首页 > 最新文献

Molecular Oral Microbiology最新文献

英文 中文
Oral biofilm dysbiosis during experimental periodontitis. 实验性牙周炎期间的口腔生物膜菌群失调。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-12-01 Epub Date: 2022-10-19 DOI: 10.1111/omi.12389
Apoena Aguiar Ribeiro, Yizu Jiao, Mustafa Girnary, Tomaz Alves, Liang Chen, Anna Farrell, Di Wu, Flavia Teles, Naohiro Inohara, Karen V Swanson, Julie T Marchesan

Objectives: We have previously characterized the main osteoimmunological events that occur during ligature periodontitis. This study aims to determine the polymicrobial community shifts that occur during disease development.

Methods: Periodontitis was induced in C57BL/6 mice using the ligature-induced periodontitis model. Healthy oral mucosa swabs and ligatures were collected every 3 days from 0 to 18 days post-ligature placement. Biofilm samples were evaluated by 16SrRNA gene sequencing (Illumina MiSeq) and QIIME. Time-course changes were determined by relative abundance, diversity, and rank analyses (PERMANOVA, Bonferroni-adjusted).

Results: Microbial differences between health and periodontal inflammation were observed at all phylogenic levels. An evident microbial community shift occurred in 25 genera during the advancement of "gingivitis" (3-6 days) to periodontitis (9-18 days). From day 0 to 18, dramatic changes were identified in Streptococcus levels, with an overall decrease (54.04%-0.02%) as well an overall increase of Enterococcus and Lactobacillus (23.7%-73.1% and 10.1%-70.2%, respectively). Alpha-diversity decreased to its lowest at 3 days, followed by an increase in diversity as disease advancement. Beta-diversity increased after ligature placement, indicating that bone loss develops in response to a greater microbial variability (p = 0.001). Levels of facultative and strict anaerobic bacteria augmented over the course of disease progression, with a total of eight species significantly different during the 18-day period.

Conclusion: The data supports that murine gingival inflammation and alveolar bone loss develop in response to microbiome shifts. Bacterial diversity increased during progression to bone loss. These findings further support the utilization of the periodontitis ligature model for microbial shift analysis under different experimental conditions.

目的:我们曾描述过结扎性牙周炎期间发生的主要骨免疫学事件。本研究旨在确定疾病发展过程中发生的多微生物群落变化:方法:使用结扎诱导牙周炎模型诱导 C57BL/6 小鼠患牙周炎。从结扎后 0 到 18 天,每隔 3 天收集一次健康口腔黏膜拭子和结扎物。通过 16SrRNA 基因测序(Illumina MiSeq)和 QIIME 对生物膜样本进行评估。通过相对丰度、多样性和等级分析(PERMANOVA,Bonferroni-adjusted)确定时间变化:结果:在所有系统发生水平上都观察到了健康与牙周炎症之间的微生物差异。在从 "牙龈炎"(3-6 天)到牙周炎(9-18 天)的过程中,25 个属发生了明显的微生物群落变化。从第 0 天到第 18 天,链球菌的含量发生了巨大变化,总体下降(54.04%-0.02%),肠球菌和乳酸杆菌的含量总体上升(分别为 23.7%-73.1% 和 10.1%-70.2% )。α-多样性在 3 天时降至最低,随后随着病情的发展而增加。结扎后,β-多样性增加,表明骨质流失是对更大的微生物变异性的反应(p = 0.001)。随着疾病的发展,兼性厌氧菌和严格厌氧菌的数量也在增加,在 18 天的时间里,共有 8 个物种存在显著差异:结论:这些数据证实,小鼠牙龈炎症和牙槽骨缺失是随着微生物群的变化而发展的。细菌多样性在骨质流失过程中有所增加。这些发现进一步支持了利用牙周炎结扎模型在不同实验条件下进行微生物变迁分析。
{"title":"Oral biofilm dysbiosis during experimental periodontitis.","authors":"Apoena Aguiar Ribeiro, Yizu Jiao, Mustafa Girnary, Tomaz Alves, Liang Chen, Anna Farrell, Di Wu, Flavia Teles, Naohiro Inohara, Karen V Swanson, Julie T Marchesan","doi":"10.1111/omi.12389","DOIUrl":"10.1111/omi.12389","url":null,"abstract":"<p><strong>Objectives: </strong>We have previously characterized the main osteoimmunological events that occur during ligature periodontitis. This study aims to determine the polymicrobial community shifts that occur during disease development.</p><p><strong>Methods: </strong>Periodontitis was induced in C57BL/6 mice using the ligature-induced periodontitis model. Healthy oral mucosa swabs and ligatures were collected every 3 days from 0 to 18 days post-ligature placement. Biofilm samples were evaluated by 16SrRNA gene sequencing (Illumina MiSeq) and QIIME. Time-course changes were determined by relative abundance, diversity, and rank analyses (PERMANOVA, Bonferroni-adjusted).</p><p><strong>Results: </strong>Microbial differences between health and periodontal inflammation were observed at all phylogenic levels. An evident microbial community shift occurred in 25 genera during the advancement of \"gingivitis\" (3-6 days) to periodontitis (9-18 days). From day 0 to 18, dramatic changes were identified in Streptococcus levels, with an overall decrease (54.04%-0.02%) as well an overall increase of Enterococcus and Lactobacillus (23.7%-73.1% and 10.1%-70.2%, respectively). Alpha-diversity decreased to its lowest at 3 days, followed by an increase in diversity as disease advancement. Beta-diversity increased after ligature placement, indicating that bone loss develops in response to a greater microbial variability (p = 0.001). Levels of facultative and strict anaerobic bacteria augmented over the course of disease progression, with a total of eight species significantly different during the 18-day period.</p><p><strong>Conclusion: </strong>The data supports that murine gingival inflammation and alveolar bone loss develop in response to microbiome shifts. Bacterial diversity increased during progression to bone loss. These findings further support the utilization of the periodontitis ligature model for microbial shift analysis under different experimental conditions.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10034670/pdf/nihms-1872862.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9533033","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Electrochemical sensors for oral biofilm-biomaterials interface characterization: A review. 口腔生物膜-生物材料界面电化学传感器研究进展。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-12-01 Epub Date: 2022-11-15 DOI: 10.1111/omi.12396
Dipankar Koley

Important processes related to the interaction of the oral microbiome with the tooth surface happen directly at the interface. For example, the chemical microenvironment that exists at the interface of microbial biofilms and the native tooth structure is directly involved in caries development. Consequentially, a critical understanding of this interface and its chemical microenvironment would provide novel avenues in caries prevention, including secondary caries that often occurs at the interface of the dental biofilm, tooth structure, and dental material. Electrochemical sensors are a unique quantitative tool and have the inherent advantages of miniaturization, stability, and selectivity. That makes the electrochemical sensors ideal tools for studying these critical biofilm microenvironments with high precision. This review highlights the development and applications of several novel electrochemical sensors such as pH, Ca2+ , and hydrogen peroxide sensors as scanning electrochemical microscope probes in addition to flexible pH wire sensors for real-time bacterial biofilm-dental surface and dental materials interface studies.

口腔微生物群与牙齿表面相互作用的重要过程直接发生在界面上。例如,存在于微生物生物膜与原生牙齿结构界面的化学微环境直接参与龋病的发生。因此,对该界面及其化学微环境的批判性理解将为预防龋齿提供新的途径,包括经常发生在牙齿生物膜,牙齿结构和牙齿材料界面的继发性龋齿。电化学传感器是一种独特的定量工具,具有小型化、稳定性和选择性等固有优势。这使得电化学传感器成为高精度研究这些关键生物膜微环境的理想工具。本文综述了几种新型电化学传感器的发展和应用,如pH、Ca2+和过氧化氢传感器作为扫描电化学显微镜探针,以及柔性pH线传感器用于实时细菌生物膜-牙表面和牙材料界面的研究。
{"title":"Electrochemical sensors for oral biofilm-biomaterials interface characterization: A review.","authors":"Dipankar Koley","doi":"10.1111/omi.12396","DOIUrl":"10.1111/omi.12396","url":null,"abstract":"<p><p>Important processes related to the interaction of the oral microbiome with the tooth surface happen directly at the interface. For example, the chemical microenvironment that exists at the interface of microbial biofilms and the native tooth structure is directly involved in caries development. Consequentially, a critical understanding of this interface and its chemical microenvironment would provide novel avenues in caries prevention, including secondary caries that often occurs at the interface of the dental biofilm, tooth structure, and dental material. Electrochemical sensors are a unique quantitative tool and have the inherent advantages of miniaturization, stability, and selectivity. That makes the electrochemical sensors ideal tools for studying these critical biofilm microenvironments with high precision. This review highlights the development and applications of several novel electrochemical sensors such as pH, Ca<sup>2+</sup> , and hydrogen peroxide sensors as scanning electrochemical microscope probes in addition to flexible pH wire sensors for real-time bacterial biofilm-dental surface and dental materials interface studies.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9759506/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10729870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transition metals and Enterococcus faecalis: Homeostasis, virulence and perspectives. 过渡金属与粪肠球菌:内稳态、毒力与展望。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-12-01 Epub Date: 2022-09-26 DOI: 10.1111/omi.12391
Islam A A Ali, Gary S P Cheung, Prasanna Neelakantan

Enterococcus faecalis, a Gram-positive bacterium, is known to be a key player in several chronic infections as well as nosocomial, heart valve, urinary tract, surgical wound, and dental root canal infections. The capability to sense different transition metal levels and tune its response accordingly endows it with the potential to thrive and cause infections in several host niches. Over the past decade, our knowledge of how transition metals play a critical role in maintaining homeostasis of E. faecalis has improved significantly. The aim of this review is to elucidate the roles of metals such as iron, manganese, zinc, and copper in the physiology, metabolism, and pathogenicity of E. faecalis. These essential micronutrients contribute to energy production, redox stress response, expression of virulence determinants, and cooperation in polymicrobial communities. The review also highlights metal homeostasis systems in E. faecalis, which respond to fluctuations in extracellular metal levels, and regulate the intracellular metal content. Regulation of intracellular metallome secures the tolerance of E. faecalis to oxidative stress and host-mediated metal sequestration strategies. Therapeutic interventions which deprive E. faecalis of its essential metal requirements or disrupt its homeostatic control have been proposed to combat E. faecalis infections.

粪肠球菌是一种革兰氏阳性细菌,已知是几种慢性感染以及医院感染、心脏瓣膜感染、尿路感染、手术伤口感染和牙根管感染的关键因素。感知不同过渡金属水平并相应地调整其反应的能力赋予了它茁壮成长的潜力,并在几个宿主壁龛中引起感染。在过去的十年中,我们对过渡金属如何在维持粪肠杆菌稳态中发挥关键作用的认识有了显著提高。本文旨在阐明铁、锰、锌、铜等金属在粪肠杆菌的生理、代谢和致病性中的作用。这些必需微量营养素有助于能量产生、氧化还原应激反应、毒力决定因素的表达以及多微生物群落的合作。该综述还强调了粪肠杆菌中的金属稳态系统,该系统对细胞外金属水平的波动作出反应,并调节细胞内金属含量。细胞内金属组的调节确保了粪肠杆菌对氧化应激和宿主介导的金属隔离策略的耐受性。已经提出了通过剥夺粪肠杆菌对必需金属的需求或破坏其稳态控制的治疗干预措施来对抗粪肠杆菌感染。
{"title":"Transition metals and Enterococcus faecalis: Homeostasis, virulence and perspectives.","authors":"Islam A A Ali,&nbsp;Gary S P Cheung,&nbsp;Prasanna Neelakantan","doi":"10.1111/omi.12391","DOIUrl":"https://doi.org/10.1111/omi.12391","url":null,"abstract":"<p><p>Enterococcus faecalis, a Gram-positive bacterium, is known to be a key player in several chronic infections as well as nosocomial, heart valve, urinary tract, surgical wound, and dental root canal infections. The capability to sense different transition metal levels and tune its response accordingly endows it with the potential to thrive and cause infections in several host niches. Over the past decade, our knowledge of how transition metals play a critical role in maintaining homeostasis of E. faecalis has improved significantly. The aim of this review is to elucidate the roles of metals such as iron, manganese, zinc, and copper in the physiology, metabolism, and pathogenicity of E. faecalis. These essential micronutrients contribute to energy production, redox stress response, expression of virulence determinants, and cooperation in polymicrobial communities. The review also highlights metal homeostasis systems in E. faecalis, which respond to fluctuations in extracellular metal levels, and regulate the intracellular metal content. Regulation of intracellular metallome secures the tolerance of E. faecalis to oxidative stress and host-mediated metal sequestration strategies. Therapeutic interventions which deprive E. faecalis of its essential metal requirements or disrupt its homeostatic control have been proposed to combat E. faecalis infections.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40356392","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Glycerol strengthens probiotic effect of Limosilactobacillus reuteri in oral biofilms: A synergistic synbiotic approach. 甘油增强罗伊氏乳酸杆菌在口腔生物膜中的益生菌作用:一种协同合成方法。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-12-01 Epub Date: 2022-09-22 DOI: 10.1111/omi.12386
Wannes Van Holm, Tim Verspecht, Rita Carvalho, Kristel Bernaerts, Nico Boon, Naiera Zayed, Wim Teughels

Both in vitro and in vivo studies have shown that the probiotic Limosilactobacillus reuteri can improve oral health. Limosilactobacillus reuteri species are known to produce the antimicrobial "reuterin" from glycerol. In order to further increase its antimicrobial activity, this study evaluated the effect of the combined use of glycerol and Limosilactobacillus reuteri (ATCC PTA 5289) in view of using a synergistic synbiotic over a probiotic. An antagonistic agar growth and a multispecies biofilm model showed that the antimicrobial potential of the probiotic was significantly enhanced against periodontal pathobionts and anaerobic commensals when supplemented with glycerol. Synbiotic biofilms also showed a significant reduction in inflammatory expression of human oral keratinocytes (HOK-18A), but only when the keratinocytes were preincubated with the probiotic. Probiotic preincubation of keratinocytes or probiotic and synbiotic treatment of biofilms alone was insufficient to significantly reduce inflammatory expression. Overall, this study shows that combining glycerol with the probiotic L. reuteri into a synergistic synbiotic can greatly improve the effectiveness of the latter.

体外和体内研究都表明,益生菌罗伊氏乳酸杆菌可以改善口腔健康。已知罗伊氏乳酸菌种类可以从甘油中产生抗微生物的“罗伊氏素”。为了进一步提高其抑菌活性,本研究对甘油和罗伊氏乳杆菌(ATCC PTA 5289)联合使用的效果进行了评价,以期在益生菌中使用增效合成菌。拮抗琼脂生长和多物种生物膜模型表明,当添加甘油时,益生菌对牙周病原体和厌氧共生菌的抗菌潜力显着增强。合成生物膜也显示出人口腔角化细胞(HOK-18A)炎症表达的显著降低,但仅当角化细胞与益生菌预孵育时。角化细胞的益生菌预孵育或单独的益生菌和生物膜的合成处理不足以显著降低炎症表达。综上所述,本研究表明,将甘油与益生菌罗伊氏乳杆菌组合成协同合成菌,可大大提高后者的有效性。
{"title":"Glycerol strengthens probiotic effect of Limosilactobacillus reuteri in oral biofilms: A synergistic synbiotic approach.","authors":"Wannes Van Holm,&nbsp;Tim Verspecht,&nbsp;Rita Carvalho,&nbsp;Kristel Bernaerts,&nbsp;Nico Boon,&nbsp;Naiera Zayed,&nbsp;Wim Teughels","doi":"10.1111/omi.12386","DOIUrl":"https://doi.org/10.1111/omi.12386","url":null,"abstract":"<p><p>Both in vitro and in vivo studies have shown that the probiotic Limosilactobacillus reuteri can improve oral health. Limosilactobacillus reuteri species are known to produce the antimicrobial \"reuterin\" from glycerol. In order to further increase its antimicrobial activity, this study evaluated the effect of the combined use of glycerol and Limosilactobacillus reuteri (ATCC PTA 5289) in view of using a synergistic synbiotic over a probiotic. An antagonistic agar growth and a multispecies biofilm model showed that the antimicrobial potential of the probiotic was significantly enhanced against periodontal pathobionts and anaerobic commensals when supplemented with glycerol. Synbiotic biofilms also showed a significant reduction in inflammatory expression of human oral keratinocytes (HOK-18A), but only when the keratinocytes were preincubated with the probiotic. Probiotic preincubation of keratinocytes or probiotic and synbiotic treatment of biofilms alone was insufficient to significantly reduce inflammatory expression. Overall, this study shows that combining glycerol with the probiotic L. reuteri into a synergistic synbiotic can greatly improve the effectiveness of the latter.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"33450488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
Antagonistic interactions by a high H2 O2 -producing commensal streptococcus modulate caries development by Streptococcus mutans. 产生高 H2 O2 的共生链球菌的拮抗相互作用可调节变异链球菌的龋齿发展。
IF 2.8 3区 医学 Q1 DENTISTRY, ORAL SURGERY & MEDICINE Pub Date : 2022-12-01 Epub Date: 2022-10-11 DOI: 10.1111/omi.12394
Dongyeop Kim, Tatsuro Ito, Anderson Hara, Yong Li, Jens Kreth, Hyun Koo

Dental caries (tooth-decay) is caused by biofilms harboring polymicrobial communities on teeth that leads to the onset of localized areas of enamel demineralization. Streptococcus mutans has been clinically associated with severe caries in childhood. Although commensal bacteria can combat S. mutans using self-generated antimicrobials such as hydrogen peroxide (H2 O2 ), constant sugar-rich diet consumption disrupts microbial homeostasis shifting toward cariogenic community. Recently, Streptococcus oralis subsp. tigurinus strain J22, an oral isolate, was identified as a uniquely potent H2 O2 producer. Here, we assess whether a high H2 O2 -producing commensal streptococcus can modulate the spatial organization and virulence of S. mutans within biofilms. Using an experimental biofilm model, we find that the presence of S. oralis J22 can effectively inhibit the clustering, accumulation, and spatial organization of S. mutans on ex vivo human tooth surface, resulting in significant reduction of enamel demineralization. Notably, the generation of H2 O2 via pyruvate oxidase (SpxB) from S. oralis J22 is not repressed by sugars (a common repressor in other mitis group streptococci), resulting in enhanced inhibition of S. mutans growth (vs. Streptococcus gordonii). We further investigate its impact on biofilm virulence using an in vivo rodent caries model under sugar-rich diet. Coinfection of S. mutans with S. oralis results in reduced caries development compared to either species infected alone, whereas coinfection with S. gordonii has negligible effects, suggesting that the presence of an efficient, high H2 O2 -producer can disrupt S. mutans virulence. This work demonstrates that oral isolates with unusual high H2 O2 production may be capable of modulating biofilm cariogenicity in vivo. The findings also highlight the importance of bacterial antagonistic interactions within polymicrobial communities in health and in disease-causing state.

龋齿(牙齿龋坏)是由牙齿上滋生的多微生物群落生物膜引起的,它会导致局部区域的釉质脱矿。在临床上,变异链球菌与儿童时期的严重龋齿有关。虽然共生细菌可以利用过氧化氢(H2 O2)等自身产生的抗菌剂来对抗变异链球菌,但持续摄入高糖饮食会破坏微生物的平衡,使其转向致龋群落。最近,口腔链球菌 tigurinus 亚种 J22 菌株(一种口腔分离物)被鉴定为一种独特的强效 H2 O2 生产者。在此,我们评估了一种高产 H2 O2 的共生链球菌是否能调节生物膜内变异杆菌的空间组织和毒力。通过一个实验性生物膜模型,我们发现口腔链球菌 J22 的存在能有效抑制人体牙齿表面变异杆菌的聚集、堆积和空间组织,从而显著减少牙釉质脱矿。值得注意的是,口腔链球菌 J22 通过丙酮酸氧化酶(SpxB)产生的 H2 O2 不会被糖(其他米氏链球菌常见的抑制因子)抑制,从而增强了对变异棒状杆菌生长的抑制作用(与戈登链球菌相比)。我们利用富含糖分的啮齿动物龋齿模型进一步研究了它对生物膜毒力的影响。与单独感染口腔链球菌相比,变异单胞菌与口腔链球菌共感染会减少龋齿的发展,而与戈登链球菌共感染的影响可以忽略不计,这表明高效、高H2 O2产生者的存在会破坏变异单胞菌的毒力。这项研究表明,具有不寻常的高 H2 O2 产量的口腔分离物可能能够调节体内生物膜的致龋性。研究结果还强调了多微生物群落中细菌拮抗相互作用在健康和致病状态中的重要性。
{"title":"Antagonistic interactions by a high H<sub>2</sub> O<sub>2</sub> -producing commensal streptococcus modulate caries development by Streptococcus mutans.","authors":"Dongyeop Kim, Tatsuro Ito, Anderson Hara, Yong Li, Jens Kreth, Hyun Koo","doi":"10.1111/omi.12394","DOIUrl":"10.1111/omi.12394","url":null,"abstract":"<p><p>Dental caries (tooth-decay) is caused by biofilms harboring polymicrobial communities on teeth that leads to the onset of localized areas of enamel demineralization. Streptococcus mutans has been clinically associated with severe caries in childhood. Although commensal bacteria can combat S. mutans using self-generated antimicrobials such as hydrogen peroxide (H<sub>2</sub> O<sub>2</sub> ), constant sugar-rich diet consumption disrupts microbial homeostasis shifting toward cariogenic community. Recently, Streptococcus oralis subsp. tigurinus strain J22, an oral isolate, was identified as a uniquely potent H<sub>2</sub> O<sub>2</sub> producer. Here, we assess whether a high H<sub>2</sub> O<sub>2</sub> -producing commensal streptococcus can modulate the spatial organization and virulence of S. mutans within biofilms. Using an experimental biofilm model, we find that the presence of S. oralis J22 can effectively inhibit the clustering, accumulation, and spatial organization of S. mutans on ex vivo human tooth surface, resulting in significant reduction of enamel demineralization. Notably, the generation of H<sub>2</sub> O<sub>2</sub> via pyruvate oxidase (SpxB) from S. oralis J22 is not repressed by sugars (a common repressor in other mitis group streptococci), resulting in enhanced inhibition of S. mutans growth (vs. Streptococcus gordonii). We further investigate its impact on biofilm virulence using an in vivo rodent caries model under sugar-rich diet. Coinfection of S. mutans with S. oralis results in reduced caries development compared to either species infected alone, whereas coinfection with S. gordonii has negligible effects, suggesting that the presence of an efficient, high H<sub>2</sub> O<sub>2</sub> -producer can disrupt S. mutans virulence. This work demonstrates that oral isolates with unusual high H<sub>2</sub> O<sub>2</sub> production may be capable of modulating biofilm cariogenicity in vivo. The findings also highlight the importance of bacterial antagonistic interactions within polymicrobial communities in health and in disease-causing state.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"33483114","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cover Image, Volume 37, Issue 6 封面图片,第37卷,第6期
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-11-24 DOI: 10.1111/omi.12401
The cover image is based on the Original Article Site-tropism of streptococci in the oral microbiome by Anthony R. McLean et al., https://doi.org/10.1111/omi.12387.
封面图片基于Anthony R. McLean等人的原创文章《口腔微生物组中链球菌的趋位性》https://doi.org/10.1111/omi.12387。
{"title":"Cover Image, Volume 37, Issue 6","authors":"","doi":"10.1111/omi.12401","DOIUrl":"https://doi.org/10.1111/omi.12401","url":null,"abstract":"The cover image is based on the Original Article <i>Site-tropism of streptococci in the oral microbiome</i> by Anthony R. McLean et al., https://doi.org/10.1111/omi.12387.","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-11-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138508325","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The functional oral microbiome: Biofilm environment, polymicrobial interactions, and community dynamics. 功能性口腔微生物群:生物膜环境、多微生物相互作用和群落动态。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-10-01 DOI: 10.1111/omi.12390
Jens Kreth, Hyun Koo, Patricia I Diaz
{"title":"The functional oral microbiome: Biofilm environment, polymicrobial interactions, and community dynamics.","authors":"Jens Kreth, Hyun Koo, Patricia I Diaz","doi":"10.1111/omi.12390","DOIUrl":"10.1111/omi.12390","url":null,"abstract":"","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9174242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Farnesol delivery via polymeric nanoparticle carriers inhibits cariogenic cross-kingdom biofilms and prevents enamel demineralization. 通过聚合物纳米颗粒载体递送法尼醇可抑制致龋跨王国生物膜并防止牙釉质脱矿。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-10-01 Epub Date: 2022-08-04 DOI: 10.1111/omi.12379
Tatsuro Ito, Kenneth R Sims, Yuan Liu, Zhenting Xiang, Rodrigo A Arthur, Anderson T Hara, Hyun Koo, Danielle S W Benoit, Marlise I Klein

Streptococcus mutans and Candida albicans are frequently detected together in the plaque from patients with early childhood caries (ECC) and synergistically interact to form a cariogenic cross-kingdom biofilm. However, this biofilm is difficult to control. Thus, to achieve maximal efficacy within the complex biofilm microenvironment, nanoparticle carriers have shown increased interest in treating oral biofilms in recent years. Here, we assessed the anti-biofilm efficacy of farnesol (Far), a hydrophobic antibacterial drug and repressor of Candida filamentous forms, against cross-kingdom biofilms employing drug delivery via polymeric nanoparticle carriers (NPCs). We also evaluated the effect of the strategy on teeth enamel demineralization. The farnesol-loaded NPCs (NPC+Far) resulted in a 2-log CFU/mL reduction of S. mutans and C. albicans (hydroxyapatite disc biofilm model). High-resolution confocal images further confirmed a significant reduction in exopolysaccharides, smaller microcolonies of S. mutans, and no hyphal form of C. albicans after treatment with NPC+Far on human tooth enamel (HT) slabs, altering the biofilm 3D structure. Furthermore, NPC+Far treatment was highly effective in preventing enamel demineralization on HT, reducing lesion depth (79% reduction) and mineral loss (85% reduction) versus vehicle PBS-treated HT, while NPC or Far alone had no differences with the PBS. The drug delivery via polymeric NPCs has the potential for targeting bacterial-fungal biofilms associated with a prevalent and costly pediatric oral disease, such as ECC.

变形链球菌和白色念珠菌经常在儿童早期龋齿(ECC)患者的牙菌斑中一起检测到,并协同作用形成致龋跨王国生物膜。然而,这种生物膜很难控制。因此,为了在复杂的生物膜微环境中实现最大功效,近年来,纳米颗粒载体对处理口腔生物膜表现出越来越大的兴趣。在这里,我们评估了法尼醇(Far)的抗生物膜功效,法尼醇是一种疏水性抗菌药物,也是丝状念珠菌的阻遏物,通过聚合物纳米颗粒载体(NPC)进行药物递送,对抗跨王国生物膜。我们还评估了该策略对牙釉质脱矿的影响。负载法尼醇的NPC(NPC+Far)导致变形链球菌和白色念珠菌(羟基磷灰石圆盘生物膜模型)的CFU/mL减少2 log。高分辨率共聚焦图像进一步证实,在人类牙釉质(HT)板上用NPC+Far处理后,胞外多糖、变形链球菌的小菌落和白色念珠菌的菌丝形式显著减少,改变了生物膜的3D结构。此外,与载体PBS处理的HT相比,NPC+Far处理在防止HT上的牙釉质脱矿、减少病变深度(减少79%)和矿物质损失(减少85%)方面非常有效,而单独使用NPC或Far与PBS没有差异。通过聚合物NPC的药物递送具有靶向与流行且昂贵的儿科口腔疾病(如ECC)相关的细菌-真菌生物膜的潜力。
{"title":"Farnesol delivery via polymeric nanoparticle carriers inhibits cariogenic cross-kingdom biofilms and prevents enamel demineralization.","authors":"Tatsuro Ito,&nbsp;Kenneth R Sims,&nbsp;Yuan Liu,&nbsp;Zhenting Xiang,&nbsp;Rodrigo A Arthur,&nbsp;Anderson T Hara,&nbsp;Hyun Koo,&nbsp;Danielle S W Benoit,&nbsp;Marlise I Klein","doi":"10.1111/omi.12379","DOIUrl":"10.1111/omi.12379","url":null,"abstract":"<p><p>Streptococcus mutans and Candida albicans are frequently detected together in the plaque from patients with early childhood caries (ECC) and synergistically interact to form a cariogenic cross-kingdom biofilm. However, this biofilm is difficult to control. Thus, to achieve maximal efficacy within the complex biofilm microenvironment, nanoparticle carriers have shown increased interest in treating oral biofilms in recent years. Here, we assessed the anti-biofilm efficacy of farnesol (Far), a hydrophobic antibacterial drug and repressor of Candida filamentous forms, against cross-kingdom biofilms employing drug delivery via polymeric nanoparticle carriers (NPCs). We also evaluated the effect of the strategy on teeth enamel demineralization. The farnesol-loaded NPCs (NPC+Far) resulted in a 2-log CFU/mL reduction of S. mutans and C. albicans (hydroxyapatite disc biofilm model). High-resolution confocal images further confirmed a significant reduction in exopolysaccharides, smaller microcolonies of S. mutans, and no hyphal form of C. albicans after treatment with NPC+Far on human tooth enamel (HT) slabs, altering the biofilm 3D structure. Furthermore, NPC+Far treatment was highly effective in preventing enamel demineralization on HT, reducing lesion depth (79% reduction) and mineral loss (85% reduction) versus vehicle PBS-treated HT, while NPC or Far alone had no differences with the PBS. The drug delivery via polymeric NPCs has the potential for targeting bacterial-fungal biofilms associated with a prevalent and costly pediatric oral disease, such as ECC.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9529802/pdf/nihms-1825297.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40632861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
A cell wall-anchored glycoprotein confers resistance to cation stress in Actinomyces oris biofilms. 细胞壁锚定的糖蛋白赋予口放线菌生物膜对阳离子应激的抗性。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-10-01 Epub Date: 2022-03-25 DOI: 10.1111/omi.12365
Abu Amar M Al Mamun, Chenggang Wu, Chungyu Chang, Belkys C Sanchez, Asis Das, Hung Ton-That

Actinomyces oris plays an important role in oral biofilm development. Like many gram-positive bacteria, A. oris produces a sizable number of surface proteins that are anchored to bacterial peptidoglycan by a conserved transpeptidase named the housekeeping sortase SrtA; however, the biological role of many A. oris surface proteins in biofilm formation is largely unknown. Here, we report that the glycoprotein GspA-a genetic suppressor of srtA deletion lethality-not only promotes biofilm formation but also maintains cell membrane integrity under cation stress. In comparison to wild-type cells, under elevated concentrations of mono- and divalent cations the formation of mono- and multi-species biofilms by mutant cells devoid of gspA was significantly diminished, although planktonic growth of both cell types in the presence of cations was indistinguishable. Because gspA overexpression is lethal to cells lacking gspA and srtA, we performed a genetic screen to identify GspA determinants involving cell viability. DNA sequencing and biochemical characterizations of viable clones revealed that mutations of two critical cysteine residues and a serine residue severely affected GspA glycosylation and biofilm formation. Furthermore, mutant cells lacking gspA were markedly sensitive to sodium dodecyl sulfate, a detergent that solubilizes the cytoplasmic membranes, suggesting the cell envelope of the gspA mutant was altered. Consistent with this observation, the gspA mutant exhibited increased membrane permeability, independent of GspA glycosylation, compared to the wild-type strain. Altogether, the results support the notion that the cell wall-anchored glycoprotein GspA provides a defense mechanism against cation stress in biofilm development promoted by A. oris.

口腔放线菌在口腔生物膜的发育中起着重要作用。像许多革兰氏阳性细菌一样,A.oris产生大量的表面蛋白,这些蛋白通过一种名为家政分拣酶SrtA的保守转肽酶锚定在细菌肽聚糖上;然而,许多口腔A.oris表面蛋白在生物膜形成中的生物学作用在很大程度上是未知的。在这里,我们报道了糖蛋白GspA-srtA缺失致死性的遗传抑制剂,它不仅促进生物膜的形成,而且在阳离子胁迫下保持细胞膜的完整性。与野生型细胞相比,在单价和二价阳离子浓度升高的情况下,缺乏gspA的突变细胞形成的单物种和多物种生物膜显著减少,尽管在阳离子存在的情况下这两种细胞类型的浮游生长难以区分。由于gspA过表达对缺乏gspA和srtA的细胞是致命的,我们进行了基因筛选,以确定涉及细胞活力的gspA决定因素。活克隆的DNA测序和生物化学特征表明,两个关键半胱氨酸残基和一个丝氨酸残基的突变严重影响GspA的糖基化和生物膜的形成。此外,缺乏gspA的突变细胞对十二烷基硫酸钠(一种溶解质膜的洗涤剂)显著敏感,这表明gspA突变株的细胞包膜发生了改变。与这一观察结果一致,与野生型菌株相比,gspA突变体表现出增加的膜渗透性,与gspA糖基化无关。总之,这些结果支持这样一种观点,即细胞壁锚定的糖蛋白GspA在a.oris促进的生物膜发育中提供了对抗阳离子应激的防御机制。
{"title":"A cell wall-anchored glycoprotein confers resistance to cation stress in Actinomyces oris biofilms.","authors":"Abu Amar M Al Mamun,&nbsp;Chenggang Wu,&nbsp;Chungyu Chang,&nbsp;Belkys C Sanchez,&nbsp;Asis Das,&nbsp;Hung Ton-That","doi":"10.1111/omi.12365","DOIUrl":"https://doi.org/10.1111/omi.12365","url":null,"abstract":"<p><p>Actinomyces oris plays an important role in oral biofilm development. Like many gram-positive bacteria, A. oris produces a sizable number of surface proteins that are anchored to bacterial peptidoglycan by a conserved transpeptidase named the housekeeping sortase SrtA; however, the biological role of many A. oris surface proteins in biofilm formation is largely unknown. Here, we report that the glycoprotein GspA-a genetic suppressor of srtA deletion lethality-not only promotes biofilm formation but also maintains cell membrane integrity under cation stress. In comparison to wild-type cells, under elevated concentrations of mono- and divalent cations the formation of mono- and multi-species biofilms by mutant cells devoid of gspA was significantly diminished, although planktonic growth of both cell types in the presence of cations was indistinguishable. Because gspA overexpression is lethal to cells lacking gspA and srtA, we performed a genetic screen to identify GspA determinants involving cell viability. DNA sequencing and biochemical characterizations of viable clones revealed that mutations of two critical cysteine residues and a serine residue severely affected GspA glycosylation and biofilm formation. Furthermore, mutant cells lacking gspA were markedly sensitive to sodium dodecyl sulfate, a detergent that solubilizes the cytoplasmic membranes, suggesting the cell envelope of the gspA mutant was altered. Consistent with this observation, the gspA mutant exhibited increased membrane permeability, independent of GspA glycosylation, compared to the wild-type strain. Altogether, the results support the notion that the cell wall-anchored glycoprotein GspA provides a defense mechanism against cation stress in biofilm development promoted by A. oris.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9474737/pdf/nihms-1788776.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41134859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
A reappraisal of microbiome dysbiosis during experimental periodontitis. 实验性牙周炎期间微生物群失调的重新评估。
IF 3.7 3区 医学 Q1 Dentistry Pub Date : 2022-10-01 DOI: 10.1111/omi.12382
Marion Arce, Natalia Endo, Nicolas Dutzan, Loreto Abusleme

Periodontitis is a chronic inflammatory disease associated with the presence of dysbiotic microbial communities. Several studies interrogating periodontitis pathogenesis have utilized the murine ligature-induced periodontitis (LIP) model and have further examined the ligature-associated microbiome relying on 16S rRNA-based sequencing techniques. However, it is often very challenging to compare microbial profiles across studies due to important differences in bioinformatic processing and databases used for taxonomic assignment. Thus, our study aim was to reanalyze microbiome sequencing datasets from studies utilizing the LIP model through a standardized bioinformatic analysis pipeline, generating a comprehensive overview of microbial dysbiosis during experimental periodontitis.We conducted a reanalysis of 16S rDNA gene sequencing datasets from nine published studies utilizing the LIP model. Reads were grouped according to the hypervariable region of the 16S rDNA gene amplified (V1-V3 and V4), preprocessed, binned into operational taxonomic units and classified utilizing relevant databases. Alpha- and beta-diversity analyses were conducted, along with relative abundance profiling of microbial communities. Our findings revealed similar microbial richness and diversity across studies and determined shifts in microbial community structure determined by periodontitis induction and study of origin. Clear variations in the relative abundance of bacterial taxa were observed starting on day 5 after ligation and onward, consistent with a distinct microbial composition during health and experimental periodontitis. We also uncovered differentially represented bacterial taxa across studies, dominating periodontal health and LIP-associated communities. Collectively, this reanalysis provides a unified overview of microbial dysbiosis during the LIP model, providing new insights that aim to inform further studies dedicated to unraveling oral host-microbial interactions.

牙周炎是一种慢性炎症性疾病,与存在益生菌群有关。一些关于牙周炎发病机制的研究利用小鼠结扎诱导的牙周炎(LIP)模型,并依靠基于16S rrna的测序技术进一步研究了结扎相关的微生物组。然而,由于生物信息学处理和用于分类分配的数据库的重要差异,在不同的研究中比较微生物概况通常是非常具有挑战性的。因此,我们的研究目的是通过标准化的生物信息学分析管道,利用LIP模型重新分析来自研究的微生物组测序数据集,从而全面概述实验性牙周炎期间的微生物生态失调。我们利用LIP模型对9项已发表的研究中的16S rDNA基因测序数据集进行了重新分析。根据扩增的16S rDNA基因高变区(V1-V3和V4)对Reads进行分组,进行预处理,将Reads分入可操作的分类单元,并利用相关数据库进行分类。进行了α和β多样性分析,以及微生物群落的相对丰度分析。我们的研究结果揭示了不同研究中相似的微生物丰富度和多样性,并确定了由牙周炎诱导和起源研究决定的微生物群落结构的变化。结扎后第5天开始观察到细菌分类群相对丰度的明显变化,这与健康和实验性牙周炎期间独特的微生物组成一致。我们还发现了不同研究中不同代表性的细菌分类群,它们主导牙周健康和lip相关群落。总的来说,这一再分析提供了LIP模型中微生物生态失调的统一概述,为进一步研究口腔宿主-微生物相互作用提供了新的见解。
{"title":"A reappraisal of microbiome dysbiosis during experimental periodontitis.","authors":"Marion Arce,&nbsp;Natalia Endo,&nbsp;Nicolas Dutzan,&nbsp;Loreto Abusleme","doi":"10.1111/omi.12382","DOIUrl":"https://doi.org/10.1111/omi.12382","url":null,"abstract":"<p><p>Periodontitis is a chronic inflammatory disease associated with the presence of dysbiotic microbial communities. Several studies interrogating periodontitis pathogenesis have utilized the murine ligature-induced periodontitis (LIP) model and have further examined the ligature-associated microbiome relying on 16S rRNA-based sequencing techniques. However, it is often very challenging to compare microbial profiles across studies due to important differences in bioinformatic processing and databases used for taxonomic assignment. Thus, our study aim was to reanalyze microbiome sequencing datasets from studies utilizing the LIP model through a standardized bioinformatic analysis pipeline, generating a comprehensive overview of microbial dysbiosis during experimental periodontitis.We conducted a reanalysis of 16S rDNA gene sequencing datasets from nine published studies utilizing the LIP model. Reads were grouped according to the hypervariable region of the 16S rDNA gene amplified (V1-V3 and V4), preprocessed, binned into operational taxonomic units and classified utilizing relevant databases. Alpha- and beta-diversity analyses were conducted, along with relative abundance profiling of microbial communities. Our findings revealed similar microbial richness and diversity across studies and determined shifts in microbial community structure determined by periodontitis induction and study of origin. Clear variations in the relative abundance of bacterial taxa were observed starting on day 5 after ligation and onward, consistent with a distinct microbial composition during health and experimental periodontitis. We also uncovered differentially represented bacterial taxa across studies, dominating periodontal health and LIP-associated communities. Collectively, this reanalysis provides a unified overview of microbial dysbiosis during the LIP model, providing new insights that aim to inform further studies dedicated to unraveling oral host-microbial interactions.</p>","PeriodicalId":18815,"journal":{"name":"Molecular Oral Microbiology","volume":null,"pages":null},"PeriodicalIF":3.7,"publicationDate":"2022-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10203226","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
期刊
Molecular Oral Microbiology
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