboxy acid phenol-d-Glucose) to clean up waste water","authors":"Safia Miloudi ,&nbsp;Messaoud Chaib ,&nbsp;Moulkheir Ayat ,&nbsp;Abdelkader Rahmouni","doi":"10.1016/j.humic.2018.11.003","DOIUrl":"10.1016/j.humic.2018.11.003","url":null,"abstract":"<div><p>In this research paper we represent a novel synthetic antibacterial phenolic polymer (pAP-DG) containing carboxylic acid group in para position. The study of poly (p-carboxylic acid phenol <span>d</span>-Glucose) as an antibacterial resin was focused on the development of antimicrobial polymers to clean up the water of pathogenic bacteria. The polycondensation reaction was realized between p-carboxy acid phenol (pAP) and <span>d</span>-Glucose (DG) as monomers; using sulfuric acid as homogeneous catalyst.</p><p>The obtained polymer was used via <em>fecal coliform</em> (TC), <em>thermotolerant coliforms</em> (CTT) and <em>fecal streptococci</em> (ST) bacteria to evaluate the water quality. The antibacterial activity of the synthesized resin was carried out by the contact method so the capacity of this resin was confirmed. In general, the polymer has a good antimicrobial activity against the microorganisms tested; the resulting polymer was characterized by various spectroscopy methods such as: <sup>1</sup>H NMR, <sup>13</sup>C NMR, DSC and FTIR and a mechanism has been proposed for this polycondensation reaction.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2018.11.003","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"48143019","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Novel targets to develop new antibacterial agents and novel alternatives to antibacterial agents 开发新的抗菌剂和新的抗菌剂替代品的新靶点
Q1 Medicine Pub Date : 2019-03-01 DOI: 10.1016/j.humic.2019.01.001
Tafere Mulaw Belete

Antibacterial agents have saved many lives and helped the growth of modern medicine over the past half century. The emergence of drug resistance, jeopardizing the effectiveness of these life-saving treatments. This clearly highlights the urgent need for new and improved antibacterial drugs with a novel target and new molecular structure agent to obviate cross-resistance. This paper reviewed the possible new ways to discover novel antibacterial agents. The most widely studied new bacterial targets for novel drug development are quorum sensor biosynthesis, bacterial virulence factor, bacteria cell division machinery, Bacterial cell wall synthesis, PDF inhibitor, isoprenoid biosynthesis, shikimate synthesis pathway, biofilm synthesis and fatty acid biosynthesis. These new discovery routes have given rise to agents that are in preclinical trials. This review also discusses the alternatives approaches that act bacteria or any approaches that target the host. The most advanced approaches that are on clinical development are phages and other approaches that are on preclinical development are antimicrobial peptides. These alternatives ways may use as adjunctive therapies, which suggest that conventional antibacterial agents are still essential.

在过去的半个世纪里,抗菌剂拯救了许多人的生命,促进了现代医学的发展。耐药性的出现,危及这些挽救生命的治疗的有效性。这清楚地表明,迫切需要新的和改进的抗菌药物,以新的靶点和新的分子结构剂来消除交叉耐药。本文综述了发现新型抗菌药物的可能新途径。目前研究最广泛的新药物靶点是群体感应生物合成、细菌毒力因子、细菌细胞分裂机制、细菌细胞壁合成、PDF抑制剂、类异戊二烯生物合成、shikimate合成途径、生物膜合成和脂肪酸生物合成。这些新的发现途径已经产生了临床前试验的药物。本综述还讨论了作用于细菌或任何针对宿主的方法的替代方法。临床开发中最先进的方法是噬菌体,临床前开发的其他方法是抗菌肽。这些替代方法可以用作辅助治疗,这表明传统的抗菌药物仍然是必不可少的。
{"title":"Novel targets to develop new antibacterial agents and novel alternatives to antibacterial agents","authors":"Tafere Mulaw Belete","doi":"10.1016/j.humic.2019.01.001","DOIUrl":"10.1016/j.humic.2019.01.001","url":null,"abstract":"<div><p>Antibacterial agents have saved many lives and helped the growth of modern medicine over the past half century. The emergence of drug resistance, jeopardizing the effectiveness of these life-saving treatments. This clearly highlights the urgent need for new and improved antibacterial drugs with a novel target and new molecular structure agent to obviate cross-resistance. This paper reviewed the possible new ways to discover novel antibacterial agents. The most widely studied new bacterial targets for novel drug development are quorum sensor biosynthesis, bacterial virulence factor, bacteria cell division machinery, Bacterial cell wall synthesis, PDF inhibitor, isoprenoid biosynthesis, shikimate synthesis pathway, biofilm synthesis and fatty acid biosynthesis. These new discovery routes have given rise to agents that are in preclinical trials. This review also discusses the alternatives approaches that act <em>bacteria</em> or any approaches that target the host. The most advanced approaches that are on clinical development are phages and other approaches that are on preclinical development are antimicrobial peptides. These alternatives ways may use as adjunctive therapies, which suggest that conventional antibacterial agents are still essential.</p></div>","PeriodicalId":37790,"journal":{"name":"Human Microbiome Journal","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.humic.2019.01.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"47988710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 66
期刊
Human Microbiome Journal
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1