首页 > 最新文献

Journal of Bacteriology最新文献

英文 中文
In memoriam: Pierre Cornelis (1949-2023). 悼念皮埃尔-科内利斯(1949-2023)。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-12 DOI: 10.1128/jb.00179-24
Jean-Paul Pirnay, Françoise Van Bambeke, Christine Baysse, Miguel Cámara, Sylvie Chevalier, Jean François Collet, Aurélie Crabbé, Jozef Dingemans, Alain Dufour, Linda Eeckhaudt, Alain Filloux, Olivier Lesouhaitier, Qing Wei, Daniel De Vos
{"title":"In memoriam: Pierre Cornelis (1949-2023).","authors":"Jean-Paul Pirnay, Françoise Van Bambeke, Christine Baysse, Miguel Cámara, Sylvie Chevalier, Jean François Collet, Aurélie Crabbé, Jozef Dingemans, Alain Dufour, Linda Eeckhaudt, Alain Filloux, Olivier Lesouhaitier, Qing Wei, Daniel De Vos","doi":"10.1128/jb.00179-24","DOIUrl":"10.1128/jb.00179-24","url":null,"abstract":"","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141758838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 sequence invariable region in TcdB2 is required for toxin escape from Clostridioides difficile. 艰难梭菌毒素逃逸需要 TcdB2 中的一个序列不变区。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-18 DOI: 10.1128/jb.00096-24
Megan L Kempher, Tyler M Shadid, Jason L Larabee, Jimmy D Ballard

Sequence differences among the subtypes of Clostridioides difficile toxin TcdB (2,366 amino acids) are broadly distributed across the entire protein, with the notable exception of 76 residues at the protein's carboxy terminus. This sequence invariable region (SIR) is identical at the DNA and protein level among the TcdB variants, suggesting this string of amino acids has undergone selective pressure to prevent alterations. The functional role of the SIR domain in TcdB has not been determined. Analysis of a recombinantly constructed TcdB mutant lacking the SIR domain did not identify changes in TcdB's enzymatic or cytopathic activities. To further assess the SIR region, we constructed a C. difficile strain with the final 228 bp deleted from the tcdB gene, resulting in the production of a truncated form of TcdB lacking the SIR (TcdB2∆2291-2366). Using a combination of approaches, we found in the absence of the SIR sequence TcdB2∆2291-2366 retained cytotoxic activity but was not secreted from C. difficile. TcdB2∆2291-2366 was not released from the cell under autolytic conditions, indicating the SIR is involved in a more discrete step in toxin escape from the bacterium. Fractionation experiments combined with antibody detection found that TcdB2∆2291-2366 accumulates at the cell membrane but is unable to complete steps in secretion beyond this point. These data suggest conservation of the SIR domain across variants of TcdB could be influenced by the sequence's role in efficient escape of the toxin from C. difficile.

Importance: Clostridioides difficile is a leading cause of antibiotic associated disease in the United States. The primary virulence factors produced by C. difficile are two large glucosylating toxins TcdA and TcdB. To date, several sequence variants of TcdB have been identified that differ in various functional properties. Here, we identified a highly conserved region among TcdB subtypes that is required for release of the toxin from C. difficile. This study reveals a putative role for the longest stretch of invariable sequence among TcdB subtypes and provides new details regarding toxin release into the extracellular environment. Improving our understanding of the functional roles of the conserved regions of TcdB variants aids in the development of new, broadly applicable strategies to treat CDI.

难辨梭状芽孢杆菌毒素 TcdB(2,366 个氨基酸)各亚型之间的序列差异广泛分布于整个蛋白质中,但位于蛋白质羧基末端的 76 个残基除外。在 TcdB 变体中,这一序列不变区(SIR)在 DNA 和蛋白质水平上都是相同的,这表明这串氨基酸经历了选择性压力,以防止发生变化。TcdB 中 SIR 结构域的功能作用尚未确定。对重组构建的缺乏 SIR 结构域的 TcdB 突变体的分析并未发现 TcdB 的酶活性或细胞病理学活性发生了变化。为了进一步评估 SIR 区域,我们构建了一株艰难梭菌菌株,删除了 tcdB 基因的最后 228 bp,从而产生了缺乏 SIR 的截短形式 TcdB(TcdB2∆2291-2366)。通过综合使用多种方法,我们发现在缺乏 SIR 序列的情况下,TcdB2∆2291-2366 仍具有细胞毒性活性,但不会从艰难梭菌中分泌出来。在自溶条件下,TcdB2Δ2291-2366 也不会从细胞中释放出来,这表明 SIR 参与了毒素从细菌中逸出的一个更为独立的步骤。结合抗体检测进行的分馏实验发现,TcdB2∆2291-2366 在细胞膜上积聚,但除此之外无法完成分泌步骤。这些数据表明,TcdB 不同变体中 SIR 结构域的保留可能受到该序列在艰难梭菌毒素有效逃逸过程中所起作用的影响:艰难梭菌是美国抗生素相关疾病的主要病因。艰难梭菌产生的主要毒力因子是两种大型葡萄糖基化毒素 TcdA 和 TcdB。迄今为止,已经发现了几种 TcdB 的序列变体,它们在各种功能特性上存在差异。在这里,我们在 TcdB 亚型中发现了一个高度保守的区域,该区域是艰难梭菌释放毒素所必需的。这项研究揭示了 TcdB 亚型中最长的一段不变序列的潜在作用,并提供了毒素释放到细胞外环境的新细节。提高我们对 TcdB 变体保守区功能作用的认识有助于开发新的、广泛适用的 CDI 治疗策略。
{"title":"A sequence invariable region in TcdB2 is required for toxin escape from <i>Clostridioides difficile</i>.","authors":"Megan L Kempher, Tyler M Shadid, Jason L Larabee, Jimmy D Ballard","doi":"10.1128/jb.00096-24","DOIUrl":"10.1128/jb.00096-24","url":null,"abstract":"<p><p>Sequence differences among the subtypes of <i>Clostridioides difficile</i> toxin TcdB (2,366 amino acids) are broadly distributed across the entire protein, with the notable exception of 76 residues at the protein's carboxy terminus. This sequence invariable region (SIR) is identical at the DNA and protein level among the TcdB variants, suggesting this string of amino acids has undergone selective pressure to prevent alterations. The functional role of the SIR domain in TcdB has not been determined. Analysis of a recombinantly constructed TcdB mutant lacking the SIR domain did not identify changes in TcdB's enzymatic or cytopathic activities. To further assess the SIR region, we constructed a <i>C. difficile</i> strain with the final 228 bp deleted from the <i>tcdB</i> gene, resulting in the production of a truncated form of TcdB lacking the SIR (TcdB2<sub>∆2291-2366</sub>). Using a combination of approaches, we found in the absence of the SIR sequence TcdB2<sub>∆2291-2366</sub> retained cytotoxic activity but was not secreted from <i>C. difficile</i>. TcdB2<sub>∆2291-2366</sub> was not released from the cell under autolytic conditions, indicating the SIR is involved in a more discrete step in toxin escape from the bacterium. Fractionation experiments combined with antibody detection found that TcdB2<sub>∆2291-2366</sub> accumulates at the cell membrane but is unable to complete steps in secretion beyond this point. These data suggest conservation of the SIR domain across variants of TcdB could be influenced by the sequence's role in efficient escape of the toxin from <i>C. difficile</i>.</p><p><strong>Importance: </strong><i>Clostridioides difficile</i> is a leading cause of antibiotic associated disease in the United States. The primary virulence factors produced by <i>C. difficile</i> are two large glucosylating toxins TcdA and TcdB. To date, several sequence variants of TcdB have been identified that differ in various functional properties. Here, we identified a highly conserved region among TcdB subtypes that is required for release of the toxin from <i>C. difficile</i>. This study reveals a putative role for the longest stretch of invariable sequence among TcdB subtypes and provides new details regarding toxin release into the extracellular environment. Improving our understanding of the functional roles of the conserved regions of TcdB variants aids in the development of new, broadly applicable strategies to treat CDI.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11323933/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141419292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
DarA-the central processing unit for the integration of osmotic with potassium and amino acid homeostasis in Bacillus subtilis. DarA--整合枯草芽孢杆菌渗透压、钾和氨基酸平衡的中央处理单元。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-04 DOI: 10.1128/jb.00190-24
Robert Warneke, Christina Herzberg, Martin Weiß, Thorben Schramm, Dietrich Hertel, Hannes Link, Jörg Stülke

Cyclic di-adenosine monophosphate (c-di-AMP) is a second messenger involved in diverse metabolic processes including osmolyte uptake, cell wall homeostasis, as well as antibiotic and heat resistance. This study investigates the role of the c-di-AMP receptor protein DarA in the osmotic stress response in Bacillus subtilis. Through a series of experiments, we demonstrate that DarA plays a central role in the cellular response to osmotic fluctuations. Our findings show that DarA becomes essential under extreme potassium limitation as well as upon salt stress, highlighting its significance in mediating osmotic stress adaptation. Suppressor screens with darA mutants reveal compensatory mechanisms involving the accumulation of osmoprotectants, particularly potassium and citrulline. Mutations affecting various metabolic pathways, including the citric acid cycle as well as glutamate and arginine biosynthesis, indicate a complex interplay between the osmotic stress response and metabolic regulation. In addition, the growth defects of the darA mutant during potassium starvation and salt stress in a strain lacking the high-affinity potassium uptake systems KimA and KtrAB can be rescued by increased affinity of the remaining potassium channel KtrCD or by increased expression of ktrD, thus resulting in increased potassium uptake. Finally, the darA mutant can respond to salt stress by the increased expression of MleN , which can export sodium ions.IMPORTANCEEnvironmental bacteria are exposed to rapidly changing osmotic conditions making an effective adaptation to these changes crucial for the survival of the cells. In Gram-positive bacteria, the second messenger cyclic di-AMP plays a key role in this adaptation by controlling (i) the influx of physiologically compatible organic osmolytes and (ii) the biosynthesis of such osmolytes. In several bacteria, cyclic di-adenosine monophosphate (c-di-AMP) can bind to a signal transduction protein, called DarA, in Bacillus subtilis. So far, no function for DarA has been discovered in any organism. We have identified osmotically challenging conditions that make DarA essential and have identified suppressor mutations that help the bacteria to adapt to those conditions. Our results indicate that DarA is a central component in the integration of osmotic stress with the synthesis of compatible amino acid osmolytes and with the homeostasis of potassium, the first response to osmotic stress.

环状二腺苷单磷酸(c-di-AMP)是一种第二信使,参与多种代谢过程,包括渗透溶质吸收、细胞壁稳态以及抗生素和耐热性。本研究探讨了 c-di-AMP 受体蛋白 DarA 在枯草芽孢杆菌渗透压应激反应中的作用。通过一系列实验,我们证明了 DarA 在细胞对渗透压波动的反应中起着核心作用。我们的研究结果表明,在极端钾限制和盐胁迫下,DarA变得至关重要,突出了它在介导渗透胁迫适应中的重要作用。利用 darA 突变体进行的抑制筛选揭示了涉及渗透保护剂(尤其是钾和瓜氨酸)积累的补偿机制。影响各种代谢途径(包括柠檬酸循环以及谷氨酸和精氨酸的生物合成)的突变表明,渗透胁迫响应与代谢调节之间存在复杂的相互作用。此外,在缺乏高亲和力钾吸收系统 KimA 和 KtrAB 的菌株中,darA 突变体在钾饥饿和盐胁迫期间的生长缺陷可以通过增加剩余钾通道 KtrCD 的亲和力或增加 ktrD 的表达来挽救,从而导致钾吸收增加。最后,darA 突变体可以通过增加 MleN 的表达来应对盐胁迫,MleN 可以输出钠离子。在革兰氏阳性细菌中,第二信使环状二-AMP 通过控制 (i) 生理相容有机渗透溶质的流入和 (ii) 此类渗透溶质的生物合成,在这种适应中发挥着关键作用。在几种细菌中,环状二腺苷单磷酸(c-di-AMP)可与枯草芽孢杆菌(Bacillus subtilis)中一种名为 DarA 的信号转导蛋白结合。迄今为止,尚未在任何生物体内发现 DarA 的功能。我们已经确定了使 DarA 变得必不可少的具有渗透压挑战性的条件,并确定了有助于细菌适应这些条件的抑制突变。我们的研究结果表明,DarA 是将渗透压与相容氨基酸渗透溶质的合成以及钾的平衡(渗透压的第一反应)结合起来的核心成分。
{"title":"DarA-the central processing unit for the integration of osmotic with potassium and amino acid homeostasis in <i>Bacillus subtilis</i>.","authors":"Robert Warneke, Christina Herzberg, Martin Weiß, Thorben Schramm, Dietrich Hertel, Hannes Link, Jörg Stülke","doi":"10.1128/jb.00190-24","DOIUrl":"10.1128/jb.00190-24","url":null,"abstract":"<p><p>Cyclic di-adenosine monophosphate (c-di-AMP) is a second messenger involved in diverse metabolic processes including osmolyte uptake, cell wall homeostasis, as well as antibiotic and heat resistance. This study investigates the role of the c-di-AMP receptor protein DarA in the osmotic stress response in <i>Bacillus subtilis</i>. Through a series of experiments, we demonstrate that DarA plays a central role in the cellular response to osmotic fluctuations. Our findings show that DarA becomes essential under extreme potassium limitation as well as upon salt stress, highlighting its significance in mediating osmotic stress adaptation. Suppressor screens with <i>darA</i> mutants reveal compensatory mechanisms involving the accumulation of osmoprotectants, particularly potassium and citrulline. Mutations affecting various metabolic pathways, including the citric acid cycle as well as glutamate and arginine biosynthesis, indicate a complex interplay between the osmotic stress response and metabolic regulation. In addition, the growth defects of the <i>darA</i> mutant during potassium starvation and salt stress in a strain lacking the high-affinity potassium uptake systems KimA and KtrAB can be rescued by increased affinity of the remaining potassium channel KtrCD or by increased expression of <i>ktrD,</i> thus resulting in increased potassium uptake. Finally, the <i>darA</i> mutant can respond to salt stress by the increased expression of MleN , which can export sodium ions.IMPORTANCEEnvironmental bacteria are exposed to rapidly changing osmotic conditions making an effective adaptation to these changes crucial for the survival of the cells. In Gram-positive bacteria, the second messenger cyclic di-AMP plays a key role in this adaptation by controlling (i) the influx of physiologically compatible organic osmolytes and (ii) the biosynthesis of such osmolytes. In several bacteria, cyclic di-adenosine monophosphate (c-di-AMP) can bind to a signal transduction protein, called DarA, in <i>Bacillus subtilis</i>. So far, no function for DarA has been discovered in any organism. We have identified osmotically challenging conditions that make DarA essential and have identified suppressor mutations that help the bacteria to adapt to those conditions. Our results indicate that DarA is a central component in the integration of osmotic stress with the synthesis of compatible amino acid osmolytes and with the homeostasis of potassium, the first response to osmotic stress.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270874/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141237576","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
ComI inhibits transformation in Bacillus subtilis by selectively killing competent cells. ComI 可选择性地杀死有能力的细胞,从而抑制枯草杆菌的转化。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-14 DOI: 10.1128/jb.00413-23
Dominique R Smith, Daniel B Kearns, Briana M Burton

Many bacteria build elaborate molecular machines to import DNA via natural competence, yet this activity is often not identified until strains have been handled and domesticated in laboratory settings. For example, one of the best studied Gram-positive model organisms, Bacillus subtilis, has a poorly transformable ancestor. Transformation in the ancestral strain is inhibited by a transmembrane peptide, ComI, which is encoded on an extrachromosomal plasmid. Although ComI was shown to be necessary and sufficient to inhibit transformation when produced at high levels under an inducible promoter, the mechanism by which ComI inhibits transformation is unknown. Here, we examine the native regulation and mechanism of transformation inhibition by ComI. We find that under native regulation, ComI expression is restricted in the absence of the plasmid. In the presence of the plasmid, we find that ComI is expressed at higher levels in cells that are differentiating into a competent state. The subcellular localization of ComI, however, does not depend on any other competence proteins, and permeabilization activity is concentration-dependent. Time-lapse microscopy reveals that competent cells producing ComI are first permeabilized and then die. Based on these observations, we propose a new model for the mechanism of ComI in which response to competence activation leads to selective elimination of the competent subpopulation.

Importance: Natural transformation mechanisms have been studied across several bacterial systems, but few examples of inhibition exist. This work investigates the mechanism of action of a plasmid-encoded transmembrane inhibitor of natural transformation. The data reveal that the peptide can cause cell permeabilization. Permeabilization is synergistic with entry of Bacillus subtilis into the "competent" state, such that cells with the ability to be transformed are preferentially killed. These findings reveal a self-preservation mechanism coupled to the physiological state of the cells that ensures that the population can maintain an unaltered plasmid and its predicted prophage.

许多细菌建立了精心设计的分子机器,通过自然能力导入 DNA,但这种活动往往要等到在实验室环境中处理和驯化菌株后才能发现。例如,研究得最好的革兰氏阳性模式生物之一枯草芽孢杆菌(Bacillus subtilis)的祖先转化能力很差。祖先菌株的转化受到一种跨膜肽 ComI 的抑制,而 ComI 是由染色体外质粒编码的。尽管 ComI 在诱导型启动子的作用下产生高浓度时对抑制转化是必要且充分的,但 ComI 抑制转化的机制尚不清楚。在这里,我们研究了 ComI 的原生调控和抑制转化的机制。我们发现,在原生调控下,ComI 的表达在没有质粒的情况下受到限制。我们发现,在有质粒存在的情况下,ComI 在分化为合格状态的细胞中表达水平较高。然而,ComI 的亚细胞定位并不依赖于任何其他能力蛋白,而且渗透活性与浓度有关。延时显微镜观察发现,产生 ComI 的有能力细胞首先被渗透,然后死亡。基于这些观察结果,我们提出了一种新的 ComI 机制模型,其中对能力激活的反应导致能力亚群的选择性淘汰:人们已经对多个细菌系统的自然转化机制进行了研究,但很少有抑制作用的实例。这项工作研究了一种质粒编码的跨膜自然转化抑制剂的作用机制。数据显示,该多肽可导致细胞渗透。渗透与枯草杆菌进入 "能转化 "状态具有协同作用,因此具有转化能力的细胞会被优先杀死。这些发现揭示了一种与细胞生理状态相耦合的自我保护机制,该机制可确保种群能够维持未改变的质粒及其预测的噬菌体。
{"title":"ComI inhibits transformation in <i>Bacillus subtilis</i> by selectively killing competent cells.","authors":"Dominique R Smith, Daniel B Kearns, Briana M Burton","doi":"10.1128/jb.00413-23","DOIUrl":"10.1128/jb.00413-23","url":null,"abstract":"<p><p>Many bacteria build elaborate molecular machines to import DNA via natural competence, yet this activity is often not identified until strains have been handled and domesticated in laboratory settings. For example, one of the best studied Gram-positive model organisms, <i>Bacillus subtilis,</i> has a poorly transformable ancestor. Transformation in the ancestral strain is inhibited by a transmembrane peptide, ComI, which is encoded on an extrachromosomal plasmid. Although ComI was shown to be necessary and sufficient to inhibit transformation when produced at high levels under an inducible promoter, the mechanism by which ComI inhibits transformation is unknown. Here, we examine the native regulation and mechanism of transformation inhibition by ComI. We find that under native regulation, ComI expression is restricted in the absence of the plasmid. In the presence of the plasmid, we find that ComI is expressed at higher levels in cells that are differentiating into a competent state. The subcellular localization of ComI, however, does not depend on any other competence proteins, and permeabilization activity is concentration-dependent. Time-lapse microscopy reveals that competent cells producing ComI are first permeabilized and then die. Based on these observations, we propose a new model for the mechanism of ComI in which response to competence activation leads to selective elimination of the competent subpopulation.</p><p><strong>Importance: </strong>Natural transformation mechanisms have been studied across several bacterial systems, but few examples of inhibition exist. This work investigates the mechanism of action of a plasmid-encoded transmembrane inhibitor of natural transformation. The data reveal that the peptide can cause cell permeabilization. Permeabilization is synergistic with entry of <i>Bacillus subtilis</i> into the \"competent\" state, such that cells with the ability to be transformed are preferentially killed. These findings reveal a self-preservation mechanism coupled to the physiological state of the cells that ensures that the population can maintain an unaltered plasmid and its predicted prophage.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270867/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317425","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 expanded genetic toolkit for inducible expression and targeted gene silencing in Rickettsia parkeri. 用于帕克氏立克次体诱导表达和靶向基因沉默的扩展基因工具包。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-06 DOI: 10.1128/jb.00091-24
Jon McGinn, Annie Wen, Desmond L Edwards, David M Brinkley, Rebecca L Lamason

Pathogenic species within the Rickettsia genus are transmitted to humans through arthropod vectors and cause a spectrum of diseases ranging from mild to life-threatening. Despite rickettsiae posing an emerging global health risk, the genetic requirements of their infectious life cycles remain poorly understood. A major hurdle toward building this understanding has been the lack of efficient tools for genetic manipulation, owing to the technical difficulties associated with their obligate intracellular nature. To this end, we implemented the Tet-On system to enable conditional gene expression in Rickettsia parkeri. Using Tet-On, we show inducible expression of antibiotic resistance and a fluorescent reporter. We further used this inducible promoter to screen the ability of R. parkeri to express four variants of the catalytically dead Cas9 (dCas9). We demonstrate that all four dCas9 variants can be expressed in R. parkeri and used for CRISPR interference (CRISPRi)-mediated targeted gene knockdown. We show targeted knockdown of an antibiotic resistance gene as well as the endogenous virulence factor sca2. Altogether, we have developed systems for inducible gene expression and CRISPRi-mediated gene knockdown for the first time in rickettsiae, laying the groundwork for more scalable, targeted mechanistic investigations into their infectious life cycles.IMPORTANCEThe spotted fever group of Rickettsia contains vector-borne pathogenic bacteria that are neglected and emerging threats to public health. Due to the obligate intracellular nature of rickettsiae, the development of tools for genetic manipulation has been stunted, and the molecular and genetic underpinnings of their infectious lifecycle remain poorly understood. Here, we expand the genetic toolkit by introducing systems for conditional gene expression and CRISPR interference (CRISPRi)-mediated gene knockdown. These systems allow for relatively easy manipulation of rickettsial gene expression. We demonstrate the effectiveness of these tools by disrupting the intracellular life cycle using CRISPRi to deplete the sca2 virulence factor. These tools will be crucial for building a more comprehensive and detailed understanding of rickettsial biology and pathogenesis.

立克次体属中的致病菌种通过节肢动物媒介传播给人类,并引起一系列从轻微到危及生命的疾病。尽管立克次体对全球健康构成了新的威胁,但人们对其传染生命周期的遗传要求仍然知之甚少。由于立克次体必须存在于细胞内的特性所带来的技术难度,缺乏有效的遗传操作工具一直是阻碍人们了解立克次体的一个主要障碍。为此,我们利用 Tet-On 系统实现了公园立克次体的条件基因表达。利用 Tet-On,我们展示了抗生素耐药性和荧光报告基因的诱导表达。我们还利用这种诱导启动子筛选了立克次体表达四种催化死亡 Cas9(dCas9)变体的能力。我们证明了所有四种 dCas9 变体都能在 R. parkeri 中表达,并用于 CRISPR 干扰(CRISPRi)介导的靶向基因敲除。我们展示了抗生素抗性基因和内源性毒力因子 sca2 的靶向基因敲除。总之,我们首次在立克次体中开发出了诱导基因表达和 CRISPRi 介导的基因敲除系统,为对立克次体的感染性生命周期进行更多可扩展的、有针对性的机理研究奠定了基础。由于立克次体必须存在于细胞内,因此遗传操作工具的开发一直处于停滞状态,人们对其感染性生命周期的分子和遗传基础仍然知之甚少。在这里,我们引入了条件基因表达系统和 CRISPR 干扰(CRISPRi)介导的基因敲除系统,从而扩展了遗传工具包。这些系统可以相对容易地操纵立克次体的基因表达。我们利用 CRISPRi 破坏细胞内的生命周期,删除 sca2 毒力因子,证明了这些工具的有效性。这些工具对于更全面、更详细地了解立克次体生物学和致病机理至关重要。
{"title":"An expanded genetic toolkit for inducible expression and targeted gene silencing in <i>Rickettsia parkeri</i>.","authors":"Jon McGinn, Annie Wen, Desmond L Edwards, David M Brinkley, Rebecca L Lamason","doi":"10.1128/jb.00091-24","DOIUrl":"10.1128/jb.00091-24","url":null,"abstract":"<p><p>Pathogenic species within the <i>Rickettsia</i> genus are transmitted to humans through arthropod vectors and cause a spectrum of diseases ranging from mild to life-threatening. Despite rickettsiae posing an emerging global health risk, the genetic requirements of their infectious life cycles remain poorly understood. A major hurdle toward building this understanding has been the lack of efficient tools for genetic manipulation, owing to the technical difficulties associated with their obligate intracellular nature. To this end, we implemented the Tet-On system to enable conditional gene expression in <i>Rickettsia parkeri</i>. Using Tet-On, we show inducible expression of antibiotic resistance and a fluorescent reporter. We further used this inducible promoter to screen the ability of <i>R. parkeri</i> to express four variants of the catalytically dead Cas9 (dCas9). We demonstrate that all four dCas9 variants can be expressed in <i>R. parkeri</i> and used for CRISPR interference (CRISPRi)-mediated targeted gene knockdown. We show targeted knockdown of an antibiotic resistance gene as well as the endogenous virulence factor <i>sca2</i>. Altogether, we have developed systems for inducible gene expression and CRISPRi-mediated gene knockdown for the first time in rickettsiae, laying the groundwork for more scalable, targeted mechanistic investigations into their infectious life cycles.IMPORTANCEThe spotted fever group of <i>Rickettsia</i> contains vector-borne pathogenic bacteria that are neglected and emerging threats to public health. Due to the obligate intracellular nature of rickettsiae, the development of tools for genetic manipulation has been stunted, and the molecular and genetic underpinnings of their infectious lifecycle remain poorly understood. Here, we expand the genetic toolkit by introducing systems for conditional gene expression and CRISPR interference (CRISPRi)-mediated gene knockdown. These systems allow for relatively easy manipulation of rickettsial gene expression. We demonstrate the effectiveness of these tools by disrupting the intracellular life cycle using CRISPRi to deplete the <i>sca2</i> virulence factor. These tools will be crucial for building a more comprehensive and detailed understanding of rickettsial biology and pathogenesis.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270864/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141261550","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Guarding the walls: the multifaceted roles of Bce modules in cell envelope stress sensing and antimicrobial resistance. 守护细胞壁:Bce 模块在细胞包膜应激传感和抗菌性中的多方面作用。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-13 DOI: 10.1128/jb.00123-24
Natasha L George, Ellen C Bennett, Benjamin J Orlando

Bacteria have developed diverse strategies for defending their cell envelopes from external threats. In Firmicutes, one widespread strategy is to use Bce modules-membrane protein complexes that unite a peptide-detoxifying ABC transporter with a stress response coordinating two-component system. These modules provide specific, front-line defense for a wide variety of antimicrobial peptides and small molecule antibiotics as well as coordinate responses for heat, acid, and oxidative stress. Because of these abilities, Bce modules play important roles in virulence and the development of antibiotic resistance in a variety of pathogens, including Staphylococcus, Streptococcus, and Enterococcus species. Despite their importance, Bce modules are still poorly understood, with scattered functional data in only a small number of species. In this review, we will discuss Bce module structure in light of recent cryo-electron microscopy structures of the B. subtilis BceABRS module and explore the common threads and variations-on-a-theme in Bce module mechanisms across species. We also highlight the many remaining questions about Bce module function. Understanding these multifunctional membrane complexes will enhance our understanding of bacterial stress sensing and may point toward new therapeutic targets for highly resistant pathogens.

细菌开发了多种策略来保护其细胞膜免受外部威胁。在真菌中,一种普遍的策略是使用 Bce 模块--将肽解毒 ABC 转运体与应激反应协调双组分系统结合在一起的膜蛋白复合物。这些模块可为多种抗菌肽和小分子抗生素提供特异性前线防御,并协调对热、酸和氧化应激的反应。由于具有这些能力,Bce 模块在多种病原体(包括葡萄球菌、链球菌和肠球菌)的毒力和抗生素耐药性发展中发挥了重要作用。尽管 Bce 模块非常重要,但人们对它们的了解仍然很少,只有少数物种中存在零散的功能数据。在这篇综述中,我们将根据最近对枯草杆菌 BceABRS 模块的冷冻电镜结构来讨论 Bce 模块的结构,并探讨不同物种 Bce 模块机制的共同点和主题变异。我们还强调了有关 Bce 模块功能的许多遗留问题。了解这些多功能膜复合物将加深我们对细菌应激传感的理解,并可能为高抗药性病原体找到新的治疗目标。
{"title":"Guarding the walls: the multifaceted roles of Bce modules in cell envelope stress sensing and antimicrobial resistance.","authors":"Natasha L George, Ellen C Bennett, Benjamin J Orlando","doi":"10.1128/jb.00123-24","DOIUrl":"10.1128/jb.00123-24","url":null,"abstract":"<p><p>Bacteria have developed diverse strategies for defending their cell envelopes from external threats. In Firmicutes, one widespread strategy is to use Bce modules-membrane protein complexes that unite a peptide-detoxifying ABC transporter with a stress response coordinating two-component system. These modules provide specific, front-line defense for a wide variety of antimicrobial peptides and small molecule antibiotics as well as coordinate responses for heat, acid, and oxidative stress. Because of these abilities, Bce modules play important roles in virulence and the development of antibiotic resistance in a variety of pathogens, including <i>Staphylococcus</i>, <i>Streptococcus</i>, and <i>Enterococcus</i> species. Despite their importance, Bce modules are still poorly understood, with scattered functional data in only a small number of species. In this review, we will discuss Bce module structure in light of recent cryo-electron microscopy structures of the <i>B. subtilis</i> BceABRS module and explore the common threads and variations-on-a-theme in Bce module mechanisms across species. We also highlight the many remaining questions about Bce module function. Understanding these multifunctional membrane complexes will enhance our understanding of bacterial stress sensing and may point toward new therapeutic targets for highly resistant pathogens.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270860/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141310749","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Identification of a copper-responsive small molecule inhibitor of uropathogenic Escherichia coli. 鉴定尿路致病性大肠杆菌的铜反应小分子抑制剂。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-10 DOI: 10.1128/jb.00112-24
Braden S Hanson, Amanuel Hailemariam, Yongjian Yang, Faras Mohamed, George L Donati, Dwight Baker, James Sacchettini, James J Cai, Sargurunathan Subashchandrabose

Urinary tract infections (UTIs) are a major global health problem and are caused predominantly by uropathogenic Escherichia coli (UPEC). UTIs are a leading cause of prescription antimicrobial use. Incessant increase in antimicrobial resistance in UPEC and other uropathogens poses a serious threat to the current treatment practices. Copper is an effector of nutritional immunity that impedes the growth of pathogens during infection. We hypothesized that copper would augment the toxicity of select small molecules against bacterial pathogens. We conducted a small molecule screening campaign with a library of 51,098 molecules to detect hits that inhibit a UPEC ΔtolC mutant in a copper-dependent manner. A molecule, denoted as E. coli inhibitor or ECIN, was identified as a copper-responsive inhibitor of wild-type UPEC strains. Our gene expression and metal content analysis results demonstrate that ECIN works in concert with copper to exacerbate Cu toxicity in UPEC. ECIN has a broad spectrum of activity against pathogens of medical and veterinary significance including Acinetobacter baumannii, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus. Subinhibitory levels of ECIN eliminate UPEC biofilm formation. Transcriptome analysis of UPEC treated with ECIN reveals induction of multiple stress response systems. Furthermore, we demonstrate that L-cysteine rescues the growth of UPEC exposed to ECIN. In summary, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC.IMPORTANCEUrinary tract infection (UTI) is a ubiquitous infectious condition affecting millions of people annually. Uropathogenic Escherichia coli (UPEC) is the predominant etiological agent of UTI. However, UTIs are becoming increasingly difficult to resolve with antimicrobials due to increased antimicrobial resistance in UPEC and other uropathogens. Here, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC. In addition to E. coli, this small molecule also inhibits pathogens of medical and veterinary significance including Acinetobacter baumannii, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus.

尿路感染(UTI)是一个重大的全球性健康问题,主要由尿路致病性大肠杆菌(UPEC)引起。尿路感染是处方抗菌药使用的主要原因。尿路感染大肠杆菌和其他尿路病原体对抗菌素的耐药性不断增加,对目前的治疗方法构成了严重威胁。铜是一种营养免疫效应物质,能在感染期间阻碍病原体的生长。我们假设铜会增强特定小分子对细菌病原体的毒性。我们利用一个包含 51,098 个分子的小分子化合物库进行了一次小分子筛选,以发现能以铜依赖性方式抑制 UPEC ΔtolC 突变体的小分子。结果发现了一种对野生型 UPEC 菌株具有铜响应抑制作用的分子,命名为大肠杆菌抑制剂或 ECIN。我们的基因表达和金属含量分析结果表明,ECIN与铜协同作用,加剧了UPEC的铜毒性。ECIN 对医用和兽用病原体具有广谱活性,包括鲍曼不动杆菌、铜绿假单胞菌和耐甲氧西林金黄色葡萄球菌。亚抑制水平的 ECIN 可消除 UPEC 生物膜的形成。用 ECIN 处理 UPEC 的转录组分析表明,ECIN 诱导了多种应激反应系统。此外,我们还证明,L-半胱氨酸能挽救暴露于 ECIN 的 UPEC 的生长。重要意义泌尿道感染(UTI)是一种无处不在的传染性疾病,每年影响数百万人。尿路致病性大肠杆菌(UPEC)是UTI的主要病原体。然而,由于 UPEC 和其他尿路病原体对抗菌药的耐药性增加,抗菌药越来越难以解决 UTI 问题。在此,我们报告了一种新型铜响应小分子 UPEC 抑制剂的鉴定和表征。除大肠杆菌外,这种小分子还能抑制医疗和兽医领域的病原体,包括鲍曼不动杆菌、铜绿假单胞菌和耐甲氧西林金黄色葡萄球菌。
{"title":"Identification of a copper-responsive small molecule inhibitor of uropathogenic <i>Escherichia coli</i>.","authors":"Braden S Hanson, Amanuel Hailemariam, Yongjian Yang, Faras Mohamed, George L Donati, Dwight Baker, James Sacchettini, James J Cai, Sargurunathan Subashchandrabose","doi":"10.1128/jb.00112-24","DOIUrl":"10.1128/jb.00112-24","url":null,"abstract":"<p><p>Urinary tract infections (UTIs) are a major global health problem and are caused predominantly by uropathogenic <i>Escherichia coli</i> (UPEC). UTIs are a leading cause of prescription antimicrobial use. Incessant increase in antimicrobial resistance in UPEC and other uropathogens poses a serious threat to the current treatment practices. Copper is an effector of nutritional immunity that impedes the growth of pathogens during infection. We hypothesized that copper would augment the toxicity of select small molecules against bacterial pathogens. We conducted a small molecule screening campaign with a library of 51,098 molecules to detect hits that inhibit a UPEC Δ<i>tolC</i> mutant in a copper-dependent manner. A molecule, denoted as <i>E. coli</i> inhibitor or ECIN, was identified as a copper-responsive inhibitor of wild-type UPEC strains. Our gene expression and metal content analysis results demonstrate that ECIN works in concert with copper to exacerbate Cu toxicity in UPEC. ECIN has a broad spectrum of activity against pathogens of medical and veterinary significance including <i>Acinetobacter baumannii, Pseudomonas aeruginosa</i>, and methicillin-resistant <i>Staphylococcus aureus</i>. Subinhibitory levels of ECIN eliminate UPEC biofilm formation. Transcriptome analysis of UPEC treated with ECIN reveals induction of multiple stress response systems. Furthermore, we demonstrate that L-cysteine rescues the growth of UPEC exposed to ECIN. In summary, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC.IMPORTANCEUrinary tract infection (UTI) is a ubiquitous infectious condition affecting millions of people annually. Uropathogenic <i>Escherichia coli</i> (UPEC) is the predominant etiological agent of UTI. However, UTIs are becoming increasingly difficult to resolve with antimicrobials due to increased antimicrobial resistance in UPEC and other uropathogens. Here, we report the identification and characterization of a novel copper-responsive small molecule inhibitor of UPEC. In addition to <i>E. coli</i>, this small molecule also inhibits pathogens of medical and veterinary significance including <i>Acinetobacter baumannii, Pseudomonas aeruginosa,</i> and methicillin-resistant <i>Staphylococcus aureus</i>.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270900/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141296058","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Non-interchangeable functions of efflux transporters of Pseudomonas aeruginosa in survival under infection-associated stress. 铜绿假单胞菌的外排转运体在感染相关压力下的生存过程中具有不可互换的功能。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-14 DOI: 10.1128/jb.00054-24
Justyna W Adamiak, Laiba Ajmal, Helen I Zgurskaya

Pseudomonas aeruginosa is a challenging opportunistic pathogen due to its intrinsic and acquired mechanisms of antibiotic resistance. A large repertoire of efflux transporters actively expels antibiotics, toxins, and metabolites from cells and enables growth of P. aeruginosa in diverse environments. In this study, we analyzed the roles of representative efflux pumps from the Resistance-Nodulation-Division (RND), Major Facilitator Superfamily (MFS), and Small Multidrug Resistance (SMR) families of proteins in the susceptibility of P. aeruginosa to antibiotics and bacterial growth under stresses imposed by human hosts during bacterial infections: an elevated temperature, osmotic stress, low iron, bile salts, and acidic pH. We selected five RND pumps MexAB-OprM, MexEF-OprN, MexCD-OprJ, MuxABC-OpmB, and TriABC-OpmH that differ in their substrate specificities and expression profiles, two MFS efflux pumps PA3136-3137 and PA5158-5160 renamed here into MfsAB and MfsCD-OpmG, respectively, and an SMR efflux transporter PA1540-1541 (MdtJI). We found that the most promiscuous RND pumps such as MexEF-OprN and MexAB-OprM are integrated into diverse survival mechanisms and enable P. aeruginosa growth under various stresses. MuxABC-OpmB and TriABC-OpmH pumps with narrower substrate spectra are beneficial only in the presence of the iron chelator 2,2'-dipyridyl and bile salts, respectively. MFS pumps do not contribute to antibiotic efflux but play orthogonal roles in acidic pH, low iron, and in the presence of bile salts. In contrast, MdtJI protects against polycationic antibiotics but does not contribute to survival under stress. Thus, efflux pumps play specific, non-interchangeable functions in P. aeruginosa cell physiology and bacterial survival under stresses.

Importance: The role of multidrug efflux pumps in the intrinsic and clinical levels of antibiotic resistance in Pseudomonas aeruginosa and other gram-negative bacteria is well-established. Their functions in bacterial physiology, however, remain unclear. The P. aeruginosa genome comprises an arsenal of efflux pumps from different protein families, the substrate specificities of which are typically assessed by measuring their impact on susceptibility to antibiotics. In this study, we analyzed how deletions and overproductions of efflux pumps affect P. aeruginosa growth under human-infection-induced stresses. Our results show that the physiological functions of multidrug efflux pumps are non-redundant and essential for the survival of this important human pathogen under stress.

铜绿假单胞菌因其固有的和后天获得的抗生素耐药性机制而成为一种具有挑战性的机会性病原体。大量的外排转运体积极地将抗生素、毒素和代谢物排出细胞,使铜绿假单胞菌能够在不同的环境中生长。在本研究中,我们分析了抗性-结节-分裂(RND)、主要促进剂超家族(MFS)和小型多药耐药性(SMR)蛋白家族中具有代表性的外排泵在铜绿假单胞菌对抗生素的敏感性以及细菌感染期间人类宿主施加的压力(温度升高、渗透压力、低铁、胆汁盐和酸性 pH 值)下细菌生长中的作用。我们选择了底物特异性和表达谱不同的五个 RND 泵 MexAB-OprM、MexEF-OprN、MexCD-OprJ、MuxABC-OpmB 和 TriABC-OpmH,两个 MFS 外排泵 PA3136-3137 和 PA5158-5160(在此分别重命名为 MfsAB 和 MfsCD-OpmG),以及一个 SMR 外排转运体 PA1540-1541 (MdtJI)。我们发现最杂乱的 RND 泵,如 MexEF-OprN 和 MexAB-OprM,融入了多种生存机制,使铜绿微囊藻在各种压力下都能生长。底物谱较窄的 MuxABC-OpmB 和 TriABC-OpmH 泵分别只在铁螯合剂 2,2'-dipyridyl 和胆汁盐存在时才有益。MFS 泵无助于抗生素外流,但在酸性 pH 值、低铁和有胆汁盐存在时发挥正交作用。相反,MdtJI 可抵御多阳离子抗生素,但无助于在压力下存活。因此,外排泵在铜绿微囊藻细胞生理和细菌在压力下的生存中发挥着特定的、不可互换的功能:多药外排泵在铜绿假单胞菌和其他革兰氏阴性菌的内在和临床抗生素耐药性中的作用已得到公认。然而,它们在细菌生理学中的功能仍不清楚。铜绿假单胞菌的基因组包括来自不同蛋白家族的外排泵,其底物特异性通常通过测量它们对抗生素敏感性的影响来评估。在这项研究中,我们分析了外排泵的缺失和过量产生如何影响铜绿假单胞菌在人类感染诱导的压力下的生长。我们的研究结果表明,多药外排泵的生理功能是非冗余的,对于这种重要的人类病原体在压力下的生存至关重要。
{"title":"Non-interchangeable functions of efflux transporters of <i>Pseudomonas aeruginosa</i> in survival under infection-associated stress.","authors":"Justyna W Adamiak, Laiba Ajmal, Helen I Zgurskaya","doi":"10.1128/jb.00054-24","DOIUrl":"10.1128/jb.00054-24","url":null,"abstract":"<p><p><i>Pseudomonas aeruginosa</i> is a challenging opportunistic pathogen due to its intrinsic and acquired mechanisms of antibiotic resistance. A large repertoire of efflux transporters actively expels antibiotics, toxins, and metabolites from cells and enables growth of <i>P. aeruginosa</i> in diverse environments. In this study, we analyzed the roles of representative efflux pumps from the Resistance-Nodulation-Division (RND), Major Facilitator Superfamily (MFS), and Small Multidrug Resistance (SMR) families of proteins in the susceptibility of <i>P. aeruginosa</i> to antibiotics and bacterial growth under stresses imposed by human hosts during bacterial infections: an elevated temperature, osmotic stress, low iron, bile salts, and acidic pH. We selected five RND pumps MexAB-OprM, MexEF-OprN, MexCD-OprJ, MuxABC-OpmB, and TriABC-OpmH that differ in their substrate specificities and expression profiles, two MFS efflux pumps PA3136-3137 and PA5158-5160 renamed here into MfsAB and MfsCD-OpmG, respectively, and an SMR efflux transporter PA1540-1541 (MdtJI). We found that the most promiscuous RND pumps such as MexEF-OprN and MexAB-OprM are integrated into diverse survival mechanisms and enable <i>P. aeruginosa</i> growth under various stresses. MuxABC-OpmB and TriABC-OpmH pumps with narrower substrate spectra are beneficial only in the presence of the iron chelator 2,2'-dipyridyl and bile salts, respectively. MFS pumps do not contribute to antibiotic efflux but play orthogonal roles in acidic pH, low iron, and in the presence of bile salts. In contrast, MdtJI protects against polycationic antibiotics but does not contribute to survival under stress. Thus, efflux pumps play specific, non-interchangeable functions in <i>P. aeruginosa</i> cell physiology and bacterial survival under stresses.</p><p><strong>Importance: </strong>The role of multidrug efflux pumps in the intrinsic and clinical levels of antibiotic resistance in <i>Pseudomonas aeruginosa</i> and other gram-negative bacteria is well-established. Their functions in bacterial physiology, however, remain unclear. The <i>P. aeruginosa</i> genome comprises an arsenal of efflux pumps from different protein families, the substrate specificities of which are typically assessed by measuring their impact on susceptibility to antibiotics. In this study, we analyzed how deletions and overproductions of efflux pumps affect <i>P. aeruginosa</i> growth under human-infection-induced stresses. Our results show that the physiological functions of multidrug efflux pumps are non-redundant and essential for the survival of this important human pathogen under stress.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11323973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141317426","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Comprehensive analysis of insertion sequences within rRNA genes of CPR bacteria and biochemical characterization of a homing endonuclease encoded by these sequences. 全面分析 CPR 细菌 rRNA 基因内的插入序列以及这些序列编码的归巢内切酶的生化特征。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-10 DOI: 10.1128/jb.00074-24
Megumi Tsurumaki, Asako Sato, Motofumi Saito, Akio Kanai

The Candidate Phyla Radiation (CPR) represents an extensive bacterial clade comprising primarily uncultured lineages and is distinguished from other bacteria by a significant prevalence of insertion sequences (ISs) within their rRNA genes. However, our understanding of the taxonomic distribution and characteristics of these ISs remains limited. In this study, we used a comprehensive approach to systematically determine the nature of the rRNA ISs in CPR bacteria. The analysis of hundreds of rRNA gene sequences across 65 CPR phyla revealed that ISs are present in 48% of 16S rRNA genes and 82% of 23S rRNA genes, indicating a broad distribution across the CPR clade, with exceptions in the 16S and 23S rRNA genes of Candidatus (Ca.) Saccharibacteria and the 16S rRNA genes of Ca. Peregrinibacteria. Over half the ISs display a group-I-intron-like structure, whereas specific 16S rRNA gene ISs display features reminiscent of group II introns. The ISs frequently encode proteins with homing endonuclease (HE) domains, centered around the LAGLIDADG motif. The LAGLIDADG HE (LHE) proteins encoded by the rRNA ISs of CPR bacteria predominantly have a single-domain structure, deviating from the usual single- or double-domain configuration observed in typical prokaryotic LHEs. Experimental analysis of one LHE protein, I-ShaI from Ca. Shapirobacteria, confirmed that its endonuclease activity targets the DNA sequence of its insertion site, and chemical cross-linking experiments demonstrated its capacity to form homodimers. These results provide robust evidence supporting the hypothesis that the explosive proliferation of rRNA ISs in CPR bacteria was facilitated by mechanisms involving LHEs.

Importance: Insertion sequences (ISs) in rRNA genes are relatively limited and infrequent in most bacterial phyla. With a comprehensive bioinformatic analysis, we show that in CPR bacteria, these ISs occur in 48% of 16S rRNA genes and 82% of 23S rRNA genes. We also report the systematic and biochemical characterization of the LAGLIDADG homing endonucleases (LHEs) encoded by these ISs in the first such analysis of the CPR bacteria. This study significantly extends our understanding of the phylogenetic positions of rRNA ISs within CPR bacteria and the biochemical features of their LHEs.

候选菌系辐射(CPR)是一个广泛的细菌支系,主要由未培养的菌系组成,其 rRNA 基因中插入序列(ISs)的大量存在使其有别于其他细菌。然而,我们对这些 ISs 的分类分布和特征的了解仍然有限。在本研究中,我们采用了一种全面的方法来系统地确定 CPR 细菌中 rRNA ISs 的性质。对 65 个 CPR 支系的数百个 rRNA 基因序列进行分析后发现,48% 的 16S rRNA 基因和 82% 的 23S rRNA 基因中存在 ISs,这表明 ISs 在 CPR 支系中分布广泛,但在 Candidatus (Ca.) Saccharibacteria 的 16S 和 23S rRNA 基因以及 Ca.Peregrinibacteria 的 16S rRNA 基因。半数以上的 ISs 显示出类似于 I 组内含子的结构,而特定的 16S rRNA 基因 ISs 则显示出类似于 II 组内含子的特征。ISs 经常编码以 LAGLIDADG 主题为中心的具有归巢内切酶(HE)结构域的蛋白质。由 CPR 细菌 rRNA ISs 编码的 LAGLIDADG HE(LHE)蛋白主要具有单链结构,不同于在典型原核生物 LHE 中观察到的通常的单链或双链结构。对来自 Ca.Shapirobacteria 中的一种 LHE 蛋白 I-ShaI 的实验分析证实,它的内切酶活性以其插入位点的 DNA 序列为目标,化学交联实验证明了它形成同源二聚体的能力。这些结果为以下假设提供了有力证据:CPR 细菌中 rRNA ISs 的爆炸性增殖是由涉及 LHEs 的机制促成的:在大多数细菌门中,rRNA 基因中的插入序列(ISs)相对有限且不常见。通过全面的生物信息学分析,我们发现在 CPR 细菌中,48% 的 16S rRNA 基因和 82% 的 23S rRNA 基因中都存在插入序列。我们还报告了由这些 ISs 编码的 LAGLIDADG 同源内切酶(LHEs)的系统和生化特征,这是首次对 CPR 细菌进行此类分析。这项研究极大地扩展了我们对 rRNA ISs 在 CPR 细菌中的系统发育位置及其 LHEs 的生化特征的了解。
{"title":"Comprehensive analysis of insertion sequences within rRNA genes of CPR bacteria and biochemical characterization of a homing endonuclease encoded by these sequences.","authors":"Megumi Tsurumaki, Asako Sato, Motofumi Saito, Akio Kanai","doi":"10.1128/jb.00074-24","DOIUrl":"10.1128/jb.00074-24","url":null,"abstract":"<p><p>The Candidate Phyla Radiation (CPR) represents an extensive bacterial clade comprising primarily uncultured lineages and is distinguished from other bacteria by a significant prevalence of insertion sequences (ISs) within their rRNA genes. However, our understanding of the taxonomic distribution and characteristics of these ISs remains limited. In this study, we used a comprehensive approach to systematically determine the nature of the rRNA ISs in CPR bacteria. The analysis of hundreds of rRNA gene sequences across 65 CPR phyla revealed that ISs are present in 48% of 16S rRNA genes and 82% of 23S rRNA genes, indicating a broad distribution across the CPR clade, with exceptions in the 16S and 23S rRNA genes of <i>Candidatus</i> (<i>Ca</i>.) Saccharibacteria and the 16S rRNA genes of <i>Ca</i>. Peregrinibacteria. Over half the ISs display a group-I-intron-like structure, whereas specific 16S rRNA gene ISs display features reminiscent of group II introns. The ISs frequently encode proteins with homing endonuclease (HE) domains, centered around the LAGLIDADG motif. The LAGLIDADG HE (LHE) proteins encoded by the rRNA ISs of CPR bacteria predominantly have a single-domain structure, deviating from the usual single- or double-domain configuration observed in typical prokaryotic LHEs. Experimental analysis of one LHE protein, I-ShaI from <i>Ca</i>. Shapirobacteria, confirmed that its endonuclease activity targets the DNA sequence of its insertion site, and chemical cross-linking experiments demonstrated its capacity to form homodimers. These results provide robust evidence supporting the hypothesis that the explosive proliferation of rRNA ISs in CPR bacteria was facilitated by mechanisms involving LHEs.</p><p><strong>Importance: </strong>Insertion sequences (ISs) in rRNA genes are relatively limited and infrequent in most bacterial phyla. With a comprehensive bioinformatic analysis, we show that in CPR bacteria, these ISs occur in 48% of 16S rRNA genes and 82% of 23S rRNA genes. We also report the systematic and biochemical characterization of the LAGLIDADG homing endonucleases (LHEs) encoded by these ISs in the first such analysis of the CPR bacteria. This study significantly extends our understanding of the phylogenetic positions of rRNA ISs within CPR bacteria and the biochemical features of their LHEs.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270868/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141296057","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Dpr-mediated H2O2 resistance contributes to streptococcus survival in a cystic fibrosis airway model system. Dpr 介导的 H2O2 抗性有助于链球菌在囊性纤维化气道模型系统中存活。
IF 2.7 3区 生物学 Q3 MICROBIOLOGY Pub Date : 2024-07-25 Epub Date: 2024-06-28 DOI: 10.1128/jb.00176-24
Rendi R Rogers, Christopher A Kesthely, Fabrice Jean-Pierre, Bassam El Hafi, George A O'Toole

The cystic fibrosis (CF) lung environment is conducive to the colonization of bacteria as polymicrobial biofilms, which are associated with poor clinical outcomes for persons with CF (pwCF). Streptococcus spp. are highly prevalent in the CF airway, but its role in the CF lung microbiome is poorly understood. Some studies have shown Streptococcus spp. to be associated with better clinical outcomes for pwCF, while others show that high abundance of Streptococcus spp. is correlated with exacerbations. Our lab previously reported a polymicrobial culture system consisting of four CF-relevant pathogens that can be used to study microbial behavior in a more clinically relevant setting. Here, we use this model system to identify genetic pathways that are important for Streptococcus sanguinis survival in the context of the polymicrobial community. We identified genes related to reactive oxygen species as differentially expressed in S. sanguinis monoculture versus growth of this microbe in the mixed community. Genetic studies identified Dpr as important for S. sanguinis survival in the community. We show that Dpr, a DNA-binding ferritin-like protein, and PerR, a peroxide-responsive transcriptional regulator of Dpr, are important for protecting S. sanguinis from phenazine-mediated toxicity in co-culture with Pseudomonas aeruginosa and when exposed to hydrogen peroxide, both of which mimic the CF lung environment. Characterizing such interactions in a clinically relevant model system contributes to our understanding of microbial behavior in the context of polymicrobial biofilm infections.

Importance: Streptococcus spp. are recognized as a highly prevalent pathogen in cystic fibrosis (CF) airway infections. However, the role of this microbe in clinical outcomes for persons with CF is poorly understood. Here, we leverage a polymicrobial community system previously developed by our group to model CF airway infections as a tool to investigate a Pseudomonas-Streptococcus interaction involving reactive oxygen species (ROS). We show that protection against ROS is required for Streptococcus sanguinis survival in a clinically relevant polymicrobial system. Using this model system to study interspecies interactions contributes to our broader understanding of the complex role of Streptococcus spp. in the CF lung.

囊性纤维化(CF)肺部环境有利于细菌定植为多微生物生物膜,而多微生物生物膜与 CF 患者(pwCF)的不良临床预后有关。链球菌在 CF 气道中非常普遍,但其在 CF 肺微生物组中的作用却鲜为人知。一些研究表明,链球菌与患者更好的临床预后有关,而另一些研究则表明,链球菌的高丰度与病情恶化有关。我们实验室以前曾报道过一种由四种与 CF 相关的病原体组成的多微生物培养系统,可用于在更贴近临床的环境中研究微生物行为。在此,我们利用这一模型系统来确定在多微生物群落背景下对血清链球菌生存至关重要的遗传途径。我们确定了与活性氧有关的基因,这些基因在血链球菌单一培养与该微生物在混合群落中生长时表达不同。基因研究发现 Dpr 对 S. sanguinis 在群落中的生存非常重要。我们的研究表明,Dpr(一种 DNA 结合铁蛋白样蛋白)和 PerR(Dpr 的过氧化物反应转录调节因子)对于保护 S. sanguinis 在与铜绿假单胞菌共培养和暴露于过氧化氢(两者都模拟了 CF 肺部环境)时免受酚嗪介导的毒性作用非常重要。在临床相关模型系统中描述这种相互作用有助于我们了解多微生物生物膜感染背景下的微生物行为:链球菌被认为是囊性纤维化(CF)气道感染的高发病原体。然而,人们对这种微生物在 CF 患者临床治疗中的作用却知之甚少。在这里,我们利用本研究小组先前开发的多微生物群落系统来模拟 CF 气道感染,并以此为工具研究假单胞菌与链球菌之间涉及活性氧(ROS)的相互作用。我们的研究表明,在临床相关的多微生物系统中,血清链球菌的存活需要对 ROS 的保护。利用这一模型系统研究种间相互作用有助于我们更广泛地了解链球菌在 CF 肺中的复杂作用。
{"title":"Dpr-mediated H<sub>2</sub>O<sub>2</sub> resistance contributes to streptococcus survival in a cystic fibrosis airway model system.","authors":"Rendi R Rogers, Christopher A Kesthely, Fabrice Jean-Pierre, Bassam El Hafi, George A O'Toole","doi":"10.1128/jb.00176-24","DOIUrl":"10.1128/jb.00176-24","url":null,"abstract":"<p><p>The cystic fibrosis (CF) lung environment is conducive to the colonization of bacteria as polymicrobial biofilms, which are associated with poor clinical outcomes for persons with CF (pwCF). <i>Streptococcus</i> spp. are highly prevalent in the CF airway, but its role in the CF lung microbiome is poorly understood. Some studies have shown <i>Streptococcus</i> spp. to be associated with better clinical outcomes for pwCF, while others show that high abundance of <i>Streptococcus</i> spp. is correlated with exacerbations. Our lab previously reported a polymicrobial culture system consisting of four CF-relevant pathogens that can be used to study microbial behavior in a more clinically relevant setting. Here, we use this model system to identify genetic pathways that are important for <i>Streptococcus sanguinis</i> survival in the context of the polymicrobial community. We identified genes related to reactive oxygen species as differentially expressed in <i>S. sanguinis</i> monoculture versus growth of this microbe in the mixed community. Genetic studies identified Dpr as important for <i>S. sanguinis</i> survival in the community. We show that Dpr, a DNA-binding ferritin-like protein, and PerR, a peroxide-responsive transcriptional regulator of Dpr, are important for protecting <i>S. sanguinis</i> from phenazine-mediated toxicity in co-culture with <i>Pseudomonas aeruginosa</i> and when exposed to hydrogen peroxide, both of which mimic the CF lung environment. Characterizing such interactions in a clinically relevant model system contributes to our understanding of microbial behavior in the context of polymicrobial biofilm infections.</p><p><strong>Importance: </strong><i>Streptococcus</i> spp. are recognized as a highly prevalent pathogen in cystic fibrosis (CF) airway infections. However, the role of this microbe in clinical outcomes for persons with CF is poorly understood. Here, we leverage a polymicrobial community system previously developed by our group to model CF airway infections as a tool to investigate a <i>Pseudomonas</i>-<i>Streptococcus</i> interaction involving reactive oxygen species (ROS). We show that protection against ROS is required for <i>Streptococcus sanguinis</i> survival in a clinically relevant polymicrobial system. Using this model system to study interspecies interactions contributes to our broader understanding of the complex role of <i>Streptococcus</i> spp. in the CF lung.</p>","PeriodicalId":15107,"journal":{"name":"Journal of Bacteriology","volume":null,"pages":null},"PeriodicalIF":2.7,"publicationDate":"2024-07-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11270861/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141468181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Journal of Bacteriology
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