1
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
2
|
Zhang J, Hao J, Wang J, Li H, Zhao D. Strategic manipulation of biofilm dispersion for controlling Listeria monocytogenes infections. Crit Rev Food Sci Nutr 2024:1-10. [PMID: 39367886 DOI: 10.1080/10408398.2024.2409340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
Listeria monocytogenes (L. monocytogenes), a gram-positive foodborne pathogen that can easily cause listeriosis. It secretes extracellular polymers and forms biofilms that are highly resistant to disinfection methods, such as UV light and germicides, posing risks to food processing equipment and food quality. Dispersion of biofilm is the cycle of its formation in which the bacteria return to planktonic state and become susceptible to antimicrobials, the strategic manipulation of biofilm dispersion is thus heralded as a novel and promising approach for the effective control of biofilm-related infections. Compared to the traditional methods, it is more effective to start with the composition of biofilms, cut off the production of their constituent substances, and genetically reduce the probability of biofilm formation. Meanwhile, the dispersion of bacteria can be supplemented with exogenous substances, making long-term control possible. This paper provides a brief but comprehensive overview of the mechanisms of L. monocytogenes biofilms or cross-contamination and their resistance properties, and facilitates our understanding and control of the prevention and containment of L. monocytogenes biofilm contamination based on the biofilm's active and passive diffusion strategies. This work provides practical guidelines for the food industry to guard against the enduring threat to food safety due to L. monocytogenes biofilms.
Collapse
Affiliation(s)
- Junyi Zhang
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, China
| | - Jianxiong Hao
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, China
| | - Jingyi Wang
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huiying Li
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, China
| | - Dandan Zhao
- College of Food Science and Biology, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
3
|
Ahmed F, Mirani ZA, Urooj S, Noor Ul Hudda H, Janees Imdad M, Zhao Y, Malakar PK. A rare biofilm dispersion strategy demonstrated by Staphylococcus aureus under oxacillin stress. Microb Pathog 2024; 194:106838. [PMID: 39111368 DOI: 10.1016/j.micpath.2024.106838] [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: 02/17/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Staphylococcus aureus (S. aureus), a versatile Gram-positive bacterium, is implicated in a spectrum of infections, and its resilience is often attributed to biofilm formation. This study investigates the effect of sub-inhibitory doses of oxacillin on biofilm formation by methicillin-resistant S. aureus (MRSA). Specifically, it examines how these doses influence biofilms' development, maturation, and dispersal. The biofilm's zenith reached 48 h of incubation, followed by a noteworthy decline at 96 h and a distinctive clearance zone around biofilm-positive cells exposed to oxacillin. Scanning electron micrographs unveiled an intriguing active biofilm dispersal mechanism, a rarity in this species. Among 180 isolates, only three carrying the elusive icaD gene exhibited this phenomenon. icaD gene was absent in their counterparts. Notably, the icaD gene emerges as a distinctive marker, crucial in regulating biofilm dispersion and setting these isolates apart. The captivating interplay of oxacillin, biofilm dynamics, and genetic signatures disintegrate novel dimensions in understanding MRSA's adaptive strategies and underscores the importance of the icaD gene in engineering biofilm resilience.
Collapse
Affiliation(s)
- Faraz Ahmed
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China.
| | - Zulfiqar Ali Mirani
- Microbiology Section, FMRRC, PCSIR Laboratories Complex Karachi, Sindh, Pakistan
| | - Shaista Urooj
- Aquatic Diagnostic and Research Center Bahria University, Karachi, Sindh, Pakistan
| | | | - Muhammad Janees Imdad
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China
| | - Yong Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China; Laboratory of Quality & Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture and Rural Affairs, Shanghai, China; Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai, China
| | - Pradeep K Malakar
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China; International Research Centre for Food and Health, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
4
|
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] [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
|
5
|
Pirușcă IA, Balaure PC, Grumezescu V, Irimiciuc SA, Oprea OC, Bîrcă AC, Vasile B, Holban AM, Voinea IC, Stan MS, Trușcă R, Grumezescu AM, Croitoru GA. New Fe 3O 4-Based Coatings with Enhanced Anti-Biofilm Activity for Medical Devices. Antibiotics (Basel) 2024; 13:631. [PMID: 39061313 PMCID: PMC11273941 DOI: 10.3390/antibiotics13070631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
With the increasing use of invasive, interventional, indwelling, and implanted medical devices, healthcare-associated infections caused by pathogenic biofilms have become a major cause of morbidity and mortality. Herein, we present the fabrication, characterization, and in vitro evaluation of biocompatibility and anti-biofilm properties of new coatings based on Fe3O4 nanoparticles (NPs) loaded with usnic acid (UA) and ceftriaxone (CEF). Sodium lauryl sulfate (SLS) was employed as a stabilizer and modulator of the polarity, dispersibility, shape, and anti-biofilm properties of the magnetite nanoparticles. The resulting Fe3O4 functionalized NPs, namely Fe3O4@SLS, Fe3O4@SLS/UA, and Fe3O4@SLS/CEF, respectively, were prepared by co-precipitation method and fully characterized by XRD, TEM, SAED, SEM, FTIR, and TGA. They were further used to produce nanostructured coatings by matrix-assisted pulsed laser evaporation (MAPLE) technique. The biocompatibility of the coatings was assessed by measuring the cell viability, lactate dehydrogenase release, and nitric oxide level in the culture medium and by evaluating the actin cytoskeleton morphology of murine pre-osteoblasts. All prepared nanostructured coatings exhibited good biocompatibility. Biofilm growth inhibition ability was tested at 24 h and 48 h against Staphylococcus aureus and Pseudomonas aeruginosa as representative models for Gram-positive and Gram-negative bacteria. The coatings demonstrated good biocompatibility, promoting osteoblast adhesion, migration, and growth without significant impact on cell viability or morphology, highlighting their potential for developing safe and effective antibacterial surfaces.
Collapse
Affiliation(s)
- Ioana Adelina Pirușcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (I.A.P.); (A.C.B.); (B.V.); (R.T.)
| | - Paul Cătălin Balaure
- Department of Organic Chemistry, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania
| | - Valentina Grumezescu
- Lasers Department, National Institute for Laser, Plasma and Radiation Physics, 077125 Magurele, Romania; (V.G.)
| | - Stefan-Andrei Irimiciuc
- Lasers Department, National Institute for Laser, Plasma and Radiation Physics, 077125 Magurele, Romania; (V.G.)
| | - Ovidiu-Cristian Oprea
- Department of Inorganic Chemistry, Physical Chemistry and Electrochemistry, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania;
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (I.A.P.); (A.C.B.); (B.V.); (R.T.)
| | - Bogdan Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (I.A.P.); (A.C.B.); (B.V.); (R.T.)
| | - Alina Maria Holban
- Microbiology and Immunology Department, Faculty of Biology, University of Bucharest, 77206 Bucharest, Romania;
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (I.C.V.); (M.S.S.)
| | - Ionela C. Voinea
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (I.C.V.); (M.S.S.)
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Miruna S. Stan
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (I.C.V.); (M.S.S.)
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Roxana Trușcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (I.A.P.); (A.C.B.); (B.V.); (R.T.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology POLITEHNICA Bucharest, 011061 Bucharest, Romania; (I.A.P.); (A.C.B.); (B.V.); (R.T.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050663 Bucharest, Romania; (I.C.V.); (M.S.S.)
| | - George-Alexandru Croitoru
- Department II, Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
6
|
Kaplan JB, Cywes-Bentley C, Pier GB, Yakandawala N, Sailer M, Edwards MS, Kridin K. Poly- β-(1→6)- N-acetyl-D-glucosamine mediates surface attachment, biofilm formation, and biocide resistance in Cutibacterium acnes. Front Microbiol 2024; 15:1386017. [PMID: 38751716 PMCID: PMC11094747 DOI: 10.3389/fmicb.2024.1386017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/18/2024] Open
Abstract
Background The commensal skin bacterium Cutibacterium acnes plays a role in the pathogenesis of acne vulgaris and also causes opportunistic infections of implanted medical devices due to its ability to form biofilms on biomaterial surfaces. Poly-β-(1→6)-N-acetyl-D-glucosamine (PNAG) is an extracellular polysaccharide that mediates biofilm formation and biocide resistance in a wide range of bacterial pathogens. The objective of this study was to determine whether C. acnes produces PNAG, and whether PNAG contributes to C. acnes biofilm formation and biocide resistance in vitro. Methods PNAG was detected on the surface of C. acnes cells by fluorescence confocal microscopy using the antigen-specific human IgG1 monoclonal antibody F598. PNAG was detected in C. acnes biofilms by measuring the ability of the PNAG-specific glycosidase dispersin B to inhibit biofilm formation and sensitize biofilms to biocide killing. Results Monoclonal antibody F598 bound to the surface of C. acnes cells. Dispersin B inhibited attachment of C. acnes cells to polystyrene rods, inhibited biofilm formation by C. acnes in glass and polypropylene tubes, and sensitized C. acnes biofilms to killing by benzoyl peroxide and tetracycline. Conclusion C. acnes produces PNAG, and PNAG contributes to C. acnes biofilm formation and biocide resistance in vitro. PNAG may play a role in C. acnes skin colonization, biocide resistance, and virulence in vivo.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Department of Biology, American University, Washington, DC, United States
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya, Israel
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | | | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya, Israel
| |
Collapse
|
7
|
Wang D, Naqvi STA, Lei F, Zhang Z, Yu H, Ma LZ. Glycosyl hydrolase from Pseudomonas fluorescens inhibits the biofilm formation of Pseudomonads. Biofilm 2023; 6:100155. [PMID: 37928620 PMCID: PMC10622837 DOI: 10.1016/j.bioflm.2023.100155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 11/07/2023] Open
Abstract
Biofilms are complex microbial communities embedded in extracellular matrix. Pathogens within the biofilm become more resistant to the antibiotics than planktonic counterparts. Novel strategies are required to encounter biofilms. Exopolysaccharides are one of the major components of biofilm matrix and play a vital role in biofilm architecture. In previous studies, a glycosyl hydrolase, PslGPA, from Pseudomonas aeruginosa was found to be able to inhibit biofilm formation by disintegrating exopolysaccharide in biofilms. Here, we investigate the potential spectrum of PslG homologous protein with anti-biofilm activity. One glycosyl hydrolase from Pseudomonas fluorescens, PslGPF, exhibits anti-biofilm activities and the key catalytic residues of PslGPF are conserved with those of PslGPA. PslGPF at concentrations as low as 50 nM efficiently inhibits the biofilm formation of P. aeruginosa and disassemble its preformed biofilm. Furthermore, PslGPF exhibits anti-biofilm activity on a series of Pseudomonads, including P. fluorescens, Pseudomonas stutzeri and Pseudomonas syringae pv. phaseolicola. PslGPF stays active under various temperatures. Our findings suggest that P. fluorescens glycosyl hydrolase PslGPF has potential to be a broad spectrum inhibitor on biofilm formation of a wide range of Pseudomonads.
Collapse
Affiliation(s)
- Di Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Syed Tatheer Alam Naqvi
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Fanglin Lei
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Yunnan University, Kunming, 650500, PR China
| | - Zhenyu Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Haiying Yu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Luyan Z. Ma
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
8
|
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: 38] [Impact Index Per Article: 19.0] [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
|
9
|
Sun H, Sun M, You Y, Xie J, Xu X, Li J. Recent progress of intelligent antibacterial nanoplatforms for treating bacterial infection. CHEMICAL ENGINEERING JOURNAL 2023; 471:144597. [DOI: 10.1016/j.cej.2023.144597] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
10
|
Kilic T, Bali EB. Biofilm control strategies in the light of biofilm-forming microorganisms. World J Microbiol Biotechnol 2023; 39:131. [PMID: 36959476 DOI: 10.1007/s11274-023-03584-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/15/2023] [Indexed: 03/25/2023]
Abstract
Biofilm is a complex consortium of microorganisms attached to biotic or abiotic surfaces and live in self-produced or acquired extracellular polymeric substances (EPSs). EPSs are mainly formed by lipids, polysaccharides, proteins, and extracellular DNAs. The adherence to the surface of microbial communities is seen in food, medical, dental, industrial, and environmental fields. Biofilm development in food processing areas challenges food hygiene, and human health. In addition, bacterial attachment and biofilm formation on medical implants inside human tissue can cause multiple critical chronic infections. More than 30 years of international research on the mechanisms of biofilm formation have been underway to address concerns about bacterial biofilm infections. Antibiofilm strategies contain cold atmospheric plasma, nanotechnological, phage-based, antimicrobial peptides, and quorum sensing inhibition. In the last years, the studies on environmentally-friendly techniques such as essential oils and bacteriophages have been intensified to reduce microbial growth. However, the mechanisms of the biofilm matrix formation are still unclear. This review aims to discuss the latest antibiofilm therapeutic strategies against biofilm-forming bacteria.
Collapse
Affiliation(s)
- Tugba Kilic
- Department of Medical Services and Techniques, Program of Medical Laboratory Techniques, Vocational School of Health Services, Gazi University, Ankara, 06830, Turkey.
| | - Elif Burcu Bali
- Department of Medical Services and Techniques, Program of Medical Laboratory Techniques, Vocational School of Health Services, Gazi University, Ankara, 06830, Turkey
| |
Collapse
|
11
|
Gill SP, Hunter WR, Coulson LE, Banat IM, Schelker J. Synthetic and biological surfactant effects on freshwater biofilm community composition and metabolic activity. Appl Microbiol Biotechnol 2022; 106:6847-6859. [PMID: 36121483 PMCID: PMC9529700 DOI: 10.1007/s00253-022-12179-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/06/2022]
Abstract
Surfactants are used to control microbial biofilms in industrial and medical settings. Their known toxicity on aquatic biota, and their longevity in the environment, has encouraged research on biodegradable alternatives such as rhamnolipids. While previous research has investigated the effects of biological surfactants on single species biofilms, there remains a lack of information regarding the effects of synthetic and biological surfactants in freshwater ecosystems. We conducted a mesocosm experiment to test how the surfactant sodium dodecyl sulfate (SDS) and the biological surfactant rhamnolipid altered community composition and metabolic activity of freshwater biofilms. Biofilms were cultured in the flumes using lake water from Lake Lunz in Austria, under high (300 ppm) and low (150 ppm) concentrations of either surfactant over a four-week period. Our results show that both surfactants significantly affected microbial diversity. Up to 36% of microbial operational taxonomic units were lost after surfactant exposure. Rhamnolipid exposure also increased the production of the extracellular enzymes, leucine aminopeptidase, and glucosidase, while SDS exposure reduced leucine aminopeptidase and glucosidase. This study demonstrates that exposure of freshwater biofilms to chemical and biological surfactants caused a reduction of microbial diversity and changes in biofilm metabolism, exemplified by shifts in extracellular enzyme activities. KEY POINTS: • Microbial biofilm diversity decreased significantly after surfactant exposure. • Exposure to either surfactant altered extracellular enzyme activity. • Overall metabolic activity was not altered, suggesting functional redundancy.
Collapse
Affiliation(s)
- Stephanie P Gill
- Department of Geography and Environmental Studies, Ulster University, Coleraine, BT52 1SA, N. Ireland, UK.
| | - William R Hunter
- Fisheries and Aquatic Ecosystems Branch, Agri-Food and Biosciences Institute, Belfast, N. Ireland, UK
| | - Laura E Coulson
- WasserCluster Lunz, Lunz am See, Austria
- Institute of Hydrobiology and Aquatic Ecosystem Management, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Ibrahim M Banat
- School of Biomedical Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - Jakob Schelker
- WasserCluster Lunz, Lunz am See, Austria
- Department of Functional and Evolutionary Ecology, University of Vienna, Vienna, Austria
| |
Collapse
|
12
|
Ramakrishnan R, Singh AK, Singh S, Chakravortty D, Das D. Enzymatic Dispersion of Biofilms: An Emerging Biocatalytic Avenue to Combat Biofilm-Mediated Microbial Infections. J Biol Chem 2022; 298:102352. [PMID: 35940306 PMCID: PMC9478923 DOI: 10.1016/j.jbc.2022.102352] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/01/2023] Open
Abstract
Drug resistance by pathogenic microbes has emerged as a matter of great concern to mankind. Microorganisms such as bacteria and fungi employ multiple defense mechanisms against drugs and the host immune system. A major line of microbial defense is the biofilm, which comprises extracellular polymeric substances that are produced by the population of microorganisms. Around 80% of chronic bacterial infections are associated with biofilms. The presence of biofilms can increase the necessity of doses of certain antibiotics up to 1000-fold to combat infection. Thus, there is an urgent need for strategies to eradicate biofilms. Although a few physicochemical methods have been developed to prevent and treat biofilms, these methods have poor efficacy and biocompatibility. In this review, we discuss the existing strategies to combat biofilms and their challenges. Subsequently, we spotlight the potential of enzymes, in particular, polysaccharide degrading enzymes, for biofilm dispersion, which might lead to facile antimicrobial treatment of biofilm-associated infections.
Collapse
Affiliation(s)
- Reshma Ramakrishnan
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Ashish Kumar Singh
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Simran Singh
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Debasis Das
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore, Karnataka, India.
| |
Collapse
|
13
|
Kaur H, Kaur A, Soni SK, Rishi P. Microbially-derived cocktail of carbohydrases as an anti-biofouling agents: a 'green approach'. BIOFOULING 2022; 38:455-481. [PMID: 35673761 DOI: 10.1080/08927014.2022.2085566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 06/15/2023]
Abstract
Enzymes, also known as biocatalysts, display vital properties like high substrate specificity, an eco-friendly nature, low energy inputs, and cost-effectiveness. Among their numerous known applications, enzymes that can target biofilms or their components are increasingly being investigated for their anti-biofouling action, particularly in healthcare, food manufacturing units and environmental applications. Enzymes can target biofilms at different levels like during the attachment of microorganisms, formation of exopolymeric substances (EPS), and their disruption thereafter. In this regard, a consortium of carbohydrases that can target heterogeneous polysaccharides present in the EPS matrix may provide an effective alternative to conventional chemical anti-biofouling methods. Further, for complete annihilation of biofilms, enzymes can be used alone or in conjunction with other antimicrobial agents. Enzymes hold the promise to replace the conventional methods with greener, more economical, and more efficient alternatives. The present article explores the potential and future perspectives of using carbohydrases as effective anti-biofilm agents.
Collapse
Affiliation(s)
- Harmanpreet Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | - Arashdeep Kaur
- Department of Microbiology, Panjab University, Chandigarh, India
| | | | - Praveen Rishi
- Department of Microbiology, Panjab University, Chandigarh, India
| |
Collapse
|
14
|
Complete Genome Sequence of Aggregatibacter actinomycetemcomitans Strain CU1000N. Microbiol Resour Announc 2022; 11:e0104221. [PMID: 35254109 PMCID: PMC9022580 DOI: 10.1128/mra.01042-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Here, we report the complete genome sequence of Aggregatibacter actinomycetemcomitans strain CU1000N. This rough strain is used extensively as a model organism to characterize localized aggressive periodontitis pathogenesis, the basic biology and oral cavity colonization of A. actinomycetemcomitans, and its interactions with other members of the oral microbiome.
Collapse
|
15
|
Redman WK, Welch GS, Williams AC, Damron AJ, Northcut WO, Rumbaugh KP. Efficacy and safety of biofilm dispersal by glycoside hydrolases in wounds. Biofilm 2021; 3:100061. [PMID: 34825176 PMCID: PMC8605310 DOI: 10.1016/j.bioflm.2021.100061] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/20/2022] Open
Abstract
Novel anti-biofilm and dispersal agents are currently being investigated in an attempt to combat biofilm-associated wound infections. Glycoside hydrolases (GHs) are enzymes that hydrolyze the glycosidic bonds between sugars, such as those found within the exopolysaccharides of the biofilm matrix. Previous studies have shown that GHs can weaken the matrix, inducing bacterial dispersal, and improving antibiotic clearance. Yet, the number of GH enzymes that have been examined for potential therapeutic effects is limited. In this study, we screened sixteen GHs for their ability to disperse mono-microbial and polymicrobial biofilms grown in different environments. Six GHs, α-amylase (source: A. oryzae), alginate lyase (source: various algae), pectinase (source: Rhizopus sp.), amyloglucosidase (source: A. niger), inulinase (source: A. niger), and xylanase (source: A. oryzae), exhibited the highest dispersal efficacy in vitro. Two GHs, α-amylase (source: Bacillus sp.) and cellulase (source: A. niger), used in conjunction with meropenem demonstrated infection clearing ability in a mouse wound model. GHs were also effective in improving antibiotic clearance in diabetic mice. To examine their safety, we screened the GHs for toxicity in cell culture. Overall, there was an inverse relationship between enzyme exposure time and cellular toxicity, with twelve out of sixteen GHs demonstrating some level of toxicity in cell culture. However, only one GH exhibited harmful effects in mice. These results further support the ability of GHs to improve antibiotic clearance of biofilm-associated infections and help lay a foundation for establishing GHs as therapeutic agents for chronic wound infections.
Collapse
Affiliation(s)
- Whitni K Redman
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Garrett S Welch
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,TTUHSC Surgery Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Avery C Williams
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Addyson J Damron
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | | | - Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,TTUHSC Surgery Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
16
|
Disrupting Irreversible Bacterial Adhesion and Biofilm Formation with an Engineered Enzyme. Appl Environ Microbiol 2021; 87:e0026521. [PMID: 33893112 DOI: 10.1128/aem.00265-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Biofilm formation is often attributed to postharvest bacterial persistence on fresh produce and food handling surfaces. In this study, a predicted glycosyl hydrolase enzyme was expressed, purified, and validated for the removal of microbial biofilms from biotic and abiotic surfaces under conditions used for chemical cleaning agents. Crystal violet biofilm staining assays revealed that 0.1 mg/ml of enzyme inhibited up to 41% of biofilm formation by Escherichia coli O157:H7, E. coli 25922, Salmonella enterica serovar Typhimurium, and Listeria monocytogenes. Furthermore, the enzyme was effective at removing mature biofilms, providing a 35% improvement over rinsing with a saline solution alone. Additionally, a parallel-plate flow cell was used to directly observe and quantify the impact of enzyme rinses on E. coli O157:H7 cells adhering to spinach leaf surfaces. The presence of 1 mg/liter enzyme resulted in nearly 6-times-higher detachment rate coefficients than a deionized (DI) water rinse, while the total cells removed from the surface increased from 10% to 25% over the 30-min rinse time, reversing the initial phases of biofilm formation. Enzyme treatment of all 4 cell types resulted in significantly reduced cell surface hydrophobicity and collapse of negatively stained E. coli 25922 cells imaged by electron microscopy, suggesting potential polysaccharide surface modification of enzyme-treated bacteria. Collectively, these results point to the broad substrate specificity and robustness of the enzyme for different types of biofilm stages, solution conditions, and pathogen biofilm types and may be useful as a method for the removal or inhibition of bacterial biofilm formation. IMPORTANCE In this study, the ability of an engineered enzyme to reduce bacterial adhesion and biofilm formation of several foodborne pathogens was demonstrated, representing a promising option for enhancing or replacing chlorine and other chemical sanitizers in food processing applications. Specifically, significant reductions of biofilms of the pathogens Escherichia coli O157:H7, Salmonella Typhimurium, and Listeria monocytogenes are observed, as are reductions in initial adhesion. Enzymes have the added benefits of being green, sustainable alternatives to chemical sanitizers, as well as having a minimal impact on food properties, in contrast to many alternative antimicrobial options such as bleach that aim to minimize food safety risks.
Collapse
|
17
|
Srinivasan R, Santhakumari S, Poonguzhali P, Geetha M, Dyavaiah M, Xiangmin L. Bacterial Biofilm Inhibition: A Focused Review on Recent Therapeutic Strategies for Combating the Biofilm Mediated Infections. Front Microbiol 2021; 12:676458. [PMID: 34054785 PMCID: PMC8149761 DOI: 10.3389/fmicb.2021.676458] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 04/14/2021] [Indexed: 12/31/2022] Open
Abstract
Biofilm formation is a major concern in various sectors and cause severe problems to public health, medicine, and industry. Bacterial biofilm formation is a major persistent threat, as it increases morbidity and mortality, thereby imposing heavy economic pressure on the healthcare sector. Bacterial biofilms also strengthen biofouling, affecting shipping functions, and the offshore industries in their natural environment. Besides, they accomplish harsh roles in the corrosion of pipelines in industries. At biofilm state, bacterial pathogens are significantly resistant to external attack like antibiotics, chemicals, disinfectants, etc. Within a cell, they are insensitive to drugs and host immune responses. The development of intact biofilms is very critical for the spreading and persistence of bacterial infections in the host. Further, bacteria form biofilms on every probable substratum, and their infections have been found in plants, livestock, and humans. The advent of novel strategies for treating and preventing biofilm formation has gained a great deal of attention. To prevent the development of resistant mutants, a feasible technique that may target adhesive properties without affecting the bacterial vitality is needed. This stimulated research is a rapidly growing field for applicable control measures to prevent biofilm formation. Therefore, this review discusses the current understanding of antibiotic resistance mechanisms in bacterial biofilm and intensely emphasized the novel therapeutic strategies for combating biofilm mediated infections. The forthcoming experimental studies will focus on these recent therapeutic strategies that may lead to the development of effective biofilm inhibitors than conventional treatments.
Collapse
Affiliation(s)
- Ramanathan Srinivasan
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, School of Life Sciences, Fujian Agriculture and Forestry University, Fujian, China.,Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fujian, China
| | - Sivasubramanian Santhakumari
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | | | - Mani Geetha
- PG Research and Department of Microbiology, St. Joseph's College of Arts and Science (Autonomous), Tamil Nadu, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Lin Xiangmin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, School of Life Sciences, Fujian Agriculture and Forestry University, Fujian, China.,Key Laboratory of Crop Ecology and Molecular Physiology (Fujian Agriculture and Forestry University), Fujian Province University, Fujian, China.,Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fujian, China
| |
Collapse
|
18
|
Abstract
Introduction: As a result of progress in medical care, a huge number of medical devices are used in the treatment of human diseases. In turn, biofilm-related infection has become a growing threat due to the tolerance of biofilms to antimicrobials, a problem magnified by the development of antimicrobial resistance worldwide. As a result, successful treatment of biofilm-disease using only antimicrobials is problematic.Areas covered: We summarize some alternative approaches to classic antimicrobials for the treatment of biofilm disease. This review is not intended to be exhaustive but to give a clinical picture of alternatives to antimicrobial agents to manage biofilm disease. We highlight those strategies that may be closer to application in clinical practice.Expert opinion: There are a number of outstanding challenges in the development of novel antibiofilm therapies. Screening for effective antibiofilm compounds requires models relevant to all clinical scenarios. Although in vitro research of anti-biofilm strategies has progressed significantly over the past decade, there is a lack of in vivo research. In addition, the complexity of biofilm biology makes it difficult to develop a compound that is likely to provide the single 'magic bullet'. The multifaceted nature of biofilms imposes the need for multi-targeted or combinatorial therapies.
Collapse
Affiliation(s)
- Jose L Del Pozo
- Infectious Diseases Division, Clínica Universidad De Navarra, Pamplona, Spain.,Department of Microbiology, Clínica Universidad De Navarra, Pamplona, Spain.,Laboratory of Microbial Biofilms, Clínica Universidad De Navarra, Pamplona, Spain
| |
Collapse
|
19
|
The Inhibitory Effects of Ficin on Streptococcus mutans Biofilm Formation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6692328. [PMID: 33860052 PMCID: PMC8009705 DOI: 10.1155/2021/6692328] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 02/15/2021] [Accepted: 03/16/2021] [Indexed: 02/05/2023]
Abstract
To investigate the effects of ficin on biofilm formation of conditionally cariogenic Streptococcus mutans (S. mutans). Biomass and metabolic activity of biofilm were assessed using crystal violet assay, colony-forming unit (CFU) counting, and MTT assay. Extracellular polysaccharide (EPS) synthesis was displayed by SEM imaging, bacteria/EPS staining, and anthrone method while acid production was revealed by lactic acid assay. Growth curve and live/dead bacterial staining were conducted to monitor bacterial growth state in both planktonic and biofilm form. Total protein and extracellular proteins of S. mutans biofilm were analyzed by protein/bacterial staining and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), severally. qRT-PCR was conducted to detect acid production, acid tolerance, and biofilm formation associated genes. Crystal violet assay, CFU counting, and MTT assay showed that the suppression effect of ficin on S. mutans biofilm formation was concentration dependent. 4 mg/mL ficin had significant inhibitory effect on S. mutans biofilm formation including biomass, metabolic activity, EPS synthesis, and lactic acid production (p < 0.05). The growth curves from 0 mg/mL to 4 mg/mL ficin were aligned with each other. There was no significant difference among different ficin groups in terms of live/dead bacterial staining result (p > 0.05). Protein/bacterial staining outcome indicated that ficin inhibit both total protein and biofilm formation during the biofilm development. There were more relatively small molecular weight protein bands in extracellular proteins of 4 mg/mL ficin group when compared with the control. Generally, ficin could inhibit biofilm formation and reduce cariogenic virulence of S. mutans effectively in vitro; thus, it could be a potential anticaries agent.
Collapse
|
20
|
Matthes R, Jablonowski L, Holtfreter B, Pink C, Kocher T. Enzymatic biofilm destabilisation to support mechanical cleansing of inserted dental implant surfaces: an in-vitro pilot study. Odontology 2021; 109:780-791. [PMID: 33740161 PMCID: PMC8387265 DOI: 10.1007/s10266-021-00599-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/27/2021] [Indexed: 12/14/2022]
Abstract
Peri-implantitis is caused by microbial contamination and biofilm formation on the implant surface. To achieve re-osseointegration, the microbes must be completely removed from the surface. Adjunctive to mechanical cleaning, chemical treatment with enzymes or other substances could optimise the treatment outcome. Therefore, we investigated the efficacy of different enzymes, a surfactant, and a chelator in destabilising dental polymicrobial biofilm. The biofilm destabilising effect of the glycosidases α-amylase, dextranase, DispersinB®, and lysozyme, as well as the proteinase subtilisin A, and the nuclease Benzonase®, the chelator EDTA, and the surfactant cocamidopropyl betaine were investigated on biofilms, inoculated with plaque on rough titanium discs. The test and the control solutions were incubated for 15 min at 36 °C on biofilms, and loosened biofilm mass was removed by shear stress with a shaker. Fluorescence-stained biofilms were microscopically analysed. Acceptable cell tolerability concentrations of test substances were determined by the MTT (tetrazolium dye) assay on the MG-63 cell line. A statistically significant biofilm destabilising effect of 10% was shown with lysozyme (2500 µg/ml).
Collapse
Affiliation(s)
- Rutger Matthes
- Department of Restorative Dentistry, Periodontology, Endodontology, Preventive and Pediatric Dentistry, Dental School, University Medicine, Greifswald Rotgerberstr. 8, 17475, Greifswald, Germany.
| | - Lukasz Jablonowski
- Department of Restorative Dentistry, Periodontology, Endodontology, Preventive and Pediatric Dentistry, Dental School, University Medicine, Greifswald Rotgerberstr. 8, 17475, Greifswald, Germany
| | - Birte Holtfreter
- Department of Restorative Dentistry, Periodontology, Endodontology, Preventive and Pediatric Dentistry, Dental School, University Medicine, Greifswald Rotgerberstr. 8, 17475, Greifswald, Germany
| | - Christiane Pink
- Department of Restorative Dentistry, Periodontology, Endodontology, Preventive and Pediatric Dentistry, Dental School, University Medicine, Greifswald Rotgerberstr. 8, 17475, Greifswald, Germany
| | - Thomas Kocher
- Department of Restorative Dentistry, Periodontology, Endodontology, Preventive and Pediatric Dentistry, Dental School, University Medicine, Greifswald Rotgerberstr. 8, 17475, Greifswald, Germany
| |
Collapse
|
21
|
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: 195] [Impact Index Per Article: 48.8] [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
|
22
|
|
23
|
Koubali H, Latrache H, Zahir H, El Louali M. Kinetics of Adhesion
Staphylococcus aureus
on Glass in the Presence of Sodium Lauryl Sulfate. J SURFACTANTS DETERG 2020. [DOI: 10.1002/jsde.12484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Hajar Koubali
- Laboratory of Bioprocess and Bio‐interfaces, Faculty of Sciences and Technics Sultan Moulay Slimane University B.P. 523 Beni Mellal 23000 Morocco
| | - Hassan Latrache
- Laboratory of Bioprocess and Bio‐interfaces, Faculty of Sciences and Technics Sultan Moulay Slimane University B.P. 523 Beni Mellal 23000 Morocco
| | - Hafida Zahir
- Laboratory of Bioprocess and Bio‐interfaces, Faculty of Sciences and Technics Sultan Moulay Slimane University B.P. 523 Beni Mellal 23000 Morocco
| | - Mostafa El Louali
- Laboratory of Bioprocess and Bio‐interfaces, Faculty of Sciences and Technics Sultan Moulay Slimane University B.P. 523 Beni Mellal 23000 Morocco
| |
Collapse
|
24
|
Wille J, Coenye T. Biofilm dispersion: The key to biofilm eradication or opening Pandora's box? Biofilm 2020; 2:100027. [PMID: 33447812 PMCID: PMC7798462 DOI: 10.1016/j.bioflm.2020.100027] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 05/28/2020] [Accepted: 05/28/2020] [Indexed: 12/16/2022] Open
Abstract
Biofilms are extremely difficult to eradicate due to their decreased antibiotic susceptibility. Inducing biofilm dispersion could be a potential strategy to help combat biofilm-related infections. Mechanisms of biofilm dispersion can basically be divided into two groups, i.e. active and passive dispersion. Active dispersion depends on a decrease in the intracellular c-di-GMP levels, leading to the production of enzymes that degrade the biofilm matrix and promote dispersion. In contrast, passive dispersion relies on triggers that directly release cells from the biofilm. In the present review, several active and passive dispersion strategies are discussed. In addition, the disadvantages and possible consequences of using dispersion as a treatment approach for biofilm-related infections are also reviewed.
Collapse
Affiliation(s)
- Jasper Wille
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Hasegawa T, Takenaka S, Oda M, Domon H, Hiyoshi T, Sasagawa K, Ohsumi T, Hayashi N, Okamoto Y, Yamamoto H, Ohshima H, Terao Y, Noiri Y. Sulfated vizantin causes detachment of biofilms composed mainly of the genus Streptococcus without affecting bacterial growth and viability. BMC Microbiol 2020; 20:361. [PMID: 33238885 PMCID: PMC7687742 DOI: 10.1186/s12866-020-02033-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/03/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sulfated vizantin, a recently developed immunostimulant, has also been found to exert antibiofilm properties. It acts not as a bactericide, but as a detachment-promoting agent by reducing the biofilm structural stability. This study aimed to investigate the mechanism underlying this activity and its species specificity using two distinct ex vivo oral biofilm models derived from human saliva. RESULTS The biofilm, composed mainly of the genus Streptococcus and containing 50 μM of sulfated vizantin, detached significantly from its basal surface with rotation at 500 rpm for only 15 s, even when 0.2% sucrose was supplied. Expression analyses for genes associated with biofilm formation and bacterial adhesion following identification of the Streptococcus species, revealed that a variety of Streptococcus species in a cariogenic biofilm showed downregulation of genes encoding glucosyltransferases involved in the biosynthesis of water-soluble glucan. The expression of some genes encoding surface proteins was also downregulated. Of the two quorum sensing systems involved in the genus Streptococcus, the expression of luxS in three species, Streptococcus oralis, Streptococcus gordonii, and Streptococcus mutans, was significantly downregulated in the presence of 50 μM sulfated vizantin. Biofilm detachment may be facilitated by the reduced structural stability due to these modulations. As a non-specific reaction, 50 μM sulfated vizantin decreased cell surface hydrophobicity by binding to the cell surface, resulting in reduced bacterial adherence. CONCLUSION Sulfated vizantin may be a candidate for a new antibiofilm strategy targeting the biofilm matrix while preserving the resident microflora.
Collapse
Affiliation(s)
- Taisuke Hasegawa
- Division of Cariology, Operative Dentistry and Endodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274, Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Shoji Takenaka
- Division of Cariology, Operative Dentistry and Endodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274, Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan.
| | - Masataka Oda
- Department of Microbiology and Infection Control Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan.,Division of Periodontology, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan
| | - Karin Sasagawa
- Division of Microbiology and Infectious Diseases, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan.,Division of Periodontology, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan
| | - Tatsuya Ohsumi
- Division of Cariology, Operative Dentistry and Endodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274, Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| | - Naoki Hayashi
- Department of Microbiology and Infection Control Science, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Yasuko Okamoto
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Hirofumi Yamamoto
- Department of Chemistry and Functional Molecule, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Japan
| | - Hayato Ohshima
- Division of Anatomy and Cell Biology of Hard Tissue, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Faculty of Dentistry & Graduate School of Medical and Dental sciences, Niigata University, Niigata, Japan
| | - Yuichiro Noiri
- Division of Cariology, Operative Dentistry and Endodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, 2-5274, Gakkocho-dori, Chuo-ku, Niigata, 951-8514, Japan
| |
Collapse
|
26
|
Abstract
The formation of microbial biofilms enables single planktonic cells to assume a multicellular mode of growth. During dispersion, the final step of the biofilm life cycle, single cells egress from the biofilm to resume a planktonic lifestyle. As the planktonic state is considered to be more vulnerable to antimicrobial agents and immune responses, dispersion is being considered a promising avenue for biofilm control. In this Review, we discuss conditions that lead to dispersion and the mechanisms by which native and environmental cues contribute to dispersion. We also explore recent findings on the role of matrix degradation in the dispersion process, and the distinct phenotype of dispersed cells. Last, we discuss the translational and therapeutic potential of dispersing bacteria during infection.
Collapse
Affiliation(s)
- Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Department of the TTUHSC Surgery Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Karin Sauer
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA.
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA.
| |
Collapse
|
27
|
Fleming D, Redman W, Welch GS, Mdluli NV, Rouchon CN, Frank KL, Rumbaugh KP. Utilizing glycoside hydrolases to improve the quantitation and visualization of biofilm bacteria. Biofilm 2020; 2:100037. [PMID: 33447822 PMCID: PMC7798457 DOI: 10.1016/j.bioflm.2020.100037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 01/06/2023] Open
Abstract
The complexity of microbial biofilms offers several challenges to the use of traditional means of microbial research. In particular, it can be difficult to calculate accurate numbers of biofilm bacteria, because even after thorough homogenization or sonication, small pieces of the biofilm remain, which contain numerous bacterial cells and result in inaccurately low colony forming units (CFU). In addition, imaging of infected tissue ex vivo often results in a disparity between the CFU and the number of bacterial cells observed under the microscope. We hypothesized that this phenomenon is due to the biofilm extracellular polymeric substance decreasing the accessibility of stains and antibodies to the embedded bacterial cells. In this study, we describe incorporating EPS-degrading glycoside hydrolases for CFU determination to obtain a more accurate estimation of the viable cells and for immunohistochemistry to disrupt the biofilm matrix and increase primary antibody binding to the bacterial cells.
Collapse
Affiliation(s)
- Derek Fleming
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Whitni Redman
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Garrett S Welch
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Nontokozo V Mdluli
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Candace N Rouchon
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kristi L Frank
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kendra P Rumbaugh
- Departments of Surgery, Immunology and Molecular Microbiology, and the Burn Center of Research Excellence, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
28
|
Jiang Y, Geng M, Bai L. Targeting Biofilms Therapy: Current Research Strategies and Development Hurdles. Microorganisms 2020; 8:microorganisms8081222. [PMID: 32796745 PMCID: PMC7465149 DOI: 10.3390/microorganisms8081222] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 01/05/2023] Open
Abstract
Biofilms are aggregate of microorganisms in which cells are frequently embedded within a self-produced matrix of extracellular polymeric substance (EPS) and adhere to each other and/or to a surface. The development of biofilm affords pathogens significantly increased tolerances to antibiotics and antimicrobials. Up to 80% of human bacterial infections are biofilm-associated. Dispersal of biofilms can turn microbial cells into their more vulnerable planktonic phenotype and improve the therapeutic effect of antimicrobials. In this review, we focus on multiple therapeutic strategies that are currently being developed to target important structural and functional characteristics and drug resistance mechanisms of biofilms. We thoroughly discuss the current biofilm targeting strategies from four major aspects—targeting EPS, dispersal molecules, targeting quorum sensing, and targeting dormant cells. We explain each aspect with examples and discuss the main hurdles in the development of biofilm dispersal agents in order to provide a rationale for multi-targeted therapy strategies that target the complicated biofilms. Biofilm dispersal is a promising research direction to treat biofilm-associated infections in the future, and more in vivo experiments should be performed to ensure the efficacy of these therapeutic agents before being used in clinic.
Collapse
|
29
|
Piarali S, Marlinghaus L, Viebahn R, Lewis H, Ryadnov MG, Groll J, Salber J, Roy I. Activated Polyhydroxyalkanoate Meshes Prevent Bacterial Adhesion and Biofilm Development in Regenerative Medicine Applications. Front Bioeng Biotechnol 2020; 8:442. [PMID: 32671021 PMCID: PMC7326089 DOI: 10.3389/fbioe.2020.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
Regenerative medicine has become an extremely valuable tool offering an alternative to conventional therapies for the repair and regeneration of tissues. The re-establishment of tissue and organ functions can be carried out by tissue engineering strategies or by using medical devices such as implants. However, with any material being implanted inside the human body, one of the conundrums that remains is the ease with which these materials can get contaminated by bacteria. Bacterial adhesion leads to the formation of mature, alive and complex three-dimensional biofilm structures, further infection of surrounding tissues and consequent development of complicated chronic infections. Hence, novel tissue engineering strategies delivering biofilm-targeted therapies, while at the same time allowing tissue formation are highly relevant. In this study our aim was to develop surface modified polyhydroxyalkanoate-based fiber meshes with enhanced bacterial anti-adhesive and juvenile biofilm disrupting properties for tissue regeneration purposes. Using reactive and amphiphilic star-shaped macromolecules as an additive to a polyhydroxyalkanoate spinning solution, a synthetic antimicrobial peptide, Amhelin, with strong bactericidal and anti-biofilm properties, and Dispersin B, an enzyme promoting the disruption of exopolysaccharides found in the biofilm matrix, were covalently conjugated to the fibers by addition to the solution before the spinning process. Staphylococcus epidermidis is one of the most problematic pathogens responsible for tissue-related infections. The initial antibacterial screening showed that Amhelin proved to be strongly bactericidal at 12 μg/ml and caused >50% reductions of biofilm formation at 6 μg/ml, while Dispersin B was found to disperse >70% of pre-formed biofilms at 3 μg/ml. Regarding the cytotoxicity of the agents toward L929 murine fibroblasts, a CC50 of 140 and 115 μg/ml was measured for Amhelin and Dispersin B, respectively. Optimization of the electrospinning process resulted in aligned fibers. Surface activated fibers with Amhelin and Dispersin B resulted in 83% reduction of adhered bacteria on the surface of the fibers. Additionally, the materials developed were found to be cytocompatible toward L929 murine fibroblasts. The strategy reported in this preliminary study suggests an alternative approach to prevent bacterial adhesion and, in turn biofilm formation, in materials used in regenerative medicine applications such as tissue engineering.
Collapse
Affiliation(s)
- Sheila Piarali
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr-University, Bochum, Germany
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | | | - Richard Viebahn
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr-University, Bochum, Germany
| | - Helen Lewis
- National Physical Laboratory, Teddington, United Kingdom
| | | | - Jürgen Groll
- Department for Functional Materials in Medicine and Dentistry and Bavarian Polymer Institute, University of Würzburg, Würzburg, Germany
| | - Jochen Salber
- Department of Surgery, Universitätsklinikum Knappschaftskrankenhaus Bochum, Ruhr-University, Bochum, Germany
| | - Ipsita Roy
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
30
|
Dey P, Parai D, Banerjee M, Hossain ST, Mukherjee SK. Naringin sensitizes the antibiofilm effect of ciprofloxacin and tetracycline against Pseudomonas aeruginosa biofilm. Int J Med Microbiol 2020; 310:151410. [PMID: 32057619 DOI: 10.1016/j.ijmm.2020.151410] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 02/03/2023] Open
Abstract
The study aims to explore the combinatorial effect of naringin with antibiotics, ciprofloxacin and tetracycline on Pseudomonas aeruginosa biofilms. The antibiofilm efficacy of selected treatment regimes against P. aeruginosa biofilm were quantified by crystal violet assay, MTT assay, Congo red binding assay, and were visualized by confocal laser scanning microscopy and scanning electron microscopy. All the assays reflected antibiofilm activities, however, combinatorial performances of naringin with antibiotics were found to be more significant. A significant reduction in swimming and swarming motilities along with pellicle formation and altered colony morphology were observed as a result of combinatorial effect. The cytotoxicity of naringin and its antibiotic combinations was assayed on murine macrophage cell line. The applicability of such combinations was tested for their relative eradication against pre-formed biofilm on urinary catheter surface. This finding indicated that naringin potentiates the efficacy of both ciprofloxacin and tetracycline on P. aeruginosa biofilm in comparison to their solo treatment. The finding would help to open hitherto unexplored possibilities of establishing naringin as a potential antibiofilm agent and suggest on the possibility of its use in drug-herb combinations for managing biofilm-associated bacterial infections.
Collapse
Affiliation(s)
- Pia Dey
- Department of Microbiology, University of Kalyani, Kalyani, WB, India
| | - Debaprasad Parai
- Department of Microbiology, University of Kalyani, Kalyani, WB, India
| | - Malabika Banerjee
- Department of Microbiology, University of Kalyani, Kalyani, WB, India
| | | | | |
Collapse
|
31
|
Ramos Y, Rocha J, Hael AL, van Gestel J, Vlamakis H, Cywes-Bentley C, Cubillos-Ruiz JR, Pier GB, Gilmore MS, Kolter R, Morales DK. PolyGlcNAc-containing exopolymers enable surface penetration by non-motile Enterococcus faecalis. PLoS Pathog 2019; 15:e1007571. [PMID: 30742693 PMCID: PMC6386517 DOI: 10.1371/journal.ppat.1007571] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 02/22/2019] [Accepted: 01/08/2019] [Indexed: 11/20/2022] Open
Abstract
Bacterial pathogens have evolved strategies that enable them to invade tissues and spread within the host. Enterococcus faecalis is a leading cause of local and disseminated multidrug-resistant hospital infections, but the molecular mechanisms used by this non-motile bacterium to penetrate surfaces and translocate through tissues remain largely unexplored. Here we present experimental evidence indicating that E. faecalis generates exopolysaccharides containing β-1,6-linked poly-N-acetylglucosamine (polyGlcNAc) as a mechanism to successfully penetrate semisolid surfaces and translocate through human epithelial cell monolayers. Genetic screening and molecular analyses of mutant strains identified glnA, rpiA and epaX as genes critically required for optimal E. faecalis penetration and translocation. Mechanistically, GlnA and RpiA cooperated to generate uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) that was utilized by EpaX to synthesize polyGlcNAc-containing polymers. Notably, exogenous supplementation with polymeric N-acetylglucosamine (PNAG) restored surface penetration by E. faecalis mutants devoid of EpaX. Our study uncovers an unexpected mechanism whereby the RpiA-GlnA-EpaX metabolic axis enables production of polyGlcNAc-containing polysaccharides that endow E. faecalis with the ability to penetrate surfaces. Hence, targeting carbohydrate metabolism or inhibiting biosynthesis of polyGlcNAc-containing exopolymers may represent a new strategy to more effectively confront enterococcal infections in the clinic. Enterococcus faecalis is a microbial inhabitant of the human gastrointestinal tract that can cause lethal infections. Typically classified as a non-motile bacterium, E. faecalis can readily migrate and translocate across epithelial barriers to invade distant organs. Nevertheless, the molecular pathways driving enterococcal invasive attributes remain poorly understood. In this study, we uncover that E. faecalis produces a polyGlcNAc-containing extracellular glycopolymer to efficiently migrate into semisolid surfaces and translocate through human epithelial cell monolayers. Our work provides evidence that non-motile bacterial pathogens can exploit endogenous carbohydrate metabolic pathways to penetrate surfaces. Thus, targeting glycopolymer biosynthetic programs might be useful to control infections by Gram-positive cocci in the clinic.
Collapse
Affiliation(s)
- Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States of America
| | - Jorge Rocha
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
| | - Ana L. Hael
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
| | - Jordi van Gestel
- Department of Evolutionary Biology and Environmental Studies, University of Zürich, Zürich, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (Eawag), Dübendorf, Switzerland
- Department of Environmental Systems Science, ETH Zürich, Zürich, Switzerland
| | - Hera Vlamakis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
| | - Colette Cywes-Bentley
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Juan R. Cubillos-Ruiz
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States of America
| | - Gerald B. Pier
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Michael S. Gilmore
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
- Department of Ophthalmology, Harvard Medical School, Boston, MA, United States of America
| | - Roberto Kolter
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, United States of America
| | - Diana K. Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
This review provides a comprehensive summary of issues associated with treating polyclonal bacterial biofilms in chronic diabetic wounds. We use this as a foundation and discuss the alternatives to conventional antibiotics and the emerging need for suitable drug delivery systems. In recent years, extraordinary advances have been made in the field of nanoparticle synthesis and packaging. However, these systems have not been incorporated into the clinic for treatments other than for cancer or severe genetic diseases. We present a unifying perspective on how the field is evolving and the need for an early amalgamation of engineering principles and a biological understanding of underlying phenomena in order to develop a therapy that is translatable to the clinic in a shorter time.
Collapse
|
33
|
Oral Microbes, Biofilms and Their Role in Periodontal and Peri-Implant Diseases. MATERIALS 2018; 11:ma11101802. [PMID: 30248991 PMCID: PMC6213094 DOI: 10.3390/ma11101802] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 01/03/2023]
Abstract
Despite many discoveries over the past 20 years regarding the etio-pathogenesis of periodontal and peri-implant diseases, as well as significant advances in our understanding of microbial biofilms, the incidence of these pathologies still continues to rise. This review presents a general overview of the main protagonists and phenomena involved in oral health and disease. A special emphasis on the role of certain keystone pathogens in periodontitis and peri-implantitis is underlined. Their capacity to bring a dysregulation of the homeostasis with their host and the microbial biofilm lifestyle are also discussed. Finally, the current treatment principles of periodontitis and peri-implantitis are presented and their limits exposed. This leads to realize that new strategies must be developed and studied to overcome the shortcomings of existing approaches.
Collapse
|
34
|
Nahar S, Mizan MFR, Ha AJW, Ha SD. Advances and Future Prospects of Enzyme-Based Biofilm Prevention Approaches in the Food Industry. Compr Rev Food Sci Food Saf 2018; 17:1484-1502. [DOI: 10.1111/1541-4337.12382] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/22/2018] [Accepted: 06/27/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Shamsun Nahar
- Dept. of Food Science and Technology; Chung-Ang Univ.; Anseong Gyeonggi-Do 456-756 Republic of Korea
| | | | - Angela Jie-won Ha
- Dept. of Food Science and Technology; Chung-Ang Univ.; Anseong Gyeonggi-Do 456-756 Republic of Korea
| | - Sang-Do Ha
- Dept. of Food Science and Technology; Chung-Ang Univ.; Anseong Gyeonggi-Do 456-756 Republic of Korea
| |
Collapse
|
35
|
Bassegoda A, Ivanova K, Ramon E, Tzanov T. Strategies to prevent the occurrence of resistance against antibiotics by using advanced materials. Appl Microbiol Biotechnol 2018; 102:2075-2089. [PMID: 29392390 DOI: 10.1007/s00253-018-8776-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/04/2018] [Accepted: 01/06/2018] [Indexed: 01/26/2023]
Abstract
Drug resistance occurrence is a global healthcare concern responsible for the increased morbidity and mortality in hospitals, time of hospitalisation and huge financial loss. The failure of the most antibiotics to kill "superbugs" poses the urgent need to develop innovative strategies aimed at not only controlling bacterial infection but also the spread of resistance. The prevention of pathogen host invasion by inhibiting bacterial virulence and biofilm formation, and the utilisation of bactericidal agents with different mode of action than classic antibiotics are the two most promising new alternative strategies to overcome antibiotic resistance. Based on these novel approaches, researchers are developing different advanced materials (nanoparticles, hydrogels and surface coatings) with novel antimicrobial properties. In this review, we summarise the recent advances in terms of engineered materials to prevent bacteria-resistant infections according to the antimicrobial strategies underlying their design.
Collapse
Affiliation(s)
- Arnau Bassegoda
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrassa, Spain
| | - Kristina Ivanova
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrassa, Spain
| | - Eva Ramon
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrassa, Spain
| | - Tzanko Tzanov
- Group of Molecular and Industrial Biotechnology, Department of Chemical Engineering, Universitat Politècnica de Catalunya, Terrassa, Spain.
| |
Collapse
|
36
|
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
|
37
|
Mohmmed SA, Vianna ME, Penny MR, Hilton ST, Mordan NJ, Knowles JC. Investigations into in situ Enterococcus faecalis biofilm removal by passive and active sodium hypochlorite irrigation delivered into the lateral canal of a simulated root canal model. Int Endod J 2017; 51:649-662. [PMID: 29178348 DOI: 10.1111/iej.12880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/22/2017] [Indexed: 12/14/2022]
Abstract
AIM To investigate in situ Enterococcus faecalis biofilm removal from the lateral canal of a simulated root canal system using passive or active irrigation protocols. METHODOLOGY Root canal models (n = 43) were manufactured from transparent resin materials using 3D printing. Each canal was created with an 18 mm length, apical size 30, a .06 taper and a lateral canal of 3 mm length, with 0.3 mm diameter. Biofilms were grown in the lateral canal and apical 3 mm of the main canal for 10 days. Three models from each group were examined for residual biofilm using SEM. The other forty models were divided into four groups (n = 10). The models were observed under a fluorescence microscope. Following 60 s of 9 mL of 2.5% NaOCl irrigation using syringe and needle, the irrigant was either left stagnant in the canal or activated using gutta-percha, sonic or ultrasonic methods for 30 s. Images were then captured every second using an external camera. The residual biofilm percentages were measured using image analysis software. The data were analysed using generalized linear mixed models. A significance level of 0.05 was used throughout. RESULTS The greatest level of biofilm removal was obtained with ultrasonic agitation (66.76%) followed by sonic (45.49%), manual agitation (43.97%) and passive irrigation groups (38.67%), respectively. The differences were significant between the residual biofilm in the passive irrigation and both sonic and ultrasonic groups (P = 0.001). CONCLUSION Agitation resulted in better penetration of 2.5% NaOCl into the lateral canal of an artificial root canal model. Ultrasonic agitation of NaOCl improved the removal of biofilm.
Collapse
Affiliation(s)
- S A Mohmmed
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.,Department of Conservative Dentistry, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - M E Vianna
- Department of Learning and Scholarship, School of Dentistry, College of Biomedical and Lifesciences, Cardiff University, Cardiff, UK
| | - M R Penny
- Faculty of Life Sciences, School of Pharmacy, University College London, London, UK
| | - S T Hilton
- Faculty of Life Sciences, School of Pharmacy, University College London, London, UK
| | - N J Mordan
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | - J C Knowles
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.,The Discoveries Centre for Regenerative and Precision Medicine, UCL Campus, London, UK.,Department of Nanobiomedical Science & BK21 Plus NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
| |
Collapse
|
38
|
Francolini I, Vuotto C, Piozzi A, Donelli G. Antifouling and antimicrobial biomaterials: an overview. APMIS 2017; 125:392-417. [PMID: 28407425 DOI: 10.1111/apm.12675] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 01/14/2017] [Indexed: 12/12/2022]
Abstract
The use of implantable medical devices is a common and indispensable part of medical care for both diagnostic and therapeutic purposes. However, as side effect, the implant of medical devices quite often leads to the occurrence of difficult-to-treat infections, as a consequence of the colonization of their abiotic surfaces by biofilm-growing microorganisms increasingly resistant to antimicrobial therapies. A promising strategy to combat device-related infections is based on anti-infective biomaterials that either repel microbes, so they cannot attach to the device surfaces, or kill them in the surrounding areas. In general, such biomaterials are characterized by antifouling coatings, exhibiting low adhesion or even repellent properties towards microorganisms, or antimicrobial coatings, able to kill microbes approaching the surface. In this light, the present overview will address the development in the last two decades of antifouling and antimicrobial biomaterials designed to potentially limit the initial stages of microbial adhesion, as well as the microbial growth and biofilm formation on medical device surfaces.
Collapse
Affiliation(s)
| | - Claudia Vuotto
- Microbial Biofilm Laboratory, IRCCS Fondazione Santa Lucia, Rome
| | | | | |
Collapse
|
39
|
Approaches to Dispersing Medical Biofilms. Microorganisms 2017; 5:microorganisms5020015. [PMID: 28368320 PMCID: PMC5488086 DOI: 10.3390/microorganisms5020015] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 03/22/2017] [Accepted: 03/31/2017] [Indexed: 02/07/2023] Open
Abstract
Biofilm-associated infections pose a complex problem to the medical community, in that residence within the protection of a biofilm affords pathogens greatly increased tolerances to antibiotics and antimicrobials, as well as protection from the host immune response. This results in highly recalcitrant, chronic infections and high rates of morbidity and mortality. Since as much as 80% of human bacterial infections are biofilm-associated, many researchers have begun investigating therapies that specifically target the biofilm architecture, thereby dispersing the microbial cells into their more vulnerable, planktonic mode of life. This review addresses the current state of research into medical biofilm dispersal. We focus on three major classes of dispersal agents: enzymes (including proteases, deoxyribonucleases, and glycoside hydrolases), antibiofilm peptides, and dispersal molecules (including dispersal signals, anti-matrix molecules, and sequestration molecules). Throughout our discussion, we provide detailed lists and summaries of some of the most prominent and extensively researched dispersal agents that have shown promise against the biofilms of clinically relevant pathogens, and we catalog which specific microorganisms they have been shown to be effective against. Lastly, we discuss some of the main hurdles to development of biofilm dispersal agents, and contemplate what needs to be done to overcome them.
Collapse
|
40
|
Glycoside Hydrolases Degrade Polymicrobial Bacterial Biofilms in Wounds. Antimicrob Agents Chemother 2017; 61:AAC.01998-16. [PMID: 27872074 PMCID: PMC5278739 DOI: 10.1128/aac.01998-16] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/15/2016] [Indexed: 01/30/2023] Open
Abstract
The persistent nature of chronic wounds leaves them highly susceptible to invasion by a variety of pathogens that have the ability to construct an extracellular polymeric substance (EPS). This EPS makes the bacterial population, or biofilm, up to 1,000-fold more antibiotic tolerant than planktonic cells and makes wound healing extremely difficult. Thus, compounds which have the ability to degrade biofilms, but not host tissue components, are highly sought after for clinical applications. In this study, we examined the efficacy of two glycoside hydrolases, α-amylase and cellulase, which break down complex polysaccharides, to effectively disrupt Staphylococcus aureus and Pseudomonas aeruginosa monoculture and coculture biofilms. We hypothesized that glycoside hydrolase therapy would significantly reduce EPS biomass and convert bacteria to their planktonic state, leaving them more susceptible to conventional antimicrobials. Treatment of S. aureus and P. aeruginosa biofilms, grown in vitro and in vivo, with solutions of α-amylase and cellulase resulted in significant reductions in biomass, dissolution of the biofilm, and an increase in the effectiveness of subsequent antibiotic treatments. These data suggest that glycoside hydrolase therapy represents a potential safe, effective, and new avenue of treatment for biofilm-related infections.
Collapse
|
41
|
Abstract
Biofilm-associated bacteria are less sensitive to antibiotics than free-living (planktonic) cells. Furthermore, with variations in the concentration of antibiotics throughout a biofilm, microbial cells are often exposed to levels below inhibitory concentrations and may develop resistance. This, as well as the irresponsible use of antibiotics, leads to the selection of pathogens that are difficult to eradicate. The Centers for Disease Control and Prevention use the terms "antibiotic" and "antimicrobial agent" interchangeably. However, a clear distinction between these two terms is required for the purpose of this assessment. Therefore, we define "antibiotics" as pharmaceutically formulated and medically administered substances and "antimicrobials" as a broad category of substances which are not regulated as drugs. This comprehensive minireview evaluates the effect of natural antimicrobials on pathogens in biofilms when used instead of, or in combination with, commonly prescribed antibiotics.
Collapse
|
42
|
Mohmmed SA, Vianna ME, Hilton ST, Boniface DR, Ng YL, Knowles JC. Investigation to test potential stereolithography materials for development of anin vitroroot canal model. Microsc Res Tech 2016; 80:202-210. [DOI: 10.1002/jemt.22788] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 09/06/2016] [Accepted: 09/21/2016] [Indexed: 01/19/2023]
Affiliation(s)
- Saifalarab A. Mohmmed
- Division of Biomaterials and Tissue Engineering; UCL Eastman Dental Institute, University College London; London UK
| | - Morgana E. Vianna
- Department of Learning and Scholarship, School of Dentistry, College of Biomedical and Life Sciences; Cardiff University; Cardiff UK
| | - Stephen T. Hilton
- School of Pharmacy, Faculty of Life Sciences; University College London; London UK
| | - David R. Boniface
- Biostatistics Unit, UCL Eastman Dental institute, University College London; London UK
| | - Yuan-Ling Ng
- Unit of Endodontology, UCL-Eastman Dental Institute, University College London; London United Kingdom
| | - Jonathan C. Knowles
- Division of Biomaterials and Tissue Engineering; UCL Eastman Dental Institute, University College London; London UK
| |
Collapse
|
43
|
A novel experimental approach to investigate the effect of different agitation methods using sodium hypochlorite as an irrigant on the rate of bacterial biofilm removal from the wall of a simulated root canal model. Dent Mater 2016; 32:1289-1300. [DOI: 10.1016/j.dental.2016.07.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/07/2016] [Accepted: 07/20/2016] [Indexed: 11/22/2022]
|
44
|
Experimental Models of Oral Biofilms Developed on Inert Substrates: A Review of the Literature. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7461047. [PMID: 27699173 PMCID: PMC5028824 DOI: 10.1155/2016/7461047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 08/14/2016] [Indexed: 01/25/2023]
Abstract
The oral ecosystem is a very complex environment where more than 700 different bacterial species can be found. Most of them are organized in biofilm on dental and mucosal surfaces. Studying this community is important because a rupture in stability can lead to the preeminence of pathogenic microorganisms, causing dental decay, gingivitis, or periodontitis. The multitude of species complicates biofilm analysis so its reproduction, collection, and counting are very delicate. The development of experimental models of dental biofilms was therefore essential and multiple in vitro designs have emerged, each of them especially adapted to observing biofilm formation of specific bacteria within specific environments. The aim of this review is to analyze oral biofilm models.
Collapse
|
45
|
Ragunath C, DiFranco K, Shanmugam M, Gopal P, Vyas V, Fine DH, Cugini C, Ramasubbu N. Surface display of Aggregatibacter actinomycetemcomitans autotransporter Aae and dispersin B hybrid act as antibiofilm agents. Mol Oral Microbiol 2016; 31:329-39. [PMID: 26280561 PMCID: PMC6118125 DOI: 10.1111/omi.12126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2015] [Indexed: 11/30/2022]
Abstract
Among the various proteins expressed by the periodontopathogen Aggregatibacter actinomycetemcomitans, two proteins play important roles for survival in the oral cavity. The autotransporter Aae facilitates the attachment of the pathogen to oral epithelial cells, which act as a reservoir, while the biofilm-degrading glycoside hydrolase dispersin B facilitates the movement of daughter cells from the mature biofilm to a new site. The objective of this study was to use the potential of these two proteins to control biofilms. To this end, we generated a hybrid construct between the Aae C-terminal translocating domain and dispersin B, and mobilized it into Escherichia coli Rosetta (DE3) pLysS cells. Immunofluorescence analysis of the modified E. coli cells confirmed the presence of dispersin B on the surface. Further, the membrane localization of the displayed dispersin B was confirmed with Western blot analysis. The integrity of the E. coli cells displaying the dispersin B was confirmed through FACS analysis. The hydrolytic activity of the surface-displayed dispersin B was confirmed by using 4-methylumbelliferyl-β-d-glucopyranoside as the substrate. The detachment ability of the dispersin B surface-displaying E. coli cells was shown using Staphylococcus epidermidis and Actinobacillus pleuropneumoniae biofilms in a microtiter assay. We concluded that the Aae β-domain is sufficient to translocate foreign enzymes in the native folded form and that the method of Aae-mediated translocation of surface displayed enzymes might be useful for control of biofilms.
Collapse
Affiliation(s)
| | | | - Mayilvahanan Shanmugam
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| | - Prerna Gopal
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| | - Vishal Vyas
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| | - Daniel H. Fine
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| | - Carla Cugini
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| | - Narayanan Ramasubbu
- Department of Oral Biology, Rutgers School of Dental Medicine, 185 South Orange Ave, Newark NJ 07103, USA
| |
Collapse
|
46
|
Hathroubi S, Mekni MA, Domenico P, Nguyen D, Jacques M. Biofilms: Microbial Shelters Against Antibiotics. Microb Drug Resist 2016; 23:147-156. [PMID: 27214143 DOI: 10.1089/mdr.2016.0087] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Biofilms are communities of aggregated bacterial cells embedded in a self-produced extracellular polymeric matrix. Biofilms are recalcitrant to antibiotic treatment and immune defenses and are implicated in many chronic bacterial and fungal infections. In this review, we provide an overview of the contribution of biofilms to persistent infections resistant to antibiotic treatment, the impact of multispecies biofilms on drug resistance and tolerance, and recent advances in the development of antibiofilm agents. Understanding the mechanisms of antibiotic resistance and tolerance in biofilms is essential for developing new preventive and therapeutic strategies and curbing drug resistance.
Collapse
Affiliation(s)
- Skander Hathroubi
- 1 Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal , Montréal, Canada
| | - Mohamed A Mekni
- 2 The National Bone Marrow Transplant Centre , UR12ES02, Faculty of Medicine, Tunis, Tunisia
| | | | - Dao Nguyen
- 4 Meakins Christie Laboratories, Department of Medicine, Research Institute of the McGill University Health Centre , Montréal, Canada
| | - Mario Jacques
- 1 Department of Pathology and Microbiology, Faculty of Veterinary Medicine, Université de Montréal , Montréal, Canada
| |
Collapse
|
47
|
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
|
48
|
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
|
49
|
Shin JM, Ateia I, Paulus JR, Liu H, Fenno JC, Rickard AH, Kapila YL. Antimicrobial nisin acts against saliva derived multi-species biofilms without cytotoxicity to human oral cells. Front Microbiol 2015; 6:617. [PMID: 26150809 PMCID: PMC4471743 DOI: 10.3389/fmicb.2015.00617] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/03/2015] [Indexed: 02/05/2023] Open
Abstract
Objectives: Nisin is a lantibiotic widely used for the preservation of food and beverages. Recently, investigators have reported that nisin may have clinical applications for treating bacterial infections. The aim of this study was to investigate the effects of ultra pure food grade Nisin ZP (>95% purity) on taxonomically diverse bacteria common to the human oral cavity and saliva derived multi-species oral biofilms, and to discern the toxicity of nisin against human cells relevant to the oral cavity. Methods: The minimum inhibitory concentrations and minimum bactericidal concentrations of taxonomically distinct oral bacteria were determined using agar and broth dilution methods. To assess the effects of nisin on biofilms, two model systems were utilized: a static and a controlled flow microfluidic system. Biofilms were inoculated with pooled human saliva and fed filter-sterilized saliva for 20–22 h at 37°C. Nisin effects on cellular apoptosis and proliferation were evaluated using acridine orange/ethidium bromide fluorescent nuclear staining and lactate dehydrogenase activity assays. Results: Nisin inhibited planktonic growth of oral bacteria at low concentrations (2.5–50 μg/ml). Nisin also retarded development of multi-species biofilms at concentrations ≥1 μg/ml. Specifically, under biofilm model conditions, nisin interfered with biofilm development and reduced biofilm biomass and thickness in a dose-dependent manner. The treatment of pre-formed biofilms with nisin resulted in dose- and time-dependent disruption of the biofilm architecture along with decreased bacterial viability. Human cells relevant to the oral cavity were unaffected by the treatment of nisin at anti-biofilm concentrations and showed no signs of apoptotic changes unless treated with much higher concentrations (>200 μg/ml). Conclusion: This work highlights the potential therapeutic value of high purity food grade nisin to inhibit the growth of oral bacteria and the development of biofilms relevant to oral diseases.
Collapse
Affiliation(s)
- Jae M Shin
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor MI, USA
| | - Islam Ateia
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor MI, USA
| | - Jefrey R Paulus
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor MI, USA
| | - Hongrui Liu
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor MI, USA
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor MI, USA
| | - Alexander H Rickard
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor MI, USA
| | - Yvonne L Kapila
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor MI, USA
| |
Collapse
|
50
|
Bansal S, Harjai K, Chhibber S. Aeromonas punctata derived depolymerase improves susceptibility of Klebsiella pneumoniae biofilm to gentamicin. BMC Microbiol 2015; 15:119. [PMID: 26063052 PMCID: PMC4461996 DOI: 10.1186/s12866-015-0455-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 05/28/2015] [Indexed: 01/28/2023] Open
Abstract
Background To overcome antibiotic resistance in biofilms, enzymes aimed at biofilm dispersal are under investigation. In the present study, applicability of an Aeromonas punctata derived depolymerase capable of degrading the capsular polysaccharide (CPS) of Klebsiella pneumoniae, in disrupting its biofilm and increasing gentamicin efficacy against biofilm was investigated. Results Intact biofilm of K. pneumoniae was recalcitrant to gentamicin due to lack of antibiotic penetration. On the other hand, gentamicin could not act on disrupted biofilm cells due to their presence in clusters. However, when depolymerase (20 units/ml) was used in combination with gentamicin (10 μg/ml), dispersal of CPS matrix by enzyme facilitated gentamicin penetration across biofilm. This resulted in significant reduction (p < 0.05) in bacterial count in intact and disrupted biofilms. Reduction in CPS after treatment with depolymerase was confirmed by confocal microscopy and enzyme linked lectinosorbent assay. Furthermore, to substantiate our study, the efficacy of bacterial depolymerase was compared with a phage borne depolymerase possessing similar application against K. pneumoniae. Although both were used at same concentration i.e. 20 units/ml, but a higher efficacy of bacterial depolymerase particularly against older biofilms was visibly clear over its phage counterpart. This could be explained due to high substrate affinity (indicated by Km value) and high turnover number (indicated by Kcat value) of the bacterial depolymerase (Km = 89.88 μM, Kcat = 285 s−1) over the phage derived one (Km = 150 μM, Kcat = 107 s−1). Conclusion Overall the study indicated that, the A. punctata derived depolymerase possesses antibiofilm potential and improves gentamicin efficacy against K. pneumoniae. Moreover, it can serve as a potential substitute to phage borne depolymerases for treating biofilms formed by K. pneumoniae. Electronic supplementary material The online version of this article (doi:10.1186/s12866-015-0455-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shruti Bansal
- Department of Microbiology, Panjab University, Sector-14, Chandigarh, 160014, India.
| | - Kusum Harjai
- Department of Microbiology, Panjab University, Sector-14, Chandigarh, 160014, India.
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Sector-14, Chandigarh, 160014, India.
| |
Collapse
|