首页 > 最新文献

npj Biofilms and Microbiomes最新文献

英文 中文
Lactobacillus plantarum L168 improves hyperoxia-induced pulmonary inflammation and hypoalveolarization in a rat model of bronchopulmonary dysplasia. 植物乳杆菌 L168 可改善支气管肺发育不良大鼠模型中高氧诱导的肺部炎症和肺泡扩张不足。
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-29 DOI: 10.1038/s41522-024-00504-w
Xian Shen, Zhaocong Yang, Qiang Wang, Xu Chen, Qihui Zhu, Zhi Liu, Nishant Patel, Xingyin Liu, Xuming Mo

Alteration of gut microbiota can affect chronic lung diseases, such as asthma and chronic obstructive pulmonary disease, through abnormal immune and inflammatory responses. Previous studies have shown a feasible connection between gut microbiota and bronchopulmonary dysplasia (BPD) in preterm infants. However, whether BPD can be ameliorated by restoring the gut microbiota remains unclear. In preterm infants with BPD, we found variance in the diversity and structure of gut microbiota. Similarly, BPD rats showed gut dysbiosis, characterized by a deficiency of Lactobacillus, which was abundant in normal rats. We therefore explored the effect and potential mechanism of action of a probiotic strain, Lactobacillus plantarum L168, in improving BPD. The BPD rats were treated with L. plantarum L168 by gavage for 2 weeks, and the effect was evaluated by lung histopathology, lung function, and serum inflammatory markers. Subsequently, we observed reduced lung injury and improved lung development in BPD rats exposed to L. plantarum L168. Further evaluation revealed that L. plantarum L168 improved intestinal permeability in BPD rats. Serum metabolomics showed altered inflammation-associated metabolites following L. plantarum L168 intervention, notably a marked increase in anti-inflammatory metabolites. In agreement with the metabolites analysis, RNA-seq analysis of the intestine and lung showed that inflammation and immune-related genes were down-regulated. Based on the information from RNA-seq, we validated that L. plantarum L168 might improve BPD relating to down-regulation of TLR4 /NF-κB /CCL4 pathway. Together, our findings suggest the potential of L. plantarum L168 to provide probiotic-based therapeutic strategies for BPD.

肠道微生物群的改变可通过异常的免疫和炎症反应影响慢性肺部疾病,如哮喘和慢性阻塞性肺病。先前的研究表明,肠道微生物群与早产儿支气管肺发育不良(BPD)之间存在可行的联系。然而,能否通过恢复肠道微生物群来改善早产儿支气管肺发育不良仍不清楚。在患有 BPD 的早产儿中,我们发现肠道微生物群的多样性和结构存在差异。同样,BPD 大鼠也表现出肠道菌群失调,其特征是缺乏乳酸杆菌,而正常大鼠体内却有大量乳酸杆菌。因此,我们探索了益生菌株植物乳杆菌 L168 在改善 BPD 方面的效果和潜在作用机制。通过灌胃植物乳杆菌 L168 治疗 BPD 大鼠 2 周,并通过肺组织病理学、肺功能和血清炎症标志物评估其效果。随后,我们观察到暴露于植物乳杆菌 L168 的 BPD 大鼠肺损伤减轻,肺发育得到改善。进一步的评估显示,植物乳杆菌 L168 改善了 BPD 大鼠的肠道通透性。血清代谢组学显示,在植物乳杆菌 L168 的干预下,炎症相关代谢物发生了改变,尤其是抗炎代谢物明显增加。与代谢物分析一致,肠道和肺部的 RNA-seq 分析表明,炎症和免疫相关基因被下调。根据 RNA-seq 的信息,我们验证了植物乳杆菌 L168 可改善 BPD,这与 TLR4 /NF-κB /CCL4 通路的下调有关。总之,我们的研究结果表明,植物乳杆菌 L168 有可能为 BPD 提供基于益生菌的治疗策略。
{"title":"Lactobacillus plantarum L168 improves hyperoxia-induced pulmonary inflammation and hypoalveolarization in a rat model of bronchopulmonary dysplasia.","authors":"Xian Shen, Zhaocong Yang, Qiang Wang, Xu Chen, Qihui Zhu, Zhi Liu, Nishant Patel, Xingyin Liu, Xuming Mo","doi":"10.1038/s41522-024-00504-w","DOIUrl":"10.1038/s41522-024-00504-w","url":null,"abstract":"<p><p>Alteration of gut microbiota can affect chronic lung diseases, such as asthma and chronic obstructive pulmonary disease, through abnormal immune and inflammatory responses. Previous studies have shown a feasible connection between gut microbiota and bronchopulmonary dysplasia (BPD) in preterm infants. However, whether BPD can be ameliorated by restoring the gut microbiota remains unclear. In preterm infants with BPD, we found variance in the diversity and structure of gut microbiota. Similarly, BPD rats showed gut dysbiosis, characterized by a deficiency of Lactobacillus, which was abundant in normal rats. We therefore explored the effect and potential mechanism of action of a probiotic strain, Lactobacillus plantarum L168, in improving BPD. The BPD rats were treated with L. plantarum L168 by gavage for 2 weeks, and the effect was evaluated by lung histopathology, lung function, and serum inflammatory markers. Subsequently, we observed reduced lung injury and improved lung development in BPD rats exposed to L. plantarum L168. Further evaluation revealed that L. plantarum L168 improved intestinal permeability in BPD rats. Serum metabolomics showed altered inflammation-associated metabolites following L. plantarum L168 intervention, notably a marked increase in anti-inflammatory metabolites. In agreement with the metabolites analysis, RNA-seq analysis of the intestine and lung showed that inflammation and immune-related genes were down-regulated. Based on the information from RNA-seq, we validated that L. plantarum L168 might improve BPD relating to down-regulation of TLR4 /NF-κB /CCL4 pathway. Together, our findings suggest the potential of L. plantarum L168 to provide probiotic-based therapeutic strategies for BPD.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10980738/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140326923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
An immune-competent human gut microphysiological system enables inflammation-modulation by Faecalibacterium prausnitzii. 具有免疫能力的人体肠道微生理系统可通过普氏粪杆菌调节炎症。
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-29 DOI: 10.1038/s41522-024-00501-z
Jianbo Zhang, Yu-Ja Huang, Martin Trapecar, Charles Wright, Kirsten Schneider, John Kemmitt, Victor Hernandez-Gordillo, Jun Young Yoon, Mathilde Poyet, Eric J Alm, David T Breault, David L Trumper, Linda G Griffith

Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we established a gut epithelium-microbe-immune (GuMI) microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), and CD4+ naive T cells circulating underneath the colonic epithelium. In GuMI-APC condition, multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines secreted into both apical and basolateral compartments compared to GuMI condition that lacks APC. In GuMI-APC with F. prausnitzii (GuMI-APC-FP), F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, without a significant effect on cytokine secretion, compared to the GuMI-APC without bacteria (GuMI-APC-NB). In contrast, in the presence of CD4+ naive T cells (GuMI-APCT-FP), TLR1, IFNA1, and IDO1 transcription levels decreased with a simultaneous increase in F. prausnitzii-induced secretion of pro-inflammatory cytokines (e.g., IL8) compared to GuMI-APC-FP that lacks T cells. These results highlight the contribution of individual innate immune cells in regulating the immune response triggered by the gut commensal F. prausnitzii. The integration of defined populations of immune cells in the gut microphysiological system demonstrated the usefulness of GuMI physiomimetic platform to study microbe-epithelial-immune interactions in healthy and disease conditions.

微生物与人体肠道上皮细胞和免疫细胞之间的相互作用对肠道健康至关重要。然而,目前还没有一种系统可以将人类先天性免疫细胞与上皮细胞和不耐受氧气的共生微生物长期共培养,这阻碍了人们以可控的方式了解微生物与免疫之间的相互作用。在这里,我们建立了一个肠道上皮-微生物-免疫(GuMI)微生理系统,以维持Faecalibacterium prausnitzii/Faecalibacterium duncaniae与结肠上皮、抗原递呈细胞(APCs,这里指树突状细胞和巨噬细胞)以及在结肠上皮下循环的CD4+幼稚T细胞长期持续共培养。在 GuMI-APC 条件下,多重细胞因子检测表明,与缺乏 APC 的 GuMI 条件相比,APC 促使分泌到顶端和基底侧的细胞因子和趋化因子水平升高。与不含细菌的 GuMI-APC (GuMI-APC-NB)相比,在含 F. prausnitzii 的 GuMI-APC (GuMI-APC-FP)中,F. prausnitzii 增加了结肠上皮细胞中促炎基因(如 toll-like receptor 1 (TLR1) 和 interferon alpha 1 (IFNA1))的转录,但对细胞因子的分泌没有显著影响。相反,与缺乏 T 细胞的 GuMI-APC-FP 相比,在有 CD4+ 天真 T 细胞(GuMI-APCT-FP)存在的情况下,TLR1、IFNA1 和 IDO1 的转录水平降低,同时 F. prausnitzi 诱导的促炎细胞因子(如 IL8)分泌增加。这些结果凸显了个体先天性免疫细胞在调节肠道寄生虫 F. prausnitzii 引发的免疫反应中的作用。在肠道微生理系统中整合确定的免疫细胞群证明了 GuMI 仿生平台在研究健康和疾病条件下微生物-上皮-免疫相互作用方面的实用性。
{"title":"An immune-competent human gut microphysiological system enables inflammation-modulation by Faecalibacterium prausnitzii.","authors":"Jianbo Zhang, Yu-Ja Huang, Martin Trapecar, Charles Wright, Kirsten Schneider, John Kemmitt, Victor Hernandez-Gordillo, Jun Young Yoon, Mathilde Poyet, Eric J Alm, David T Breault, David L Trumper, Linda G Griffith","doi":"10.1038/s41522-024-00501-z","DOIUrl":"10.1038/s41522-024-00501-z","url":null,"abstract":"<p><p>Crosstalk of microbes with human gut epithelia and immune cells is crucial for gut health. However, there is no existing system for a long-term co-culture of human innate immune cells with epithelium and oxygen-intolerant commensal microbes, hindering the understanding of microbe-immune interactions in a controlled manner. Here, we established a gut epithelium-microbe-immune (GuMI) microphysiological system to maintain the long-term continuous co-culture of Faecalibacterium prausnitzii/Faecalibacterium duncaniae with colonic epithelium, antigen-presenting cells (APCs, herein dendritic cells and macrophages), and CD4<sup>+</sup> naive T cells circulating underneath the colonic epithelium. In GuMI-APC condition, multiplex cytokine assays suggested that APCs contribute to the elevated level of cytokines and chemokines secreted into both apical and basolateral compartments compared to GuMI condition that lacks APC. In GuMI-APC with F. prausnitzii (GuMI-APC-FP), F. prausnitzii increased the transcription of pro-inflammatory genes such as toll-like receptor 1 (TLR1) and interferon alpha 1 (IFNA1) in the colonic epithelium, without a significant effect on cytokine secretion, compared to the GuMI-APC without bacteria (GuMI-APC-NB). In contrast, in the presence of CD4<sup>+</sup> naive T cells (GuMI-APCT-FP), TLR1, IFNA1, and IDO1 transcription levels decreased with a simultaneous increase in F. prausnitzii-induced secretion of pro-inflammatory cytokines (e.g., IL8) compared to GuMI-APC-FP that lacks T cells. These results highlight the contribution of individual innate immune cells in regulating the immune response triggered by the gut commensal F. prausnitzii. The integration of defined populations of immune cells in the gut microphysiological system demonstrated the usefulness of GuMI physiomimetic platform to study microbe-epithelial-immune interactions in healthy and disease conditions.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10980819/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140326921","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Disentangling direct vs indirect effects of microbiome manipulations in a habitat-forming marine holobiont. 在生境形成的海洋全生物体中区分微生物组操作的直接和间接影响。
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-29 DOI: 10.1038/s41522-024-00503-x
Alexander Harry McGrath, Kimberley Lema, Suhelen Egan, Georgina Wood, Sebastian Vadillo Gonzalez, Staffan Kjelleberg, Peter D Steinberg, Ezequiel M Marzinelli

Host-associated microbiota are critical for eukaryotic host functioning, to the extent that hosts and their associated microbial communities are often considered "holobionts". Most studies of holobionts have focused on descriptive approaches or have used model systems, usually in the laboratory, to understand host-microbiome interactions. To advance our understanding of host-microbiota interactions and their wider ecological impacts, we need experimental frameworks that can explore causation in non-model hosts, which often have highly diverse microbiota, and in their natural ecological setting (i.e. in the field). We used a dominant habitat-forming seaweed, Hormosira banksii, to explore these issues and to experimentally test host-microbiota interactions in a non-model holobiont. The experimental protocols were aimed at trying to disentangle microbially mediated effects on hosts from direct effects on hosts associated with the methods employed to manipulate host-microbiota. This was done by disrupting the microbiome, either through removal/disruption using a combination of antimicrobial treatments, or additions of specific taxa via inoculations, or a combination of thew two. The experiments were done in mesocosms and in the field. Three different antibiotic treatments were used to disrupt seaweed-associated microbiota to test whether disturbances of microbiota, particularly bacteria, would negatively affect host performance. Responses of bacteria to these disturbances were complex and differed substantially among treatments, with some antibacterial treatments having little discernible effect. However, the temporal sequence of responses antibiotic treatments, changes in bacterial diversity and subsequent decreases in host performance, strongly suggested an effect of the microbiota on host performance in some treatments, as opposed to direct effects of the antibiotics. To further test these effects, we used 16S-rRNA-gene sequencing to identify bacterial taxa that were either correlated, or uncorrelated, with poor host performance following antibiotic treatment. These were then isolated and used in inoculation experiments, independently or in combination with the previously used antibiotic treatments. Negative effects on host performance were strongest where specific microbial antimicrobials treatments were combined with inoculations of strains that were correlated with poor host performance. For these treatments, negative host effects persisted the entire experimental period (12 days), even though treatments were only applied at the beginning of the experiment. Host performance recovered in all other treatments. These experiments provide a framework for exploring causation and disentangling microbially mediated vs. direct effects on hosts for ecologically important, non-model holobionts in the field. This should allow for better predictions of how these systems will respond to, and potentially mitigate, environmental disturbances in their nat

宿主相关微生物群对真核生物宿主的功能至关重要,因此宿主及其相关微生物群落通常被视为 "整体生物"。对整体生物的大多数研究都侧重于描述性方法,或使用模型系统(通常是在实验室中)来了解宿主与微生物群的相互作用。为了增进我们对宿主-微生物群相互作用及其更广泛生态影响的了解,我们需要能够在非模型宿主(通常具有高度多样化的微生物群)的自然生态环境(即野外)中探索因果关系的实验框架。我们利用一种主要的生境形成海藻 Hormosira banksii 来探讨这些问题,并在非模型整体生物体中对宿主与微生物群的相互作用进行实验测试。实验方案旨在尝试将微生物介导的对宿主的影响与宿主-微生物区系操纵方法对宿主的直接影响区分开来。具体做法是破坏微生物群,或通过抗微生物处理组合去除/破坏微生物群,或通过接种添加特定类群,或两者结合。实验在中置培养箱和野外进行。实验采用了三种不同的抗生素处理方法来破坏海藻相关的微生物群,以检验微生物群(尤其是细菌)的干扰是否会对宿主的表现产生负面影响。细菌对这些干扰的反应很复杂,不同处理之间差异很大,有些抗菌处理几乎没有明显的影响。然而,抗生素处理、细菌多样性变化和宿主表现随之下降的时间顺序强烈表明,在某些处理中,微生物群对宿主表现有影响,而不是抗生素的直接影响。为了进一步检验这些影响,我们使用 16S-rRNA 基因测序来确定与抗生素治疗后宿主表现不佳相关或不相关的细菌类群。然后,我们分离出这些细菌类群,并在接种实验中将其单独或与先前使用的抗生素处理方法结合使用。当特定微生物抗菌剂处理与接种与宿主表现不佳相关的菌株相结合时,对宿主表现的负面影响最大。在这些处理中,尽管处理只是在实验开始时进行的,但对宿主的负面影响却持续了整个实验期(12 天)。在所有其他处理中,寄主表现都有所恢复。这些实验提供了一个框架,用于探索因果关系,并区分微生物介导与直接影响宿主的关系,以研究生态学上重要的非模式整体生物。这将有助于更好地预测这些系统在自然环境中如何应对并减轻环境干扰。
{"title":"Disentangling direct vs indirect effects of microbiome manipulations in a habitat-forming marine holobiont.","authors":"Alexander Harry McGrath, Kimberley Lema, Suhelen Egan, Georgina Wood, Sebastian Vadillo Gonzalez, Staffan Kjelleberg, Peter D Steinberg, Ezequiel M Marzinelli","doi":"10.1038/s41522-024-00503-x","DOIUrl":"10.1038/s41522-024-00503-x","url":null,"abstract":"<p><p>Host-associated microbiota are critical for eukaryotic host functioning, to the extent that hosts and their associated microbial communities are often considered \"holobionts\". Most studies of holobionts have focused on descriptive approaches or have used model systems, usually in the laboratory, to understand host-microbiome interactions. To advance our understanding of host-microbiota interactions and their wider ecological impacts, we need experimental frameworks that can explore causation in non-model hosts, which often have highly diverse microbiota, and in their natural ecological setting (i.e. in the field). We used a dominant habitat-forming seaweed, Hormosira banksii, to explore these issues and to experimentally test host-microbiota interactions in a non-model holobiont. The experimental protocols were aimed at trying to disentangle microbially mediated effects on hosts from direct effects on hosts associated with the methods employed to manipulate host-microbiota. This was done by disrupting the microbiome, either through removal/disruption using a combination of antimicrobial treatments, or additions of specific taxa via inoculations, or a combination of thew two. The experiments were done in mesocosms and in the field. Three different antibiotic treatments were used to disrupt seaweed-associated microbiota to test whether disturbances of microbiota, particularly bacteria, would negatively affect host performance. Responses of bacteria to these disturbances were complex and differed substantially among treatments, with some antibacterial treatments having little discernible effect. However, the temporal sequence of responses antibiotic treatments, changes in bacterial diversity and subsequent decreases in host performance, strongly suggested an effect of the microbiota on host performance in some treatments, as opposed to direct effects of the antibiotics. To further test these effects, we used 16S-rRNA-gene sequencing to identify bacterial taxa that were either correlated, or uncorrelated, with poor host performance following antibiotic treatment. These were then isolated and used in inoculation experiments, independently or in combination with the previously used antibiotic treatments. Negative effects on host performance were strongest where specific microbial antimicrobials treatments were combined with inoculations of strains that were correlated with poor host performance. For these treatments, negative host effects persisted the entire experimental period (12 days), even though treatments were only applied at the beginning of the experiment. Host performance recovered in all other treatments. These experiments provide a framework for exploring causation and disentangling microbially mediated vs. direct effects on hosts for ecologically important, non-model holobionts in the field. This should allow for better predictions of how these systems will respond to, and potentially mitigate, environmental disturbances in their nat","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10980776/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140326922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
A genetic screen identifies a role for oprF in Pseudomonas aeruginosa biofilm stimulation by subinhibitory antibiotics. 基因筛选确定了oprF在铜绿假单胞菌生物膜受亚抑制性抗生素刺激过程中的作用。
IF 7.8 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-03-23 DOI: 10.1038/s41522-024-00496-7
Luke N Yaeger, Michael R M Ranieri, Jessica Chee, Sawyer Karabelas-Pittman, Madeleine Rudolph, Alessio M Giovannoni, Hanjeong Harvey, Lori L Burrows

Biofilms are surface-associated communities of bacteria that grow in a self-produced matrix of polysaccharides, proteins, and extracellular DNA (eDNA). Sub-minimal inhibitory concentrations (sub-MIC) of antibiotics induce biofilm formation, potentially as a defensive response to antibiotic stress. However, the mechanisms behind sub-MIC antibiotic-induced biofilm formation are unclear. We show that treatment of Pseudomonas aeruginosa with multiple classes of sub-MIC antibiotics with distinct targets induces biofilm formation. Further, addition of exogenous eDNA or cell lysate failed to increase biofilm formation to the same extent as antibiotics, suggesting that the release of cellular contents by antibiotic-driven bacteriolysis is insufficient. Using a genetic screen for stimulation-deficient mutants, we identified the outer membrane porin OprF and the ECF sigma factor SigX as important. Similarly, loss of OmpA - the Escherichia coli OprF homolog - prevented sub-MIC antibiotic stimulation of E. coli biofilms. Our screen also identified the periplasmic disulfide bond-forming enzyme DsbA and a predicted cyclic-di-GMP phosphodiesterase encoded by PA2200 as essential for biofilm stimulation. The phosphodiesterase activity of PA2200 is likely controlled by a disulfide bond in its regulatory domain, and folding of OprF is influenced by disulfide bond formation, connecting the mutant phenotypes. Addition of reducing agent dithiothreitol prevented sub-MIC antibiotic biofilm stimulation. Finally, activation of a c-di-GMP-responsive promoter follows treatment with sub-MIC antibiotics in the wild-type but not an oprF mutant. Together, these results show that antibiotic-induced biofilm formation is likely driven by a signaling pathway that translates changes in periplasmic redox state into elevated biofilm formation through increases in c-di-GMP.

生物膜是由细菌组成的表面相关群落,它们在由多糖、蛋白质和细胞外 DNA(eDNA)组成的自产基质中生长。亚微量抑制浓度(sub-MIC)的抗生素会诱导生物膜的形成,这可能是对抗生素压力的一种防御性反应。然而,亚微量抑制浓度抗生素诱导生物膜形成的机制尚不清楚。我们的研究表明,用具有不同靶点的多种亚微量抗生素处理铜绿假单胞菌可诱导生物膜形成。此外,添加外源 eDNA 或细胞裂解液也不能在与抗生素相同的程度上增加生物膜的形成,这表明抗生素驱动的细菌溶解不足以释放细胞内容物。通过对刺激缺陷突变体的基因筛选,我们发现外膜孔蛋白 OprF 和 ECF 西格玛因子 SigX 是重要的突变体。同样,大肠杆菌 OprF 同源物 OmpA 的缺失也阻止了亚微克级抗生素对大肠杆菌生物膜的刺激。我们的筛选还发现,外质二硫键形成酶 DsbA 和 PA2200 编码的预测环二-GMP 磷酸二酯酶对生物膜刺激至关重要。PA2200 的磷酸二酯酶活性可能受其调节结构域中的二硫键控制,而 OprF 的折叠则受二硫键形成的影响,这两者将突变体表型联系在一起。添加还原剂二硫苏糖醇可阻止亚微克级抗生素对生物膜的刺激。最后,野生型而非oprF突变体在接受亚微克抗生素处理后,c-di-GMP响应启动子会被激活。这些结果表明,抗生素诱导的生物膜形成可能是由一种信号通路驱动的,这种信号通路通过增加 c-di-GMP 将周质氧化还原状态的变化转化为生物膜形成的增加。
{"title":"A genetic screen identifies a role for oprF in Pseudomonas aeruginosa biofilm stimulation by subinhibitory antibiotics.","authors":"Luke N Yaeger, Michael R M Ranieri, Jessica Chee, Sawyer Karabelas-Pittman, Madeleine Rudolph, Alessio M Giovannoni, Hanjeong Harvey, Lori L Burrows","doi":"10.1038/s41522-024-00496-7","DOIUrl":"10.1038/s41522-024-00496-7","url":null,"abstract":"<p><p>Biofilms are surface-associated communities of bacteria that grow in a self-produced matrix of polysaccharides, proteins, and extracellular DNA (eDNA). Sub-minimal inhibitory concentrations (sub-MIC) of antibiotics induce biofilm formation, potentially as a defensive response to antibiotic stress. However, the mechanisms behind sub-MIC antibiotic-induced biofilm formation are unclear. We show that treatment of Pseudomonas aeruginosa with multiple classes of sub-MIC antibiotics with distinct targets induces biofilm formation. Further, addition of exogenous eDNA or cell lysate failed to increase biofilm formation to the same extent as antibiotics, suggesting that the release of cellular contents by antibiotic-driven bacteriolysis is insufficient. Using a genetic screen for stimulation-deficient mutants, we identified the outer membrane porin OprF and the ECF sigma factor SigX as important. Similarly, loss of OmpA - the Escherichia coli OprF homolog - prevented sub-MIC antibiotic stimulation of E. coli biofilms. Our screen also identified the periplasmic disulfide bond-forming enzyme DsbA and a predicted cyclic-di-GMP phosphodiesterase encoded by PA2200 as essential for biofilm stimulation. The phosphodiesterase activity of PA2200 is likely controlled by a disulfide bond in its regulatory domain, and folding of OprF is influenced by disulfide bond formation, connecting the mutant phenotypes. Addition of reducing agent dithiothreitol prevented sub-MIC antibiotic biofilm stimulation. Finally, activation of a c-di-GMP-responsive promoter follows treatment with sub-MIC antibiotics in the wild-type but not an oprF mutant. Together, these results show that antibiotic-induced biofilm formation is likely driven by a signaling pathway that translates changes in periplasmic redox state into elevated biofilm formation through increases in c-di-GMP.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":7.8,"publicationDate":"2024-03-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10960818/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140194294","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Promastigote EPS secretion and haptomonad biofilm formation as evolutionary adaptations of trypanosomatid parasites for colonizing honeybee hosts. 原体EPS分泌和钩端螺旋体生物膜形成是锥虫寄生体在蜜蜂宿主上定植的进化适应。
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-21 DOI: 10.1038/s41522-024-00492-x
Jéssica Carreira de Paula, Pedro García Olmedo, Tamara Gómez-Moracho, María Buendía-Abad, Mariano Higes, Raquel Martín-Hernández, Antonio Osuna, Luis Miguel de Pablos

Bees are major pollinators involved in the maintenance of all terrestrial ecosystems. Biotic and abiotic factors placing these insects at risk is a research priority for ecological and agricultural sustainability. Parasites are one of the key players of this global decline and the study of their mechanisms of action is essential to control honeybee colony losses. Trypanosomatid parasites and particularly the Lotmaria passim are widely spread in honeybees, however their lifestyle is poorly understood. In this work, we show how these parasites are able to differentiate into a new parasitic lifestyle: the trypanosomatid biofilms. Using different microscopic techniques, we demonstrated that the secretion of Extracellular Polymeric Substances by free-swimming unicellular promastigote forms is a prerequisite for the generation and adherence of multicellular biofilms to solid surfaces in vitro and in vivo. Moreover, compared to human-infective trypanosomatid parasites our study shows how trypanosomatid parasites of honeybees increases their resistance and thus resilience to drastic changes in environmental conditions such as ultralow temperatures and hypoosmotic shock, which would explain their success thriving within or outside their hosts. These results set up the basis for the understanding of the success of this group of parasites in nature and to unveil the impact of such pathogens in honeybees, a keystones species in most terrestrial ecosystems.

蜜蜂是维持所有陆地生态系统的主要传粉昆虫。使这些昆虫面临危险的生物和非生物因素是生态和农业可持续发展的研究重点。寄生虫是导致全球蜜蜂数量下降的关键因素之一,研究寄生虫的作用机制对于控制蜂群损失至关重要。锥虫寄生虫,尤其是Lotmaria passim在蜜蜂中广泛传播,但人们对它们的生活方式知之甚少。在这项工作中,我们展示了这些寄生虫如何能够分化成一种新的寄生生活方式:锥虫生物膜。我们利用不同的显微技术证明,自由游动的单细胞原虫分泌胞外聚合物物质是在体外和体内产生多细胞生物膜并将其附着在固体表面的先决条件。此外,与人类感染的锥吸虫寄生虫相比,我们的研究显示了蜜蜂锥吸虫寄生虫如何增强其抵抗力,从而适应超低温和低渗透休克等环境条件的剧烈变化,这也是它们在宿主体内或体外成功茁壮成长的原因。这些结果为了解这类寄生虫在自然界中的成功奠定了基础,并揭示了这类病原体对蜜蜂--大多数陆地生态系统中的关键物种--的影响。
{"title":"Promastigote EPS secretion and haptomonad biofilm formation as evolutionary adaptations of trypanosomatid parasites for colonizing honeybee hosts.","authors":"Jéssica Carreira de Paula, Pedro García Olmedo, Tamara Gómez-Moracho, María Buendía-Abad, Mariano Higes, Raquel Martín-Hernández, Antonio Osuna, Luis Miguel de Pablos","doi":"10.1038/s41522-024-00492-x","DOIUrl":"10.1038/s41522-024-00492-x","url":null,"abstract":"<p><p>Bees are major pollinators involved in the maintenance of all terrestrial ecosystems. Biotic and abiotic factors placing these insects at risk is a research priority for ecological and agricultural sustainability. Parasites are one of the key players of this global decline and the study of their mechanisms of action is essential to control honeybee colony losses. Trypanosomatid parasites and particularly the Lotmaria passim are widely spread in honeybees, however their lifestyle is poorly understood. In this work, we show how these parasites are able to differentiate into a new parasitic lifestyle: the trypanosomatid biofilms. Using different microscopic techniques, we demonstrated that the secretion of Extracellular Polymeric Substances by free-swimming unicellular promastigote forms is a prerequisite for the generation and adherence of multicellular biofilms to solid surfaces in vitro and in vivo. Moreover, compared to human-infective trypanosomatid parasites our study shows how trypanosomatid parasites of honeybees increases their resistance and thus resilience to drastic changes in environmental conditions such as ultralow temperatures and hypoosmotic shock, which would explain their success thriving within or outside their hosts. These results set up the basis for the understanding of the success of this group of parasites in nature and to unveil the impact of such pathogens in honeybees, a keystones species in most terrestrial ecosystems.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10957890/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140185056","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Extracellular vesicles from vaginal Gardnerella vaginalis and Mobiluncus mulieris contain distinct proteomic cargo and induce inflammatory pathways. 阴道加德纳菌和莫比伦菌的胞外囊泡含有不同的蛋白质组,并能诱导炎症途径。
IF 7.8 1区 生物学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-03-21 DOI: 10.1038/s41522-024-00502-y
Andrea Joseph, Lauren Anton, Yuxia Guan, Briana Ferguson, Isabella Mirro, Nova Meng, Michael France, Jacques Ravel, Michal A Elovitz

Colonization of the vaginal space with bacteria such as Gardnerella vaginalis and Mobiluncus mulieris is associated with increased risk for STIs, bacterial vaginosis, and preterm birth, while Lactobacillus crispatus is associated with optimal reproductive health. Although host-microbe interactions are hypothesized to contribute to reproductive health and disease, the bacterial mediators that are critical to this response remain unclear. Bacterial extracellular vesicles (bEVs) are proposed to participate in host-microbe communication by providing protection of bacterial cargo, delivery to intracellular targets, and ultimately induction of immune responses from the host. We evaluated the proteome of bEVs produced in vitro from G. vaginalis, M. mulieris, and L. crispatus, identifying specific proteins of immunologic interest. We found that bEVs from each bacterial species internalize within cervical and vaginal epithelial cells, and that epithelial and immune cells express a multi-cytokine response when exposed to bEVs from G. vaginalis and M. mulieris but not L. crispatus. Further, we demonstrate that the inflammatory response induced by G. vaginalis and M. mulieris bEVs is TLR2-specific. Our results provide evidence that vaginal bacteria communicate with host cells through secreted bEVs, revealing a mechanism by which bacteria lead to adverse reproductive outcomes associated with inflammation. Elucidating host-microbe interactions in the cervicovaginal space will provide further insight into the mechanisms contributing to microbiome-mediated adverse outcomes and may reveal new therapeutic targets.

阴道加德纳菌和阴道毛杆菌等细菌在阴道内的定植与性传播感染、细菌性阴道病和早产风险的增加有关,而脆片乳杆菌则与最佳生殖健康有关。虽然宿主与微生物之间的相互作用被认为有助于生殖健康和疾病,但对这种反应至关重要的细菌介质仍不清楚。细菌胞外囊泡(bEVs)被认为可通过保护细菌货物、向细胞内靶标输送以及最终诱导宿主的免疫反应来参与宿主与微生物的交流。我们评估了阴道球菌、毛霉菌和栗色球菌体外产生的 bEVs 的蛋白质组,确定了具有免疫学意义的特定蛋白质。我们发现,每种细菌的 bEVs 都能在宫颈和阴道上皮细胞内发生内化,当暴露于 G. vaginalis 和 M. mulieris 的 bEVs 而不是 L. crispatus 的 bEVs 时,上皮细胞和免疫细胞会表达多种细胞因子反应。此外,我们还证明了阴道杆菌和毛霉菌 bEV 诱导的炎症反应是 TLR2 特异性的。我们的研究结果提供了阴道细菌通过分泌的 bEV 与宿主细胞沟通的证据,揭示了细菌导致与炎症相关的不良生殖结果的机制。阐明宫颈阴道空间中宿主与微生物之间的相互作用将有助于进一步了解微生物介导不良后果的机制,并可能揭示新的治疗靶点。
{"title":"Extracellular vesicles from vaginal Gardnerella vaginalis and Mobiluncus mulieris contain distinct proteomic cargo and induce inflammatory pathways.","authors":"Andrea Joseph, Lauren Anton, Yuxia Guan, Briana Ferguson, Isabella Mirro, Nova Meng, Michael France, Jacques Ravel, Michal A Elovitz","doi":"10.1038/s41522-024-00502-y","DOIUrl":"10.1038/s41522-024-00502-y","url":null,"abstract":"<p><p>Colonization of the vaginal space with bacteria such as Gardnerella vaginalis and Mobiluncus mulieris is associated with increased risk for STIs, bacterial vaginosis, and preterm birth, while Lactobacillus crispatus is associated with optimal reproductive health. Although host-microbe interactions are hypothesized to contribute to reproductive health and disease, the bacterial mediators that are critical to this response remain unclear. Bacterial extracellular vesicles (bEVs) are proposed to participate in host-microbe communication by providing protection of bacterial cargo, delivery to intracellular targets, and ultimately induction of immune responses from the host. We evaluated the proteome of bEVs produced in vitro from G. vaginalis, M. mulieris, and L. crispatus, identifying specific proteins of immunologic interest. We found that bEVs from each bacterial species internalize within cervical and vaginal epithelial cells, and that epithelial and immune cells express a multi-cytokine response when exposed to bEVs from G. vaginalis and M. mulieris but not L. crispatus. Further, we demonstrate that the inflammatory response induced by G. vaginalis and M. mulieris bEVs is TLR2-specific. Our results provide evidence that vaginal bacteria communicate with host cells through secreted bEVs, revealing a mechanism by which bacteria lead to adverse reproductive outcomes associated with inflammation. Elucidating host-microbe interactions in the cervicovaginal space will provide further insight into the mechanisms contributing to microbiome-mediated adverse outcomes and may reveal new therapeutic targets.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":7.8,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10957959/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140185107","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Integrative metagenomic and metabolomic analyses reveal the potential of gut microbiota to exacerbate acute pancreatitis. 元基因组和代谢组的综合分析揭示了肠道微生物群加剧急性胰腺炎的可能性。
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-21 DOI: 10.1038/s41522-024-00499-4
Jianjun Liu, Qiulong Yan, Shenghui Li, Juying Jiao, Yiming Hao, Guixin Zhang, Qingkai Zhang, Fei Luo, Yue Zhang, Qingbo Lv, Wenzhe Zhang, Aiqin Zhang, Huiyi Song, Yi Xin, Yufang Ma, Lawrence Owusu, Xiaochi Ma, Peiyuan Yin, Dong Shang

Early dysbiosis in the gut microbiota may contribute to the severity of acute pancreatitis (AP), however, a comprehensive understanding of the gut microbiome, potential pathobionts, and host metabolome in individuals with AP remains elusive. Hence, we employed fecal whole-metagenome shotgun sequencing in 82 AP patients and 115 matched healthy controls, complemented by untargeted serum metabolome and lipidome profiling in a subset of participants. Analyses of the gut microbiome in AP patients revealed reduced diversity, disrupted microbial functions, and altered abundance of 77 species, influenced by both etiology and severity. AP-enriched species, mostly potential pathobionts, correlated positively with host liver function and serum lipid indicators. Conversely, many AP-depleted species were short-chain fatty acid producers. Gut microflora changes were accompanied by shifts in the serum metabolome and lipidome. Specifically, certain gut species, like enriched Bilophila wadsworthia and depleted Bifidobacterium spp., appeared to contribute to elevated triglyceride levels in biliary or hyperlipidemic AP patients. Through culturing and whole-genome sequencing of bacterial isolates, we identified virulence factors and clinically relevant antibiotic resistance in patient-derived strains, suggesting a predisposition to opportunistic infections. Finally, our study demonstrated that gavage of specific pathobionts could exacerbate pancreatitis in a caerulein-treated mouse model. In conclusion, our comprehensive analysis sheds light on the gut microbiome and serum metabolome in AP, elucidating the role of pathobionts in disease progression. These insights offer valuable perspectives for etiologic diagnosis, prevention, and intervention in AP and related conditions.

肠道微生物群的早期菌群失调可能会导致急性胰腺炎(AP)的严重程度,然而,对急性胰腺炎患者的肠道微生物群、潜在致病菌和宿主代谢组的全面了解仍是空白。因此,我们对82名急性胰腺炎患者和115名匹配的健康对照者进行了粪便全基因组枪式测序,并对一部分参与者进行了非靶向血清代谢组和脂质组分析。对 AP 患者肠道微生物组的分析表明,受病因和严重程度的影响,77 种微生物的多样性降低、微生物功能紊乱、丰度改变。AP富集的物种(主要是潜在的致病菌)与宿主肝功能和血清脂质指标呈正相关。相反,许多缺乏 AP 的物种是短链脂肪酸的生产者。肠道微生物区系的变化伴随着血清代谢组和脂质组的变化。具体来说,某些肠道菌群,如富集的双歧杆菌(Bilophila wadsworthia)和枯竭的双歧杆菌(Bifidobacterium spp.),似乎会导致胆汁性或高脂血症 AP 患者的甘油三酯水平升高。通过对细菌分离物进行培养和全基因组测序,我们在患者来源菌株中发现了毒力因子和临床相关的抗生素耐药性,这表明患者有机会性感染的倾向。最后,我们的研究表明,在经尾叶素处理的小鼠模型中,灌胃特定病原菌可加重胰腺炎。总之,我们的综合分析揭示了 AP 的肠道微生物组和血清代谢组,阐明了病原菌在疾病进展中的作用。这些见解为 AP 及其相关疾病的病因诊断、预防和干预提供了宝贵的视角。
{"title":"Integrative metagenomic and metabolomic analyses reveal the potential of gut microbiota to exacerbate acute pancreatitis.","authors":"Jianjun Liu, Qiulong Yan, Shenghui Li, Juying Jiao, Yiming Hao, Guixin Zhang, Qingkai Zhang, Fei Luo, Yue Zhang, Qingbo Lv, Wenzhe Zhang, Aiqin Zhang, Huiyi Song, Yi Xin, Yufang Ma, Lawrence Owusu, Xiaochi Ma, Peiyuan Yin, Dong Shang","doi":"10.1038/s41522-024-00499-4","DOIUrl":"10.1038/s41522-024-00499-4","url":null,"abstract":"<p><p>Early dysbiosis in the gut microbiota may contribute to the severity of acute pancreatitis (AP), however, a comprehensive understanding of the gut microbiome, potential pathobionts, and host metabolome in individuals with AP remains elusive. Hence, we employed fecal whole-metagenome shotgun sequencing in 82 AP patients and 115 matched healthy controls, complemented by untargeted serum metabolome and lipidome profiling in a subset of participants. Analyses of the gut microbiome in AP patients revealed reduced diversity, disrupted microbial functions, and altered abundance of 77 species, influenced by both etiology and severity. AP-enriched species, mostly potential pathobionts, correlated positively with host liver function and serum lipid indicators. Conversely, many AP-depleted species were short-chain fatty acid producers. Gut microflora changes were accompanied by shifts in the serum metabolome and lipidome. Specifically, certain gut species, like enriched Bilophila wadsworthia and depleted Bifidobacterium spp., appeared to contribute to elevated triglyceride levels in biliary or hyperlipidemic AP patients. Through culturing and whole-genome sequencing of bacterial isolates, we identified virulence factors and clinically relevant antibiotic resistance in patient-derived strains, suggesting a predisposition to opportunistic infections. Finally, our study demonstrated that gavage of specific pathobionts could exacerbate pancreatitis in a caerulein-treated mouse model. In conclusion, our comprehensive analysis sheds light on the gut microbiome and serum metabolome in AP, elucidating the role of pathobionts in disease progression. These insights offer valuable perspectives for etiologic diagnosis, prevention, and intervention in AP and related conditions.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10957925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140185108","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Metabolic profiles outperform the microbiota in assessing the response of vaginal microenvironments to the changed state of HPV infection 在评估阴道微环境对人类乳头瘤病毒感染变化状态的反应方面,代谢特征优于微生物群
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-20 DOI: 10.1038/s41522-024-00500-0
Wenkui Dai, Hui Du, Qian Zhou, Sumei Li, Yinan Wang, Jun Hou, Chunlei Guo, Qing Yang, Changzhong Li, Shouxia Xie, Shuai Cheng Li, Ruifang Wu

There is a deficiency in population-based studies investigating the impact of HPV infection on vaginal microenvironment, which influences the risk of persistent HPV infection. This prospective study aimed to unravel the dynamics of vaginal microbiota (VM) and vaginal metabolome in reaction to the changed state of HPV infection. Our results propose that the vaginal metabolome may be a superior indicator to VM when assessing the impact of altered HPV state on the vaginal microenvironment.

基于人群的研究缺乏对 HPV 感染对阴道微环境影响的调查,而阴道微环境会影响 HPV 持续感染的风险。这项前瞻性研究旨在揭示阴道微生物群(VM)和阴道代谢组在HPV感染状态改变时的动态变化。我们的研究结果表明,在评估HPV状态改变对阴道微环境的影响时,阴道代谢组可能是优于VM的指标。
{"title":"Metabolic profiles outperform the microbiota in assessing the response of vaginal microenvironments to the changed state of HPV infection","authors":"Wenkui Dai, Hui Du, Qian Zhou, Sumei Li, Yinan Wang, Jun Hou, Chunlei Guo, Qing Yang, Changzhong Li, Shouxia Xie, Shuai Cheng Li, Ruifang Wu","doi":"10.1038/s41522-024-00500-0","DOIUrl":"https://doi.org/10.1038/s41522-024-00500-0","url":null,"abstract":"<p>There is a deficiency in population-based studies investigating the impact of HPV infection on vaginal microenvironment, which influences the risk of persistent HPV infection. This prospective study aimed to unravel the dynamics of vaginal microbiota (VM) and vaginal metabolome in reaction to the changed state of HPV infection. Our results propose that the vaginal metabolome may be a superior indicator to VM when assessing the impact of altered HPV state on the vaginal microenvironment.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140172419","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lactobacillus rhamnosus GG ameliorates hyperuricemia in a novel model 鼠李糖乳杆菌 GG 在新型模型中改善高尿酸血症
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-20 DOI: 10.1038/s41522-024-00486-9
Yang Fu, Yong-Song Chen, Dai-Yang Xia, Xiao-Dan Luo, Hao-Tong Luo, Jie Pan, Wei-Qing Ma, Jin-Ze Li, Qian-Yuan Mo, Qiang Tu, Meng-Meng Li, Yue Zhao, Yu Li, Yi-Teng Huang, Zhi-Xian Chen, Zhen-Jun Li, Lukuyu Bernard, Michel Dione, You-Ming Zhang, Kai Miao, Jian-Ying Chen, Shan-Shan Zhu, Jie Ren, Ling-Juan Zhou, Xian-Zhi Jiang, Juan Chen, Zhen-Ping Lin, Jun-Peng Chen, Hui Ye, Qing-Yun Cao, Yong-Wen Zhu, Lin Yang, Xue Wang, Wen-Ce Wang

Hyperuricemia (HUA) is a metabolic syndrome caused by abnormal purine metabolism. Although recent studies have noted a relationship between the gut microbiota and gout, whether the microbiota could ameliorate HUA-associated systemic purine metabolism remains unclear. In this study, we constructed a novel model of HUA in geese and investigated the mechanism by which Lactobacillus rhamnosus GG (LGG) could have beneficial effects on HUA. The administration of antibiotics and fecal microbiota transplantation (FMT) experiments were used in this HUA goose model. The effects of LGG and its metabolites on HUA were evaluated in vivo and in vitro. Heterogeneous expression and gene knockout of LGG revealed the mechanism of LGG. Multi-omics analysis revealed that the Lactobacillus genus is associated with changes in purine metabolism in HUA. This study showed that LGG and its metabolites could alleviate HUA through the gut-liver-kidney axis. Whole-genome analysis, heterogeneous expression, and gene knockout of LGG enzymes ABC-type multidrug transport system (ABCT), inosine-uridine nucleoside N-ribohydrolase (iunH), and xanthine permease (pbuX) demonstrated the function of nucleoside degradation in LGG. Multi-omics and a correlation analysis in HUA patients and this goose model revealed that a serum proline deficiency, as well as changes in Collinsella and Lactobacillus, may be associated with the occurrence of HUA. Our findings demonstrated the potential of a goose model of diet-induced HUA, and LGG and proline could be promising therapies for HUA.

高尿酸血症(HUA)是一种由嘌呤代谢异常引起的代谢综合征。尽管最近的研究指出了肠道微生物群与痛风之间的关系,但微生物群能否改善 HUA 相关的全身性嘌呤代谢仍不清楚。在这项研究中,我们构建了一种新型鹅 HUA 模型,并研究了鼠李糖乳杆菌 GG(LGG)对 HUA 产生有益影响的机制。该HUA鹅模型采用了抗生素给药和粪便微生物群移植(FMT)实验。在体内和体外评估了 LGG 及其代谢物对 HUA 的影响。LGG 的异质性表达和基因敲除揭示了 LGG 的作用机制。多组学分析表明,乳酸杆菌属与 HUA 的嘌呤代谢变化有关。这项研究表明,LGG 及其代谢产物可通过肠道-肝脏-肾脏轴缓解 HUA。LGG酶ABC型多药转运系统(ABCT)、肌苷尿苷核苷酸N-核苷酸水解酶(iunH)和黄嘌呤渗透酶(pbuX)的全基因组分析、异质性表达和基因敲除证明了LGG中核苷降解的功能。对 HUA 患者和这种鹅模型进行的多组学和相关分析表明,血清脯氨酸缺乏以及柯林斯菌和乳酸杆菌的变化可能与 HUA 的发生有关。我们的研究结果证明了饮食诱发 HUA 的鹅模型的潜力,LGG 和脯氨酸可能是治疗 HUA 的有前途的疗法。
{"title":"Lactobacillus rhamnosus GG ameliorates hyperuricemia in a novel model","authors":"Yang Fu, Yong-Song Chen, Dai-Yang Xia, Xiao-Dan Luo, Hao-Tong Luo, Jie Pan, Wei-Qing Ma, Jin-Ze Li, Qian-Yuan Mo, Qiang Tu, Meng-Meng Li, Yue Zhao, Yu Li, Yi-Teng Huang, Zhi-Xian Chen, Zhen-Jun Li, Lukuyu Bernard, Michel Dione, You-Ming Zhang, Kai Miao, Jian-Ying Chen, Shan-Shan Zhu, Jie Ren, Ling-Juan Zhou, Xian-Zhi Jiang, Juan Chen, Zhen-Ping Lin, Jun-Peng Chen, Hui Ye, Qing-Yun Cao, Yong-Wen Zhu, Lin Yang, Xue Wang, Wen-Ce Wang","doi":"10.1038/s41522-024-00486-9","DOIUrl":"https://doi.org/10.1038/s41522-024-00486-9","url":null,"abstract":"<p>Hyperuricemia (HUA) is a metabolic syndrome caused by abnormal purine metabolism. Although recent studies have noted a relationship between the gut microbiota and gout, whether the microbiota could ameliorate HUA-associated systemic purine metabolism remains unclear. In this study, we constructed a novel model of HUA in geese and investigated the mechanism by which <i>Lactobacillus rhamnosus</i> GG (LGG) could have beneficial effects on HUA. The administration of antibiotics and fecal microbiota transplantation (FMT) experiments were used in this HUA goose model. The effects of LGG and its metabolites on HUA were evaluated in vivo and in vitro. Heterogeneous expression and gene knockout of LGG revealed the mechanism of LGG. Multi-omics analysis revealed that the <i>Lactobacillus</i> genus is associated with changes in purine metabolism in HUA. This study showed that LGG and its metabolites could alleviate HUA through the gut-liver-kidney axis. Whole-genome analysis, heterogeneous expression, and gene knockout of LGG enzymes ABC-type multidrug transport system (<i>ABCT</i>), inosine-uridine nucleoside N-ribohydrolase (<i>iunH</i>), and xanthine permease (<i>pbuX</i>) demonstrated the function of nucleoside degradation in LGG. Multi-omics and a correlation analysis in HUA patients and this goose model revealed that a serum proline deficiency, as well as changes in <i>Collinsella</i> and <i>Lactobacillus</i>, may be associated with the occurrence of HUA. Our findings demonstrated the potential of a goose model of diet-induced HUA, and LGG and proline could be promising therapies for HUA.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140172456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Competition quenching strategies reduce antibiotic tolerance in polymicrobial biofilms 竞争淬灭策略降低了多微生物生物膜的抗生素耐受性
IF 9.2 1区 生物学 Q1 Immunology and Microbiology Pub Date : 2024-03-19 DOI: 10.1038/s41522-024-00489-6
Bram Lories, Tom E. R. Belpaire, Bart Smeets, Hans P. Steenackers

Bacteria typically live in dense communities where they are surrounded by other species and compete for a limited amount of resources. These competitive interactions can induce defensive responses that also protect against antimicrobials, potentially complicating the antimicrobial treatment of pathogens residing in polymicrobial consortia. Therefore, we evaluate the potential of alternative antivirulence strategies that quench this response to competition. We test three competition quenching approaches: (i) interference with the attack mechanism of surrounding competitors, (ii) inhibition of the stress response systems that detect competition, and (iii) reduction of the overall level of competition in the community by lowering the population density. We show that either strategy can prevent the induction of antimicrobial tolerance of Salmonella Typhimurium in response to competitors. Competition quenching strategies can thus reduce tolerance of pathogens residing in polymicrobial communities and could contribute to the improved eradication of these pathogens via traditional methods.

细菌通常生活在密集的群落中,它们被其他物种包围,争夺有限的资源。这些竞争性的相互作用会诱发防御反应,同时也能抵御抗微生物药物,这可能会使多微生物群落中病原体的抗微生物治疗复杂化。因此,我们评估了抑制这种竞争反应的替代抗病毒策略的潜力。我们测试了三种抑制竞争的方法:(i) 干扰周围竞争者的攻击机制,(ii) 抑制检测竞争的应激反应系统,(iii) 通过降低种群密度来减少群落中的整体竞争水平。我们的研究表明,这两种策略都能阻止鼠伤寒沙门氏菌对竞争者产生抗菌耐受性。因此,竞争淬灭策略可以降低多微生物群落中病原体的耐受性,并有助于通过传统方法更好地消灭这些病原体。
{"title":"Competition quenching strategies reduce antibiotic tolerance in polymicrobial biofilms","authors":"Bram Lories, Tom E. R. Belpaire, Bart Smeets, Hans P. Steenackers","doi":"10.1038/s41522-024-00489-6","DOIUrl":"https://doi.org/10.1038/s41522-024-00489-6","url":null,"abstract":"<p>Bacteria typically live in dense communities where they are surrounded by other species and compete for a limited amount of resources. These competitive interactions can induce defensive responses that also protect against antimicrobials, potentially complicating the antimicrobial treatment of pathogens residing in polymicrobial consortia. Therefore, we evaluate the potential of alternative antivirulence strategies that quench this response to competition. We test three competition quenching approaches: (i) interference with the attack mechanism of surrounding competitors, (ii) inhibition of the stress response systems that detect competition, and (iii) reduction of the overall level of competition in the community by lowering the population density. We show that either strategy can prevent the induction of antimicrobial tolerance of <i>Salmonella</i> Typhimurium in response to competitors. Competition quenching strategies can thus reduce tolerance of pathogens residing in polymicrobial communities and could contribute to the improved eradication of these pathogens via traditional methods.</p>","PeriodicalId":19370,"journal":{"name":"npj Biofilms and Microbiomes","volume":null,"pages":null},"PeriodicalIF":9.2,"publicationDate":"2024-03-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140172449","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
npj Biofilms and Microbiomes
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