1
|
Wu R, Jiang Y, Yan J, Shen N, Liu S, Yin H, Zhu S, Qiao J. Beneficial changes in gut microbiota after phototherapy for neonatal hyperbilirubinemia. Biomed Rep 2024; 20:101. [PMID: 38765854 PMCID: PMC11099589 DOI: 10.3892/br.2024.1789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/22/2024] [Indexed: 05/22/2024] Open
Abstract
Phototherapy is the most commonly used treatment for neonatal hyperbilirubinemia (NH). Gut microbiota is involved in bilirubin metabolism; however, it is uncertain whether this is affected by phototherapy. The present study included 43 newborns with hyperbilirubinemia and collected fecal samples for high-throughput sequencing before and after phototherapy. Selection α diversity analysis was used to determine the differences in diversity and abundance between the two groups, whereas similarity was determined using β diversity analysis. Linear discriminant analysis effect size analysis was used to screen for markedly different bacteria. The structure of the gut microbiota in newborns with hyperbilirubinemia changed after phototherapy, with a significant decrease in abundance and diversity. The changes in the key bacterial species were characterized by an increase in the abundance of Streptococcus salivarius and a decrease in the abundance of Escherichia, Klebsiella pneumoniae, Rothia mucilaginosa and Streptococcus oralis. These changes mainly manifested as an increase in beneficial bacteria and a decrease in opportunistic bacteria, which may not be related to the side effects of phototherapy. These results can provide theoretical assistance for microbiological research on the later stages of NH.
Collapse
Affiliation(s)
- Rang Wu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Yazhou Jiang
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Jingjing Yan
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Nan Shen
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Song Liu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Hanjun Yin
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Suyue Zhu
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Jibing Qiao
- Department of Pediatrics, Suqian Hospital Affiliated to Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| |
Collapse
|
2
|
Liu WQ, Ji X, Ba F, Zhang Y, Xu H, Huang S, Zheng X, Liu Y, Ling S, Jewett MC, Li J. Cell-free biosynthesis and engineering of ribosomally synthesized lanthipeptides. Nat Commun 2024; 15:4336. [PMID: 38773100 PMCID: PMC11109155 DOI: 10.1038/s41467-024-48726-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/13/2024] [Indexed: 05/23/2024] Open
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a major class of natural products with diverse chemical structures and potent biological activities. A vast majority of RiPP gene clusters remain unexplored in microbial genomes, which is partially due to the lack of rapid and efficient heterologous expression systems for RiPP characterization and biosynthesis. Here, we report a unified biocatalysis (UniBioCat) system based on cell-free gene expression for rapid biosynthesis and engineering of RiPPs. We demonstrate UniBioCat by reconstituting a full biosynthetic pathway for de novo biosynthesis of salivaricin B, a lanthipeptide RiPP. Next, we delete several protease/peptidase genes from the source strain to enhance the performance of UniBioCat, which then can synthesize and screen salivaricin B variants with enhanced antimicrobial activity. Finally, we show that UniBioCat is generalizable by synthesizing and evaluating the bioactivity of ten uncharacterized lanthipeptides. We expect UniBioCat to accelerate the discovery, characterization, and synthesis of RiPPs.
Collapse
Affiliation(s)
- Wan-Qiu Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiangyang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Fang Ba
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yufei Zhang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Huiling Xu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shuhui Huang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiao Zheng
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yifan Liu
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Shengjie Ling
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Michael C Jewett
- Department of Bioengineering, Stanford University, Stanford, CA, US.
| | - Jian Li
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China.
- State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
3
|
Sadaoka N, Le MNT, Kawada-Matsuo M, Eng S, Zendo T, Nakanishi J, Takeda K, Shiba H, Komatsuzawa H. Opposing genetic polymorphisms of two ABC transporters contribute to the variation of nukacin resistance in Streptococcus mutans. Appl Environ Microbiol 2024; 90:e0208423. [PMID: 38411065 PMCID: PMC10952377 DOI: 10.1128/aem.02084-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/23/2024] [Indexed: 02/28/2024] Open
Abstract
Streptococcus mutans is a cariogenic bacterium that produces a variety of bacteriocins and retains resistance to these bacteriocins. In this study, we investigated the susceptibility of 127 S. mutans strains to nukacins produced by Staphylococcus spp., which are commensal bacteria in humans. We detected diverse susceptibilities among strains. Nineteen strains had a disrupted LctF (type I), which is responsible for nukacin susceptibility, whereas the remaining 108 strains had an intact LctF (type II) and displayed resistance to nukacins. However, the type I strains still showed resistance to nukacins to some extent. Interestingly, 18/19 (94.7%) type I strains carried a mukA-T locus, which is related to the synthesis of mutacin K8, and mukFEG, an ABC transporter. In contrast, among type II strains, only 6/108 strains (5.6%) had both the mukA-T locus and mukFEG, 19/108 strains (17.6%) carried only mukFEG, and 83/108 strains (76.9%) harbored neither mukA-T nor mukFEG. We also found that MukF had two variants: 305 amino acids (type α) and 302 amino acids (type β). All type I strains showed a type α (MukFα), whereas most type II strains with mukFEG (22/25 strains) had a type β (MukFβ). Then, we constructed a mukFEG-deletion mutant complemented with MukFαEG or MukFβEG and found that only MukFαEG was involved in nukacin resistance. The nukacin resistance capability of type II-LctFEG was stronger than that of MukFαEG. In conclusion, we identified a novel nukacin resistance factor, MukFEG, and either LctFEG or MukFEG was active in most strains via genetic polymorphisms depending on mukA-T genes. IMPORTANCE Streptococcus mutans is an important pathogenic bacterium not only for dental caries but also for systemic diseases. S. mutans is known to produce a variety of bacteriocins and to retain resistance these bacteriocins. In this study, two ABC transporters, LctFEG and MukFEG, were implicated in nukacin resistance and each ABC transporter has two subtypes, active and inactive. Of the two ABC transporters, only one ABC transporter was always resistant, while the other ABC transporter was inactivated by genetic mutation. Interestingly, this phenomenon was defined by the presence or absence of the mutacin K8 synthesis gene region, one of the bacteriocins of S. mutans. This suggests that the resistance acquisition is tightly controlled in each strain. This study provides important evidence that the insertion of bacteriocin synthesis genes is involved in the induction of genetic polymorphisms and suggests that bacteriocin synthesis genes may play an important role in bacterial evolution.
Collapse
Affiliation(s)
- Naoki Sadaoka
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Oral Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Oral Infectious Diseases, Hiroshima University, Hiroshima, Japan
| | - Sopongselamuny Eng
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Takeshi Zendo
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School, Kyushu University, Fukuoka, Japan
| | - Jun Nakanishi
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsuhiro Takeda
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Shiba
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
- Project Research Centre for Oral Infectious Diseases, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
4
|
Li W, Liang H, Lin X, Hu T, Wu Z, He W, Wang M, Zhang J, Jie Z, Jin X, Xu X, Wang J, Yang H, Zhang W, Kristiansen K, Xiao L, Zou Y. A catalog of bacterial reference genomes from cultivated human oral bacteria. NPJ Biofilms Microbiomes 2023; 9:45. [PMID: 37400465 DOI: 10.1038/s41522-023-00414-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/20/2023] [Indexed: 07/05/2023] Open
Abstract
The oral cavity harbors highly diverse communities of microorganisms. However, the number of isolated species and high-quality genomes is limited. Here we present a Cultivated Oral Bacteria Genome Reference (COGR), comprising 1089 high-quality genomes based on large-scale aerobic and anaerobic cultivation of human oral bacteria isolated from dental plaques, tongue, and saliva. COGR covers five phyla and contains 195 species-level clusters of which 95 include 315 genomes representing species with no taxonomic annotation. The oral microbiota differs markedly between individuals, with 111 clusters being person-specific. Genes encoding CAZymes are abundant in the genomes of COGR. Members of the Streptococcus genus make up the largest proportion of COGR and many of these harbor entire pathways for quorum sensing important for biofilm formation. Several clusters containing unknown bacteria are enriched in individuals with rheumatoid arthritis, emphasizing the importance of culture-based isolation for characterizing and exploiting oral bacteria.
Collapse
Affiliation(s)
- Wenxi Li
- BGI-Shenzhen, 518083, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, 510006, Guangzhou, China
| | | | - Xiaoqian Lin
- BGI-Shenzhen, 518083, Shenzhen, China
- School of Biology and Biological Engineering, South China University of Technology, 510006, Guangzhou, China
| | | | - Zhinan Wu
- BGI-Shenzhen, 518083, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wenxin He
- BGI-Shenzhen, 518083, Shenzhen, China
| | | | | | - Zhuye Jie
- BGI-Shenzhen, 518083, Shenzhen, China
| | - Xin Jin
- BGI-Shenzhen, 518083, Shenzhen, China
| | - Xun Xu
- BGI-Shenzhen, 518083, Shenzhen, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, 518120, Shenzhen, China
| | - Jian Wang
- BGI-Shenzhen, 518083, Shenzhen, China
- James D. Watson Institute of Genome Sciences, 310058, Hangzhou, China
| | - Huanming Yang
- BGI-Shenzhen, 518083, Shenzhen, China
- James D. Watson Institute of Genome Sciences, 310058, Hangzhou, China
| | | | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark.
- Qingdao-Europe Advanced Institute for Life Sciences, BGI-Shenzhen, 266555, Qingdao, China.
- PREDICT, Center for Molecular Prediction of Inflammatory Bowel Disease, Faculty of Medicine, Aalborg University, 2450, Copenhagen, Denmark.
| | - Liang Xiao
- BGI-Shenzhen, 518083, Shenzhen, China.
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China.
- Qingdao-Europe Advanced Institute for Life Sciences, BGI-Shenzhen, 266555, Qingdao, China.
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, China.
| | - Yuanqiang Zou
- BGI-Shenzhen, 518083, Shenzhen, China.
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Universitetsparken 13, 2100, Copenhagen, Denmark.
- Qingdao-Europe Advanced Institute for Life Sciences, BGI-Shenzhen, 266555, Qingdao, China.
- Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI-Shenzhen, Shenzhen, China.
| |
Collapse
|
5
|
Tagg JR, Harold LK, Jain R, Hale JDF. Beneficial modulation of human health in the oral cavity and beyond using bacteriocin-like inhibitory substance-producing streptococcal probiotics. Front Microbiol 2023; 14:1161155. [PMID: 37056747 PMCID: PMC10086258 DOI: 10.3389/fmicb.2023.1161155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
The human oral cavity contains a diversity of microbial habitats that have been adopted and adapted to as homeland by an amazingly heterogeneous population of microorganisms collectively referred to as the oral microbiota. These microbes generally co-habit in harmonious homeostasis. However, under conditions of imposed stress, as with changes to the host’s physiology or nutritional status, or as a response to foreign microbial or antimicrobial incursions, some components of the oral “microbiome” (viz. the in situ microbiota) may enter a dysbiotic state. This microbiome dysbiosis can manifest in a variety of guises including streptococcal sore throats, dental caries, oral thrush, halitosis and periodontal disease. Most of the strategies currently available for the management or treatment of microbial diseases of the oral cavity focus on the repetitive “broad sweep” and short-term culling of oral microbe populations, hopefully including the perceived principal pathogens. Both physical and chemical techniques are used. However, the application of more focused approaches to the harnessing or elimination of key oral cavity pathogens is now feasible through the use of probiotic strains that are naturally adapted for oral cavity colonization and also are equipped to produce anti-competitor molecules such as the bacteriocins and bacteriocin-like inhibitory substances (viz BLIS). Some of these probiotics are capable of suppressing the proliferation of a variety of recognized microbial pathogens of the human mouth, thereby assisting with the restoration of oral microbiome homeostasis. BLIS K12 and BLIS M18, the progenitors of the BLIS-producing oral probiotics, are members of the human oral cavity commensal species Streptococcus salivarius. More recently however, a number of other streptococcal and some non-streptococcal candidate oral probiotics have also been promoted. What is becoming increasingly apparent is that the future for oral probiotic applications will probably extend well beyond the attempted limitation of the direct pathological consequences of oral microbiome dysbiosis to also encompass a plethora of systemic diseases and disorders of the human host. The background to and the evolving prospects for the beneficial modulation of the oral microbiome via the application of BLIS-producing S. salivarius probiotics comprises the principal focus of the present review.
Collapse
|
6
|
In Vitro Evaluation of Commercial Probiotic Products Containing Streptococcus salivarius K12 by Assessment of Probiotic Viability and Inhibitory Potency against Respiratory Pathogens. Processes (Basel) 2023. [DOI: 10.3390/pr11020622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
Upper respiratory infections (URI) are the most frequent illnesses, especially in children. The majority of those infections are prescribed broad-spectrum antibiotics, which are associated with various side effects and with the increase in multi-drug-resistant strains. A promising alternative approach is the administration of the probiotic strain Streptococcus salivarius K12 (SSK12) that colonizes the upper respiratory tract (URT) and produces the salivaricins A2 and B, which strongly antagonize the growth of key respiratory pathogens. However, since for food supplements no quality controls of the active probiotic ingredient are mandatory, the efficacy of commercial products containing SSK12 may vary. This study aimed to investigate the in vitro efficacy of several commercial SSK12-containing probiotics, positioned for the prevention of respiratory infections. The parameters evaluated to determine the in vitro efficacy included the viability of the probiotic bacterial strain and the minimum inhibitory dilution (MID) of the probiotic, determined by the agar spot method, against the pathogenic/potential pathogenic bacterial strains Streptococcus pyogenes FF22 and Micrococcus luteus T18. All tests were carried out both 12 and 24 months after manufacturing (AM) for each commercial product. The viability ranged from 9 × 108 to 4.4 × 109 CFU/serving at 12 months AM and from 8.5 × 107 to 2.8 × 109 CFU/serving at 24 months AM. The MID was, in general, positively correlated with the probiotic bacterium viability and varied between the commercial products, ranging from 10−5 to 10−7 at 12 months AM and from 10−4 to 10−7 at 24 months AM. Moreover, the inhibition zones related to the two indicator strains were variable in diameter for different products. The high variation of the in vitro efficacy of commercial products containing SSK12 may explain the different results reported in the literature regarding the clinical benefits of these preparations, and the determination of this parameter may be useful to evaluate the quality of probiotic products containing this bacterial strain.
Collapse
|
7
|
Li J, Jin J, Li S, Zhong Y, Jin Y, Zhang X, Xia B, Zhu Y, Guo R, Sun X, Guo J, Hu F, Xiao W, Huang F, Ye H, Li R, Zhou Y, Xiang X, Yao H, Yan Q, Su L, Wu L, Luo T, Liu Y, Guo X, Qin J, Qi H, He J, Wang J, Li Z. Tonsillar Microbiome-Derived Lantibiotics Induce Structural Changes of IL-6 and IL-21 Receptors and Modulate Host Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202706. [PMID: 36031409 PMCID: PMC9596850 DOI: 10.1002/advs.202202706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/22/2022] [Indexed: 06/15/2023]
Abstract
Emerging evidence emphasizes the functional impacts of host microbiome on the etiopathogenesis of autoimmune diseases, including rheumatoid arthritis (RA). However, there are limited mechanistic insights into the contribution of microbial biomolecules especially microbial peptides toward modulating immune homeostasis. Here, by mining the metagenomics data of tonsillar microbiome, a deficiency of the encoding genes of lantibiotic peptides salivaricins in RA patients is identified, which shows strong correlation with circulating immune cells. Evidence is provided that the salivaricins exert immunomodulatory effects in inhibiting T follicular helper (Tfh) cell differentiation and interleukin-21 (IL-21) production. Mechanically, salivaricins directly bind to and induce conformational changes of IL-6 and IL-21 receptors, thereby inhibiting the bindings of IL-6 and IL-21 to their receptors and suppressing the downstream signaling pathway. Finally, salivaricin administration exerts both prophylactic and therapeutic effects against experimental arthritis in a murine model of RA. Together, these results provide a mechanism link of microbial peptides-mediated immunomodulation.
Collapse
|
8
|
Ongpipattanakul C, Desormeaux EK, DiCaprio A, van der Donk WA, Mitchell DA, Nair SK. Mechanism of Action of Ribosomally Synthesized and Post-Translationally Modified Peptides. Chem Rev 2022; 122:14722-14814. [PMID: 36049139 PMCID: PMC9897510 DOI: 10.1021/acs.chemrev.2c00210] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ribosomally synthesized and post-translationally modified peptides (RiPPs) are a natural product class that has undergone significant expansion due to the rapid growth in genome sequencing data and recognition that they are made by biosynthetic pathways that share many characteristic features. Their mode of actions cover a wide range of biological processes and include binding to membranes, receptors, enzymes, lipids, RNA, and metals as well as use as cofactors and signaling molecules. This review covers the currently known modes of action (MOA) of RiPPs. In turn, the mechanisms by which these molecules interact with their natural targets provide a rich set of molecular paradigms that can be used for the design or evolution of new or improved activities given the relative ease of engineering RiPPs. In this review, coverage is limited to RiPPs originating from bacteria.
Collapse
Affiliation(s)
- Chayanid Ongpipattanakul
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Emily K. Desormeaux
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Adam DiCaprio
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
| | - Wilfred A. van der Donk
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Howard Hughes Medical Institute, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Douglas A. Mitchell
- Department of Chemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Department of Microbiology, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| | - Satish K. Nair
- Department of Biochemistry, University of Illinois at Urbana-Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, USA
- Departments of Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 1206 West Gregory Drive, Urbana, Illinois 61801, USA
| |
Collapse
|
9
|
Yoo JM, Mendoza RM, Hwang IC, Kang DK. Whole genome sequence analysis of Ligilactobacillus agilis C7 isolated from pig feces revealed three bacteriocin gene clusters. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1008-1011. [PMID: 36287741 PMCID: PMC9574610 DOI: 10.5187/jast.2022.e55] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 11/05/2022]
Abstract
We here report the whole genome sequence of Ligilactobacillus agilis C7 with anti-listerial activity, which was isolated from pig feces. The genome size of L. agilis C7 (~ 3.0 Mb) is relatively larger compared with other L. agilis strains. L. agilis C7 carries three bacteriocin gene clusters encoding garvicin Q, salivaricin A, and Blp family class II bacteriocin. Garvicin Q and salivaricin A are reported to be active against Listeria monocytogenes and Micrococcus luteus, respectively, as well as against other Gram-positive bacteria. Meanwhile, the bacteriocin encoded in the blp cassette was shown to be active against pneumococci, mediating intraspecies competition. This report highlights the potential of L. agilis C7 for the production of bacteriocins inhibiting pathogenic bacteria.
Collapse
Affiliation(s)
- Jeong Min Yoo
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Remilyn M. Mendoza
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - In-Chan Hwang
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science,
Dankook University, Cheonan 31116, Korea,Corresponding author: Dae-Kyung Kang,
Department of Animal Resources Science, Dankook University, Cheonan 31116,
Korea., Tel: +82-41-550-3655, E-mail:
| |
Collapse
|
10
|
Lawrence GW, McCarthy N, Walsh CJ, Kunyoshi TM, Lawton EM, O’Connor PM, Begley M, Cotter PD, Guinane CM. Effect of a bacteriocin-producing Streptococcus salivarius on the pathogen Fusobacterium nucleatum in a model of the human distal colon. Gut Microbes 2022; 14:2100203. [PMID: 35877697 PMCID: PMC9318236 DOI: 10.1080/19490976.2022.2100203] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The gut microbiome is a vast reservoir of microbes, some of which produce antimicrobial peptides called bacteriocins that may inhibit specific bacteria associated with disease. Fusobacterium nucleatum is an emerging human bacterial pathogen associated with gastrointestinal diseases including colorectal cancer (CRC). In this study, fecal samples of healthy donors were screened for potential bacteriocin-producing probiotics with antimicrobial activity against F. nucleatum. A novel isolate, designated as Streptococcus salivarius DPC6993 demonstrated a narrow-spectrum of antimicrobial activity against F. nucleatum in vitro. In silico analysis of the S. salivarius DPC6993 genome revealed the presence of genes involved in the production of the bacteriocins salivaricin A5 and salivaricin B. After 6 h in a colon fermentation model, there was a significant drop in the number of F. nucleatum in samples that had been simultaneously inoculated with S. salivarius DPC6993 + F. nucleatum DSM15643 compared to those inoculated with F. nucleatum DSM15643 alone (mean ± SD: 9243.3 ± 3408.4 vs 29688.9 ± 4993.9 copies/μl). Furthermore, 16S rRNA amplicon analysis revealed a significant difference in the mean relative abundances of Fusobacterium between samples inoculated with both S. salivarius DPC6993 and F. nucleatum DSM15643 (0.05%) and F. nucleatum DSM15643 only (0.32%). Diversity analysis indicated minimal impact exerted by S. salivarius DPC6993 on the surrounding microbiota. Overall, this study highlights the ability of a natural gut bacterium to target a bacterial pathogen associated with CRC. The specific targeting of CRC-associated pathogens by biotherapeutics may ultimately reduce the risk of CRC development and positively impact CRC outcomes.
Collapse
Affiliation(s)
- Garreth W. Lawrence
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Niamh McCarthy
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Calum J. Walsh
- Food Biosciences, Teagasc Food Research Centre Moorepark, Cork, Ireland,VistaMilk SFI Research Centre, Moorepark, Fermoy, Cork, Ireland
| | | | | | - Paula M. O’Connor
- Food Biosciences, Teagasc Food Research Centre Moorepark, Cork, Ireland,VistaMilk SFI Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Máire Begley
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Paul D. Cotter
- Food Biosciences, Teagasc Food Research Centre Moorepark, Cork, Ireland,VistaMilk SFI Research Centre, Moorepark, Fermoy, Cork, Ireland,Paul D. Cotter Food Biosciences, Teagasc Food Research Centre Moorepark, APC Microbiome Ireland, Cork, Ireland
| | - Caitriona M. Guinane
- Department of Biological Sciences, Munster Technological University, Cork, Ireland,CONTACT Caitriona M. Guinane Department of Biological Sciences, Munster Technological University, Cork, Ireland
| |
Collapse
|
11
|
Guo H, Xiang X, Lin X, Wang Q, Qin S, Lu X, Xu J, Fang Y, Liu Y, Cui J, Li Z. Oropharyngeal Probiotic ENT-K12 as an Effective Dietary Intervention for Children With Recurrent Respiratory Tract Infections During Cold Season. Front Nutr 2022; 9:900448. [PMID: 35634421 PMCID: PMC9132010 DOI: 10.3389/fnut.2022.900448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Recurrent respiratory tract infections (RRTi) cause a high burden of disease and lead to negative impact on quality of life, frequent school/work absenteeism, and doctor visits, which remain a great challenge to pediatricians because RRTi can increase the risk of various complications including antibiotic overuse and resistance, which is one of the biggest threats to global health, and there is no confirmed effective treatment. In this study, we aimed to assess the clinical efficacy and safety of oropharyngeal probiotic ENT-K12 as a dietary intervention or a complementary treatment along with standard medical treatment during acute respiratory infections among children with RRTi during cold season. The results of this study show that when comparing to practicing of standard medical treatment only, the complementary intake of oropharyngeal probiotic ENT-K12 can effectively reduce episodes of both acute and RRTi in school children, shorten the course of respiratory symptoms onset, reduce the use of antibiotics and antiviral drugs, and reduce the absence days from both children's school and parents' work. Using oropharyngeal probiotics as a complementary dietary intervention to stabilize oropharyngeal microflora, specifically inhibiting respiratory pathogens and enhancing host immunity, could possibly be a promising approach to reduce RRTi burden and combating antibiotic resistance in long term, more clinical studies will be needed to further confirm the clinical practicing guide to ensure its clinical benefit.
Collapse
Affiliation(s)
- Hongyan Guo
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaochen Xiang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xuan Lin
- Department of Endocrinology, CR & WISCO General Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Si Qin
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xinyan Lu
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jiawei Xu
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Ying Fang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Yang Liu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jing Cui
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Vidal Amaral JR, Jucá Ramos RT, Almeida Araújo F, Bentes Kato R, Figueira Aburjaile F, de Castro Soares S, Góes-Neto A, Matiuzzi da Costa M, Azevedo V, Brenig B, Soares de Oliveira S, Soares Rosado A. Bacteriocin Producing Streptococcus agalactiae Strains Isolated from Bovine Mastitis in Brazil. Microorganisms 2022; 10:microorganisms10030588. [PMID: 35336163 PMCID: PMC8953382 DOI: 10.3390/microorganisms10030588] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 11/18/2022] Open
Abstract
Antibiotic resistance is one of the biggest health challenges of our time. We are now facing a post-antibiotic era in which microbial infections, currently treatable, could become fatal. In this scenario, antimicrobial peptides such as bacteriocins represent an alternative solution to traditional antibiotics because they are produced by many organisms and can inhibit bacteria, fungi, and/or viruses. Herein, we assessed the antimicrobial activity and biotechnological potential of 54 Streptococcus agalactiae strains isolated from bovine mastitis. Deferred plate antagonism assays revealed an inhibition spectrum focused on species of the genus Streptococcus—namely, S. pyogenes, S. agalactiae, S. porcinus, and S. uberis. Three genomes were successfully sequenced, allowing for their taxonomic confirmation via a multilocus sequence analysis (MLSA). Virulence potential and antibiotic resistance assessments showed that strain LGMAI_St_08 is slightly more pathogenic than the others. Moreover, the mreA gene was identified in the three strains. This gene is associated with resistance against erythromycin, azithromycin, and spiramycin. Assessments for secondary metabolites and antimicrobial peptides detected the bacteriocin zoocin A. Finally, comparative genomics evidenced high similarity among the genomes, with more significant similarity between the LGMAI_St_11 and LGMAI_St_14 strains. Thus, the current study shows promising antimicrobial and biotechnological potential for the Streptococcus agalactiae strains.
Collapse
Affiliation(s)
- João Ricardo Vidal Amaral
- Institute of Microbiology, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | | | - Fabrício Almeida Araújo
- Socio-Environmental and Water Resources Institute, Universidade Federal Rural da Amazônia, Belém 66077-830, PA, Brazil
| | - Rodrigo Bentes Kato
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Flávia Figueira Aburjaile
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Siomar de Castro Soares
- Institute of Biological and Natural Sciences, Universidade Federal do Triângulo Mineiro, Uberaba 38025-180, MG, Brazil
| | - Aristóteles Góes-Neto
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Mateus Matiuzzi da Costa
- Department of Biological Sciences, Universidade Federal do Vale do São Francisco, Petrolina 56304-917, PE, Brazil
| | - Vasco Azevedo
- Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil
| | - Bertram Brenig
- Department of Molecular Biology of Livestock, Institute of Veterinary Medicine, Georg August University Göttingen, 37077 Göttingen, Germany
| | - Selma Soares de Oliveira
- Institute of Microbiology, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | - Alexandre Soares Rosado
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Makkah 23955, Saudi Arabia
| |
Collapse
|
13
|
Hale JDF, Jain R, Wescombe PA, Burton JP, Simon RR, Tagg JR. Safety assessment of Streptococcus salivarius M18 a probiotic for oral health. Benef Microbes 2022; 13:47-60. [PMID: 35098909 DOI: 10.3920/bm2021.0107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of probiotics targeting non-intestinal body sites continues to generate interest amongst researchers, biotech companies and consumers alike. A key consideration for any bacterial strain to be developed into a probiotic is a robust assessment of its safety profile. Streptococcus salivarius strain M18 was originally isolated from a healthy adult and evaluated for its probiotic capabilities targeted to dental and oral health applications. This publication presents the safety characterisation of strain M18. Application of a diverse range of techniques showed that strain M18 can be specifically distinguished from other S. salivarius using a variety of molecular and phenotypic methodologies and that it lacks any relevant antibiotic resistance or virulence determinants. Direct comparison of the strain M18 safety profile with that of the prototype S. salivarius probiotic, S. salivarius strain K12, supports the proposition that strain M18 is indeed safe for probiotic application in humans.
Collapse
Affiliation(s)
- J D F Hale
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| | - R Jain
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| | - P A Wescombe
- Yili Innovation Center Oceania, Lincoln University, Christchurch, New Zealand
| | - J P Burton
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, Canada
| | - R R Simon
- Intertek Health Sciences Inc, Mississauga, ON, Canada
| | - J R Tagg
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| |
Collapse
|
14
|
Vogel V, Spellerberg B. Bacteriocin Production by Beta-Hemolytic Streptococci. Pathogens 2021; 10:pathogens10070867. [PMID: 34358017 PMCID: PMC8308785 DOI: 10.3390/pathogens10070867] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/17/2023] Open
Abstract
Beta-hemolytic streptococci cause a variety of infectious diseases associated with high morbidity and mortality. A key factor for successful infection is host colonization, which can be difficult in a multispecies environment. Secreting bacteriocins can be beneficial during this process. Bacteriocins are small, ribosomally produced, antimicrobial peptides produced by bacteria to inhibit the growth of other, typically closely related, bacteria. In this systematic review, bacteriocin production and regulation of beta-hemolytic streptococci was surveyed. While Streptococcus pyogenes produces eight different bacteriocins (Streptococcin A-FF22/A-M49, Streptin, Salivaricin A, SpbMN, Blp1, Blp2, Streptococcin A-M57), only one bacteriocin of Streptococcus agalactiae (Agalacticin = Nisin P) and one of Streptococcus dysgalactiae subsp. equisimilis (Dysgalacticin) has been described. Expression of class I bacteriocins is regulated by a two-component system, typically with autoinduction by the bacteriocin itself. In contrast, a separate quorum sensing system regulates expression of class II bacteriocins. Both identified class III bacteriocins are plasmid-encoded and regulation has not been elucidated.
Collapse
|
15
|
Chandrasekhar SN, Mallikarjun SB, Salim HP. Comparative Evaluation of Antibacterial Activity of Probiotics SK12 and SM18: An In Vitro Study. Int J Clin Pediatr Dent 2021; 13:611-616. [PMID: 33976484 PMCID: PMC8060948 DOI: 10.5005/jp-journals-10005-1838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Aim To assess the antimicrobial activity of probiotics SK12 and SM18 on Streptococcus mutans and also to compare the antimicrobial activity of SK12 and SM18. Materials and methods Synthetic strains of Streptococcus mutans were used to study the antimicrobial activity of probiotics SK12 and SM18 using various tests such as disk diffusion, minimum inhibitory concentration (MIC), and minimum bactericidal concentration (MBC). In disk diffusion, the zone of inhibition was measured to assess the antimicrobial activity. Chlorhexidine was used as a control for this test. The MIC and MBC were assessed at different dilutions of the probiotic sample (100 mg/mL, 50 mg/mL, 25 mg/mL, 12.5 mg/mL, 6.25 mg/mL, 3.12 mg/mL, 1.6 mg/mL, 0.8 mg/mL, 0.4 mg/mL, and 0.2 mg/mL). Result SM18 demonstrated 20 mm of zone of inhibition, whereas SK12 demonstrated 15 mm showing a less antibacterial activity in comparison to SM18. SM18 was found to be bactericidal and effective at a minimum concentration of 0.8 mg/mL, whereas SK12 was bactericidal and effective at a minimum concentration of 1.6 mg/mL. Conclusion Probiotics demonstrate antibacterial activity against cariogenic microflora. SM is 18 having a better antibacterial activity at lower concentrations than SK12 in reducing cariogenic microorganisms. Clinical significance: BLIS K12 and M18 both demonstrated an antibacterial effect on Streptococcus mutans, wherein the use of probiotic in caries prevention is found to be limited. Hence, it is suggestive to reap the bacterial effects of BLIS K12 and M18 in caries prevention. How to cite this article Chandrasekhar SN, Mallikarjun SB, Salim HP. Comparative Evaluation of Antibacterial Activity of Probiotics SK12 and SM18: An In Vitro Study. Int J Clin Pediatr Dent 2020;13(6):611–616.
Collapse
Affiliation(s)
- Srihari Nirguna Chandrasekhar
- Department of Pediatric and Preventive Dentistry, Coorg Institute of Dental Sciences, Virajpet, Karnataka, India
- Srihari Nirguna Chandrasekhar, Department of Pediatric and Preventive Dentistry, Coorg Institute of Dental Sciences, Virajpet, Karnataka, India, Phone: +919480174728, e-mail:
| | - Shanthala B Mallikarjun
- Department of Pediatric and Preventive Dentistry, Coorg Institute of Dental Sciences, Virajpet, Karnataka, India
| | - Henna P Salim
- Department of Pediatric and Preventive Dentistry, Coorg Institute of Dental Sciences, Virajpet, Karnataka, India
| |
Collapse
|
16
|
García-Curiel L, Del Rocío López-Cuellar M, Rodríguez-Hernández AI, Chavarría-Hernández N. Toward understanding the signals of bacteriocin production by Streptococcus spp. and their importance in current applications. World J Microbiol Biotechnol 2021; 37:15. [PMID: 33394178 DOI: 10.1007/s11274-020-02973-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/01/2020] [Indexed: 12/25/2022]
Abstract
Microorganisms have developed quorum sensing (QS) systems to detect small signaling molecules that help to control access to additional nutrients and space in highly competitive polymicrobial niches. Many bacterial processes are QS-regulated; two examples are the highly related traits of the natural genetic competence state and the production of antimicrobial peptides such as bacteriocins. The Streptococcus genus is widely studied for its competence and for its ability to produce bacteriocins, as these antimicrobial peptides have significant potential in the treatment of infections caused by multiple-resistant pathogens, a severe public health issue. The transduction of a two-component system controls competence in streptococci: (1) ComD/E, which controls the competence in the Mitis and Anginosus groups, and (2) ComR/S, which performs the same function in the Bovis, Mutans, Salivarius, and Pyogenic groups. The cell-to-cell communication required for bacteriocin production in the Streptococcus groups is controlled mainly by a paralog of the ComD/E system. The relationships between pheromone signals and induction pathways are related to the bacteriocin production systems. In this review, we discuss the recent advances in the understanding of signaling and the induction of bacteriocin biosynthesis by QS regulation in streptococci. This information could aid in the design of better methods for the development and production of these antimicrobial peptides. It could also contribute to the analysis and emerging applications of bacteriocins in terms of their safety, quality, and human health benefits.
Collapse
Affiliation(s)
- Laura García-Curiel
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Ma Del Rocío López-Cuellar
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México.
| | - Adriana Inés Rodríguez-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Norberto Chavarría-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| |
Collapse
|
17
|
Barbour A, Wescombe P, Smith L. Evolution of Lantibiotic Salivaricins: New Weapons to Fight Infectious Diseases. Trends Microbiol 2020; 28:578-593. [PMID: 32544444 DOI: 10.1016/j.tim.2020.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 01/20/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023]
Abstract
Lantibiotic salivaricins are polycyclic peptides containing lanthionine and/or β-methyllanthionine residues produced by certain strains of Streptococcus salivarius, which almost exclusively reside in the human oral cavity. The importance of these molecules stems from their antimicrobial activity towards relevant oral pathogens which has so far been applied through the development of salivaricin-producing probiotic strains. However, salivaricins may also prove to be of great value in the development of new and novel antibacterial therapies in this era of emerging antibiotic resistance. In this review, we describe the biosynthesis, antimicrobial activity, structure, and mode of action of the lantibiotic salivaricins characterized to date. Moreover, we also provide an expert opinion and suggestions for future development of this important field of microbiology.
Collapse
Affiliation(s)
| | - Philip Wescombe
- Yili Innovation Center Oceania, Lincoln University, Christchurch, New Zealand
| | - Leif Smith
- Department of Biology, College of Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
18
|
Derovs A, Laivacuma S, Krumina A. Targeting Microbiota: What Do We Know about It at Present? ACTA ACUST UNITED AC 2019; 55:medicina55080459. [PMID: 31405111 PMCID: PMC6723830 DOI: 10.3390/medicina55080459] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022]
Abstract
The human microbiota is a variety of different microorganisms. The composition of microbiota varies from host to host, and it changes during the lifetime. It is known that microbiome may be changed because of a diet, bacteriophages and different processes for example, such as inflammation. Like all other areas of medicine, there is a continuous growth in the area of microbiology. Different microbes can reside in all sites of a human body, even in locations that were previously considered as sterile; for example, liver, pancreas, brain and adipose tissue. Presently one of the etiological factors for liver disease is considered to be pro-inflammatory changes in a host’s organism. There are lot of supporting data about intestinal dysbiosis and increased intestinal permeability and its effect on development of liver disease pointing to the gut–liver axis. The gut–liver axis affects pathogenesis of many liver diseases, such as chronic hepatitis B, chronic hepatitis C, alcoholic liver disease, non-alcoholic liver disease, non-alcoholic steatohepatitis, liver cirrhosis and hepatocellular carcinoma. Gut microbiota has been implicated in the regulation of brain health, emphasizing the gut–brain axis. Also, experiments with mice showed that microorganisms have significant effects on the blood–brain barrier integrity. Microbiota can modulate a variety of mechanisms through the gut–liver axis and gut–brain axis. Normal intestinal flora impacts the health of a host in many positive ways, but there is now significant evidence that intestinal microbiota, especially altered, have the ability to impact the pathologies of many diseases through different inflammatory mechanisms. At this point, many of the pathophysiological reactions in case of microbial disbyosis are still unclear.
Collapse
Affiliation(s)
- Aleksejs Derovs
- Riga Stradins University, Department of Internal Medicine, LV-1007 Riga, Latvia.
- Riga East Clinical University Hospital, LV-1038 Riga, Latvia.
- Riga Stradins University, Department of Infectology and Dermatology, LV-1007 Riga, Latvia.
| | - Sniedze Laivacuma
- Riga East Clinical University Hospital, LV-1038 Riga, Latvia
- Riga Stradins University, Department of Infectology and Dermatology, LV-1007 Riga, Latvia
| | - Angelika Krumina
- Riga East Clinical University Hospital, LV-1038 Riga, Latvia
- Riga Stradins University, Department of Infectology and Dermatology, LV-1007 Riga, Latvia
| |
Collapse
|
19
|
Jia GC, Che N, Xia YJ, Lai PFH, Xiong ZQ, Wang GQ, Zhang H, Ai LZ. Adhesion to pharyngeal epithelium and modulation of immune response: Lactobacillus salivarius AR809, a potential probiotic strain isolated from the human oral cavity. J Dairy Sci 2019; 102:6738-6749. [PMID: 31178178 DOI: 10.3168/jds.2018-16117] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
Abstract
Microbiome modulators such as probiotics are known to modulate oral diseases. Very few probiotics are commercially available for use in the oral cavity. In this context, we selected human-origin Lactobacillus salivarius AR809 as a promising oropharyngeal probiotic and characterized its functional and immunomodulatory properties. Results demonstrated that AR809 could efficiently adhere to pharyngeal epithelial FaDu cells, antagonize Staphylococcus aureus, adapt to the oral environment, and modulate host innate immunity by inducing potentially protective effects. Particularly, AR809 diminished proinflammatory activity by enhancing the production of IL10 and inhibiting the expression of tumor necrosis factor-α, IL1B, inducible nitric oxide synthase, and RELA. Finally, we observed that AR809 grew efficiently when cultured in milk, suggesting that the preparation of a fermented milk product containing AR809 could be a practical way to administer this probiotic to humans. In conclusion, AR809 has high potential to adhere to the pharyngeal mucosa and could be applied in novel milk-based probiotic fermented food products.
Collapse
Affiliation(s)
- G C Jia
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - N Che
- Department of Otolaryngology, Tongji Hospital, Tongji University, Shanghai 200065, China
| | - Y J Xia
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Phoency F-H Lai
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Z Q Xiong
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - G Q Wang
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - H Zhang
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - L Z Ai
- Shanghai Engineering Research Center of Food Microbiology, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
20
|
Wholey WY, Abu-Khdeir M, Yu EA, Siddiqui S, Esimai O, Dawid S. Characterization of the Competitive Pneumocin Peptides of Streptococcus pneumoniae. Front Cell Infect Microbiol 2019; 9:55. [PMID: 30915281 PMCID: PMC6422914 DOI: 10.3389/fcimb.2019.00055] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 02/20/2019] [Indexed: 01/01/2023] Open
Abstract
In the polymicrobial environment of the human nasopharynx, Streptococcus pneumoniae (pneumococcus) competes with other members of the microbial community for limited nutrients in part by secreting small peptide bacteriocins called pneumocins. Pneumocin production is controlled by a quorum sensing system encoded by the blp locus. Although the locus is found in all pneumococci, there is significant variability in the repertoire of pneumocins and associated immunity proteins encoded in the Bacteriocin Immunity Region (BIR) and in the presence or absence of a functional Blp transporter. Strains without an active Blp transporter are inactive in plate overlay assays and rely on a homologous transporter that is only produced during brief periods of competence to stimulate the blp locus and secrete pneumocins. The variability of the locus suggests that selective pressure is influencing the content to promote the optimal competitive environment. Much of the variability in the blp locus has been described at the genome level; the phenotypic activity attributable to the various BIR genes has not been fully described. To examine the role of the predicted pneumocin peptides in competition, 454 isolates were screened for competence independent blp pheromone secretion using plate assays. Active strains were characterized for inhibition, BIR content, BlpC pherotype and serotype. Deletion analysis on inhibitory strains demonstrated that BlpI and BlpJ peptides function as a two-peptide bacteriocin and that BlpIJ immunity is encoded by the co-transcribed blpU4/5 genes. BlpIJ secretion promotes inhibitory activity against the majority of pneumococcal isolates when expressed in a Blp transporter intact background. Intermediate levels of competition in biofilms were noted when BlpIJ containing strains carried the non-functional Blp transporter. Based on genome data, the combination of BlpIJ in a Blp transporter intact strain is surprisingly rare, despite clear advantages during colonization and biofilm growth. In contrast, we show that the blpK/pncF operon encoding the single-peptide pneumocin BlpK and its immunity protein is found in the majority of isolates. Unlike, BlpIJ and BlpK were shown to promote a limited spectrum of inhibition due in part to immunity that is independent of activation of the blp locus.
Collapse
Affiliation(s)
- Wei-Yun Wholey
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maha Abu-Khdeir
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Emily A Yu
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Saher Siddiqui
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Ogenna Esimai
- Department of Computer Science and Engineering, University of Texas at Arlington, Arlington, TX, United States
| | - Suzanne Dawid
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
21
|
Geng M, Deng P, Mire T, Austin F, Smith L. Draft Genome Sequence of the Lantibiotic-Producing Strain Streptococcus salivarius HS0302. Microbiol Resour Announc 2019; 8:e01410-18. [PMID: 30637392 PMCID: PMC6318363 DOI: 10.1128/mra.01410-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/16/2018] [Indexed: 11/29/2022] Open
Abstract
Streptococcus salivarius is a prevalent commensal species of human oral mucosal surfaces. S. salivarius strain HS0302 produces the type AII lantibiotic salivaricin A2. Here, we report its draft genome sequence, revealing its potential to produce a variety of bacteriocins.
Collapse
Affiliation(s)
- Mengxin Geng
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Peng Deng
- Chinese Academy of Financial Inclusion, Renmin University of China, Beijing, China
| | - Timothy Mire
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Frank Austin
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Leif Smith
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
22
|
Renye JA, Somkuti GA, Steinberg DH. Thermophilin 109 is a naturally produced broad spectrum bacteriocin encoded within the blp gene cluster of Streptococcus thermophilus. Biotechnol Lett 2018; 41:283-292. [DOI: 10.1007/s10529-018-02637-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/05/2018] [Indexed: 11/28/2022]
|
23
|
Covalent Structure and Bioactivity of the Type AII Lantibiotic Salivaricin A2. Appl Environ Microbiol 2018; 84:AEM.02528-17. [PMID: 29269497 DOI: 10.1128/aem.02528-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/18/2017] [Indexed: 01/31/2023] Open
Abstract
Lantibiotics are a class of lanthionine-containing, ribosomally synthesized, and posttranslationally modified peptides (RiPPs) produced by Gram-positive bacteria. Salivaricin A2 belongs to the type AII lantibiotics, which are generally considered to kill Gram-positive bacteria by binding to the cell wall precursor lipid II via a conserved ring A structure. Salivaricin A2 was first reported to be isolated from a probiotic strain, Streptococcus salivarius K12, but the structural and bioactivity characterizations of the antibiotic have remained limited. In this study, salivaricin A2 was purified and its covalent structure was characterized. N-terminal analogues of salivaricin A2 were generated to study the importance for bioactivity of the length and charge of the N-terminal amino acids. Analogue salivaricin A2(3-22) has no antibacterial activity and does not have an antagonistic effect on the native compound. The truncated analogue also lost its ability to bind to lipid II in a thin-layer chromatography (TLC) assay, suggesting that the N-terminal amino acids are important for binding to lipid II. The creation of N-terminal analogues of salivaricin A2 promoted a better understanding of the bioactivity of this antibiotic and further elucidated the structural importance of the N-terminal leader peptide. The antibacterial activity of salivaricin A2 is due not only to the presence of the positively charged N-terminal amino acid residues, but to the length of the N-terminal linear peptide.IMPORTANCE The amino acid composition of the N-terminal linear peptide of salivaricin A2 is crucial for function. Our study shows that the length of the amino acid residues in the linear peptide is crucial for salivaricin A2 antimicrobial activity. Very few type AII lantibiotic covalent structures have been confirmed. The characterization of the covalent structure of salivaricin A2 provides additional support for the predicted lanthionine and methyl-lanthionine ring formations present in this structural class of lantibiotics. Removal of the N-terminal Lys1 and Arg2 residues from the peptide causes a dramatic shift in the chemical shift values of amino acid residues 7 through 9, suggesting that the N-terminal amino acids contribute to a distinct structural conformer for the linear peptide region. The demonstration that the bioactivity could be partially restored with the substitution of N-terminal alanine residues supports further studies aimed at determining whether new analogues of salivaricin A2 for novel applications can be synthesized.
Collapse
|
24
|
Mousa WK, Athar B, Merwin NJ, Magarvey NA. Antibiotics and specialized metabolites from the human microbiota. Nat Prod Rep 2017; 34:1302-1331. [DOI: 10.1039/c7np00021a] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human microbiota associated with each body site produce specialized molecules to kill human pathogens. Advanced bioinformatics tools will help to discover unique microbiome chemistry.
Collapse
Affiliation(s)
- Walaa K. Mousa
- Departments of Biochemistry and Biomedical Sciences & Chemistry and Chemical Biology
- M. G. DeGroote Institute for Infectious Disease Research
- McMaster University
- Hamilton
- Canada L8S 4K1
| | - Bilal Athar
- Departments of Biochemistry and Biomedical Sciences & Chemistry and Chemical Biology
- M. G. DeGroote Institute for Infectious Disease Research
- McMaster University
- Hamilton
- Canada L8S 4K1
| | - Nishanth J. Merwin
- Departments of Biochemistry and Biomedical Sciences & Chemistry and Chemical Biology
- M. G. DeGroote Institute for Infectious Disease Research
- McMaster University
- Hamilton
- Canada L8S 4K1
| | - Nathan A. Magarvey
- Departments of Biochemistry and Biomedical Sciences & Chemistry and Chemical Biology
- M. G. DeGroote Institute for Infectious Disease Research
- McMaster University
- Hamilton
- Canada L8S 4K1
| |
Collapse
|
25
|
Identification of a two-component Class IIb bacteriocin in Streptococcus pyogenes by recombinase-based in vivo expression technology. Sci Rep 2016; 6:36233. [PMID: 27808235 PMCID: PMC5093712 DOI: 10.1038/srep36233] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 10/10/2016] [Indexed: 12/30/2022] Open
Abstract
Streptococcus pyogenes is a globally prominent bacterial pathogen that exhibits strict tropism for the human host, yet bacterial factors responsible for the ability of S. pyogenes to compete within this limited biological niche are not well understood. Using an engineered recombinase-based in vivo expression technology (RIVET) system, we identified an in vivo-induced promoter region upstream of a predicted Class IIb bacteriocin system in the M18 serotype S. pyogenes strain MGAS8232. This promoter element was not active under in vitro laboratory conditions, but was highly induced within the mouse nasopharynx. Recombinant expression of the predicted mature S. pyogenes bacteriocin peptides (designated SpbM and SpbN) revealed that both peptides were required for antimicrobial activity. Using a gain of function experiment in Lactococcus lactis, we further demonstrated S. pyogenes immunity function is encoded downstream of spbN. These data highlight the importance of bacterial gene regulation within appropriate environments to help understand mechanisms of niche adaptation by bacterial pathogens.
Collapse
|
26
|
Palma TH, Harth-Chú EN, Scott J, Stipp RN, Boisvert H, Salomão MF, Theobaldo JD, Possobon RF, Nascimento LC, McCafferty JW, Faller L, Duncan MJ, Mattos-Graner RO. Oral cavities of healthy infants harbour high proportions of Streptococcus salivarius strains with phenotypic and genotypic resistance to multiple classes of antibiotics. J Med Microbiol 2016; 65:1456-1464. [PMID: 27902365 DOI: 10.1099/jmm.0.000377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Emerging antibiotic resistance in the oropharyngeal microbiota, of which Streptococcus salivarius is a prominent species, represents a challenge for treating paediatric populations. In this study, we investigated the role of Streptococcussalivarius as a reservoir for antibiotic resistance genes (ARG) in the oral microbiota by analysing 95 Streptococcussalivarius isolates from 22 healthy infants (2-16 months of age). MICs of penicillin G, amoxicillin, erythromycin, tetracycline, doxycycline and streptomycin were determined. ARG profiles were assessed in a subset of 21 strains by next-generation sequencing of genomes, followed by searches of assembled reads against the Comprehensive Antibiotic Resistance Database. Strains resistant to erythromycin, penicillins and tetracyclines were isolated from 83.3, 33.3 and 16.6 %, respectively, of infants aged 2 to 8 months with no prior antibiotic treatment. These percentages were100.0, 66.6 and 50.0 %, by 13 to 16 months of age. ARG or polymorphisms associated with antibiotic resistance were the most prevalent and involved genes for macrolide efflux (mel, mefA/E and macB), ribosomal protection [erm(B), tet(M) and tet(O)] and β-lactamase-like proteins. Phylogenetically related strains showing multidrug-resistant phenotypes harboured multidrug efflux ARG. Polymorphic genes associated with antibiotic resistance to drugs affecting DNA replication, folate synthesis, RNA/protein synthesis and regulators of antibiotic stress responses were detected. These data imply that Streptococcussalivarius strains established during maturation of the oral microbiota harbour a diverse array of functional ARG, even in the absence of antibiotic selective pressures, highlighting a potential role for this species in shaping antibiotic susceptibility profiles of oropharyngeal communities.
Collapse
Affiliation(s)
- Thaís H Palma
- Department of Oral Diagnosis, University of Campinas, São Paulo, Brazil
| | - Erika N Harth-Chú
- Department of Oral Diagnosis, University of Campinas, São Paulo, Brazil
| | - Jodie Scott
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Rafael N Stipp
- Department of Oral Diagnosis, University of Campinas, São Paulo, Brazil
| | - Heike Boisvert
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | | | | | - Rosana F Possobon
- Cepae - Research and Dental Care Center for Special Needs Patients of Piracicaba Dental School, University of Campinas, São Paulo, Brazil
| | - Leandro C Nascimento
- Laboratory of Genomics and Gene Expression (LGE), University of Campinas, São Paulo, Brazil
| | | | - Lina Faller
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Margaret J Duncan
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | | |
Collapse
|
27
|
Genome Sequence of the Oral Probiotic Streptococcus salivarius JF. GENOME ANNOUNCEMENTS 2016; 4:4/5/e00971-16. [PMID: 27660775 PMCID: PMC5034126 DOI: 10.1128/genomea.00971-16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Streptococcus salivarius is a nonpathogenic Gram-positive bacterium and the predominant colonizer of the oral microbiota. It finds a wide application in the prevention of upper respiratory tract infections, also reducing the frequency of other main pathogens. Here, we present the complete genome sequence of the oral probiotic S. salivarius JF.
Collapse
|
28
|
Barbour A, Tagg J, Abou-Zied OK, Philip K. New insights into the mode of action of the lantibiotic salivaricin B. Sci Rep 2016; 6:31749. [PMID: 27526944 PMCID: PMC4985645 DOI: 10.1038/srep31749] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 07/26/2016] [Indexed: 01/19/2023] Open
Abstract
Salivaricin B is a 25 amino acid polycyclic peptide belonging to the type AII lantibiotics and first shown to be produced by Streptococcus salivarius. In this study we describe the bactericidal mode of action of salivaricin B against susceptible Gram-positive bacteria. The killing action of salivaricin B required micro-molar concentrations of lantibiotic whereas the prototype lantibiotic nisin A was shown to be potent at nano-molar levels. Unlike nisin A, salivaricin B did not induce pore formation or dissipate the membrane potential in susceptible cells. This was established by measuring the fluorescence of the tryptophan residue at position 17 when salivaricin B interacted with bacterial membrane vesicles. The absence of a fluorescence blue shift indicates a failure of salivaricin B to penetrate the membranes. On the other hand, salivaricin B interfered with cell wall biosynthesis, as shown by the accumulation of the final soluble cell wall precursor UDP-MurNAc-pentapeptide which is the backbone of the bacterial peptidoglycan. Transmission electron microscopy of salivaricin B-treated cells showed a reduction in cell wall thickness together with signs of aberrant septum formation in the absence of visible changes to cytoplasmic membrane integrity.
Collapse
Affiliation(s)
- Abdelahhad Barbour
- Division of Microbiology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala lumpur, Malaysia
| | - John Tagg
- Department of Microbiology and Immunology, University of Otago, P.O. Box 56, Dunedin 9054, New Zealand
| | - Osama K Abou-Zied
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, P.O. Box 36, Postal Code 123, Muscat, Sultanate of Oman
| | - Koshy Philip
- Division of Microbiology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala lumpur, Malaysia
| |
Collapse
|
29
|
Ge X, Teng K, Wang J, Zhao F, Wang F, Zhang J, Zhong J. Ligand determinants of nisin for its induction activity. J Dairy Sci 2016; 99:5022-5031. [PMID: 27132090 DOI: 10.3168/jds.2015-10809] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/23/2016] [Indexed: 11/19/2022]
Abstract
Nisin has been widely used in the food industry as a safe and natural preservative and has the potential to be used as a biomedicine. Improving nisin production is important for its enormous applications. Nisin A is produced in Lactococcus lactis and its biosynthesis is induced through the regulation of the 2-component system NisKR. In this study, alanine-scanning mutagenesis was applied to study the key structure or AA in nisin for inducing the 2-component system NisKR to regulate downstream gene expression. Assay of β-galactosidase activity revealed that either ring A or ring B was necessary for nisin to induce lacZ reporter gene expression. A substituted first ring formed by Thr2 and Cys7 in S3A instead of ring A (formed by Ser3 and Cys7) fully retained nisin induction activity. Mutation of cationic AA and addition of cationic ions hardly affected nisin induction activity. These results demonstrated that the N-terminal ring structures in nisin were involved in activating NisKR to act as an inducing molecule, whereas the electrostatic force might not contribute to this process. In addition, 2 specific residues were revealed to have potential for improving both nisin induction and antimicrobial activity, which might be used for increasing nisin production.
Collapse
Affiliation(s)
- Xiaoxuan Ge
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Kunling Teng
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.
| | - Jian Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; College of Engineering, University of Georgia, Athens 30602
| | - Fangyuan Zhao
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Fangfang Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Jie Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; University of Chinese Academy of Sciences, Beijing 100101, People's Republic of China
| | - Jin Zhong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.
| |
Collapse
|
30
|
Gashti MP, Asselin J, Barbeau J, Boudreau D, Greener J. A microfluidic platform with pH imaging for chemical and hydrodynamic stimulation of intact oral biofilms. LAB ON A CHIP 2016; 16:1412-9. [PMID: 26956837 DOI: 10.1039/c5lc01540e] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
A microfluidic platform with a fluorescent nanoparticle-based sensor is demonstrated for real-time, ratiometric pH imaging of biofilms. Sensing is accomplished by a thin patterned layer of covalently bonded Ag@SiO2+FiTC nanoparticles on an embedded planar glass substrate. The system is designed to be sensitive, responsive and give sufficient spatial resolution to enable new micro-scale studies of the dynamic response of oral biofilms to well-controlled chemical and hydrodynamic stimulation. Performance under challenging operational conditions is demonstrated, which include long-duration exposure to sheer stresses, photoexcitation and pH sensor biofouling. After comprehensive validation, the device was used to monitor pH changes at the attachment surface of a biofilm of the oral bacteria, Streptococcus salivarius. By controlling flow and chemical concentration conditions in the microchannel, biochemical and mass transport contributions to the Stephan curve could be probed individually. This opens the way for the analysis of separate contributions to dental caries due to localized acidification directly at the biofilm tooth interface.
Collapse
Affiliation(s)
| | - J Asselin
- Département de chimie, Université Laval, Québec (QC), G1V 0A6 Canada. and Centre d'optique, photonique et laser (COPL), Québec (QC), G1V 0A6 Canada
| | - J Barbeau
- Faculté de médecine dentaire, Université de Montréal (QC), H3C 3J4 Canada
| | - D Boudreau
- Département de chimie, Université Laval, Québec (QC), G1V 0A6 Canada. and Centre d'optique, photonique et laser (COPL), Québec (QC), G1V 0A6 Canada
| | - J Greener
- Département de chimie, Université Laval, Québec (QC), G1V 0A6 Canada.
| |
Collapse
|
31
|
Walker GV, Heng NCK, Carne A, Tagg JR, Wescombe PA. Salivaricin E and abundant dextranase activity may contribute to the anti-cariogenic potential of the probiotic candidate Streptococcus salivarius JH. Microbiology (Reading) 2016; 162:476-486. [DOI: 10.1099/mic.0.000237] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Glenn V. Walker
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Nicholas C. K. Heng
- Sir John Walsh Research Institute (Faculty of Dentistry), University of Otago, Dunedin, New Zealand
| | - Alan Carne
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - John R. Tagg
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Blis Technologies Ltd, Dunedin, New Zealand
| | | |
Collapse
|
32
|
Wescombe PA, Heng NCK, Burton JP, Tagg JR. Something Old and Something New: An Update on the Amazing Repertoire of Bacteriocins Produced by Streptococcus salivarius. Probiotics Antimicrob Proteins 2016; 2:37-45. [PMID: 26780899 DOI: 10.1007/s12602-009-9026-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Streptococcus salivarius has an exclusive and intimate association with humans. We are its sole natural host, and its contribution to the relationship appears overwhelmingly benevolent. Beautifully adapted to its preferred habitat, the human tongue, it only rarely ventures far from this location in the healthy host and indeed appears ill-equipped to become invasive due to a scarcity of virulence attributes. We consider that its strategically advantageous lingual location and numerical predominance allow S. salivarius to carry out a population surveillance and modulation role within the oral microbiota. Some strains are armed with complex arrays of targeted antibiotic weaponry, much of which belongs to the lantibiotic class of bacteriocins and a key to their ability to assemble and utilize this armament is their possession of transmissible multi-bacteriocin-encoding megaplasmid DNA. This review traces the origins of research into S. salivarius bacteriocins and bacteriocin-like inhibitory substances, showcases some of the inhibitory activities that we currently have knowledge of, and speculates about potential directions for ongoing investigation and probiotic application of this previously under-rated human commensal.
Collapse
Affiliation(s)
- Philip A Wescombe
- BLIS Technologies Ltd., Centre for Innovation, University of Otago, Dunedin, New Zealand
| | - Nicholas C K Heng
- Department of Oral Sciences, University of Otago, Dunedin, New Zealand
| | - Jeremy P Burton
- BLIS Technologies Ltd., Centre for Innovation, University of Otago, Dunedin, New Zealand
| | - John R Tagg
- BLIS Technologies Ltd., Centre for Innovation, University of Otago, Dunedin, New Zealand. .,Department of Microbiology and Immunology, University of Otago, P. O. Box 56, Dunedin, New Zealand.
| |
Collapse
|
33
|
Abstract
Developments in the use of genomics to guide natural product discovery and a recent emphasis on understanding the molecular mechanisms of microbiota-host interactions have converged on the discovery of small molecules from the human microbiome. Here, we review what is known about small molecules produced by the human microbiota. Numerous molecules representing each of the major metabolite classes have been found that have a variety of biological activities, including immune modulation and antibiosis. We discuss technologies that will affect how microbiota-derived molecules are discovered in the future and consider the challenges inherent in finding specific molecules that are critical for driving microbe-host and microbe-microbe interactions and understanding their biological relevance.
Collapse
Affiliation(s)
- Mohamed S Donia
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| | - Michael A Fischbach
- Department of Bioengineering and Therapeutic Sciences and the California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
34
|
Jimenez JC, Federle MJ. Quorum sensing in group A Streptococcus. Front Cell Infect Microbiol 2014; 4:127. [PMID: 25309879 PMCID: PMC4162386 DOI: 10.3389/fcimb.2014.00127] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 08/26/2014] [Indexed: 01/05/2023] Open
Abstract
Quorum sensing (QS) is a widespread phenomenon in the microbial world that has important implications in the coordination of population-wide responses in several bacterial pathogens. In Group A Streptococcus (GAS), many questions surrounding QS systems remain to be solved pertaining to their function and their contribution to the GAS lifestyle in the host. The QS systems of GAS described to date can be categorized into four groups: regulator gene of glucosyltransferase (Rgg), Sil, lantibiotic systems, and LuxS/AI-2. The Rgg family of proteins, a conserved group of transcription factors that modify their activity in response to signaling peptides, has been shown to regulate genes involved in virulence, biofilm formation and competence. The sil locus, whose expression is regulated by the activity of signaling peptides and a putative two-component system (TCS), has been implicated on regulating genes involved with invasive disease in GAS isolates. Lantibiotic regulatory systems are involved in the production of bacteriocins and their autoregulation, and some of these genes have been shown to target both bacterial organisms as well as processes of survival inside the infected host. Finally AI-2 (dihydroxy pentanedione, DPD), synthesized by the LuxS enzyme in several bacteria including GAS, has been proposed to be a universal bacterial communication molecule. In this review we discuss the mechanisms of these four systems, the putative functions of their targets, and pose critical questions for future studies.
Collapse
Affiliation(s)
- Juan Cristobal Jimenez
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Michael J Federle
- Department of Medicinal Chemistry and Pharmacognosy, Center for Pharmaceutical Biotechnology, College of Pharmacy, University of Illinois at Chicago Chicago, IL, USA
| |
Collapse
|
35
|
Barbour A, Philip K. Variable characteristics of bacteriocin-producing Streptococcus salivarius strains isolated from Malaysian subjects. PLoS One 2014; 9:e100541. [PMID: 24941127 PMCID: PMC4062538 DOI: 10.1371/journal.pone.0100541] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/28/2014] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Salivaricins are bacteriocins produced by Streptococcus salivarius, some strains of which can have significant probiotic effects. S. salivarius strains were isolated from Malaysian subjects showing variable antimicrobial activity, metabolic profile, antibiotic susceptibility and lantibiotic production. METHODOLOGY/PRINCIPAL FINDINGS In this study we report new S. salivarius strains isolated from Malaysian subjects with potential as probiotics. Safety assessment of these strains included their antibiotic susceptibility and metabolic profiles. Genome sequencing using Illumina's MiSeq system was performed for both strains NU10 and YU10 and demonstrating the absence of any known streptococcal virulence determinants indicating that these strains are safe for subsequent use as probiotics. Strain NU10 was found to harbour genes encoding salivaricins A and 9 while strain YU10 was shown to harbour genes encoding salivaricins A3, G32, streptin and slnA1 lantibiotic-like protein. Strain GT2 was shown to harbour genes encoding a large non-lantibiotic bacteriocin (salivaricin-MPS). A new medium for maximum biomass production buffered with 2-(N-morpholino)ethanesulfonic acid (MES) was developed and showed better biomass accumulation compared with other commercial media. Furthermore, we extracted and purified salivaricin 9 (by strain NU10) and salivaricin G32 (by strain YU10) from S. salivarius cells grown aerobically in this medium. In addition to bacteriocin production, S. salivarius strains produced levan-sucrase which was detected by a specific ESI-LC-MS/MS method which indicates additional health benefits from the developed strains. CONCLUSION The current study established the bacteriocin, levan-sucrase production and basic safety features of S. salivarius strains isolated from healthy Malaysian subjects demonstrating their potential for use as probiotics. A new bacteriocin-production medium was developed with potential scale up application for pharmaceuticals and probiotics from S. salivarius generating different lantibiotics. This is relevant for the clinical management of oral cavity and upper respiratory tract in the human population.
Collapse
Affiliation(s)
- Abdelahhad Barbour
- Institute of Biological Sciences, Microbiology Division, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Koshy Philip
- Institute of Biological Sciences, Microbiology Division, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Blin K, Kazempour D, Wohlleben W, Weber T. Improved lanthipeptide detection and prediction for antiSMASH. PLoS One 2014; 9:e89420. [PMID: 24586765 PMCID: PMC3930743 DOI: 10.1371/journal.pone.0089420] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 01/21/2014] [Indexed: 11/18/2022] Open
Abstract
Lanthipeptides are a class of ribosomally synthesised and post-translationally modified peptide (RiPP) natural products from the bacterial secondary metabolism. Their name is derived from the characteristic lanthionine or methyl-lanthionine residues contained in the processed peptide. Lanthipeptides that possess an antibacterial activity are called lantibiotics. Whereas multiple tools exist to identify lanthipeptide gene clusters from genomic data, no programs are available to predict the post-translational modifications of lanthipeptides, such as the proteolytic cleavage of the leader peptide part or tailoring modifications based on the analysis of the gene cluster sequence. antiSMASH is a software pipeline for the identification of secondary metabolite biosynthetic clusters from genomic input and the prediction of products produced by the identified clusters. Here we present a novel antiSMASH module using a rule-based approach to combine signature motifs for biosynthetic enzymes and lanthipeptide-specific cleavage site motifs to identify lanthipeptide clusters in genomic data, assign the specific lanthipeptide class, predict prepeptide cleavage, tailoring reactions, and the processed molecular weight of the mature peptide products.
Collapse
Affiliation(s)
- Kai Blin
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University of Tübingen, Tübingen, Germany
- German Centre for Infection Research, Tübingen, Germany
| | - Daniyal Kazempour
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University of Tübingen, Tübingen, Germany
| | - Wolfgang Wohlleben
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University of Tübingen, Tübingen, Germany
- German Centre for Infection Research, Tübingen, Germany
| | - Tilmann Weber
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University of Tübingen, Tübingen, Germany
- German Centre for Infection Research, Tübingen, Germany
- * E-mail:
| |
Collapse
|
37
|
Teng K, Zhang J, Zhang X, Ge X, Gao Y, Wang J, Lin Y, Zhong J. Identification of ligand specificity determinants in lantibiotic bovicin HJ50 and the receptor BovK, a multitransmembrane histidine kinase. J Biol Chem 2014; 289:9823-32. [PMID: 24526683 DOI: 10.1074/jbc.m113.513150] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lantibiotic bovicin HJ50 is produced by Streptococcus bovis HJ50 and acts as the extracellular signal to autoregulate its own biosynthesis through BovK/R two-component system. Bovicin HJ50 shows a linear N-terminal and glubolar C-terminal structure, and the sensor histidine kinase BovK contains eight transmembrane segments lacking any extensive surface-exposed sensory domain. The signal recognition mechanism between bovicin HJ50 and BovK is still unknown. We performed saturated alanine scanning mutagenesis and other amino acid substitutions on bovicin HJ50 using a semi-in vitro biosynthesis. Results of the mutants inducing activities indicated that several charged and hydrophobic amino acids in ring B of bovicin HJ50, as well as two glycines were key residues to recognize BovK. Circular dichroism analyses indicated that both glycines contributed to bovicin HJ50 structural changes in the membrane. Biotin-labeled bovicin HJ50 could interact with the N-terminal sensor of BovK, and several charged residues and a conserved hydrophobic region in the N-terminal portion of BovK sensor domain were important for interacting with the signal bovicin HJ50. By combining the results, we suggested a mechanism of bovicin HJ50 recognizing and activating BovK mainly through electrostatic and hydrophobic interactions.
Collapse
Affiliation(s)
- Kunling Teng
- From the State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences and
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Jakubovics NS, Yassin SA, Rickard AH. Community interactions of oral streptococci. ADVANCES IN APPLIED MICROBIOLOGY 2014; 87:43-110. [PMID: 24581389 DOI: 10.1016/b978-0-12-800261-2.00002-5] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
It is now clear that the most common oral diseases, dental caries and periodontitis, are caused by mixed-species communities rather than by individual pathogens working in isolation. Oral streptococci are central to these disease processes since they are frequently the first microorganisms to colonize oral surfaces and they are numerically the dominant microorganisms in the human mouth. Numerous interactions between oral streptococci and other bacteria have been documented. These are thought to be critical for the development of mixed-species oral microbial communities and for the transition from oral health to disease. Recent metagenomic studies are beginning to shed light on the co-occurrence patterns of streptococci with other oral bacteria. Refinements in microscopy techniques and biofilm models are providing detailed insights into the spatial distribution of streptococci in oral biofilms. Targeted genetic manipulation is increasingly being applied for the analysis of specific genes and networks that modulate interspecies interactions. From this work, it is clear that streptococci produce a range of extracellular factors that promote their integration into mixed-species communities and enable them to form social networks with neighboring taxa. These "community integration factors" include coaggregation-mediating adhesins and receptors, small signaling molecules such as peptides or autoinducer-2, bacteriocins, by-products of metabolism including hydrogen peroxide and lactic acid, and a range of extracellular enzymes. Here, we provide an overview of various types of community interactions between oral streptococci and other microorganisms, and we consider the possibilities for the development of new technologies to interfere with these interactions to help control oral biofilms.
Collapse
Affiliation(s)
- Nicholas S Jakubovics
- Oral Biology, School of Dental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | - Sufian A Yassin
- Oral Biology, School of Dental Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Alexander H Rickard
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
39
|
Isolation and identification of a new intracellular antimicrobial peptide produced by Paenibacillus alvei AN5. World J Microbiol Biotechnol 2013; 30:1377-85. [DOI: 10.1007/s11274-013-1558-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 11/11/2013] [Indexed: 10/26/2022]
|
40
|
Abstract
The sal lantibiotic locus plays an important role in the virulence of Streptococcus pyogenes. Our transcriptional analysis of the sal locus provides new information on the complex regulation of this operon. Transcription of the operon is regulated by a promoter upstream of the operon and by a second internal promoter upstream of the salKRZ genes. Here we identify the location of the internal promoter and provide information on how this promoter is autoregulated by proteins within the locus. We determined by primer extension that the salKR promoter is located within the salY gene and identified several regulatory regions important for expression. The higher activity of the promoter in a salKR deletion strain indicates a role in repression by the SalR response regulator. Further, this promoter had higher activity in a salA deletion strain, implicating corepression or a signaling role for the SalA peptide. Finally, we demonstrate that this promoter can be controlled by host factors. Analysis of transcriptional regulation of this locus provides a better understanding of the function of the sal locus in S. pyogenes pathogenesis.
Collapse
|
41
|
Barbour A, Philip K, Muniandy S. Enhanced production, purification, characterization and mechanism of action of salivaricin 9 lantibiotic produced by Streptococcus salivarius NU10. PLoS One 2013; 8:e77751. [PMID: 24147072 PMCID: PMC3797685 DOI: 10.1371/journal.pone.0077751] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 09/06/2013] [Indexed: 01/28/2023] Open
Abstract
Background Lantibiotics are small lanthionine-containing bacteriocins produced by lactic acid bacteria. Salivaricin 9 is a newly discovered lantibiotic produced by Streptococcus salivarius. In this study we present the mechanism of action of salivaricin 9 and some of its properties. Also we developed new methods to produce and purify the lantibiotic from strain NU10. Methodology / Principal Findings Salivaricin 9 was found to be auto-regulated when an induction assay was applied and this finding was used to develop a successful salivaricin 9 production system in liquid medium. A combination of XAD-16 and cation exchange chromatography was used to purify the secondary metabolite which was shown to have a molecular weight of approximately 3000 Da by SDS-PAGE. MALDI-TOF MS analysis indicated the presence of salivaricin 9, a 2560 Da lantibiotic. Salivaricin 9 is a bactericidal molecule targeting the cytoplasmic membrane of sensitive cells. The membrane permeabilization assay showed that salivaricin 9 penetrated the cytoplasmic membrane and induced pore formation which resulted in cell death. The morphological changes of test bacterial strains incubated with salivaricin 9 were visualized using Scanning Electron Microscopy which confirmed a pore forming mechanism of inhibition. Salivaricin 9 retained biological stability when exposed to high temperature (90-100°C) and stayed bioactive at pH ranging 2 to 10. When treated with proteinase K or peptidase, salivaricin 9 lost all antimicrobial activity, while it remained active when treated with lyticase, catalase and certain detergents. Conclusion The mechanism of antimicrobial action of a newly discovered lantibiotic salivaricin 9 was elucidated in this study. Salivaricin 9 penetrated the cytoplasmic membrane of its targeted cells and induced pore formation. This project has given new insights on lantibiotic peptides produced by S. salivarius isolated from the oral cavities of Malaysian subjects.
Collapse
Affiliation(s)
- Abdelahhad Barbour
- Institute of Biological Sciences, Microbiology Division, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | | | | |
Collapse
|
42
|
GET_HOMOLOGUES, a versatile software package for scalable and robust microbial pangenome analysis. Appl Environ Microbiol 2013; 79:7696-701. [PMID: 24096415 DOI: 10.1128/aem.02411-13] [Citation(s) in RCA: 615] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
GET_HOMOLOGUES is an open-source software package that builds on popular orthology-calling approaches making highly customizable and detailed pangenome analyses of microorganisms accessible to nonbioinformaticians. It can cluster homologous gene families using the bidirectional best-hit, COGtriangles, or OrthoMCL clustering algorithms. Clustering stringency can be adjusted by scanning the domain composition of proteins using the HMMER3 package, by imposing desired pairwise alignment coverage cutoffs, or by selecting only syntenic genes. The resulting homologous gene families can be made even more robust by computing consensus clusters from those generated by any combination of the clustering algorithms and filtering criteria. Auxiliary scripts make the construction, interrogation, and graphical display of core genome and pangenome sets easy to perform. Exponential and binomial mixture models can be fitted to the data to estimate theoretical core genome and pangenome sizes, and high-quality graphics can be generated. Furthermore, pangenome trees can be easily computed and basic comparative genomics performed to identify lineage-specific genes or gene family expansions. The software is designed to take advantage of modern multiprocessor personal computers as well as computer clusters to parallelize time-consuming tasks. To demonstrate some of these capabilities, we survey a set of 50 Streptococcus genomes annotated in the Orthologous Matrix (OMA) browser as a benchmark case. The package can be downloaded at http://www.eead.csic.es/compbio/soft/gethoms.php and http://maya.ccg.unam.mx/soft/gethoms.php.
Collapse
|
43
|
Wescombe PA, Hale JDF, Heng NCK, Tagg JR. Developing oral probiotics from Streptococcus salivarius. Future Microbiol 2013; 7:1355-71. [PMID: 23231486 DOI: 10.2217/fmb.12.113] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Considerable human illness can be linked to the development of oral microbiota disequilibria. The predominant oral cavity commensal, Streptococcus salivarius has emerged as an important source of safe and efficacious probiotics, capable of fostering more balanced, health-associated oral microbiota. Strain K12, the prototype S. salivarius probiotic, originally introduced to counter Streptococcus pyogenes infections, now has an expanded repertoire of health-promoting applications. K12 and several more recently proposed S. salivarius probiotics are now being applied to control diverse bacterial consortia infections including otitis media, halitosis and dental caries. Other potential applications include upregulation of immunological defenses against respiratory viral infections and treatment of oral candidosis. An overview of the key steps required for probiotic development is also presented.
Collapse
Affiliation(s)
- Philip A Wescombe
- BLIS Technologies Ltd. Centre for Innovation, PO Box 56, Dunedin, 9054, New Zealand
| | | | | | | |
Collapse
|
44
|
Macedovicin, the second food-grade lantibiotic produced by Streptococcus macedonicus ACA-DC 198. Food Microbiol 2013; 33:124-30. [DOI: 10.1016/j.fm.2012.09.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 07/14/2012] [Accepted: 09/19/2012] [Indexed: 11/20/2022]
|
45
|
Protective mechanisms of respiratory tract Streptococci against Streptococcus pyogenes biofilm formation and epithelial cell infection. Appl Environ Microbiol 2012; 79:1265-76. [PMID: 23241973 DOI: 10.1128/aem.03350-12] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Streptococcus pyogenes (group A streptococci [GAS]) encounter many streptococcal species of the physiological microbial biome when entering the upper respiratory tract of humans, leading to the question how GAS interact with these bacteria in order to establish themselves at this anatomic site and initiate infection. Here we show that S. oralis and S. salivarius in direct contact assays inhibit growth of GAS in a strain-specific manner and that S. salivarius, most likely via bacteriocin secretion, also exerts this effect in transwell experiments. Utilizing scanning electron microscopy documentation, we identified the tested strains as potent biofilm producers except for GAS M49. In mixed-species biofilms, S. salivarius dominated the GAS strains, while S. oralis acted as initial colonizer, building the bottom layer in mixed biofilms and thereby allowing even GAS M49 to form substantial biofilms on top. With the exception of S. oralis, artificial saliva reduced single-species biofilms and allowed GAS to dominate in mixed biofilms, although the overall two-layer structure was unchanged. When covered by S. oralis and S. salivarius biofilms, epithelial cells were protected from GAS adherence, internalization, and cytotoxic effects. Apparently, these species can have probiotic effects. The use of Affymetrix array technology to assess HEp-2 cell transcription levels revealed modest changes after exposure to S. oralis and S. salivarius biofilms which could explain some of the protective effects against GAS attack. In summary, our study revealed a protection effect of respiratory tract bacteria against an important airway pathogen and allowed a first in vitro insight into local environmental processes after GAS enter the respiratory tract.
Collapse
|
46
|
Lefébure T, Richards VP, Lang P, Pavinski-Bitar P, Stanhope MJ. Gene repertoire evolution of Streptococcus pyogenes inferred from phylogenomic analysis with Streptococcus canis and Streptococcus dysgalactiae. PLoS One 2012; 7:e37607. [PMID: 22666370 PMCID: PMC3364286 DOI: 10.1371/journal.pone.0037607] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/24/2012] [Indexed: 01/08/2023] Open
Abstract
Streptococcus pyogenes, is an important human pathogen classified within the pyogenic group of streptococci, exclusively adapted to the human host. Our goal was to employ a comparative evolutionary approach to better understand the genomic events concomitant with S. pyogenes human adaptation. As part of ascertaining these events, we sequenced the genome of one of the potential sister species, the agricultural pathogen S. canis, and combined it in a comparative genomics reconciliation analysis with two other closely related species, Streptococcus dysgalactiae and Streptococcus equi, to determine the genes that were gained and lost during S. pyogenes evolution. Genome wide phylogenetic analyses involving 15 Streptococcus species provided convincing support for a clade of S. equi, S. pyogenes, S. dysgalactiae, and S. canis and suggested that the most likely S. pyogenes sister species was S. dysgalactiae. The reconciliation analysis identified 113 genes that were gained on the lineage leading to S. pyogenes. Almost half (46%) of these gained genes were phage associated and 14 showed significant matches to experimentally verified bacteria virulence factors. Subsequent to the origin of S. pyogenes, over half of the phage associated genes were involved in 90 different LGT events, mostly involving different strains of S. pyogenes, but with a high proportion involving the horse specific pathogen S. equi subsp. equi, with the directionality almost exclusively (86%) in the S. pyogenes to S. equi direction. Streptococcus agalactiae appears to have played an important role in the evolution of S. pyogenes with a high proportion of LGTs originating from this species. Overall the analysis suggests that S. pyogenes adaptation to the human host was achieved in part by (i) the integration of new virulence factors (e.g. speB, and the sal locus) and (ii) the construction of new regulation networks (e.g. rgg, and to some extent speB).
Collapse
Affiliation(s)
| | | | | | | | - Michael J. Stanhope
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
47
|
In vitro functional and immunomodulatory properties of the Lactobacillus helveticus MIMLh5-Streptococcus salivarius ST3 association that are relevant to the development of a pharyngeal probiotic product. Appl Environ Microbiol 2012; 78:4209-16. [PMID: 22504812 DOI: 10.1128/aem.00325-12] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The use of proper bacterial strains as probiotics for the pharyngeal mucosa is a potential prophylactic strategy for upper respiratory tract infections. In this context, we characterized in vitro the functional and immunomodulatory properties of the strains Lactobacillus helveticus MIMLh5 and Streptococcus salivarius ST3 that were selected during previous investigations as promising pharyngeal probiotics. In this study, we demonstrated in vitro that strains MIMLh5 and ST3, alone and in combination, can efficiently adhere to pharyngeal epithelial cells, antagonize Streptococcus pyogenes, and modulate host innate immunity by inducing potentially protective effects. In particular, we found that the strains MIMLh5 and ST3 activate U937 human macrophages by significantly inducing the expression of the proinflammatory cytokine tumor necrosis factor alpha (TNF-α). Nonetheless, the induction of the anti-inflammatory interleukin-10 (IL-10) by MIMLh5 or ST3 was never lower than that of TNF-α, suggesting that these bacteria can potentially exert a regulatory rather than a proinflammatory effect. We also found that the strains MIMLh5 and ST3 induce cyclooxygenase 2 (COX-2) expression and demonstrated that toll-like receptor 2 (TLR-2) participates in the recognition of the strains MIMLh5 and ST3 by U937 cells. Finally, we observed that these microorganisms grow efficiently when cocultured in milk, suggesting that the preparation of a milk-based fermented product containing both MIMLh5 and ST3 can be a practical solution for the administration of these bacteria. In conclusion, we propose the combined use of L. helveticus MIMLh5 and S. salivarius ST3 for the preparation of novel products that display probiotic properties for the pharyngeal mucosa.
Collapse
|
48
|
Salivaricin G32, a Homolog of the Prototype Streptococcus pyogenes Nisin-Like Lantibiotic SA-FF22, Produced by the Commensal Species Streptococcus salivarius. Int J Microbiol 2012; 2012:738503. [PMID: 22567013 PMCID: PMC3332205 DOI: 10.1155/2012/738503] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 12/05/2011] [Indexed: 01/08/2023] Open
Abstract
Salivaricin G32, a 2667 Da novel member of the SA-FF22 cluster of lantibiotics, has been purified and characterized from Streptococcus salivarius strain G32. The inhibitory peptide differs from the Streptococcus pyogenes—produced SA-FF22 in the absence of lysine in position 2. The salivaricin G32 locus was widely distributed in BLIS-producing S. salivarius, with 6 (23%) of 26 strains PCR-positive for the structural gene, slnA. As for most other lantibiotics produced by S. salivarius, the salivaricin G32 locus can be megaplasmid encoded. Another member of the SA-FF22 family was detected in two Streptococcus dysgalactiae of bovine origin, an observation supportive of widespread distribution of this lantibiotic within the genus Streptococcus. Since the inhibitory spectrum of salivaricin G32 includes Streptococcus pyogenes, its production by S. salivarius, either as a member of the normal oral microflora or as a commercial probiotic, could serve to enhance protection of the human host against S. pyogenes infection.
Collapse
|
49
|
Abstract
Aided by genome-mining strategies, knowledge of the prevalence and diversity of ribosomally synthesized natural products (RNPs) is rapidly increasing. Among these are the lantipeptides, posttranslationally modified peptides containing characteristic thioether cross-links imperative for bioactivity and stability. Though this family was once thought to be a limited class of antimicrobial compounds produced by gram-positive bacteria, new insights have revealed a much larger diversity of activity, structure, biosynthetic machinery, and producing organisms than previously appreciated. Detailed investigation of the enzymes responsible for installing the posttranslational modifications has resulted in improved in vivo and in vitro engineering systems focusing on enhancement of the therapeutic potential of these compounds. Although dozens of new lantipeptides have been isolated in recent years, bioinformatic analyses indicate that many hundreds more await discovery owing to the widespread frequency of lantipeptide biosynthetic machinery in bacterial genomes.
Collapse
Affiliation(s)
- Patrick J Knerr
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA.
| | | |
Collapse
|
50
|
Genome sequence of the bacteriocin-producing oral probiotic Streptococcus salivarius strain M18. J Bacteriol 2011; 193:6402-3. [PMID: 22038965 DOI: 10.1128/jb.06001-11] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus salivarius is a Gram-positive bacterial commensal and pioneer colonizer of the human oral cavity. Many strains produce ribosomally synthesized proteinaceous antibiotics (bacteriocins), and some strains have been developed for use as oral probiotics. Here, we present the draft genome sequence of the bacteriocin-producing oral probiotic S. salivarius strain M18.
Collapse
|