1
|
Sangavi R, Malligarjunan N, Satish L, Raja V, Pandian SK, Gowrishankar S. Anticariogenic activity of marine brown algae Padina boergesenii and its active components towards Streptococcus mutans. Front Cell Infect Microbiol 2024; 14:1458825. [PMID: 39654980 PMCID: PMC11625749 DOI: 10.3389/fcimb.2024.1458825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Streptococcus mutans is a well-recognized bacterium that plays a predominant role in the progression of dental caries. Its pathogenicity is linked to several key characteristics, including the ability to produce organic acids (acidogenicity), thrive in low pH environments (aciduricity), synthesize exopolysaccharides (EPS) via glucosyltransferases, and form retentive biofilms. The treatment of dental caries with conventional antibiotics is often ineffective due to the bacterium's capacity to form recalcitrant biofilms. To address these challenges, strategies that specifically target the pathogen's virulence without affecting its viability have emerged as promising alternatives. In this context, we investigated the anticariogenic properties of the methanolic extract of Padina boergesenii (MEPB). MEPB demonstrated substantial, dose-dependent antibiofilm activity, with a maximum inhibition of 93% at 128 μg/mL, without compromising the viability of S. mutans. Anti-virulence assays using sub-MIC (minimum inhibitory concentration) levels of MEPB showed significant reductions in key virulence factors: 75% reduction in sucrose-dependent adherence, 65% reduction in sucrose-independent adherence, along with notable decreases in acid production, acid tolerance, and water-insoluble (85%) and water-soluble (52%) glucan synthesis. Additionally, MEPB significantly reduced cell surface hydrophobicity (55%) and extracellular DNA (eDNA) production (64%). qPCR analysis corroborated these in vitro findings, revealing that MEPB suppresses the expression of genes involved in S. mutans virulence, particularly genes related to EPS synthesis (gtfB, gtfC & gtfD) biofilm formation(gbpB & gbpC) and two-component regulatory system (vicR) were downregulated. Toxicity testing on human buccal epithelial cells confirmed the non-toxic nature of MEPB, suggesting its safety for potential therapeutic use. Furthermore, GC-MS/MS analysis identified palmitic acid, myristic acid, and stearic acid as the major active constituents of the MEPB extract. Subsequent biofilm inhibitory assays confirmed the potent antibiofilm efficacy of these compounds: palmitic acid (85%), myristic acid (72%) and stearic acid (83%). In conclusion, this study identifies P. boergesenii and its active biomolecules as potential anticariogenic agents, offering an alternative approach to combat dental caries by targeting bacterial virulence mechanisms rather than viability.
Collapse
Affiliation(s)
| | | | - Lakkakula Satish
- Applied Phycology and Biotechnology Division, Marine Algal Research Station, CSIR-Central Salt & Marine Chemical Research Institute, Mandapam, India
| | - Veerapandian Raja
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | | | | |
Collapse
|
2
|
do Amaral COF, Kantovitiz KR, de Araújo VC, Marega T, Teixeira LN, Martinez EF. Assessment of dental and periodontal indices and Streptococcus mutans virulence in fragile X syndrome patients. JOURNAL OF INTELLECTUAL DISABILITY RESEARCH : JIDR 2024; 68:1026-1035. [PMID: 38717133 DOI: 10.1111/jir.13142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/16/2024] [Accepted: 04/11/2024] [Indexed: 08/23/2024]
Abstract
INTRODUCTION Fragile X syndrome (FXS) is the most common cause of hereditary genetic disorder in a single gene characterised by intellectual disability. Behavioural features such as autism, hyperactivity and anxiety disorder may be present. Biofilm development and pathogenicity of Streptococcus mutans may be altered because FXS renders the dental approach and oral hygiene more complex. OBJECTIVES The purpose of this study was to compare the levels of transcripts for VicRK and CovR of S. mutans isolated from FXS patients with the levels of transcripts for VicRK and CovR of standard strain ATCC, using a quantitative polymerase chain reaction (qPCR). METHODS The caries experience index was assessed by the International Caries Detection and Assessment System (ICDAS), Periodontal Condition Index (PCI) and Invasive Dental Treatment Need Index (INI). RESULTS The clinical index findings revealed a high rate of caries cavities and bleeding on probing of FXS patients. When VicRK and CovR transcript levels were compared with the reference strain, Fragile X patients were found to have significantly higher values. CONCLUSION The present study demonstrated that FXS patients have more adverse clinical conditions, with increased biofilm accumulation and virulence. When combined with behavioural abnormalities, these patients become even more vulnerable to dental caries.
Collapse
Affiliation(s)
- Cristhiane Olivia Ferreira do Amaral
- Division of Special Care, Faculdade São Leopoldo Mandic, Campinas, Brazil
- Department of Special Care Dentistry, Dental School, Universidade do Oeste Paulista - UNOESTE, Presidente Prudente, Brazil
| | | | - V C de Araújo
- Division of Oral Pathology, Faculdade São Leopoldo Mandic, Campinas, Brazil
| | - T Marega
- Division of Special Care, Faculdade São Leopoldo Mandic, Campinas, Brazil
| | - L N Teixeira
- Division of Oral Pathology, Faculdade São Leopoldo Mandic, Campinas, Brazil
| | - E F Martinez
- Division of Cell Biology, Faculdade São Leopoldo Mandic, Campinas, Brazil
| |
Collapse
|
3
|
Mattos-Graner RO, Klein MI, Alves LA. The complement system as a key modulator of the oral microbiome in health and disease. Crit Rev Microbiol 2024; 50:138-167. [PMID: 36622855 DOI: 10.1080/1040841x.2022.2163614] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/10/2023]
Abstract
In this review, we address the interplay between the complement system and host microbiomes in health and disease, focussing on oral bacteria known to contribute to homeostasis or to promote dysbiosis associated with dental caries and periodontal diseases. Host proteins modulating complement activities in the oral environment and expression profiles of complement proteins in oral tissues were described. In addition, we highlight a sub-set of bacterial proteins involved in complement evasion and/or dysregulation previously characterized in pathogenic species (or strains), but further conserved among prototypical commensal species of the oral microbiome. Potential roles of these proteins in host-microbiome homeostasis and in the emergence of commensal strain lineages with increased virulence were also addressed. Finally, we provide examples of how commensal bacteria might exploit the complement system in competitive or cooperative interactions within the complex microbial communities of oral biofilms. These issues highlight the need for studies investigating the effects of the complement system on bacterial behaviour and competitiveness during their complex interactions within oral and extra-oral host sites.
Collapse
Affiliation(s)
- Renata O Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Marlise I Klein
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
| | - Lívia Araújo Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Sao Paulo, Brazil
- School of Dentistry, Cruzeiro do Sul University (UNICSUL), Sao Paulo, Brazil
| |
Collapse
|
4
|
Wang X, Wang S, Yuan L, Liang Z, Zhang X, Lin D, Hu X. Influence of adhesion force on croRS gene expression and antibiotic resistance of Enterococcus faecalis. J Biomed Mater Res A 2024; 112:44-52. [PMID: 37695122 DOI: 10.1002/jbm.a.37610] [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: 03/19/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Surface properties of materials influence bacterial adhesion and play important roles in biofilm antibiotic resistance. The two-component system CroRS of Enterococcus faecalis (E. faecalis) can be activated by antibiotics and is involved in cephalosporin resistance. We hypothesized that surfaces properties could influence the expression of croRS in E. faecalis biofilm and contribute to cephalosporin resistance. In this study, the hydrophobicity of poly-ethylene (PE) and stainless steel (SS) was characterized. Adhesion forces were measured using atomic force microscopy. The transcript levels of croRS in E. faecalis adhering to surfaces exerting different adhesion forces were compared, in presence and absence of cephalosporin. The ceftriaxone susceptibility of E. faecalis biofilms was investigated using colony forming units (CFU) counting. The water contact angles of PE and SS were 97.1 ± 0.3° and 33.5 ± 0.3°, respectively (p < .05). The adhesion force of E. faecalis on PE was 7.6 ± 1.0 nN, which was higher than that on SS surfaces (3.5 ± 0.5 nN, p < .05). The gene expression of croS and croR in E. faecalis was higher on PE compared to that on SS (p < .05). E. faecalis on the hydrophobic PE surfaces, exerting stronger adhesion force, was more resistant to ceftriaxone compared to that on more hydrophilic SS surfaces. Results revealed the surface properties of materials can modulate the expression of croRS system and interfere with the outcome of antimicrobial therapy of E. faecalis biofilm. The modification of surface properties of biomedical devices may be used as a strategy to increase the efficacy of antimicrobial therapy.
Collapse
Affiliation(s)
- Xue Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Siyu Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lingling Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhihua Liang
- Guangdong Provincial Key Laboratory of Magnetoelectric Physics and Devices, School of Physics, Sun Yat-sen University, Guangzhou, China
- Centre for Physical Mechanics and Biophysics, School of Physics, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyue Zhang
- Guangdong Provincial Key Laboratory of Magnetoelectric Physics and Devices, School of Physics, Sun Yat-sen University, Guangzhou, China
- Centre for Physical Mechanics and Biophysics, School of Physics, Sun Yat-sen University, Guangzhou, China
| | - Danle Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaoli Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Zuber P, Kreth J. Aspects of oral streptococcal metabolic diversity: Imagining the landscape beneath the fog. Mol Microbiol 2023; 120:508-524. [PMID: 37329112 DOI: 10.1111/mmi.15106] [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: 03/15/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023]
Abstract
It is widely acknowledged that the human-associated microbial community influences host physiology, systemic health, disease progression, and even behavior. There is currently an increased interest in the oral microbiome, which occupies the entryway to much of what the human initially encounters from the environment. In addition to the dental pathology that results from a dysbiotic microbiome, microbial activity within the oral cavity exerts significant systemic effects. The composition and activity of the oral microbiome is influenced by (1) host-microbial interactions, (2) the emergence of niche-specific microbial "ecotypes," and (3) numerous microbe-microbe interactions, shaping the underlying microbial metabolic landscape. The oral streptococci are central players in the microbial activity ongoing in the oral cavity, due to their abundance and prevalence in the oral environment and the many interspecies interactions in which they participate. Streptococci are major determinants of a healthy homeostatic oral environment. The metabolic activities of oral Streptococci, particularly the metabolism involved in energy generation and regeneration of oxidative resources vary among the species and are important factors in niche-specific adaptations and intra-microbiome interactions. Here we summarize key differences among streptococcal central metabolic networks and species-specific differences in how the key glycolytic intermediates are utilized.
Collapse
Affiliation(s)
- Peter Zuber
- Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Jens Kreth
- School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
6
|
Dufour D, Li H, Gong SG, Lévesque CM. Transcriptome Analysis of Streptococcus mutans Quorum Sensing-Mediated Persisters Reveals an Enrichment in Genes Related to Stress Defense Mechanisms. Genes (Basel) 2023; 14:1887. [PMID: 37895236 PMCID: PMC10606796 DOI: 10.3390/genes14101887] [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: 09/01/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Persisters are a small fraction of growth-arrested phenotypic variants that can survive lethal concentrations of antibiotics but are able to resume growth once antibiotics are stopped. Their formation can be a stochastic process or one triggered by environmental cues. In the human pathogen Streptococcus mutans, the canonical peptide-based quorum-sensing system is an inducible DNA repair system that is pivotal for bacterial survival. Previous work has shown that the CSP-signaling peptide is a stress-signaling alarmone that promotes the formation of stress-induced persisters. In this study, we exposed S. mutans to the CSP pheromone to mimic DNA damage conditions and isolated the antibiotic persisters by treating the cultures with ofloxacin. A transcriptome analysis was then performed to evaluate the differential gene expression between the normal stationary-phase cells and the persisters. RNA sequencing revealed that triggered persistence was associated with the upregulation of genes related to several stress defense mechanisms, notably, multidrug efflux pumps, the arginine deaminase pathway, and the Opu/Opc system. In addition, we showed that inactivation of the VicK kinase of the YycFG essential two-component regulatory system abolished the formation of triggered persisters via the CSP pheromone. These data contribute to the understanding of the triggered persistence phenotype and may suggest new therapeutic strategies for treating persistent streptococcal infections.
Collapse
Affiliation(s)
| | | | | | - Céline M. Lévesque
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada; (D.D.); (H.L.); (S.-G.G.)
| |
Collapse
|
7
|
Drummond IY, DePaolo A, Krieger M, Driscoll H, Eckstrom K, Spatafora GA. Small regulatory RNAs are mediators of the Streptococcus mutans SloR regulon. J Bacteriol 2023; 205:e0017223. [PMID: 37695854 PMCID: PMC10521355 DOI: 10.1128/jb.00172-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/08/2023] [Indexed: 09/13/2023] Open
Abstract
Dental caries is among the most prevalent chronic diseases worldwide. Streptococcus mutans, the chief causative agent of caries, uses a 25-kDa manganese-dependent SloR protein to coordinate the uptake of essential manganese with the transcription of its virulence attributes. Small non-coding RNAs (sRNAs) can either enhance or repress gene expression, and reports in the literature ascribe an emerging role for sRNAs in the environmental stress response. Herein, we focused our attention on 18-50 nt sRNAs as mediators of the S. mutans SloR and manganese regulons. Specifically, the results of RNA sequencing revealed 19 sRNAs in S. mutans, which were differentially transcribed in the SloR-proficient UA159 and SloR-deficient GMS584 strains, and 10 sRNAs that were differentially expressed in UA159 cells grown in the presence of low vs high manganese. We describe SmsR1532 and SmsR1785 as SloR- and manganese-responsive sRNAs that are processed from large transcripts and that bind SloR directly in their promoter regions. The predicted targets of these sRNAs include regulators of metal ion transport, growth management via a toxin-antitoxin operon, and oxidative stress tolerance. These findings support a role for sRNAs in coordinating intracellular metal ion homeostasis with virulence gene control in an important oral cariogen. IMPORTANCE Small regulatory RNAs (sRNAs) are critical mediators of environmental signaling, particularly in bacterial cells under stress, but their role in Streptococcus mutans is poorly understood. S. mutans, the principal causative agent of dental caries, uses a 25-kDa manganese-dependent protein, called SloR, to coordinate the regulated uptake of essential metal ions with the transcription of its virulence genes. In the present study, we identified and characterized sRNAs that are both SloR and manganese responsive. Taken together, this research can elucidate the details of regulatory networks that engage sRNAs in an important oral pathogen and that can enable the development of an effective anti-caries therapeutic.
Collapse
Affiliation(s)
| | | | - Madeline Krieger
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Heather Driscoll
- Department of Biology, Vermont Biomedical Research Network, Norwich University, Northfield, Vermont, USA
| | - Korin Eckstrom
- Department of Microbiology and Molecular Genetics, The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, Vermont, USA
| | | |
Collapse
|
8
|
Zheng T, Jing M, Gong T, Yan J, Wang X, Xu M, Zhou X, Zeng J, Li Y. Regulatory mechanisms of exopolysaccharide synthesis and biofilm formation in Streptococcus mutans. J Oral Microbiol 2023; 15:2225257. [PMID: 37346997 PMCID: PMC10281425 DOI: 10.1080/20002297.2023.2225257] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Background Dental caries is a chronic, multifactorial and biofilm-mediated oral bacterial infection affecting almost every age group and every geographical region. Streptococcus mutans is considered an important pathogen responsible for the initiation and development of dental caries. It produces exopolysaccharides in situ to promote the colonization of cariogenic bacteria and coordinate dental biofilm development. Objective The understanding of the regulatory mechanism of S. mutans biofilm formation can provide a theoretical basis for the prevention and treatment of caries. Design At present, an increasing number of studies have identified many regulatory systems in S. mutans that regulate biofilm formation, including second messengers (e.g. c-di-AMP, Ap4A), transcription factors (e.g. EpsR, RcrR, StsR, AhrC, FruR), two-component systems (e.g. CovR, VicR), small RNA (including sRNA0426, srn92532, and srn133489), acetylation modifications (e.g. ActG), CRISPR-associated proteins (e.g. Cas3), PTS systems (e.g. EIIAB), quorum-sensing signaling system (e.g. LuxS), enzymes (including Dex, YidC, CopZ, EzrA, lmrB, SprV, RecA, PdxR, MurI) and small-molecule metabolites. Results This review summarizes the recent progress in the molecular regulatory mechanisms of exopolysaccharides synthesis and biofilm formation in S. mutans.
Collapse
Affiliation(s)
- Ting Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Meiling Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiangchuan Yan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaowan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mai Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Drummond IY, DePaolo A, Krieger M, Driscoll H, Eckstrom K, Spatafora GA. Small regulatory RNAs are mediators of the Streptococcus mutans SloR regulon. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543485. [PMID: 37398324 PMCID: PMC10312646 DOI: 10.1101/2023.06.02.543485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Dental caries is among the most prevalent chronic infectious diseases worldwide. Streptococcus mutans , the chief causative agent of caries, uses a 25 kDa manganese dependent SloR protein to coordinate the uptake of essential manganese with the transcription of its virulence attributes. Small non-coding RNAs (sRNAs) can either enhance or repress gene expression and reports in the literature ascribe an emerging role for sRNAs in the environmental stress response. Herein, we identify 18-50 nt sRNAs as mediators of the S. mutans SloR and manganese regulons. Specifically, the results of sRNA-seq revealed 56 sRNAs in S. mutans that were differentially transcribed in the SloR-proficient UA159 and SloR-deficient GMS584 strains, and 109 sRNAs that were differentially expressed in UA159 cells grown in the presence of low versus high manganese. We describe SmsR1532 and SmsR1785 as SloR- and/or manganese-responsive sRNAs that are processed from large transcripts, and that bind SloR directly in their promoter regions. The predicted targets of these sRNAs include regulators of metal ion transport, growth management via a toxin-antitoxin operon, and oxidative stress tolerance. These findings support a role for sRNAs in coordinating intracellular metal ion homeostasis with virulence gene control in an important oral cariogen. IMPORTANCE Small regulatory RNAs (sRNAs) are critical mediators of environmental signaling, particularly in bacterial cells under stress, but their role in Streptococcus mutans is poorly understood. S. mutans, the principal causative agent of dental caries, uses a 25 kDa manganese-dependent protein, called SloR, to coordinate the regulated uptake of essential metal ions with the transcription of its virulence genes. In the present study, we identified and characterize sRNAs that are both SloR- and manganese-responsive. Taken together, this research can elucidate the details of regulatory networks that engage sRNAs in an important oral pathogen, and that can enable the development of an effective anti-caries therapeutic.
Collapse
|
10
|
Yan J, Gong T, Ma Q, Zheng T, Chen J, Li J, Jing M, Lin Y, Wang X, Lei L, Wang S, Zeng J, Li Y. vicR overexpression in Streptococcus mutans causes aggregation and affects interspecies competition. Mol Oral Microbiol 2023; 38:224-236. [PMID: 36779415 DOI: 10.1111/omi.12407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/14/2023]
Abstract
Streptococcus mutans is considered to be a major causative agent of dental caries. VicRK is a two-component signal transduction system (TCSTS) of S. mutans, which can regulate the virulence of S. mutans, such as biofilm formation, exopolysaccharide production, acid production, and acid resistance. Meanwhile, it can also regulate the production of mutacins (nlmC) through the TCSTS ComDE. In this study, we found that the vicR-overexpressing strain was more likely to aggregate to form cell clusters, leading to the formation of abnormal biofilm; the overexpression of vicR increased the length of the chain of S. mutans. Furthermore, the expression of the mutacins in the vicR overexpression strain was increased under aerobic conditions. Compared with the control strain and the parental strain, the vicR overexpression strain was more competitive against Streptococcus gordonii. But there was no significant difference against Streptococcus sanguinis. In clinical strains, the expression level of vicR was positively correlated with their competitive ability against S. gordonii. Transcriptional profiling revealed 24 significantly upregulated genes in the vicR-overexpressing strain, including nlmA, nlmB, nlmC, and nlmD encoding mutacins. Electrophoretic mobility shift assays and DNase I footprinting assays confirmed that VicR can directly bind to the promoter sequence of nlmD. Taken together, our findings further demonstrate that VicRK, an important TCSTS of S. mutans, is involved in S. mutans cell morphology and biofilm formation. VicRK regulates the production of more mutacins in S. mutans in response to oxygen stimulation. VicR can bind to the promoter sequence of nlmD, thereby directly regulating the production of mutacins NlmD.
Collapse
Affiliation(s)
- Jiangchuan Yan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tao Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qizhao Ma
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ting Zheng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiamin Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Meiling Jing
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yongwang Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaowan Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shida Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Operative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
The Regulations of Essential WalRK Two-Component System on Enterococcus faecalis. J Clin Med 2023; 12:jcm12030767. [PMID: 36769415 PMCID: PMC9917794 DOI: 10.3390/jcm12030767] [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: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Enterococcus faecalis (E. faecalis) is a Gram-positive, facultative anaerobic bacterium that is highly adaptable to its environment. In humans, it can cause serious infections with biofilm formation. With increasing attention on its health threat, prevention and control of biofilm formation in E. faecalis have been observed. Many factors including polysaccharides as well as autolysis, proteases, and eDNA regulate biofilm formation. Those contributors are regulated by several important regulatory systems involving the two-component signal transduction system (TCS) for its adaptation to the environment. Highly conserved WalRK as one of 17 TCSs is the only essential TCS in E. faecalis. In addition to biofilm formation, various metabolisms, including cell wall construction, drug resistance, as well as interactions among regulatory systems and resistance to the host immune system, can be modulated by the WalRK system. Therefore, WalRK has been identified as a key target for E. faecalis infection control. In the present review, the regulation of WalRK on E. faecalis pathogenesis and associated therapeutic strategies are demonstrated.
Collapse
|
12
|
Chen R, Du M, Liu C. Strategies for dispersion of cariogenic biofilms: applications and mechanisms. Front Microbiol 2022; 13:981203. [PMID: 36134140 PMCID: PMC9484479 DOI: 10.3389/fmicb.2022.981203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/11/2022] [Indexed: 11/05/2022] Open
Abstract
Bacteria residing within biofilms are more resistant to drugs than planktonic bacteria. They can thus play a significant role in the onset of chronic infections. Dispersion of biofilms is a promising avenue for the treatment of biofilm-associated diseases, such as dental caries. In this review, we summarize strategies for dispersion of cariogenic biofilms, including biofilm environment, signaling pathways, biological therapies, and nanovehicle-based adjuvant strategies. The mechanisms behind these strategies have been discussed from the components of oral biofilm. In the future, these strategies may provide great opportunities for the clinical treatment of dental diseases. Graphical Abstract.
Collapse
|
13
|
Kong L, Su M, Sang J, Huang S, Wang M, Cai Y, Xie M, Wu J, Wang S, Foster SJ, Zhang J, Han A. The W-Acidic Motif of Histidine Kinase WalK Is Required for Signaling and Transcriptional Regulation in Streptococcus mutans. Front Microbiol 2022; 13:820089. [PMID: 35558126 PMCID: PMC9087282 DOI: 10.3389/fmicb.2022.820089] [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: 11/22/2021] [Accepted: 02/14/2022] [Indexed: 02/05/2023] Open
Abstract
In Streptococcus mutans, we find that the histidine kinase WalK possesses the longest C-terminal tail (CTT) among all 14 TCSs, and this tail plays a key role in the interaction of WalK with its response regulator WalR. We demonstrate that the intrinsically disordered CTT is characterized by a conserved tryptophan residue surrounded by acidic amino acids. Mutation in the tryptophan not only disrupts the stable interaction, but also impairs the efficient phosphotransferase and phosphatase activities of WalRK. In addition, the tryptophan is important for WalK to compete with DNA containing a WalR binding motif for the WalR interaction. We further show that the tryptophan is important for in vivo transcriptional regulation and bacterial biofilm formation by S. mutans. Moreover, Staphylococcus aureus WalK also has a characteristic CTT, albeit relatively shorter, with a conserved W-acidic motif, that is required for the WalRK interaction in vitro. Together, these data reveal that the W-acidic motif of WalK is indispensable for its interaction with WalR, thereby playing a key role in the WalRK-dependent signal transduction, transcriptional regulation and biofilm formation.
Collapse
Affiliation(s)
- Lingyuan Kong
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mingyang Su
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jiayan Sang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shanshan Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Min Wang
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Yongfei Cai
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Mingquan Xie
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Jun Wu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shida Wang
- State Key Laboratory for Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Simon J Foster
- Department of Molecular Biology and Biotechnology, The Florey Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Jiaqin Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Aidong Han
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
14
|
Zhang H, Xia M, Zhang B, Zhang Y, Chen H, Deng Y, Yang Y, Lei L, Hu T. Sucrose selectively regulates
Streptococcus mutans
polysaccharide by
GcrR. Environ Microbiol 2022; 24:1395-1410. [PMID: 35064734 DOI: 10.1111/1462-2920.15887] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/21/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Hongyu Zhang
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Mengying Xia
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Bin Zhang
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Yue Zhang
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Hong Chen
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Yalan Deng
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Yingming Yang
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Lei Lei
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| | - Tao Hu
- Department of Preventive Dentistry, West China School & Hospital of Stomatology Sichuan University, 14#, 3rd Section, Renmin South Road Chengdu Sichuan 610041 China
| |
Collapse
|
15
|
Chen Z, Xiong Y, Tang Y, Zhao Y, Chen J, Zheng J, Wu Y, Deng Q, Qu D, Yu Z. In vitro activities of thiazolidione derivatives combined with daptomycin against clinical Enterococcus faecium strains. BMC Microbiol 2022; 22:16. [PMID: 34996348 PMCID: PMC8740470 DOI: 10.1186/s12866-021-02423-8] [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] [Received: 06/17/2021] [Accepted: 12/06/2021] [Indexed: 11/19/2022] Open
Abstract
Background Previous reports have demonstrated two thiazolidione derivatives (H2-60 and H2-81) can robustly inhibit the planktonic growth and biofilm formation of S. epidermidis and S. aureus by targeting the histidine kinase YycG. Whereas the antibacterial and anti-biofilm activity of these two thiazolidione derivatives (H2-60 and H2-81) against Enterococcus faecium remains elusive. Here, the pET28a-YycG recombinant plasmid were in vitro expressed in E. coli competent cell BL21 (DE3) and induced to express YycG’ protein (conding HisKA and HATPase_c domain) by 0.5 mM IPTG and was purified by Ni – NTA agarose and then for the autophosphorylation test. Antimicrobial testing and time-killing assay were also be determined. Anti-biofilm activity of two derivatives with sub-MIC concentration towards positive biofilm producers of clinical E. faecium were detected using polystyrene microtiter plate and CLSM. Results The MICs of H2-60 and H2-81 in the clinical isolates of E. faecium were in the range from 3.125 mg/L to 25 mg/L. Moreover, either H2-60 or H2-81 showed the excellent bactericidal activity against E. faecium with monotherapy or its combination with daptomycin by time-killing assay. E. faecium planktonic cells can be decreased by H2-60 or H2-81 for more than 3 × log10 CFU/mL after 24 h treatment when combined with daptomycin. Furthermore, over 90% of E. faecium biofilm formation could markedly be inhibited by H2-60 and H2-81 at 1/4 × MIC value. In addition, the frequency of the eradicated viable cells embedded in mature biofilm were evaluated by the confocal laser microscopy, suggesting that of H2-60 combined with ampicillin or daptomycin was significantly high when compared with single treatment (78.17 and 74.48% vs. 41.59%, respectively, P < 0.01). Conclusion These two thiazolidione derivatives (H2-60 and H2-81) could directly impact the kinase phosphoration activity of YycG of E. faecium. H2-60 combined with daptomycin exhibit the excellent antibacterial and anti-biofilm activity against E. faecium by targeting YycG. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02423-8.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Yanpeng Xiong
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Yuanyuan Tang
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Yuxi Zhao
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Junwen Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China
| | - Qiwen Deng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, China.
| | - Zhijian Yu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518052, China.
| |
Collapse
|
16
|
Oliveira LT, Alves LA, Harth-Chu EN, Nomura R, Nakano K, Mattos-Graner RO. VicRK and CovR polymorphisms in Streptococcus mutans strains associated with cardiovascular infections. J Med Microbiol 2021; 70. [PMID: 34939562 DOI: 10.1099/jmm.0.001457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Introduction. Streptococcus mutans, a common species of the oral microbiome, expresses virulence genes promoting cariogenic dental biofilms, persistence in the bloodstream and cardiovascular infections.Gap statement. Virulence gene expression is variable among S. mutans strains and controlled by the transcription regulatory systems VicRK and CovR.Aim. This study investigates polymorphisms in the vicRK and covR loci in S. mutans strains isolated from the oral cavity or from the bloodstream, which were shown to differ in expression of covR, vicRK and downstream genes.Methodology. The transcriptional activities of covR, vicR and vicK were compared by RT-qPCR between blood and oral strains after exposure to human serum. PCR-amplified promoter and/or coding regions of covR and vicRK of 18 strains (11 oral and 7 blood) were sequenced and compared to the reference strain UA159.Results. Serum exposure significantly reduced covR and vicR/K transcript levels in most strains (P<0.05), but reductions were higher in oral than in blood strains. Single-nucleotide polymorphisms (SNPs) were detected in covR regulatory and coding regions, but SNPs affecting the CovR effector domain were only present in two blood strains. Although vicR was highly conserved, vicK showed several SNPs, and SNPs affecting VicK regions important for autokinase activity were found in three blood strains.Conclusions. This study reveals transcriptional and structural diversity in covR and vicR/K, and identifies polymorphisms of functional relevance in blood strains, indicating that covR and vicRK might be important loci for S. mutans adaptation to host selective pressures associated with virulence diversity.
Collapse
Affiliation(s)
- Letícia T Oliveira
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Lívia A Alves
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Erika N Harth-Chu
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| | - Ryota Nomura
- Department of Pediatric Dentistry, Osaka University, Graduate School of Dentistry, Osaka, Japan
| | - Kazuhiko Nakano
- Department of Pediatric Dentistry, Osaka University, Graduate School of Dentistry, Osaka, Japan
| | - Renata O Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School - State University of Campinas, Piracicaba, SP, Brazil
| |
Collapse
|
17
|
The vicK gene of Streptococcus mutans mediates its cariogenicity via exopolysaccharides metabolism. Int J Oral Sci 2021; 13:45. [PMID: 34916484 PMCID: PMC8677823 DOI: 10.1038/s41368-021-00149-x] [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: 12/08/2020] [Revised: 07/01/2021] [Accepted: 10/25/2021] [Indexed: 02/05/2023] Open
Abstract
Streptococcus mutans (S. mutans) is generally regarded as a major contributor to dental caries because of its ability to synthesize extracellular polysaccharides (EPS) that aid in the formation of plaque biofilm. The VicRKX system of S. mutans plays an important role in biofilm formation. The aim of this study was to investigate the effects of vicK gene on specific characteristics of EPS in S. mutans biofilm. We constructed single-species biofilms formed by different mutants of vicK gene. Production and distribution of EPS were detected through atomic force microscopy, scanning electron microscopy and confocal laser scanning microscopy. Microcosmic structures of EPS were analyzed by gel permeation chromatography and gas chromatography-mass spectrometry. Cariogenicity of the vicK mutant was assessed in a specific pathogen-free rat model. Transcriptional levels of cariogenicity-associated genes were confirmed by quantitative real-time polymerase chain reaction. The results showed that deletion of vicK gene suppressed biofilm formation as well as EPS production, and EPS were synthesized mostly around the cells. Molecular weight and monosaccharide components underwent evident alterations. Biofilms formed in vivo were sparse and contributed a decreased degree of caries. Moreover, expressional levels of genes related to EPS synthesis were down-regulated, except for gtfB. Our report demonstrates that vicK gene enhances biofilm formation and subsequent caries development. And this may due to its regulations on EPS metabolism, like synthesis or microcosmic features of EPS. This study suggests that vicK gene and EPS can be considered as promising targets to modulate dental caries.
Collapse
|
18
|
Katsumata T, Nguyen-Tra Le M, Kawada-Matsuo M, Taniguchi Y, Ouhara K, Oogai Y, Nakata M, Mizuno N, Nishitani Y, Komatsuzawa H. KATSUMATA et al.Comprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutansComprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutans. Microbiol Immunol 2021; 66:145-156. [PMID: 34888908 DOI: 10.1111/1348-0421.12958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
Streptococcus mutans, a cariogenic pathogen, adheres to the tooth surface and forms a biofilm. Bacterial cell surface proteins are associated with adherence to substrates. Sortase A (SrtA) mediates the localization of proteins with an LPXTG motif-containing proteins to the cell surface by covalent binding to peptidoglycan. In S. mutans UA159, 6 SrtA-dependent proteins, SpaP, WapA, WapE, DexA, FruA, and GbpC, were identified. Although some of these proteins were characterized, a comprehensive analysis of the 6 proteins has not been reported. In this study, we constructed mutants deficient in each of these proteins and the SrtA-deficient mutant. The SrtA-deficient mutant showed drastically decreased binding to salivary components, biofilm formation, bacterial coaggregation activity, hydrophobicity, and cellular matrix binding (collagen type I, fibronectin, and laminin). The SpaP-deficient mutant showed significantly reduced binding to salivary components and partially increased coaggregation with Porphyromonas gingivalis, and decreased hydrophobicity, and collagen binding. The WapA-deficient mutant showed slightly decreased coaggregation with Fusobacterium nucleatum. Although the SrtA-deficient mutant showed drastically altered phenotypes, all SrtA-dependent protein-deficient mutants, except the SpaP-deficient mutant, did not show considerable alterations in binding to salivary components. These results indicate that the 6 proteins may coordinately contribute to these activities. In addition, using genomic data of 125 S. mutans strains, we compared the amino acid sequences of each surface protein and found many variations among strains, which may affect the phenotype of cell surface proteins in S. mutans. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tamaki Katsumata
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuri Taniguchi
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
19
|
Kant S, Pancholi V. Novel Tyrosine Kinase-Mediated Phosphorylation With Dual Specificity Plays a Key Role in the Modulation of Streptococcus pyogenes Physiology and Virulence. Front Microbiol 2021; 12:689246. [PMID: 34950110 PMCID: PMC8689070 DOI: 10.3389/fmicb.2021.689246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 10/25/2021] [Indexed: 11/15/2022] Open
Abstract
Streptococcus pyogenes (Group A Streptococcus, GAS) genomes do not contain a gene encoding a typical bacterial-type tyrosine kinase (BY-kinase) but contain an orphan gene-encoding protein Tyr-phosphatase (SP-PTP). Hence, the importance of Tyr-phosphorylation is underappreciated and not recognized for its role in GAS pathophysiology and pathogenesis. The fact that SP-PTP dephosphorylates Abl-tyrosine kinase-phosphorylated myelin basic protein (MBP), and SP-STK (S. pyogenes Ser/Thr kinase) also autophosphorylates its Tyr101-residue prompted us to identify a putative tyrosine kinase and Tyr-phosphorylation in GAS. Upon a genome-wide search of kinases possessing a classical Walker motif, we identified a non-canonical tyrosine kinase M5005_Spy_1476, a ∼17 kDa protein (153 aa) (SP-TyK). The purified recombinant SP-TyK autophosphorylated in the presence of ATP. In vitro and in vivo phosphoproteomic analyses revealed two key phosphorylated tyrosine residues located within the catalytic domain of SP-TyK. An isogenic mutant lacking SP-TyK derived from the M1T1 strain showed a retarded growth pattern. It displayed defective cell division and long chains with multiple parallel septa, often resulting in aggregates. Transcriptomic analysis of the mutant revealed 287 differentially expressed genes responsible for GAS pathophysiology and pathogenesis. SP-TyK also phosphorylated GAS CovR, WalR, SP-STP, and SDH/GAPDH proteins with dual specificity targeting their Tyr/Ser/Thr residues as revealed by biochemical and mass-spectrometric-based phosphoproteomic analyses. SP-TyK-phosphorylated CovR bound to PcovR efficiently. The mutant displayed sustained release of IL-6 compared to TNF-α during co-culturing with A549 lung cell lines, attenuation in mice sepsis model, and significantly reduced ability to adhere to and invade A549 lung cells and form biofilms on abiotic surfaces. SP-TyK, thus, plays a critical role in fine-tuning the regulation of key cellular functions essential for GAS pathophysiology and pathogenesis through post-translational modifications and hence, may serve as a promising target for future therapeutic developments.
Collapse
|
20
|
Deng X, Zhang C, Chen J, Shi Y, Ma X, Wang Y, Wang Z, Yu Z, Zheng J, Chen Z. Antibacterial and anti-biofilm activities of histidine kinase YycG inhibitors against Streptococcus agalactiae. J Antibiot (Tokyo) 2021; 74:874-883. [PMID: 34489569 DOI: 10.1038/s41429-021-00475-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
This study aims to investigate the antibacterial and anti-biofilm activities of YycG inhibitors H2-60 and H2-81 against Streptococcus agalactiae. A total of 118 nonduplicate S. agalactiae clinical isolates were collected, and the minimal inhibitory concentrations (MICs) of H2-60 and H2-81 were determined. H2-60 and H2-81 inhibit biofilm formation of S. agalactiae were detected by crystal violet staining, and against established biofilms of S. agalactiae were observed by confocal laser scanning microscope. Inhibitory effect of H2-60 and H2-81 on the phosphorylation activity of the HisKA domain of YycG' protein was measured. The MIC50/MIC90 was 3.13/6.25 μM for H2-60 and 6.25/12.5 μM for H2-81 against S. agalactiae, respectively. S. agalactiae planktonic cells can be decreased by H2-60 or H2-81 for more than 3 × log10 CFU ml-1 after 24 h treatment. Biofilm formation of 8 S. agalactiae strains (strong biofilm producers) was significantly reduced after treated with 1/4 × MIC of H2-60 or H2-81 for 24 h. H2-60 and H2-81 could reduce 45.79% and 29.56% of the adherent cells in the established biofilm of S. agalactiae after 72 h treatment, respectively. H2-60 combined with daptomycin reduced 83.63% of the adherent cells in the established biofilm of S. agalactiae, which was significantly better than that of H2-60 (45.79%) or daptomycin (55.07%) alone. The half maximal inhibitory concentrations (IC50) were 35.6 μM for H2-60 and 46.3 μM for H2-81 against the HisKA domain of YycG' protein. In conclusion, YycG inhibitors H2-60 and H2-81 exhibit excellent antibacterial and anti-biofilm activities against S. agalactiae.
Collapse
Affiliation(s)
- Xiangbin Deng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Chaoqin Zhang
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Junwen Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yiyi Shi
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoyu Ma
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Yu Wang
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhanwen Wang
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Zhijian Yu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Quality Control Center of Hospital Infection management of Shenzhen, Guang Dong Medical University, Shenzhen, China
| | - Jinxin Zheng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China. .,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China. .,Quality Control Center of Hospital Infection management of Shenzhen, Guang Dong Medical University, Shenzhen, China.
| | - Zhong Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China. .,Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China. .,Quality Control Center of Hospital Infection management of Shenzhen, Guang Dong Medical University, Shenzhen, China.
| |
Collapse
|
21
|
Mao M, Zhang W, Huang Z, Huang J, Wang J, Li W, Gu S. Graphene Oxide-Copper Nanocomposites Suppress Cariogenic Streptococcus mutans Biofilm Formation. Int J Nanomedicine 2021; 16:7727-7739. [PMID: 34824531 PMCID: PMC8610231 DOI: 10.2147/ijn.s303521] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 10/26/2021] [Indexed: 01/22/2023] Open
Abstract
Introduction Dental caries is a biofilm-dependent disease that largely relies on the ability of Streptococcus mutans to synthesize exopolysaccharide matrix. Graphene oxide-based metal nanomaterials, as the derivatives of graphene, are potent agents against pathogens by their impressive antibacterial and anti-biofilm biofunctions. Previously, we fabricated the novel graphene oxide-copper nanocomposites (GO-Cu), maintaining a long-term release of copper nanoparticles. Here, the biofunctionalization of GO-Cu nanocomposites against cariogenic S. mutans is investigated. Methods Growth curve observation and colony forming units counting were applied to detect the antibacterial effect of GO-Cu nanocomposites on S. mutans. Scanning electron microscopy and the crystal violet assay were used to detect nanocomposite effects on biofilm forming ability. The production and distribution of exopolysaccharides within biofilm was analyzed and the expression of genes required for biofilm formation was explored. Moreover, the regulatory landscape of GO-Cu nanocomposites on S. mutans pathogenicity was probed. Results It has been found that GO-Gu nanocomposites were antibacterial to S. mutans and 10 μg/mL GO-Cu nanocomposites could inhibit the bacteria bioactivity instead of killing them. The biomass of S. mutans biofilm was significantly reduced when treated with 10 μg/mL GO-Cu nanocomposites. Also, 10 μg/mL GO-Cu nanocomposites could alter the biofilm architecture and impair exopolysaccharides production and distribution, and dysregulated the expression of exopolysaccharide-associated genes. Conclusion In all, we found low-dose GO-Cu nanocomposites could disrupt exopolysaccharide matrix assembly and further impair optimal biofilm development with minimal cytotoxicity. Therefore, GO-Cu nanocomposites can open up a new avenue for the development of alternative anti-caries biomaterials.
Collapse
Affiliation(s)
- Mengying Mao
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| | - Wenjie Zhang
- National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China.,Department of Prosthodontics, Oral Bioengineering and Regenerative Medicine Lab, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhengwei Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| | - Jing Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| | - Jia Wang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| | - Weiping Li
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| | - Shensheng Gu
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,National Clinical Research Center for Oral Diseases, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, People's Republic of China
| |
Collapse
|
22
|
Potassium iodide enhances inactivation of Streptococcus mutans biofilm in antimicrobial photodynamic therapy with red laser. Photodiagnosis Photodyn Ther 2021; 37:102622. [PMID: 34775066 DOI: 10.1016/j.pdpdt.2021.102622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/25/2021] [Accepted: 11/08/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To evaluate the effect of potassium iodide (KI) addition on antimicrobial photodynamic therapy (aPDT) mediated by red laser (λ = 660 nm) and methylene blue in Streptococcus mutans biofilm model. METHODS S. mutans biofilms were cultured in 96-well plates containing BHI broth with 1% sucrose for 18 h, 10% CO2 and 37°C and divided in groups (n = 3, in triplicate): C (NaCl 0.9%); CX (0.2% chlorhexidine); P (photosensitizer); KI (10, 25 and 50 mM); PKI (10, 25 and 50 mM); L (L 1: : 100 J/cm2, 9 J; L2: 200 J/cm2, 18 J); PL (photosensitizer + L1 or L2); KIL (KI at 10, 25 and 50 mM + L1 or L2); and PKIL (photosensitizer + 10, 25 and 50 mM KI + L1 or L2). Biofilms were submitted to three pre-irradiation (PI) times (5, 10, and 15 min). After the treatments, microbial counting's reduction was analyzed by Kruskal-Wallis and post-hoc Dunn's tests, respectively, and the interaction between light parameters and the PI times by two-way ANOVA (p < 0.05). RESULTS The S. mutans viability significantly reduced in all aPDT groups, in the presence or absence of KI (p < 0.05). For all PI times, PKIL groups (10, 25, and 50 mM) significantly differed from PL groups (p < 0.05) with a reduction of 9.0 logs reached at 50 mM of KI with 15 min of PI, irradiated at 18 J. We found no significant interaction between PI time and irradiation (p > 0.05). CONCLUSION Addition KI to TFDA mediated by methylene blue and red laser promoted an additional effect in reducing the microbial viability of S. mutans biofilm.
Collapse
|
23
|
Chen Z, Song K, Shang Y, Xiong Y, Lyu Z, Chen J, Zheng J, Li P, Wu Y, Gu C, Xie Y, Deng Q, Yu Z, Zhang J, Qu D. Selection and Identification of Novel Antibacterial Agents against Planktonic Growth and Biofilm Formation of Enterococcus faecalis. J Med Chem 2021; 64:15037-15052. [PMID: 34657423 DOI: 10.1021/acs.jmedchem.1c00939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
YycFG, one of the two-component systems involved in the regulation of biofilm formation, has attracted increasing interest as a potential target of antibacterial and antibiofilm agents. YycG inhibitors for Staphylococcus aureus and Staphylococcus epidermidis have been developed, but Enterococcus faecalis remains underexplored. Herein, we selected and identified novel candidate molecules against E. faecalis targeting histidine kinase YycG using high-throughput virtual screening; six molecules (compound-16, -30, -42, -46, -59, and -62) with low cytotoxicity toward mammalian cells were verified as potential YycG inhibitors through an autophosphorylation test and binding kinetics. Compound-16 inhibited planktonic cells of E. faecalis, including the vancomycin- or linezolid-resistant strains. In contrast, compound-62 did not affect planktonic growth but significantly inhibited biofilm formation in static and dynamic conditions. Compound-62 combined with ampicillin could synergistically eradicate the biofilm-embedded viable bacteria. The study demonstrates that YycG inhibitors may be valuable approaches for the development of novel antimicrobial agents for difficult-to-treat bacterial infections.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Kun Song
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yongpeng Shang
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Yanpeng Xiong
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Zhihui Lyu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Junwen Chen
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Peiyu Li
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Chenjian Gu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Youhua Xie
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Qiwen Deng
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases and the Key Lab of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China
| | - Jian Zhang
- Medicinal Bioinformatics Center, Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Department of Medical Microbiology and Parasitology, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China
| |
Collapse
|
24
|
Araújo IJDS, Ricardo MG, Gomes OP, Giovani PA, Puppin-Rontani J, Pecorari VA, Martinez EF, Napimoga MH, Nociti Junior FH, Puppin-Rontani RM, Lisboa-Filho PN, Kantovitz KR. Titanium dioxide nanotubes added to glass ionomer cements affect S. mutans viability and mechanisms of virulence. Braz Oral Res 2021; 35:e062. [PMID: 34133579 DOI: 10.1590/1807-3107bor-2021.vol35.0062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022] Open
Abstract
This in vitro study evaluated the impact of TiO2 nanotubes (n-TiO2) incorporated into glass ionomer cement (GIC) on Streptococcus mutans (S. mutans) characteristics at cellular and molecular levels. n-TiO2, synthesized by the alkaline method (20 nm in size), was added to Ketac Molar EasyMix® at 0%, 3%, 5%, and 7% by weight. S. mutans strains were cultured on GIC disks with addition or not of n-TiO2 for 1, 3, and 7 days and the following parameters were assessed: inhibition halo (mm) (n=3/group); cell viability (live/dead) (n=5/group); cell morphology (SEM) (n=3/group); and gene expression by real-time PCR (vicR, covR, gtfB, gtfC, and gtfD) (n=6/group). The data were analyzed by the Kruskal-Wallis test, repeated-measures ANOVA or two-way ANOVA, and Tukey's and Dunn's post-hoc tests (α=0.05). The agar diffusion test showed a higher antibacterial property for 5% n-TiO2 compared with 3% and 7% (p<0.05) with no effect of time (1, 3, and 7 days). The cell number was significantly affected by all n-TiO2 groups, while viability was mostly affected by 3% and 5% n-TiO2, which also affected cell morphology and organization. Real-time PCR demonstrated that n-TiO2 reduced the expression of covR when compared with GIC with no n-TiO2 (p<0.05), with no effect of time, except for 3% n-TiO2 on vicR expression. Within-group and between-group analyses revealed n-TiO2 did not affect mRNA levels of gtfB, gtfC, and gtfD (p>0.05). Incorporation of n-TiO2 at 3% and 5% potentially affected S. mutans viability and the expression of key genes for bacterial survival and growth, improving the anticariogenic properties of GIC.
Collapse
Affiliation(s)
- Isaac Jordão de Souza Araújo
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | | | - Orisson Ponce Gomes
- Universidade Estadual Paulista Júlio Mesquita Filho - UNESP, School of Sciences, Department of Physics, Bauru, SP, Brazil
| | - Priscila Alves Giovani
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Pediatric Dentistry, Piracicaba, SP, Brazil
| | - Júlia Puppin-Rontani
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Vanessa Arias Pecorari
- Universidade Paulista - UNIP, School of Dentistry, Department of Bioestatistics, São Paulo, São Paulo, Brazil
| | | | | | - Francisco Humberto Nociti Junior
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Prosthodontics and Periodontology, Piracicaba, SP, Brazil
| | - Regina Maria Puppin-Rontani
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Pediatric Dentistry, Piracicaba, SP, Brazil
| | - Paulo Noronha Lisboa-Filho
- Universidade Estadual Paulista Júlio Mesquita Filho - UNESP, School of Sciences, Department of Physics, Bauru, SP, Brazil
| | | |
Collapse
|
25
|
Rizzardi KF, Indiani CMDSP, Mattos-Graner RDO, de Sousa ET, Nobre-Dos-Santos M, Parisotto TM. Firmicutes Levels in the Mouth Reflect the Gut Condition With Respect to Obesity and Early Childhood Caries. Front Cell Infect Microbiol 2021; 11:593734. [PMID: 34123864 PMCID: PMC8190403 DOI: 10.3389/fcimb.2021.593734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
The present cross-sectional study investigated whether Firmicutes (F) and Bacteroidetes (B) levels in the mouth reflected the gut condition in obesity and early childhood caries (ECC). Eighty preschoolers (3-5 years) were equally assigned into four groups: 1. obese + ECC, 2. obese + caries-free (CF), 3. eutrophic + ECC, and 4. eutrophic + CF. Nutritional status and ECC were assessed based on the WHO criteria. Dental biofilm and fecal samples were collected for F and B quantification using RT-PCR analysis. Data were evaluated using three-way-ANOVA and Pearson’s correlation (α = 0.05). Regardless of the anatomical location effect (p = 0.22), there were higher values for F in the obese children + ECC compared with those in obese + caries-free (CF) in both mouth and gut (p < 0.05). The correlation for F at these sites was negative in obese children + ECC (r = −0.48; p = 0.03) and positive in obese children + CF (r=0.50; p = 0.03). Bacteroidetes were influenced by ECC (p = 0.03) and the anatomical location (p = 0.00), and the levels tended to be higher in the mouth of the obese children + ECC (p = 0.04). The F/B ratio was higher in the gut and was affected by the anatomical location (p = 0.00). This preliminary study suggested that modulated by ECC, counts of oral Firmicutes reflected corresponding condition in the gut of obese preschoolers. In addition, we first evidenced that the Firmicutes phylum behave differently according to the nutritional status and caries experience and that supragingival biofilm and gut could share levels of similarity.
Collapse
Affiliation(s)
- Karina Ferreira Rizzardi
- Laboratory of Clinical and Molecular Microbiology, University São Francisco, Bragança Paulista, Brazil
| | | | | | - Emerson Tavares de Sousa
- Department of Health Sciences and Pediatric Dentistry, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Marinês Nobre-Dos-Santos
- Department of Health Sciences and Pediatric Dentistry, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Thaís Manzano Parisotto
- Laboratory of Clinical and Molecular Microbiology, University São Francisco, Bragança Paulista, Brazil
| |
Collapse
|
26
|
Chen J, Zhang A, Xiang Z, Lu M, Huang P, Gong T, Pan Y, Lin Y, Zhou X, Li Y. EpsR Negatively Regulates Streptococcus mutans Exopolysaccharide Synthesis. J Dent Res 2021; 100:968-976. [PMID: 33749354 DOI: 10.1177/00220345211000668] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Streptococcus mutans is considered the primary etiological agent of human dental caries. Glucosyltransferases (Gtfs) from S. mutans play important roles in the formation of biofilm matrix and the development of cariogenic oral biofilm. Therefore, Gtfs are considered an important target to prevent the development of dental caries. However, the role of transcription factors in regulating gtf expression is not yet clear. Here, we identify a MarR (multiple antibiotic resistance regulator) family transcription factor named EpsR (exopolysaccharide synthesis regulator), which negatively regulates gtfB expression and exopolysaccharide (EPS) production in S. mutans. The epsR in-frame deletion strain grew slowly, aggregated more easily in the presence of dextran, and displayed different colony morphology and biofilm structure. Notably, epsR deletion resulted in altered 3-dimensional biofilm architecture, increased water-insoluble EPS production, and upregulated GtfB protein content and activity. In addition, global gene expression profiling revealed differences in the expression levels of 69 genes in which gtfB was markedly upregulated. The conserved DNA motif for EpsR binding was determined by electrophoretic mobility shift assay and DNase I footprinting assays. Moreover, analysis of β-galactosidase activity suggested that EpsR acted as a repressor and inhibited gtfB expression. Taken together, our findings indicate that EpsR is an important transcription factor that regulates gtfB expression and EPS production in S. mutans. These results add new aspects to the complexity of regulating the expression of genes involved in the cariogenicity of S. mutans, which might lead to novel strategies to prevent the formation of cariogenic biofilm that may favor diseases.
Collapse
Affiliation(s)
- J Chen
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - A Zhang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Xiang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - M Lu
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - P Huang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - T Gong
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Pan
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Lin
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Zhou
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Li
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Cortês IT, Rosalen PL, Berto LA, Castro ML, Pedrini DL, Porto AN, Cogo-Müller K, Nobre Franco GC. Effect of adrenaline and noradrenaline on biofilm formation and virulence factors of Streptococcus mutans UA159. Arch Oral Biol 2021; 125:105091. [PMID: 33652302 DOI: 10.1016/j.archoralbio.2021.105091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To evaluate in vitro the effects of adrenaline and noradrenaline on the biofilm formation on orthodontic brackets, acid production and expression of virulence genes of Streptococcus mutans UA159 (S. mutans). DESIGN S. mutans UA159 biofilm was formed on orthodontic brackets under exposure to adrenaline (100 μM), noradrenaline (50 μM) or PBS solution (control group) in triptone-yeast extract with 1 % sucrose. After 24 h, biofilm formation was quantified through Colony Forming Units / mL (CFU/mL) and RNA was extracted to perform gene expression analysis through real-time reverse transcriptase-PCR (RT-qPCR). Evaluation of acid production was carried out on planktonic cultures for 6 h. One-way ANOVA followed by Tukey's test was carried to determine statistical difference. The level of significance was set at 5 %. RESULTS Catecholamines stimulated biofilm formation of S. mutans in orthodontic brackets (p < 0,05) but did not interfere with acid production (pH reduction) or the expression of the tested genes related to biofilm formation (gtfB, gtfC, gbpA, gbpB, gbpC, gbpD and brpA), aciduric (relA) and acidogenic properties (ldh). CONCLUSIONS The present study was the first to demonstrate that catecholamines can stimulate S. mutans UA159 biofilm formation. These findings can contribute to clarify the role of stress on bacterial metabolism and contribute to the understanding of a possible role on caries development, mainly in orthodontic patients.
Collapse
Affiliation(s)
- Iago Torres Cortês
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil.
| | - Pedro Luiz Rosalen
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil; Graduate Program in Biological Sciences, Federal University of Alfenas, Alfenas, MG, Brazil.
| | - Luciana Aranha Berto
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil.
| | | | | | | | - Karina Cogo-Müller
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil; State University of Campinas, Faculty of Pharmaceutical Sciences, Campinas, SP, Brazil.
| | | |
Collapse
|
28
|
Al-Dahmash ND, Al-Ansari MM, Al-Otibi FO, Singh AR. Frankincense, an aromatic medicinal exudate of Boswellia carterii used to mediate silver nanoparticle synthesis: Evaluation of bacterial molecular inhibition and its pathway. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102337] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
29
|
Wen ZT, Jorgensen AN, Huang X, Ellepola K, Chapman L, Wu H, Brady LJ. Multiple factors are involved in regulation of extracellular membrane vesicle biogenesis in Streptococcus mutans. Mol Oral Microbiol 2021; 36:12-24. [PMID: 33040492 PMCID: PMC7940556 DOI: 10.1111/omi.12318] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/01/2020] [Accepted: 10/04/2020] [Indexed: 12/26/2022]
Abstract
Streptococcus mutans, a major etiological agent of human dental caries, produces membrane vesicles (MVs) that contain protein and extracellular DNA. In this study, functional genomics, along with in vitro biofilm models, was used to identify factors that regulate MV biogenesis. Our results showed that when added to growth medium, MVs significantly enhanced biofilm formation by S. mutans, especially during growth in sucrose. This effect occurred in the presence and absence of added human saliva. Functional genomics revealed several genes, including sfp, which have a major effect on S. mutans MVs. In Bacillus sp. sfp encodes a 4'-phosphopantetheinyl transferase that contributes to surfactin biosynthesis and impacts vesiculogenesis. In S. mutans, sfp resides within the TnSmu2 Genomic Island that supports pigment production associated with oxidative stress tolerance. Compared to the UA159 parent, the Δsfp mutant, TW406, demonstrated a 1.74-fold (p < .05) higher MV yield as measured by BCA protein assay. This mutant also displayed increased susceptibility to low pH and oxidative stressors, as demonstrated by acid killing and hydrogen peroxide challenge assays. Deficiency of bacA, a putative surfactin synthetase homolog within TnSmu2, and especially dac and pdeA that encode a di-adenylyl cyclase and a phosphodiesterase, respectively, also significantly increased MV yield (p < .05). However, elimination of bacA2, a bacitracin synthetase homolog, resulted in a >1.5-fold (p < .05) reduction of MV yield. These results demonstrate that S. mutans MV properties are regulated by genes within and outside of the TnSmu2 island, and that as a major particulate component of the biofilm matrix, MVs significantly influence biofilm formation.
Collapse
Affiliation(s)
- Zezhang T. Wen
- Department of Oral and Craniofacial Biology, School of Dentistry and Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health, New Orleans, LA
| | - Ashton N. Jorgensen
- Department of Oral and Craniofacial Biology, School of Dentistry and Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health, New Orleans, LA
| | - Xiaochang Huang
- Department of Oral and Craniofacial Biology, School of Dentistry and Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health, New Orleans, LA
| | - Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry and Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health, New Orleans, LA
| | - Lynne Chapman
- Department of Oral and Craniofacial Biology, School of Dentistry and Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health, New Orleans, LA
| | - Hui Wu
- Integrative Biomedical and Diagnostics Science, School of Dentistry, Oregon Health and Science University, Portland, OR
| | - L. Jeannine Brady
- Department of Oral Biology, School of Dentistry, the University of Florida, Gainesville, FL
| |
Collapse
|
30
|
Priya A, Kumar CBM, Valliammai A, Selvaraj A, Pandian SK. Usnic acid deteriorates acidogenicity, acidurance and glucose metabolism of Streptococcus mutans through downregulation of two-component signal transduction systems. Sci Rep 2021; 11:1374. [PMID: 33446778 PMCID: PMC7809355 DOI: 10.1038/s41598-020-80338-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
The principal etiological agent of human dental caries, Streptococcus mutans is a multi-virulent pathogen that can transform commensal oral microbial community to plaque biofilms. Major virulence factors that are associated with the cariogenicity of S. mutans include adhesion, acidogenicity and acidurity. All these pathogenic traits coordinate and alter the dental plaque ecology which provide room for interaction with other similar acidogenic and aciduric bacteria. This cariogenic flora increases the possibility of enamel demineralization which headway to caries development. The present study was aimed at evaluating the antimicrobial and antiinfective potential of a lichen secondary metabolite usnic acid (UA) against S. mutans. Minimum inhibitory concentration (MIC), Minimum bactericidal concentration (MBC) and growth kinetics were evaluated to determine the antimicrobial potential of UA against S. mutans. UA at 5 µg mL-1 and 10 µg mL-1 concentration were considered as MIC and MBC respectively. Effect on biofilm formation was microscopically assessed and found to be reduced in a concentration dependent manner. Gene expression of gtfB, gtfC, gtfD, vicR, ComDE and smu0630 was found to be downregulated upon treatment with sub-MIC of UA. Acidogenicity, acidurity, eDNA synthesis and response to oxidative stress were found to be attenuated by the influence of UA. It was also demonstrated to act on preformed mature biofilm of S. mutans. Moreover, UA was shown to possess very low frequency to acquire spontaneous resistance development in S. mutans. Besides, no morphological aberrations or toxic effect was instigated by UA in the human buccal epithelial cells as well as to the oral commensals. Altogether, these results demonstrate the therapeutic potential of usnic acid in the treatment of S. mutans infection.
Collapse
Affiliation(s)
- Arumugam Priya
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Chandra Bose Manish Kumar
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Alaguvel Valliammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | - Anthonymuthu Selvaraj
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, 630003, India
| | | |
Collapse
|
31
|
Amino Sugars Reshape Interactions between Streptococcus mutans and Streptococcus gordonii. Appl Environ Microbiol 2020; 87:AEM.01459-20. [PMID: 33097515 DOI: 10.1128/aem.01459-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
Amino sugars, particularly glucosamine (GlcN) and N-acetylglucosamine (GlcNAc), are abundant carbon and nitrogen sources supplied in host secretions and in the diet to the biofilms colonizing the human oral cavity. Evidence is emerging that these amino sugars provide ecological advantages to beneficial commensals over oral pathogens and pathobionts. Here, we performed transcriptome analysis on Streptococcus mutans and Streptococcus gordonii growing in single-species or dual-species cultures with glucose, GlcN, or GlcNAc as the primary carbohydrate source. Compared to glucose, GlcN caused drastic transcriptomic shifts in each species of bacteria when it was cultured alone. Likewise, cocultivation in the presence of GlcN yielded transcriptomic profiles that were dramatically different from the single-species results from GlcN-grown cells. In contrast, GlcNAc elicited only minor changes in the transcriptome of either organism in single- and dual-species cultures. Interestingly, genes involved in pyruvate metabolism were among the most significantly affected by GlcN in both species, and these changes were consistent with measurements of pyruvate in culture supernatants. Differing from what was found in a previous report, growth of S. mutans alone with GlcN inhibited the expression of multiple operons required for mutacin production. Cocultivation with S. gordonii consistently increased the expression of two manganese transporter operons (slo and mntH) and decreased expression of mutacin genes in S. mutans Conversely, S. gordonii appeared to be less affected by the presence of S. mutans but did show increases in genes for biosynthetic processes in the cocultures. In conclusion, amino sugars profoundly alter the interactions between pathogenic and commensal streptococci by reprogramming central metabolism.IMPORTANCE Carbohydrate metabolism is central to the development of dental caries. A variety of sugars available to dental microorganisms influence the development of caries by affecting the physiology, ecology, and pathogenic potential of tooth biofilms. Using two well-characterized oral bacteria, one pathogen (Streptococcus mutans) and one commensal (Streptococcus gordonii), in an RNA deep-sequencing analysis, we studied the impact of two abundant amino sugars on bacterial gene expression and interspecies interactions. The results indicated large-scale remodeling of gene expression induced by GlcN in particular, affecting bacterial energy generation, acid production, protein synthesis, and release of antimicrobial molecules. Our study provides novel insights into how amino sugars modify bacterial behavior, information that will be valuable in the design of new technologies to detect and prevent oral infectious diseases.
Collapse
|
32
|
Genetic and physiological effects of subinhibitory concentrations of oral antimicrobial agents on Streptococcus mutans biofilms. Microb Pathog 2020; 150:104669. [PMID: 33278519 DOI: 10.1016/j.micpath.2020.104669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/01/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022]
Abstract
Streptococcus mutans is the main etiological agent of dental caries because of its capacity to adhere to enamel structure and form biofilms. This study aimed to evaluate the effects of the anticariogenic agents - sodium fluoride (NaF) and chlorhexidine (CHX) - at levels below minimum inhibitory concentrations (sub-MICs) on the growth of planktonic cells and biofilms and on the expression of vicR and covR genes associated with the regulation of biofilm formation. MICs and minimum bactericidal concentrations (MBCs) of NaF and CHX were determined for S. mutans strains ATCC25175, UA159 and 3VF2. Growth curves were constructed for planktonic cells cultured in brain heart infusion (BHI) broth supplemented with NaF (0.125-0.75MIC) or CHX (0.25-0.75MIC). Biofilm formation assays were performed in microplates containing CHX or NaF at 0.5-1.0MIC and stained with violet crystal. Quantitative polymerase chain reaction determined the alterations in covR and vicR expression in cells exposed to antimicrobials at sub-MIC levels. NaF and CHX at sub-MIC levels affected the growth of planktonic cells of all three S. mutans strains, depending on the concentration tested. The biofilm formation in UA159 and 3VF2 was reduced by NaF at concentrations ≥0.5 MIC, while that of ATCC 25175 was reduced significantly irrespective of dose. In contrast, UA159 and 3VF2 biofilms were not affected by CHX at these levels, whereas those of ATCC 25175 were reduced significantly at all concentrations tested. Under sub-MIC conditions, CHX and (to a lesser degree) NaF increased vicR and covR expression in all three strains, although there were large differences between strains and treatment conditions employed. CHX and NaF at sub-MIC levels influence on the growth of S. mutans in planktonic and biofilm conditions and on transcript levels of biofilm-associated genes vicR and covR, in a dose-dependent manner.
Collapse
|
33
|
Alves LA, Ganguly T, Harth-Chú ÉN, Kajfasz J, Lemos JA, Abranches J, Mattos-Graner RO. PepO is a target of the two-component systems VicRK and CovR required for systemic virulence of Streptococcus mutans. Virulence 2020; 11:521-536. [PMID: 32427040 PMCID: PMC7239026 DOI: 10.1080/21505594.2020.1767377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/10/2020] [Accepted: 03/29/2020] [Indexed: 12/14/2022] Open
Abstract
Streptococcus mutans, a cariogenic species, is often associated with cardiovascular infections. Systemic virulence of specific S. mutans serotypes has been associated with the expression of the collagen- and laminin-binding protein Cnm, which is transcriptionally regulated by VicRK and CovR. In this study, we characterized a VicRK- and CovR-regulated gene, pepO, coding for a conserved endopeptidase. Transcriptional and protein analyses revealed that pepO is highly expressed in S. mutans strains resistant to complement immunity (blood isolates) compared to oral isolates. Gel mobility assay, transcriptional, and Western blot analyses revealed that pepO is repressed by VicR and induced by CovR. Deletion of pepO in the Cnm+ strain OMZ175 (OMZpepO) or in the Cnm- UA159 (UApepO) led to an increased susceptibility to C3b deposition, and to low binding to complement proteins C1q and C4BP. Additionally, pepO mutants showed diminished ex vivo survival in human blood and impaired capacity to kill G. mellonella larvae. Inactivation of cnm in OMZ175 (OMZcnm) resulted in increased resistance to C3b deposition and unaltered blood survival, although both pepO and cnm mutants displayed attenuated virulence in G. mellonella. Unlike OMZcnm, OMZpepO could invade HCAEC endothelial cells. Supporting these phenotypes, recombinant proteins rPepO and rCnmA showed specific profiles of binding to C1q, C4BP, and to other plasma (plasminogen, fibronectin) and extracellular matrix proteins (type I collagen, laminin). Therefore this study identifies a novel VicRK/CovR-target required for immune evasion and host persistence, pepO, expanding the roles of VicRK and CovR in regulating S. mutans virulence.
Collapse
Affiliation(s)
- Lívia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| | - Tridib Ganguly
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Érika N. Harth-Chú
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| | - Jessica Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - José A. Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Jacqueline Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School – State University of Campinas, Piracicaba, SP, Brazil
| |
Collapse
|
34
|
Lei L, Zeng J, Wang L, Gong T, Zheng X, Qiu W, Zhang R, Yun L, Yang Y, Li Y. Quantitative acetylome analysis reveals involvement of glucosyltransferase acetylation in Streptococcus mutans biofilm formation. ENVIRONMENTAL MICROBIOLOGY REPORTS 2020; 13:86-97. [PMID: 33185947 DOI: 10.1111/1758-2229.12907] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 11/11/2020] [Indexed: 02/05/2023]
Abstract
Streptococcus mutans (S. mutans) effectively utilizes dietary sucrose for the exopolysaccharide productions, which are mostly synthesized by the effects of glucosyltransferases (Gtfs). In the present study, the acetylome of S. mutans was identified and quantitative acetylome analysis of the bacterial biofilm growth (SMB) was compared with that of planktonic growth (SMP). The dynamic changes of protein acetylation were quantified using the integrated approach involving TMT labeling and Kac affinity enrichment followed by high-resolution mass spectrometry-based quantitative proteomics. In total, 973 acetylation sites in 445 proteins were identified, among which 617 acetylation sites in 302 proteins were quantitated. The overall analysis indicated that 22.7% of proteins were acetylated. Among the quantified proteins in SMB, the acetylation degree of lysine in 56 sites increased, while that of lysine decreased in 52 sites. In the acetylome of S. mutans, six significantly enriched motifs were identified and obtained including Kac****K, KacF, Kac****R, KacY, KacH, F*Kac. In addition, KEGG pathway-based enrichment analysis indicated significant enrichments in glycolysis/gluconeogenesis, and RNA degradation. Particularly, most downregulated acetylated lysine proteins were glucosyltransferase-SI, glucosyltransferase-I, and glucosyltransferase-S in S. mutans biofilm, which probably reveals a switch-off mechanism for the regulation of glucosyltransferases function during the biofilm development.
Collapse
Affiliation(s)
- Lei Lei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jumei Zeng
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Lingyun Wang
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tao Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Qiu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ru Zhang
- Department of Endodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Libing Yun
- Department of Forensic Pathology, West China School of Basic Medical Sciences & Forensic Science, Sichuan University, Chengdu, China
| | - Yingming Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuqing Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
35
|
Wu S, Liu Y, Lei L, Zhang H. Endogenous antisense walR RNA modulates biofilm organization and pathogenicity of Enterococcus faecalis. Exp Ther Med 2020; 21:69. [PMID: 33365069 PMCID: PMC7716642 DOI: 10.3892/etm.2020.9501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 06/10/2020] [Indexed: 02/05/2023] Open
Abstract
Enterococcus faecalis (E. faecalis) is regarded as the major pathogen for persistent periapical periodontitis. The aim of the present study was to investigate the role of antisense walR RNA in the regulation of adjacent downstream genes. Reverse transcription-PCR assays were performed to validate walR. Adjacent downstream genes walK, EF1195, EF1196, and EF1197 were co-transcribed and detect antisense walR RNA. Northern blotting and 5'-rapid amplification of cDNA ends (5'-RACE) assays were conducted to detect and confirm a novel walR antisense (ASwalR) RNA. ASwalR overexpression mutants were constructed, and the biofilm biomass was determined using a crystal violet microtiter assay. The present study detected and confirmed a 550-bp noncoding antisense RNA with the potential to attenuate the activities of the essential response regulator WalR. The levels of antisense walR RNA transcripts were inversely associated with the production of WalR protein. It was showed that overexpression of ASwalR leads to reduced biofilm formation and exopolysaccharide synthesis. Furthermore, the pathogenicity of E. faecalis was markedly decreased by ASwalR overexpression in an in vivo periapical periodontitis model. In summary, the present study detected a novel antisense walR RNA that leads to a reduction in biofilm formation and the pathogenicity of E. faecalis. Collectively, the data suggest a role for ASwalR as a post-transcriptional modulator of the WalR regulator in E. faecalis.
Collapse
Affiliation(s)
- Shizhou Wu
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China.,Department of Advanced Oral Sciences and Therapeutics, University of Maryland School of Dentistry, Baltimore, MD 21201, USA
| | - Yunjie Liu
- West China School of Public Health, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Hui Zhang
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
36
|
Deciphering Streptococcal Biofilms. Microorganisms 2020; 8:microorganisms8111835. [PMID: 33233415 PMCID: PMC7700319 DOI: 10.3390/microorganisms8111835] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Streptococci are a diverse group of bacteria, which are mostly commensals but also cause a considerable proportion of life-threatening infections. They colonize many different host niches such as the oral cavity, the respiratory, gastrointestinal, and urogenital tract. While these host compartments impose different environmental conditions, many streptococci form biofilms on mucosal membranes facilitating their prolonged survival. In response to environmental conditions or stimuli, bacteria experience profound physiologic and metabolic changes during biofilm formation. While investigating bacterial cells under planktonic and biofilm conditions, various genes have been identified that are important for the initial step of biofilm formation. Expression patterns of these genes during the transition from planktonic to biofilm growth suggest a highly regulated and complex process. Biofilms as a bacterial survival strategy allow evasion of host immunity and protection against antibiotic therapy. However, the exact mechanisms by which biofilm-associated bacteria cause disease are poorly understood. Therefore, advanced molecular techniques are employed to identify gene(s) or protein(s) as targets for the development of antibiofilm therapeutic approaches. We review our current understanding of biofilm formation in different streptococci and how biofilm production may alter virulence-associated characteristics of these species. In addition, we have summarized the role of surface proteins especially pili proteins in biofilm formation. This review will provide an overview of strategies which may be exploited for developing novel approaches against biofilm-related streptococcal infections.
Collapse
|
37
|
Balasubramanian AR, Vasudevan S, Shanmugam K, Lévesque CM, Solomon AP, Neelakantan P. Combinatorial effects of trans-cinnamaldehyde with fluoride and chlorhexidine on Streptococcus mutans. J Appl Microbiol 2020; 130:382-393. [PMID: 32707601 DOI: 10.1111/jam.14794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/06/2020] [Accepted: 07/18/2020] [Indexed: 01/03/2023]
Abstract
AIMS The aim of this study was to investigate the effects of trans-cinnamaldehyde (TC) and its synergistic activity with chlorhexidine (CHX) and fluoride against Streptococcus mutans. METHODS AND RESULTS Streptococcus mutans UA159 was treated with TC alone and in combination with CHX or sodium fluoride. The synergy profile was analysed using the Zero Interaction Potency model. TC showed strong synergism (synergy score of 21·697) with CHX, but additive effect (synergy score of 5·298) with fluoride. TC and the combinations were tested for acid production (glycolytic pH drop) and biofilm formation by S. mutans, and nitric oxide production in macrophages. TC significantly inhibited sucrose-dependent biofilm formation and acid production by S. mutans. Mechanistic studies were carried out by qRT-PCR-based transcriptomic studies which showed that TC acts by impairing genes related to metabolism, quorum sensing, bacteriocin expression, stress tolerance and biofilm formation. CONCLUSIONS trans-Cinnamaldehyde potentiates CHX and sodium fluoride in inhibiting S. mutans biofilms and virulence through multiple mechanisms. This study sheds significant new light on the potential to develop TC as an anti-caries treatment. SIGNIFICANCE AND IMPACT OF THE STUDY Oral diseases were classified as a 'silent epidemic' in the US Surgeon General's Report on Oral Health. Two decades later, >4 billion people are still affected worldwide by caries, having significant effects on the quality of life. There is an urgent need to develop novel compounds and strategies to combat dental caries. Here, we prove that TC downregulates multiple pathways and potentiates the CHX and fluoride to prevent S. mutans biofilms and virulence. This study sheds significant new light on the potential to develop TC in combination with CHX or fluoride as novel treatments to arrest dental caries.
Collapse
Affiliation(s)
- A R Balasubramanian
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - S Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - K Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - C M Lévesque
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - A P Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - P Neelakantan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR
| |
Collapse
|
38
|
Multi-targeted Antisense Oligonucleotide Delivery by a Framework Nucleic Acid for Inhibiting Biofilm Formation and Virulence. NANO-MICRO LETTERS 2020; 12:74. [PMID: 34138282 PMCID: PMC7770702 DOI: 10.1007/s40820-020-0409-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
A framework nucleic acid delivery system was developed through self-assembly, which can deliver antisense oligonucleotides against multiple targets in bacterial cells. The ASOs-tFNAs (750 nM) was found to simultaneously inhibit the expression of gtfBCD, gbpB, and ftf, and significantly reduce the extracellular polysaccharide synthesis and biofilm thickness.
Biofilm formation is responsible for numerous chronic infections and represents a serious health challenge. Bacteria and the extracellular polysaccharides (EPS) cause biofilms to become adherent, toxic, resistant to antibiotics, and ultimately difficult to remove. Inhibition of EPS synthesis can prevent the formation of bacterial biofilms, reduce their robustness, and promote removal. Here, we have developed a framework nucleic acid delivery system with a tetrahedral configuration. It can easily access bacterial cells and functions by delivering antisense oligonucleotides that target specific genes. We designed antisense oligonucleotide sequences with multiple targets based on conserved regions of the VicK protein-binding site. Once delivered to bacterial cells, they significantly decreased EPS synthesis and biofilm thickness. Compared to existing approaches, this system is highly efficacious because it simultaneously reduces the expression of all targeted genes (gtfBCD, gbpB, ftf). We demonstrate a novel nucleic acid-based nanomaterial with multi-targeted inhibition that has great potential for the treatment of chronic infections caused by biofilms.![]()
Collapse
|
39
|
Lei L, Zhang B, Mao M, Chen H, Wu S, Deng Y, Yang Y, Zhou H, Hu T. Carbohydrate Metabolism Regulated by Antisense vicR RNA in Cariogenicity. J Dent Res 2019; 99:204-213. [PMID: 31821772 DOI: 10.1177/0022034519890570] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Streptococcus mutans is a major cariogenic pathogen that resides in multispecies oral microbial biofilms. The VicRK 2-component system is crucial for bacterial adaptation, virulence, and biofilm organization and contains a global and vital response regulator, VicR. Notably, we identified an antisense vicR RNA (AS vicR) associated with an adjacent RNase III–encoding ( rnc) gene that was relevant to microRNA-size small RNAs (msRNAs). Here, we report that ASvicR overexpression significantly impeded bacterial growth, biofilm exopolysaccharide synthesis, and cariogenicity in vivo. Transcriptome analysis revealed that the AS vicR RNA mainly regulated carbohydrate metabolism. In particular, overproducing AS vicR demonstrated a reduction in galactose and glucose metabolism by monosaccharide composition analysis. The results of high-performance gel permeation chromatography revealed that the water-insoluble glucans isolated from AS vicR presented much lower molecular weights. Furthermore, direct evidence showed that total RNAs were disrupted by rnc-encoded RNase III. With the coexpression of T4 RNA ligase, putative msRNA1657, which is an rnc-related messenger RNA, was verified to bind to the 5′-UTR regions of the vicR gene. Furthermore, AS vicR regulation revealed a sponge regulatory-mediated network for msRNA associated with adjacent RNase III–encoding genes. There was an increase in AS vicR transcript levels in clinical S. mutans strains from caries-free children, while the expression of AS vicR was decreased in early childhood caries patients; this outcome may be explored as a potential strategy contributing to the management of dental caries. Taken together, our findings suggest an important role of AS vicR-mediated sponge regulation in S. mutans, indicating the characterization of lactose metabolism by a vital response regulator in cariogenicity. These findings have a number of implications and have reshaped our understanding of bacterial gene regulation from its transcriptional conception to the key roles of regulatory RNAs.
Collapse
Affiliation(s)
- L. Lei
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - B. Zhang
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - M. Mao
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Endodontics, College of Stomatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - H. Chen
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - S. Wu
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Y. Deng
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Y. Yang
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - H. Zhou
- Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - T. Hu
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Harth-Chu EN, Alves LA, Theobaldo JD, Salomão MF, Höfling JF, King WF, Smith DJ, Mattos-Graner RO. PcsB Expression Diversity Influences on Streptococcus mitis Phenotypes Associated With Host Persistence and Virulence. Front Microbiol 2019; 10:2567. [PMID: 31798545 PMCID: PMC6861525 DOI: 10.3389/fmicb.2019.02567] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022] Open
Abstract
S. mitis is an abundant member of the commensal microbiota of the oral cavity and pharynx, which has the potential to promote systemic infections. By analyzing a collection of S. mitis strains isolated from the oral cavity at commensal states or from systemic infections (blood strains), we established that S. mitis ubiquitously express the surface immunodominant protein, PcsB (also called GbpB), required for binding to sucrose-derived exopolysaccharides (EPS). Immuno dot blot assays with anti-PcsB antibodies and RT-qPCR transcription analyses revealed strain-specific profiles of PcsB production associated with diversity in pcsB transcriptional activities. Additionally, blood strains showed significantly higher levels of PcsB expression compared to commensal isolates. Because Streptococcus mutans co-colonizes S. mitis dental biofilms, and secretes glucosyltransferases (GtfB/C/D) for the synthesis of highly insoluble EPS from sucrose, profiles of S. mitis binding to EPS, biofilm formation and evasion of the complement system were assessed in sucrose-containing BHI medium supplemented or not with filter-sterilized S. mutans culture supernatants. These analyses showed significant S. mitis binding to EPS and biofilm formation in the presence of S. mutans supernatants supplemented with sucrose, compared to BHI or BHI-sucrose medium. In addition, these phenotypes were abolished if strains were grown in culture supernatants of a gtfBCD-defective S. mutans mutant. Importantly, GtfB/C/D-associated phenotypes were enhanced in high PcsB-expressing strains, compared to low PcsB producers. Increased PcsB expression was further correlated with increased resistance to deposition of C3b/iC3b of the complement system after exposure to human serum, when strains were previously grown in the presence of S. mutans supernatants. Finally, analyses of PcsB polymorphisms and bioinformatic prediction of epitopes with significant binding to MHC class II alleles revealed that blood isolates harbor PcsB polymorphisms in its functionally conserved CHAP-domain, suggesting antigenic variation. These findings reveal important roles of PcsB in S. mitis-host interactions under commensal and pathogenic states, highlighting the need for studies to elucidate mechanisms regulating PcsB expression in this species.
Collapse
Affiliation(s)
- Erika N Harth-Chu
- Department of Oral Diagnosis, Piracicaba Dental School, UNICAMP, Piracicaba, Brazil
| | - Lívia A Alves
- Department of Oral Diagnosis, Piracicaba Dental School, UNICAMP, Piracicaba, Brazil
| | - Jéssica D Theobaldo
- Department of Oral Diagnosis, Piracicaba Dental School, UNICAMP, Piracicaba, Brazil
| | - Mariana F Salomão
- Department of Oral Diagnosis, Piracicaba Dental School, UNICAMP, Piracicaba, Brazil
| | - José F Höfling
- Department of Oral Diagnosis, Piracicaba Dental School, UNICAMP, Piracicaba, Brazil
| | - William F King
- Department of Immunology and Infectious Disease, The Forsyth Institute, Cambridge, MA, United States
| | - Daniel J Smith
- Department of Immunology and Infectious Disease, The Forsyth Institute, Cambridge, MA, United States
| | | |
Collapse
|
41
|
Trans,trans-farnesol, an antimicrobial natural compound, improves glass ionomer cement properties. PLoS One 2019; 14:e0220718. [PMID: 31430298 PMCID: PMC6701760 DOI: 10.1371/journal.pone.0220718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/22/2019] [Indexed: 12/26/2022] Open
Abstract
A series of experiments were conducted to characterize a novel restorative material. We explored the effect on biological, physical and chemical properties of glass ionomer cement (GIC) adding-the naturally occurring tt-farnesol (900 mM). Two groups were accomplished for all assays: GIC+tt-farnesol and GIC (control). Biological assays: 1) agar diffusion against some cariogenic bacteria; 2) S. mutans biofilm formation and confocal laser scanning microscopy-CLSM. 3) gtfB, gtfC, gtfD, gbpB, vicR, and covR expression; 4) MTT and microscopic morphology. Physical properties assays: 1) roughness; 2) hardness; 3) compressive strength and 4) diametral tensile strength. Chemical assay: Raman spectroscopy. The adding of tt-farnesol to GIC led to larger zones of inhibition (p<0.05), biofilms with a short-term reduction in bacterial viability but similar biomass (p>0.05). Polysaccharides levels increased over time, similarly over groups (p>0.05). Viable and non-viable S. mutans were seen on the specimens’ surface by CLSM but their virulence was not modulated by tt-farnesol. The tt-farnesol increased the HaCaT cell viability without impact on compressive and diametral tensile strength and roughness although the hardness was positively affected (p<0.05). Raman confirmed the presence of tt-farnesol. The incorporation of tt-farnesol into GIC inhibited the growth of cariogenic bacteria but had a little effect on the composition, structure and physiology of the biofilm matrices. Also, the tt-farnesol increased the hardness and the biocompatibility of the GIC, not influencing negatively other physical properties of the restorative material.
Collapse
|
42
|
Park M, Sutherland JB, Rafii F. Effects of nano-hydroxyapatite on the formation of biofilms by Streptococcus mutans in two different media. Arch Oral Biol 2019; 107:104484. [PMID: 31382161 DOI: 10.1016/j.archoralbio.2019.104484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/20/2019] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES The aim of this study was to examine the effect of nano-hydroxyapatite (nHA) on biofilm formation by Streptococcus mutans, which is actively involved in the initiation of dental caries. DESIGN The effects of nHA on growth and biofilm formation by S. mutans were investigated in two media: a saliva analog medium, basal medium mucin (BMM); and a nutrient-rich medium, brain heart infusion (BHI); in the presence and absence of sucrose. RESULTS Sucrose enhanced the growth of S. mutans in both media. In the presence of sucrose, nHA enhanced bacterial growth and biofilm formation more in BMM medium than in BHI. nHA also affected the transcription of glucosyltransferase (gtf) genes and production of polysaccharide differently in the two media. In BHI medium, the transcription of all three gtf genes, coding for enzymes that synthesize soluble and insoluble glucans from sucrose, was increased more than 3-fold by nHA. However, in BMM medium, only the transcription of gtfB and gtfC, coding for insoluble glucans, was substantially enhanced by nHA. CONCLUSIONS nHA appeared to enhance biofilm formation by increasing glucosyltransferase transcription, which resulted in an increase in production of insoluble glucans. This effect was influenced by the growth conditions.
Collapse
Affiliation(s)
- Miseon Park
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - John B Sutherland
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Fatemeh Rafii
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA.
| |
Collapse
|
43
|
Wu X, Song Q, Han A. Interacting proteins of the essential two-component system YycFG in Bacillus subtilis. J Basic Microbiol 2019; 59:950-959. [PMID: 31339578 DOI: 10.1002/jobm.201800701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 06/14/2019] [Accepted: 06/24/2019] [Indexed: 11/06/2022]
Abstract
Two-component signal transduction systems (TCSs) play a major role in adaption and survival of microorganisms in a dynamic and sometimes dangerous environment. YycFG is an essential TCS for many Gram-positive bacteria, such as Bacillus subtilis, which regulates many important biological processes. However, its functional essentiality remains largely unknown. Here, we report several YycFG interacting proteins through coimmunoprecipitation (Co-IP) and mass spectrometry (MS) analyses. We engineered the B. subtilis genome by a knock-in approach to express YycG with a C-terminal Flag and YycF with an N-terminal HA tag. Immunoprecipitated fractions using anti-Flag or anti-HA agarose were subjected to MS analyses. A total of 41 YycG interacting proteins and four YycF interacting proteins were identified, most of which are involved in cellular metabolic processes, including cell wall synthesis and modification. The interactions of YycG with AsnB and FabL, as examples, were further validated in vitro. This study provided a clue that YycFG may be directly involved in regulation of bacterial central metabolic pathways.
Collapse
Affiliation(s)
- Xuanang Wu
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, China
| | - Qi Song
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, China
| | - Aidong Han
- State Key Laboratory for Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, China
| |
Collapse
|
44
|
Lin TH, Pan TM. Characterization of an antimicrobial substance produced by Lactobacillus plantarum NTU 102. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2019; 52:409-417. [DOI: 10.1016/j.jmii.2017.08.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 12/11/2016] [Accepted: 08/11/2017] [Indexed: 11/15/2022]
|
45
|
Tierney AR, Rather PN. Roles of two-component regulatory systems in antibiotic resistance. Future Microbiol 2019; 14:533-552. [PMID: 31066586 DOI: 10.2217/fmb-2019-0002] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Two-component regulatory systems (TCSs) are a major mechanism by which bacteria sense and respond to changes in their environment. TCSs typically consist of two proteins that bring about major regulation of the cell genome through coordinated action mediated by phosphorylation. Environmental conditions that activate TCSs are numerous and diverse and include exposure to antibiotics as well as conditions inside a host. The resulting regulatory action often involves activation of antibiotic defenses and changes to cell physiology that increase antibiotic resistance. Examples of resistance mechanisms enacted by TCSs contained in this review span those found in both Gram-negative and Gram-positive species and include cell surface modifications, changes in cell permeability, increased biofilm formation, and upregulation of antibiotic-degrading enzymes.
Collapse
Affiliation(s)
- Aimee Rp Tierney
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, 30322 USA
| | - Philip N Rather
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA, 30322 USA.,Research Service, Department of Veterans' Affairs, Atlanta VA Health Care System, Decatur, GA, 30033 USA
| |
Collapse
|
46
|
Machado AHS, Garcia IM, Motta ADSD, Leitune VCB, Collares FM. Triclosan-loaded chitosan as antibacterial agent for adhesive resin. J Dent 2019; 83:33-39. [DOI: 10.1016/j.jdent.2019.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/17/2019] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
|
47
|
Wang S, Huang Q, Liu X, Li Z, Yang H, Lu Z. Rapid Antibiofilm Effect of Ag/ZnO Nanocomposites Assisted by Dental LED Curing Light against Facultative Anaerobic Oral Pathogen Streptococcus mutans. ACS Biomater Sci Eng 2019; 5:2030-2040. [PMID: 33405515 DOI: 10.1021/acsbiomaterials.9b00118] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The integration of nanomaterials with clinical therapeutic instruments is a promising approach to improve the effects of nanomaterials. We reported an efficient synergistic antibacterial strategy formed through the combination of Ag/ZnO nanocomposites with a light-emitting diode (LED) curing light, which is a commonly used small instrument in dental clinics. The as-designed integration depicted a significantly enhanced bactericidal effect on facultative anaerobic oral pathogen Streptococcus mutans (S. mutans) both in planktonic and biofilm phases over a very short irradiation time (≤5 min). Further study showed that the combination of LED and Ag/ZnO nanocomposites induced more ·OH and ·O2- generation, which is responsible for the enhanced antibacterial activity. Moreover, this combination could destroy S. mutans biofilm by killing the bacteria embedded within biofilm, inhibiting exopolysaccharide production and down-regulating the biofilm-related gene expression. Therefore, it is proposed that this combination could be applied in dental clinics to realize dental caries prevention and dental restoration simultaneously.
Collapse
Affiliation(s)
- Shilei Wang
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| | - Qiaomu Huang
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| | - Xiangyu Liu
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| | - Zhao Li
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| | - Hao Yang
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| | - Zhong Lu
- Research Center for Environmental Ecology and Engineering, Key Laboratory for Green Chemical Process of Ministry of Education, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No. 206, Guanggu first road, Wuhan 430073, PR China
| |
Collapse
|
48
|
Lemos JA, Palmer SR, Zeng L, Wen ZT, Kajfasz JK, Freires IA, Abranches J, Brady LJ. The Biology of Streptococcus mutans. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0051-2018. [PMID: 30657107 PMCID: PMC6615571 DOI: 10.1128/microbiolspec.gpp3-0051-2018] [Citation(s) in RCA: 356] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
As a major etiological agent of human dental caries, Streptococcus mutans resides primarily in biofilms that form on the tooth surfaces, also known as dental plaque. In addition to caries, S. mutans is responsible for cases of infective endocarditis with a subset of strains being indirectly implicated with the onset of additional extraoral pathologies. During the past 4 decades, functional studies of S. mutans have focused on understanding the molecular mechanisms the organism employs to form robust biofilms on tooth surfaces, to rapidly metabolize a wide variety of carbohydrates obtained from the host diet, and to survive numerous (and frequent) environmental challenges encountered in oral biofilms. In these areas of research, S. mutans has served as a model organism for ground-breaking new discoveries that have, at times, challenged long-standing dogmas based on bacterial paradigms such as Escherichia coli and Bacillus subtilis. In addition to sections dedicated to carbohydrate metabolism, biofilm formation, and stress responses, this article discusses newer developments in S. mutans biology research, namely, how S. mutans interspecies and cross-kingdom interactions dictate the development and pathogenic potential of oral biofilms and how next-generation sequencing technologies have led to a much better understanding of the physiology and diversity of S. mutans as a species.
Collapse
Affiliation(s)
- J A Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - S R Palmer
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH 43210
| | - L Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - Z T Wen
- Department of Comprehensive Dentistry and Biomaterials and Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - J K Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - I A Freires
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - J Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - L J Brady
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| |
Collapse
|
49
|
CovR and VicRKX Regulate Transcription of the Collagen Binding Protein Cnm of Streptococcus mutans. J Bacteriol 2018; 200:JB.00141-18. [PMID: 30201780 DOI: 10.1128/jb.00141-18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 09/04/2018] [Indexed: 11/20/2022] Open
Abstract
Cnm is a surface-associated protein present in a subset of Streptococcus mutans strains that mediates binding to extracellular matrices, intracellular invasion, and virulence. Here, we showed that cnm transcription is controlled by the global regulators CovR and VicRKX. In silico analysis identified multiple putative CovR- and VicR-binding motifs in the regulatory region of cnm as well as in the downstream gene pgfS, which is associated with the posttranslational modification of Cnm. Electrophoretic mobility shift assays revealed that CovR and VicR specifically and independently bind to the cnm and pgfS promoter regions. Quantitative real-time PCR and Western blot analyses of ΔcovR and ΔvicK strains as well as of a strain overexpressing vicRKX revealed that CovR functions as a positive regulator of cnm, whereas VicRKX acts as a negative regulator. In agreement with the role of VicRKX as a repressor, the ΔvicK strain showed enhanced binding to collagen and laminin and higher intracellular invasion rates. Overexpression of vicRKX was associated with decreased rates of intracellular invasion but did not affect collagen or lamin binding activities, suggesting that this system controls additional genes involved in binding to these extracellular matrix proteins. As expected, based on the role of CovR in cnm regulation, the ΔcovR strain showed decreased intracellular invasion rates, but, unexpectedly collagen and laminin binding activities were increased in this mutant strain. Collectively, the results presented here expand the repertoire of virulence-related genes regulated by CovR and VicRKX to include the core gene pgfS and the noncore gene cnm IMPORTANCE Streptococcus mutans is a major pathogen associated with dental caries and also implicated in systemic infections, in particular, infective endocarditis. The Cnm adhesin of S. mutans is an important virulence factor associated with systemic infections and caries severity. Despite its role in virulence, the regulatory mechanisms governing cnm expression are poorly understood. Here, we describe the identification of two independent regulatory systems controlling the transcription of cnm and the downstream pgfS-pgfM1-pgfE-pgfM2 operon. A better understanding of the mechanisms controlling expression of virulence factors like Cnm can facilitate the development of new strategies to treat bacterial infections.
Collapse
|
50
|
Zheng C, Li L, Ge H, Meng H, Li Y, Bei W, Zhou X. Role of two-component regulatory systems in the virulence of Streptococcus suis. Microbiol Res 2018; 214:123-128. [PMID: 30031474 DOI: 10.1016/j.micres.2018.07.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/19/2018] [Accepted: 07/07/2018] [Indexed: 01/01/2023]
Abstract
Streptococcus suis is an important zoonotic pathogen that causes severe infections and great economic losses worldwide. Understanding how this pathogen senses and responds to environmental signals during the infectious process can offer insight into its pathogenesis and may be helpful in the development of drug targets. Two-component regulatory systems (TCSs) play an essential role in this environmental response. In S. suis, at least 15 groups of TCSs have been predicted. Among them, several have been demonstrated to be involved in virulence and/or stress response. In this review, we discuss the progress in the study of TCSs in S. suis, focusing on the role of these systems in the virulence of this bacterium.
Collapse
Affiliation(s)
- Chengkun Zheng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; State Key Laboratory of Agricultural Microbiology/The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Lingzhi Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Haojie Ge
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Hongmei Meng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Yang Li
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China
| | - Weicheng Bei
- State Key Laboratory of Agricultural Microbiology/The Cooperative Innovation Center for Sustainable Pig Production, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xiaohui Zhou
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety/Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, The Ministry of Education of China, Yangzhou University, Yangzhou, 225009, China; Department of Pathobiology and Veterinary Science, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|