1
|
Mba IE, Mba TO, Uwazie CK, Aina FA, Kemisola AO, Uwazie IJ. New insights and perspectives on the virulence of hypervirulent Klebsiella pneumoniae. Folia Microbiol (Praha) 2025:10.1007/s12223-025-01261-9. [PMID: 40198504 DOI: 10.1007/s12223-025-01261-9] [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: 06/22/2024] [Accepted: 03/16/2025] [Indexed: 04/10/2025]
Abstract
Klebsiella pneumoniae, a Gram-negative bacterium, comprises strains with diverse virulence potentials, ranging from classical to hypervirulent variants. Understanding the genetic basis underlying the virulence disparities between hypervirulent (hvKp) and classical K. pneumoniae (cKp) strains is crucial. hvKp strains are characterized by hypermucoviscosity, attributed to the presence of specific virulence genes and the production of molecules that aid in their ability to survive, evade host immune defenses, and cause infection. In contrast, classical strains exhibit a broader array of antimicrobial resistance determinants, conferring resistance to multiple antibiotics. Although current definitions of hvKp incorporate clinical features, phenotypes, and genotypes, identifying hvKp strains in clinical settings remains challenging. Genomic studies have been pivotal and have helped to identify distinct genetic profiles in hvKp strains, including unique virulence plasmids and chromosomal variations, underscoring the genetic diversity within K. pneumoniae populations. This review examines the virulence and genetic determinants associated with hvKp. The presence of genes defining hypervirulence, alongside considerations of their utility as biomarkers and targets for therapeutic strategies, is discussed, while also providing insight into biofilm formation by hvKp and key questions that need urgent responses in understanding hvKp.
Collapse
Affiliation(s)
- Ifeanyi Elibe Mba
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Ibadan, 200005, Nigeria.
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria Nsukka, Enugu, 410001, Nigeria.
| | - Toluwalase Odunayo Mba
- Department of Public Health, Faculty of Basic Medical and Health Sciences, Lead City University, Ibadan, 200255, Nigeria
- Medical Institute, Sumy State University, Sumy, 40007, Ukraine
| | | | - Fetuata Aminat Aina
- Department of Microbiology, College of Natural Sciences, Federal University of Agriculture, Abeokuta, 111101, Nigeria
| | | | | |
Collapse
|
2
|
Liu D, Lu Y, Li Z, Pang X, Gao X. Quorum Sensing: Not Just a Bridge Between Bacteria. Microbiologyopen 2025; 14:e70016. [PMID: 40159675 PMCID: PMC11955508 DOI: 10.1002/mbo3.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
The study of quorum sensing (QS) has gained critical importance, offering insights into bacterial and microorganism communication. QS, regulated by autoinducers, synchronizes collective bacterial behaviors across diverse chemical signals and target genes. This review highlights innovative approaches to regulating QS, emphasizing the potential of quorum quenching and QS inhibitors to mitigate bacterial pathogenicity. These strategies have shown promise in aquaculture and plant resistance, disrupting QS pathways to combat infections. QS also provides opportunities for developing biosensors for early disease detection and preventing biofilm formation, which is critical to overcoming antimicrobial resistance. The applications of QS extend to cancer therapy, with targeted drug delivery systems utilizing QS mechanisms. Advancements in QS regulation, such as the use of nanomaterials, hydrogels, and microplastics, provide novel methods to modulate QS systems. This review explores the latest developments in QS, recognizing its significance in controlling bacterial behavior and its broad impacts on human health and disease management. Integrating these insights into therapeutic strategies and diagnostics represents a pivotal opportunity for medical progress.
Collapse
Affiliation(s)
- Derun Liu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Yonglin Lu
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
| | - Ziyun Li
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xin Pang
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| | - Xueyan Gao
- Medical Science and Technology Innovation CenterShandong First Medical University & Shandong Academy of Medical SciencesjinanChina
- State Key Laboratory of Microbial TechnologyShandong UniversityQingdaoChina
| |
Collapse
|
3
|
Pu X, Fang B, Wu J, Zhao Z, Liu Y, Li J, Gao H, Wang R, Zhang M. Effects of Lacticaseibacillus paracasei L9 on Oral Microbiota and Cariogenic Factors in Streptococcus mutans-Infected Mice. Foods 2024; 13:4118. [PMID: 39767060 PMCID: PMC11675566 DOI: 10.3390/foods13244118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
In the pathogenesis of dental caries, Streptococcus mutans (S. mutans) plays a central role. S. mutans can produce extracellular polysaccharides, which can help the bacteria form biofilms on the tooth surface, create a stable living environment, and hinder the removal of bacteria by natural defense substances in the oral cavity such as saliva. Meanwhile, the oral microbiota and dietary habits exert long-term influences on its development. This study, employing the BALB/c mouse model, explored the effects of L. paracasei L9 on dental caries. In the experiment, mice underwent the S. mutans inoculation and were subsequently treated with L. paracasei L9 or S. salivarius K12 for 28 consecutive days. The results showed that L. paracasei L9 significantly ameliorated early enamel caries, and both L. paracasei L9 and S. salivarius K12 cooperatively downregulated the expressions of critical cariogenic factors, effectively suppressing the initial adhesion of S. mutans and the formation of dental plaques. L. paracasei L9 reshaped the oral microbiota of caries-affected mice, selectively reducing pathogens abundances and augmenting abundances of probiotics such as Lactobacillaceae and Streptococcus salivarius. This study offers a strategic approach for the management of dental caries, highlighting the potential of these probiotics in the field of oral health.
Collapse
Affiliation(s)
- Xinyao Pu
- School of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China;
| | - Bing Fang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.F.); (J.W.); (Z.Z.)
| | - Jianmin Wu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.F.); (J.W.); (Z.Z.)
| | - Zhi Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.F.); (J.W.); (Z.Z.)
| | - Yue Liu
- School of Food and Health, Beijing Technology and Business University, No. 11 Fucheng Road, Beijing 100024, China; (Y.L.); (J.L.); (H.G.)
| | - Jingyu Li
- School of Food and Health, Beijing Technology and Business University, No. 11 Fucheng Road, Beijing 100024, China; (Y.L.); (J.L.); (H.G.)
| | - Haina Gao
- School of Food and Health, Beijing Technology and Business University, No. 11 Fucheng Road, Beijing 100024, China; (Y.L.); (J.L.); (H.G.)
| | - Ran Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; (B.F.); (J.W.); (Z.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, No. 11 Fucheng Road, Beijing 100024, China; (Y.L.); (J.L.); (H.G.)
| |
Collapse
|
4
|
Hsu JE, Matsen FA, Whitson AJ, Waalkes A, Almazan J, Bourassa LA, Salipante SJ, Long DR. 2023 Neer Award for Basic Science: Genetics of Cutibacterium acnes in revision shoulder arthroplasty: a large-scale bacterial whole-genome sequencing study. J Shoulder Elbow Surg 2024; 33:2400-2410. [PMID: 38604398 PMCID: PMC11663454 DOI: 10.1016/j.jse.2024.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/24/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Cutibacterium acnes is the bacterium most commonly responsible for shoulder periprosthetic joint infection (PJI) and is often cultured from samples obtained at the time of revision for failed shoulder arthroplasty. We sought to determine whether these bacteria originate from the patient or from exogenous sources. We also sought to identify which C. acnes genetic traits were associated with the development of shoulder PJI. METHODS We performed bacterial whole-genome sequencing of C. acnes from a single-institution repository of cultures obtained before or during primary and revision shoulder arthroplasty and correlated the molecular epidemiology and genetic content of strains with clinical features of infection. RESULTS A total of 341 isolates collected over a 4-year period from 88 patients were sequenced. C. acnes cultured from surgical specimens demonstrated significant similarity to the strains colonizing the skin of the same patient (P < .001). Infrequently, there was evidence of strains shared across unrelated patients, suggesting that exogenous sources of C. acnes culture-positivity were uncommon. Phylotypes IB and II were modestly associated with clinical features of PJI, but all phylotypes appeared inherently capable of causing disease. Chronic shoulder PJI was associated with the absence of common C. acnes genes involved in bacterial quorum-sensing (luxS, tqsA). CONCLUSION C. acnes strains cultured from deep intraoperative sources during revision shoulder arthroplasty demonstrate strong genetic similarity to the strains colonizing a patient's skin. Some phylotypes of C. acnes commonly colonizing human skin are modestly more virulent than others, but all phylotypes have a capacity for PJI. C. acnes cultured from cases of PJI commonly demonstrated genetic hallmarks associated with adaptation from acute to chronic phases of infection. This is the strongest evidence to date supporting the role of the patient's own, cutaneous C. acnes strains in the pathogenesis of shoulder arthroplasty infection. Our findings support the importance of further research focused on perioperative decolonization and management of endogenous bacteria that are likely to be introduced into the arthroplasty wound at the time of skin incision.
Collapse
Affiliation(s)
- Jason E Hsu
- Department of Orthopaedics and Sports Medicine, University of Washington Medical Center, Seattle, WA, USA.
| | - Frederick A Matsen
- Department of Orthopaedics and Sports Medicine, University of Washington Medical Center, Seattle, WA, USA
| | - Anastasia J Whitson
- Department of Orthopaedics and Sports Medicine, University of Washington Medical Center, Seattle, WA, USA
| | - Adam Waalkes
- Department of Laboratory Medicine & Pathology, University of Washington Medical Center, Seattle, WA, USA
| | - Jared Almazan
- Department of Laboratory Medicine & Pathology, University of Washington Medical Center, Seattle, WA, USA
| | - Lori A Bourassa
- Department of Laboratory Medicine & Pathology, University of Washington Medical Center, Seattle, WA, USA
| | - Stephen J Salipante
- Department of Laboratory Medicine & Pathology, University of Washington Medical Center, Seattle, WA, USA
| | - Dustin R Long
- Department of Anesthesiology & Pain Medicine, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
5
|
Alothaim AS, Alhoqail WA, Menakha M, Vijayakumar R. Combining molecular modelling and experimental approaches to gain mechanistic insights into the LuxP drug target in Streptococcus pyogens. J Biomol Struct Dyn 2024; 42:9494-9504. [PMID: 37642991 DOI: 10.1080/07391102.2023.2252079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/20/2023] [Indexed: 08/31/2023]
Abstract
Autoinducer-2 can mediate inter- and intra-species communication signal between bacteria and these signals from AI-2 is noted from limited species of bacteria. In humans, S. pyogenes is a pathogen that causes a wide range of illnesses and can survive in the host system and transmit infection. The process by which S. pyogenes acquires the competence to live and disseminate infection remains unknown. We hypothesized that AI-2 and their receptors would play a significant role during infection, and for that present investigation provides the experimental and molecular insights. In the absence of details about the receptor LuxP and LuxQ, the screening approach provides supporting insights. The evolutionary relationship and similarities of the PBP domain (Spy 1535) and the signal transmission PDZ domain (Spy 1536) were studied in relation to their counterparts in other bacteria. Molecular docking and modeling confirmed the domain-enhanced specificity for AI-2 binding. In vitro studies showed that AI-2, which is present in the cell-free supernatant of S. pyogenes, regulates luminescence in P. luminous and biofilm development in E. coli using the LuxS reporter genes. Examination of S. pyogenes gene expression revealed modulation of virulence genes when the pathogen was exposed to V. harveyi HSL and AI-2. Therefore, S. pyogenes pathogenicity is sequentially regulated by AI-2 it acquires from other commensal bacteria. Overall, this study lays the groundwork for understanding the signalling mechanism from AI-2, which are critical to the pathogenic mechanism of S. pyogenes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Abdulaziz S Alothaim
- Department of Biology, College of Science, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Wardah A Alhoqail
- Department of Biology, College of Education, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Muniraj Menakha
- Department of Bio-informatics, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Rajendran Vijayakumar
- Department of Biology, College of Science, Majmaah University, Al-Majmaah, Saudi Arabia
| |
Collapse
|
6
|
Sikdar R, Beauclaire MV, Lima BP, Herzberg MC, Elias MH. N-acyl homoserine lactone signaling modulates bacterial community associated with human dental plaque. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585217. [PMID: 38559107 PMCID: PMC10980036 DOI: 10.1101/2024.03.15.585217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
N-acyl homoserine lactones (AHLs) are small diffusible signaling molecules that mediate a cell density-dependent bacterial communication system known as quorum sensing (QS). AHL-mediated QS regulates gene expression to control many critical bacterial behaviors including biofilm formation, pathogenicity, and antimicrobial resistance. Dental plaque is a complex multispecies oral biofilm formed by successive colonization of the tooth surface by groups of commensal, symbiotic, and pathogenic bacteria, which can contribute to tooth decay and periodontal diseases. While the existence and roles of AHL-mediated QS in oral microbiota have been debated, recent evidence indicates that AHLs play significant roles in oral biofilm development and community dysbiosis. The underlying mechanisms, however, remain poorly characterized. To better understand the importance of AHL signaling in dental plaque formation, we manipulated AHL signaling by adding AHL lactonases or exogenous AHL signaling molecules. We find that AHLs can be detected in dental plaque grown under 5% CO2 conditions, but not when grown under anaerobic conditions, and yet anaerobic cultures are still responsive to AHLs. QS signal disruption using lactonases leads to changes in microbial population structures in both planktonic and biofilm states, changes that are dependent on the substrate preference of the used lactonase but mainly result in the increase in the abundance of commensal and pioneer colonizer species. Remarkably, the opposite manipulation, that is the addition of exogenous AHLs increases the abundance of late colonizer bacterial species. Hence, this work highlights the importance of AHL-mediated QS in dental plaque communities, its potential different roles in anaerobic and aerobic parts of dental plaque, and underscores the potential of QS interference in the control of periodontal diseases.
Collapse
Affiliation(s)
- Rakesh Sikdar
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Mai V. Beauclaire
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Saint Paul, MN 55108, USA
| | - Bruno P. Lima
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark C. Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mikael H. Elias
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
7
|
Huq M, Wahid SUH, Istivan T. Biofilm Formation in Campylobacter concisus: The Role of the luxS Gene. Microorganisms 2023; 12:46. [PMID: 38257873 PMCID: PMC10820981 DOI: 10.3390/microorganisms12010046] [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: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Campylobacter concisus is a bacterium that inhabits human oral cavities and is an emerging intestinal tract pathogen known to be a biofilm producer and one of the bacterial species found in dental plaque. In this study, biofilms of oral and intestinal C. concisus isolates were phenotypically characterized. The role of the luxS gene, which is linked to the regulation of biofilm formation in other pathogens, was assessed in relation to the pathogenic potential of this bacterium. Biofilm formation capacity was assessed using phenotypic assays. Oral strains were shown to be the highest producers. A luxS mutant was created by inserting a kanamycin cassette within the luxS gene of the highest biofilm-forming isolate. The loss of the polar flagellum was observed with scanning and transmission electron microscopy (SEM and TEM). Furthermore, the luxS mutant exhibited a significant reduction (p < 0.05) in biofilm formation, motility, and its expression of flaB, in addition to the capability to invade intestinal epithelial cells, compared to the parental strain. The study concluded that C. concisus oral isolates are significantly higher biofilm producers than the intestinal isolates and that LuxS plays a role in biofilm formation, invasion, and motility in this bacterium.
Collapse
Affiliation(s)
- Mohsina Huq
- School of Science, STEM College, RMIT University, Bundoora, Melbourne, VIC 3083, Australia
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | | | - Taghrid Istivan
- School of Science, STEM College, RMIT University, Bundoora, Melbourne, VIC 3083, Australia
| |
Collapse
|
8
|
Man LL, Xiang DJ. Effect of LuxS/AI-2-mediated quorum sensing system on bacteriocin production of Lactobacillus plantarum NMD-17. Folia Microbiol (Praha) 2023; 68:855-866. [PMID: 37156969 DOI: 10.1007/s12223-023-01060-0] [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: 12/14/2022] [Accepted: 04/21/2023] [Indexed: 05/10/2023]
Abstract
Lactobacillus plantarum NMD-17 separated from koumiss could produce a bacteriocin named plantaricin MX against Gram-positive bacteria and Gram-negative bacteria. The bacteriocin synthesis of L. plantarum NMD-17 was remarkably induced in co-cultivation with Lactobacillus reuteri NMD-86 as the increase of cell numbers and AI-2 activity, and the expressions of luxS encoding signal AI-2 synthetase, plnB encoding histidine protein kinase, plnD encoding response regulator, and plnE and plnF encoding structural genes of bacteriocin were significantly upregulated in co-cultivation, showing that the bacteriocin synthesis of L. plantarum NMD-17 in co-cultivation may be regulated by LuxS/AI-2-mediated quorum sensing system. In order to further demonstrate the role of LuxS/AI-2-mediated quorum sensing system in the bacteriocin synthesis of L. plantarum NMD-17, plasmids pUC18 and pMD18-T simple were used as the skeleton to construct the suicide plasmids pUC18-UF-tet-DF and pMD18-T simple-plnB-tet-plnD for luxS and plnB-plnD gene deletion, respectively. luxS and plnB-plnD gene knockout mutants were successfully obtained by homologous recombination. luxS gene knockout mutant lost its AI-2 synthesis ability, suggesting that LuxS protein encoded by luxS gene is key enzyme for AI-2 synthesis. plnB-plnD gene knockout mutant lost the ability to synthesize bacteriocin against Salmonella typhimurium ATCC14028, indicating that plnB-plnD gene was a necessary gene for bacteriocin synthesis of L. plantarum NMD-17. Bacteriocin synthesis, cell numbers, and AI-2 activity of luxS or plnB-plnD gene knockout mutants in co-cultivation with L. reuteri NMD-86 were obviously lower than those of wild-type strain in co-cultivation at 6-9 h (P < 0.01). The results showed that LuxS/AI-2-mediated quorum sensing system played an important role in the bacteriocin synthesis of L. plantarum NMD-17 in co-cultivation.
Collapse
Affiliation(s)
- Li-Li Man
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, 028042, People's Republic of China
| | - Dian-Jun Xiang
- College of Agriculture, Inner Mongolia Minzu University, Tongliao, 028042, People's Republic of China.
| |
Collapse
|
9
|
Lv X, Feng Z, Luo J, Liu Z, Lu J, Han S, Wang K, Zhang L. Effects of caffeic acid phenethyl ester against multi-species cariogenic biofilms. Folia Microbiol (Praha) 2023; 68:977-989. [PMID: 37289416 DOI: 10.1007/s12223-023-01064-w] [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: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023]
Abstract
Dental caries is a biofilm-related disease, widely perceived to be caused by oral ecological imbalance when cariogenic/aciduric bacteria obtain an ecological advantage. Compared with planktonic bacteria, dental plaques are difficult to remove under extracellular polymeric substance protection. In this study, the effect of caffeic acid phenethyl ester (CAPE) on a preformed cariogenic multi-species biofilm was evaluated, which was comprised of cariogenic bacteria (Streptococcus mutans), commensal bacteria (Streptococcus gordonii), and a pioneer colonizer (Actinomyces naeslundii). Our result revealed that treatment with 0.08 mg/mL CAPE reduced live S. mutans in the preformed multi-species biofilm while not significantly changing the quantification of live S. gordonii. CAPE significantly reduced the production of lactic acid, extracellular polysaccharide, and extracellular DNA and made the biofilm looser. Moreover, CAPE could promote the H2O2 production of S. gordonii and inhibit the expression of SMU.150 encoding mutacin to modulate the interaction among species in biofilms. Overall, our results suggested that CAPE could inhibit the cariogenic properties and change the microbial composition of the multi-species biofilms, indicating its application potential in dental caries prevention and management.
Collapse
Affiliation(s)
- Xiaohui Lv
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zening Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyuan Luo
- Department of Endodontics, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenqi Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junzhuo Lu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sili Han
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
10
|
Monteith W, Pascoe B, Mourkas E, Clark J, Hakim M, Hitchings MD, McCarthy N, Yahara K, Asakura H, Sheppard SK. Contrasting genes conferring short- and long-term biofilm adaptation in Listeria. Microb Genom 2023; 9:001114. [PMID: 37850975 PMCID: PMC10634452 DOI: 10.1099/mgen.0.001114] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/28/2023] [Indexed: 10/19/2023] Open
Abstract
Listeria monocytogenes is an opportunistic food-borne bacterium that is capable of infecting humans with high rates of hospitalization and mortality. Natural populations are genotypically and phenotypically variable, with some lineages being responsible for most human infections. The success of L. monocytogenes is linked to its capacity to persist on food and in the environment. Biofilms are an important feature that allow these bacteria to persist and infect humans, so understanding the genetic basis of biofilm formation is key to understanding transmission. We sought to investigate the biofilm-forming ability of L. monocytogenes by identifying genetic variation that underlies biofilm formation in natural populations using genome-wide association studies (GWAS). Changes in gene expression of specific strains during biofilm formation were then investigated using RNA sequencing (RNA-seq). Genetic variation associated with enhanced biofilm formation was identified in 273 genes by GWAS and differential expression in 220 genes by RNA-seq. Statistical analyses show that the number of overlapping genes flagged by either type of experiment is less than expected by random sampling. This novel finding is consistent with an evolutionary scenario where rapid adaptation is driven by variation in gene expression of pioneer genes, and this is followed by slower adaptation driven by nucleotide changes within the core genome.
Collapse
Affiliation(s)
- William Monteith
- Department of Biology, University of Oxford, Oxford, UK
- Department of Biology, University of Bath, Claverton Down, Bath, UK
| | - Ben Pascoe
- Department of Biology, University of Oxford, Oxford, UK
- Big Data Institute, University of Oxford, Oxford, UK
| | | | - Jack Clark
- Department of Genetics, University of Leicester, University Road, Leicester, UK
| | - Maliha Hakim
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, UK
| | - Matthew D. Hitchings
- Swasnsea University Medical School, Swansea University, Singleton Campus, Swansea, UK
| | - Noel McCarthy
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Koji Yahara
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroshi Asakura
- Division of Biomedical Food Research, National Institute of Health Sciences, Tonomachi 3-25-26, Kawasaki-ku, Kawasaki, Kanagawa 210-9501, Japan
| | | |
Collapse
|
11
|
Rudin L, Bornstein MM, Shyp V. Inhibition of biofilm formation and virulence factors of cariogenic oral pathogen Streptococcus mutans by natural flavonoid phloretin. J Oral Microbiol 2023; 15:2230711. [PMID: 37416858 PMCID: PMC10321187 DOI: 10.1080/20002297.2023.2230711] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Objectives To evaluate the effect and mechanism of action of the flavonoid phloretin on the growth and sucrose-dependent biofilm formation of Streptococcus mutans. Methods Minimum inhibitory concentration, viability, and biofilm susceptibility assays were conducted to assess antimicrobial and antibiofilm effect of phloretin. Biofilm composition and structure were analysed with scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Water-soluble (WSG) and water-insoluble glucan (WIG) were determined using anthrone method. Lactic acid measurements and acid tolerance assay were performed to assess acidogenicity and aciduricity. Reverse transcription quantitative PCR (RT-qPCR) was used to measure the expression of virulence genes essential for surface attachment, biofilm formation, and quorum sensing. Results Phloretin inhibited S. mutans growth and viability in a dose-dependent manner. Furthermore, it reduced gtfB and gtfC gene expression, correlating with the reduction of extracellular polysaccharides (EPS)/bacteria and WIG/WSG ratio. Inhibition of comED and luxS gene expression, involved in stress tolerance, was associated with compromised acidogenicity and aciduricity of S. mutans. Conclusions Phloretin exhibits antibacterial properties against S. mutans, modulates acid production and tolerance, and reduces biofilm formation. Clinical significance Phloretin is a promising natural compound with pronounced inhibitory effect on key virulence factors of the cariogenic pathogen, S. mutans.
Collapse
Affiliation(s)
- Lucille Rudin
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Michael M. Bornstein
- Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of BaselBaselSwitzerland
- Head of the Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel. Mattenstrasse 40, Basel, Switzerland
| | - Viktoriya Shyp
- Postdoctoral Researcher. Department Research, University Center for Dental Medicine Basel UZB
- Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel. Mattenstrasse 40, Basel, Switzerland
| |
Collapse
|
12
|
Gonçalves ASC, Leitão MM, Simões M, Borges A. The action of phytochemicals in biofilm control. Nat Prod Rep 2023; 40:595-627. [PMID: 36537821 DOI: 10.1039/d2np00053a] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: 2009 to 2021Antimicrobial resistance is now rising to dangerously high levels in all parts of the world, threatening the treatment of an ever-increasing range of infectious diseases. This has becoming a serious public health problem, especially due to the emergence of multidrug-resistance among clinically important bacterial species and their ability to form biofilms. In addition, current anti-infective therapies have low efficacy in the treatment of biofilm-related infections, leading to recurrence, chronicity, and increased morbidity and mortality. Therefore, it is necessary to search for innovative strategies/antibacterial agents capable of overcoming the limitations of conventional antibiotics. Natural compounds, in particular those obtained from plants, have been exhibiting promising properties in this field. Plant secondary metabolites (phytochemicals) can act as antibiofilm agents through different mechanisms of action from the available antibiotics (inhibition of quorum-sensing, motility, adhesion, and reactive oxygen species production, among others). The combination of different phytochemicals and antibiotics have revealed synergistic or additive effects in biofilm control. This review aims to bring together the most relevant reports on the antibiofilm properties of phytochemicals, as well as insights into their structure and mechanistic action against bacterial pathogens, spanning December 2008 to December 2021.
Collapse
Affiliation(s)
- Ariana S C Gonçalves
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Miguel M Leitão
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| | - Anabela Borges
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal
| |
Collapse
|
13
|
Zhang B, Jiang C, Cao H, Zeng W, Ren J, Hu Y, Li W, He Q. Transcriptome analysis of heat resistance regulated by quorum sensing system in Glaesserella parasuis. Front Microbiol 2022; 13:968460. [PMID: 36033895 PMCID: PMC9403865 DOI: 10.3389/fmicb.2022.968460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The ability of bacteria to resist heat shock allows them to adapt to different environments. In addition, heat shock resistance is known for their virulence. Our previous study showed that the AI-2/luxS quorum sensing system affects the growth characteristics, biofilm formation, and virulence of Glaesserella parasuis. The resistance of quorum sensing system deficient G. parasuis to heat shock was obviously weaker than that of wild type strain. However, the regulatory mechanism of this phenotype remains unclear. To illustrate the regulatory mechanism by which the quorum sensing system provides resistance to heat shock, the transcriptomes of wild type (GPS2), ΔluxS, and luxS complemented (C-luxS) strains were analyzed. Four hundred forty-four differentially expressed genes were identified in quorum sensing system deficient G. parasuis, which participated in multiple regulatory pathways. Furthermore, we found that G. parasuis regulates the expression of rseA, rpoE, rseB, degS, clpP, and htrA genes to resist heat shock via the quorum sensing system. We further confirmed that rseA and rpoE genes exerted an opposite regulatory effect on heat shock resistance. In conclusion, the findings of this study provide a novel insight into how the quorum sensing system affects the transcriptome of G. parasuis and regulates its heat shock resistance property.
Collapse
Affiliation(s)
- Bingzhou Zhang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Changsheng Jiang
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Hua Cao
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Wei Zeng
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Jingping Ren
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Yaofang Hu
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
| | - Wentao Li
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, China
- *Correspondence: Qigai He,
| |
Collapse
|
14
|
Salazar-Sánchez A, Baztarrika I, Alonso R, Fernández-Astorga A, Martínez-Ballesteros I, Martinez-Malaxetxebarria I. Arcobacter butzleri Biofilms: Insights into the Genes Beneath Their Formation. Microorganisms 2022; 10:1280. [PMID: 35888999 PMCID: PMC9324650 DOI: 10.3390/microorganisms10071280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 12/19/2022] Open
Abstract
Arcobacter butzleri, the most prevalent species of the genus, has the demonstrated ability to adhere to various surfaces through biofilm production. The biofilm formation capability has been related to the expression of certain genes, which have not been characterized in A. butzleri. In order to increase the knowledge of this foodborne pathogen, the aim of this study was to assess the role of six biofilm-associated genes in campylobacteria (flaA, flaB, fliS, luxS, pta and spoT) in the biofilm formation ability of A. butzleri. Knockout mutants were constructed from different foodborne isolates, and static biofilm assays were conducted on polystyrene (PS), reinforced glass and stainless steel. Additionally, motility and Congo red binding assays were performed. In general, mutants in flaAB, fliS and luxS showed a decrease in the biofilm production irrespective of the surface; mutants in spoT showed an increase on stainless steel, and mutants in pta and spoT showed a decrease on reinforced glass but an increase on PS. Our work sheds light on the biofilm-related pathogenesis of A. butzleri, although future studies are necessary to achieve a satisfactory objective.
Collapse
Affiliation(s)
- Adrián Salazar-Sánchez
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
| | - Itsaso Baztarrika
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
| | - Rodrigo Alonso
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria-Gasteiz, Spain
| | - Aurora Fernández-Astorga
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
| | - Ilargi Martínez-Ballesteros
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria-Gasteiz, Spain
| | - Irati Martinez-Malaxetxebarria
- MikroIker Research Group, Department of Immunology, Microbiology and Parasitology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.S.-S.); (I.B.); (R.A.); (A.F.-A.); (I.M.-B.)
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01006 Vitoria-Gasteiz, Spain
| |
Collapse
|
15
|
Candidate Phyla Radiation, an Underappreciated Division of the Human Microbiome, and Its Impact on Health and Disease. Clin Microbiol Rev 2022; 35:e0014021. [PMID: 35658516 DOI: 10.1128/cmr.00140-21] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Candidate phyla radiation (CPR) is an emerging division of the bacterial domain within the human microbiota. Still poorly known, these microorganisms were first described in the environment in 1981 as "ultramicrobacteria" with a cell volume under 0.1 μm3 and were first associated with the human oral microbiota in 2007. The evolution of technology has been paramount for the study of CPR within the human microbiota. In fact, since these ultramicrobacteria have yet to be axenically cultured despite ongoing efforts, progress in imaging technology has allowed their observation and morphological description. Although their genomic abilities and taxonomy are still being studied, great strides have been made regarding their taxonomic classification, as well as their lifestyle. In addition, advancements in next-generation sequencing and the continued development of bioinformatics tools have allowed their detection as commensals in different human habitats, including the oral cavity and gastrointestinal and genital tracts, thus highlighting CPR as a nonnegligible part of the human microbiota with an impact on physiological settings. Conversely, several pathologies present dysbiosis affecting CPR levels, including inflammatory, mucosal, and infectious diseases. In this exhaustive review of the literature, we provide a historical perspective on the study of CPR, an overview of the methods available to study these organisms and a description of their taxonomy and lifestyle. In addition, their distribution in the human microbiome is presented in both homeostatic and dysbiotic settings. Future efforts should focus on developing cocultures and, if possible, axenic cultures to obtain isolates and therefore genomes that would provide a better understanding of these ultramicrobacteria, the importance of which in the human microbiome is undeniable.
Collapse
|
16
|
Mahto KU, Kumari S, Das S. Unraveling the complex regulatory networks in biofilm formation in bacteria and relevance of biofilms in environmental remediation. Crit Rev Biochem Mol Biol 2021; 57:305-332. [PMID: 34937434 DOI: 10.1080/10409238.2021.2015747] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Biofilms are assemblages of bacteria embedded within a matrix of extracellular polymeric substances (EPS) attached to a substratum. The process of biofilm formation is a complex phenomenon regulated by the intracellular and intercellular signaling systems. Various secondary messenger molecules such as cyclic dimeric guanosine 3',5'-monophosphate (c-di-GMP), cyclic adenosine 3',5'-monophosphate (cAMP), and cyclic dimeric adenosine 3',5'-monophosphate (c-di-AMP) are involved in complex signaling networks to regulate biofilm development in several bacteria. Moreover, the cell to cell communication system known as Quorum Sensing (QS) also regulates biofilm formation via diverse mechanisms in various bacterial species. Bacteria often switch to the biofilm lifestyle in the presence of toxic pollutants to improve their survivability. Bacteria within a biofilm possess several advantages with regard to the degradation of harmful pollutants, such as increased protection within the biofilm to resist the toxic pollutants, synthesis of extracellular polymeric substances (EPS) that helps in the sequestration of pollutants, elevated catabolic gene expression within the biofilm microenvironment, higher cell density possessing a large pool of genetic resources, adhesion ability to a wide range of substrata, and metabolic heterogeneity. Therefore, a comprehensive account of the various factors regulating biofilm development would provide valuable insights to modulate biofilm formation for improved bioremediation practices. This review summarizes the complex regulatory networks that influence biofilm development in bacteria, with a major focus on the applications of bacterial biofilms for environmental restoration.
Collapse
Affiliation(s)
- Kumari Uma Mahto
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| | - Swetambari Kumari
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| | - Surajit Das
- Department of Life Science, Laboratory of Environmental Microbiology and Ecology (LEnME), National Institute of Technology, Odisha, India
| |
Collapse
|
17
|
Shaheer P, Sreejith VN, Joseph TC, Murugadas V, Lalitha KV. Quorum quenching Bacillus spp.: an alternative biocontrol agent for Vibrio harveyi infection in aquaculture. DISEASES OF AQUATIC ORGANISMS 2021; 146:117-128. [PMID: 34617517 DOI: 10.3354/dao03619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Quorum sensing (QS) is a type of cell to cell communication in bacteria that can also regulate the virulence potential in pathogenic strains. Hence, QS disruption, i.e. the quorum quenching (QQ) mechanism, is presently being explored as a novel bio-control strategy to counter bacterial infections. In the present study, we characterized the QQ ability of Bacillus spp. strains to reduce the expression of some virulence factors of a shrimp pathogen, Vibrio harveyi. We screened a total of 118 spore-forming bacterial isolates from aquaculture ponds and mangrove soil for their ability to degrade the synthetic N-acyl-homoserine lactones (AHLs) C4-HSL, C6-HSL, C8-HSL, and C10-HSL. We then selected the top 17 isolates with high AHL-degradation ability for further study. Among them, B. subtilis MFB10, B. lentus MFB2, and B. firmus MFB7 had the highest ability for degradation. These 3 isolates suppressed the expression of virulence genes encoding protease, lipase, phospholipase, caseinase, chitinase, and gelatinase, and potentially inhibited the biofilm formation of V. harveyi MFB32. The reduction in expression of virulence genes like those coding for metalloprotease, serine protease, and haemolysin were confirmed by real-time PCR analysis. Moreover, in an in vivo challenge experiment, these Bacillus spp. protected Penaeus monodon post-larvae against V. harveyi MFB3 infection. Our results demonstrate the potential application of AHL-degrading Bacillus spp. as an alternative to antibiotics in shrimp hatcheries to control luminescent vibriosis. This novel bio-therapeutic method is a promising approach towards disease control in shrimp aquaculture.
Collapse
Affiliation(s)
- P Shaheer
- Microbiology Fermentation and Biotechnology Division, Central Institute of Fisheries Technology (CIFT), Matsyapuri PO, Cochin 682029, Kerala, India
| | | | | | | | | |
Collapse
|
18
|
Cai S, Meng K, Liu P, Cao X, Wang G. Suppressive effects of gecko cathelicidin on biofilm formation and cariogenic virulence factors of Streptococcus mutans. Arch Oral Biol 2021; 129:105205. [PMID: 34237581 DOI: 10.1016/j.archoralbio.2021.105205] [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] [Received: 04/21/2021] [Revised: 06/17/2021] [Accepted: 06/26/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aims of this study were to investigate the effectivity of gecko cathelicidin Gj-CATH2 on biofilm formation and cariogenic virulence factors of S. mutans, and preliminary explore its function mechanisms. DESIGN Minimum inhibitory concentration and bacterial killing kinetics assays were performed to assess the antimicrobial effect of Gj-CATH2.The influence of Gj-CATH2 on S. mutans biofilm formation was determined by crystal violet staining method and observed by SEM. The effects of Gj-CATH2 on exopolysaccharides (EPS) synthesis, bacterial aggregation, acidogenicity and aciduricity of S. mutans were also investigated. Quantitative real-time PCR was conducted to acquire the expression profile of related genes. RESULTS Gj-CATH2 showed strong bactericidal and anti-biofilm effects on S. mutans. SEM confirmed the reduction of the dense structure in S. mutans biofilm in Gj-CATH2-treated groups. Gj-CATH2 significantly inhibited EPS synthesis, cell aggregation, acid production of S. mutans, but showed no influence on its acid proof. Furthermore, the expression of genes related to biofilm formation (gtfB/C/D, gbpB/D), quorum sensing system (luxS and comD/E) and acidogenicity (ldh) was significantly suppressed by Gj-CATH2. Gj-CATH2 also displayed advantageous resistance in human saliva and exhibited negligible toxicity against mammalian cells. CONCLUSIONS Gj-CATH2 inhibited S. mutans biofilm formation by targeting the bacterial adhesion and the biofilm maturation stages. Gj-CATH2 significantly suppressed virulence factors production of S. mutans, resulting in decreased EPS synthesis and reduced acidogenicity of bacteria. These findings suggest Gj-CATH2 might be a promising agent for clinical application in prevention of dental caries.
Collapse
Affiliation(s)
- Shasha Cai
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China.
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, 272067, China
| | - Peng Liu
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China
| | - Xianting Cao
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China
| | - Guannan Wang
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
19
|
Li E, Wu J, Zhang D. Exogenous autoinducer-2 inhibits biofilm development of Desulfovibrio sp. Huiquan2017. World J Microbiol Biotechnol 2021; 37:124. [PMID: 34170406 DOI: 10.1007/s11274-021-03071-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 05/16/2021] [Indexed: 11/28/2022]
Abstract
Sulfate-reducing bacteria (SRB) are culprits for microbiologically influenced corrosion, and biofilms are believed to play essential roles in the corrosion induced by SRB. However, little is known about the regulation of SRB biofilms. Quorum sensing signal molecules acyl-homoserine lactones (AHLs) and autoinducer-2 (AI-2) regulate biofilm formation of many bacteria. In this study, the production of AHLs and AI-2 by one SRB strain, Desulfovibrio sp. Huiquan2017, was detected, and the effect of exogenous AI-2 on bacterial biofilm formation was discussed. It was found that the cell-free supernatants of Desulfovibrio sp. Huiquan2017 induced luminescence in a ∆luxS mutant strain Vibrio harveyi BB170, indicating the production of functional AI-2 by the bacterium. In the presence of exogenous AI-2, the growth of Desulfovibrio sp. Huiquan2017 and early biofilm formation were not affected, but the later stage of biofilm development was inhibited significantly. The biofilms became looser, smaller, and thinner, and contained less bacteria and extracellular polymeric substances (EPS). The inhibition effect of AI-2 on the biofilm development of Desulfovibrio sp. Huiquan2017 was mainly achieved through reducing the amount of EPS in biofilms. These findings shed light on the biofilm regulation of SRB.
Collapse
Affiliation(s)
- Ee Li
- Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China
- Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China
| | - Jiajia Wu
- Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China.
- Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| | - Dun Zhang
- Key Laboratory of Marine Environmental Corrosion and Bio-Fouling, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, China.
- Open Studio for Marine Corrosion and Protection, Qingdao National Laboratory for Marine Science and Technology, 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
20
|
Liu Z, Zhang L, Wang J, Li Y, Chang Y, Huang X, Duan J, Ai Y, Zeng X, Guo J. Virtual Screening and Biological Evaluation of Anti-Biofilm Agents Targeting LuxS in the Quorum Sensing System. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211019625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Biofilm formation is considered as a crucial factor in various oral diseases, such as dental caries. The quorum sensing (QS) signaling system was proved to have a crucial role in the microbial dental plaque biofilm formation of Streptococcus mutans ( S. mutans). LuxS was critical to regulating the QS system and survival of the bacterium, and, therefore, compounds which target LuxS may be a potential therapy for dental caries. The binding activities of 37,170 natural compounds to LuxS were virtually screened in this study. Baicalein and paeonol were chosen for further research of the binding mode and ΔG values with LuxS. Both baicalein and paeonol inhibited the biofilm formation without influence on the growth of S. mutans. Baicalein also distinctly reduced the production of both rhamnolipids and acids. The results provide us with a new approach to combat dental caries instead of the traditional use of antibacterial chemicals.
Collapse
Affiliation(s)
- Zheng Liu
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Lihua Zhang
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Jincai Wang
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Guangzhou, China
| | - Yanping Li
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Yiqun Chang
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Xiaoling Huang
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jun Duan
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Yilong Ai
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Xuxin Zeng
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
| | - Jialiang Guo
- Foshan Stomatology Hospital & School of Medicine, Foshan University, China
- Institute of Pharmaceutical Analysis, College of Pharmacy, Jinan University, Guangzhou, China
- Guangdong Engineering Research Centre of Digital Stomatology & Foshan Engineering Research Centre of Digital Stomatology Medicine, Foshan, China
| |
Collapse
|
21
|
Yuan K, Hou L, Jin Q, Niu C, Mao M, Wang R, Huang Z. Comparative transcriptomics analysis of Streptococcus mutans with disruption of LuxS/AI-2 quorum sensing and recovery of methyl cycle. Arch Oral Biol 2021; 127:105137. [PMID: 33965851 DOI: 10.1016/j.archoralbio.2021.105137] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The LuxS/AI-2 quorum sensing (QS) system has critical roles in Streptococcus mutans cariogenicity. Whereas the molecular and cellular mechanisms of the LuxS/AI-2 QS system are not thoroughly understood. Given that LuxS has roles in QS and methyl cycle, its mutation can cause QS deficiency and methyl cycle disruption. The aim of this study was to investigate effects of the LuxS/AI-2 QS system on gene expression in Streptococcus mutans when methyl cycle was recovered with exogenous sahH gene. METHODS Our previous study introduced the exogenous sahH gene from Pseudomonas aeruginosa into an S. mutans luxS-null strain to restore the disrupted methyl cycle, and this produced the solely the LuxS/AI-2 QS system deficient strain. Here, we analyzed the transcriptomics of this strain to get insights into the molecular mechanisms of the LuxS/AI-2 QS system applying RNA-seq. RESULTS With recovery of methyl cycle, 84 genes didn't change in expression trends in S. mutans luxS-null strain. These genes mainly encode the ABC transporters, sugar transporter EII and enzymes of carbohydrate metabolism, and are rich in the Phosphotransferase system, Fructose and mannose metabolism, Amino sugar and nucleotide sugar metabolism, Galactose metabolism, Glycolysis/Gluconeogenesis, RNA degradation, Lysine biosynthesis, and Glycine, serine and threonine metabolism. CONCLUSIONS The LuxS/AI-2 QS system may mainly affect ABC transporters and carbohydrate transport, transformation and metabolism via EII subunits and enzymes to influence virulence-associated traits without effects of methyl cycle inStreptococcus mutans.
Collapse
Affiliation(s)
- Keyong Yuan
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lili Hou
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiaoqiao Jin
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chenguang Niu
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mengying Mao
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ruirui Wang
- Department of Stomatology, Minhang Branch, Zhongshan Hospital, Fudan University, China.
| | - Zhengwei Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
22
|
The Role of luxS in Histophilus somni Virulence and Biofilm Formation. Infect Immun 2021; 89:IAI.00567-20. [PMID: 33139386 DOI: 10.1128/iai.00567-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 10/29/2020] [Indexed: 01/09/2023] Open
Abstract
S-Ribosylhomocysteinase (LuxS) is required for the synthesis of the autoinducer-2 (AI-2) quorum-sensing signaling molecule in many Gram-negative bacteria. The bovine (and ovine) opportunistic pathogen Histophilus somni contains luxS and forms a biofilm containing an exopolysaccharide (EPS) in the matrix. Since biofilm formation is regulated by quorum sensing in many bacteria, the roles of luxS in H. somni virulence and biofilm formation were investigated. Although culture supernatants from H. somni were ineffective at inducing bioluminescence in the Vibrio harveyi reporter strain BB170, H. somni luxS complemented the biosynthesis of AI-2 in the luxS-deficient Escherichia coli strain DH5α. H. somni strain 2336 luxS was inactivated by transposon mutagenesis. RNA expression profiles revealed that many genes were significantly differentially expressed in the luxS mutant compared to that in the wild-type, whether the bacteria were grown planktonically or in a biofilm. Furthermore, the luxS mutant had a truncated and asialylated lipooligosaccharide (LOS) and was substantially more serum sensitive than the wild-type. Not surprisingly, the luxS mutant was attenuated in a mouse model for H. somni virulence, and some of the altered phenotypes were partially restored after the mutation was complemented with a functional luxS However, no major differences were observed between the wild-type and the luxS mutant in regard to outer membrane protein profiles, biofilm formation, EPS production, or intracellular survival. These results indicate that luxS plays a role in H. somni virulence in the context of LOS biosynthesis but not biofilm formation or other phenotypic properties examined.
Collapse
|
23
|
Alves-Barroco C, Paquete-Ferreira J, Santos-Silva T, Fernandes AR. Singularities of Pyogenic Streptococcal Biofilms - From Formation to Health Implication. Front Microbiol 2021; 11:584947. [PMID: 33424785 PMCID: PMC7785724 DOI: 10.3389/fmicb.2020.584947] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023] Open
Abstract
Biofilms are generally defined as communities of cells involved in a self-produced extracellular matrix adhered to a surface. In biofilms, the bacteria are less sensitive to host defense mechanisms and antimicrobial agents, due to multiple strategies, that involve modulation of gene expression, controlled metabolic rate, intercellular communication, composition, and 3D architecture of the extracellular matrix. These factors play a key role in streptococci pathogenesis, contributing to therapy failure and promoting persistent infections. The species of the pyogenic group together with Streptococcus pneumoniae are the major pathogens belonging the genus Streptococcus, and its biofilm growth has been investigated, but insights in the genetic origin of biofilm formation are limited. This review summarizes pyogenic streptococci biofilms with details on constitution, formation, and virulence factors associated with formation.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - João Paquete-Ferreira
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Teresa Santos-Silva
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| |
Collapse
|
24
|
Souissi M, Ben Lagha A, Chaieb K, Grenier D. Effect of a Berry Polyphenolic Fraction on Biofilm Formation, Adherence Properties and Gene Expression of Streptococcus mutans and Its Biocompatibility with Oral Epithelial Cells. Antibiotics (Basel) 2021; 10:antibiotics10010046. [PMID: 33466319 PMCID: PMC7824760 DOI: 10.3390/antibiotics10010046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 02/02/2023] Open
Abstract
The ability of Streptococcus mutans to adhere to oral surfaces and form biofilm is a key step in the tooth decay process. The aim of this study was to investigate a berry (wild blueberry, cranberry, and strawberry) polyphenolic fraction, commercialized as Orophenol®, for its antibacterial, anti-biofilm, and anti-adhesion properties on S. mutans. Moreover, the biocompatibility of the fraction with human oral epithelial cells was assessed. Phenolic acids, flavonoids (flavonols, anthocyanins, flavan-3-ols), and procyanidins made up 10.71%, 19.76%, and 5.29% of the berry polyphenolic fraction, respectively, as determined by chromatography and mass spectrometry. The berry polyphenolic preparation dose-dependently inhibited S. mutans biofilm formation while not reducing bacterial growth. At concentrations ranging from 250 to 1000 µg/mL, the fraction inhibited the adhesion of S. mutans to both saliva-coated hydroxyapatite and saliva-coated nickel-chrome alloy. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis showed that incubating S. mutans with the berry polyphenolic fraction was associated with a reduced expression of luxS gene, which regulates quorum sensing in S. mutans. The berry fraction did not show any significant cytotoxicity in an oral epithelial cell model. In conclusion, Orophenol®, which is a mixture of polyphenols from wild blueberry, cranberry and strawberry, possesses interesting anti-caries properties while being compatible with oral epithelial cells.
Collapse
Affiliation(s)
- Mariem Souissi
- Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (M.S.); (K.C.)
- Faculty of Sciences of Bizerta, University of Carthage, Zarzouna 7021, Tunisia
| | - Amel Ben Lagha
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC G1V 0A6, Canada;
| | - Kamel Chaieb
- Laboratory of Analysis, Treatment and Valorization of Pollutants of the Environment and Products, Faculty of Pharmacy, University of Monastir, Monastir 5000, Tunisia; (M.S.); (K.C.)
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC G1V 0A6, Canada;
- Correspondence:
| |
Collapse
|
25
|
Li J, Zhao X. Effects of quorum sensing on the biofilm formation and viable but non-culturable state. Food Res Int 2020; 137:109742. [DOI: 10.1016/j.foodres.2020.109742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/08/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023]
|
26
|
Zhang Y, Zhu Y, Zuo Y, Tang C, Zhou F, Cui X, Wang L. Effects of Rhein-8-O-β-D-glucopyranoside on the Biofilm Formation of Streptococcus mutans. Curr Microbiol 2020; 78:323-328. [PMID: 33128581 DOI: 10.1007/s00284-020-02248-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/09/2020] [Indexed: 11/26/2022]
Abstract
Dental caries is the most frequent biofilm-related human infectious disease in the oral cavity. Streptococcus mutans is one of the primary etiological agents of dental caries. The aim of our study was to investigate the effects of rhein-8-O-β-D-glucopyranoside (Rg) on the development of S. mutans biofilms. Growth curves were generated, and biofilm oxygen sensitivity was detected after Rg treatment. The expression levels of luxS, brpA, ffh, recA, nth, and smx were analyzed by real-time PCR. The trypan blue exclusion assay was used to measure the effect of Rg on monocyte viability. The results showed that Rg could significantly inhibit the growth of S. mutans and suppress the biofilm formation of S. mutans in a concentration-dependent manner. In Rg-treated biofilms, the expression levels of luxS, brpA, ffh, recA, nth, and smx were all decreased. Our results further showed that Rg was nontoxic, as Rg did not affect monocyte viability or lactate dehydrogenase activity in the exposed cells. These results suggested that Rg inhibited the biofilm formation of S. mutans, and the decrease in luxS, brpA, ffh, recA, nth, and smx expression might contribute to the antibacterial effects of Rg.
Collapse
Affiliation(s)
- Yaochao Zhang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China.
| | - Yong Zhu
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Yanping Zuo
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Chengfang Tang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Fang Zhou
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Xiaoming Cui
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Lin Wang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| |
Collapse
|
27
|
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
|
28
|
Zhang J, Chen C, Chen J, Zhou S, Zhao Y, Xu M, Xu H. Dual Mode of Anti-Biofilm Action of G3 against Streptococcus mutans. ACS APPLIED MATERIALS & INTERFACES 2020; 12:27866-27875. [PMID: 32484655 DOI: 10.1021/acsami.0c00771] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Oral biofilms, formed by multiple microorganisms and their extracellular polymeric substances, seriously affect people's life. The emergence of the resistance of biofilms to conventional antibiotics and their side effects on the oral cavity have posed a great challenge in the treatment of dental diseases. Recently, antimicrobial peptides have been recognized as promising alternatives to conventional antibiotics due to their broad antibacterial spectrum, high antibacterial activity, and specific mechanism. However, the research of their anti-biofilm behaviors is still in its infancy, and the underlying mechanism remains unclear. In this study, we investigated the anti-biofilm activities of a designed helical peptide (G3) against Streptococcus mutans (S. mutans), one of the primary causative pathogens of caries. The results indicated that G3 inhibited S. mutans biofilm formation by interfering with different stages of biofilm development. At the initial stage, G3 inhibited the bacterial adhesion by decreasing the bacterial surface charges, hydrophobicity, membrane integrity, and adhesion-related gene transcription. At the later stage, G3 interacted with extracellular DNA to destabilize the 3D architecture of mature biofilms and thus dispersed them. The high activity of G3 against S. mutans biofilms, along with its specific modes of action, endows it great application potential in preventing and treating dental plaque diseases.
Collapse
Affiliation(s)
- Jiangyu Zhang
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Cuixia Chen
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Jiaxi Chen
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Shasha Zhou
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Yurong Zhao
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Minglu Xu
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| | - Hai Xu
- Centre for Bioengineering and Biotechnology, China University of Petroleum (East China), 66 Changjiang West Road, Qingdao 266580, China
| |
Collapse
|
29
|
Kuebutornye FKA, Abarike ED, Lu Y, Hlordzi V, Sakyi ME, Afriyie G, Wang Z, Li Y, Xie CX. Mechanisms and the role of probiotic Bacillus in mitigating fish pathogens in aquaculture. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:819-841. [PMID: 31953625 DOI: 10.1007/s10695-019-00754-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/26/2019] [Indexed: 06/10/2023]
Abstract
Diseases are natural components of the environment, and many have economic implications for aquaculture and fisheries. Aquaculture is a fast-growing industry with the aim to meet the high protein demand of the ever-increasing global population; however, the emergence of diseases is a major setback to the industry. Probiotics emerged as a better solution to curb the disease problem in aquaculture among many alternatives. Probiotic Bacillus has been proven to better combat a wide range of fish pathogens relative to other probiotics in aquaculture; therefore, understanding the various mechanisms used by Bacillus in combating diseases will help improve their mode of action hence yielding better results in their combat against pathogens in the aquaculture industry. Thus, an overview of the mechanisms (production of bacteriocins, suppression of virulence gene expression, competition for adhesion sites, production of lytic enzymes, production of antibiotics, immunostimulation, competition for nutrients and energy, and production of organic acids) used by Bacillus probiotics in mitigating fish pathogens ranging from Aeromonas, Vibrio, Streptococcus, Yersinia, Pseudomonas, Clostridium, Acinetobacter, Edwardsiella, Flavobacterium, white spot syndrome virus, and infectious hypodermal and hematopoietic necrosis virus proven to be mitigated by Bacillus have been provided.
Collapse
Affiliation(s)
- Felix K A Kuebutornye
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Emmanuel Delwin Abarike
- Department of Fisheries and Aquatic Resources Management, University for Development Studies, Tamale, Ghana
| | - Yishan Lu
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China.
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China.
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China.
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China.
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China.
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China.
| | - Vivian Hlordzi
- Laboratory of Aquatic Animal Nutrition and Feed, College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong, China
| | - Michael Essien Sakyi
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Gyamfua Afriyie
- Key Laboratory of Aquaculture in South China Sea for Aquatic Economic Animal of Guangdong Higher Education Institutes, Fisheries College, Guangdong Ocean University, Zhanjiang, 524025, China
| | - Zhiwen Wang
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Yuan Li
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China
| | - Cai Xia Xie
- College of Fisheries, Guangdong Ocean University, Huguang Yan East, Zhanjiang, 524088, Guangdong Province, China
- Shenzhen Institute of Guangdong Ocean University, Shenzhen, 518120, Guangdong, China
- Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Animals, Zhanjiang, 524088, China
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, Shenzhen, 518120, China
- Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen, 518120, China
- Guangdong Key Laboratory of Control for Diseases of Aquatic Economic Animals, Zhanjiang, 524088, China
| |
Collapse
|
30
|
Expression of Meiothermus ruber luxS in E. coli alters the antibiotic susceptibility and biofilm formation. Appl Microbiol Biotechnol 2020; 104:4457-4469. [PMID: 32215705 DOI: 10.1007/s00253-020-10480-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/28/2020] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
Abstract
Quorum sensing (QS) and signal molecules used for interspecies communication are well defined in mesophiles, but there is still a plethora of microorganisms in which existence and mechanisms of QS need to be explored, thermophiles being among them. In silico analysis has revealed the presence of autoinducer-2 (AI-2) class of QS signaling molecules in thermophiles, synthesized by LuxS (AI-2 synthase), though the functions of this system are not known. In this study, LuxS of Meiothermus ruber was used for understanding the mechanism and functions of AI-2 based QS among thermophilic bacteria. The luxS gene of M. ruber was expressed in luxS- deletion mutant of Escherichia coli. Complementation of luxS resulted in significant AI-2 activity, enhanced biofilm formation, and antibiotic susceptibility. Transcriptome analysis showed significant differential expression of 204 genes between the luxS-complemented and luxS- deletion mutant of E. coli. Majority of the genes regulated by luxS belonged to efflux pumps. This elucidation may contribute towards finding novel alternatives against incessant antibiotic resistance in bacteria.Key Points• Expression of luxS in luxS-E. coli resulted in increase in biofilm index. • Reduction in the MIC of antibiotics was observed after complementation of luxS. • Downregulation of efflux pump genes was observed after complementation of luxS. • Transcriptome analysis showed that 204 genes were differentially regulated significantly.
Collapse
|
31
|
Short-Chain N-Acylhomoserine Lactone Quorum-Sensing Molecules Promote Periodontal Pathogens in In Vitro Oral Biofilms. Appl Environ Microbiol 2020; 86:AEM.01941-19. [PMID: 31757829 DOI: 10.1128/aem.01941-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Acylhomoserine lactones (AHLs), the quorum-sensing (QS) signals produced by a range of Gram-negative bacteria, are involved in biofilm formation in many pathogenic and environmental bacteria. Nevertheless, the current paradigm excludes a role of AHLs in dental plaque formation, while other QS signals, such as AI-2 and autoinducer peptides, have been demonstrated to play an important role in biofilm formation and virulence-related gene expression in oral pathogens. In the present work, we have explored the effect of externally added AHLs on in vitro oral biofilm models for commensal, cariogenic, and periodontal dental plaque. While little effect on bacterial growth was observed, some AHLs specifically affected the lactic acid production and protease activity of the biofilms. Most importantly, the analysis of bacterial diversity in the biofilms showed that the addition of C6-homoserine lactone (C6-HSL) results in a shift toward a periodontal bacterial composition profile by increasing the relative presence of the orange-complex bacteria Peptostreptococcus and Prevotella These results point to a relevant role of AHL-mediated QS in dental plaque formation and might be involved in the development of dysbiosis, the mechanism of which should be further investigated. This finding potentially opens new opportunities for the prevention or treatment of the periodontal disease.IMPORTANCE Dental plaque is omnipresent in healthy oral cavities and part of our commensal microbial colonization. At the same time, dental plaque is the cause of the most common human diseases, caries and gum disease. Dental plaque consists of billions of microbes attached to the surface of your teeth. Communication among these microbes is pivotal for development of these complex communities yet poorly studied in dental plaque. In the present study, we show that a specific communication molecule induces changes within the community related to the development of gum disease. This finding suggests that interfering with microbial communication may represent an interesting novel strategy to prevent gum disease that should be further investigated.
Collapse
|
32
|
Ryu EJ, An SJ, Sim J, Sim J, Lee J, Choi BK. Use of d-galactose to regulate biofilm growth of oral streptococci. Arch Oral Biol 2020; 111:104666. [PMID: 31955046 DOI: 10.1016/j.archoralbio.2020.104666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/12/2019] [Accepted: 01/12/2020] [Indexed: 12/31/2022]
Abstract
In the oral microbial community, commensals can compete with pathogens and reduce their colonization in the oral cavity. A substance that can inhibit harmful bacteria and enrich beneficial bacteria is required to maintain oral health. The purpose of this study was to examine the effect of d-galactose on the biofilm formation of the cariogenic bacteria Streptococcus mutans and oral commensal streptococci and to evaluate their use in solution and in paste form. Biofilms of S. mutans, Streptococcus oralis, and Streptococcus mitis were formed on saliva-coated glass slips in the absence or presence of d-galactose and evaluated by staining with 1 % crystal violet. d-Galactose significantly inhibited the biofilm formation of S. mutans at concentrations ranging from 2 μM to 200 mM but increased the biofilm formation of S. oralis and S. mitis at concentrations of 2-200 mM. d-Galactose significantly inhibited three glucosyltransferase genes, gtfB, gtfC, and gtfD. The effect of d-galactose in the form of solution and paste was evaluated using bovine teeth. Pretreatment with 100 mM d-galactose on bovine teeth resulted in significantly reduced S. mutans biofilm formation. Our results suggest that d-galactose can be a candidate substance for the development of oral hygiene products to prevent caries by inhibiting the biofilm formation of S. mutans and simultaneously increasing the biofilm formation of commensal oral streptococci.
Collapse
Affiliation(s)
- Eun-Ju Ryu
- Quorum Bio Co., Ltd, Bioresearch Center 501-1, Seoul National University, Republic of Korea
| | - Sun-Jin An
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Republic of Korea
| | - Jaehyun Sim
- Quorum Bio Co., Ltd, Bioresearch Center 501-1, Seoul National University, Republic of Korea
| | - Jun Sim
- Quorum Bio Co., Ltd, Bioresearch Center 501-1, Seoul National University, Republic of Korea
| | - Julian Lee
- Department of Bioinformatics and Life Science, Soongsil University, Seoul 06978, Republic of Korea
| | - Bong-Kyu Choi
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Republic of Korea.
| |
Collapse
|
33
|
Yi J, Zhang D, Cheng Y, Tan J, Luo Y. The impact of Paenibacillus polymyxa HY96-2 luxS on biofilm formation and control of tomato bacterial wilt. Appl Microbiol Biotechnol 2019; 103:9643-9657. [PMID: 31686149 PMCID: PMC6867978 DOI: 10.1007/s00253-019-10162-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 08/27/2019] [Accepted: 09/28/2019] [Indexed: 12/24/2022]
Abstract
The focus of this study was to investigate the effects of luxS, a key regulatory gene of the autoinducer-2 (AI-2) quorum sensing (QS) system, on the biofilm formation and biocontrol efficacy against Ralstonia solanacearum by Paenibacillus polymyxa HY96-2. luxS mutants were constructed and assayed for biofilm formation of the wild-type (WT) strain and luxS mutants of P. polymyxa HY96-2 in vitro and in vivo. The results showed that luxS positively regulated the biofilm formation of HY96-2. Greenhouse experiments of tomato bacterial wilt found that from the early stage to late stage postinoculation, the biocontrol efficacy of the luxS deletion strain was the lowest with 50.70 ± 1.39% in the late stage. However, the luxS overexpression strain had the highest biocontrol efficacy with 75.66 ± 1.94% in the late stage. The complementation of luxS could restore the biocontrol efficacy of the luxS deletion strain with 69.84 ± 1.09% in the late stage, which was higher than that of the WT strain with 65.94 ± 2.73%. Therefore, we deduced that luxS could promote the biofilm formation of P. polymyxa HY96-2 and further promoted its biocontrol efficacy against R. solanacearum.
Collapse
Affiliation(s)
- Jincui Yi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Daojing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuejuan Cheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Jingjing Tan
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuanchan Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
34
|
Zhang B, Ku X, Zhang X, Zhang Y, Chen G, Chen F, Zeng W, Li J, Zhu L, He Q. The AI-2/ luxS Quorum Sensing System Affects the Growth Characteristics, Biofilm Formation, and Virulence of Haemophilus parasuis. Front Cell Infect Microbiol 2019; 9:62. [PMID: 30941317 PMCID: PMC6434701 DOI: 10.3389/fcimb.2019.00062] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Haemophilus parasuis (H. parasuis) is a kind of opportunistic pathogen of the upper respiratory tract of piglets. Under certain circumstances, virulent strains can breach the mucosal barrier and enter the bloodstream, causing severe Glässer's disease. Many virulence factors are found to be related to the pathogenicity of H. parasuis strain, but the pathogenic mechanism remains unclear. LuxS/AI-2, as a kind of very important quorum sensing system, affects the growth characteristics, biofilm formation, antibiotic production, virulence, and metabolism of different strains. In order to investigate the effect of luxS/AI-2 quorum sensing system on the virulence of H. parasuis, a deletion mutant strain (ΔluxS) and complemented strain (C-luxS) were constructed and characterized. The results showed that the luxS gene participated in regulating and controlling stress resistance, biofilm formation and virulence. Compared with wild-type strain, ΔluxS strain decreased the production of AI-2 molecules and the tolerance toward oxidative stress and heat shock, and it reduced the abilities of autoagglutination, hemagglutination, and adherence, whereas it increased the abilities to form biofilm in vitro. In vivo experiments showed that ΔluxS strain attenuated its virulence about 10-folds and significantly decreased its tissue burden of bacteria in mice, compared with the wild-type strain. Taken together, the luxS/AI-2 quorum sensing system in H. parasuis not only plays an important role in growth and biofilm formation, but also affects the pathogenicity of H. parasuis.
Collapse
Affiliation(s)
- Bingzhou Zhang
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xugang Ku
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xiaoqian Zhang
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yan Zhang
- College of Animal Sciences and Veterinary Medicine, Xinjiang Agricultural University, Urumqi, China
| | - Guo Chen
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Fangzhou Chen
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Wei Zeng
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jing Li
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Ling Zhu
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology, Division of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
35
|
Parrino B, Schillaci D, Carnevale I, Giovannetti E, Diana P, Cirrincione G, Cascioferro S. Synthetic small molecules as anti-biofilm agents in the struggle against antibiotic resistance. Eur J Med Chem 2019; 161:154-178. [PMID: 30347328 DOI: 10.1016/j.ejmech.2018.10.036] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Biofilm formation significantly contributes to microbial survival in hostile environments and it is currently considered a key virulence factor for pathogens responsible for serious chronic infections. In the last decade many efforts have been made to identify new agents able to modulate bacterial biofilm life cycle, and many compounds have shown interesting activities in inhibiting biofilm formation or in dispersing pre-formed biofilms. However, only a few of these compounds were tested using in vivo models for their clinical significance. Contrary to conventional antibiotics, most of the anti-biofilm compounds act as anti-virulence agents as they do not affect bacterial growth. In this review we selected the most relevant literature of the last decade, focusing on the development of synthetic small molecules able to prevent bacterial biofilm formation or to eradicate pre-existing biofilms of clinically relevant Gram-positive and Gram-negative pathogens. In addition, we provide a comprehensive list of the possible targets to counteract biofilm formation and development, as well as a detailed discussion the advantages and disadvantages of the different current biofilm-targeting strategies.
Collapse
Affiliation(s)
- Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Domenico Schillaci
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Ilaria Carnevale
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, via Paradisa, 56100, Pisa, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands; Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, via Paradisa, 56100, Pisa, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| |
Collapse
|
36
|
Di Domenico EG, Cavallo I, Bordignon V, D'Agosto G, Pontone M, Trento E, Gallo MT, Prignano G, Pimpinelli F, Toma L, Ensoli F. The Emerging Role of Microbial Biofilm in Lyme Neuroborreliosis. Front Neurol 2018; 9:1048. [PMID: 30559713 PMCID: PMC6287027 DOI: 10.3389/fneur.2018.01048] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/19/2018] [Indexed: 01/04/2023] Open
Abstract
Lyme borreliosis (LB) is the most common tick-borne disease caused by the spirochete Borrelia burgdorferi in North America and Borrelia afzelii or Borrelia garinii in Europe and Asia, respectively. The infection affects multiple organ systems, including the skin, joints, and the nervous system. Lyme neuroborreliosis (LNB) is the most dangerous manifestation of Lyme disease, occurring in 10-15% of infected individuals. During the course of the infection, bacteria migrate through the host tissues altering the coagulation and fibrinolysis pathways and the immune response, reaching the central nervous system (CNS) within 2 weeks after the bite of an infected tick. The early treatment with oral antimicrobials is effective in the majority of patients with LNB. Nevertheless, persistent forms of LNB are relatively common, despite targeted antibiotic therapy. It has been observed that the antibiotic resistance and the reoccurrence of Lyme disease are associated with biofilm-like aggregates in B. burgdorferi, B. afzelii, and B. garinii, both in vitro and in vivo, allowing Borrelia spp. to resist to adverse environmental conditions. Indeed, the increased tolerance to antibiotics described in the persisting forms of Borrelia spp., is strongly reminiscent of biofilm growing bacteria, suggesting a possible role of biofilm aggregates in the development of the different manifestations of Lyme disease including LNB.
Collapse
Affiliation(s)
- Enea Gino Di Domenico
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Ilaria Cavallo
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Valentina Bordignon
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Martina Pontone
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Elisabetta Trento
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Maria Teresa Gallo
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Grazia Prignano
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Fulvia Pimpinelli
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Luigi Toma
- Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, Regina Elena National Cancer Institute IRCCS, Rome, Italy
| | - Fabrizio Ensoli
- Clinical Pathology and Microbiology Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| |
Collapse
|
37
|
Lee K, Yu H, Zhang X, Choo KH. Quorum sensing and quenching in membrane bioreactors: Opportunities and challenges for biofouling control. BIORESOURCE TECHNOLOGY 2018; 270:656-668. [PMID: 30213542 DOI: 10.1016/j.biortech.2018.09.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/01/2018] [Accepted: 09/03/2018] [Indexed: 05/06/2023]
Abstract
Membrane biofouling, due to biofilm growth after planktonic bacteria attachment to a membrane, is a major bottleneck limiting the energy-efficient operation and maintenance of membrane bioreactors (MBRs). Microbial communications, known as quorum sensing (QS), are responsible for this biofouling behavior. Novel strategies for stopping this communication, known as quorum quenching (QQ), appear to be successful for biofouling control in MBRs used for wastewater treatment. This review describes recent information regarding the signal molecules and mechanisms responsible for QS behaviors, promising approaches for QQ (enzymatic, bacterial, fungal, photocatalytic, mimicking, and biostimulating methods), and efficient fabrication and use of QQ media for MBR applications. We discuss the opportunities and challenges of QQ techniques for their further improvement and practical use in MBRs.
Collapse
Affiliation(s)
- Kibaek Lee
- Advanced Institute of Water Industry, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Huarong Yu
- School of Municipal and Environmental Engineering, Harbin Institute of Technology, 73 Huanghe Road, Nangang, Harbin 150090, People's Republic of China
| | - Xiaolei Zhang
- Advanced Institute of Water Industry, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea
| | - Kwang-Ho Choo
- Advanced Institute of Water Industry, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea; Department of Environmental Engineering, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu 41566, Republic of Korea.
| |
Collapse
|
38
|
Bedree JK, Bor B, Cen L, Edlund A, Lux R, McLean JS, Shi W, He X. Quorum Sensing Modulates the Epibiotic-Parasitic Relationship Between Actinomyces odontolyticus and Its Saccharibacteria epibiont, a Nanosynbacter lyticus Strain, TM7x. Front Microbiol 2018; 9:2049. [PMID: 30319555 PMCID: PMC6166536 DOI: 10.3389/fmicb.2018.02049] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/13/2018] [Indexed: 11/13/2022] Open
Abstract
The ultra-small, obligate parasitic epibiont, TM7x, the first and only current member of the long-elusive Saccharibacteria (formerly the TM7 phylum) phylum to be cultivated, was isolated in co-culture with its bacterial host, Actinomyces odontolyticus subspecies actinosynbacter, XH001. Initial phenotypic characterization of the TM7x-associated XH001 co-culture revealed enhanced biofilm formation in the presence of TM7x compared to XH001 as monoculture. Genomic analysis and previously published transcriptomic profiling of XH001 also revealed the presence of a putative AI-2 quorum sensing (QS) operon, which was highly upregulated upon association of TM7x with XH001. This analysis revealed that the most highly induced gene in XH001 was an lsrB ortholog, which encodes a putative periplasmic binding protein for the auto inducer (AI)-2 QS signaling molecule. Further genomic analyses suggested the lsrB operon in XH001 is a putative hybrid AI-2/ribose transport operon as well as the existence of a luxS ortholog, which encodes the AI-2 synthase. In this study, the potential role of AI-2 QS in the epibiotic-parasitic relationship between XH001 and TM7x in the context of biofilm formation was investigated. A genetic system for XH001 was developed to generate lsrB and luxS gene deletion mutants in XH001. Phenotypic characterization demonstrated that deletion mutations in either lsrB or luxS did not affect XH001's growth dynamic, mono-species biofilm formation capability, nor its ability to associate with TM7x. TM7x association with XH001 induced lsrB gene expression in a luxS-dependent manner. Intriguingly, unlike wild type XH001, which displayed significantly increased biofilm formation upon establishing the epibiotic-parasitic relationship with TM7x, XH001ΔlsrB, and XH001ΔluxS mutants failed to achieve enhanced biofilm formation when associated with TM7x. In conclusion, we demonstrated a significant role for AI-2 QS in modulating dual-species biofilm formation when XH001 and TM7x establish their epibiotic-parasitic relationship.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| | - Batbileg Bor
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| | - Lujia Cen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| | - Anna Edlund
- Department of Genomic Medicine, J. Craig Venter Institute, La Jolla, CA, United States
| | - Renate Lux
- Section of Periodontics, Division of Constitutive and Regenerative Sciences, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA, United States
| | - Wenyuan Shi
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, United States
| |
Collapse
|
39
|
Ben Amara H, Song HY, Ryu E, Park JS, Schwarz F, Kim BM, Choi BK, Koo KT. Effects of quorum-sensing inhibition on experimental periodontitis induced by mixed infection in mice. Eur J Oral Sci 2018; 126:449-457. [PMID: 30230039 DOI: 10.1111/eos.12570] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
This study aimed to verify, in in vivo settings, whether quorum-sensing inhibition molecules could attenuate alveolar bone loss induced by Porphyromonas gingivalis/Fusobacterium nucleatum co-infection and reduce the bacterial colonization of periodontal tissues. In BALB/c mice, periodontitis was induced through oral inoculation with P. gingivalis and F. nucleatum six times during a 42-d period. Quorum sensing inhibitors (a furanone compound and D-ribose) were administered simultaneously with bacterial infection. Linear and volumetric modifications of interproximal alveolar bone levels were compared between groups using micro-computed tomography. Total bacteria, and P. gingivalis and F. nucleatum DNA in periodontal tissues, were quantified using real-time PCR. Radiographic linear measurements demonstrated a significant reduction of alveolar bone loss, of approximately 40%, in mice treated with quorum sensing inhibitors when compared with the co-infection group. This was confirmed by a significant increase of residual bone volume in the test group. While total bacterial genes in the treatment group significantly decreased by 93% in periodontal tissue samples when quorum sensing inhibitors were administered, no significant differences of P. gingivalis DNA were found. Quorum sensing inhibitors reduced periodontal breakdown and bacterial infection in periodontal tissues after co-infection with P. gingivalis and F. nucleatum.
Collapse
Affiliation(s)
- Heithem Ben Amara
- Department of Periodontology and Dental Research Institute, Translational Research Laboratory for Tissue Engineering (TTE), School of Dentistry, Seoul National University, Seoul, Korea
| | - Hyun Y Song
- Department of Periodontology and Dental Research Institute, Translational Research Laboratory for Tissue Engineering (TTE), School of Dentistry, Seoul National University, Seoul, Korea
| | - Eunju Ryu
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Korea
| | - Ji S Park
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Frank Schwarz
- Department of Oral Surgery, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
| | - Byeong M Kim
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Bong-Kyu Choi
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Korea
| | - Ki-Tae Koo
- Department of Periodontology and Dental Research Institute, Translational Research Laboratory for Tissue Engineering (TTE), School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
40
|
Influence of naringenin on the biofilm formation of Streptococcus mutans. J Dent 2018; 76:24-31. [DOI: 10.1016/j.jdent.2018.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/03/2018] [Accepted: 04/15/2018] [Indexed: 12/18/2022] Open
|
41
|
Wang Y, Wang Y, Sun L, Grenier D, Yi L. The LuxS/AI-2 system of Streptococcus suis. Appl Microbiol Biotechnol 2018; 102:7231-7238. [PMID: 29938319 DOI: 10.1007/s00253-018-9170-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 11/24/2022]
Abstract
Quorum sensing (QS) is an important protective mechanism that allows bacteria to adapt to its environment. A limited number of signal molecules play the key role of transmitting information in this mechanism. Signals are transmitted between individual bacterium through QS systems, resulting in the expression of specific genes. QS plays an important role in a variety of bacterial processes, including drug resistance, biofilm formation, motility, adherence, and virulence. Most Gram-positive and Gram-negative bacteria possess QS systems, mainly the LuxS/AI-2-mediated QS system. Evidence has been brought that LuxS/AI-2 system controls major virulence determinants in Streptococcus suis and, as such, the ability of this bacterial species to cause infections in humans and pigs. Understanding the S. suis LuxS/AI-2 system may open up novel avenues for decreasing the drug resistance and infectivity of S. suis. This article focuses on the progress made to date on the S. suis LuxS/AI-2-mediated QS system.
Collapse
Affiliation(s)
- Yang Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China.
| | - Yuxin Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Liyun Sun
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang, China
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada
| | - Li Yi
- Groupe de Recherche en Écologie Buccale (GREB), Faculté de Médecine Dentaire, Université Laval, Quebec City, QC, Canada. .,College of Life Science, Luoyang Normal University, Luoyang, China.
| |
Collapse
|
42
|
Lim SY, Teh CSJ, Thong KL. Biofilm-Related Diseases and Omics: Global Transcriptional Profiling of Enterococcus faecium Reveals Different Gene Expression Patterns in the Biofilm and Planktonic Cells. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2018; 21:592-602. [PMID: 29049010 DOI: 10.1089/omi.2017.0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Enterococcus faecium is an opportunistic pathogen with a remarkable ability to acquire resistance toward multiple antibiotics, including those of last-resort drugs such as vancomycin and daptomycin. The occurrence of vancomycin-resistant E. faecium is on the rise and there is a need to understand the virulence of this organism. One of the factors that contributes to the virulence is the ability to form biofilms. Since bacteria in biofilm state are more resistant to antibiotics and host immune response, understanding the molecular mechanism of biofilm development is important to control biofilm-related diseases. The aim of this study was to determine the global gene expression profiles of an E. faecium strain, VREr5, during the early event of sessile growth compared with its planktonic phase through RNA-sequencing approach. The results clearly illustrated distinct expression profiles of the planktonic and biofilm cells. A total of 177 genes were overexpressed in the biofilm cells. Most of them encode for proteins involved in adherence, such as the ebpABCfm locus. Genes associated with plasmid replication, gene exchange, and protein synthesis were also upregulated during the early event of biofilm development. Furthermore, the transcriptome analysis also identified genes such as fsrB, luxS, and spx that might suppress biofilm formation in VREr5. The putative biofilm-related bee locus was found to be downregulated. These new findings could provide caveats for future studies on the regulation and maintenance of biofilm and development of biomarkers for biofilm-related diseases.
Collapse
Affiliation(s)
- Shu Yong Lim
- 1 Institute of Biological Sciences, Faculty of Science, University of Malaya , Kuala Lumpur, Malaysia
| | - Cindy Shuan Ju Teh
- 2 Department of Medical Microbiology, Faculty of Medicine, University of Malaya , Kuala Lumpur, Malaysia
| | - Kwai Lin Thong
- 1 Institute of Biological Sciences, Faculty of Science, University of Malaya , Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Hu X, Wang Y, Gao L, Jiang W, Lin W, Niu C, Yuan K, Ma R, Huang Z. The Impairment of Methyl Metabolism From luxS Mutation of Streptococcus mutans. Front Microbiol 2018; 9:404. [PMID: 29657574 PMCID: PMC5890193 DOI: 10.3389/fmicb.2018.00404] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/21/2018] [Indexed: 12/19/2022] Open
Abstract
The luxS gene is present in a wide range of bacteria and is involved in many cellular processes. LuxS mutation can cause autoinducer(AI)-2 deficiency and methyl metabolism disorder. The objective of this study was to demonstrate that, in addition to AI-2-mediated quorum sensing (QS), methyl metabolism plays an important role in LuxS regulation in Streptococcus mutans. The sahH gene from Pseudomonas aeruginosa was amplified and introduced into the S. mutans luxS-null strain to complement the methyl metabolism disruption in a defective QS phenotype. The intracellular activated methyl cycle (AMC) metabolites [S-adenosylmethionine (SAM), S-adenosylhomocysteine (SAH), homocysteine (HCY), and methionine] were quantified in wild-type S. mutans and its three derivatives to determine the metabolic effects of disrupting the AMC. Biofilm mass and structure, acid tolerance, acid production, exopolysaccharide synthesis of multispecies biofilms and the transcriptional level of related genes were determined. The results indicated that SAH and SAM were relatively higher in S. mutans luxS-null strain and S. mutans luxS null strain with plasmid pIB169 when cultured overnight, and HCY was significantly higher in S. mutans UA159. Consistent with the transcriptional profile, luxS deletion-mediated impairment of biofilm formation and acid tolerance was restored to wild-type levels using transgenic SahH. These results also suggest that methionine methyl metabolism contributes to LuxS regulation in S. mutans to a significant degree.
Collapse
Affiliation(s)
- Xuchen Hu
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Yuxia Wang
- Department of Endodontics, Tianjin Stomatological Hospital, Nankai University, Tianjin, China
| | - Li Gao
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Wenxin Jiang
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Wenzhen Lin
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Chenguang Niu
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Keyong Yuan
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Rui Ma
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| | - Zhengwei Huang
- Department of Endodontics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, China
| |
Collapse
|
44
|
Muras A, Mayer C, Romero M, Camino T, Ferrer MD, Mira A, Otero A. Inhibition of Steptococcus mutans biofilm formation by extracts of Tenacibaculum sp. 20J, a bacterium with wide-spectrum quorum quenching activity. J Oral Microbiol 2018; 10:1429788. [PMID: 29410771 PMCID: PMC5795696 DOI: 10.1080/20002297.2018.1429788] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023] Open
Abstract
Background: Previous studies have suggested the quorum sensing signal AI-2 as a potential target to prevent the biofilm formation by Streptococcus mutans, a pathogen involved in tooth decay. Objective: To obtain inhibition of biofilm formation by S. mutans by extracts obtained from the marine bacterium Tenacibaculum sp. 20J interfering with the AI-2 quorum sensing system. Design: The AI-2 inhibitory activity was tested with the biosensors Vibrio harveyi BB170 and JMH597. S. mutans ATCC25175 biofilm formation was monitored using impedance real-time measurements with the xCELLigence system®, confocal laser microscopy, and the crystal violet quantification method. Results: The addition of the cell extract from Tenacibaculum sp. 20J reduced biofilm formation in S. mutans ATCC25175 by 40–50% compared to the control without significantly affecting growth. A decrease of almost 40% was also observed in S. oralis DSM20627 and S. dentisani 7747 biofilms. Conclusions: The ability of Tenacibaculum sp. 20J to interfere with AI-2 and inhibit biofilm formation in S. mutans was demonstrated. The results indicate that the inhibition of quorum sensing processes may constitute a suitable strategy for inhibiting dental plaque formation, although additional experiments using mixed biofilm models would be required.
Collapse
Affiliation(s)
- Andrea Muras
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Celia Mayer
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Manuel Romero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Tamara Camino
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria D Ferrer
- Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Alex Mira
- Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Ana Otero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
45
|
Effects of sub-minimum inhibitory concentrations of lemon essential oil on the acid tolerance and biofilm formation of Streptococcus mutans. Arch Oral Biol 2017; 87:235-241. [PMID: 29331510 DOI: 10.1016/j.archoralbio.2017.12.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Lemon essential oil (LEO) is a kind of secondary metabolite from lemon peels and has been found to inhibit cariogenic bacteria for decades. However, its effects on main cariogenic virulence factors are rarely reported. The present study aimed to investigate the effects of sub-minimum inhibitory concentrations (sub-MICs) of LEO on the acid tolerance and biofilm formation of Streptococcus mutans (S. mutans) and preliminarily reveal the possible underlying mechanisms. DESIGNS Effects of LEO on the acid tolerance and biofilm formation of S. mutans were investigated by the broth dilution method and crystal violet staining method respectively. The expression of luxS, srtA and spaP gene was also determined to explore the underlying mechanism. In addition, Tea polyphenols (TP), a major natural inhibitor of cariogenic virulence factors, and limonene (LIM), the major component of LEO, were selected as comparisons to evaluate the effects of LEO. RESULTS Sub-MICs of LEO, LIM and TP exhibited a dose-dependent inhibition of growth of S. mutans at pH ranging from 4.0 to 7.0. The formation of S. mutans biofilm was remarkably inhibited and the inhibitory rates of LEO, LIM and TP were 97.87%, 94.88% and 96.01% respectively at 1/2 MIC. Similarly, a down-regulation was observed in the expression of luxS, srtA and spaP gene at sub-MIC levels. CONCLUSIONS Effects of LEO were similar or slightly stronger than LIM and TP, suggesting that LEO might represent a novel, natural anticarious agent that inhibited the specific genes associated with bacterial acid tolerance and biofilm formation without necessarily affecting the growth of oral bacteria.
Collapse
|
46
|
Actinobacillus pleuropneumoniae biofilms: Role in pathogenicity and potential impact for vaccination development. Anim Health Res Rev 2017; 19:17-30. [DOI: 10.1017/s146625231700010x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AbstractActinobacillus pleuropneumoniae is a Gram-negative bacterium that belongs to the family Pasteurellaceae. It is the causative agent of porcine pleuropneumonia, a highly contagious respiratory disease that is responsible for major economic losses in the global pork industry. The disease may present itself as a chronic or an acute infection characterized by severe pathology, including hemorrhage, fibrinous and necrotic lung lesions, and, in the worst cases, rapid death. A. pleuropneumoniae is transmitted via aerosol route, direct contact with infected pigs, and by the farm environment. Many virulence factors associated with this bacterium are well characterized. However, much less is known about the role of biofilm, a sessile mode of growth that may have a critical impact on A. pleuropneumoniae pathogenicity. Here we review the current knowledge on A. pleuropneumoniae biofilm, factors associated with biofilm formation and dispersion, and the impact of biofilm on the pathogenesis A. pleuropneumoniae. We also provide an overview of current vaccination strategies against A. pleuropneumoniae and consider the possible role of biofilms vaccines for controlling the disease.
Collapse
|
47
|
Engevik MA, Versalovic J. Biochemical Features of Beneficial Microbes: Foundations for Therapeutic Microbiology. Microbiol Spectr 2017; 5:10.1128/microbiolspec.BAD-0012-2016. [PMID: 28984235 PMCID: PMC5873327 DOI: 10.1128/microbiolspec.bad-0012-2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Indexed: 12/15/2022] Open
Abstract
Commensal and beneficial microbes secrete myriad products which target the mammalian host and other microbes. These secreted substances aid in bacterial niche development, and select compounds beneficially modulate the host and promote health. Microbes produce unique compounds which can serve as signaling factors to the host, such as biogenic amine neuromodulators, or quorum-sensing molecules to facilitate inter-bacterial communication. Bacterial metabolites can also participate in functional enhancement of host metabolic capabilities, immunoregulation, and improvement of intestinal barrier function. Secreted products such as lactic acid, hydrogen peroxide, bacteriocins, and bacteriocin-like substances can also target the microbiome. Microbes differ greatly in their metabolic potential and subsequent host effects. As a result, knowledge about microbial metabolites will facilitate selection of next-generation probiotics and therapeutic compounds derived from the mammalian microbiome. In this article we describe prominent examples of microbial metabolites and their effects on microbial communities and the mammalian host.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| | - James Versalovic
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030 and Department of Pathology, Texas Children's Hospital, Houston, TX 77030
| |
Collapse
|
48
|
Wang X, Li X, Ling J. Streptococcus gordonii LuxS/autoinducer-2 quorum-sensing system modulates the dual-species biofilm formation with Streptococcus mutans. J Basic Microbiol 2017; 57:605-616. [PMID: 28485524 DOI: 10.1002/jobm.201700010] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/04/2017] [Accepted: 04/21/2017] [Indexed: 01/26/2023]
Abstract
Dental plaques are mixed-species biofilms that are related to the development of dental caries. Streptococcus mutans (S. mutans) is an important cariogenic bacterium that forms mixed-species biofilms with Streptococcus gordonii (S. gordonii), an early colonizer of the tooth surface. The LuxS/autoinducer-2(AI-2) quorum sensing system is involved in the regulation of mixed-species biofilms, and AI-2 is proposed as a universal signal for the interaction between bacterial species. In this work, a S. gordonii luxS deficient strain was constructed to investigate the effect of the S. gordonii luxS gene on dual-species biofilm formed by S. mutans and S. gordonii. In addition, AI-2 was synthesized in vitro by incubating recombinant LuxS and Pfs enzymes of S. gordonii together. The effect of AI-2 on S. mutans single-species biofilm formation and cariogenic virulence gene expression were also assessed. The results showed that luxS disruption in S. gordonii altered dual-species biofilm formation, architecture, and composition, as well as the susceptibility to chlorhexidine. And the in vitro synthesized AI-2 had a concentration-dependent effect on S. mutans biofilm formation and virulence gene expression. These findings indicate that LuxS/AI-2 quorum-sensing system of S. gordonii plays a role in regulating the dual-species biofilm formation with S. mutans.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Xiaolan Li
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Junqi Ling
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
49
|
Leal CRL, Alvarenga LH, Oliveira-Silva T, Kato IT, Godoy-Miranda B, Bussadori SK, Ribeiro MS, Prates RA. Antimicrobial photodynamic therapy on Streptococcus mutans is altered by glucose in the presence of methylene blue and red LED. Photodiagnosis Photodyn Ther 2017; 19:1-4. [PMID: 28414082 DOI: 10.1016/j.pdpdt.2017.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/24/2017] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND Dental caries are a multifactorial disease that progressively produces tooth destruction as a result of bacterial colonization of enamel surface, especially Streptococcus mutans. The objective of this work was to investigate the role of glucose in antimicrobial photodynamic therapy (aPDT) on S. mutans. METHODS S. mutans ATCC 25175 were cultured on microaerophilia at 37°C for 48h, and we tested aPDT in the presence of 50mM glucose. Bacterial suspension was used to investigate aPDT with 100μM methylene blue (MB) under LED emitting radiation at ʎ=660nm and parameters as following (P=473 mW; I=166.8 mW/cm2, and doses of 5, 10 and 20J/cm2). A seventy-two hours biofilm was grown on 96 flat buttoned well-plate and irradiation was performed from 10 to 80J/cm2 at similar conditions. RESULTS There was no dark toxicity nor bacterial death regarding LED irradiation on suspension and on biofilm. Nevertheless, aPDT presented expressive bacterial inactivation following 1 and 2min of irradiation on cell suspension. On the other hand, there was no inactivation in the presence of glucose under the same conditions. Biofilm was completely inactivated by MB-mediated aPDT after 6min of irradiation. However, the presence of glucose delayed the complete inactivation of the biofilm. CONCLUSION The presence of glucose in the suspension drastically delayed the effect of aPDT on S. mutans and this effect is more pronounced in bacterial suspension than on biofilm.
Collapse
Affiliation(s)
- Cintia R Lima Leal
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Letícia H Alvarenga
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil.
| | - Tamires Oliveira-Silva
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Ilka T Kato
- Department of Biomedical Engineering, ABC Federal University (UFABC), Santo Andre, SP, Brazil
| | - Bianca Godoy-Miranda
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Sandra K Bussadori
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil
| | - Martha S Ribeiro
- Center for Lasers and Applications, IPEN-CNEN/SP, São Paulo, SP, Brazil
| | - Renato A Prates
- Dep. of Biophotonics, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil; School of Dentistry, Universidade Nove de Julho (UNINOVE), São Paulo, Brazil.
| |
Collapse
|
50
|
Ou M, Ling J. Norspermidine changes the basic structure of S. mutans biofilm. Mol Med Rep 2016; 15:210-220. [PMID: 27922663 PMCID: PMC5355703 DOI: 10.3892/mmr.2016.5979] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 10/26/2016] [Indexed: 01/08/2023] Open
Abstract
The factors regulating the assembly of the three-dimensional structure of Streptococcus mutans biofilms remain obscure. Polyamines are essential in biofilm formation of certain bacteria. Norspermidine, an unusual polyamine, has been a controversial polyamine that can lead to biofilm disassembly. However, the role of norspermidine in S. mutans biofilms remains unknown. Therefore, the present study investigated the impact of norspermidine on S. mutans biofilms. The different architectures of the biofilms in norspermidine and control groups indicated that the basic units, bacteria-exopolysaccharide units (BEUs), represent the exopolysaccharide (EPS) and bacterial assembly pattern in S. mutans biofilms. In addition, norspermidine inhibited S. mutans biofilm formation and changed the basic composition of the biofilm, which led to an unusual EPS architecture. Therefore, 5 mM norspermidine inhibited biofilm formation both by decreasing the rate of cell viability and changing the biofilm structure. Gene-expression microarray analysis indicated that the formation of an irregular architecture in the norspermidine group was potentially attributable to the downregulation of elements of the quorum-sensing system (by 2.7–15-fold). The present study suggested that the BEUs are a basic structure of S. mutans biofilm and its assembly is regulated majorly by the quorum-sensing system. Norspermidine can lead to structure change in BEUs by influencing S. mutans quorum-sensing system.
Collapse
Affiliation(s)
- Meizhen Ou
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Provincial Key Laboratory of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| | - Junqi Ling
- Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Provincial Key Laboratory of Stomatology, Sun Yat‑sen University, Guangzhou, Guangdong 510055, P.R. China
| |
Collapse
|