1
|
Geng F, Liu J, Liu J, Lu Z, Pan Y. Recent progress in understanding the role of bacterial extracellular DNA: focus on dental biofilm. Crit Rev Microbiol 2024:1-19. [PMID: 39648406 DOI: 10.1080/1040841x.2024.2438117] [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: 01/31/2024] [Revised: 05/11/2024] [Accepted: 11/30/2024] [Indexed: 12/10/2024]
Abstract
Dental biofilm is a highly complicated and dynamic structure comprising not only microbial communities but also the surrounding matrix of extracellular polymeric substances (EPS), including polysaccharides, proteins, extracellular DNA (eDNA) and other biopolymers. In recent years, the important role of bacterial eDNA in dental biofilms has gradually attracted attention. In this review, we present recent studies on the presence, dynamic conformation and release of oral bacterial eDNA. Moreover, updated information on functions associated with oral bacterial eDNA in biofilm formation, antibiotic resistance, activation of the immune system and immune evasion is highlighted. Finally, we summarize the role of oral bacterial eDNA as a promising target for the treatment of oral diseases. Increasing insight into the versatile roles of bacterial eDNA in dental biofilms will facilitate the prevention and treatment of biofilm-induced oral infections.
Collapse
Affiliation(s)
- Fengxue Geng
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Junchao Liu
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Jinwen Liu
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Ze Lu
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yaping Pan
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
- Liaoning Provincial Key Laboratory of Oral Diseases, Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
2
|
Zayed N, Vertommen R, Simoens K, Bernaerts K, Boon N, Srivastava MG, Braem A, Van Holm W, Castro AB, Teughels W. How well do antimicrobial mouth rinses prevent dysbiosis in an in vitro periodontitis biofilm model? J Periodontol 2024; 95:880-891. [PMID: 38946115 DOI: 10.1002/jper.23-0674] [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: 11/11/2023] [Revised: 02/02/2024] [Accepted: 02/11/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND Periodontal diseases are associated with dysbiosis in the oral microbial communities. Managing oral biofilms is therefore key for preventing these diseases. Management protocols often include over-the-counter antimicrobial mouth rinses, which lack data on their effects on the oral microbiome's ecology, bacterial composition, metabolic activity, and dysbiosis resilience. This study examined the efficacy of antimicrobial mouth rinses to halt dysbiosis in in vitro oral biofilms under periodontitis-simulating conditions. METHODS Multispecies oral biofilms were grown on hydroxyapatite discs (HADs) and rinsed daily with one of six mouth rinses. Positive and negative controls were included. After three rinses, biofilms were analyzed with viability quantitative polymerase chain reaction and visualized using scanning electron microscopy. Supernatants of rinsed biofilms were used for metabolic activity analysis. In addition, human oral keratinocytes were exposed to rinsed biofilms to assess their inflammatory response. All outputs were analyzed for correlation using Spearman coefficient. RESULTS Product-related changes were observed in the rinsed biofilms. Three of the six tested mouth rinses could significantly prevent dysbiosis with ≥30% reduction in pathobiont abundance relative to the control. These biofilms had lower metabolic activity, and the exposed human oral keratinocyte produced less interleukin-8. Interleukin-8 production correlated to both pathobiont quantity and the metabolic activity of the biofilms. CONCLUSION Some mouth rinses could support biofilm resilience and stop dysbiosis evolution in the biofilm model, with a clear product-related effect. Such mouth rinses can be considered for patients under maintenance/supportive periodontal therapy to prevent/delay disease recurrence. Others are more useful for different periodontal therapy stages.
Collapse
Affiliation(s)
- Naiera Zayed
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Rik Vertommen
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Kenneth Simoens
- Chemical and Biochemical Reactor Engineering and Safety, Department of Chemical Engineering, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Kristel Bernaerts
- Chemical and Biochemical Reactor Engineering and Safety, Department of Chemical Engineering, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Nico Boon
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Mrinal Gaurav Srivastava
- Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Annabel Braem
- Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Wannes Van Holm
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Ana B Castro
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Wim Teughels
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
3
|
Kaplan JB, Sukhishvili SA, Sailer M, Kridin K, Ramasubbu N. Aggregatibacter actinomycetemcomitans Dispersin B: The Quintessential Antibiofilm Enzyme. Pathogens 2024; 13:668. [PMID: 39204268 PMCID: PMC11357414 DOI: 10.3390/pathogens13080668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
The extracellular matrix of most bacterial biofilms contains polysaccharides, proteins, and nucleic acids. These biopolymers have been shown to mediate fundamental biofilm-related phenotypes including surface attachment, intercellular adhesion, and biocide resistance. Enzymes that degrade polymeric biofilm matrix components, including glycoside hydrolases, proteases, and nucleases, are useful tools for studying the structure and function of biofilm matrix components and are also being investigated as potential antibiofilm agents for clinical use. Dispersin B is a well-studied, broad-spectrum antibiofilm glycoside hydrolase produced by Aggregatibacter actinomycetemcomitans. Dispersin B degrades poly-N-acetylglucosamine, a biofilm matrix polysaccharide that mediates biofilm formation, stress tolerance, and biocide resistance in numerous Gram-negative and Gram-positive pathogens. Dispersin B has been shown to inhibit biofilm and pellicle formation; detach preformed biofilms; disaggregate bacterial flocs; sensitize preformed biofilms to detachment by enzymes, detergents, and metal chelators; and sensitize preformed biofilms to killing by antiseptics, antibiotics, bacteriophages, macrophages, and predatory bacteria. This review summarizes the results of nearly 100 in vitro and in vivo studies that have been carried out on dispersin B since its discovery 20 years ago. These include investigations into the biological function of the enzyme, its structure and mechanism of action, and its in vitro and in vivo antibiofilm activities against numerous bacterial species. Also discussed are potential clinical applications of dispersin B.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
| | - Svetlana A. Sukhishvili
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX 77843, USA;
| | | | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel;
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Narayanan Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ 07103, USA;
| |
Collapse
|
4
|
Naseef Pathoor N, Viswanathan A, Wadhwa G, Ganesh PS. Understanding the biofilm development of Acinetobacter baumannii and novel strategies to combat infection. APMIS 2024; 132:317-335. [PMID: 38444124 DOI: 10.1111/apm.13399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/22/2024] [Indexed: 03/07/2024]
Abstract
Acinetobacter baumannii (A. baumannii) is a Gram-negative, nonmotile, and aerobic bacillus emerged as a superbug, due to increasing the possibility of infection and accelerating rates of antimicrobial agents. It is recognized as a nosocomial pathogen due to its ability to form biofilms. These biofilms serve as a defensive barrier, increase antibiotic resistance, and make treatment more difficult. As a result, the current situation necessitates the rapid emergence of novel therapeutic approaches to ensure successful treatment outcomes. This review explores the intricate relationship between biofilm formation and antibiotic resistance in A. baumannii, emphasizing the role of key virulence factors and quorum sensing (QS) mechanisms that will lead to infections and facilitate insight into developing innovative method to control A. baumannii infections. Furthermore, the review article looks into promising approaches for preventing biofilm formation on medically important surfaces and potential therapeutic methods for eliminating preformed biofilms, which can address biofilm-associated A. baumannii infections. Modern advances in emerging therapeutic options such as antimicrobial peptide (AMPs), nanoparticles (NPs), bacteriophage therapy, photodynamic therapy (PDT), and other biofilm inhibitors can assist readers understand the current landscape and future prospects for effectively treating A. baumannii biofilm infections.
Collapse
Affiliation(s)
- Naji Naseef Pathoor
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| | - Akshaya Viswanathan
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| | - Gulshan Wadhwa
- Department of Biotechnology, Ministry of Science and Technology, New Delhi, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, India
| |
Collapse
|
5
|
Zafer MM, Mohamed GA, Ibrahim SRM, Ghosh S, Bornman C, Elfaky MA. Biofilm-mediated infections by multidrug-resistant microbes: a comprehensive exploration and forward perspectives. Arch Microbiol 2024; 206:101. [PMID: 38353831 PMCID: PMC10867068 DOI: 10.1007/s00203-023-03826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/30/2023] [Indexed: 02/16/2024]
Abstract
A biofilm is a collection of microorganisms organized in a matrix of extracellular polymeric material. Biofilms consist of microbial cells that attach to both surfaces and each other, whether they are living or non-living. These microbial biofilms can lead to hospital-acquired infections and are generally detrimental. They possess the ability to resist the human immune system and antibiotics. The National Institute of Health (NIH) states that biofilm formation is associated with 65% of all microbial illnesses and 80% of chronic illnesses. Additionally, non-device-related microbial biofilm infections include conditions like cystic fibrosis, otitis media, infective endocarditis, and chronic inflammatory disorders. This review aims to provide an overview of research on chronic infections caused by microbial biofilms, methods used for biofilm detection, recent approaches to combat biofilms, and future perspectives, including the development of innovative antimicrobial strategies such as antimicrobial peptides, bacteriophages, and agents that disrupt biofilms.
Collapse
Affiliation(s)
- Mai M Zafer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ahram Canadian University, Cairo, Egypt.
| | - Gamal A Mohamed
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sabrin R M Ibrahim
- Department of Chemistry, Preparatory Year Program, Batterjee Medical College, 21442, Jeddah, Saudi Arabia
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt
| | - Soumya Ghosh
- Natural and Medical Science Research Center, University of Nizwa, Nizwa, 616, Oman
| | - Charné Bornman
- Department of Engineering Sciences, Faculty of Natural and Agricultural Sciences, University of the Free State, Bloemfontein, 9301, South Africa
| | - Mahmoud A Elfaky
- Department of Natural Products and Alternative Medicine, Faculty of Pharmacy, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
- Center for Artificial Intelligence in Precision Medicine, King Abdulaziz University, 21589, Jeddah, Saudi Arabia.
| |
Collapse
|
6
|
Wang S, Zhao Y, Breslawec AP, Liang T, Deng Z, Kuperman LL, Yu Q. Strategy to combat biofilms: a focus on biofilm dispersal enzymes. NPJ Biofilms Microbiomes 2023; 9:63. [PMID: 37679355 PMCID: PMC10485009 DOI: 10.1038/s41522-023-00427-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/15/2023] [Indexed: 09/09/2023] Open
Abstract
Bacterial biofilms, which consist of three-dimensional extracellular polymeric substance (EPS), not only function as signaling networks, provide nutritional support, and facilitate surface adhesion, but also serve as a protective shield for the residing bacterial inhabitants against external stress, such as antibiotics, antimicrobials, and host immune responses. Biofilm-associated infections account for 65-80% of all human microbial infections that lead to serious mortality and morbidity. Tremendous effort has been spent to address the problem by developing biofilm-dispersing agents to discharge colonized microbial cells to a more vulnerable planktonic state. Here, we discuss the recent progress of enzymatic eradicating strategies against medical biofilms, with a focus on dispersal mechanisms. Particularly, we review three enzyme classes that have been extensively investigated, namely glycoside hydrolases, proteases, and deoxyribonucleases.
Collapse
Affiliation(s)
- Shaochi Wang
- Otorhinolaryngology Hospital, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Yanteng Zhao
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Alexandra P Breslawec
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20740, USA
| | - Tingting Liang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus, 475004, Kaifeng, Henan, China
| | - Zhifen Deng
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China
| | - Laura L Kuperman
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD, 20740, USA.
- Mirimus Inc., 760 Parkside Avenue, Brooklyn, NY, 11226, USA.
| | - Qiuning Yu
- Otorhinolaryngology Hospital, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| |
Collapse
|
7
|
Perry EK, Tan MW. Bacterial biofilms in the human body: prevalence and impacts on health and disease. Front Cell Infect Microbiol 2023; 13:1237164. [PMID: 37712058 PMCID: PMC10499362 DOI: 10.3389/fcimb.2023.1237164] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/11/2023] [Indexed: 09/16/2023] Open
Abstract
Bacterial biofilms can be found in most environments on our planet, and the human body is no exception. Consisting of microbial cells encased in a matrix of extracellular polymers, biofilms enable bacteria to sequester themselves in favorable niches, while also increasing their ability to resist numerous stresses and survive under hostile circumstances. In recent decades, biofilms have increasingly been recognized as a major contributor to the pathogenesis of chronic infections. However, biofilms also occur in or on certain tissues in healthy individuals, and their constituent species are not restricted to canonical pathogens. In this review, we discuss the evidence for where, when, and what types of biofilms occur in the human body, as well as the diverse ways in which they can impact host health under homeostatic and dysbiotic states.
Collapse
Affiliation(s)
| | - Man-Wah Tan
- Department of Infectious Diseases, Genentech, South San Francisco, CA, United States
| |
Collapse
|
8
|
Bezdekova J, Canfarotta F, Grillo F, Yesilkaya H, Vaculovicova M, Piletsky S. Molecularly imprinted nanoparticles for pathogen visualisation. NANOSCALE ADVANCES 2023; 5:2602-2609. [PMID: 37143801 PMCID: PMC10153071 DOI: 10.1039/d2na00913g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/01/2023] [Indexed: 05/06/2023]
Abstract
Saccharides displayed on the cell surface of pathogens play critical roles in many activities such as adhesion, recognition and pathogenesis, as well as in prokaryotic development. In this work, we report the synthesis of molecularly imprinted nanoparticles (nanoMIPs) against pathogen surface monosaccharides using an innovative solid-phase approach. These nanoMIPs can serve as robust and selective artificial lectins specific to one particular monosaccharide. The evaluation of their binding capabilities has been implemented against bacterial cells (E. coli and S. pneumoniae) as model pathogens. The nanoMIPs were produced against two different monosaccharides: mannose (Man), which is present mainly on the surface of Gram-negative bacteria, and N-acetylglucosamine (GlcNAc) exposed on the surface of the majority of bacteria. Herein, we assessed the potential use of nanoMIPs for pathogen cell imaging and detection via flow cytometry and confocal microscopy.
Collapse
Affiliation(s)
| | | | - Fabiana Grillo
- University of Leicester University Rd Leicester LE1 7RH UK
| | | | | | | |
Collapse
|
9
|
Lai SJ, Tu IF, Tseng TS, Tsai YH, Wu SH. The deficiency of poly-β-1,6-N-acetyl-glucosamine deacetylase trigger A. baumannii to convert to biofilm-independent colistin-tolerant cells. Sci Rep 2023; 13:2800. [PMID: 36797306 PMCID: PMC9935895 DOI: 10.1038/s41598-023-30065-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen that can be resistant to antibiotics by rapidly modulating its anti-drug mechanisms. The multidrug-resistant A. baumannii has been considered one of the most threatening pathogens to our society. Biofilm formation and persistent cells within the biofilm matrix are recognized as intractable problems, especially in hospital-acquired infections. Poly-β-1,6-N-acetyl-glucosamine (PNAG) is one of the important building blocks in A. baumannii's biofilm. Here, we discover a protein phosphoryl-regulation on PNAG deacetylase, AbPgaB1, in which residue Ser411 was phosphorylated. The phosphoryl-regulation on AbPgaB1 modulates the product turnover rate in which deacetylated PNAG is produced and reflected in biofilm production. We further uncovered the PgaB deficient A. baumannii strain shows the lowest level of biofilm production but has a high minimal inhibition concentration to antibiotic colistin and tetracycline. Based on bactericidal post-antibiotic effects and time-dependent killing assays with antibacterial drugs, we claim that the PgaB-deficient A. baumannii converts to colistin-tolerant cells. This study utilizes a biofilm-independent colistin-tolerant model of A. baumannii to further investigate its characteristics and mechanisms to better understand clinical outcomes.
Collapse
Affiliation(s)
- Shu-Jung Lai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan. .,Research Center for Cancer Biology, China Medical University, Taichung, 404333, Taiwan.
| | - I-Fan Tu
- grid.28665.3f0000 0001 2287 1366Institute of Biological Chemistry, Academia Sinica, Taipei, 11529 Taiwan
| | - Tien-Sheng Tseng
- grid.260542.70000 0004 0532 3749Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Hsuan Tsai
- grid.510951.90000 0004 7775 6738Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518132 China
| | - Shih-Hsiung Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, 11529, Taiwan. .,Department of Chemistry, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
10
|
Duque C, Chrisostomo DA, Souza ACA, de Almeida Braga GP, Dos Santos VR, Caiaffa KS, Pereira JA, de Oliveira WC, de Aguiar Ribeiro A, Parisotto TM. Understanding the Predictive Potential of the Oral Microbiome in the Development and Progression of Early Childhood Caries. Curr Pediatr Rev 2023; 19:121-138. [PMID: 35959611 DOI: 10.2174/1573396318666220811124848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/24/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Early childhood caries (ECC) is the most common chronic disease in young children and a public health problem worldwide. It is characterized by the presence of atypical and fast progressive caries lesions. The aggressive form of ECC, severe early childhood caries (S-ECC), can lead to the destruction of the whole crown of most of the deciduous teeth and cause pain and sepsis, affecting the child's quality of life. Although the multifactorial etiology of ECC is known, including social, environmental, behavioral, and genetic determinants, there is a consensus that this disease is driven by an imbalance between the oral microbiome and host, or dysbiosis, mediated by high sugar consumption and poor oral hygiene. Knowledge of the microbiome in healthy and caries status is crucial for risk monitoring, prevention, and development of therapies to revert dysbiosis and restore oral health. Molecular biology tools, including next-generation sequencing methods and proteomic approaches, have led to the discovery of new species and microbial biomarkers that could reveal potential risk profiles for the development of ECC and new targets for anti-caries therapies. This narrative review summarized some general aspects of ECC, such as definition, epidemiology, and etiology, the influence of oral microbiota in the development and progression of ECC based on the current evidence from genomics, transcriptomic, proteomic, and metabolomic studies and the effect of antimicrobial intervention on oral microbiota associated with ECC. CONCLUSION The evaluation of genetic and proteomic markers represents a promising approach to predict the risk of ECC before its clinical manifestation and plan efficient therapeutic interventions for ECC in its initial stages, avoiding irreversible dental cavitation.
Collapse
Affiliation(s)
- Cristiane Duque
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Daniela Alvim Chrisostomo
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Amanda Caselato Andolfatto Souza
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Gabriela Pacheco de Almeida Braga
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Vanessa Rodrigues Dos Santos
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Karina Sampaio Caiaffa
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Jesse Augusto Pereira
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Warlley Campos de Oliveira
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, State University of São Paulo (UNESP), Araçatuba, Brazil
| | - Apoena de Aguiar Ribeiro
- Division of Diagnostic Sciences, University of North Carolina at Chapel Hill - Adams School of Dentistry, Chapel Hill, North Carolina, United State
| | - Thaís Manzano Parisotto
- Laboratory of Clinical and Molecular Microbiology, São Francisco University, Bragança Paulista, Brazil
| |
Collapse
|
11
|
Shakya S, Danshiitsoodol N, Noda M, Inoue Y, Sugiyama M. 3-Phenyllactic acid generated in medicinal plant extracts fermented with plant-derived lactic acid bacteria inhibits the biofilm synthesis of Aggregatibacter actinomycetemcomitans. Front Microbiol 2022; 13:991144. [PMID: 36212837 PMCID: PMC9539679 DOI: 10.3389/fmicb.2022.991144] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
In the present study, the effect of PLA on a periodontic pathogen, Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans), the biofilm, and virulence-related genes was investigated. We confirmed that two lactic acid bacteria (LAB) strains isolated from plant sources, Lactiplantibacillus plantarum MSC-C2 and Pediococcus pentosaceus K40, secrete PLA into the de Man, Rogosa & Sharpe (MRS) broth when supplemented with phenyl pyruvic acid (PPA) as a precursor to PLA. Moreover, PLA was generated in the fermentation broths of two medicinal plant extracts, Paeonia lactiflora Pall (PR) and Carthamus tinctorius (CT), when used by each LAB strain and each extract supplemented with PPA. We determined that the minimum inhibitory concentration (MIC) of PLA against A. actinomycetemcomitans was 20 mM. PLA significantly decreased biofilm formation and suppressed the transcription of pgA, ltxA, and cdtB genes, which encode the poly-N-acetylglucosamine (PGA) polysaccharide of biofilm matrix and exotoxins leukotoxin and cytolethal distending toxin (CDT), respectively. The PLA produced by the MSC-C2 and K40 strains was increased several times by the addition of PPA to the MRS broth. The anti-biofilm effect of the extracts from the fermentation broth was proportional to the increasing PLA concentration, while a cumulatively higher effect than that of PLA alone suggested a combinational effect of PLA and the other metabolites, such as lactic acid (LA). Among the two medicinal plants, PLA, produced after the addition of PPA, was higher in PR extract in case of both the LAB strains. PLA production by the MSC-C2 strain in the PR extract reached 4.8 ± 0.23 mM, which was obviously higher than that in the MRS broth (3.88 ± 0.12 mM) supplemented with 1 mg/ml PPA. The activity to inhibit biofilm formation in the fermented PR extract was clearly high. PLA formed in the fermented PR extract downregulated the dispersin B encoding the dspB gene together with pgA, ltxA, and cdtB. In conclusion, this study shows a promising activity of PLA against the A. actinomycetemcomitans biofilm and virulence genes. In addition, the combinational effect of PLA and the medicinal plant extract can be achieved by fermentation with a specific plant-derived LAB strain.
Collapse
|
12
|
Costa RC, Bertolini M, Costa Oliveira BE, Nagay BE, Dini C, Benso B, Klein MI, Barāo VAR, Souza JGS. Polymicrobial biofilms related to dental implant diseases: unravelling the critical role of extracellular biofilm matrix. Crit Rev Microbiol 2022; 49:370-390. [PMID: 35584310 DOI: 10.1080/1040841x.2022.2062219] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biofilms are complex tri-dimensional structures that encase microbial cells in an extracellular matrix comprising self-produced polymeric substances. The matrix rich in extracellular polymeric substance (EPS) contributes to the unique features of biofilm lifestyle and structure, enhancing microbial accretion, biofilm virulence, and antimicrobial resistance. The role of the EPS matrix of biofilms growing on biotic surfaces, especially dental surfaces, is largely unravelled. To date, there is a lack of a broad overview of existing literature concerning the relationship between the EPS matrix and the dental implant environment and its role in implant-related infections. Here, we discuss recent advances in the critical role of the EPS matrix on biofilm growth and virulence on the dental implant surface and its effect on the etiopathogenesis and progression of implant-related infections. Similar to other biofilms associated with human diseases/conditions, EPS-enriched biofilms on implant surfaces promote microbial accumulation, microbiological shift, cross-kingdom interaction, antimicrobial resistance, biofilm virulence, and, consequently, peri-implant tissue damage. But intriguingly, the protagonism of EPS role on implant-related infections and the development of matrix-target therapeutic strategies has been neglected. Finally, we highlight the need for more in-depth analyses of polymicrobial interactions within EPS matrix and EPS-targeting technologies' rationale for disrupting the complex biofilm microenvironment with more outstanding translation to implant applications in the near future.
Collapse
Affiliation(s)
- Raphael C Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Martinna Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | | | - Bruna E Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Bruna Benso
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, CA, Chile
| | - Marlise I Klein
- Department of Dental Materials and Prosthodontics, São Paulo State University, São Paulo, Brazil
| | - Valentim A R Barāo
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Joāo Gabriel S Souza
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil.,Dental Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Brazil.,Dental Research Division, Guarulhos University, Sāo Paulo, Brazil
| |
Collapse
|
13
|
Fu J, Liu C, Li L, Liu J, Tie Y, Wen X, Zhao Q, Qiao Z, An Z, Zheng J. Adaptive response and tolerance to weak acids in
Saccharomyces cerevisiae boulardii
: a metabolomics approach. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Junjie Fu
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
| | - Chaolan Liu
- Antibiotics Research and Re‐evalution Key Laboratory of Sichuan Province Sichuan Industrial Institute of Antibiotics Chengdu University Chengdu 610052 China
| | - Li Li
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
| | - Jun Liu
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
| | - Yu Tie
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
- Solid‐State Fermentation Resource Utilisation Key Laboratory of Sichuan Province Yibin 644000 China
| | - Xueping Wen
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
| | - Qikai Zhao
- College of Biotechnology Engineering Sichuan University of Science and Engineering Yibin 644000 China
- HengfengHuaBang Biological Science and Technology Co., Ltd. Leshan 614000 China
| | | | - Zheming An
- Wuliangye Yibin Co, Ltd Yibin 644000 China
| | - Jia Zheng
- Wuliangye Yibin Co, Ltd Yibin 644000 China
| |
Collapse
|
14
|
Manoharan A, Das T, Whiteley GS, Glasbey T, Kriel FH, Manos J. The effect of N-acetylcysteine in a combined antibiofilm treatment against antibiotic-resistant Staphylococcus aureus. J Antimicrob Chemother 2021; 75:1787-1798. [PMID: 32363384 DOI: 10.1093/jac/dkaa093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/31/2020] [Accepted: 02/17/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The WHO declared Staphylococcus aureus as a 'pathogen of high importance' in 2017. One-fifth of all bloodstream-related infections in Australia and 12 000 cases of bacteraemia in the UK (2017-18) were caused by the MRSA variant. To address the need for novel therapies, we investigated several permutations of an innovative combination therapy containing N-acetylcysteine (NAC), an antibiotic and an enzyme of choice in eradicating MRSA and MSSA biofilms. METHODS Biofilm viability (resazurin assay) and colony count methods were used to investigate the effect of NAC, antibiotics and enzymes on S. aureus biofilm disruption and killing. The effects of NAC and enzymes on the polysaccharide content of biofilm matrices were analysed using the phenol/sulphuric acid method and the effect of NAC on DNA cleavage was determined using the Qubit fluorometer technique. Changes in biofilm architecture when subjected to NAC and enzymes were visualized using confocal laser scanning microscopy (CLSM). RESULTS NAC alone displayed bacteriostatic effects when tested on planktonic bacterial growth. Combination treatments containing 30 mM NAC resulted in ≥90% disruption of biofilms across all MRSA and MSSA strains with a 2-3 log10 decrease in cfu/mL in treated biofilms. CLSM showed that NAC treatment drastically disrupted S. aureus biofilm architecture. There was also reduced polysaccharide production in MRSA biofilms in the presence of NAC. CONCLUSIONS Our results indicate that inclusion of NAC in a combination treatment is a promising strategy for S. aureus biofilm eradication. The intrinsic acidity of NAC was identified as key to maximum biofilm disruption and degradation of matrix components.
Collapse
Affiliation(s)
- Arthika Manoharan
- Department of Infectious Diseases and Immunology, Central Clinical School, The University of Sydney, Sydney, Australia
| | - Theerthankar Das
- Department of Infectious Diseases and Immunology, Central Clinical School, The University of Sydney, Sydney, Australia
| | | | - Trevor Glasbey
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| | - Frederik H Kriel
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| | - Jim Manos
- Whiteley Corporation, 19-23 Laverick Avenue, Tomago NSW 2319, Australia
| |
Collapse
|
15
|
Lahiri D, Nag M, Banerjee R, Mukherjee D, Garai S, Sarkar T, Dey A, Sheikh HI, Pathak SK, Edinur HA, Pati S, Ray RR. Amylases: Biofilm Inducer or Biofilm Inhibitor? Front Cell Infect Microbiol 2021; 11:660048. [PMID: 33987107 PMCID: PMC8112260 DOI: 10.3389/fcimb.2021.660048] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Biofilm is a syntrophic association of sessile groups of microbial cells that adhere to biotic and abiotic surfaces with the help of pili and extracellular polymeric substances (EPS). EPSs also prevent penetration of antimicrobials/antibiotics into the sessile groups of cells. Hence, methods and agents to avoid or remove biofilms are urgently needed. Enzymes play important roles in the removal of biofilm in natural environments and may be promising agents for this purpose. As the major component of the EPS is polysaccharide, amylase has inhibited EPS by preventing the adherence of the microbial cells, thus making amylase a suitable antimicrobial agent. On the other hand, salivary amylase binds to amylase-binding protein of plaque-forming Streptococci and initiates the formation of biofilm. This review investigates the contradictory actions and microbe-associated genes of amylases, with emphasis on their structural and functional characteristics.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Ritwik Banerjee
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Dipro Mukherjee
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Sayantani Garai
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Tanmay Sarkar
- Department of Food Technology and Bio-Chemical Engineering, Jadavpur University, Kolkata, India.,Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda, India
| | - Ankita Dey
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| | - Hassan I Sheikh
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus, Malaysia
| | - Sushil Kumar Pathak
- Department of Bioscience and Bioinformatics, Khallikote University, Berhampur, India
| | | | - Siddhartha Pati
- Centre of Excellence, Khallikote University, Berhampur, India.,Research Division, Association for Biodiversity Conservation and Research (ABC), Balasore, India
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Haringhata, India
| |
Collapse
|
16
|
Jakubovics NS, Goodman SD, Mashburn-Warren L, Stafford GP, Cieplik F. The dental plaque biofilm matrix. Periodontol 2000 2021; 86:32-56. [PMID: 33690911 PMCID: PMC9413593 DOI: 10.1111/prd.12361] [Citation(s) in RCA: 220] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
| | - Steven D Goodman
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Lauren Mashburn-Warren
- Center for Microbial Pathogenesis, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Graham P Stafford
- Integrated Biosciences, School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Fabian Cieplik
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
17
|
Dawson LF, Peltier J, Hall CL, Harrison MA, Derakhshan M, Shaw HA, Fairweather NF, Wren BW. Extracellular DNA, cell surface proteins and c-di-GMP promote biofilm formation in Clostridioides difficile. Sci Rep 2021; 11:3244. [PMID: 33547340 PMCID: PMC7865049 DOI: 10.1038/s41598-020-78437-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Clostridioides difficile is the leading cause of nosocomial antibiotic-associated diarrhoea worldwide, yet there is little insight into intestinal tract colonisation and relapse. In many bacterial species, the secondary messenger cyclic-di-GMP mediates switching between planktonic phase, sessile growth and biofilm formation. We demonstrate that c-di-GMP promotes early biofilm formation in C. difficile and that four cell surface proteins contribute to biofilm formation, including two c-di-GMP regulated; CD2831 and CD3246, and two c-di-GMP-independent; CD3392 and CD0183. We demonstrate that C. difficile biofilms are composed of extracellular DNA (eDNA), cell surface and intracellular proteins, which form a protective matrix around C. difficile vegetative cells and spores, as shown by a protective effect against the antibiotic vancomycin. We demonstrate a positive correlation between biofilm biomass, sporulation frequency and eDNA abundance in all five C. difficile lineages. Strains 630 (RT012), CD305 (RT023) and M120 (RT078) contain significantly more eDNA in their biofilm matrix than strains R20291 (RT027) and M68 (RT017). DNase has a profound effect on biofilm integrity, resulting in complete disassembly of the biofilm matrix, inhibition of biofilm formation and reduced spore germination. The addition of exogenous DNase could be exploited in treatment of C. difficile infection and relapse, to improve antibiotic efficacy.
Collapse
Affiliation(s)
- Lisa F Dawson
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK.
| | - Johann Peltier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Catherine L Hall
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Mark A Harrison
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Maria Derakhshan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Helen A Shaw
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
- National Institute for Biological Standards and Control, Potters Bar, UK
| | - Neil F Fairweather
- Department of Life Sciences, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Brendan W Wren
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
18
|
Biofilms as Promoters of Bacterial Antibiotic Resistance and Tolerance. Antibiotics (Basel) 2020; 10:antibiotics10010003. [PMID: 33374551 PMCID: PMC7822488 DOI: 10.3390/antibiotics10010003] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 12/19/2020] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistant bacteria are a global threat for human and animal health. However, they are only part of the problem of antibiotic failure. Another bacterial strategy that contributes to their capacity to withstand antimicrobials is the formation of biofilms. Biofilms are associations of microorganisms embedded a self-produced extracellular matrix. They create particular environments that confer bacterial tolerance and resistance to antibiotics by different mechanisms that depend upon factors such as biofilm composition, architecture, the stage of biofilm development, and growth conditions. The biofilm structure hinders the penetration of antibiotics and may prevent the accumulation of bactericidal concentrations throughout the entire biofilm. In addition, gradients of dispersion of nutrients and oxygen within the biofilm generate different metabolic states of individual cells and favor the development of antibiotic tolerance and bacterial persistence. Furthermore, antimicrobial resistance may develop within biofilms through a variety of mechanisms. The expression of efflux pumps may be induced in various parts of the biofilm and the mutation frequency is induced, while the presence of extracellular DNA and the close contact between cells favor horizontal gene transfer. A deep understanding of the mechanisms by which biofilms cause tolerance/resistance to antibiotics helps to develop novel strategies to fight these infections.
Collapse
|
19
|
Cetylpyridinium Chloride: Mechanism of Action, Antimicrobial Efficacy in Biofilms, and Potential Risks of Resistance. Antimicrob Agents Chemother 2020; 64:AAC.00576-20. [PMID: 32513792 DOI: 10.1128/aac.00576-20] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Antimicrobial resistance is a serious issue for public health care all over the world. While resistance toward antibiotics has attracted strong interest among researchers and the general public over the last 2 decades, the directly related problem of resistance toward antiseptics and biocides has been somewhat left untended. In the field of dentistry, antiseptics are routinely used in professional care, but they are also included in lots of oral care products such as mouthwashes or dentifrices, which are easily available for consumers over-the-counter. Despite this fact, there is little awareness among the dental community about potential risks of the widespread, unreflected, and potentially even needless use of antiseptics in oral care. Cetylpyridinium chloride (CPC), a quaternary ammonium compound, which was first described in 1939, is one of the most commonly used antiseptics in oral care products and included in a wide range of over-the-counter products such as mouthwashes and dentifrices. The aim of the present review is to summarize the current literature on CPC, particularly focusing on its mechanism of action, its antimicrobial efficacy toward biofilms, and on potential risks of resistance toward this antiseptic as well as underlying mechanisms. Furthermore, this work aims to raise awareness among the dental community about the risk of resistance toward antiseptics in general.
Collapse
|
20
|
Morrison ZA, Nitz M. Synthesis of C6-substituted UDP-GlcNAc derivatives. Carbohydr Res 2020; 495:108071. [PMID: 32634644 DOI: 10.1016/j.carres.2020.108071] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 02/05/2023]
Abstract
UDP-sugar analogs are useful for the study of glycosyltransferases and the production of unnatural glycans. The preparation of five UDP-GlcNAc derivatives is reported with 6-deoxy, 6-azido, 6-amino, 6-mercapto, or 6-fluoro substitutions. A concise chemoenzymatic synthesis was developed using the kinase NahK (B. longum JCM1217) and the uridyl transferase GlmU (E. coli K12).
Collapse
Affiliation(s)
- Zachary A Morrison
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, Ontario, M5S 3H6, Canada.
| |
Collapse
|
21
|
Szymańska M, Karakulska J, Sobolewski P, Kowalska U, Grygorcewicz B, Böttcher D, Bornscheuer UT, Drozd R. Glycoside hydrolase (PelA h) immobilization prevents Pseudomonas aeruginosa biofilm formation on cellulose-based wound dressing. Carbohydr Polym 2020; 246:116625. [PMID: 32747262 DOI: 10.1016/j.carbpol.2020.116625] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/30/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022]
Abstract
Bacterial cellulose (BC) is recognized as a wound dressing material well-suited for chronic wounds; however, it has no intrinsic antimicrobial activity. Further, the formation of biofilms can limit the effectiveness of the pre-saturation of BC with antimicrobial agents. Here, to hinder biofilm formation by P. aeruginosa, we immobilized the hydrolytic domain of PelA (a glycohydrolase involved in the synthesis of biofilm polysaccharide Pel) on the surface of BC. The immobilization of 32.35 ± 1.05 mg PelAh per g BC membrane resulted in an eight-fold higher P. aeruginosa cell detachment from BC membrane, indicating reduced biofilm matrix stability. Further, 1D and 2D infrared spectroscopy analysis indicated systematic reduction of polysaccharide biofilm elements, confirming the specificity of immobilized PelAh. Importantly, BC-PelAh was not cytotoxic towards L929 fibroblast cells. Thus, we conclude that PelAh can be used in BC wound dressings for safe and specific protection against biofilm formation by P. aeruginosa.
Collapse
Affiliation(s)
- Magdalena Szymańska
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, 45 Piastow Avenue, 71-311, Szczecin, Poland
| | - Jolanta Karakulska
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, 45 Piastow Avenue, 71-311, Szczecin, Poland
| | - Peter Sobolewski
- Department of Polymer and Biomaterials Science, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, 45 Piastów Avenue, 71-311, Szczecin, Poland
| | - Urszula Kowalska
- Center of Bioimmobilisation and Innovative Packaging Materials, Faculty of Food Science and Fisheries, West Pomeranian University of Technology in Szczecin, 35 Klemensa Janickiego Str., 71-270, Szczecin, Poland
| | - Bartłomiej Grygorcewicz
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, 72 Powstańców Wielkopolskich Str., 70-111, Szczecin, Poland
| | - Dominique Böttcher
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487, Greifswald, Germany
| | - Uwe T Bornscheuer
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, D-17487, Greifswald, Germany
| | - Radosław Drozd
- Department of Microbiology and Biotechnology, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology in Szczecin, 45 Piastow Avenue, 71-311, Szczecin, Poland.
| |
Collapse
|
22
|
Sadiq FA, Yan B, Zhao J, Zhang H, Chen W. Untargeted metabolomics reveals metabolic state of Bifidobacterium bifidum in the biofilm and planktonic states. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2019.108772] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Forman A, Pfoh R, Eddenden A, Howell PL, Nitz M. Synthesis of defined mono-de-N-acetylated β-(1→6)-N-acetyl-d-glucosamine oligosaccharides to characterize PgaB hydrolase activity. Org Biomol Chem 2019; 17:9456-9466. [PMID: 31642455 DOI: 10.1039/c9ob02079a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Many clinically-relevant biofilm-forming bacterial strains produce partially de-N-acetylated poly-β-(1→6)-N-acetyl-d-glucosamine (dPNAG) as an exopolysaccharide. In Gram-negative bacteria, the periplasmic protein PgaB is responsible for partial de-N-acetylation of PNAG prior to its export to the extracellular space. In addition to de-N-acetylase activity found in the N-terminal domain, PgaB contains a C-terminal hydrolase domain that can disrupt dPNAG-dependent biofilms and hydrolyzes dPNAG but not fully acetylated PNAG. The role of this C-terminal domain in biofilm formation has yet to be determined in vivo. Further characterization of the enzyme's hydrolase activity has been hampered by a lack of specific dPNAG oligosaccharides. Here, we report the synthesis of a defined mono de-N-acetylated dPNAG penta- and hepta-saccharide. Using mass spectrometry analysis and a fluorescence-based thin-layer chromatography (TLC) assay, we found that our defined dPNAG oligosaccharides are hydrolase substrates. In addition to the expected cleavage site, two residues to the reducing side of the de-N-acetylated residue, minor cleavage products on the non-reducing side of the de-N-acetylation site were observed. These findings provide quantitative data to support how PNAG is processed in Gram-negative bacteria.
Collapse
Affiliation(s)
- Adam Forman
- Department of Chemistry, University of Toronto, 80 St. George Street, Toronto, ON, Canada M5S 3H6.
| | | | | | | | | |
Collapse
|
24
|
Lysocins: Bioengineered Antimicrobials That Deliver Lysins across the Outer Membrane of Gram-Negative Bacteria. Antimicrob Agents Chemother 2019; 63:AAC.00342-19. [PMID: 30962344 DOI: 10.1128/aac.00342-19] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 04/03/2019] [Indexed: 11/20/2022] Open
Abstract
The prevalence of multidrug-resistant Pseudomonas aeruginosa has stimulated development of alternative therapeutics. Bacteriophage peptidoglycan hydrolases, termed lysins, represent an emerging antimicrobial option for targeting Gram-positive bacteria. However, lysins against Gram-negatives are generally deterred by the outer membrane and their inability to work in serum. One solution involves exploiting evolved delivery systems used by colicin-like bacteriocins (e.g., S-type pyocins of P. aeruginosa) to translocate through the outer membrane. Following surface receptor binding, colicin-like bacteriocins form Tol- or TonB-dependent translocons to actively import bactericidal domains through outer membrane protein channels. With this understanding, we developed lysocins, which are bioengineered lysin-bacteriocin fusion molecules capable of periplasmic import. In our proof-of-concept studies, components from the P. aeruginosa bacteriocin pyocin S2 (PyS2) responsible for surface receptor binding and outer membrane translocation were fused to the GN4 lysin to generate the PyS2-GN4 lysocin. PyS2-GN4 delivered the GN4 lysin to the periplasm to induce peptidoglycan cleavage and log-fold killing of P. aeruginosa with minimal endotoxin release. While displaying narrow-spectrum antipseudomonal activity in human serum, PyS2-GN4 also efficiently disrupted biofilms, outperformed standard-of-care antibiotics, exhibited no cytotoxicity toward eukaryotic cells, and protected mice from P. aeruginosa challenge in a bacteremia model. In addition to targeting P. aeruginosa, lysocins can be constructed to target other prominent Gram-negative bacterial pathogens.
Collapse
|
25
|
Cugini C, Shanmugam M, Landge N, Ramasubbu N. The Role of Exopolysaccharides in Oral Biofilms. J Dent Res 2019; 98:739-745. [PMID: 31009580 DOI: 10.1177/0022034519845001] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The oral cavity contains a rich consortium of exopolysaccharide-producing microbes. These extracellular polysaccharides comprise a major component of the oral biofilm. Together with extracellular proteins, DNA, and lipids, they form the biofilm matrix, which contributes to bacterial colonization, biofilm formation and maintenance, and pathogenesis. While a number of oral microbes have been studied in detail with regard to biofilm formation and pathogenesis, the exopolysaccharides have been well characterized for only select organisms, namely Streptococcus mutans and Aggregatibacter actinomycetemcomitans. Studies on the exopolysaccharides of other oral organisms, however, are in their infancy. In this review, we present the current research on exopolysaccharides of oral microbes regarding their biosynthesis, regulation, contributions to biofilm formation and stability of the matrix, and immune evasion. In addition, insight into the role of exopolysaccharides in biofilms is highlighted through the evaluation of emerging techniques such as pH probing of biofilm colonies, solid-state nuclear magnetic resonance for macromolecular interactions within biofilms, and super-resolution microscopy analysis of biofilm development. Finally, exopolysaccharide as a potential nutrient source for species within a biofilm is discussed.
Collapse
Affiliation(s)
- C Cugini
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - M Shanmugam
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - N Landge
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - N Ramasubbu
- 1 Department of Oral Biology, Center for Oral Biology Research, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
26
|
Ahlstrand T, Kovesjoki L, Maula T, Oscarsson J, Ihalin R. Aggregatibacter actinomycetemcomitans LPS binds human interleukin-8. J Oral Microbiol 2018; 11:1549931. [PMID: 34917288 PMCID: PMC8670607 DOI: 10.1080/20002297.2018.1549931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Various gram-negative species sequester host cytokines using outer membrane proteins or surface modulation by sulfated polysaccharides. An outer membrane lipoprotein (BilRI) of the periodontal pathogen Aggregatibacter actinomycetemcomitans binds several cytokines, including interleukin (IL)-8. Because IL-8 is positively charged at physiological pH, we aimed to determine whether IL-8 interacts with negatively charged lipopolysaccharide (LPS). Binding was investigated using electrophoretic mobility shift assays and microwell-based time-resolved fluorometric immunoassay. LPS from each tested strain of A. actinomycetemcomitans (N = 13), Pseudomonas aeruginosa (N = 1) and Escherichia coli (N = 1) bound IL-8. The Kd value of the A. actinomycetemcomitans LPS-IL-8 interaction varied between 1.2–17 μM irrespective of the serotype and the amount of phosphorus in LPS and was significantly lower than that of the BilRI-IL-8 interaction. Moreover, IL-8 interacted with whole A. actinomycetemcomitans cells and outer membrane vesicles. Hence, LPS might be involved in binding of IL-8 to the outer membrane of A. actinomycetemcomitans. This raises an interesting question regarding whether other gram-negative periodontal pathogens use LPS for IL-8 sequestering in vivo.
Collapse
Affiliation(s)
- Tuuli Ahlstrand
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Laura Kovesjoki
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Terhi Maula
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| | - Riikka Ihalin
- Department of Biochemistry, University of Turku, Turku, Finland
| |
Collapse
|
27
|
Ahlstrand T, Torittu A, Elovaara H, Välimaa H, Pöllänen MT, Kasvandik S, Högbom M, Ihalin R. Interactions between the Aggregatibacter actinomycetemcomitans secretin HofQ and host cytokines indicate a link between natural competence and interleukin-8 uptake. Virulence 2018; 9:1205-1223. [PMID: 30088437 PMCID: PMC6086316 DOI: 10.1080/21505594.2018.1499378] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Naturally competent bacteria acquire DNA from their surroundings to survive in nutrient-poor environments and incorporate DNA into their genomes as new genes for improved survival. The secretin HofQ from the oral pathogen Aggregatibacter actinomycetemcomitans has been associated with DNA uptake. Cytokine sequestering is a potential virulence mechanism in various bacteria and may modulate both host defense and bacterial physiology. The objective of this study was to elucidate a possible connection between natural competence and cytokine uptake in A. actinomycetemcomitans. The extramembranous domain of HofQ (emHofQ) was shown to interact with various cytokines, of which IL-8 exhibited the strongest interaction. The dissociation constant between emHofQ and IL-8 was 43 nM in static settings and 2.4 μM in dynamic settings. The moderate binding affinity is consistent with the hypothesis that emHofQ recognizes cytokines before transporting them into the cells. The interaction site was identified via crosslinking and mutational analysis. By structural comparison, relateda type I KH domain with a similar interaction site was detected in the Neisseria meningitidis secretin PilQ, which has been shown to participate in IL-8 uptake. Deletion of hofQ from the A. actinomycetemcomitans genome decreased the overall biofilm formation of this organism, abolished the response to cytokines, i.e., decreased eDNA levels in the presence of cytokines, and increased the susceptibility of the biofilm to tested β-lactams. Moreover, we showed that recombinant IL-8 interacted with DNA. These results can be used in further studies on the specific role of cytokine uptake in bacterial virulence without interfering with natural-competence-related DNA uptake.
Collapse
Affiliation(s)
- Tuuli Ahlstrand
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Annamari Torittu
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Heli Elovaara
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Hannamari Välimaa
- b Department of Virology , University of Helsinki , Helsinki , Finland.,c Department of Oral and Maxillofacial Surgery , Helsinki University Hospital , Helsinki , Finland
| | - Marja T Pöllänen
- d Institute of Dentistry , University of Turku , Turku , Finland
| | - Sergo Kasvandik
- e Institute of Technology , University of Tartu , Tartu , Estonia
| | - Martin Högbom
- f Department of Biochemistry and Biophysics , Stockholm University , Stockholm , Sweden
| | - Riikka Ihalin
- a Department of Biochemistry , University of Turku , Turku , Finland
| |
Collapse
|
28
|
Maigaard Hermansen GM, Boysen A, Krogh TJ, Nawrocki A, Jelsbak L, Møller-Jensen J. HldE Is Important for Virulence Phenotypes in Enterotoxigenic Escherichia coli. Front Cell Infect Microbiol 2018; 8:253. [PMID: 30131942 PMCID: PMC6090259 DOI: 10.3389/fcimb.2018.00253] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the most common causes of diarrheal illness in third world countries and it especially affects children and travelers visiting these regions. ETEC causes disease by adhering tightly to the epithelial cells in a concerted effort by adhesins, flagella, and other virulence-factors. When attached ETEC secretes toxins targeting the small intestine host-cells, which ultimately leads to osmotic diarrhea. HldE is a bifunctional protein that catalyzes the nucleotide-activated heptose precursors used in the biosynthesis of lipopolysaccharide (LPS) and in post-translational protein glycosylation. Both mechanisms have been linked to ETEC virulence: Lipopolysaccharide (LPS) is a major component of the bacterial outer membrane and is needed for transport of heat-labile toxins to the host cells, and ETEC glycoproteins have been shown to play an important role for bacterial adhesion to host epithelia. Here, we report that HldE plays an important role for ETEC virulence. Deletion of hldE resulted in markedly reduced binding to the human intestinal cells due to reduced expression of colonization factor CFA/I on the bacterial surface. Deletion of hldE also affected ETEC motility in a flagella-dependent fashion. Expression of both colonization factors and flagella was inhibited at the level of transcription. In addition, the hldE mutant displayed altered growth, increased biofilm formation and clumping in minimal growth medium. Investigation of an orthogonal LPS-deficient mutant combined with mass spectrometric analysis of protein glycosylation indicated that HldE exerts its role on ETEC virulence both through protein glycosylation and correct LPS configuration. These results place HldE as an attractive target for the development of future antimicrobial therapeutics.
Collapse
Affiliation(s)
| | - Anders Boysen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Thøger J Krogh
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Arkadiusz Nawrocki
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Lars Jelsbak
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jakob Møller-Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
29
|
Sismaet HJ, Goluch ED. Electrochemical Probes of Microbial Community Behavior. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2018; 11:441-461. [PMID: 29490192 DOI: 10.1146/annurev-anchem-061417-125627] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Advances in next-generation sequencing technology along with decreasing costs now allow the microbial population, or microbiome, of a location to be determined relatively quickly. This research reveals that microbial communities are more diverse and complex than ever imagined. New and specialized instrumentation is required to investigate, with high spatial and temporal resolution, the dynamic biochemical environment that is created by microbes, which allows them to exist in every corner of the Earth. This review describes how electrochemical probes and techniques are being used and optimized to learn about microbial communities. Described approaches include voltammetry, electrochemical impedance spectroscopy, scanning electrochemical microscopy, separation techniques coupled with electrochemical detection, and arrays of complementary metal-oxide-semiconductor circuits. Microbial communities also interact with and influence their surroundings; therefore, the review also includes a discussion of how electrochemical probes optimized for microbial analysis are utilized in healthcare diagnostics and environmental monitoring applications.
Collapse
Affiliation(s)
- Hunter J Sismaet
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA;
| | - Edgar D Goluch
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts 02115, USA;
- Department of Bioengineering, Department of Biology, and Department of Civil and Environmental Engineering, Northeastern University, Boston, Massachusetts 02115, USA
| |
Collapse
|
30
|
Bao K, Bostanci N, Thurnheer T, Grossmann J, Wolski WE, Thay B, Belibasakis GN, Oscarsson J. Aggregatibacter actinomycetemcomitans H-NS promotes biofilm formation and alters protein dynamics of other species within a polymicrobial oral biofilm. NPJ Biofilms Microbiomes 2018; 4:12. [PMID: 29844920 PMCID: PMC5964231 DOI: 10.1038/s41522-018-0055-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/03/2018] [Accepted: 04/19/2018] [Indexed: 12/12/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans is a Gram-negative organism, strongly associated with aggressive forms of periodontitis. An important virulence property of A. actinomycetemcomitans is its ability to form tenacious biofilms that can attach to abiotic as well as biotic surfaces. The histone-like (H-NS) family of nucleoid-structuring proteins act as transcriptional silencers in many Gram-negative bacteria. To evaluate the role of H-NS in A. actinomycetemcomitans, hns mutant derivatives of serotype a strain D7S were generated. Characteristics of the hns mutant phenotype included shorter and fewer pili, and substantially lower monospecies biofilm formation relative to the wild type. Furthermore, the D7S hns mutant exhibited significantly reduced growth within a seven-species oral biofilm model. However, no apparent difference was observed regarding the numbers and proportions of the remaining six species regardless of being co-cultivated with D7S hns or its parental strain. Proteomics analysis of the strains grown in monocultures confirmed the role of H-NS as a repressor of gene expression in A. actinomycetemcomitans. Interestingly, proteomics analysis of the multispecies biofilms indicated that the A. actinomycetemcomitans wild type and hns mutant imposed different regulatory effects on the pattern of protein expression in the other species, i.e., mainly Streptococcus spp., Fusobacterium nucleatum, and Veillonella dispar. Gene ontology analysis revealed that a large portion of the differentially regulated proteins was related to translational activity. Taken together, our data suggest that, apart from being a negative regulator of protein expression in A. actinomycetemcomitans, H-NS promotes biofilm formation and may be an important factor for survival of this species within a multispecies biofilm. A member of a specific group of gene-regulating proteins promotes biofilm formation by a bacterium associated with aggressive forms of gum disease. Forming biofilms helps the bacterium to cause persistent infections. Researchers at Karolinska Institutet and Umeå University (Sweden), and University of Zürich (Switzerland), led by Jan Oscarsson at Umeå University, investigated the role of the “histone-like” protein H-NS in Aggregatibacter actinomycetemcomitans infections. These proteins are known to suppress the activity of specific genes in many bacteria, a property confirmed in this research. By studying mutant bacterial strains deficient in H-NS protein, the researchers demonstrated that this protein promotes the formation of biofilms by the bacteria. The results suggest that H-NS plays a significant role in allowing Aggregatibacter actinomycetemcomitans to thrive in biofilms containing mixed populations of bacteria. This effect appears to involve activating production of hair-like appendages called pili on the bacterial surface.
Collapse
Affiliation(s)
- Kai Bao
- 1Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Solnavägen, Sweden.,2Division of Oral Microbiology and Immunology, Center of Dental Medicine, University of Zürich, Zürich, Switzerland
| | - Nagihan Bostanci
- 1Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Solnavägen, Sweden
| | - Thomas Thurnheer
- 2Division of Oral Microbiology and Immunology, Center of Dental Medicine, University of Zürich, Zürich, Switzerland
| | - Jonas Grossmann
- 3Functional Genomics Center, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Witold E Wolski
- 3Functional Genomics Center, ETH Zürich and University of Zürich, Zürich, Switzerland
| | - Bernard Thay
- 4Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| | - Georgios N Belibasakis
- 1Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Solnavägen, Sweden
| | - Jan Oscarsson
- 4Oral Microbiology, Department of Odontology, Umeå University, Umeå, Sweden
| |
Collapse
|
31
|
Stumpp NS, Konze SA, Gerardy-Schahn R, Stiesch M, Buettner FFR. A single-step transconjugation system for gene deletion in Aggregatibacter actinomycetemcomitans. J Microbiol Methods 2018; 148:74-77. [PMID: 29627600 DOI: 10.1016/j.mimet.2018.04.002] [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: 02/05/2018] [Revised: 04/03/2018] [Accepted: 04/03/2018] [Indexed: 11/25/2022]
Abstract
Aggregatibacter (A.) actinomycetemcomitans is a periodontopathogenic bacterium causing aggressive periodontitis. Here we describe a single-step transconjugation system as novel and easily applicable protocol for site-specific genetic manipulation of A. actinomycetemcomitans. Deletion of PgaC, which is involved in the synthesis of biofilm matrix, led to a reduced biofilm formation.
Collapse
Affiliation(s)
- Nico S Stumpp
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Germany
| | - Sarah A Konze
- Institute of Clinical Biochemistry, Hannover Medical School, Germany
| | | | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Germany.
| | - Falk F R Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Germany.
| |
Collapse
|
32
|
Schneider B, Weigel W, Sztukowska M, Demuth DR. Identification and functional characterization of type II toxin/antitoxin systems in Aggregatibacter actinomycetemcomitans. Mol Oral Microbiol 2018; 33:224-233. [PMID: 29319934 PMCID: PMC5969271 DOI: 10.1111/omi.12215] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 11/30/2022]
Abstract
Type II toxin/antitoxin (TA) systems contribute to the formation of persister cells and biofilm formation for many organisms. Aggregatibacter actinomycetemcomitans thrives in the complex oral microbial community subjected to continual environmental flux. Little is known regarding the presence and function of type II TA systems in this organism or their contribution to adaptation and persistence in the biofilm. We identified 11 TA systems that are conserved across all seven serotypes of A. actinomycetemcomitans and represent the RelBE, MazEF and HipAB families of type II TA systems. The systems selectively responded to various environmental conditions that exist in the oral cavity. Two putative RelBE‐like TA systems, D11S_1194‐1195 and D11S_1718‐1719 were induced in response to low pH and deletion of D11S_1718‐1719 significantly reduced metabolic activity of stationary phase A. actinomycetemcomitans cells upon prolonged exposure to acidic conditions. The deletion mutant also exhibited reduced biofilm biomass when cultured under acidic conditions. The D11S_1194 and D11S_1718 toxin proteins inhibited in vitro translation of dihydrofolate reductase (DHFR) and degraded ribosome‐associated, but not free, MS2 virus RNA. In contrast, the corresponding antitoxins (D11S_1195 and D11S_1719), or equimolar mixtures of toxin and antitoxin, had no effect on DHFR production or RNA degradation. Together, these results suggest that D11S_1194‐1195 and D11S_1718‐1719 are RelBE‐like type II TA systems that are activated under acidic conditions and may function to cleave ribosome‐associated mRNA to inhibit translation in A. actinomycetemcomitans. In vivo, these systems may facilitate A. actinomycetemcomitans adaptation and persistence in acidic local environments in the dental biofilm.
Collapse
Affiliation(s)
- B Schneider
- Department of Oral Immunology and Infectious Disease, University of Louisville School of Dentistry, Louisville, KY, USA
| | - W Weigel
- Department of Oral Immunology and Infectious Disease, University of Louisville School of Dentistry, Louisville, KY, USA
| | - M Sztukowska
- Department of Oral Immunology and Infectious Disease, University of Louisville School of Dentistry, Louisville, KY, USA
| | - D R Demuth
- Department of Oral Immunology and Infectious Disease, University of Louisville School of Dentistry, Louisville, KY, USA
| |
Collapse
|
33
|
Abstract
Surface-attached colonies of bacteria known as biofilms play a major role in the pathogenesis of medical device infections. Biofilm colonies are notorious for their resistance to antibiotics and host defenses, which makes most device infections difficult or impossible to eradicate. Bacterial cells in a biofilm are held together by an extracellular polymeric matrix that is synthesized by the bacteria themselves. Enzymes that degrade biofilm matrix polymers have been shown to inhibit bio film formation, detach established bio film colonies, and render biofilm cells sensitive to killing by antimicrobial agents. This review discusses the potential use of biofilm matrix-degrading enzymes as anti-biofilm agents for the treatment and prevention of device infections. Two enzymes, deoxyribonuclease I and the glycoside hydrolase dispersin B, will be reviewed in detail. In vitro and in vivo studies demonstrating the anti-biofilm activities of these two enzymes will be summarized, and the therapeutic potential and possible drawbacks of using these enzymes as clinical agents will be discussed.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Department of Oral Biology, New Jersey Dental School, Newark, NJ - USA
| |
Collapse
|
34
|
Parthiban C, Varudharasu D, Shanmugam M, Gopal P, Ragunath C, Thomas L, Nitz M, Ramasubbu N. Structural and functional analysis of de-N-acetylase PgaB from periodontopathogen Aggregatibacter actinomycetemcomitans. Mol Oral Microbiol 2017; 32:324-340. [PMID: 27706922 PMCID: PMC11471279 DOI: 10.1111/omi.12175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2016] [Indexed: 10/15/2024]
Abstract
The oral pathogen Aggregatibacter actinomycetemcomitans uses pga gene locus for the production of an exopolysaccharide made up of a linear homopolymer of β-1,6-N-acetyl-d-glucosamine (PGA). An enzyme encoded by the pgaB of the pga operon in A. actinomycetemcomitans is a de-N-acetylase, which is used to alter the PGA. The full length enzyme (AaPgaB) and the N-terminal catalytic domain (residues 25-290, AaPgaBN) from A. actinomycetemcomitans were cloned, expressed and purified. The enzymatic activities of the AaPgaB enzymes were determined using 7-acetoxycoumarin-3-carboxylic acid as the substrate. The AaPgaB enzymes displayed significantly lower de-N-acetylase activity compared with the activity of the deacetylase PdaA from Bacillus subtilis, a member of the CE4 family of enzymes. To delineate the differences in the activity and the active site architecture, the structure of AaPgaBN was determined. The AaPgaBN structure has two metal ions in the active site instead of one found in other CE4 enzymes. Based on the crystal structure comparisons among the various CE4 enzymes, two residues, Q51 and R271, were identified in AaPgaB, which could potentially affect the enzyme activity. Of the two mutants generated, Q51E and R271K, the variant Q51E showed enhanced activity compared with AaPgaB, validating the requirement that an activating aspartate residue in the active site is essential for higher activity. In summary, our study provides the first structural evidence for a di-nuclear metal site at the active site of a member of the CE4 family of enzymes, evidence that AaPgaBN is catalytically active and that mutant Q51E exhibits higher de-N-acetylase activity.
Collapse
Affiliation(s)
- C Parthiban
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - D Varudharasu
- Selvam Structure Based Drug Design Laboratory, Selvam College of Technology, Namakkal, Tamilnadu, India
| | - M Shanmugam
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - P Gopal
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - C Ragunath
- Scientific Chemical Technologies, Malden, MA, USA
| | - L Thomas
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
| | - M Nitz
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| | - N Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
35
|
Schlafer S, Meyer RL, Dige I, Regina VR. Extracellular DNA Contributes to Dental Biofilm Stability. Caries Res 2017; 51:436-442. [PMID: 28728145 DOI: 10.1159/000477447] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 05/08/2017] [Indexed: 02/05/2023] Open
Abstract
Extracellular DNA (eDNA) is a major matrix component of many bacterial biofilms. While the presence of eDNA and its role in biofilm stability have been demonstrated for several laboratory biofilms of oral bacteria, there is no data available on the presence and function of eDNA in in vivo grown dental biofilms. This study aimed to determine whether eDNA was part of the matrix in biofilms grown in situ in the absence of sucrose and whether treatment with DNase dispersed biofilms grown for 2.5, 5, 7.5, 16.5, or 24 h. Three hundred biofilms from 10 study participants were collected and treated with either DNase or heat-inactivated DNase for 1 h. The bacterial biovolume was determined with digital image analysis. Staining with TOTO®-1 allowed visualization of eDNA both on bacterial cell surfaces and, with a cloud-like appearance, in the intercellular space. DNase treatment strongly reduced the amount of biofilm in very early stages of growth (up to 7.5 h), but the treatment effect decreased with increasing biofilm age. This study proves the involvement of eDNA in dental biofilm formation and its importance for biofilm stability in the earliest stages. Further research is required to uncover the interplay of eDNA and other matrix components and to explore the therapeutic potential of DNase treatment for biofilm control.
Collapse
Affiliation(s)
- Sebastian Schlafer
- Department of Dentistry and Oral Health,y, Aarhus University, Aarhus, Denmark
| | | | | | | |
Collapse
|
36
|
Shanmugam M, Oyeniyi AO, Parthiban C, Gujjarlapudi SK, Pier GB, Ramasubbu N. Role of de-N-acetylase PgaB from Aggregatibacter actinomycetemcomitans in exopolysaccharide export in biofilm mode of growth. Mol Oral Microbiol 2017; 32:500-510. [PMID: 28548373 DOI: 10.1111/omi.12188] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2017] [Indexed: 11/29/2022]
Abstract
Aggregatibacter actinomycetemcomitans, a Gram-negative bacterium, is the causative agent of localized aggressive periodontitis. Attachment to a biotic surface is a critical first step in the A. actinomycetemcomitans infection process for which exopolysaccharides have been shown to be essential. In addition, the pga operon, containing genes encoding for biosynthetic proteins for poly-N-acetyl glucosamine (PNAG), plays a key role in A. actinomycetemcomitans virulence, as a mutant strain lacking the pga operon induces significantly less bone resorption. Among the genes in the pga operon, pgaB codes for a de-N-acetylase that is responsible for the deacetylation of the PNAG exopolysaccharide. Here we report the role of PgaB in regulation of virulence genes using a markerless, scarless deletion mutant targeting the coding region of the N-terminal catalytic domain of PgaB. The results demonstrate that the N-terminal, catalytic domain of PgaB is crucial for exopolysaccharide export.
Collapse
Affiliation(s)
- M Shanmugam
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - A O Oyeniyi
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - C Parthiban
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - S K Gujjarlapudi
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| | - G B Pier
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - N Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, USA
| |
Collapse
|
37
|
Bacteriophage Lysin CF-301, a Potent Antistaphylococcal Biofilm Agent. Antimicrob Agents Chemother 2017; 61:AAC.02666-16. [PMID: 28461319 DOI: 10.1128/aac.02666-16] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/21/2017] [Indexed: 02/07/2023] Open
Abstract
Biofilms pose a unique therapeutic challenge because of the antibiotic tolerance of constituent bacteria. Treatments for biofilm-based infections represent a major unmet medical need, requiring novel agents to eradicate mature biofilms. Our objective was to evaluate bacteriophage lysin CF-301 as a new agent to target Staphylococcus aureus biofilms. We used minimum biofilm-eradicating concentration (MBEC) assays on 95 S. aureus strains to obtain a 90% MBEC (MBEC90) value of ≤0.25 μg/ml for CF-301. Mature biofilms of coagulase-negative staphylococci, Streptococcus pyogenes, and Streptococcus agalactiae were also sensitive to disruption, with MBEC90 values ranging from 0.25 to 8 μg/ml. The potency of CF-301 was demonstrated against S. aureus biofilms formed on polystyrene, glass, surgical mesh, and catheters. In catheters, CF-301 removed all biofilm within 1 h and killed all released bacteria by 6 h. Mixed-species biofilms, formed by S. aureus and Staphylococcus epidermidis on several surfaces, were removed by CF-301, as were S. aureus biofilms either enriched for small-colony variants (SCVs) or grown in human synovial fluid. The antibacterial activity of CF-301 was further demonstrated against S. aureus persister cells in exponential-phase and stationary-phase populations. Finally, the antibiofilm activity of CF-301 was greatly improved in combinations with the cell wall hydrolase lysostaphin when tested against a range of S. aureus strains. In all, the data show that CF-301 is highly effective at disrupting biofilms and killing biofilm bacteria, and, as such, it may be an efficient new agent for treating staphylococcal infections with a biofilm component.
Collapse
|
38
|
Abstract
Candida albicans, the most pervasive fungal pathogen that colonizes humans, forms biofilms that are architecturally complex. They consist of a basal yeast cell polylayer and an upper region of hyphae encapsulated in extracellular matrix. However, biofilms formed in vitro vary as a result of the different conditions employed in models, the methods used to assess biofilm formation, strain differences, and, in a most dramatic fashion, the configuration of the mating type locus (MTL). Therefore, integrating data from different studies can lead to problems of interpretation if such variability is not taken into account. Here we review the conditions and factors that cause biofilm variation, with the goal of engendering awareness that more attention must be paid to the strains employed, the methods used to assess biofilm development, every aspect of the model employed, and the configuration of the MTL locus. We end by posing a set of questions that may be asked in comparing the results of different studies and developing protocols for new ones. This review should engender the notion that not all biofilms are created equal.
Collapse
Affiliation(s)
- David R Soll
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| | - Karla J Daniels
- Developmental Studies Hybridoma Bank, Department of Biology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
39
|
Ahlstrand T, Tuominen H, Beklen A, Torittu A, Oscarsson J, Sormunen R, Pöllänen MT, Permi P, Ihalin R. A novel intrinsically disordered outer membrane lipoprotein of Aggregatibacter actinomycetemcomitans binds various cytokines and plays a role in biofilm response to interleukin-1β and interleukin-8. Virulence 2016; 8:115-134. [PMID: 27459270 PMCID: PMC5383217 DOI: 10.1080/21505594.2016.1216294] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) do not have a well-defined and stable 3-dimensional fold. Some IDPs can function as either transient or permanent binders of other proteins and may interact with an array of ligands by adopting different conformations. A novel outer membrane lipoprotein, bacterial interleukin receptor I (BilRI) of the opportunistic oral pathogen Aggregatibacter actinomycetemcomitans binds a key gatekeeper proinflammatory cytokine interleukin (IL)-1β. Because the amino acid sequence of the novel lipoprotein resembles that of fibrinogen binder A of Haemophilus ducreyi, BilRI could have the potential to bind other proteins, such as host matrix proteins. However, from the tested host matrix proteins, BilRI interacted with neither collagen nor fibrinogen. Instead, the recombinant non-lipidated BilRI, which was intrinsically disordered, bound various pro/anti-inflammatory cytokines, such as IL-8, tumor necrosis factor (TNF)-α, interferon (IFN)-γ and IL-10. Moreover, BilRI played a role in the in vitro sensing of IL-1β and IL-8 because low concentrations of cytokines did not decrease the amount of extracellular DNA in the matrix of bilRI− mutant biofilm as they did in the matrix of wild-type biofilm when the biofilms were exposed to recombinant cytokines for 22 hours. BilRI played a role in the internalization of IL-1β in the gingival model system but did not affect either IL-8 or IL-6 uptake. However, bilRI deletion did not entirely prevent IL-1β internalization, and the binding of cytokines to BilRI was relatively weak. Thus, BilRI might sequester cytokines on the surface of A. actinomycetemcomitans to facilitate the internalization process in low local cytokine concentrations.
Collapse
Affiliation(s)
- Tuuli Ahlstrand
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Heidi Tuominen
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Arzu Beklen
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Annamari Torittu
- a Department of Biochemistry , University of Turku , Turku , Finland
| | - Jan Oscarsson
- b Oral Microbiology , Department of Odontology, Umeå University , Umeå , Sweden
| | - Raija Sormunen
- c Biocenter Oulu and Department of Pathology , University of Oulu , Oulu Finland
| | | | - Perttu Permi
- e Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki , Helsinki , Finland.,f Department of Biological and Environmental Sciences , Nanoscience Center, University of Jyväskylä , Jyväskylä , Finland.,g Department of Chemistry , Nanoscience Center, University of Jyväskylä , Jyväskylä , Finland
| | - Riikka Ihalin
- a Department of Biochemistry , University of Turku , Turku , Finland
| |
Collapse
|
40
|
Majed R, Faille C, Kallassy M, Gohar M. Bacillus cereus Biofilms-Same, Only Different. Front Microbiol 2016; 7:1054. [PMID: 27458448 PMCID: PMC4935679 DOI: 10.3389/fmicb.2016.01054] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/23/2016] [Indexed: 12/24/2022] Open
Abstract
Bacillus cereus displays a high diversity of lifestyles and ecological niches and include beneficial as well as pathogenic strains. These strains are widespread in the environment, are found on inert as well as on living surfaces and contaminate persistently the production lines of the food industry. Biofilms are suspected to play a key role in this ubiquitous distribution and in this persistency. Indeed, B. cereus produces a variety of biofilms which differ in their architecture and mechanism of formation, possibly reflecting an adaptation to various environments. Depending on the strain, B. cereus has the ability to grow as immersed or floating biofilms, and to secrete within the biofilm a vast array of metabolites, surfactants, bacteriocins, enzymes, and toxins, all compounds susceptible to act on the biofilm itself and/or on its environment. Within the biofilm, B. cereus exists in different physiological states and is able to generate highly resistant and adhesive spores, which themselves will increase the resistance of the bacterium to antimicrobials or to cleaning procedures. Current researches show that, despite similarities with the regulation processes and effector molecules involved in the initiation and maturation of the extensively studied Bacillus subtilis biofilm, important differences exists between the two species. The present review summarizes the up to date knowledge on biofilms produced by B. cereus and by two closely related pathogens, Bacillus thuringiensis and Bacillus anthracis. Economic issues caused by B. cereus biofilms and management strategies implemented to control these biofilms are included in this review, which also discuss the ecological and functional roles of biofilms in the lifecycle of these bacterial species and explore future developments in this important research area.
Collapse
Affiliation(s)
- Racha Majed
- Micalis Institute, INRA, AgroParisTech, CNRS, Université Paris-SaclayJouy-en-Josas, France; Unité de Recherche Technologies et Valorisation Alimentaire, Laboratoire de Biotechnologie, Université Saint-JosephBeirut, Lebanon
| | - Christine Faille
- UMR UMET: Unité Matériaux et Transformations, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Université de Lille Villeneuve d'Ascq, France
| | - Mireille Kallassy
- Unité de Recherche Technologies et Valorisation Alimentaire, Laboratoire de Biotechnologie, Université Saint-Joseph Beirut, Lebanon
| | - Michel Gohar
- Micalis Institute, INRA, AgroParisTech, CNRS, Université Paris-SaclayJouy-en-Josas, France; Unité de Recherche Technologies et Valorisation Alimentaire, Laboratoire de Biotechnologie, Université Saint-JosephBeirut, Lebanon
| |
Collapse
|
41
|
Velusamy SK, Sampathkumar V, Godboley D, Fine DH. Profound Effects of Aggregatibacter actinomycetemcomitans Leukotoxin Mutation on Adherence Properties Are Clarified in in vitro Experiments. PLoS One 2016; 11:e0151361. [PMID: 26977924 PMCID: PMC4792451 DOI: 10.1371/journal.pone.0151361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/27/2016] [Indexed: 02/02/2023] Open
Abstract
Leukotoxin (Ltx) is a prominent virulence factor produced by Aggregatibacter actinomycetemcomitans, an oral microorganism highly associated with aggressive periodontitis. Ltx compromises host responsiveness by altering the viability of neutrophils, lymphocytes, and macrophages. Previously, we developed a Rhesus (Rh) monkey colonization model designed to determine the effect of virulence gene mutations on colonization of A. actinomycetemcomitans. Unexpectedly, an A. actinomycetemcomitans leukotoxin (ltxA) mutant (RhAa-VS2) failed to colonize in the Rh model. No previous literature suggested that Ltx was associated with A. actinomycetemcomitans binding to tooth surfaces. These results led us to explore the broad effects of the ltxA mutation in vitro. Results indicated that LtxA activity was completely abolished in RhAa-VS2 strain, while complementation significantly (P<0.0001) restored leukotoxicity compared to RhAa-VS2 strain. RT-PCR analysis of ltx gene expression ruled out polar effects. Furthermore, binding of RhAa-VS2 to salivary-coated hydroxyapatite (SHA) was significantly decreased (P<0.0001) compared to wild type RhAa3 strain. Real time RT-PCR analysis of the genes related to SHA binding in RhAa-VS2 showed that genes related to binding were downregulated [rcpA (P = 0.018), rcpB (P = 0.02), tadA (P = 0.002)] as compared to wild type RhAa3. RhAa-VS2 also exhibited decreased biofilm depth (P = 0.008) and exo-polysaccharide production (P<0.0001). Buccal epithelial cell (BEC) binding of RhAa-VS2 was unaffected. Complementation with ltxA restored binding to SHA (P<0.002) but had no effect on biofilm formation when compared to RhAa3. In conclusion, mutation of ltxA diminished hard tissue binding in vitro, which helps explain the previous in vivo failure of a ltxA knockout to colonize the Rh oral cavity. These results suggest that; 1) one specific gene knockout (in this case ltxA) could affect other seemingly unrelated genes (such as rcpA, rcpB tadA etc), and 2) some caution should be used when interpreting the effect attributed to targeted gene mutations when seen in a competitive in vivo environment.
Collapse
Affiliation(s)
- Senthil Kumar Velusamy
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark, New Jersey, United States of America
| | - Vandana Sampathkumar
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark, New Jersey, United States of America
| | - Dipti Godboley
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark, New Jersey, United States of America
| | - Daniel H. Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
42
|
Sager M, Benten WPM, Engelhardt E, Gougoula C, Benga L. Characterization of Biofilm Formation in [Pasteurella] pneumotropica and [Actinobacillus] muris Isolates of Mouse Origin. PLoS One 2015; 10:e0138778. [PMID: 26430880 PMCID: PMC4592018 DOI: 10.1371/journal.pone.0138778] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/03/2015] [Indexed: 12/24/2022] Open
Abstract
[Pasteurella] pneumotropica biotypes Jawetz and Heyl and [Actinobacillus] muris are the most prevalent Pasteurellaceae species isolated from laboratory mouse. However, mechanisms contributing to their high prevalence such as the ability to form biofilms have not been studied yet. In the present investigation we analyze if these bacterial species can produce biofilms in vitro and investigate whether proteins, extracellular DNA and polysaccharides are involved in the biofilm formation and structure by inhibition and dispersal assays using proteinase K, DNase I and sodium periodate. Finally, the capacity of the biofilms to confer resistance to antibiotics is examined. We demonstrate that both [P.] pneumotropica biotypes but not [A.] muris are able to form robust biofilms in vitro, a phenotype which is widely spread among the field isolates. The biofilm inhibition and dispersal assays by proteinase and DNase lead to a strong inhibition in biofilm formation when added at the initiation of the biofilm formation and dispersed pre-formed [P.] pneumotropica biofilms, revealing thus that proteins and extracellular DNA are essential in biofilm formation and structure. Sodium periodate inhibited the bacterial growth when added at the beginning of the biofilm formation assay, making difficult the assessment of the role of β-1,6-linked polysaccharides in the biofilm formation, and had a biofilm stimulating effect when added on pre-established mature biofilms of [P.] pneumotropica biotype Heyl and a majority of [P.] pneumotropica biotype Jawetz strains, suggesting that the presence of β-1,6-linked polysaccharides on the bacterial surface might attenuate the biofilm production. Conversely, no effect or a decrease in the biofilm quantity was observed by biofilm dispersal using sodium periodate on further biotype Jawetz isolates, suggesting that polysaccharides might be incorporated in the biofilm structure. We additionally show that [P.] pneumotropica cells enclosed in biofilms were less sensitive to treatment with amoxicillin and enrofloxacin than planktonic bacteria. Taken together, these findings provide a first step in understanding of the biofilm mechanisms in [P.] pneumotropica, which might contribute to elucidation of colonization and pathogenesis mechanisms for these obligate inhabitants of the mouse mucosa.
Collapse
Affiliation(s)
- Martin Sager
- Central Animal Research Facility, Heinrich—Heine—University, University Hospital, Düsseldorf, Germany
| | - W. Peter M. Benten
- Central Animal Research Facility, Heinrich—Heine—University, University Hospital, Düsseldorf, Germany
| | - Eva Engelhardt
- Central Animal Research Facility, Heinrich—Heine—University, University Hospital, Düsseldorf, Germany
| | - Christina Gougoula
- Central Animal Research Facility, Heinrich—Heine—University, University Hospital, Düsseldorf, Germany
| | - Laurentiu Benga
- Central Animal Research Facility, Heinrich—Heine—University, University Hospital, Düsseldorf, Germany
| |
Collapse
|
43
|
Herbert BA, Novince CM, Kirkwood KL. Aggregatibacter actinomycetemcomitans, a potent immunoregulator of the periodontal host defense system and alveolar bone homeostasis. Mol Oral Microbiol 2015. [PMID: 26197893 DOI: 10.1111/omi.12119] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aggregatibacter actinomycetemcomitans is a perio-pathogenic bacteria that has long been associated with localized aggressive periodontitis. The mechanisms of its pathogenicity have been studied in humans and preclinical experimental models. Although different serotypes of A. actinomycetemcomitans have differential virulence factor expression, A. actinomycetemcomitans cytolethal distending toxin (CDT), leukotoxin, and lipopolysaccharide (LPS) have been most extensively studied in the context of modulating the host immune response. Following colonization and attachment in the oral cavity, A. actinomycetemcomitans employs CDT, leukotoxin, and LPS to evade host innate defense mechanisms and drive a pathophysiologic inflammatory response. This supra-physiologic immune response state perturbs normal periodontal tissue remodeling/turnover and ultimately has catabolic effects on periodontal tissue homeostasis. In this review, we have divided the host response into two systems: non-hematopoietic and hematopoietic. Non-hematopoietic barriers include epithelium and fibroblasts that initiate the innate immune host response. The hematopoietic system contains lymphoid and myeloid-derived cell lineages that are responsible for expanding the immune response and driving the pathophysiologic inflammatory state in the local periodontal microenvironment. Effector systems and signaling transduction pathways activated and utilized in response to A. actinomycetemcomitans will be discussed to further delineate immune cell mechanisms during A. actinomycetemcomitans infection. Finally, we will discuss the osteo-immunomodulatory effects induced by A. actinomycetemcomitans and dissect the catabolic disruption of balanced osteoclast-osteoblast-mediated bone remodeling, which subsequently leads to net alveolar bone loss.
Collapse
Affiliation(s)
- B A Herbert
- Department of Oral Health Sciences and the Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - C M Novince
- Department of Oral Health Sciences and the Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - K L Kirkwood
- Department of Oral Health Sciences and the Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
44
|
Transcriptome Profiling of Wild-Type and pga-Knockout Mutant Strains Reveal the Role of Exopolysaccharide in Aggregatibacter actinomycetemcomitans. PLoS One 2015. [PMID: 26221956 PMCID: PMC4519337 DOI: 10.1371/journal.pone.0134285] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Exopolysaccharides have a diverse set of functions in most bacteria including a mechanistic role in protecting bacteria against environmental stresses. Among the many functions attributed to the exopolysaccharides, biofilm formation, antibiotic resistance, immune evasion and colonization have been studied most extensively. The exopolysaccharide produced by many Gram positive as well as Gram negative bacteria including the oral pathogen Aggregatibacter actinomycetemcomitans is the homopolymer of β(1,6)-linked N-acetylglucosamine. Recently, we reported that the PGA-deficient mutant of A. actinomycetemcomitans failed to colonize or induce bone resorption in a rat model of periodontal disease, and the colonization genes, apiA and aae, were significantly down regulated in the mutant strain. To understand the role of exopolysaccharide and the pga locus in the global expression of A. actinomycetemcomitans, we have used comparative transcriptome profiling to identify differentially expressed genes in the wild-type strain in relation to the PGA-deficient strain. Transcriptome analysis revealed that about 50% of the genes are differently expressed (P < 0.05 and fold change >1.5). Our study demonstrated that the absence of the pga locus affects the genes involved in peptidoglycan recycling, glycogen storage, and virulence. Further, using confocal microscopy and plating assays, we show that the viability of pga mutant strain is significantly reduced during biofilm growth. Thus, this study highlights the importance of pga genes and the exopolysaccharide in the virulence of A. actinomycetemcomitans.
Collapse
|
45
|
Richter AM, Povolotsky TL, Wieler LH, Hengge R. Cyclic-di-GMP signalling and biofilm-related properties of the Shiga toxin-producing 2011 German outbreak Escherichia coli O104:H4. EMBO Mol Med 2015; 6:1622-37. [PMID: 25361688 PMCID: PMC4287979 DOI: 10.15252/emmm.201404309] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
In 2011, nearly 4,000 people in Germany were infected by Shiga toxin (Stx)-producing Escherichia coli O104:H4 with > 22% of patients developing haemolytic uraemic syndrome (HUS). Genome sequencing showed the outbreak strain to be related to enteroaggregative E. coli (EAEC), suggesting its high virulence results from EAEC-typical strong adherence and biofilm formation combined to Stx production. Here, we report that the outbreak strain contains a novel diguanylate cyclase (DgcX)--producing the biofilm-promoting second messenger c-di-GMP--that shows higher expression than any other known E. coli diguanylate cyclase. Unlike closely related E. coli, the outbreak strain expresses the c-di-GMP-controlled biofilm regulator CsgD and amyloid curli fibres at 37°C, but is cellulose-negative. Moreover, it constantly generates derivatives with further increased and deregulated production of CsgD and curli. Since curli fibres are strongly proinflammatory, with cellulose counteracting this effect, high c-di-GMP and curli production by the outbreak O104:H4 strain may enhance not only adherence but may also contribute to inflammation, thereby facilitating entry of Stx into the bloodstream and to the kidneys where Stx causes HUS.
Collapse
Affiliation(s)
- Anja M Richter
- Institute of Biology / Microbiology Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tatyana L Povolotsky
- Institute of Biology / Microbiology Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lothar H Wieler
- Institute of Microbiology and Epizootics Freie Universität Berlin, Berlin, Germany
| | - Regine Hengge
- Institute of Biology / Microbiology Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
46
|
Whitfield GB, Marmont LS, Howell PL. Enzymatic modifications of exopolysaccharides enhance bacterial persistence. Front Microbiol 2015; 6:471. [PMID: 26029200 PMCID: PMC4432689 DOI: 10.3389/fmicb.2015.00471] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/29/2015] [Indexed: 12/25/2022] Open
Abstract
Biofilms are surface-attached communities of bacterial cells embedded in a self-produced matrix that are found ubiquitously in nature. The biofilm matrix is composed of various extracellular polymeric substances, which confer advantages to the encapsulated bacteria by protecting them from eradication. The matrix composition varies between species and is dependent on the environmental niche that the bacteria inhabit. Exopolysaccharides (EPS) play a variety of important roles in biofilm formation in numerous bacterial species. The ability of bacteria to thrive in a broad range of environmental settings is reflected in part by the structural diversity of the EPS produced both within individual bacterial strains as well as by different species. This variability is achieved through polymerization of distinct sugar moieties into homo- or hetero-polymers, as well as post-polymerization modification of the polysaccharide. Specific enzymes that are unique to the production of each polymer can transfer or remove non-carbohydrate moieties, or in other cases, epimerize the sugar units. These modifications alter the physicochemical properties of the polymer, which in turn can affect bacterial pathogenicity, virulence, and environmental adaptability. Herein, we review the diversity of modifications that the EPS alginate, the Pel polysaccharide, Vibrio polysaccharide, cepacian, glycosaminoglycans, and poly-N-acetyl-glucosamine undergo during biosynthesis. These are EPS produced by human pathogenic bacteria for which studies have begun to unravel the effect modifications have on their physicochemical and biological properties. The biological advantages these polymer modifications confer to the bacteria that produce them will be discussed. The expanding list of identified modifications will allow future efforts to focus on linking these modifications to specific biosynthetic genes and biofilm phenotypes.
Collapse
Affiliation(s)
- Gregory B Whitfield
- Program in Molecular Structure and Function, Research Institute, The Hospital for Sick Children Toronto, ON, Canada ; Department of Biochemistry, Faculty of Medicine, University of Toronto Toronto, ON, Canada
| | - Lindsey S Marmont
- Program in Molecular Structure and Function, Research Institute, The Hospital for Sick Children Toronto, ON, Canada ; Department of Biochemistry, Faculty of Medicine, University of Toronto Toronto, ON, Canada
| | - P Lynne Howell
- Program in Molecular Structure and Function, Research Institute, The Hospital for Sick Children Toronto, ON, Canada ; Department of Biochemistry, Faculty of Medicine, University of Toronto Toronto, ON, Canada
| |
Collapse
|
47
|
Karched M, Bhardwaj RG, Inbamani A, Asikainen S. Quantitation of biofilm and planktonic life forms of coexisting periodontal species. Anaerobe 2015; 35:13-20. [PMID: 25926392 DOI: 10.1016/j.anaerobe.2015.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/21/2015] [Accepted: 04/24/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Complexity of oral polymicrobial communities has prompted a need for developing in vitro models to study behavior of coexisting bacteria. Little knowledge is available of in vitro co-growth of several periodontitis-associated species without early colonizers of dental plaque. THE AIM was to determine temporal changes in the quantities of six periodontal species in an in vitro biofilm model in comparison with parallel planktonic cultures. MATERIAL AND METHODS Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, Prevotella intermedia, Parvimonas micra, Campylobacter rectus and Fusobacterium nucleatum were anaerobically grown as multispecies and monospecies biofilms and parallel planktonic cultures using cell culture plates and microfuge tubes, respectively. After incubating 2, 4, 6, 8 days, biofilms and planktonic cultures were harvested, DNA extracted and the target species quantified using qPCR with species-specific 16S rDNA primers. Biofilm growth as monocultures was visualized at day 2 and 8 with confocal microscopy and crystal violet staining. RESULTS The six species were found throughout the test period in all culture conditions, except that P. gingivalis and F. nucleatum were not detected in multispecies planktonic cultures at day 8. In multispecies biofilm, P. gingivalis qPCR counts (cells/ml) increased (P<0.05) from day 2-8 and were then higher (P<0.05) than those of A. actinomycetemcomitans and C. rectus, whereas in monospecies biofilm, P. gingivalis counts were lower (P<0.05) than those of the other species, except A. actinomycetemcomitans. When multi- and monospecies biofilm cultures were compared, P. gingivalis counts were higher (P<0.05) but those of the other species, except P. intermedia, lower (P<0.05) in multispecies biofilm. Comparison between planktonic and biofilm cultures showed that A. actinomycetemcomitans, P. micra and C. rectus had higher (P<0.05) counts in planktonic cultures no matter whether grown in mono- or multispecies environment. CONCLUSIONS Six periodontal species were able to form multispecies biofilm up to 8 days in vitro without pioneer plaque bacteria. P. gingivalis seemed to prefer multispecies biofilm environment whereas P. micra and A. actinomycetemcomitans planktonic culture.
Collapse
Affiliation(s)
- Maribasappa Karched
- Oral Microbiology, General Facility Laboratory, Faculty of Dentistry, Kuwait University, Kuwait
| | - Radhika G Bhardwaj
- Oral Microbiology, General Facility Laboratory, Faculty of Dentistry, Kuwait University, Kuwait
| | - Anandavalli Inbamani
- Oral Microbiology, General Facility Laboratory, Faculty of Dentistry, Kuwait University, Kuwait
| | - Sirkka Asikainen
- Oral Microbiology, General Facility Laboratory, Faculty of Dentistry, Kuwait University, Kuwait.
| |
Collapse
|
48
|
Shanmugam M, Gopal P, El Abbar F, Schreiner HC, Kaplan JB, Fine DH, Ramasubbu N. Role of exopolysaccharide in Aggregatibacter actinomycetemcomitans-induced bone resorption in a rat model for periodontal disease. PLoS One 2015; 10:e0117487. [PMID: 25706999 PMCID: PMC4338281 DOI: 10.1371/journal.pone.0117487] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 12/24/2014] [Indexed: 11/19/2022] Open
Abstract
Aggregatibacter actinomycetemcomitans a causative agent of periodontal disease in humans, forms biofilm on biotic and abiotic surfaces. A. actinomycetemcomitans biofilm is heterogeneous in nature and is composed of proteins, extracellular DNA and exopolysaccharide. To explore the role played by the exopolysaccharide in the colonization and disease progression, we employed genetic reduction approach using our rat model of A. actinomycetemcomitans-induced periodontitis. To this end, a genetically modified strain of A. actinomycetemcomitans lacking the pga operon was compared with the wild-type strain in the rat infection model. The parent and mutant strains were primarily evaluated for bone resorption and disease. Our study showed that colonization, bone resorption/disease and antibody response were all elevated in the wild-type fed rats. The bone resorption/disease caused by the pga mutant strain, lacking the exopolysaccharide, was significantly less (P < 0.05) than the bone resorption/disease caused by the wild-type strain. Further analysis of the expression levels of selected virulence genes through RT-PCR showed that the decrease in colonization, bone resorption and antibody titer in the absence of the exopolysaccharide might be due to attenuated levels of colonization genes, flp-1, apiA and aae in the mutant strain. This study demonstrates that the effect exerted by the exopolysaccharide in A. actinomycetemcomitans-induced bone resorption has hitherto not been recognized and underscores the role played by the exopolysaccharide in A. actinomycetemcomitans-induced disease.
Collapse
Affiliation(s)
- Mayilvahanan Shanmugam
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Prerna Gopal
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Faiha El Abbar
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Helen C Schreiner
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Jeffrey B Kaplan
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Daniel H Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| | - Narayanan Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, 07103, United States of America
| |
Collapse
|
49
|
Alvarenga LH, Prates RA, Yoshimura TM, Kato IT, Suzuki LC, Ribeiro MS, Ferreira LR, Pereira SADS, Martinez EF, Saba-Chujfi E. Aggregatibacter actinomycetemcomitans biofilm can be inactivated by methylene blue-mediated photodynamic therapy. Photodiagnosis Photodyn Ther 2014; 12:131-5. [PMID: 25461964 DOI: 10.1016/j.pdpdt.2014.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 10/01/2014] [Accepted: 10/05/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the antibacterial effects of photodynamic action of methylene blue (MB) against Aggregatibacter actinomycetemcomitans organized on biofilm. METHODS After the biofilm growth in 96 flat-bottom well plate, the following groups were used: control group, untreated by either laser or photosensitizer (PS); MB group or dark toxicity group, which was exposed to MB alone (100μM) for 1min (pre-irradiation time); laser group, irradiated with laser for 5min in the absence of PS and three antimicrobial photodynamic inactivation (APDI) groups, with three exposure times of 1, 3 and 5min of irradiation, corresponding to fluences of 15, 45, and 75J/cm(2) respectively. The results were compared to the control group for statistical proposes. Scanning electronic microscope analysis was used to access structural changes in biofilm. RESULTS Red laser alone and MB alone were not able to inactivate bacterial biofilm. APDI groups showed differences when compared to the control group and they were dependent on the exposure time. No statistically significant differences were observed among the APDI groups at 1 and 3min of irradiation. On the other hand, 5min of APDI showed 99.85% of bacterial reduction (p=0.0004). In addition, the biofilm loose its structure following 5min APDI. CONCLUSIONS The results of this study suggest that A. actinomycetemcomitans biofilm can be inactivated by MB mediated APDI.
Collapse
Affiliation(s)
| | - Renato Araujo Prates
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil; School of Dentistry, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil.
| | - Tania Mateus Yoshimura
- Center for Lasers and Applications, Nuclear and Energy Research Institute IPEN-CNEN/SP, São Paulo, Brazil
| | - Ilka Tiemy Kato
- Department of Biomedical Engineering, ABC Federal University (UFABC), Santo Andre, SP, Brazil
| | - Luis Cláudio Suzuki
- Center for Lasers and Applications, Nuclear and Energy Research Institute IPEN-CNEN/SP, São Paulo, Brazil
| | - Martha Simões Ribeiro
- Center for Lasers and Applications, Nuclear and Energy Research Institute IPEN-CNEN/SP, São Paulo, Brazil
| | - Luis Rodolfo Ferreira
- Postgraduate Program in Biophotonics Applied to Health Sciences, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil; School of Dentistry, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | | | | | | |
Collapse
|
50
|
Jaglic Z, Desvaux M, Weiss A, Nesse LL, Meyer RL, Demnerova K, Schmidt H, Giaouris E, Sipailiene A, Teixeira P, Kačániová M, Riedel CU, Knøchel S. Surface adhesins and exopolymers of selected foodborne pathogens. MICROBIOLOGY-SGM 2014; 160:2561-2582. [PMID: 25217529 DOI: 10.1099/mic.0.075887-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The ability of bacteria to bind different compounds and to adhere to biotic and abiotic surfaces provides them with a range of advantages, such as colonization of various tissues, internalization, avoidance of an immune response, and survival and persistence in the environment. A variety of bacterial surface structures are involved in this process and these promote bacterial adhesion in a more or less specific manner. In this review, we will focus on those surface adhesins and exopolymers in selected foodborne pathogens that are involved mainly in primary adhesion. Their role in biofilm development will also be considered when appropriate. Both the clinical impact and the implications for food safety of such adhesion will be discussed.
Collapse
Affiliation(s)
- Zoran Jaglic
- Veterinary Research Institute, Brno, Czech Republic
| | - Mickaël Desvaux
- INRA, UR454 Microbiologie, F-63122 Saint-Genès Champanelle, France
| | - Agnes Weiss
- Department of Food Microbiology, Institute of Food Science and Biotechnology, University of Hohenheim, Garbenstrasse 28, 70599 Stuttgart, Germany
| | | | - Rikke L Meyer
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, DK-8000 Aarhus C, Denmark
| | - Katerina Demnerova
- Institute of Chemical Technology, Faculty of Food and Biochemical Technology, Department of Biochemistry and Microbiology, Technicka 5, Prague, 166 28, Czech Republic
| | - Herbert Schmidt
- Department of Food Microbiology, Institute of Food Science and Biotechnology, University of Hohenheim, Garbenstrasse 28, 70599 Stuttgart, Germany
| | - Efstathios Giaouris
- Department of Food Science and Nutrition, Faculty of the Environment, University of the Aegean, 81400 Myrina, Lemnos Island, Greece
| | | | - Pilar Teixeira
- CEB - Centre of Biological Engineering, University of Minho, Braga, Portugal
| | | | - Christian U Riedel
- Institute of Microbiology and Biotechnology, University of Ulm, Ulm, Germany
| | - Susanne Knøchel
- Department of Food Science, University of Copenhagen, Rolighedsvej 30, Frederiksberg C 1958, Denmark
| |
Collapse
|