首页 > 最新文献

mSystems最新文献

英文 中文
Genomic insights into the evolution of secondary metabolism of Escovopsis and its allies, specialized fungal symbionts of fungus-farming ants. 从基因组学角度看蚁蚕共生真菌(Escovopsis)及其盟友的次级代谢进化。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-21 DOI: 10.1128/msystems.00576-24
Aileen Berasategui, Hassan Salem, Abraham G Moller, Yuliana Christopher, Quimi Vidaurre Montoya, Caitlin Conn, Timothy D Read, Andre Rodrigues, Nadine Ziemert, Nicole Gerardo

The metabolic intimacy of symbiosis often demands the work of specialists. Natural products and defensive secondary metabolites can drive specificity by ensuring infection and propagation across host generations. But in contrast to bacteria, little is known about the diversity and distribution of natural product biosynthetic pathways among fungi and how they evolve to facilitate symbiosis and adaptation to their host environment. In this study, we define the secondary metabolism of Escovopsis and closely related genera, symbionts in the gardens of fungus-farming ants. We ask how the gain and loss of various biosynthetic pathways correspond to divergent lifestyles. Long-read sequencing allowed us to define the chromosomal features of representative Escovopsis strains, revealing highly reduced genomes composed of seven to eight chromosomes. The genomes are highly syntenic with macrosynteny decreasing with increasing phylogenetic distance, while maintaining a high degree of mesosynteny. An ancestral state reconstruction analysis of biosynthetic pathways revealed that, while many secondary metabolites are shared with non-ant-associated Sordariomycetes, 56 pathways are unique to the symbiotic genera. Reflecting adaptation to diverging ant agricultural systems, we observe that the stepwise acquisition of these pathways mirrors the ecological radiations of attine ants and the dynamic recruitment and replacement of their fungal cultivars. As different clades encode characteristic combinations of biosynthetic gene clusters, these delineating profiles provide important insights into the possible mechanisms underlying specificity between these symbionts and their fungal hosts. Collectively, our findings shed light on the evolutionary dynamic nature of secondary metabolism in Escovopsis and its allies, reflecting adaptation of the symbionts to an ancient agricultural system.IMPORTANCEMicrobial symbionts interact with their hosts and competitors through a remarkable array of secondary metabolites and natural products. Here, we highlight the highly streamlined genomic features of attine-associated fungal symbionts. The genomes of Escovopsis species, as well as species from other symbiont genera, many of which are common with the gardens of fungus-growing ants, are defined by seven chromosomes. Despite a high degree of metabolic conservation, we observe some variation in the symbionts' potential to produce secondary metabolites. As the phylogenetic distribution of the encoding biosynthetic gene clusters coincides with attine transitions in agricultural systems, we highlight the likely role of these metabolites in mediating adaptation by a group of highly specialized symbionts.

共生过程中的代谢密切关系往往需要专家来完成。天然产物和防御性次生代谢物可以通过确保宿主的感染和跨代繁殖来驱动特异性。但与细菌相比,人们对真菌天然产物生物合成途径的多样性和分布,以及它们如何进化以促进共生和适应宿主环境知之甚少。在这项研究中,我们确定了蚁蛆属和密切相关属的次生代谢,它们是真菌养殖蚂蚁花园中的共生体。我们想知道各种生物合成途径的增减是如何与不同的生活方式相对应的。通过长读测序,我们确定了具有代表性的 Escovopsis 菌株的染色体特征,发现了由 7 至 8 条染色体组成的高度缩小的基因组。这些基因组高度同源,大同源度随着系统发育距离的增加而降低,同时保持了高度的中同源度。对生物合成途径的祖先状态重建分析表明,虽然许多次级代谢物与非共生的尾柄霉菌共享,但有 56 种途径是共生属所独有的。我们观察到,这些途径的逐步获得反映了蚂蚁的生态辐射及其真菌栽培种的动态招募和替换,这反映了对蚂蚁农业系统分化的适应。由于不同支系编码生物合成基因簇的特征组合,这些划分特征为了解这些共生体与其真菌宿主之间的特异性可能机制提供了重要的启示。总之,我们的发现揭示了 Escovopsis 及其盟友次生代谢的进化动态性质,反映了共生体对古老农业系统的适应。在这里,我们重点介绍了啮齿动物相关真菌共生体高度精简的基因组特征。Escovopsis物种以及其他共生菌属的物种(其中许多与真菌生长蚂蚁的花园很相似)的基因组由七条染色体组成。尽管新陈代谢保持高度一致,但我们观察到共生体产生次级代谢物的潜力存在一些差异。由于编码生物合成基因簇的系统发育分布与农业系统中的蚂蚁过渡相吻合,我们强调了这些代谢物在调解一群高度特化的共生体的适应性方面可能发挥的作用。
{"title":"Genomic insights into the evolution of secondary metabolism of <i>Escovopsis</i> and its allies, specialized fungal symbionts of fungus-farming ants.","authors":"Aileen Berasategui, Hassan Salem, Abraham G Moller, Yuliana Christopher, Quimi Vidaurre Montoya, Caitlin Conn, Timothy D Read, Andre Rodrigues, Nadine Ziemert, Nicole Gerardo","doi":"10.1128/msystems.00576-24","DOIUrl":"10.1128/msystems.00576-24","url":null,"abstract":"<p><p>The metabolic intimacy of symbiosis often demands the work of specialists. Natural products and defensive secondary metabolites can drive specificity by ensuring infection and propagation across host generations. But in contrast to bacteria, little is known about the diversity and distribution of natural product biosynthetic pathways among fungi and how they evolve to facilitate symbiosis and adaptation to their host environment. In this study, we define the secondary metabolism of <i>Escovopsis</i> and closely related genera, symbionts in the gardens of fungus-farming ants. We ask how the gain and loss of various biosynthetic pathways correspond to divergent lifestyles. Long-read sequencing allowed us to define the chromosomal features of representative <i>Escovopsis</i> strains, revealing highly reduced genomes composed of seven to eight chromosomes. The genomes are highly syntenic with macrosynteny decreasing with increasing phylogenetic distance, while maintaining a high degree of mesosynteny. An ancestral state reconstruction analysis of biosynthetic pathways revealed that, while many secondary metabolites are shared with non-ant-associated <i>Sordariomycetes</i>, 56 pathways are unique to the symbiotic genera. Reflecting adaptation to diverging ant agricultural systems, we observe that the stepwise acquisition of these pathways mirrors the ecological radiations of attine ants and the dynamic recruitment and replacement of their fungal cultivars. As different clades encode characteristic combinations of biosynthetic gene clusters, these delineating profiles provide important insights into the possible mechanisms underlying specificity between these symbionts and their fungal hosts. Collectively, our findings shed light on the evolutionary dynamic nature of secondary metabolism in <i>Escovopsis</i> and its allies, reflecting adaptation of the symbionts to an ancient agricultural system.IMPORTANCEMicrobial symbionts interact with their hosts and competitors through a remarkable array of secondary metabolites and natural products. Here, we highlight the highly streamlined genomic features of attine-associated fungal symbionts. The genomes of <i>Escovopsis</i> species, as well as species from other symbiont genera, many of which are common with the gardens of fungus-growing ants, are defined by seven chromosomes. Despite a high degree of metabolic conservation, we observe some variation in the symbionts' potential to produce secondary metabolites. As the phylogenetic distribution of the encoding biosynthetic gene clusters coincides with attine transitions in agricultural systems, we highlight the likely role of these metabolites in mediating adaptation by a group of highly specialized symbionts.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265373/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141432344","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unassigning bacterial species for microbiome studies. 为微生物组研究重新分配细菌种类。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-24 DOI: 10.1128/msystems.00515-24
Ceylan Tanes, Vincent Tu, Scott Daniel, Kyle Bittinger

The method of 16S rRNA marker gene sequencing has fueled microbiome research and continues to be relevant. A perceived weakness of the method is that taxonomic assignments are not possible to make at the rank of species. We show that by working to rule out bacterial or archaeal species membership, we can provide an answer that is more accurate and useful. The Unassigner software operates on 16S rRNA marker gene data and computes a rule-out probability for species membership using a beta-binomial distribution. We demonstrate that our approach is accurate based on full-genome comparisons. Our method is consistent with existing approaches and dramatically improves on them based on the percentage of reads it can associate with a species in a sample. The software is available at https://github.com/PennChopMicrobiomeProgram/unassigner.IMPORTANCEWhile existing methods do not provide reliable species-level assignments for 16S rRNA marker gene data, the Unassigner software solves this problem by ruling out species membership, allowing researchers to reason at the species level.

16S rRNA 标记基因测序法推动了微生物组的研究,并将继续发挥重要作用。人们认为该方法的一个弱点是无法按物种等级进行分类。我们的研究表明,通过排除细菌或古细菌的物种成员资格,我们可以提供更准确、更有用的答案。Unassigner 软件可在 16S rRNA 标记基因数据上运行,并利用贝塔二叉分布计算物种成员资格的排除概率。我们在全基因组比较的基础上证明了我们的方法是准确的。我们的方法与现有的方法一致,而且根据样本中与物种相关联的读数百分比,我们的方法大大提高了现有方法的性能。该软件可从以下网址获取:https://github.com/PennChopMicrobiomeProgram/unassigner.IMPORTANCEWhile 现有方法无法为 16S rRNA 标记基因数据提供可靠的物种级分配,而 Unassigner 软件通过排除物种成员资格解决了这一问题,使研究人员能够在物种级别进行推理。
{"title":"Unassigning bacterial species for microbiome studies.","authors":"Ceylan Tanes, Vincent Tu, Scott Daniel, Kyle Bittinger","doi":"10.1128/msystems.00515-24","DOIUrl":"10.1128/msystems.00515-24","url":null,"abstract":"<p><p>The method of 16S rRNA marker gene sequencing has fueled microbiome research and continues to be relevant. A perceived weakness of the method is that taxonomic assignments are not possible to make at the rank of species. We show that by working to <i>rule out</i> bacterial or archaeal species membership, we can provide an answer that is more accurate and useful. The Unassigner software operates on 16S rRNA marker gene data and computes a rule-out probability for species membership using a beta-binomial distribution. We demonstrate that our approach is accurate based on full-genome comparisons. Our method is consistent with existing approaches and dramatically improves on them based on the percentage of reads it can associate with a species in a sample. The software is available at https://github.com/PennChopMicrobiomeProgram/unassigner.IMPORTANCEWhile existing methods do not provide reliable species-level assignments for 16S rRNA marker gene data, the Unassigner software solves this problem by ruling out species membership, allowing researchers to reason at the species level.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264914/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141443029","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bile acid-gut microbiota imbalance in cholestasis and its long-term effect in mice. 胆汁淤积症中胆汁酸-肠道微生物群失衡及其对小鼠的长期影响
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-27 DOI: 10.1128/msystems.00127-24
Xin Yang, Yuesong Xu, Jie Li, Ximing Ran, Zhihao Gu, Linfeng Song, Lei Zhang, Li Wen, Guang Ji, Ruirui Wang

Cholestasis is a common morbid state that may occur in different phases; however, a comprehensive evaluation of the long-term effect post-recovery is still lacking. In the hepatic cholestasis mouse model, which was induced by a temporary complete blockage of the bile duct, the stasis of bile acids and liver damage typically recovered within a short period. However, we found that the temporary hepatic cholestasis had a long-term effect on gut microbiota dysbiosis, including overgrowth of small intestinal bacteria, decreased diversity of the gut microbiota, and an overall imbalance in its composition accompanied by an elevated inflammation level. Additionally, we observed an increase in Escherichia-Shigella (represented by ASV136078), rich in virulence factors, in both small and large intestines following cholestasis. To confirm the causal role of dysregulated gut microbiota in promoting hepatic inflammation and injury, we conducted gut microbiota transplantation into germ-free mice. We found that recipient mice transplanted with feces from cholestasis mice exhibited liver inflammation, damage, and accumulation of hepatic bile acids. In conclusion, our study demonstrates that cholestasis disrupts the overall load and structural composition of the gut microbiota in mice, and these adverse effects persist after recovery from cholestatic liver injury. This finding suggests the importance of monitoring the structural composition of the gut microbiota in patients with cholestasis and during their recovery.

Importance: Our pre-clinical study using a mouse model of cholestasis underscores that cholestasis not only disrupts the equilibrium and structural configuration of the gut microbiota but also emphasizes the persistence of these adverse effects even after bile stasis restoration. This suggests the need of monitoring and initiating interventions for gut microbiota structural restoration in patients with cholestasis during and after recovery. We believe that our study contributes to novel and better understanding of the intricate interplay among bile acid homeostasis, gut microbiota, and cholestasis-associated complications. Our pre-clinical findings may provide implications for the clinical management of patients with cholestasis.

胆汁淤积症是一种常见的病态,可发生在不同阶段,但目前仍缺乏对恢复后长期影响的全面评估。在肝胆汁淤积小鼠模型中,通过暂时性完全阻断胆管诱导胆汁淤积,胆汁酸淤积和肝损伤通常会在短期内恢复。然而,我们发现暂时性肝胆汁淤积会对肠道微生物群失调产生长期影响,包括小肠细菌过度生长、肠道微生物群多样性降低、肠道微生物群组成整体失衡以及炎症水平升高。此外,我们还观察到,胆汁淤积症发生后,小肠和大肠中富含毒力因子的埃希氏菌(以 ASV136078 为代表)都有所增加。为了证实肠道微生物群失调在促进肝脏炎症和损伤中的因果作用,我们将肠道微生物群移植到无菌小鼠体内。我们发现,移植了胆汁淤积症小鼠粪便的受体小鼠表现出肝脏炎症、损伤和肝胆汁酸积聚。总之,我们的研究表明,胆汁淤积症会破坏小鼠肠道微生物群的整体负荷和结构组成,这些不利影响在胆汁淤积性肝损伤恢复后仍会持续。这一发现表明,在胆汁淤积症患者及其恢复期间监测肠道微生物群的结构组成非常重要:我们利用胆汁淤积症小鼠模型进行的临床前研究强调,胆汁淤积症不仅会破坏肠道微生物群的平衡和结构配置,而且即使在胆汁淤积恢复后,这些不利影响也会持续存在。这表明,有必要在胆汁淤积症患者康复期间和康复后对其肠道微生物群结构的恢复进行监测并采取干预措施。我们相信,我们的研究有助于更好地理解胆汁酸平衡、肠道微生物群和胆汁淤积症相关并发症之间错综复杂的相互作用。我们的临床前研究结果可能会对胆汁淤积症患者的临床治疗产生影响。
{"title":"Bile acid-gut microbiota imbalance in cholestasis and its long-term effect in mice.","authors":"Xin Yang, Yuesong Xu, Jie Li, Ximing Ran, Zhihao Gu, Linfeng Song, Lei Zhang, Li Wen, Guang Ji, Ruirui Wang","doi":"10.1128/msystems.00127-24","DOIUrl":"10.1128/msystems.00127-24","url":null,"abstract":"<p><p>Cholestasis is a common morbid state that may occur in different phases; however, a comprehensive evaluation of the long-term effect post-recovery is still lacking. In the hepatic cholestasis mouse model, which was induced by a temporary complete blockage of the bile duct, the stasis of bile acids and liver damage typically recovered within a short period. However, we found that the temporary hepatic cholestasis had a long-term effect on gut microbiota dysbiosis, including overgrowth of small intestinal bacteria, decreased diversity of the gut microbiota, and an overall imbalance in its composition accompanied by an elevated inflammation level. Additionally, we observed an increase in <i>Escherichia-Shigella</i> (represented by ASV136078), rich in virulence factors, in both small and large intestines following cholestasis. To confirm the causal role of dysregulated gut microbiota in promoting hepatic inflammation and injury, we conducted gut microbiota transplantation into germ-free mice. We found that recipient mice transplanted with feces from cholestasis mice exhibited liver inflammation, damage, and accumulation of hepatic bile acids. In conclusion, our study demonstrates that cholestasis disrupts the overall load and structural composition of the gut microbiota in mice, and these adverse effects persist after recovery from cholestatic liver injury. This finding suggests the importance of monitoring the structural composition of the gut microbiota in patients with cholestasis and during their recovery.</p><p><strong>Importance: </strong>Our pre-clinical study using a mouse model of cholestasis underscores that cholestasis not only disrupts the equilibrium and structural configuration of the gut microbiota but also emphasizes the persistence of these adverse effects even after bile stasis restoration. This suggests the need of monitoring and initiating interventions for gut microbiota structural restoration in patients with cholestasis during and after recovery. We believe that our study contributes to novel and better understanding of the intricate interplay among bile acid homeostasis, gut microbiota, and cholestasis-associated complications. Our pre-clinical findings may provide implications for the clinical management of patients with cholestasis.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265269/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141458182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
zDB: bacterial comparative genomics made easy. zDB:细菌比较基因组学变得简单。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-28 DOI: 10.1128/msystems.00473-24
Bastian Marquis, Trestan Pillonel, Alessia Carrara, Claire Bertelli

The analysis and comparison of genomes rely on different tools for tasks such as annotation, orthology prediction, and phylogenetic inference. Most tools are specialized for a single task, and additional efforts are necessary to integrate and visualize the results. To fill this gap, we developed zDB, an application integrating a Nextflow analysis pipeline and a Python visualization platform built on the Django framework. The application is available on GitHub (https://github.com/metagenlab/zDB) and from the bioconda channel. Starting from annotated Genbank files, zDB identifies orthologs and infers a phylogeny for each orthogroup. A species phylogeny is also constructed from shared single-copy orthologs. The results can be enriched with Pfam protein domain prediction, Cluster of Orthologs Genes and Kyoto Encyclopedia of Genes and Genomes annotations, and Swissprot homologs. The web application allows searching for specific genes or annotations, running Blast queries, and comparing genomic regions and whole genomes. The metabolic capacities of organisms can be compared at either the module or pathway levels. Finally, users can run queries to examine the conservation of specific genes or annotations across a chosen subset of genomes and display the results as a list of genes, Venn diagram, or heatmaps. Those features make zDB useful for both bioinformaticians and researchers more accustomed to laboratory research.IMPORTANCEGenome comparison and analysis rely on many independent tools, leaving to scientists the burden to integrate and visualize their results for interpretation. To alleviate this burden, we have built zDB, a comparative genomics tool that includes both an analysis pipeline and a visualization platform. The analysis pipeline automates gene annotation, orthology prediction, and phylogenetic inference, while the visualization platform allows scientists to easily explore the results in a web browser. Among other features, the interface allows users to visually compare whole genomes and targeted regions, assess the conservation of genes or metabolic pathways, perform Blast searches, or look for specific annotations. Altogether, this tool will be useful for a broad range of applications in comparative studies between two and hundred genomes. Furthermore, it is designed to allow sharing of data sets easily at a local or international scale, thereby supporting exploratory analyses for non-bioinformaticians on the genome of their favorite organisms.

基因组的分析和比较依赖于不同的工具,如注释、选集预测和系统发育推断。大多数工具都是针对单一任务的专用工具,因此需要额外的努力来整合和可视化结果。为了填补这一空白,我们开发了zDB,一个集成了Next-flow分析管道和基于Django框架的Python可视化平台的应用程序。该应用程序可在 GitHub (https://github.com/metagenlab/zDB) 和 bioconda 频道上下载。从注释的 Genbank 文件开始,zDB 可识别直向同源物,并推断出每个直向群的系统发生。还可从共享的单拷贝直向同源物中构建物种系统发育。研究结果可通过 Pfam 蛋白结构域预测、同源物基因簇和京都基因和基因组百科全书注释以及 Swissprot 同源物进行丰富。该网络应用程序允许搜索特定基因或注释,运行 Blast 查询,以及比较基因组区域和整个基因组。生物体的代谢能力可在模块或通路水平上进行比较。最后,用户可以运行查询来检查特定基因或注释在所选基因组子集中的保护情况,并以基因列表、维恩图或热图的形式显示结果。这些功能使 zDB 对生物信息学家和更习惯于实验室研究的研究人员都很有用。重要意义基因组比较和分析依赖于许多独立的工具,这就给科学家们留下了整合和可视化其结果以进行解释的负担。为了减轻这一负担,我们开发了 zDB,这是一款比较基因组学工具,包括分析管道和可视化平台。分析管道可自动进行基因注释、正选预测和系统发育推断,而可视化平台则可让科学家在网络浏览器中轻松探索结果。除其他功能外,该界面还允许用户直观地比较全基因组和目标区域、评估基因或代谢途径的保护情况、执行爆炸搜索或查找特定注释。总之,该工具将在两个或上百个基因组之间的比较研究中发挥广泛的作用。此外,它的设计还允许在本地或国际范围内轻松共享数据集,从而支持非生物信息学家对他们最喜欢的生物的基因组进行探索性分析。
{"title":"zDB: bacterial comparative genomics made easy.","authors":"Bastian Marquis, Trestan Pillonel, Alessia Carrara, Claire Bertelli","doi":"10.1128/msystems.00473-24","DOIUrl":"10.1128/msystems.00473-24","url":null,"abstract":"<p><p>The analysis and comparison of genomes rely on different tools for tasks such as annotation, orthology prediction, and phylogenetic inference. Most tools are specialized for a single task, and additional efforts are necessary to integrate and visualize the results. To fill this gap, we developed zDB, an application integrating a Nextflow analysis pipeline and a Python visualization platform built on the Django framework. The application is available on GitHub (https://github.com/metagenlab/zDB) and from the bioconda channel. Starting from annotated Genbank files, zDB identifies orthologs and infers a phylogeny for each orthogroup. A species phylogeny is also constructed from shared single-copy orthologs. The results can be enriched with Pfam protein domain prediction, Cluster of Orthologs Genes and Kyoto Encyclopedia of Genes and Genomes annotations, and Swissprot homologs. The web application allows searching for specific genes or annotations, running Blast queries, and comparing genomic regions and whole genomes. The metabolic capacities of organisms can be compared at either the module or pathway levels. Finally, users can run queries to examine the conservation of specific genes or annotations across a chosen subset of genomes and display the results as a list of genes, Venn diagram, or heatmaps. Those features make zDB useful for both bioinformaticians and researchers more accustomed to laboratory research.IMPORTANCEGenome comparison and analysis rely on many independent tools, leaving to scientists the burden to integrate and visualize their results for interpretation. To alleviate this burden, we have built zDB, a comparative genomics tool that includes both an analysis pipeline and a visualization platform. The analysis pipeline automates gene annotation, orthology prediction, and phylogenetic inference, while the visualization platform allows scientists to easily explore the results in a web browser. Among other features, the interface allows users to visually compare whole genomes and targeted regions, assess the conservation of genes or metabolic pathways, perform Blast searches, or look for specific annotations. Altogether, this tool will be useful for a broad range of applications in comparative studies between two and hundred genomes. Furthermore, it is designed to allow sharing of data sets easily at a local or international scale, thereby supporting exploratory analyses for non-bioinformaticians on the genome of their favorite organisms.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264898/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469575","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phage SEP1 hijacks Staphylococcus epidermidis stationary cells' metabolism to replicate. 噬菌体 SEP1 劫持表皮葡萄球菌固定细胞的新陈代谢进行复制。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-21 DOI: 10.1128/msystems.00263-24
Maria Daniela Silva, Graça Pinto, Angela França, Joana Azeredo, Luís D R Melo

In nature, bacteria often survive in a stationary state with low metabolic activity. Phages use the metabolic machinery of the host cell to replicate, and, therefore, their efficacy against non-dividing cells is usually limited. Nevertheless, it was previously shown that the Staphylococcus epidermidis phage SEP1 has the remarkable capacity to actively replicate in stationary-phase cells, reducing their numbers. Here, we studied for the first time the transcriptomic profiles of both exponential and stationary cells infected with SEP1 phage using RNA-seq to gain a better understanding of this rare phenomenon. We showed that SEP1 successfully takes over the transcriptional apparatus of both exponential and stationary cells. Infection was, however, delayed in stationary cells, with genes within the gp142-gp154 module putatively implicated in host takeover. S. epidermidis responded to SEP1 infection by upregulating three genes involved in a DNA modification system, with this being observed already 5 min after infection in exponential cells and later in stationary cells. In stationary cells, a significant number of genes involved in translation and RNA metabolic and biosynthetic processes were upregulated after 15 and 30 min of SEP1 infection in comparison with the uninfected control, showing that SEP1 activates metabolic and biosynthetic pathways necessary to its successful replication.IMPORTANCEMost phage-host interaction studies are performed with exponentially growing cells. However, this cell state is not representative of what happens in natural environments. Additionally, most phages fail to replicate in stationary cells. The Staphylococcus epidermidis phage SEP1 is one of the few phages reported to date to be able to infect stationary cells. Here, we unveiled the interaction of SEP1 with its host in both exponential and stationary states of growth at the transcriptomic level. The findings of this study provide valuable insights for a better implementation of phage therapy since phages able to infect stationary cells could be more efficient in the treatment of recalcitrant infections.

在自然界中,细菌通常以新陈代谢活性较低的静止状态生存。噬菌体利用宿主细胞的新陈代谢机制进行复制,因此,它们对非分裂细胞的作用通常有限。不过,以前的研究表明,表皮葡萄球菌噬菌体 SEP1 具有在静止期细胞中积极复制的显著能力,能减少细胞数量。在这里,我们首次利用 RNA-seq 技术研究了感染 SEP1 噬菌体的指数期细胞和静止期细胞的转录组图谱,以更好地了解这一罕见现象。我们发现,SEP1成功地接管了指数细胞和静止细胞的转录装置。不过,静止细胞的感染延迟了,gp142-gp154 模块中的基因可能与宿主接管有关。表皮葡萄球菌对 SEP1 感染的反应是上调涉及 DNA 修饰系统的三个基因,这在指数细胞感染后 5 分钟就能观察到,在静止细胞中则稍晚。在静止细胞中,与未感染的对照组相比,SEP1 感染 15 分钟和 30 分钟后,大量参与翻译和 RNA 代谢及生物合成过程的基因上调,这表明 SEP1 激活了其成功复制所必需的代谢和生物合成途径。然而,这种细胞状态并不代表自然环境中发生的情况。此外,大多数噬菌体无法在静止细胞中复制。表皮葡萄球菌噬菌体 SEP1 是迄今为止报道的能够感染静止细胞的少数噬菌体之一。在这里,我们从转录组水平揭示了 SEP1 在指数生长和静止生长两种状态下与宿主的相互作用。这项研究的发现为更好地实施噬菌体疗法提供了宝贵的见解,因为能够感染静止细胞的噬菌体在治疗顽固性感染时可能会更有效。
{"title":"Phage SEP1 hijacks <i>Staphylococcus epidermidis</i> stationary cells' metabolism to replicate.","authors":"Maria Daniela Silva, Graça Pinto, Angela França, Joana Azeredo, Luís D R Melo","doi":"10.1128/msystems.00263-24","DOIUrl":"10.1128/msystems.00263-24","url":null,"abstract":"<p><p>In nature, bacteria often survive in a stationary state with low metabolic activity. Phages use the metabolic machinery of the host cell to replicate, and, therefore, their efficacy against non-dividing cells is usually limited. Nevertheless, it was previously shown that the <i>Staphylococcus epidermidis</i> phage SEP1 has the remarkable capacity to actively replicate in stationary-phase cells, reducing their numbers. Here, we studied for the first time the transcriptomic profiles of both exponential and stationary cells infected with SEP1 phage using RNA-seq to gain a better understanding of this rare phenomenon. We showed that SEP1 successfully takes over the transcriptional apparatus of both exponential and stationary cells. Infection was, however, delayed in stationary cells, with genes within the <i>gp142-gp154</i> module putatively implicated in host takeover. <i>S. epidermidis</i> responded to SEP1 infection by upregulating three genes involved in a DNA modification system, with this being observed already 5 min after infection in exponential cells and later in stationary cells. In stationary cells, a significant number of genes involved in translation and RNA metabolic and biosynthetic processes were upregulated after 15 and 30 min of SEP1 infection in comparison with the uninfected control, showing that SEP1 activates metabolic and biosynthetic pathways necessary to its successful replication.IMPORTANCEMost phage-host interaction studies are performed with exponentially growing cells. However, this cell state is not representative of what happens in natural environments. Additionally, most phages fail to replicate in stationary cells. The <i>Staphylococcus epidermidis</i> phage SEP1 is one of the few phages reported to date to be able to infect stationary cells. Here, we unveiled the interaction of SEP1 with its host in both exponential and stationary states of growth at the transcriptomic level. The findings of this study provide valuable insights for a better implementation of phage therapy since phages able to infect stationary cells could be more efficient in the treatment of recalcitrant infections.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265418/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141432345","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Microbial diversity, genomics, and phage-host interactions of cyanobacterial harmful algal blooms. 蓝藻有害藻华的微生物多样性、基因组学和噬菌体-宿主相互作用。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-10 DOI: 10.1128/msystems.00709-23
Lauren E Krausfeldt, Elizaveta Shmakova, Hyo Won Lee, Viviana Mazzei, Keith A Loftin, Robert P Smith, Emily Karwacki, P Eric Fortman, Barry H Rosen, Hidetoshi Urakawa, Manoj Dadlani, Rita R Colwell, Jose V Lopez

The occurrence of cyanobacterial harmful algal blooms (cyanoHABs) is related to their physical and chemical environment. However, less is known about their associated microbial interactions and processes. In this study, cyanoHABs were analyzed as a microbial ecosystem, using 1 year of 16S rRNA sequencing and 70 metagenomes collected during the bloom season from Lake Okeechobee (Florida, USA). Biogeographical patterns observed in microbial community composition and function reflected ecological zones distinct in their physical and chemical parameters that resulted in bloom "hotspots" near major lake inflows. Changes in relative abundances of taxa within multiple phyla followed increasing bloom severity. Functional pathways that correlated with increasing bloom severity encoded organic nitrogen and phosphorus utilization, storage of nutrients, exchange of genetic material, phage defense, and protection against oxidative stress, suggesting that microbial interactions may promote cyanoHAB resilience. Cyanobacterial communities were highly diverse, with picocyanobacteria ubiquitous and oftentimes most abundant, especially in the absence of blooms. The identification of novel bloom-forming cyanobacteria and genomic comparisons indicated a functionally diverse cyanobacterial community with differences in its capability to store nitrogen using cyanophycin and to defend against phage using CRISPR and restriction-modification systems. Considering blooms in the context of a microbial ecosystem and their interactions in nature, physiologies and interactions supporting the proliferation and stability of cyanoHABs are proposed, including a role for phage infection of picocyanobacteria. This study displayed the power of "-omics" to reveal important biological processes that could support the effective management and prediction of cyanoHABs.

Importance: Cyanobacterial harmful algal blooms pose a significant threat to aquatic ecosystems and human health. Although physical and chemical conditions in aquatic systems that facilitate bloom development are well studied, there are fundamental gaps in the biological understanding of the microbial ecosystem that makes a cyanobacterial bloom. High-throughput sequencing was used to determine the drivers of cyanobacteria blooms in nature. Multiple functions and interactions important to consider in cyanobacterial bloom ecology were identified. The microbial biodiversity of blooms revealed microbial functions, genomic characteristics, and interactions between cyanobacterial populations that could be involved in bloom stability and more coherently define cyanobacteria blooms. Our results highlight the importance of considering cyanobacterial blooms as a microbial ecosystem to predict, prevent, and mitigate them.

蓝藻有害藻华(cyanoHABs)的发生与其物理和化学环境有关。然而,人们对其相关的微生物相互作用和过程却知之甚少。本研究将蓝藻有害藻华作为一个微生物生态系统进行分析,使用了一年的 16S rRNA 测序和在藻华季节从奥基乔比湖(美国佛罗里达州)收集的 70 个元基因组。在微生物群落组成和功能方面观察到的生物地理模式反映了物理和化学参数不同的生态区,这些生态区在主要入湖口附近形成了水华 "热点"。随着水华严重程度的增加,多个门类中类群的相对丰度也发生了变化。与水华严重程度增加相关的功能途径包括有机氮和磷的利用、营养物质的储存、遗传物质的交换、噬菌体的防御以及对氧化应激的保护,这表明微生物的相互作用可能会促进蓝藻水华的恢复能力。蓝藻群落具有高度的多样性,其中微囊藻无处不在,有时数量最多,尤其是在没有藻华的情况下。新型水华形成蓝藻的鉴定和基因组比较表明,蓝藻群落的功能多种多样,它们利用蓝藻素储存氮的能力以及利用 CRISPR 和限制性修饰系统抵御噬菌体的能力也各不相同。从微生物生态系统及其在自然界中的相互作用的角度来考虑蓝藻水华,提出了支持蓝藻水华增殖和稳定的生理机制和相互作用,包括噬菌体感染皮蓝藻的作用。这项研究展示了 "组学 "在揭示重要生物过程方面的威力,可为有效管理和预测蓝藻有害藻华提供支持:蓝藻有害藻华对水生生态系统和人类健康构成重大威胁。尽管人们对水生系统中促进藻华发展的物理和化学条件进行了深入研究,但对蓝藻藻华产生的微生物生态系统的生物学认识还存在根本性的差距。高通量测序被用来确定自然界中蓝藻藻华的驱动因素。研究发现了蓝藻水华生态学中需要考虑的多种功能和相互作用。藻华的微生物生物多样性揭示了蓝藻种群之间的微生物功能、基因组特征和相互作用,这些可能参与藻华的稳定性,并能更一致地定义蓝藻藻华。我们的研究结果强调了将蓝藻水华视为一个微生物生态系统来预测、预防和缓解蓝藻水华的重要性。
{"title":"Microbial diversity, genomics, and phage-host interactions of cyanobacterial harmful algal blooms.","authors":"Lauren E Krausfeldt, Elizaveta Shmakova, Hyo Won Lee, Viviana Mazzei, Keith A Loftin, Robert P Smith, Emily Karwacki, P Eric Fortman, Barry H Rosen, Hidetoshi Urakawa, Manoj Dadlani, Rita R Colwell, Jose V Lopez","doi":"10.1128/msystems.00709-23","DOIUrl":"10.1128/msystems.00709-23","url":null,"abstract":"<p><p>The occurrence of cyanobacterial harmful algal blooms (cyanoHABs) is related to their physical and chemical environment. However, less is known about their associated microbial interactions and processes. In this study, cyanoHABs were analyzed as a microbial ecosystem, using 1 year of 16S rRNA sequencing and 70 metagenomes collected during the bloom season from Lake Okeechobee (Florida, USA). Biogeographical patterns observed in microbial community composition and function reflected ecological zones distinct in their physical and chemical parameters that resulted in bloom \"hotspots\" near major lake inflows. Changes in relative abundances of taxa within multiple phyla followed increasing bloom severity. Functional pathways that correlated with increasing bloom severity encoded organic nitrogen and phosphorus utilization, storage of nutrients, exchange of genetic material, phage defense, and protection against oxidative stress, suggesting that microbial interactions may promote cyanoHAB resilience. Cyanobacterial communities were highly diverse, with picocyanobacteria ubiquitous and oftentimes most abundant, especially in the absence of blooms. The identification of novel bloom-forming cyanobacteria and genomic comparisons indicated a functionally diverse cyanobacterial community with differences in its capability to store nitrogen using cyanophycin and to defend against phage using CRISPR and restriction-modification systems. Considering blooms in the context of a microbial ecosystem and their interactions in nature, physiologies and interactions supporting the proliferation and stability of cyanoHABs are proposed, including a role for phage infection of picocyanobacteria. This study displayed the power of \"-omics\" to reveal important biological processes that could support the effective management and prediction of cyanoHABs.</p><p><strong>Importance: </strong>Cyanobacterial harmful algal blooms pose a significant threat to aquatic ecosystems and human health. Although physical and chemical conditions in aquatic systems that facilitate bloom development are well studied, there are fundamental gaps in the biological understanding of the microbial ecosystem that makes a cyanobacterial bloom. High-throughput sequencing was used to determine the drivers of cyanobacteria blooms in nature. Multiple functions and interactions important to consider in cyanobacterial bloom ecology were identified. The microbial biodiversity of blooms revealed microbial functions, genomic characteristics, and interactions between cyanobacterial populations that could be involved in bloom stability and more coherently define cyanobacteria blooms. Our results highlight the importance of considering cyanobacterial blooms as a microbial ecosystem to predict, prevent, and mitigate them.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265339/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141296440","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lactobacillus acidophilus protects against Corynebacterium pseudotuberculosis infection by regulating the autophagy of macrophages and maintaining gut microbiota homeostasis in C57BL/6 mice. 嗜酸乳杆菌通过调节巨噬细胞的自噬和维持C57BL/6小鼠肠道微生物群的平衡来防止假结核棒状杆菌感染。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-27 DOI: 10.1128/msystems.00484-24
Dengliang Li, Yuecai Jiang, Zhanding Cui, Mengzhen Ma, Fang Zhu, Guanhua Li, Haoyue Yang, Shaofei Li, Tianliang Zhang, Dekun Chen, Wentao Ma

Corynebacterium pseudotuberculosis (C. p), a facultative intracellular bacterium, is an important zoonotic pathogen that causes abscesses and pyogenic granulomas. The relationship between gut microbiota and host health or diseases has received increasing attention. However, the role of gut microbiota in the process of C. p infection is still unclear. In this study, we established a C. p infection model in C57BL/6 mice and examined the impact of preemptive oral administration Lactobacillus acidophilus (L. acidophilus) on infection. Our findings revealed that C. p infection led to pronounced pathological alterations in the liver and kidneys, characterized by abscess formation, intense inflammatory responses, and bacterial overload. Remarkably, these deleterious effects were greatly relieved by oral administration of L. acidophilus before infection with C. p. Additionally, we further found that during C. p infection, peritoneal macrophages (PMs) of mice orally administered with L. acidophilus accumulated more rapidly at sites of infection. Furthermore, our results showed that PMs from mice with oral L. acidophilus administration showed a stronger C. p clearance effect, and this was mediated by high expression of LC3-II protein. Meanwhile, oral administration of L. acidophilus protected the gut microbiota disorder in C57BL/6 mice caused by C. p infection. In summary, our study demonstrates that oral administration of L. acidophilus confers effective protection against C. p infection in C57BL/6 mice by modulating macrophage autophagy, thereby augmenting bacterial clearance and preserving gut microbiota and function stability. These findings position L. acidophilus as a viable probiotic candidate for the clinical prevention of C. p infection.

Importance: Corynebacterium pseudotuberculosis (C. p) is known to induce a range of chronic diseases in both animals and humans. Currently, clinical treatment for C. p infection mainly relies on antibiotic therapy or surgical intervention. However, excessive use of antibiotics may increase the risk of drug-resistant strains, and the effectiveness of treatment remains unsatisfactory. Furthermore, surgical procedures do not completely eradicate pathogens and can easily cause environmental pollution. Probiotic interventions are receiving increasing attention for improving the body's immune system and maintaining health. In this study, we established a C. p infection model in C57BL/6 mice to explore the impact of Lactobacillus acidophilus during C. p infection. Our results showed that L. acidophilus effectively protected against C. p infection by regulating the autophagy of macrophages and maintaining intestinal microbiota homeostasis. This study may provide a new strategy for the prevention of C. p

假结核棒状杆菌(Corynebacterium pseudotuberculosis,C. p)是一种兼性细胞内细菌,是一种重要的人畜共患病原体,可引起脓肿和化脓性肉芽肿。肠道微生物群与宿主健康或疾病之间的关系日益受到关注。然而,肠道微生物群在 C. p. 感染过程中的作用仍不清楚。在本研究中,我们在 C57BL/6 小鼠中建立了嗜酸乳杆菌感染模型,并研究了预先口服嗜酸乳杆菌对感染的影响。我们的研究结果表明,C. p感染会导致肝脏和肾脏发生明显的病理改变,其特点是脓肿形成、强烈的炎症反应和细菌超载。值得注意的是,在感染 C. p.之前口服嗜酸乳杆菌可大大缓解这些有害影响。此外,我们进一步发现,在 C. p.感染期间,口服嗜酸乳杆菌的小鼠腹腔巨噬细胞(PMs)在感染部位聚集得更快。此外,我们的研究结果表明,口服嗜酸乳杆菌的小鼠腹腔巨噬细胞具有更强的C. p清除效果,而这是由LC3-II蛋白的高表达所介导的。同时,口服嗜酸乳杆菌可保护C57BL/6小鼠因C. p感染而导致的肠道微生物群紊乱。总之,我们的研究表明,口服嗜酸乳杆菌可通过调节巨噬细胞自噬作用有效保护C57BL/6小鼠免受C. p感染,从而提高细菌清除率并保护肠道微生物群和功能稳定性。这些发现将嗜酸乳杆菌定位为临床预防假丝酵母感染的一种可行的候选益生菌:众所周知,假结核棒状杆菌(C. p)会诱发动物和人类的一系列慢性疾病。目前,假丝酵母菌感染的临床治疗主要依靠抗生素治疗或手术干预。然而,过度使用抗生素可能会增加耐药菌株的风险,而且治疗效果仍不尽如人意。此外,外科手术并不能完全根除病原体,而且容易造成环境污染。益生菌干预措施在改善人体免疫系统和维护健康方面正受到越来越多的关注。在本研究中,我们在 C57BL/6 小鼠中建立了梭状芽孢杆菌感染模型,以探讨嗜酸乳杆菌在梭状芽孢杆菌感染过程中的影响。结果表明,嗜酸乳杆菌通过调节巨噬细胞的自噬和维持肠道微生物群的平衡,有效地防止了 C. p 的感染。这项研究可为预防嗜酸乳杆菌感染提供一种新策略。
{"title":"<i>Lactobacillus acidophilus</i> protects against <i>Corynebacterium pseudotuberculosis</i> infection by regulating the autophagy of macrophages and maintaining gut microbiota homeostasis in C57BL/6 mice.","authors":"Dengliang Li, Yuecai Jiang, Zhanding Cui, Mengzhen Ma, Fang Zhu, Guanhua Li, Haoyue Yang, Shaofei Li, Tianliang Zhang, Dekun Chen, Wentao Ma","doi":"10.1128/msystems.00484-24","DOIUrl":"10.1128/msystems.00484-24","url":null,"abstract":"<p><p><i>Corynebacterium pseudotuberculosis</i> (<i>C. p</i>), a facultative intracellular bacterium, is an important zoonotic pathogen that causes abscesses and pyogenic granulomas. The relationship between gut microbiota and host health or diseases has received increasing attention. However, the role of gut microbiota in the process of <i>C. p</i> infection is still unclear. In this study, we established a <i>C. p</i> infection model in C57BL/6 mice and examined the impact of preemptive oral administration <i>Lactobacillus acidophilus</i> (<i>L. acidophilus</i>) on infection. Our findings revealed that <i>C. p</i> infection led to pronounced pathological alterations in the liver and kidneys, characterized by abscess formation, intense inflammatory responses, and bacterial overload. Remarkably, these deleterious effects were greatly relieved by oral administration of <i>L. acidophilus</i> before infection with <i>C. p</i>. Additionally, we further found that during <i>C. p</i> infection, peritoneal macrophages (PMs) of mice orally administered with <i>L. acidophilus</i> accumulated more rapidly at sites of infection. Furthermore, our results showed that PMs from mice with oral <i>L. acidophilus</i> administration showed a stronger <i>C. p</i> clearance effect, and this was mediated by high expression of LC3-II protein. Meanwhile, oral administration of <i>L. acidophilus</i> protected the gut microbiota disorder in C57BL/6 mice caused by <i>C. p</i> infection. In summary, our study demonstrates that oral administration of <i>L. acidophilus</i> confers effective protection against <i>C. p</i> infection in C57BL/6 mice by modulating macrophage autophagy, thereby augmenting bacterial clearance and preserving gut microbiota and function stability. These findings position <i>L. acidophilus</i> as a viable probiotic candidate for the clinical prevention of <i>C. p</i> infection.</p><p><strong>Importance: </strong><i>Corynebacterium pseudotuberculosis</i> (C. <i>p</i>) is known to induce a range of chronic diseases in both animals and humans. Currently, clinical treatment for C. <i>p</i> infection mainly relies on antibiotic therapy or surgical intervention. However, excessive use of antibiotics may increase the risk of drug-resistant strains, and the effectiveness of treatment remains unsatisfactory. Furthermore, surgical procedures do not completely eradicate pathogens and can easily cause environmental pollution. Probiotic interventions are receiving increasing attention for improving the body's immune system and maintaining health. In this study, we established a C. <i>p</i> infection model in C57BL/6 mice to explore the impact of <i>Lactobacillus</i> acidophilus during C. <i>p</i> infection. Our results showed that <i>L. acidophilus</i> effectively protected against C. <i>p</i> infection by regulating the autophagy of macrophages and maintaining intestinal microbiota homeostasis. This study may provide a new strategy for the prevention of C. <i>p</","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141458112","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pangenome comparison of Bacteroides fragilis genomospecies unveils genetic diversity and ecological insights. 脆弱拟杆菌(Bacteroides fragilis)基因组比较揭示了遗传多样性和生态学意义。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-27 DOI: 10.1128/msystems.00516-24
Renee E Oles, Marvic Carrillo Terrazas, Luke R Loomis, Chia-Yun Hsu, Caitlin Tribelhorn, Pedro Belda-Ferre, Allison C Ea, MacKenzie Bryant, Jocelyn A Young, Hannah C Carrow, William J Sandborn, Parambir S Dulai, Mamata Sivagnanam, David Pride, Rob Knight, Hiutung Chu

Bacteroides fragilis is a Gram-negative commensal bacterium commonly found in the human colon, which differentiates into two genomospecies termed divisions I and II. Through a comprehensive collection of 694 B. fragilis whole genome sequences, we identify novel features distinguishing these divisions. Our study reveals a distinct geographic distribution with division I strains predominantly found in North America and division II strains in Asia. Additionally, division II strains are more frequently associated with bloodstream infections, suggesting a distinct pathogenic potential. We report differences between the two divisions in gene abundance related to metabolism, virulence, stress response, and colonization strategies. Notably, division II strains harbor more antimicrobial resistance (AMR) genes than division I strains. These findings offer new insights into the functional roles of division I and II strains, indicating specialized niches within the intestine and potential pathogenic roles in extraintestinal sites.

Importance: Understanding the distinct functions of microbial species in the gut microbiome is crucial for deciphering their impact on human health. Classifying division II strains as Bacteroides fragilis can lead to erroneous associations, as researchers may mistakenly attribute characteristics observed in division II strains to the more extensively studied division I B. fragilis. Our findings underscore the necessity of recognizing these divisions as separate species with distinct functions. We unveil new findings of differential gene prevalence between division I and II strains in genes associated with intestinal colonization and survival strategies, potentially influencing their role as gut commensals and their pathogenicity in extraintestinal sites. Despite the significant niche overlap and colonization patterns between these groups, our study highlights the complex dynamics that govern strain distribution and behavior, emphasizing the need for a nuanced understanding of these microorganisms.

脆弱拟杆菌(Bacteroides fragilis)是一种常见于人类结肠的革兰氏阴性共生菌,分为两个基因组种,分别称为 I 和 II。通过全面收集 694 个脆弱拟杆菌全基因组序列,我们发现了区分这两个分支的新特征。我们的研究揭示了其独特的地理分布,I分部菌株主要分布在北美,II分部菌株主要分布在亚洲。此外,II 部菌株更常见于血液感染,这表明它们具有不同的致病潜力。我们报告了两个分部在新陈代谢、毒力、应激反应和定殖策略方面的基因丰度差异。值得注意的是,II 部菌株比 I 部菌株携带更多抗菌素耐药性(AMR)基因。这些发现为了解 I 部和 II 部菌株的功能作用提供了新的视角,表明了它们在肠道内的特殊生态位以及在肠道外的潜在致病作用:重要意义:了解肠道微生物组中微生物物种的不同功能对于解读它们对人类健康的影响至关重要。将第二分部菌株归类为脆弱拟杆菌可能会导致错误的关联,因为研究人员可能会错误地将在第二分部菌株中观察到的特征归因于研究更为广泛的第一分部脆弱拟杆菌。我们的研究结果强调,有必要将这些分部视为具有不同功能的独立物种。我们揭示了新的发现,即在与肠道定殖和生存策略相关的基因中,I 部和 II 部菌株的基因流行率不同,这可能会影响它们作为肠道共生菌的角色及其在肠道外的致病性。尽管这两类菌株之间存在明显的生态位重叠和定植模式,但我们的研究强调了制约菌株分布和行为的复杂动态,强调了对这些微生物进行细致入微的了解的必要性。
{"title":"Pangenome comparison of <i>Bacteroides fragilis</i> genomospecies unveils genetic diversity and ecological insights.","authors":"Renee E Oles, Marvic Carrillo Terrazas, Luke R Loomis, Chia-Yun Hsu, Caitlin Tribelhorn, Pedro Belda-Ferre, Allison C Ea, MacKenzie Bryant, Jocelyn A Young, Hannah C Carrow, William J Sandborn, Parambir S Dulai, Mamata Sivagnanam, David Pride, Rob Knight, Hiutung Chu","doi":"10.1128/msystems.00516-24","DOIUrl":"10.1128/msystems.00516-24","url":null,"abstract":"<p><p><i>Bacteroides fragilis</i> is a Gram-negative commensal bacterium commonly found in the human colon, which differentiates into two genomospecies termed divisions I and II. Through a comprehensive collection of 694 <i>B. fragilis</i> whole genome sequences, we identify novel features distinguishing these divisions. Our study reveals a distinct geographic distribution with division I strains predominantly found in North America and division II strains in Asia. Additionally, division II strains are more frequently associated with bloodstream infections, suggesting a distinct pathogenic potential. We report differences between the two divisions in gene abundance related to metabolism, virulence, stress response, and colonization strategies. Notably, division II strains harbor more antimicrobial resistance (AMR) genes than division I strains. These findings offer new insights into the functional roles of division I and II strains, indicating specialized niches within the intestine and potential pathogenic roles in extraintestinal sites.</p><p><strong>Importance: </strong>Understanding the distinct functions of microbial species in the gut microbiome is crucial for deciphering their impact on human health. Classifying division II strains as <i>Bacteroides fragilis</i> can lead to erroneous associations, as researchers may mistakenly attribute characteristics observed in division II strains to the more extensively studied division I <i>B. fragilis</i>. Our findings underscore the necessity of recognizing these divisions as separate species with distinct functions. We unveil new findings of differential gene prevalence between division I and II strains in genes associated with intestinal colonization and survival strategies, potentially influencing their role as gut commensals and their pathogenicity in extraintestinal sites. Despite the significant niche overlap and colonization patterns between these groups, our study highlights the complex dynamics that govern strain distribution and behavior, emphasizing the need for a nuanced understanding of these microorganisms.</p>","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265264/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141458187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Real-world bisphenol A exposure not linked to microbiota dynamics in childhood obesity. 现实世界中的双酚 A 暴露与儿童肥胖症的微生物群动态无关。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-07-03 DOI: 10.1128/msystems.00556-24
Robyn L Prueitt, Julie E Goodman
{"title":"Real-world bisphenol A exposure not linked to microbiota dynamics in childhood obesity.","authors":"Robyn L Prueitt, Julie E Goodman","doi":"10.1128/msystems.00556-24","DOIUrl":"10.1128/msystems.00556-24","url":null,"abstract":"","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265450/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492669","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bacillus coagulans restores pathogen-induced intestinal dysfunction via acetate-FFAR2-NF-κB-MLCK-MLC axis in Apostichopus japonicus. 凝结芽孢杆菌通过醋酸-FFAR2-NF-[式中:见正文]B-MLCK-MLC 轴恢复日本钝鳃蝇由病原体诱发的肠道功能障碍。
IF 5 2区 生物学 Q1 MICROBIOLOGY Pub Date : 2024-07-23 Epub Date: 2024-06-28 DOI: 10.1128/msystems.00602-24
Mingshan Song, Shanshan Zhang, Zhen Zhang, Liyuan Guo, Weikang Liang, Chenghua Li, Zhonghua Wang

Skin ulceration syndrome (SUS) is currently the main disease threatening Apostichopus japonicus aquaculture due to its higher mortality rate and infectivity, which is caused by Vibrio splendidus. Our previous studies have demonstrated that SUS is accompanied by intestinal microbiota (IM) dysbiosis, alteration of short-chain fatty acids (SCFAs) content and the damage to the intestinal barrier. However, the mediating effect of IM on intestine dysfunction is largely unknown. Herein, we conducted comprehensive intestinal microbiota transplantation (IMT) to explore the link between IM and SUS development. Furthermore, we isolated and identified a Bacillus coagulans strain with an ability to produce acetic acid from both healthy individual and SUS individual with IM from healthy donors. We found that dysbiotic IM and intestinal barrier function in SUS recipients A. japonicus could be restored by IM from healthy donors. The B. coagulans strain could restore IM community and intestinal barrier function. Consistently, acetate supply also restores intestinal homeostasis of SUS-diseased and V. splendidus-infected A. japonicus. Mechanically, acetate was found to specifically bind to its receptor-free fatty acid receptor 2 (FFAR2) to mediate IM structure community and intestinal barrier function. Knockdown of FFAR2 by transfection of specific FFAR2 siRNA could hamper acetate-mediated intestinal homeostasis in vivo. Furthermore, we confirmed that acetate/FFAR2 could inhibit V. splendidus-activated NF-κB-MLCK-MLC signaling pathway to restore intestinal epithelium integrity and upregulated the expression of ZO-1 and Occludin. Our findings provide the first evidence that B. coagulans restores pathogen-induced intestinal barrier dysfunction via acetate/FFAR2-NF-κB-MLCK-MLC axis, which provides new insights into the control and prevention of SUS outbreak from an ecological perspective.IMPORTANCESkin ulceration syndrome (SUS) as a main disease in Apostichopus japonicus aquaculture has severely restricted the developmental A. japonicus aquaculture industry. Intestinal microbiota (IM) has been studied extensively due to its immunomodulatory properties. Short-chain fatty acids (SCFAs) as an essential signal molecule for microbial regulation of host health also have attracted wide attention. Therefore, it is beneficial to explore the link between IM and SUS for prevention and control of SUS. In the study, the contribution of IM to SUS development has been examined. Additionally, our research further validated the restoration of SCFAs on intestinal barrier dysfunction caused by SUS via isolating SCFAs-producing bacteria. Notably, this restoration might be achieved by inhibition of NF-κB-MLCK-MLC signal pathway, which could be activated by V. splendidus. These findings may have important implications for exploration of the role of IM in SUS occurrence and provide insight into

皮肤溃疡综合征(SUS)是目前威胁日本狎鸥鱼养殖的主要疾病,因其死亡率和感染性较高,由绚丽弧菌引起。我们之前的研究表明,SUS伴随着肠道微生物群(IM)失调、短链脂肪酸(SCFAs)含量的改变和肠道屏障的破坏。然而,IM 对肠道功能障碍的介导作用在很大程度上是未知的。在此,我们进行了全面的肠道微生物群移植(IMT),以探索 IM 与 SUS 发生之间的联系。此外,我们还从健康个体和SUS个体的健康供体中分离并鉴定出了具有产生醋酸能力的凝结芽孢杆菌菌株。我们发现,健康供体的 IM 可以恢复 SUS 受体 A. japonicus 中菌群失调的 IM 和肠道屏障功能。凝结芽孢杆菌菌株可以恢复肠内生物群落和肠道屏障功能。同样,醋酸盐也能恢复 SUS 疾病和白芨感染的日本豚鼠的肠道平衡。在机制上,研究发现醋酸与其无受体的脂肪酸受体 2(FFAR2)特异性结合,以介导 IM 结构群落和肠屏障功能。通过转染特异性 FFAR2 siRNA 来敲除 FFAR2 可阻碍醋酸介导的体内肠道平衡。此外,我们还证实醋酸/FFAR2能抑制白芨激活的NF-κB-MLCK-MLC信号通路,从而恢复肠上皮细胞的完整性,并上调ZO-1和Occludin的表达。我们的研究结果首次证明了凝结芽孢杆菌可通过醋酸酯/FFAR2-NF-κB-MLCK-MLC 轴恢复病原体诱导的肠屏障功能障碍,这为从生态学角度控制和预防 SUS 的爆发提供了新的见解。 重要意义皮肤溃疡综合征(SUS)作为日本狎鸥鱼养殖过程中的一种主要疾病,严重制约了日本狎鸥鱼养殖业的发展。肠道微生物群(IM)因其免疫调节特性而被广泛研究。短链脂肪酸(SCFAs)作为微生物调节宿主健康的重要信号分子也引起了广泛关注。因此,探讨 IM 与 SUS 之间的联系有利于预防和控制 SUS。本研究探讨了 IM 对 SUS 发展的贡献。此外,我们的研究还通过分离产生 SCFAs 的细菌,进一步验证了 SCFAs 对 SUS 引起的肠屏障功能障碍的修复作用。值得注意的是,这种修复作用可能是通过抑制芨芨草菌激活的 NF-κB-MLCK-MLC 信号通路实现的。这些发现可能对探索 IM 在 SUS 发生中的作用具有重要意义,并为 SUS 的治疗提供了启示。
{"title":"<i>Bacillus coagulans</i> restores pathogen-induced intestinal dysfunction via acetate-FFAR2-NF-κB-MLCK-MLC axis in <i>Apostichopus japonicus</i>.","authors":"Mingshan Song, Shanshan Zhang, Zhen Zhang, Liyuan Guo, Weikang Liang, Chenghua Li, Zhonghua Wang","doi":"10.1128/msystems.00602-24","DOIUrl":"10.1128/msystems.00602-24","url":null,"abstract":"<p><p>Skin ulceration syndrome (SUS) is currently the main disease threatening <i>Apostichopus japonicus</i> aquaculture due to its higher mortality rate and infectivity, which is caused by <i>Vibrio splendidus</i>. Our previous studies have demonstrated that SUS is accompanied by intestinal microbiota (IM) dysbiosis, alteration of short-chain fatty acids (SCFAs) content and the damage to the intestinal barrier. However, the mediating effect of IM on intestine dysfunction is largely unknown. Herein, we conducted comprehensive intestinal microbiota transplantation (IMT) to explore the link between IM and SUS development. Furthermore, we isolated and identified a <i>Bacillus coagulans</i> strain with an ability to produce acetic acid from both healthy individual and SUS individual with IM from healthy donors. We found that dysbiotic IM and intestinal barrier function in SUS recipients <i>A. japonicus</i> could be restored by IM from healthy donors. The <i>B. coagulans</i> strain could restore IM community and intestinal barrier function. Consistently, acetate supply also restores intestinal homeostasis of SUS-diseased and <i>V. splendidus</i>-infected <i>A. japonicus</i>. Mechanically, acetate was found to specifically bind to its receptor-free fatty acid receptor 2 (FFAR2) to mediate IM structure community and intestinal barrier function. Knockdown of FFAR2 by transfection of specific FFAR2 siRNA could hamper acetate-mediated intestinal homeostasis <i>in vivo</i>. Furthermore, we confirmed that acetate/FFAR2 could inhibit <i>V. splendidus</i>-activated NF-κB-MLCK-MLC signaling pathway to restore intestinal epithelium integrity and upregulated the expression of ZO-1 and Occludin. Our findings provide the first evidence that <i>B. coagulans</i> restores pathogen-induced intestinal barrier dysfunction via acetate/FFAR2-NF-κB-MLCK-MLC axis, which provides new insights into the control and prevention of SUS outbreak from an ecological perspective.IMPORTANCESkin ulceration syndrome (SUS) as a main disease in <i>Apostichopus japonicus</i> aquaculture has severely restricted the developmental <i>A. japonicus</i> aquaculture industry. Intestinal microbiota (IM) has been studied extensively due to its immunomodulatory properties. Short-chain fatty acids (SCFAs) as an essential signal molecule for microbial regulation of host health also have attracted wide attention. Therefore, it is beneficial to explore the link between IM and SUS for prevention and control of SUS. In the study, the contribution of IM to SUS development has been examined. Additionally, our research further validated the restoration of SCFAs on intestinal barrier dysfunction caused by SUS via isolating SCFAs-producing bacteria. Notably, this restoration might be achieved by inhibition of NF-κB-MLCK-MLC signal pathway, which could be activated by <i>V. splendidus</i>. These findings may have important implications for exploration of the role of IM in SUS occurrence and provide insight into ","PeriodicalId":18819,"journal":{"name":"mSystems","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11265352/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
mSystems
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
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