1
|
Lormand JD, Savelle CH, Teschler JK, López E, Little RH, Malone JG, Yildiz FH, García-García MJ, Sondermann H. Secreted retropepsin-like enzymes are essential for stress tolerance and biofilm formation in Pseudomonas aeruginosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643919. [PMID: 40166241 PMCID: PMC11957051 DOI: 10.1101/2025.03.18.643919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Proteases regulate important biological functions. Here we present the structural and functional characterization of three previously uncharacterized aspartic proteases in Pseudomonas aeruginosa. We show that these proteases have structural hallmarks of retropepsin peptidases and play redundant roles for cell survival under hypoosmotic stress conditions. Consequently, we named them retropepsin-like osmotic stress tolerance peptidases (Rlo). Our research shows that while Rlo proteases are homologous to RimB, an aspartic peptidase involved in rhizosphere colonization and plant infection, they contain N-terminal signal peptides and perform distinct biological functions. Mutants lacking all three secreted Rlo peptidases show defects in antibiotic resistance, biofilm formation, and cell morphology. These defects are rescued by mutations in the inactive transglutaminase transmembrane protein RloB and the cytoplasmic ATP-grasp protein RloC, two previously uncharacterized genes in the same operon as one of the Rlo proteases. These studies identify Rlo proteases and rlo operon products as critical factors in clinically relevant processes, making them appealing targets for therapeutic strategies against Pseudomonas infections.
Collapse
Affiliation(s)
- Justin D. Lormand
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Charles H. Savelle
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Jennifer K. Teschler
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California, USA
| | - Eva López
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Richard H. Little
- Department of Molecular Microbiology, John Innes Centre, Norwich, UK
| | - Jacob G. Malone
- Department of Molecular Microbiology, John Innes Centre, Norwich, UK
- School of Biological Sciences, University of East Anglia, Norwich, UK
| | - Fitnat H. Yildiz
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California, USA
| | - María J. García-García
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
| | - Holger Sondermann
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Hamburg, Germany
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
- Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
2
|
Yang H, Dong P, Huo S, Nychas GJE, Luo X, Zhu L, Mao Y, Han G, Liu M, Liu Y, Zhang Y. Deciphering the inhibitory mechanisms of cinnamaldehyde on biofilm formation of Listeria monocytogenes and implement these strategies to control its transfer to beef surfaces. Food Res Int 2025; 204:115946. [PMID: 39986790 DOI: 10.1016/j.foodres.2025.115946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 02/05/2025] [Indexed: 02/24/2025]
Abstract
Natural essential oils have received widespread attention as promising microbial inhibitors, whereas a comprehensive understanding of their mechanisms underlying biofilm control and impact on biofilm cross-contamination on meat remains poorly understood. In this study, Listeria monocytogenes (Lm) biofilms were treated with sub-inhibitory concentrations of cinnamaldehyde (CA) and characterized over a 4-day period. Both 1/2 MIC (160 μg/mL) and 1/4 MIC (80 μg/mL) CA delayed the development of Lm biofilm on abiotic surfaces and reduced the maximum biofilm formation. The limited effect of 1/4 MIC CA on the flagellar-mediated motility of Lm during initial adhesion indicated that hindering bacterial motility was not the main reason for CA inhibition of biofilm formation. Transcriptomics results showed that CA was involved in inhibitory pathways dominated by energy metabolism and peptidoglycan synthesis during the initial adhesion period and the maturation period of the biofilm, respectively. This posed an obstacle to the polymers required for biofilm cell adhesion and the energy consumption required for their production. Down-regulation of genes associated with multiple signalling systems and virulence factors also suggested that CA further mitigated resistance and virulence in residual biofilm cells. In addition, quantification of biofilm cells transferred to beef surfaces confirmed that CA significantly reduces the biomass transferred and the risk of persistent biofilm contamination. This study provided the theoretical basis for the control of Lm biofilm and its cross-contamination in the food industry by natural essential oils.
Collapse
Affiliation(s)
- Huixuan Yang
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Pengcheng Dong
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China
| | - Shengnan Huo
- Shandong Institute for Food and Drug Control, Jinan 250101, China; Key Laboratory of Supervising Technology for Meat and Meat Products, State Administration for Market Regulation, Jinan 250101, China
| | - George-John E Nychas
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China; Agricultural University of Athens, Iera Odos 75, Athens 11855, Greece
| | - Xin Luo
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China
| | - Lixian Zhu
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China
| | - Yanwei Mao
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China
| | - Guangxing Han
- Lilnyi Station of China Agriculture Research System (beef), Linyi, Shandong 276000, China
| | - Minze Liu
- Yangxin Yiliyuan Halal Meat Co., Ltd., Binzhou 251800, China
| | - Yunge Liu
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China; Yangxin Yiliyuan Halal Meat Co., Ltd., Binzhou 251800, China.
| | - Yimin Zhang
- Lab of Beef Processing and Quality Control, College of Food Science and Engineering, Shandong Agricultural University, Tai'an, Shandong 271018, China; International Joint Research Lab (China and Greece) of Digital Transformation as an Enabler for Food Safety and Sustainability, Tai'an, Shandong 271018, China.
| |
Collapse
|
3
|
Sangha JS, Gogulancea V, Curtis TP, Jakubovics NS, Barrett P, Metris A, Ofiţeru ID. Advancing dental biofilm models: the integral role of pH in predicting S. mutans colonization. mSphere 2025; 10:e0074324. [PMID: 39660862 PMCID: PMC11774048 DOI: 10.1128/msphere.00743-24] [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: 09/03/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
Mathematical models can provide insights into complex interactions and dynamics within microbial communities to complement and extend experimental laboratory approaches. For dental biofilms, they can give a basis for evaluating biofilm growth or the transition from health to disease. We have developed mathematical models to simulate the transition toward a cariogenic microbial biofilm, modeled as the overgrowth of Streptococcus mutans within a five-species dental community. This work builds on experimental data from a continuous flow reactor with hydroxyapatite coupons for biofilm growth, in a chemically defined medium with varying concentrations of glucose and lactic acid. The biofilms formed on the coupons were simulated using individual-based models (IbMs), with bacterial growth modeled using experimentally measured kinetic parameters. The IbM assumes that the maximum theoretical growth yield for biomass is dependent on the local concentration of reactants and products, while the growth rates were described using traditional Monod equations. We have simulated all the conditions studied experimentally, considering different initial relative abundance of the five species, and also different initial clustering in the biofilm. The simulation results only reproduced the experimental dominance of S. mutans at high glucose concentration after we considered the species-specific effect of pH on growth rates. This highlights the significance of the aciduric property of S. mutans in the development of caries. Our study demonstrates the potential of combining in vitro and in silico studies to gain a new understanding of the factors that influence dental biofilm dynamics.IMPORTANCEWe have developed in silico models able to reproduce the relative abundance measured in vitro in the synthetic dental biofilm communities growing in a chemically defined medium. The advantage of this combination of in vitro and in silico models is that we can study the influence of one parameter at a time and aim for direct validation. Our work demonstrates the utility of individual-based models for simulating diverse conditions affecting dental biofilm scenarios, such as the frequency of glucose intake, sucrose pulsing, or integration of pathogenic or probiotic species. Although in silico models are reductionist approaches, they have the advantage of not being limited in the scenarios they can test by the ethical consideration of an in vivo system, thus significantly contributing to dental biofilm research.
Collapse
Affiliation(s)
- Jay S. Sangha
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Valentina Gogulancea
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Thomas P. Curtis
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nicholas S. Jakubovics
- Faculty of Medical Sciences, Newcastle University, School of Dental Sciences, Newcastle upon Tyne, United Kingdom
| | - Paul Barrett
- Safety and Environmental Assurance Centre, Unilever, Bedfordshire, United Kingdom
| | - Aline Metris
- Safety and Environmental Assurance Centre, Unilever, Bedfordshire, United Kingdom
| | - Irina D. Ofiţeru
- School of Engineering, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Afzal M, Carda-Diéguez M, Bloch S, Thies LGS, Mira A, Schäffer C. Decoding gene expression dynamics in planktonic and biofilm cells of Streptococcus mutans: regulation and role of mutanofactin genes in biofilm formation. FRONTIERS IN ORAL HEALTH 2025; 6:1535034. [PMID: 39896144 PMCID: PMC11782227 DOI: 10.3389/froh.2025.1535034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Introduction Dental caries is the most prevalent chronic infectious disease globally, with Streptococcus mutans recognized as a primary causative agent due to its acidogenicity and robust biofilm-forming ability. In S. mutans biofilm formation, the role of autoinducers has been extensively studied, while the influence of other small molecules remains largely unexplored. Mutanofactins, a class of polyketide/non-ribosomal lipopeptide secondary metabolites, are emerging as potential modulators of S. mutans biofilm development. Methods Transcriptomic analysis was conducted to examine gene expression patterns in S. mutans NMT4863 across distinct growth phases and lifestyles, aiming to identify metabolic factors influencing biofilm formation. Transcriptomic profiles were compared between cells in early-, mid-, and late-exponential-, and stationary phase, as well as between planktonic and biofilm cells. Differentially expressed genes were identified, and pathway analyses revealed significant alterations in key metabolic and regulatory pathways. Specifically, the biosynthetic mutanofactin gene cluster was analyzed via quantitative real-time polymerase chain reaction. Results Several genes and operons were differentially expressed across the tested growth phases, with 1,095 genes showing differential expression between stationary-phase, planktonic and biofilm cells. Pathway analysis revealed significant changes in ascorbate metabolism, carbohydrate utilization and transport systems, lipoic acid metabolism, bacterial toxin pathways, two-component regulatory systems, and secondary metabolite biosynthesis. Notably, expression of the muf gene cluster, was elevated in early exponential-phase cells relative to stationary-phase cells. Additionally, the mufCDEFGHIJ genes were identified as components of a single transcriptional unit (muf operon). MufC, a transcriptional regulator of the TetR/AcrR-family, acts as a positive regulator of the muf operon in strain NMT4863. Bioinformatic analysis pinpointed a 20-bp regulatory sequence in the muf operon promoter region (5'-AAATGAGCTATAATTCATTT-3'). Interestingly, the muf operon was found to be significantly downregulated in biofilm cells. Conclusion This study provides key insights into gene expression dynamics that drive biofilm formation in S. mutans NMT4863, with a particular emphasis on the role of the muf operon. This operon is governed by the TetR/AcrR-family regulator MufC and plays a central role in biofilm development, offering a novel perspective on the molecular basis of S. mutans biofilm formation and resilience.
Collapse
Affiliation(s)
- Muhammad Afzal
- Department of Natural Sciences and Sustainable Resources, Institue of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| | | | - Susanne Bloch
- Department of Natural Sciences and Sustainable Resources, Institue of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Leon G. S. Thies
- Department of Natural Sciences and Sustainable Resources, Institue of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| | - Alex Mira
- Department of Genomics and Health, FISABIO Foundation, Valencia, Spain
| | - Christina Schäffer
- Department of Natural Sciences and Sustainable Resources, Institue of Biochemistry, NanoGlycobiology Research Group, Universität für Bodenkultur Wien, Vienna, Austria
| |
Collapse
|
5
|
Zhu M, Hao C, Zou T, Jiang S, Wu B. Phage therapy as an alternative strategy for oral bacterial infections: a systematic review. BMC Oral Health 2025; 25:44. [PMID: 39780179 PMCID: PMC11715224 DOI: 10.1186/s12903-024-05399-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Oral infectious diseases, such as dental caries, periodontitis and periapical periodontitis, are often complicated by causative bacterial biofilm formation and significantly impact human oral health and quality of life. Bacteriophage (phage) therapy has emerged as a potential alternative with successful applications in antimicrobial trials. While therapeutic use of phages has been considered as effective treatment of some infectious diseases, related research focusing on oral infectious diseases is few and lacks attention. Therefore, a systematic review was conducted to comprehensively evaluate the overall efficacy of phages in reducing bacterial infections associated with various oral diseases. METHODS A systematic search of PubMed, MEDLINE and Web of Science for literature published up to May 2024 was conducted according to inclusion criteria to identify studies assessing bacteriophages as potential therapy for oral infectious diseases. A total of four authors assessed study eligibility and performed data extraction. RESULTS A total of 487 articles published between 1975 and 2024 were retrieved. Among the 10 eligible reports, preliminary studies have been conducted on seven types of phages and reported their antibacterial effect. To be more specific, 3 contained data on dental caries (n = 32), 5 focused on periodontitis (n = 105) and 2 examined periapical diseases (n = 7). The majority of publications (9 out of 10) discussed the impact of phages on biofilm formation. Only one report (1 out of 10) mentioned the safety concern in phage application. CONCLUSIONS This review strongly suggests that phages isolated from oral cavity with certain characteristics can be highly effective and are considered suitable candidates for phage therapy in treating oral/odontogenic infections caused by bacteria.
Collapse
Affiliation(s)
- Mingqi Zhu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- School of Stomatology, Southern Medical University, Guangzhou, 510515, China
| | - Chunxiu Hao
- Shenzhen Clinical College of Stomatology, Southern Medical University, Shenzhen, 518118, China
- Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Shenzhen, 518118, China
| | - Ting Zou
- Shenzhen Clinical College of Stomatology, Southern Medical University, Shenzhen, 518118, China
- Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Shenzhen, 518118, China
| | - Shan Jiang
- Shenzhen Clinical College of Stomatology, Southern Medical University, Shenzhen, 518118, China.
- Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Shenzhen, 518118, China.
| | - Buling Wu
- Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Shenzhen Clinical College of Stomatology, Southern Medical University, Shenzhen, 518118, China.
- Shenzhen Stomatology Hospital (Pingshan) of Southern Medical University, Shenzhen, 518118, China.
| |
Collapse
|
6
|
Jaroch M, Savage K, Kuipers P, Bacusmo JM, Hu J, Sun J, Dedon PC, Rice KC, de Crécy-Lagard V. Environmental Control of Queuosine Levels in Streptococcus mutans tRNAs. Mol Microbiol 2025; 123:48-59. [PMID: 39719891 PMCID: PMC11724357 DOI: 10.1111/mmi.15336] [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: 08/19/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024]
Abstract
Queuosine (Q) is a modification of the wobble base in tRNAs that decode NA(C/U) codons. It is ubiquitous in bacteria, including many pathogens. Streptococcus mutans is an early colonizer of dental plaque biofilm and a key player in dental caries. Using a combination of genetic and physiological approaches, the predicted Q synthesis and salvage pathways were validated in this organism. These experiments confirmed that S. mutans can synthesize Q de novo through similar pathways found in Bacillus subtilis and Escherichia coli. However, S. mutans has a distinct salvage pathway compared to these model organisms, as it uses a transporter belonging to the energy coupling factor (ECF) family controlled by a preQ1-dependent riboswitch. Furthermore, Q levels in this oral pathogen depended heavily on the media composition, suggesting that micronutrients can affect Q-mediated translation efficiency.
Collapse
Affiliation(s)
- Marshall Jaroch
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
| | - Kathryn Savage
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
| | - Paul Kuipers
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
| | - Jo Marie Bacusmo
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
| | - Jennifer Hu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jingjing Sun
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Peter C. Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Kelly C. Rice
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
| | - Valérie de Crécy-Lagard
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL 32611
- Genetics Institute, University of Florida, Gainesville, FL 32611
| |
Collapse
|
7
|
Ahirwar P, Kozlovskaya V, Pukkanasut P, Nikishau P, Nealy S, Harber G, Michalek SM, Antony L, Wu H, Kharlampieva E, Velu SE. Polymer vesicles for the delivery of inhibitors of cariogenic biofilm. Dent Mater 2024; 40:1937-1953. [PMID: 39317560 PMCID: PMC11580801 DOI: 10.1016/j.dental.2024.09.006] [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: 06/14/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVES The goal of this study is to develop a novel drug delivery platform for the pH-responsive delivery of biofilm inhibitors as a potential avenue to prevent and treat dental caries. METHODS Biofilm and growth inhibition assays were performed in polystyrene microtiter 96-well plates. Docking analysis was performed using the reported GtfB + HA5 co-crystal structure (PDB code: 8fg8) in SeeSAR 13.0.1 software. Polymersome vesicles were assembled from poly(N-vinylpyrrolidone)8-block-poly(dimethylsiloxane)64-block-poly(N-vinylpyrrolidone)8 (PVPON8-PDMS64-PVPON8) triblock copolymer using a nanoprecipitation method. Microbiome analysis of biofilm inhibitors and the in vivo drug release and antivirulence activities of polymersome encapsulated inhibitors have been carried out in a S. mutans induced rat caries model. RESULTS Biofilm inhibitors for HA5 and HA6 have shown species-specific selectivity towards S. mutans and the ability to preserve the oral microbiome in a S. mutans induced dental caries model. The inhibitors were encapsulated into pH-responsive block copolymer vesicles to generate polymersome-encapsulated biofilm inhibitors, and their biofilm and growth inhibitory activities against S. mutans and representative strains of oral commensal streptococci have been assessed. A 4-week treatment of S. mutans UA159 infected gnotobiotic rats with 100 µM of polymersome-encapsulated biofilm inhibitor, PEHA5 showed significant reductions in buccal, sulcal, and proximal caries scores compared to an untreated control group. SIGNIFICANCE Taken together, our data suggests that the biofilm-selective therapy using the polymersome-encapsulated biofilm inhibitors is a viable approach for the prevention and treatment of dental caries while preserving the oral microbiome.
Collapse
Affiliation(s)
- Parmanand Ahirwar
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Piyasuda Pukkanasut
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pavel Nikishau
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sarah Nealy
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gregory Harber
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Suzanne M Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Linto Antony
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hui Wu
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center of Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Sadanandan E Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Microbiome Center, Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Global Center for Craniofacial Oral and Dental Disorders, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
8
|
Fang H, Guo C, Mei X, Hao M, Zhang J, Luo L, Liu H, Liu Y, Huang H, He X, Zhu Y, Yang M, Zhu S. Light stress elicits soilborne disease suppression mediated by root-secreted flavonoids in Panax notoginseng. HORTICULTURE RESEARCH 2024; 11:uhae213. [PMID: 39386000 PMCID: PMC11462611 DOI: 10.1093/hr/uhae213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/21/2024] [Indexed: 10/12/2024]
Abstract
Developing disease-suppressive soils is an effective approach for managing soilborne diseases, which can be achieved through crop metabolism and root secretion modification to recruit beneficial soil microbiota. Many factors, such as light, can elicit and modify plant metabolomic activities, resulting in disease suppression. To investigate the impact of light, Panax notoginseng was planted in a greenhouse and forest, conditioned with three levels of light intensities, including the optimal (15% light transmittance of full light), suboptimal low (5% light transmittance of full light) and suboptimal high (30% light transmittance of full light) intensities. We assessed the rhizosphere microbiota of P. notoginseng and root rot disease caused by soilborne pathogen Ilyonectria destructans, and elucidated the mechanism. Results showed that suboptimal light conditions alleviated root rot disease of P. notoginseng by enriching beneficial microbiota in the rhizosphere. Both low and high light stresses enhanced the secondary metabolism profile in favor of plant defense, particularly the flavonoid pathway. Notably, high light stress demonstrated a robust ability to promote flavonoid metabolism and secretion, resulting in the enrichment of more beneficial microorganisms that suppressed the soilborne pathogen I. destructans. These findings highlight the potential for adjusting canopy light intensities to improve soil health and promote sustainable agriculture.
Collapse
Affiliation(s)
- Haiyan Fang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Cunwu Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Xinyue Mei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Minwen Hao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Yuanjiang County Tobacco Monopoly Bureau, Yuxi, 653399, China
| | - Jiayin Zhang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Lifen Luo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Haijiao Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Yixiang Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | - Huichuan Huang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | - Xiahong He
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | - Youyong Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | - Min Yang
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | - Shusheng Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory for Agro-Biodiversity and Pest Control of Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| |
Collapse
|
9
|
Naumova OY, Dobrynin PV, Khafizova GV, Grigorenko EL. The Association of the Oral Microbiota with Cognitive Functioning in Adolescence. Genes (Basel) 2024; 15:1263. [PMID: 39457387 PMCID: PMC11507344 DOI: 10.3390/genes15101263] [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: 08/30/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
Background: A growing body of research supports the role of the microbial communities residing in the digestive system in the host's cognitive functioning. Most of these studies have been focused on the gut microbiome and its association with clinical phenotypes in middle-aged and older adults. There is an insufficiency of population-based research exploring the association of normative cognitive functioning with the microbiome particularly with the oral microbiota. Methods: In this study, using metagenomics and metabolomics, we characterized the salivary microbiome diversity in a sample of 51 males of Hispanic and African American origin aged 12-18 years and explored the associations between the microbiome and the youths' cognitive performance captured with the Kaufman Assessment Battery for Children II (KABC-II). Results: Several bacterial species of the oral microbiota and related metabolic pathways were associated with cognitive function. In particular, we found negative associations between indicators of general intelligence and the relative abundance of Bacteroidetes and Lachnospiraceae and positive associations with Bifidobacteriaceae and Prevotella histicola sp. Among metabolic pathways, the super pathways related to bacterial cell division and GABA metabolism were linked to cognitive function. Conclusions: The results of our work are consistent with the literature reporting on the association between microbiota and cognitive function and support further population work to elucidate the potential for a healthy oral microbiome to improve cognitive health.
Collapse
Affiliation(s)
- Oxana Y. Naumova
- Department of Psychology, University of Houston, Houston, TX 77204, USA; (O.Y.N.); (P.V.D.); (G.V.K.)
- Vavilov Institute of General Genetics RAS, Moscow 119991, Russia
| | - Pavel V. Dobrynin
- Department of Psychology, University of Houston, Houston, TX 77204, USA; (O.Y.N.); (P.V.D.); (G.V.K.)
| | - Galina V. Khafizova
- Department of Psychology, University of Houston, Houston, TX 77204, USA; (O.Y.N.); (P.V.D.); (G.V.K.)
| | - Elena L. Grigorenko
- Department of Psychology, University of Houston, Houston, TX 77204, USA; (O.Y.N.); (P.V.D.); (G.V.K.)
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Taylor ZA, Chen P, Noeparvar P, Pham DN, Walker AR, Kitten T, Zeng L. Glycerol metabolism contributes to competition by oral streptococci through production of hydrogen peroxide. J Bacteriol 2024; 206:e0022724. [PMID: 39171915 PMCID: PMC11411925 DOI: 10.1128/jb.00227-24] [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: 06/10/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
As a biological byproduct from both humans and microbes, glycerol's contribution to microbial homeostasis in the oral cavity remains understudied. In this study, we examined glycerol metabolism by Streptococcus sanguinis, a commensal associated with oral health. Genetic mutants of glucose-PTS enzyme II (manL), glycerol metabolism (glp and dha pathways), and transcriptional regulators were characterized with regard to glycerol catabolism, growth, production of hydrogen peroxide (H2O2), transcription, and competition with Streptococcus mutans. Biochemical assays identified the glp pathway as a novel source for H2O2 production by S. sanguinis that is independent of pyruvate oxidase (SpxB). Genetic analysis indicated that the glp pathway requires glycerol and a transcriptional regulator, GlpR, for expression and is negatively regulated by PTS, but not the catabolite control protein, CcpA. Conversely, deletion of either manL or ccpA increased the expression of spxB and a second, H2O2-non-producing glycerol metabolic pathway (dha), indicative of a mode of regulation consistent with conventional carbon catabolite repression (CCR). In a plate-based antagonism assay and competition assays performed with planktonic and biofilm-grown cells, glycerol greatly benefited the competitive fitness of S. sanguinis against S. mutans. The glp pathway appears to be conserved in several commensal streptococci and actively expressed in caries-free plaque samples. Our study suggests that glycerol metabolism plays a more significant role in the ecology of the oral cavity than previously understood. Commensal streptococci, though not able to use glycerol as a sole carbohydrate source for growth, benefit from the catabolism of glycerol through production of both ATP and H2O2. IMPORTANCE Glycerol is an abundant carbohydrate in the oral cavity. However, little is understood regarding the metabolism of glycerol by commensal streptococci, some of the most abundant oral bacteria. This was in part because most streptococci cannot grow on glycerol as the sole carbon source. In this study, we show that Streptococcus sanguinis, a commensal associated with dental health, can degrade glycerol for persistence and competition through two pathways, one of which generates hydrogen peroxide at levels capable of inhibiting Streptococcus mutans. Preliminary studies suggest that several additional commensal streptococci are also able to catabolize glycerol, and glycerol-related genes are actively expressed in human dental plaque samples. Our findings reveal the potential of glycerol to significantly impact microbial homeostasis, which warrants further exploration.
Collapse
Affiliation(s)
- Zachary A. Taylor
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Ping Chen
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Payam Noeparvar
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Danniel N. Pham
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Alejandro R. Walker
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Todd Kitten
- Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| | - Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| |
Collapse
|
11
|
Rice KC, Davis KAT. Brief Communication: Confocal microscopy of oral streptococcal biofilms grown in simulated microgravity using a random positioning machine. NPJ Microgravity 2024; 10:89. [PMID: 39251626 PMCID: PMC11385976 DOI: 10.1038/s41526-024-00427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Biofilms are a concern for spaceflight missions, given their propensity for biofouling systems and their potential threat to astronaut health. Herein, we describe a random positioning machine-based method for growing fluorescent protein-expressing streptococcal biofilms under simulated microgravity. Biofilms can be subsequently imaged by confocal microscopy without further manipulation, minimizing disruption of architecture. This methodology could be adaptable to other bacteria, potentially standardizing biofilm growth and study under simulated microgravity.
Collapse
Affiliation(s)
- Kelly C Rice
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL, 32611, USA.
| | - Ke Aira T Davis
- Department of Microbiology and Cell Science, IFAS, University of Florida, Gainesville, FL, 32611, USA
- Health Sciences (Biotechnology), Central Georgia Technical College, Macon, GA, 31206, USA
| |
Collapse
|
12
|
Taylor ZA, Chen P, Noeparvar P, Pham DN, Walker AR, Kitten T, Zeng L. Glycerol Metabolism Contributes to Competition by Oral Streptococci through Production of Hydrogen Peroxide. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.598274. [PMID: 38979179 PMCID: PMC11230354 DOI: 10.1101/2024.06.28.598274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
As a biological byproduct from both humans and microbes, glycerol's contribution to microbial homeostasis in the oral cavity remains understudied. Here we examined glycerol metabolism by Streptococcus sanguinis, a commensal associated with oral health. Genetic mutants of glucose-PTS enzyme II ( manL ), glycerol metabolism ( glp and dha pathways), and transcriptional regulators were characterized with regard to glycerol catabolism, growth, production of hydrogen peroxide (H 2 O 2 ), transcription, and competition with Streptococcus mutans . Biochemical assays identified the glp pathway as a novel source of H 2 O 2 production by S. sanguinis that is independent of pyruvate oxidase (SpxB). Genetic analysis indicated that the glp pathway requires glycerol and a transcriptional regulator, GlpR, for expression and is negatively regulated by PTS, but not the catabolite control protein, CcpA. Conversely, deletion of either manL or ccpA increased expression of spxB and a second, H 2 O 2 -non-producing glycerol metabolic pathway ( dha ), indicative of a mode of regulation consistent with conventional carbon catabolite repression (CCR). In a plate-based antagonism assay and competition assays performed with planktonic and biofilm-grown cells, glycerol greatly benefited the competitive fitness of S. sanguinis against S. mutans. The glp pathway appears to be conserved in several commensal streptococci and actively expressed in caries-free plaque samples. Our study suggests that glycerol metabolism plays a more significant role in the ecology of the oral cavity than previously understood. Commensal streptococci, though not able to use glycerol as a sole carbohydrate for growth, benefit from catabolism of glycerol through production of both ATP and H 2 O 2 . Importance Glycerol is an abundant carbohydrate found in oral cavity, both due to biological activities of humans and microbes, and as a common ingredient of foods and health care products. However, very little is understood regarding the metabolism of glycerol by some of the most abundant oral bacteria, commensal streptococci. This was in part because most streptococci cannot grow on glycerol as the sole carbon source. Here we show that Streptococcus sanguinis , an oral commensal associated with dental health, can degrade glycerol for persistence and competition through two independent pathways, one of which generates hydrogen peroxide at levels capable of inhibiting a dental pathobiont, Streptococcus mutans . Preliminary studies suggest that several other commensal streptococci are also able to catabolize glycerol, and glycerol-related genes are being actively expressed in human dental plaque samples. Our findings reveal the potential of glycerol to significantly impact microbial homeostasis which warrants further exploration.
Collapse
|
13
|
Chen YYM, Yang YC, Shieh HR, Lin YJ, Ke WJ, Chiu CH. Functional Analysis of the Major Pilin Proteins of Type IV Pili in Streptococcus sanguinis CGMH010. Int J Mol Sci 2024; 25:5402. [PMID: 38791440 PMCID: PMC11121087 DOI: 10.3390/ijms25105402] [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: 03/29/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
The pil gene cluster for Type IV pilus (Tfp) biosynthesis is commonly present and highly conserved in Streptococcus sanguinis. Nevertheless, Tfp-mediated twitching motility is less common among strains, and the factors determining twitching activity are not fully understood. Here, we analyzed the functions of three major pilin proteins (PilA1, PilA2, and PilA3) in the assembly and activity of Tfp in motile S. sanguinis CGMH010. Using various recombinant pilA deletion strains, we found that Tfp composed of different PilA proteins varied morphologically and functionally. Among the three PilA proteins, PilA1 was most critical in the assembly of twitching-active Tfp, and recombinant strains expressing motility generated more structured biofilms under constant shearing forces compared to the non-motile recombinant strains. Although PilA1 and PilA3 shared 94% identity, PilA3 could not compensate for the loss of PilA1, suggesting that the nature of PilA proteins plays an essential role in twitching activity. The single deletion of individual pilA genes had little effect on the invasion of host endothelia by S. sanguinis CGMH010. In contrast, the deletion of all three pilA genes or pilT, encoding the retraction ATPase, abolished Tfp-mediated invasion. Tfp- and PilT-dependent invasion were also detected in the non-motile S. sanguinis SK36, and thus, the retraction of Tfp, but not active twitching, was found to be essential for invasion.
Collapse
Affiliation(s)
- Yi-Ywan M. Chen
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (H.-R.S.); (Y.-J.L.); (W.-J.K.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan;
| | - Yuan-Chen Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Hui-Ru Shieh
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (H.-R.S.); (Y.-J.L.); (W.-J.K.)
| | - Yu-Juan Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (H.-R.S.); (Y.-J.L.); (W.-J.K.)
| | - Wan-Ju Ke
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (H.-R.S.); (Y.-J.L.); (W.-J.K.)
| | - Cheng-Hsun Chiu
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan;
| |
Collapse
|
14
|
Dai J, Fischer NG, Rahimi JR, Wang H, Hu C, Chen W, Lin Y, Sang T, Chew HP, Kong L, Aparicio C, Ye Z, Huang S. Interpenetrating nanofibrillar membrane of self-assembled collagen and antimicrobial peptides for enhanced bone regeneration. Int J Biol Macromol 2024; 267:131480. [PMID: 38599427 DOI: 10.1016/j.ijbiomac.2024.131480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/27/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
Bone regeneration remains a major clinical challenge, especially when infection necessitates prolonged antibiotic treatment. This study presents a membrane composed of self-assembled and interpenetrating GL13K, an antimicrobial peptide (AMP) derived from a salivary protein, in a collagen membrane for antimicrobial activity and enhanced bone regeneration. Commercially available collagen membranes were immersed in GL13K solution, and self-assembly was initiated by raising the solution pH to synthesize the multifunctional membrane called COL-GL. COL-GL was composed of interpenetrating large collagen fibers and short GL13K nanofibrils, which increased hydrophobicity, reduced biodegradation from collagenase, and stiffened the matrix compared to control collagen membranes. Incorporation of GL13K led to antimicrobial and anti-fouling activity against early oral surface colonizer Streptococcus gordonii while not affecting fibroblast cytocompatibility or pre-osteoblast osteogenic differentiation. GL13K in solution also reduced macrophage inflammatory cytokine expression and increased pro-healing cytokine expression. Bone formation in a rat calvarial model was accelerated at eight weeks with COL-GL compared to the gold-standard collagen membrane based on microcomputed tomography and histology. Interpenetration of GL13K within collagen sidesteps challenges with antimicrobial coatings on bone regeneration scaffolds while increasing bone regeneration. This strength makes COL-GL a promising approach to reduce post-surgical infections and aid bone regeneration in dental and orthopedic applications. STATEMENT OF SIGNIFICANCE: The COL-GL membrane, incorporating the antimicrobial peptide GL13K within a collagen membrane, signifies a noteworthy breakthrough in bone regeneration strategies for dental and orthopedic applications. By integrating self-assembled GL13K nanofibers into the membrane, this study successfully addresses the challenges associated with antimicrobial coatings, exhibiting improved antimicrobial and anti-fouling activity while preserving compatibility with fibroblasts and pre-osteoblasts. The accelerated bone formation observed in a rat calvarial model emphasizes the potential of this innovative approach to minimize post-surgical infections and enhance bone regeneration outcomes. As a promising alternative for future therapeutic interventions, this material tackles the clinical challenges of extended antibiotic treatments and antibiotic resistance in bone regeneration scenarios.
Collapse
Affiliation(s)
- Jinhong Dai
- Institute of Stomatology & Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang, China
| | - Nicholas G Fischer
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, United States
| | - Joseph R Rahimi
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, United States
| | - Hongning Wang
- Institute of Stomatology & Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang, China
| | - Chaoming Hu
- Institute of Stomatology & Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang, China
| | - Wener Chen
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Yifan Lin
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Ting Sang
- School of Stomatology of Nanchang University & The Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, China
| | - Hooi Pin Chew
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, United States
| | - Liang Kong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Conrado Aparicio
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, United States; Division of Basic Research, Faculty of Odontology UIC Barcelona, Universitat Internacional de Catalunya, Barcelona, Spain; Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.
| | - Zhou Ye
- MDRCBB, Minnesota Dental Research Center for Biomaterials and Biomechanics, University of Minnesota, Minneapolis, MN, United States; Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| | - Shengbin Huang
- Institute of Stomatology & Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, 373 Xueyuan Xi Road, Wenzhou, Zhejiang, China.
| |
Collapse
|
15
|
Zeng L, Noeparvar P, Burne RA, Glezer BS. Genetic characterization of glyoxalase pathway in oral streptococci and its contribution to interbacterial competition. J Oral Microbiol 2024; 16:2322241. [PMID: 38440286 PMCID: PMC10911100 DOI: 10.1080/20002297.2024.2322241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
Objectives To analyze contributions to microbial ecology of Reactive Electrophile Species (RES), including methylglyoxal, generated during glycolysis. Methods Genetic analyses were performed on the glyoxalase pathway in Streptococcus mutans (SM) and Streptococcus sanguinis (SS), followed by phenotypic assays and transcription analysis. Results Deleting glyoxalase I (lguL) reduced RES tolerance to a far greater extent in SM than in SS, decreasing the competitiveness of SM against SS. Although SM displays a greater RES tolerance than SS, lguL-null mutants of either species showed similar tolerance; a finding consistent with the ability of methylglyoxal to induce the expression of lguL in SM, but not in SS. A novel paralogue of lguL (named gloA2) was identified in most streptococci. SM mutant ∆gloA2SM showed little change in methylglyoxal tolerance yet a significant growth defect and increased autolysis on fructose, a phenotype reversed by the addition of glutathione, or by the deletion of a fructose: phosphotransferase system (PTS) that generates fructose-1-phosphate (F-1-P). Conclusions Fructose contributes to RES generation in a PTS-specific manner, and GloA2 may be required to degrade certain RES derived from F-1-P. This study reveals the critical roles of RES in fitness and interbacterial competition and the effects of PTS in modulating RES metabolism.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Payam Noeparvar
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Robert A. Burne
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Benjamin S. Glezer
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
16
|
Hara T, Sakanaka A, Lamont RJ, Amano A, Kuboniwa M. Interspecies metabolite transfer fuels the methionine metabolism of Fusobacterium nucleatum to stimulate volatile methyl mercaptan production. mSystems 2024; 9:e0076423. [PMID: 38289043 PMCID: PMC10878106 DOI: 10.1128/msystems.00764-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/19/2023] [Indexed: 02/21/2024] Open
Abstract
The major oral odor compound methyl mercaptan (CH3SH) is strongly associated with halitosis and periodontitis. CH3SH production stems from the metabolism of polymicrobial communities in periodontal pockets and on the tongue dorsum. However, understanding of CH3SH-producing oral bacteria and their interactions is limited. This study aimed to investigate CH3SH production by major oral bacteria and the impact of interspecies interactions on its generation. Using a newly constructed large-volume anaerobic noncontact coculture system, Fusobacterium nucleatum was found to be a potent producer of CH3SH, with that production stimulated by metabolic interactions with Streptococcus gordonii, an early dental plaque colonizer. Furthermore, analysis of extracellular amino acids using an S. gordonii arginine-ornithine antiporter (ArcD) mutant demonstrated that ornithine excreted from S. gordonii is a key contributor to increased CH3SH production by F. nucleatum. Further study with 13C, 15N-methionine, as well as gene expression analysis, revealed that ornithine secreted by S. gordonii increased the demand for methionine through accelerated polyamine synthesis by F. nucleatum, leading to elevated methionine pathway activity and CH3SH production. Collectively, these findings suggest that interaction between S. gordonii and F. nucleatum plays a key role in CH3SH production, providing a new insight into the mechanism of CH3SH generation in oral microbial communities. A better understanding of the underlying interactions among oral bacteria involved in CH3SH generation can lead to the development of more appropriate prophylactic approaches to treat halitosis and periodontitis. An intervention approach like selectively disrupting this interspecies network could also offer a powerful therapeutic strategy.IMPORTANCEHalitosis can have a significant impact on the social life of affected individuals. Among oral odor compounds, CH3SH has a low olfactory threshold and halitosis is a result of its production. Recently, there has been a growing interest in the collective properties of oral polymicrobial communities, regarded as important for the development of oral diseases, which are shaped by physical and metabolic interactions among community participants. However, it has yet to be investigated whether interspecies interactions have an impact on the production of volatile compounds, leading to the development of halitosis. The present findings provide mechanistic insights indicating that ornithine, a metabolite excreted by Streptococcus gordonii, promotes polyamine synthesis by Fusobacterium nucleatum, resulting in a compensatory increase in demand for methionine, which results in elevated methionine pathway activity and CH3SH production. Elucidation of the mechanisms related to CH3SH production is expected to lead to the development of new strategies for managing halitosis.
Collapse
Affiliation(s)
- Takeshi Hara
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Advanced Technology Institute, Mandom Corporation, Osaka, Japan
| | - Akito Sakanaka
- Department of Preventive Density, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Richard J. Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - Atsuo Amano
- Department of Preventive Density, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| | - Masae Kuboniwa
- Department of Preventive Density, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
17
|
Waldman LJ, Butera T, Boyd JD, Grady ME. Sucrose-mediated formation and adhesion strength of Streptococcus mutans biofilms on titanium. Biofilm 2023; 6:100143. [PMID: 37534044 PMCID: PMC10391683 DOI: 10.1016/j.bioflm.2023.100143] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 08/04/2023] Open
Abstract
Biofilms consist of bacterial cells surrounded by a matrix of extracellular polymeric substance (EPS), which protects the colony from many countermeasures, including antibiotic treatments. Growth and formation of bacterial biofilms are affected by nutrients available in the environment. In the oral cavity, the presence of sucrose affects the growth of Streptococcus mutans that produce acids that erode enamel and form dental caries. Biofilm formation on dental implants commonly leads to severe infections and can restrict osseointegration necessary for the implant to be successful. This work determines the effect of sucrose concentration on biofilm EPS formation and adhesion of Streptococcus mutans, a common oral colonizer, to titanium substrates simulating common dental implants. Biofilm formation and profiles are visualized at high magnification with scanning electron microscopy (SEM). Large mounds and complex structures consisting of bacterial cells and EPS can be seen in biofilms at sucrose concentrations that are favorable for biofilm growth. The laser spallation technique is used to apply stress wave loading to the biofilm, causing the biofilm to delaminate at a critical tensile stress threshold. The critical tensile stress threshold is the adhesion strength. Because laser spallation applies the stress loading to the rear of the substrate, bulk adhesion properties of the biofilm can be determined despite the heterogenous composition and low cohesion strength of the biofilm. Statistical analysis reveals that adhesion strength of biofilms initially increase with increasing sucrose concentration and then decrease as sucrose concentration continues to increase. The adhesion strength of bacterial biofilms to the substrate in this study is compared to the adhesion of osteoblast-like cells to the same substrates published previously. When sucrose is present in the biofilm growth environment, S. mutans adhesion is higher than that of the osteoblast-like cells. Results of this study suggest sucrose-mediated S. mutans biofilms may outcompete osteoblasts in terms of adhesion during osseointegration, which could explain higher rates of peri-implant disease associated with high sugar diets. Further studies demonstrating adhesion differentials between biofilms and cells including co-cultures are needed and motivated by the present work.
Collapse
Affiliation(s)
- Laura J. Waldman
- Department of Mechanical and Aerospace Engineering University of Kentucky, Lexington, KY, 40506, USA
| | - Tony Butera
- Department of Mechanical and Aerospace Engineering University of Kentucky, Lexington, KY, 40506, USA
| | - James D. Boyd
- Department of Mechanical Engineering Clemson University, Clemson, SC, 29634, USA
| | - Martha E. Grady
- Department of Mechanical and Aerospace Engineering University of Kentucky, Lexington, KY, 40506, USA
| |
Collapse
|
18
|
Rudin L, Roth N, Kneubühler J, Dubey BN, Bornstein MM, Shyp V. Inhibitory effect of natural flavone luteolin on Streptococcus mutans biofilm formation. Microbiol Spectr 2023; 11:e0522322. [PMID: 37732737 PMCID: PMC10581090 DOI: 10.1128/spectrum.05223-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 07/24/2023] [Indexed: 09/22/2023] Open
Abstract
Streptococcus mutans is one of the key pathogens responsible for dental caries, which is known to be one of the most prevalent biofilm-associated diseases worldwide. S. mutans virulence strongly depends on its biofilm formation and enamel demineralization abilities due to the production of surface adhesins, exopolysaccharides, and acid in the presence of sugar. Luteolin is an abundant natural flavone with a prominent anti-bacterial function. However, it remains unclear how luteolin affects S. mutans pathogenicity including its acidogenicity and biofilm formation. In this study, the effect of luteolin on S. mutans growth, acid production, and its early and late biofilm formation and biofilm disruption was tested. Luteolin shows strong anti-biofilm activity, while it remains non-toxic for bacterial cell viability. In the biofilm, luteolin reduces the expression of S. mutans virulence genes such as gbpC, spaP, gtfBCD, and ftf encoding for surface adhesins and extracellular polysaccharides (EPS)-producing enzymes, which reflects in the strong reduction of bacteria and EPS. Further, it reduces water-insoluble glucan production in the biofilm, potentially, via direct interference with glucosyltransfereases (Gtfs). Moreover, at biofilm inhibitory concentrations, luteolin significantly reduces acid production by S. mutans. Finally, luteolin could target S. mutans amyloid proteins to disrupt the biofilm based on the observation that it inhibits the uptake of the amyloid dye, thioflavin T, by S. mutans extracellular proteins and failed to inhibit biofilm formation by the mutant strain lacking three main amyloid proteins. In conclusion, luteolin appears to be a potent natural compound with pleiotropic anti-biofilm properties against one of the main cariogenic human pathogens, S. mutans. IMPORTANCE Flavonoids are natural compounds with proven anti-bacterial and anti-biofilm properties. Here, we describe the anti-biofilm properties of natural flavone luteolin against the main cariogenic bacteria, S. mutans. Luteolin inhibited gene expression of cell surface adhesins, fructosyltransferases, and glucosyltransferases, which promotes a significant reduction of bacterial and EPS biomass in early and late biofilms. Moreover, luteolin could directly target S. mutans Gtfs and functional amyloids to modulate pathogenic biofilms. These observations provide important insights into the anti-biofilm properties of luteolin while laying out a framework for future therapeutic strategies targeting biofilm-associated virulence factors of oral pathogens.
Collapse
Affiliation(s)
- Lucille Rudin
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Noelle Roth
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Julien Kneubühler
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Badri Nath Dubey
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Notkestr, Hamburg, Germany
| | - Michael M. Bornstein
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
- Department of Oral Health and Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Viktoriya Shyp
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
- Department of Oral Health and Medicine, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| |
Collapse
|
19
|
Paredes A, Iheacho C, Smith AT. Metal Messengers: Communication in the Bacterial World through Transition-Metal-Sensing Two-Component Systems. Biochemistry 2023; 62:2339-2357. [PMID: 37539997 PMCID: PMC10530140 DOI: 10.1021/acs.biochem.3c00296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Bacteria survive in highly dynamic and complex environments due, in part, to the presence of systems that allow the rapid control of gene expression in the presence of changing environmental stimuli. The crosstalk between intra- and extracellular bacterial environments is often facilitated by two-component signal transduction systems that are typically composed of a transmembrane histidine kinase and a cytosolic response regulator. Sensor histidine kinases and response regulators work in tandem with their modular domains containing highly conserved structural features to control a diverse array of genes that respond to changing environments. Bacterial two-component systems are widespread and play crucial roles in many important processes, such as motility, virulence, chemotaxis, and even transition metal homeostasis. Transition metals are essential for normal prokaryotic physiological processes, and the presence of these metal ions may also influence pathogenic virulence if their levels are appropriately controlled. To do so, bacteria use transition-metal-sensing two-component systems that bind and respond to rapid fluctuations in extracytosolic concentrations of transition metals. This perspective summarizes the structural and metal-binding features of bacterial transition-metal-sensing two-component systems and places a special emphasis on understanding how these systems are used by pathogens to establish infection in host cells and how these systems may be targeted for future therapeutic developments.
Collapse
Affiliation(s)
- Alexander Paredes
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Chioma Iheacho
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| | - Aaron T Smith
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
20
|
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
|
21
|
Skilbeck MG, Cannon RD, Farella M, Mei L. The effect of surface roughening of orthodontic elastomers on hydrophobicity and in vitro adherence of Streptococcus gordonii. J Mech Behav Biomed Mater 2023; 143:105881. [PMID: 37209593 DOI: 10.1016/j.jmbbm.2023.105881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/22/2023]
Abstract
OBJECTIVES Biofilm formation around orthodontic appliances causes gingivitis, enamel decalcification and caries. Bacteria adhere less readily to superhydrophobic surfaces. The aim of this study was to determine whether a superhydrophobic surface could be generated on orthodontic elastomers by surface modification in order to reduce bacterial adhesion. MATERIALS AND METHODS Orthodontic elastomers were modified with sandpapers of various grit sizes (80-600 grit). Surface roughness of the modified and unmodified surfaces was assessed qualitatively with scanning electron microscopy and quantitatively with confocal microscopy. Water contact angles were measured with a goniometer to quantify hydrophobicity. Measurements were performed on unextended elastomers (100% original length) and elastomers extended to 150%, and 200% of the original length. Adhesion of Streptococcus gordonii to saliva coated elastomers was measured by counting colony forming units on agar plates. RESULTS Abrasion with different sandpapers produced elastomers with surface roughness (Ra) ranging from 2 to 12 μm. Contact angles followed a quadratic trend with a maximum contact angle of 104° at an Ra of 7-9 μm. Average water contact angles, when viewed perpendicular to the direction of extension, decreased from 99° to 90° when the extension was increased from 100% to 200% and increased from 100° to 103° when viewed parallel to the direction of extension. Bacterial adhesion increased as roughness increased and this effect was more pronounced with elastomer extension. CONCLUSION The surface roughness of orthodontic elastomers influences both their hydrophobicity and bacterial adhesion. Superhydrophobicity of elastomers could not be achieved with sandpaper abrasion.
Collapse
Affiliation(s)
- Michael G Skilbeck
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, New Zealand.
| | - Richard D Cannon
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, New Zealand.
| | - Mauro Farella
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, New Zealand; Department of Surgical Sciences, University of Cagliari, Italy.
| | - Li Mei
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, New Zealand.
| |
Collapse
|
22
|
Yesilay G, Dos Santos OAL, A BR, Hazeem LJ, Backx BP, J JV, Kamel AH, Bououdina M. Impact of pathogenic bacterial communities present in wastewater on aquatic organisms: Application of nanomaterials for the removal of these pathogens. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 261:106620. [PMID: 37399782 DOI: 10.1016/j.aquatox.2023.106620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 07/05/2023]
Abstract
Contaminated wastewater (WW) can cause severe hazards to numerous delicate ecosystems and associated life forms. In addition, human health is negatively impacted by the presence of microorganisms in water. Multiple pathogenic microorganisms in contaminated water, including bacteria, fungi, yeast, and viruses, are vectors for several contagious diseases. To avoid the negative impact of these pathogens, WW must be free from pathogens before being released into stream water or used for other reasons. In this review article, we have focused on pathogenic bacteria in WW and summarized the impact of the different types of pathogenic bacteria on marine organisms. Moreover, we presented a variety of physical and chemical techniques that have been developed to provide a pathogen-free aquatic environment. Among the techniques, membrane-based techniques for trapping hazardous biological contaminants are gaining popularity around the world. Besides, novel and recent advancements in nanotechnological science and engineering suggest that many waterborne pathogens could be inactivated using nano catalysts, bioactive nanoparticles, nanostructured catalytic membranes, nanosized photocatalytic structures, and electrospun nanofibers and processes have been thoroughly examined.
Collapse
Affiliation(s)
- Gamze Yesilay
- Molecular Biology and Genetics Department, Hamidiye Institute of Health Sciences, University of Health Sciences-Türkiye, Istanbul 34668, Türkiye; Experimental Medicine Application & Research Center, University of Health Sciences, Validebag Research Park, Uskudar, Istanbul 34662, Türkiye
| | | | - Bevin Roger A
- Department of Chemistry, Catalysis and Nanomaterials Research Laboratory, Loyola College, Chennai 600 034, India
| | - Layla J Hazeem
- Department of Biology, College of Science, University of Bahrain, 32038, Bahrain
| | | | - Judith Vijaya J
- Department of Chemistry, Catalysis and Nanomaterials Research Laboratory, Loyola College, Chennai 600 034, India
| | - Ayman H Kamel
- Department of Chemistry, College of Science, University of Bahrain, 32038, Bahrain; Department of Chemistry, Faculty of Science, Ain Shams University, Abbasia, Cairo 11566, Egypt
| | - Mohamed Bououdina
- Department of Mathematics and Science, Faculty of Humanities and Sciences, Prince Sultan University, Riyadh, Saudi Arabia.
| |
Collapse
|
23
|
Nasir SN, Iftikhar A, Zubair F, Alshammari A, Alharbi M, Alasmari AF, Khan A, Waseem M, Ali SS, Ali L, Waheed Y, Wei DQ. Structural vaccinology-based design of multi-epitopes vaccine against Streptococcus gordonii and validation using molecular modeling and immune simulation approaches. Heliyon 2023; 9:e16148. [PMID: 37234653 PMCID: PMC10208844 DOI: 10.1016/j.heliyon.2023.e16148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
Streptococcus gordonii is an oral bacterium colonizing the dental cavity and leading to plaque formation. This pervasive colonizer is also the etiologic agent of bacterial endocarditis and has a major role in infective endocarditis. The bacteria reach the heart through oral bleeding, leading to inflammation of cardiovascular valves. Over the past 50 years, it has shown a significant pathogenic role in immunocompromised and neutropenic patients. Since antibiotic resistance has created prophylaxis failure towards infective endocarditis, a potent therapeutic candidate is needed. Therefore, multi-epitopes vaccine offers advantages over the other approaches. Thus, herein, numerous molecular-omics tools were exploited to mine immunogenic peptides, i.e., T-cell and B-cell epitopes, and construct a vaccine sequence. Our findings revealed a total of 24 epitopes, including CTL, HTL, and B-cell are responsible for imparting immune responses, which were combined with the help of different linkers, and MEVC was constructed. Multifactorial validation of the candidate vaccine was performed to minimize the risk factors. The final sequence was docked with TLR2 to validate its conformation compatibility with receptor and long-term interactions stability. Our analysis revealed that the vaccine construct is immunogenic and non-allergenic. The construct also established various contacts with the immune receptor. Finally, the vaccine sequence was reverse-translated, optimized for codon usage, and analyzed for expression in the Escherichia coli K12 strain. Maximum expression was noted with a CAI score of 0.95. In silico immune simulation revealed that the antigen was neutralized on the 3rd day after injection. In conclusion, the current study warrants validation of the vaccine construct both in in vitro and in vivo models for accurate therapeutic intervention.
Collapse
Affiliation(s)
- Syed Nouman Nasir
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Punjab, Pakistan
| | - Ayesha Iftikhar
- Government Khwaja Muhammad Safdar Medical College, Sialkot, Punjab, Pakistan
| | - Farukh Zubair
- Rashid Latif Medical College, Lahore, Punjab, Pakistan
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abbas Khan
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nayang, Henan, 473006, PR China
| | - Muhammad Waseem
- Faculty of Rehabilitation and Allied Health Science, Riphah International University, Islamabad, Pakistan
| | - Syed Shujait Ali
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Punjab, Pakistan
| | - Liaqat Ali
- Fisch College of Pharmacy, The University of Texas at Tyler, Tyler, TX, USA
| | - Yasir Waheed
- Office of Research, Innovation, and Commercialization (ORIC), Shaheed Zulfiqar Ali Bhutto Medical University (SZABMU), Islamabad, 44000, Pakistan
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, 1401, Lebanon
| | - Dong-Qing Wei
- Department of Bioinformatics and Biological Statistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, PR China
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nayang, Henan, 473006, PR China
| |
Collapse
|
24
|
Zaidi S, Ali K, Chawla YM, Khan AU. mltG gene deletion mitigated virulence potential of Streptococcus mutans: An in-vitro, ex-situ and in-vivo study. AMB Express 2023; 13:19. [PMID: 36806997 PMCID: PMC9941400 DOI: 10.1186/s13568-023-01526-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Bacterial cells are surrounded by a peptidoglycan (PG) cell wall, which is essential for cell integrity and intrinsic biogenesis pathways; hence, the cell wall is a potential target for several antibiotics. Among several lytic transglycosylases (LTs), the mltG gene plays a crucial role in the synthesis of peripheral PG. It localises the re-modelled PGs for septum formation and cleavage across the bacterial cell wall during daughter cells separation. However, the role of mltG gene in bacterial virulence, particularly in Gram-positive bacteria during dentine biofilm and caries development, has remained unexplored. Hence, we exploited Gram-positive Streptococcus mutans cells for the very first time to construct a mltG knock-out bacterial strain, e.g., ΔmltG S. mutans. Systematic comparative investigations revealed that doubling time (Td), survival, enzymatic efficiencies, pH tolerance, bio-synthesise of lipid, proteins and DNA, biofilm formation and dentine lesions were significantly (p < 0.001) compromised in case of ΔmltG S. mutans than wild type strain. The qRT-PCR based gene expression profiling revealed that transcriptional expression of critically important genes involved in biofilm, metabolism, and stress response were dysregulated in the mutant. Besides, an incredible reduction in dentine caries development was found in the molar teeth of Wistar rats and also in human extracted teeth. Concisely, these trends obtained evidently advocated the fact that the deletion of mltG gene can be a potential target to impair the S. mutans virulence through severe growth retardation, thereby reducing the virulence potential of S. mutans.
Collapse
Affiliation(s)
- Sahar Zaidi
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| | - Khursheed Ali
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| | - Yadya M. Chawla
- grid.425195.e0000 0004 0498 7682ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Asad U. Khan
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| |
Collapse
|
25
|
Gloag ES, Khosravi Y, Masters JG, Wozniak DJ, Amorin Daep C, Stoodley P. A Combination of Zinc and Arginine Disrupt the Mechanical Integrity of Dental Biofilms. Microbiol Spectr 2023; 11:e0335122. [PMID: 36472465 PMCID: PMC9927089 DOI: 10.1128/spectrum.03351-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Mechanical cleaning remains the standard of care for maintaining oral hygiene. However, mechanical cleaning is often augmented with active therapeutics that further promote oral health. A dentifrice, consisting of the "Dual Zinc plus Arginine" (DZA) technology, was found to be effective at controlling bacteria using in vitro laboratory studies, translating to clinical efficacy to deliver plaque and gingivitis reduction benefits. Here, we used biophysical analyses and confocal laser scanning microscopy to understand how a DZA dentifrice impacted the mechanical properties of dental plaque biofilms and determine if changes to biofilm rheology enhanced the removal of dental plaque. Using both uniaxial mechanical indentation and an adapted rotating-disc rheometry assay, it was found that DZA treatment compromised biofilm mechanical integrity, resulting in the biofilm being more susceptible to removal by shear forces compared to treatment with either arginine or zinc alone. Confocal laser scanning microscopy revealed that DZA treatment reduced the amount of extracellular polymeric slime within the biofilm, likely accounting for the reduced mechanical properties. We propose a model where arginine facilitates the entry of zinc into the biofilm, resulting in additive effects of the two activities toward dental plaque biofilms. Together, our results support the use of a dentifrice containing Dual Zinc plus Arginine as part of daily oral hygiene regimens. IMPORTANCE Mechanical removal of dental plaque is augmented with therapeutic compounds to promote oral health. A dentifrice containing the ingredients zinc and arginine has shown efficacy at reducing dental plaque both in vitro and in vivo. However, how these active compounds interact together to facilitate dental plaque removal is unclear. Here, we used a combination of biophysical analyses and microscopy to demonstrate that combined treatment with zinc and arginine targets the matrix of dental plaque biofilms, which destabilized the mechanical integrity of these microbial communities, making them more susceptible to removal by shear forces.
Collapse
Affiliation(s)
- Erin S. Gloag
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Yalda Khosravi
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - James G. Masters
- Colgate-Palmolive Technology Center, Piscataway, New Jersey, USA
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | | | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
- National Biofilm Innovation Centre (NBIC), University of Southampton, Southampton, United Kingdom
- National Centre for Advanced Tribology at Southampton (nCATS), Mechanical Engineering, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
26
|
Yun Z, Xianghong L, Qianhua G, Qin D. Copper ions inhibit Streptococcus mutans-Veillonella parvula dual biofilm by activating Streptococcus mutans reactive nitrogen species. BMC Oral Health 2023; 23:48. [PMID: 36709299 PMCID: PMC9883903 DOI: 10.1186/s12903-023-02738-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/11/2023] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND To investigate the inhibition mechanism of copper ions on Streptococcus mutans-Veillonella parvula dual biofilm. METHODS S. mutans-V. parvula dual biofilm was constructed and copper ions were added at different concentrations. After the biofilm was collected, RNA-seq and qRT-PCR were then performed to get gene information. RESULTS The coculture of S. mutans and V. parvula formed a significantly better dual biofilm of larger biomass than S. mutans mono biofilm. And copper ions showed a more significant inhibitory effect on S. mutans-V. parvula dual biofilm than on S. mutans mono biofilm when copper ions concentration reached 100 µM, and copper ions showed a decreased inhibitory effect on S. gordonii-V. parvula dual biofilm and S. sanguis-V.parvula dual biofilm than on the two mono biofilms as the concentration of copper ions increased. And common trace elements such as iron, magnesium, and zinc showed no inhibitory effect difference on S. mutans-V. parvula dual biofilm. The RNA-seq results showed a significant difference in the expression of a new ABC transporter SMU_651c, SMU_652c, SMU_653c, and S. mutans copper chaperone copYAZ. SMU_651c, SMU_652c, and SMU_653c were predicted to function as nitrite/nitrate transporter-related proteins, which suggested the specific inhibition of copper ions on S. mutans-V. parvula dual biofilm may be caused by the activation of S. mutans reactive nitrogen species. CONCLUSIONS Streptococcus mutans and Veillonella parvula are symbiotic, forming a dual biofilm of larger biomass to better resist the external antibacterial substances, which may increase the virulence of S. mutans. While common trace elements such as iron, magnesium, and zinc showed no specific inhibitory effect on S. mutans-V. parvula dual biofilm, copper ion had a unique inhibitory effect on S. mutans-V. parvula dual biofilm which may be caused by activating S. mutans RNS when copper ions concentration reached 250 µM.
Collapse
Affiliation(s)
- Zhang Yun
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, 610041 Sichuan China ,grid.13291.380000 0001 0807 1581Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041 Sichuan China
| | - Liu Xianghong
- grid.54549.390000 0004 0369 4060Department of Stomatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 China
| | - Gao Qianhua
- grid.54549.390000 0004 0369 4060Department of Stomatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 China
| | - Du Qin
- grid.54549.390000 0004 0369 4060Department of Stomatology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, 610072 China
| |
Collapse
|
27
|
Zeng L, Walker AR, Burne RA, Taylor ZA. Glucose Phosphotransferase System Modulates Pyruvate Metabolism, Bacterial Fitness, and Microbial Ecology in Oral Streptococci. J Bacteriol 2023; 205:e0035222. [PMID: 36468868 PMCID: PMC9879115 DOI: 10.1128/jb.00352-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Spontaneous mutants with defects in the primary glucose phosphotransferase permease (manLMNO) of Streptococcus sanguinis SK36 showed enhanced fitness at low pH. Transcriptomics and metabolomics with a manL deletion mutant (SK36/manL) revealed redirection of pyruvate to production of acetate and formate, rather than lactate. These observations were consistent with measurements of decreased lactic acid accumulation and increased excretion of acetate, formate, pyruvate, and H2O2. Genes showing increased expression in SK36/manL included those encoding carbohydrate transporters, extracellular glycosidases, intracellular polysaccharide metabolism, and arginine deiminase and pathways for metabolism of acetoin, ethanolamine, ascorbate, and formate, along with genes required for membrane biosynthesis and adhesion. Streptococcus mutans UA159 persisted much better in biofilm cocultures with SK36/manL than with SK36, an effect that was further enhanced by culturing the biofilms anaerobically but dampened by adding arginine to the medium. We posited that the enhanced persistence of S. mutans with SK36/manL was in part due to excess excretion of pyruvate by the latter, as addition of pyruvate to S. mutans-S. sanguinis cocultures increased the proportions of UA159 in the biofilms. Reducing the buffer capacity or increasing the concentration of glucose benefited UA159 when cocultured with SK36, but not with SK36/manL, likely due to the altered metabolism and enhanced acid tolerance of the mutant. When manL was deleted in S. mutans or Streptococcus gordonii, the mutants presented altered fitness characteristics. Our study demonstrated that phosphotransferase system (PTS)-dependent modulation of central metabolism can profoundly affect streptococcal fitness and metabolic interactions, revealing another dimension in commensal-pathogen relationships influencing dental caries development. IMPORTANCE Dental caries is underpinned by a dysbiotic microbiome and increased acid production. As beneficial bacteria that can antagonize oral pathobionts, oral streptococci such as S. sanguinis and S. gordonii can ferment many carbohydrates, despite their relative sensitivity to low pH. We characterized the molecular basis for why mutants of glucose transporter ManLMNO of S. sanguinis showed enhanced production of hydrogen peroxide and ammonia and improved persistence under acidic conditions. A metabolic shift involving more than 300 genes required for carbohydrate transport, energy production, and envelope biogenesis was observed. Significantly, manL mutants engineered in three different oral streptococci displayed altered capacities for acid production and interspecies antagonism, highlighting the potential for targeting the glucose-PTS to modulate the pathogenicity of oral biofilms.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Alejandro R. Walker
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Robert A. Burne
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Zachary A. Taylor
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
28
|
Polizzi A, Donzella M, Nicolosi G, Santonocito S, Pesce P, Isola G. Drugs for the Quorum Sensing Inhibition of Oral Biofilm: New Frontiers and Insights in the Treatment of Periodontitis. Pharmaceutics 2022; 14:2740. [PMID: 36559234 PMCID: PMC9781207 DOI: 10.3390/pharmaceutics14122740] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Chemical molecules are used by microorganisms to communicate with each other. Quorum sensing is the mechanism through which microorganisms regulate their population density and activity with chemical signaling. The inhibition of quorum sensing, called quorum quenching, may disrupt oral biofilm formation, which is the main etiological factor of oral diseases, including periodontitis. Periodontitis is a chronic inflammatory disorder of infectious etiology involving the hard and soft periodontal tissues and which is related to various systemic disorders, including cardiovascular diseases, diabetes and obesity. The employment of adjuvant therapies to traditional scaling and root planing is currently being studied to further reduce the impact of periodontitis. In this sense, using antibiotics and antiseptics involves non-negligible risks, such as antibiotic resistance phenomena and hinders the re-establishment of eubiosis. Different quorum sensing signal molecules have been identified in periodontal pathogenic oral bacteria. In this regard, quorum sensing inhibitors are emerging as some interesting solutions for the management of periodontitis. Therefore, the aim of this review is to summarize the current state of knowledge on the mechanisms of quorum sensing signal molecules produced by oral biofilm and to analyze the potential of quorum sensing inhibitors for the management of periodontitis.
Collapse
Affiliation(s)
- Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
- Department of Surgical Sciences (DISC), University of Genova, 16132 Genoa, Italy
| | - Martina Donzella
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Giada Nicolosi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Paolo Pesce
- Department of Surgical Sciences (DISC), University of Genova, 16132 Genoa, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| |
Collapse
|
29
|
Prasad V, Washburn F, Barouni B, Saeed M. A Rare Case of Prosthetic Joint Infection with Streptococcus gordonii. Am J Case Rep 2022; 23:e937271. [PMID: 36355628 PMCID: PMC9664417 DOI: 10.12659/ajcr.937271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Chronic prosthetic joint infection (PJI) is a devastating complication following total joint arthroplasty, resulting in significant morbidity and mortality. The criterion standard of treatment for chronic PJI is two-stage revision arthroplasty consisting of complete hardware removal, thorough irrigation and debridement, placement of an antibiotic spacer, prolonged intravenous antibiotics based on culture sensitivities, and revision total knee arthroplasty once the infection resolves. The most common organism implicated in chronic PJI is Staphylococcus aureus. CASE REPORT In this report, we have summarized the case of a 75-year-old woman who developed chronic PJI caused by an unusual organism, Streptococcus gordonii, 1 year after a right total knee arthroplasty. S. gordonii is a gram-positive organism that is an oral flora and a colonizer of human teeth. This organism is known to create biofilm on the human teeth, more commonly known as dental plaque. S. gordonii has the ability to travel to extraoral sites and cause infection. It has been found to be a cause of subacute bacterial endocarditis, but it has been rarely described in the literature as a cause of prosthetic joint infection. Treatment of S. gordonii requires a tailored approach. CONCLUSIONS This case report highlights the clinical presentation, diagnosis, and treatment of chronic prosthetic joint infection caused by S. gordonii and identifies a rare cause of PJI that is not well documented in the literature. Streptococcal PJI portends a poorer prognosis, and identification of this organism is crucial for prompt treatment and improved outcomes for PJI.
Collapse
Affiliation(s)
- Varsha Prasad
- Department of Internal Medicine, Community Memorial Health System, Ventura, CA, USA,Corresponding Author: Varsha Prasad, e-mail:
| | - Frederic Washburn
- Department of Orthopedic Surgery, Community Memorial Health System, Ventura, CA, USA
| | - Baina Barouni
- Graduate Medical Education, Western University School of Medicine, Ventura, CA, USA
| | - Musab Saeed
- Department of Infectious Disease, Community Memorial Health System, Ventura, CA, USA
| |
Collapse
|
30
|
Rabe A, Gesell Salazar M, Michalik S, Kocher T, Below H, Völker U, Welk A. Impact of different oral treatments on the composition of the supragingival plaque microbiome. J Oral Microbiol 2022; 14:2138251. [PMID: 36338832 PMCID: PMC9629129 DOI: 10.1080/20002297.2022.2138251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Dental plaque consists of a diverse microbial community embedded in a complex structure of exopolysaccharides. Dental biofilms form a natural barrier against pathogens but lead to oral diseases in a dysbiotic state. Objective Using a metaproteome approach combined with a standard plaque-regrowth study, this pilot study examined the impact of different concentrations of lactoperoxidase (LPO) on early plaque formation, and active biological processes. Design Sixteen orally healthy subjects received four local treatments as a randomized single-blind study based on a cross-over design. Two lozenges containing components of the LPO-system in different concentrations were compared to a placebo and Listerine®. The newly formed dental plaque was analyzed by mass spectrometry (nLC-MS/MS). Results On average 1,916 metaproteins per sample were identified, which could be assigned to 116 genera and 1,316 protein functions. Listerine® reduced the number of metaproteins and their relative abundance, confirming the plaque inhibiting effect. The LPO-lozenges triggered mainly higher metaprotein abundances of early and secondary colonizers as well as bacteria associated with dental health but also periodontitis. Functional information indicated plaque biofilm growth. Conclusion In conclusion, the mechanisms on plaque biofilm formation of Listerine® and the LPO-system containing lozenges are different. In contrast to Listerine®, the lozenges led to a higher bacterial diversity.
Collapse
Affiliation(s)
- Alexander Rabe
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany,CONTACT Alexander Rabe University Medicine Greifswald, Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, Felix-Hausdorff-Str. 8, 17489Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Thomas Kocher
- Center for Dentistry, Oral and Maxillofacial Medicine, Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, Dental School of University Medicine Greifswald, Fleischmannstraße 42-44, 17489
| | - Harald Below
- Institute for Hygiene and Environmental Medicine, University Medicine Greifswald, Walter-Rathenau-Straße 49 A17475Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Alexander Welk
- Center for Dentistry, Oral and Maxillofacial Medicine, Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, Dental School of University Medicine Greifswald, Fleischmannstraße 42-44, 17489
| |
Collapse
|
31
|
Prevalence of Type IV Pili-Mediated Twitching Motility in Streptococcus sanguinis Strains and Its Impact on Biofilm Formation and Host Adherence. Appl Environ Microbiol 2022; 88:e0140322. [PMID: 36094177 PMCID: PMC9499025 DOI: 10.1128/aem.01403-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type IV pili (Tfp) are known to mediate several biological activities, including surface-dependent twitching motility. Although a pil gene cluster for Tfp biosynthesis is found in all sequenced Streptococcus sanguinis strains, Tfp-mediated twitching motility is less commonly detected. Upon examining 81 clinical strains, 39 strains generated twitching zones on blood agar plates (BAP), while 27 strains displayed twitching on Todd-Hewitt (TH) agar. Although BAP appears to be more suitable for the development of twitching zones, 5 strains exhibited twitching motility only on TH agar, indicating that twitching motility is not only strain specific but also sensitive to growth media. Furthermore, different twitching phenotypes were observed in strains expressing comparable levels of pilT, encoding the retraction ATPase, suggesting that the twitching phenotype on agar plates is regulated by multiple factors. By using a PilT-null and a pilin protein-null derivative (CHW02) of twitching-active S. sanguinis CGMH010, we found that Tfp retraction was essential for biofilm stability. Further, biofilm growth was amplified in CHW02 in the absence of shearing force, indicating that S. sanguinis may utilize other ligands for biofilm formation in the absence of Tfp. Similar to SK36, Tfp from CGMH010 were required for colonization of host cells, but PilT only marginally affected adherence and only in the twitching-active strain. Taken together, the results suggest that Tfp participates in host cell adherence and that Tfp retraction facilitates biofilm stability. IMPORTANCE Although the gene clusters encoding Tfp are commonly present in Streptococcus sanguinis, not all strains express surface-dependent twitching motility on agar surfaces. Regardless of whether the Tfp could drive motility, Tfp can serve as a ligand for the colonization of host cells. Though many S. sanguinis strains lack twitching activity, motility can enhance biofilm stability in a twitching-active strain; thus, perhaps motility provides little or no advantage to the survival of bacteria within dental plaque. Rather, Tfp retraction could provide additional advantages for the bacteria to establish infections outside the oral cavity.
Collapse
|
32
|
Weng L, Wu L, Guo R, Ye J, Liang W, Wu W, Chen L, Yang D. Lactobacillus cell envelope-coated nanoparticles for antibiotic delivery against cariogenic biofilm and dental caries. J Nanobiotechnology 2022; 20:356. [PMID: 35918726 PMCID: PMC9344742 DOI: 10.1186/s12951-022-01563-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/20/2022] [Indexed: 12/31/2022] Open
Abstract
Background Due to their prevalence, dental caries ranks first among all diseases endangering human health. Therefore, the prevention of caries is of great significance, as caries have become a serious public health problem worldwide. Currently, using nanoscale drug delivery systems to prevent caries has received increased attention. However, the preventive efficacy of these systems is substantially limited due to the unique physiological structure of cariogenic biofilms. Thus, novel strategies aimed at combating cariogenic biofilms to improve preventive efficiency against caries are meaningful and very necessary. Herein, inspired by cell membrane coating technology and Lactobacillus strains, we coated triclosan (TCS)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (TCS@PLGA-NPs) with an envelope of Lactobacillus (LA/TCS@PLGA-NPs) and investigated their potential as a nanoparticle delivery system against cariogenic biofilms and dental caries. Results LA/TCS@PLGA-NPs were successfully prepared with favorable properties, including a coated envelope, controllable size, negative charge, sustained drug-release kinetics and so on. The LA/TCS@PLGA-NPs inherited native properties from the source cell surface, thus the LA/TCS@PLGA-NPs adhered to S. mutans, integrated into the S. mutans biofilm, and interfered with the biofilm formation of S. mutans. The nanoparticles significantly inhibited the activity, biomass and virulence gene expression of S. mutans biofilms in vitro. Additionally, LA/TCS@PLGA-NPs exhibited a long-lasting inhibitory effect on the progression of caries in vivo. The safety performance of the nanoparticles is also favorable. Conclusions Our findings reveal that the antibiofilm effect of LA/TCS@PLGA-NPs relies not only on the inheritance of native properties from the Lactobacillus cell surface but also on the inhibitory effect on the activity, biomass and virulence of S. mutans biofilms. Thus, these nanoparticles could be considered feasible candidates for a new class of effective drug delivery systems for the prevention of caries. Furthermore, this work provides new insights into cell membrane coating technology and presents a novel strategy to combat bacterial biofilms and associated infections. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01563-x.
Collapse
Affiliation(s)
- Luting Weng
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
| | - Lang Wu
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, 401147, China
| | - Rongjuan Guo
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Jiajia Ye
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Wen Liang
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, 401147, China
| | - Wei Wu
- Bioengineering College of Chongqing University, No.174 Shazhengjie, Shapingba, Chongqing, 400044, China.
| | - Liang Chen
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.
| | - Deqin Yang
- Stomatological Hospital of Chongqing Medical University, No. 426, Songshi North Road, Yubei District, Chongqing, 401147, China.
| |
Collapse
|
33
|
Host–Bacterial Interactions: Outcomes of Antimicrobial Peptide Applications. MEMBRANES 2022; 12:membranes12070715. [PMID: 35877918 PMCID: PMC9317001 DOI: 10.3390/membranes12070715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 02/04/2023]
Abstract
The bacterial membrane is part of a secretion system which plays an integral role to secrete proteins responsible for cell viability and pathogenicity; pathogenic bacteria, for example, secrete virulence factors and other membrane-associated proteins to invade the host cells through various types of secretion systems (Type I to Type IX). The bacterial membrane can also mediate microbial communities’ communication through quorum sensing (QS), by secreting auto-stimulants to coordinate gene expression. QS plays an important role in regulating various physiological processes, including bacterial biofilm formation while providing increased virulence, subsequently leading to antimicrobial resistance. Multi-drug resistant (MDR) bacteria have emerged as a threat to global health, and various strategies targeting QS and biofilm formation have been explored by researchers worldwide. Since the bacterial secretion systems play such a crucial role in host–bacterial interactions, this review intends to outline current understanding of bacterial membrane systems, which may provide new insights for designing approaches aimed at antimicrobials discovery. Various mechanisms pertaining interaction of the bacterial membrane with host cells and antimicrobial agents will be highlighted, as well as the evolution of bacterial membranes in evasion of antimicrobial agents. Finally, the use of antimicrobial peptides (AMPs) as a cellular device for bacterial secretion systems will be discussed as emerging potential candidates for the treatment of multidrug resistance infections.
Collapse
|
34
|
Quantification of Extracellular DNA Network Abundance and Architecture within Streptococcus gordonii Biofilms Reveals Modulatory Factors. Appl Environ Microbiol 2022; 88:e0069822. [PMID: 35695569 PMCID: PMC9275248 DOI: 10.1128/aem.00698-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Extracellular DNA (eDNA) is an important component of biofilm matrix that serves to maintain biofilm structural integrity, promotes genetic exchange within the biofilm, and provides protection against antimicrobial compounds. Advances in microscopy techniques have provided evidence of the cobweb- or lattice-like structures of eDNA within biofilms from a range of environmental niches. However, methods to reliably assess the abundance and architecture of eDNA remain lacking. This study aimed to address this gap by development of a novel, high-throughput image acquisition and analysis platform for assessment of eDNA networks in situ within biofilms. Utilizing Streptococcus gordonii as the model, the capacity for this imaging system to reliably detect eDNA networks and monitor changes in abundance and architecture (e.g., strand length and branch number) was verified. Evidence was provided of a synergy between glucans and eDNA matrices, while it was revealed that surface-bound nuclease SsnA could modify these eDNA structures under conditions permissive for enzymatic activity. Moreover, cross talk between the competence and hexaheptapeptide permease systems was shown to regulate eDNA release by S. gordonii. This novel imaging system can be applied across the wider field of biofilm research, with potential to significantly advance interrogation of the mechanisms by which the eDNA network architecture develops, how it can influence biofilm properties, and how it may be targeted for therapeutic benefit. IMPORTANCE Extracellular DNA (eDNA) is critical for maintaining the structural integrity of many microbial biofilms, making it an attractive target for the management of biofilms. However, our knowledge and targeting of eDNA are currently hindered by a lack of tools for the quantitative assessment of eDNA networks within biofilms. Here, we demonstrate use of a novel image acquisition and analysis platform with the capacity to reliably monitor the abundance and architecture of eDNA networks. Application of this tool to Streptococcus gordonii biofilms has provided new insights into how eDNA networks are stabilized within the biofilm and the pathways that can regulate eDNA release. This highlights how exploitation of this novel imaging system across the wider field of biofilm research has potential to significantly advance interrogation of the mechanisms by which the eDNA network architecture develops, how it can influence biofilm properties, and how it may be targeted for therapeutic benefit.
Collapse
|
35
|
Santore MM. Interplay of physico-chemical and mechanical bacteria-surface interactions with transport processes controls early biofilm growth: A review. Adv Colloid Interface Sci 2022; 304:102665. [PMID: 35468355 DOI: 10.1016/j.cis.2022.102665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/01/2022]
Abstract
Biofilms initiate when bacteria encounter and are retained on surfaces. The surface orchestrates biofilm growth through direct physico-chemical and mechanical interactions with different structures on bacterial cells and, in turn, through its influence on cell-cell interactions. Individual cells respond directly to a surface through mechanical or chemical means, initiating "surface sensing" pathways that regulate gene expression, for instance producing extra cellular matrix or altering phenotypes. The surface can also physically direct the evolving colony morphology as cells divide and grow. In either case, the physico-chemistry of the surface influences cells and cell communities through mechanisms that involve additional factors. For instance the numbers of cells arriving on a surface from solution relative to the generation of new cells by division depends on adhesion and transport kinetics, affecting early colony density and composition. Separately, the forces experienced by adhering cells depend on hydrodynamics, gravity, and the relative stiffnesses and viscoelasticity of the cells and substrate materials, affecting mechanosensing pathways. Physical chemistry and surface functionality, along with interfacial mechanics also influence cell-surface friction and control colony morphology, in particular 2D and 3D shape. This review focuses on the current understanding of the mechanisms in which physico-chemical interactions, deriving from surface functionality, impact individual cells and cell community behavior through their coupling with other interfacial processes.
Collapse
|
36
|
Wang N, Ma Y, Shi H, Song Y, Guo S, Yang S. Mg-, Zn-, and Fe-Based Alloys With Antibacterial Properties as Orthopedic Implant Materials. Front Bioeng Biotechnol 2022; 10:888084. [PMID: 35677296 PMCID: PMC9168471 DOI: 10.3389/fbioe.2022.888084] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022] Open
Abstract
Implant-associated infection (IAI) is one of the major challenges in orthopedic surgery. The development of implants with inherent antibacterial properties is an effective strategy to resolve this issue. In recent years, biodegradable alloy materials have received considerable attention because of their superior comprehensive performance in the field of orthopedic implants. Studies on biodegradable alloy orthopedic implants with antibacterial properties have gradually increased. This review summarizes the recent advances in biodegradable magnesium- (Mg-), iron- (Fe-), and zinc- (Zn-) based alloys with antibacterial properties as orthopedic implant materials. The antibacterial mechanisms of these alloy materials are also outlined, thus providing more basis and insights on the design and application of biodegradable alloys with antibacterial properties as orthopedic implants.
Collapse
Affiliation(s)
- Ning Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yutong Ma
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Huixin Shi
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Yiping Song
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Shu Guo, ; Shude Yang,
| | - Shude Yang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, School of Stomatology and Department of Oral Pathology, School of Stomatology, China Medical University, Shenyang, China
- *Correspondence: Shu Guo, ; Shude Yang,
| |
Collapse
|
37
|
Quorum quenching of Streptococcus mutans via the nano-quercetin-based antimicrobial photodynamic therapy as a potential target for cariogenic biofilm. BMC Microbiol 2022; 22:125. [PMID: 35538403 PMCID: PMC9088123 DOI: 10.1186/s12866-022-02544-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Background Quorum sensing (QS) system can regulate the expression of virulence factors and biofilm formation in Streptococcus mutans. Antimicrobial photodynamic therapy (aPDT) inhibits quorum quenching (QQ), and can be used to prevent microbial biofilm. We thereby aimed to evaluate the anti-biofilm potency and anti-metabolic activity of nano-quercetin (N-QCT)-mediated aPDT against S. mutans. Also, in silico evaluation of the inhibitory effect of N-QCT on the competence-stimulating peptide (CSP) of S. mutans was performed to elucidate the impact of aPDT on various QS-regulated genes. Methods Cytotoxicity and intracellular reactive oxygen species (ROS) generation were assessed following synthesis and confirmation of N-QCT. Subsequently, the minimum biofilm inhibitory concentration (MBIC) of N-QCT against S. mutans and anti-biofilm effects of aPDT were assessed using colorimetric assay and plate counting. Molecular modeling and docking analysis were performed to confirm the connection of QCT to CSP. The metabolic activity of S. mutans and the expression level of various genes involved in QS were evaluated by flow cytometry and reverse transcription quantitative real-time PCR, respectively. Results Successful synthesis of non-toxic N-QCT was confirmed through several characterization tests. The MBIC value of N-QCT against S. mutans was 128 μg/mL. Similar to the crystal violet staining, the results log10 CFU/mL showed a significant degradation of preformed biofilms in the group treated with aPDT compared to the control group (P < 0.05). Following aPDT, metabolic activity of S. mutans also decreased by 85.7% (1/2 × MBIC of N-QCT) and 77.3% (1/4 × MBIC of N-QCT), as compared to the control values (P < 0.05). In silico analysis showed that the QCT molecule was located in the site formed by polypeptide helices of CSP. The relative expression levels of the virulence genes were significantly decreased in the presence of N-QCT-mediated aPDT (P < 0.05). Conclusions The combination of N-QCT with blue laser as a QQ-strategy leads to maximum ROS generation, disrupts the microbial biofilm of S. mutans, reduces metabolic activity, and downregulates the expression of genes involved in the QS pathway by targeting genes of the QS signaling system of S. mutans.
Collapse
|
38
|
Nadar S, Khan T, Patching SG, Omri A. Development of Antibiofilm Therapeutics Strategies to Overcome Antimicrobial Drug Resistance. Microorganisms 2022; 10:microorganisms10020303. [PMID: 35208758 PMCID: PMC8879831 DOI: 10.3390/microorganisms10020303] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
A biofilm is a community of stable microorganisms encapsulated in an extracellular matrix produced by themselves. Many types of microorganisms that are found on living hosts or in the environment can form biofilms. These include pathogenic bacteria that can serve as a reservoir for persistent infections, and are culpable for leading to a broad spectrum of chronic illnesses and emergence of antibiotic resistance making them difficult to be treated. The absence of biofilm-targeting antibiotics in the drug discovery pipeline indicates an unmet opportunity for designing new biofilm inhibitors as antimicrobial agents using various strategies and targeting distinct stages of biofilm formation. The strategies available to control biofilm formation include targeting the enzymes and proteins specific to the microorganism and those involved in the adhesion pathways leading to formation of resistant biofilms. This review primarily focuses on the recent strategies and advances responsible for identifying a myriad of antibiofilm agents and their mechanism of biofilm inhibition, including extracellular polymeric substance synthesis inhibitors, adhesion inhibitors, quorum sensing inhibitors, efflux pump inhibitors, and cyclic diguanylate inhibitors. Furthermore, we present the structure–activity relationships (SAR) of these agents, including recently discovered biofilm inhibitors, nature-derived bioactive scaffolds, synthetic small molecules, antimicrobial peptides, bioactive compounds isolated from fungi, non-proteinogenic amino acids and antibiotics. We hope to fuel interest and focus research efforts on the development of agents targeting the uniquely complex, physical and chemical heterogeneous biofilms through a multipronged approach and combinatorial therapeutics for a more effective control and management of biofilms across diseases.
Collapse
Affiliation(s)
- Sahaya Nadar
- Department of Pharmaceutical Chemistry, St. John Institute of Pharmacy and Research, Mumbai 400056, India;
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry & Quality Assurance, SVKM’s Dr. Bhanuben Nanavati College of Pharmacy, Mumbai 400056, India;
| | - Simon G. Patching
- School of Biomedical Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
- Correspondence: or (S.G.P.); (A.O.)
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, ON P3E 2C6, Canada
- Correspondence: or (S.G.P.); (A.O.)
| |
Collapse
|
39
|
Enrichment of Burkholderia in the Rhizosphere by Autotoxic Ginsenosides to Alleviate Negative Plant-Soil Feedback. Microbiol Spectr 2021; 9:e0140021. [PMID: 34756064 PMCID: PMC8579924 DOI: 10.1128/spectrum.01400-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The accumulation of autotoxins and soilborne pathogens in soil was shown to be the primary driver of negative plant-soil feedback (NPSF). There is a concerted understanding that plants could enhance their adaptability to biotic or abiotic stress by modifying the rhizosphere microbiome. However, it is not clear whether autotoxins could enrich microbes to degrade themselves or antagonize soilborne pathogens. Here, we found that the microbiome degraded autotoxic ginsenosides, belonging to triterpenoid glycosides, and antagonized pathogens in the rhizosphere soil of Panax notoginseng (sanqi). Deep analysis by 16S rRNA sequencing showed that the bacterial community was obviously changed in the rhizosphere soil and identified the Burkholderia-Caballeronia-Paraburkholderia (BCP) group as the main ginsenoside-enriched bacteria in the rhizosphere soil. Eight strains belonging to the BCP group were isolated, and Burkholderia isolate B36 showed a high ability to simultaneously degrade autotoxic ginsenosides (Rb1, Rg1, and Rd) and antagonize the soilborne pathogen Ilyonectria destructans. Interestingly, ginsenosides could stimulate the growth and biofilm formation of B36, eventually enhancing the antagonistic ability of B36 to I. destructans and the colonization ability in the rhizosphere soil. In summary, autotoxic ginsenosides secreted by P. notoginseng could enrich beneficial microbes in the rhizosphere to simultaneously degrade autotoxins and antagonize pathogen, providing a novel ecological strategy to alleviate NPSF. IMPORTANCE Autotoxic ginsenosides, secreted by sanqi into soil, could enrich Burkholderia sp. to alleviate negative plant-soil feedback (NPSF) by degrading autotoxins and antagonizing the root rot pathogen. In detail, ginsenosides could stimulate the growth and biofilm formation of Burkholderia sp. B36, eventually enhancing the antagonistic ability of Burkholderia sp. B36 to a soilborne pathogen and the colonization of B36 in soil. This ecological strategy could alleviate NPSF by manipulating the rhizosphere microbiome to simultaneously degrade autotoxins and antagonize pathogen.
Collapse
|
40
|
Lee D, Im J, Park DH, Jeong S, Park M, Yoon S, Park J, Han SH. Lactobacillus plantarum Lipoteichoic Acids Possess Strain-Specific Regulatory Effects on the Biofilm Formation of Dental Pathogenic Bacteria. Front Microbiol 2021; 12:758161. [PMID: 34867884 PMCID: PMC8636137 DOI: 10.3389/fmicb.2021.758161] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Bacterial biofilm residing in the oral cavity is closely related to the initiation and persistence of various dental diseases. Previously, we reported the anti-biofilm activity of Lactobacillus plantarum lipoteichoic acid (Lp.LTA) on a representative dental cariogenic pathogen, Streptococcus mutans. Since LTA structure varies in a bacterial strain-specific manner, LTAs from various L. plantarum strains may have differential anti-biofilm activity due to their distinct molecular structures. In the present study, we isolated Lp.LTAs from four different strains of L. plantarum (LRCC 5193, 5194, 5195, and 5310) and compared their anti-biofilm effects on the dental pathogens, including S. mutans, Enterococcus faecalis, and Streptococcus gordonii. All Lp.LTAs similarly inhibited E. faecalis biofilm formation in a dose-dependent manner. However, their effects on S. gordonii and S. mutans biofilm formation were different: LRCC 5310 Lp.LTA most effectively suppressed the biofilm formation of all strains of dental pathogens, while Lp.LTAs from LRCC 5193 and 5194 hardly inhibited or even enhanced the biofilm formation. Furthermore, LRCC 5310 Lp.LTA dramatically reduced the biofilm formation of the dental pathogens on the human dentin slice infection model. Collectively, these results suggest that Lp.LTAs have strain-specific regulatory effects on biofilm formation of dental pathogens and LRCC 5310 Lp.LTA can be used as an effective anti-biofilm agent for the prevention of dental infectious diseases.
Collapse
Affiliation(s)
- Dongwook Lee
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Dong Hyun Park
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Sungho Jeong
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Miri Park
- Bio Research Team, Lotte R&D Center, Seoul, South Korea
| | - Seokmin Yoon
- Bio Research Team, Lotte R&D Center, Seoul, South Korea
| | - Jaewoong Park
- Bio Research Team, Lotte R&D Center, Seoul, South Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and DRI, School of Dentistry, Seoul National University, Seoul, South Korea
| |
Collapse
|
41
|
Park T, Im J, Kim AR, Lee D, Jeong S, Yun CH, Han SH. Short-chain fatty acids inhibit the biofilm formation of Streptococcus gordonii through negative regulation of competence-stimulating peptide signaling pathway. J Microbiol 2021; 59:1142-1149. [PMID: 34865199 DOI: 10.1007/s12275-021-1576-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022]
Abstract
Streptococcus gordonii, a Gram-positive commensal bacterium, is an opportunistic pathogen closely related to initiation and progression of various oral diseases, such as periodontitis and dental caries. Its biofilm formation is linked with the development of such diseases by enhanced resistance against antimicrobial treatment or host immunity. In the present study, we investigated the effect of short-chain fatty acids (SCFAs) on the biofilm formation of S. gordonii. SCFAs, including sodium acetate (NaA), sodium propionate (NaP), and sodium butyrate (NaB), showed an effective inhibitory activity on the biofilm formation of S. gordonii without reduction in bacterial growth. SCFAs suppressed S. gordonii biofilm formation at early time points whereas SCFAs did not affect its preformed biofilm. A quorum-sensing system mediated by competence-stimulating peptide (CSP) is known to regulate biofilm formation of streptococci. Interestingly, SCFAs substantially decreased mRNA expression of comD and comE, which are CSP-sensor and its response regulator responsible for CSP pathway, respectively. Although S. gordonii biofilm formation was enhanced by exogenous synthetic CSP treatment, such effect was not observed in the presence of SCFAs. Collectively, these results suggest that SCFAs have an anti-biofilm activity on S. gordonii through inhibiting comD and comE expression which results in negative regulation of CSP quorum-sensing system. SCFAs could be an effective anti-biofilm agent against S. gordonii for the prevention of oral diseases.
Collapse
Affiliation(s)
- Taehwan Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - A Reum Kim
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sungho Jeong
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, 25354, Republic of Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
42
|
Puccio T, An SS, Schultz AC, Lizarraga CA, Bryant AS, Culp DJ, Burne RA, Kitten T. Manganese transport by Streptococcus sanguinis in acidic conditions and its impact on growth in vitro and in vivo. Mol Microbiol 2021; 117:375-393. [PMID: 34862691 PMCID: PMC8844241 DOI: 10.1111/mmi.14854] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023]
Abstract
Streptococcus sanguinis is an oral commensal and an etiological agent of infective endocarditis. Previous studies have identified the SsaACB manganese transporter as essential for endocarditis virulence; however, the significance of SsaACB in the oral environment has never been examined. Here we report that a ΔssaACB deletion mutant of strain SK36 exhibits reduced growth and manganese uptake under acidic conditions. Further studies revealed that these deficits resulted from the decreased activity of TmpA, shown in the accompanying paper to function as a ZIP‐family manganese transporter. Transcriptomic analysis of fermentor‐grown cultures of SK36 WT and ΔssaACB strains identified pH‐dependent changes related to carbon catabolite repression in both strains, though their magnitude was generally greater in the mutant. In strain VMC66, which possesses a MntH transporter, loss of SsaACB did not significantly alter growth or cellular manganese levels under the same conditions. Interestingly, there were only modest differences between SK36 and its ΔssaACB mutant in competition with Streptococcus mutans in vitro and in a murine oral colonization model. Our results suggest that the heterogeneity of the oral environment may provide a rationale for the variety of manganese transporters found in S. sanguinis.
Collapse
Affiliation(s)
- Tanya Puccio
- Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| | - Seon-Sook An
- Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| | - Alexander C Schultz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Claudia A Lizarraga
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Ashley S Bryant
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - David J Culp
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Robert A Burne
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, Florida, USA
| | - Todd Kitten
- Philips Institute for Oral Health Research, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| |
Collapse
|
43
|
Mayumi S, Kuboniwa M, Sakanaka A, Hashino E, Ishikawa A, Ijima Y, Amano A. Potential of Prebiotic D-Tagatose for Prevention of Oral Disease. Front Cell Infect Microbiol 2021; 11:767944. [PMID: 34804997 PMCID: PMC8604381 DOI: 10.3389/fcimb.2021.767944] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Recent studies have shown phenotypic and metabolic heterogeneity in related species including Streptococcus oralis, a typical oral commensal bacterium, Streptococcus mutans, a cariogenic bacterium, and Streptococcus gordonii, which functions as an accessory pathogen in periodontopathic biofilm. In this study, metabolites characteristically contained in the saliva of individuals with good oral hygiene were determined, after which the effects of an identified prebiotic candidate, D-tagatose, on phenotype, gene expression, and metabolic profiles of those three key bacterial species were investigated. Examinations of the saliva metabolome of 18 systemically healthy volunteers identified salivary D-tagatose as associated with lower dental biofilm abundance in the oral cavity (Spearman’s correlation coefficient; r = -0.603, p = 0.008), then the effects of D-tagatose on oral streptococci were analyzed in vitro. In chemically defined medium (CDM) containing D-tagatose as the sole carbohydrate source, S. mutans and S. gordonii each showed negligible biofilm formation, whereas significant biofilms were formed in cultures of S. oralis. Furthermore, even in the presence of glucose, S. mutans and S. gordonii showed growth suppression and decreases in the final viable cell count in a D-tagatose concentration-dependent manner. In contrast, no inhibitory effects of D-tagatose on the growth of S. oralis were observed. To investigate species-specific inhibition by D-tagatose, the metabolomic profiles of D-tagatose-treated S. mutans, S. gordonii, and S. oralis cells were examined. The intracellular amounts of pyruvate-derived amino acids in S. mutans and S. gordonii, but not in S. oralis, such as branched-chain amino acids and alanine, tended to decrease in the presence of D-tagatose. This phenomenon indicates that D-tagatose inhibits growth of those bacteria by affecting glycolysis and its downstream metabolism. In conclusion, the present study provides evidence that D-tagatose is abundant in saliva of individuals with good oral health. Additionally, experimental results demonstrated that D-tagatose selectively inhibits growth of the oral pathogens S. mutans and S. gordonii. In contrast, the oral commensal S. oralis seemed to be negligibly affected, thus highlighting the potential of administration of D-tagatose as an oral prebiotic for its ability to manipulate the metabolism of those targeted oral streptococci.
Collapse
Affiliation(s)
- Shota Mayumi
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Akito Sakanaka
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Ei Hashino
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Asuka Ishikawa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Yura Ijima
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Atsuo Amano
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Japan
| |
Collapse
|
44
|
Novel Virulent Bacteriophage ΦSG005, Which Infects Streptococcus gordonii, Forms a Distinct Clade among Streptococcus Viruses. Viruses 2021; 13:v13101964. [PMID: 34696394 PMCID: PMC8537203 DOI: 10.3390/v13101964] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/14/2021] [Accepted: 09/25/2021] [Indexed: 11/16/2022] Open
Abstract
Bacteriophages are viruses that specifically infect bacteria and are classified as either virulent phages or temperate phages. Despite virulent phages being promising antimicrobial agents due to their bactericidal effects, the implementation of phage therapy depends on the availability of virulent phages against target bacteria. Notably, virulent phages of Streptococcus gordonii, which resides in the oral cavity and is an opportunistic pathogen that can cause periodontitis and endocarditis have previously never been found. We thus attempted to isolate virulent phages against S. gordonii. In the present study, we report for the first time a virulent bacteriophage against S. gordonii, ΦSG005, discovered from drainage water. ΦSG005 is composed of a short, non-contractile tail and a long head, revealing Podoviridae characteristics via electron microscopic analysis. In turbidity reduction assays, ΦSG005 showed efficient bactericidal effects on S. gordonii. Whole-genome sequencing showed that the virus has a DNA genome of 16,127 bp with 21 coding sequences. We identified no prophage-related elements such as integrase in the ΦSG005 genome, demonstrating that the virus is a virulent phage. Phylogenetic analysis indicated that ΦSG005 forms a distinct clade among the streptococcus viruses and is positioned next to streptococcus virus C1. Molecular characterization revealed the presence of an anti-CRISPR (Acr) IIA5-like protein in the ΦSG005 genome. These findings facilitate our understanding of streptococcus viruses and advance the development of phage therapy against S. gordonii infection.
Collapse
|
45
|
Sharma A, Vashistt J, Shrivastava R. Response surface modeling integrated microtiter plate assay for Mycobacterium fortuitum biofilm quantification. BIOFOULING 2021; 37:830-843. [PMID: 34503352 DOI: 10.1080/08927014.2021.1974846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 06/13/2023]
Abstract
In this study, the effects of agitation, temperature, and pH on biofilm formation by Mycobacterium fortuitum were studied and quantified through response surface modeling. The microtiter plate assay was optimized to achieve conditions favoring maximum mycobacterial biofilm quantification. Optical density (OD) measurement using a crystal violet assay was performed to estimate the amount of biofilm formed. Response surface methodology (RSM) results revealed an R2 value of 96.18%, exhibiting a maximum OD of 2.119 (λ570 nm) at a temperature of 37 °C and pH 7.0, under a static environment. The conditions were experimentally validated. Statistically significant results showed that the maximum biofilm was produced 96 h after mycobacterial inoculation. Thus, the results provide a basis for using RSM as an efficient optimization method for M. fortuitum biofilm assays. This approach can also be incorporated into strategies for screening anti-biofilm compounds, synthetic chemicals, drugs, or inhibitors against pathogenic mycobacteria.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan (H.P.), India
| | - Jitendraa Vashistt
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan (H.P.), India
| | - Rahul Shrivastava
- Department of Biotechnology & Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan (H.P.), India
| |
Collapse
|
46
|
Wang Y, Hoffmann JP, Baker SM, Bentrup KHZ, Wimley WC, Fuselier JA, Bitoun JP, Morici LA. Inhibition of Streptococcus mutans biofilms with bacterial-derived outer membrane vesicles. BMC Microbiol 2021; 21:234. [PMID: 34429066 PMCID: PMC8386047 DOI: 10.1186/s12866-021-02296-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Biofilms are microbial communities surrounded by a self-produced extracellular matrix which protects them from environmental stress. Bacteria within biofilms are 10- to 1000-fold more resistant to antibiotics, making it challenging but imperative to develop new therapeutics that can disperse biofilms and eradicate infection. Gram-negative bacteria produce outer membrane vesicles (OMV) that play critical roles in communication, genetic exchange, cargo delivery, and pathogenesis. We have previously shown that OMVs derived from Burkholderia thailandensis inhibit the growth of drug-sensitive and drug-resistant bacteria and fungi. RESULTS Here, we examine the antibiofilm activity of Burkholderia thailandensis OMVs against the oral biofilm-forming pathogen Streptococcus mutans. We demonstrate that OMV treatment reduces biofilm biomass, biofilm integrity, and bacterial cell viability. Both heat-labile and heat-stable components, including 4-hydroxy-3-methyl-2-(2-non-enyl)-quinoline and long-chain rhamnolipid, contribute to the antibiofilm activity of OMVs. When OMVs are co-administered with gentamicin, the efficacy of the antibiotic against S. mutans biofilms is enhanced. CONCLUSION These studies indicate that bacterial-derived OMVs are highly effective biological nanoparticles that can inhibit and potentially eradicate biofilms.
Collapse
Affiliation(s)
- Yihui Wang
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Joseph P. Hoffmann
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Sarah M. Baker
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Kerstin Höner zu Bentrup
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - William C. Wimley
- grid.265219.b0000 0001 2217 8588Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA USA
| | - Joseph A. Fuselier
- grid.265219.b0000 0001 2217 8588Department of Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Jacob P. Bitoun
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Lisa A. Morici
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| |
Collapse
|
47
|
Panyakorn T, Makeudom A, Kangvonkit P, Pattamapun K, Wanachantararak P, Charumanee S, Krisanaprakornkit S. Efficacy of double antibiotics in hydroxypropyl methylcellulose for bactericidal activity against Enterococcus faecalis and Streptococcus gordonii in biofilm. Arch Oral Biol 2021; 129:105210. [PMID: 34293645 DOI: 10.1016/j.archoralbio.2021.105210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 11/19/2022]
Abstract
OBJECTIVE This study aimed to investigate the efficacy of double antibiotics, including ciprofloxacin and metronidazole, in a new vehicle, hydroxypropyl methylcellulose (HPMC), against Enterococcus faecalis and Streptococcus gordonii grown in biofilm. DESIGN Human mandibular premolars were prepared and divided into four groups: (i) negative control, (ii) positive control, (iii) infected with E. faecalis and S. gordonii for 21 days and intracanally medicated with double antibiotics in HPMC, and (iv) infected with E. faecalis and S. gordonii for 21 days and intracanally medicated with calcium hydroxide (UltraCal™). The efficacy of medication for 14 or 28 days was determined by bacterial cultures and RT-qPCR for absolute quantities of E. faecalis and S. gordonii cDNA and for relative mRNA expressions of pbp5 and gtfG genes. RESULTS There were significant decreases in the mean colony forming units and mean cDNA amounts of E. faecalis and S. gordonii in group (iii) on days 14 and 28 compared to those in group (ii) (p < 0.01). However, the mean cDNA amounts of E. faecalis and S. gordonii in group (iv) were found to be significantly increased on day 28 (p < 0.05). The mRNA expression of gtfG was significantly decreased in groups (iii) and (iv) on days 14 and 28, whereas that of pbp5 was significantly increased in group (iv) on days 14 and 28 (p < 0.01). CONCLUSION Double antibiotics in HPMC gel showed an in vitro efficacy against E. faecalis and S. gordonii grown in biofilm, suggesting its clinical application as an intracanal medicament for both primary and persistent infections.
Collapse
Affiliation(s)
- Theeraphop Panyakorn
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Anupong Makeudom
- School of Dentistry, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Paisan Kangvonkit
- School of Dentistry, Mae Fah Luang University, Chiang Rai, 57100, Thailand
| | - Kassara Pattamapun
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | | | - Suporn Charumanee
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Suttichai Krisanaprakornkit
- Center of Excellence in Oral and Maxillofacial Biology, Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
48
|
Ellepola K, Huang X, Riley RP, Bitoun JP, Wen ZT. Streptococcus mutans Lacking sufCDSUB Is Viable, but Displays Major Defects in Growth, Stress Tolerance Responses and Biofilm Formation. Front Microbiol 2021; 12:671533. [PMID: 34248879 PMCID: PMC8264796 DOI: 10.3389/fmicb.2021.671533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Streptococcus mutans appears to possess a sole iron-sulfur (Fe-S) cluster biosynthesis system encoded by the sufCDSUB cluster. This study was designed to examine the role of sufCDSUB in S. mutans physiology. Allelic exchange mutants deficient of the whole sufCDSUB cluster and in individual genes were constructed. Compared to the wild-type, UA159, the sufCDSUB-deficient mutant, Δsuf::kanr, had a significantly reduced growth rate, especially in medium with the absence of isoleucine, leucine or glutamate/glutamine, amino acids that require Fe-S clusters for biosynthesis and when grown with medium adjusted to pH 6.0 and under oxidative and nitrosative stress conditions. Relative to UA159, Δsuf::kanr had major defects in stress tolerance responses with reduced survival rate of > 2-logs following incubation at low pH environment or after hydrogen peroxide challenge. When compared to UA159, Δsuf::kanr tended to form aggregates in broth medium and accumulated significantly less biofilm. As shown by luciferase reporter fusion assays, the expression of sufCDSUB was elevated by > 5.4-fold when the reporter strain was transferred from iron sufficient medium to iron-limiting medium. Oxidative stress induced by methyl viologen increased sufCDSUB expression by > 2-fold, and incubation in a low pH environment led to reduction of sufCDSUB expression by > 7-fold. These results suggest that lacking of SufCDSUB in S. mutans causes major defects in various cellular processes of the deficient mutant, including growth, stress tolerance responses and biofilm formation. In addition, the viability of the deficient mutant also suggests that SUF, the sole Fe-S cluster machinery identified is non-essential in S. mutans, which is not known in any other bacterium lacking the NIF and/or ISC system. However, how the bacterium compensates the Fe-S deficiency and if any novel Fe-S assembly systems exist in this bacterium await further investigation.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Xiaochang Huang
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Ryan P Riley
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jacob P Bitoun
- Department of Microbiology, Tulane University, New Orleans, LA, United States
| | - Zezhang Tom Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
49
|
Lahiri D, Nag M, Banerjee R, Mukherjee D, Garai S, Sarkar T, Dey A, Sheikh HI, Pathak SK, Edinur HA, Pati S, Ray RR. Amylases: Biofilm Inducer or Biofilm Inhibitor? Front Cell Infect Microbiol 2021; 11:660048. [PMID: 33987107 PMCID: PMC8112260 DOI: 10.3389/fcimb.2021.660048] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Biofilm is a syntrophic association of sessile groups of microbial cells that adhere to biotic and abiotic surfaces with the help of pili and extracellular polymeric substances (EPS). EPSs also prevent penetration of antimicrobials/antibiotics into the sessile groups of cells. Hence, methods and agents to avoid or remove biofilms are urgently needed. Enzymes play important roles in the removal of biofilm in natural environments and may be promising agents for this purpose. As the major component of the EPS is polysaccharide, amylase has inhibited EPS by preventing the adherence of the microbial cells, thus making amylase a suitable antimicrobial agent. On the other hand, salivary amylase binds to amylase-binding protein of plaque-forming Streptococci and initiates the formation of biofilm. This review investigates the contradictory actions and microbe-associated genes of amylases, with emphasis on their structural and functional characteristics.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Ritwik Banerjee
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Dipro Mukherjee
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Sayantani Garai
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Tanmay Sarkar
- Department of Food Technology and Bio-Chemical Engineering, Jadavpur University, Kolkata, India.,Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, India
| | - Ankita Dey
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| | - Hassan I Sheikh
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Malaysia
| | - Sushil Kumar Pathak
- Department of Bioscience and Bioinformatics, Khallikote University, Berhampur, India
| | | | - Siddhartha Pati
- Centre of Excellence, Khallikote University, Berhampur, India.,Research Division, Association for Biodiversity Conservation and Research (ABC), Balasore, India
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| |
Collapse
|
50
|
Mehla J, Liechti G, Morgenstein RM, Caufield JH, Hosseinnia A, Gagarinova A, Phanse S, Goodacre N, Brockett M, Sakhawalkar N, Babu M, Xiao R, Montelione GT, Vorobiev S, den Blaauwen T, Hunt JF, Uetz P. ZapG (YhcB/DUF1043), a novel cell division protein in gamma-proteobacteria linking the Z-ring to septal peptidoglycan synthesis. J Biol Chem 2021; 296:100700. [PMID: 33895137 PMCID: PMC8163987 DOI: 10.1016/j.jbc.2021.100700] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/14/2021] [Accepted: 04/21/2021] [Indexed: 01/26/2023] Open
Abstract
YhcB, a poorly understood protein conserved across gamma-proteobacteria, contains a domain of unknown function (DUF1043) and an N-terminal transmembrane domain. Here, we used an integrated approach including X-ray crystallography, genetics, and molecular biology to investigate the function and structure of YhcB. The Escherichia coli yhcB KO strain does not grow at 45 °C and is hypersensitive to cell wall–acting antibiotics, even in the stationary phase. The deletion of yhcB leads to filamentation, abnormal FtsZ ring formation, and aberrant septum development. The Z-ring is essential for the positioning of the septa and the initiation of cell division. We found that YhcB interacts with proteins of the divisome (e.g., FtsI, FtsQ) and elongasome (e.g., RodZ, RodA). Seven of these interactions are also conserved in Yersinia pestis and/or Vibrio cholerae. Furthermore, we mapped the amino acid residues likely involved in the interactions of YhcB with FtsI and RodZ. The 2.8 Å crystal structure of the cytosolic domain of Haemophilus ducreyi YhcB shows a unique tetrameric α-helical coiled-coil structure likely to be involved in linking the Z-ring to the septal peptidoglycan-synthesizing complexes. In summary, YhcB is a conserved and conditionally essential protein that plays a role in cell division and consequently affects envelope biogenesis. Based on these findings, we propose to rename YhcB to ZapG (Z-ring-associated protein G). This study will serve as a starting point for future studies on this protein family and on how cells transit from exponential to stationary survival.
Collapse
Affiliation(s)
- Jitender Mehla
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, USA.
| | - George Liechti
- Department of Microbiology and Immunology, Henry Jackson Foundation, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Randy M Morgenstein
- Department of Microbiology and Molecular Genetics, Oklahoma State University, Stillwater, Oklahoma, USA
| | - J Harry Caufield
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ali Hosseinnia
- Department of Biochemistry, Research and Innovation Centre, University of Regina, Regina, Saskatchewan, Canada
| | - Alla Gagarinova
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Sadhna Phanse
- Department of Biochemistry, Research and Innovation Centre, University of Regina, Regina, Saskatchewan, Canada
| | - Norman Goodacre
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mary Brockett
- Department of Microbiology and Immunology, Henry Jackson Foundation, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Neha Sakhawalkar
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Mohan Babu
- Department of Biochemistry, Research and Innovation Centre, University of Regina, Regina, Saskatchewan, Canada
| | - Rong Xiao
- Nexomics Biosciences Inc., Rocky Hill, New Jersey, USA; Department of Chemistry and Chemical Biology, and Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Gaetano T Montelione
- Department of Chemistry and Chemical Biology, and Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Chemistry and Chemical Biology, Rensselaer Polytechnic Institute, Troy, New York, USA
| | - Sergey Vorobiev
- Department of Chemistry and Chemical Biology, and Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Tanneke den Blaauwen
- Bacterial Cell Biology & Physiology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - John F Hunt
- Department of Chemistry and Chemical Biology, and Center for Biotechnology and Interdisciplinary Sciences, Rensselaer Polytechnic Institute, Troy, New York, USA; Department of Biological Sciences, Columbia University, New York, New York, USA
| | - Peter Uetz
- Center for the Study of Biological Complexity, Virginia Commonwealth University, Richmond, Virginia, USA.
| |
Collapse
|