51
|
Inhibitory effects of sodium new houttuyfonate on growth and biofilm formation of Streptococcus mutans. Microb Pathog 2021; 157:104957. [PMID: 34022356 DOI: 10.1016/j.micpath.2021.104957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/14/2021] [Accepted: 05/14/2021] [Indexed: 02/08/2023]
Abstract
The present study aimed to assess the impact of sodium new houttuyfonate (SNH) on growth and biofilm formation of Streptococcus mutans, and the combinatorial effects of SNH with cariostatic agents. The effects of SNH on S. mutans planktonic cultures were assessed by growth curve assay. The effects of SNH on S. mutans biofilm and extracellular polysaccharides (EPS) production were observed via crystal violet (CV) assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, colony-forming unit (CFU) counting assay, scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Quantitative real-time polymerase chain reaction (qPCR) was applied to investigate the regulatory effects of SNH on the expression of virulence genes of S. mutans. Checkerboard microdilution assay was performed to investigate the combinatorial effects of SNH with two common cariostatic agents. SNH acted as an inhibitor on planktonic cell growth, biofilm formation and EPS production of S. mutans. SNH also downregulated the expression of gtfBCD and comDE systems and exhibited synergism with chlorhexidine (CHX). In conclusion, this study indicated a possibility for SNH to become an anticaries agents by its antimicrobial activity and synergistic effects with CHX against S. mutans.
Collapse
|
52
|
Zhang K, Raju C, Zhong W, Pethe K, Gründling A, Chan-Park MB. Cationic Glycosylated Block Co-β-peptide Acts on the Cell Wall of Gram-Positive Bacteria as Anti-biofilm Agents. ACS APPLIED BIO MATERIALS 2021; 4:3749-3761. [PMID: 35006805 DOI: 10.1021/acsabm.0c01241] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antimicrobial resistance is a global threat. In addition to the emergence of resistance to last resort drugs, bacteria escape antibiotics killing by forming complex biofilms. Strategies to tackle antibiotic resistance as well as biofilms are urgently needed. Wall teichoic acid (WTA), a generic anionic glycopolymer present on the cell surface of many Gram-positive bacteria, has been proposed as a possible therapeutic target, but its druggability remains to be demonstrated. Here we report a cationic glycosylated block co-β-peptide that binds to WTA. By doing so, the co-β-peptide not only inhibits biofilm formation, it also disperses preformed biofilms in several Gram-positive bacteria and resensitizes methicillin-resistant Staphylococcus aureus to oxacillin. The cationic block of the co-β-peptide physically interacts with the anionic WTA within the cell envelope, whereas the glycosylated block forms a nonfouling corona around the bacteria. This reduces physical interaction between bacteria-substrate and bacteria-biofilm matrix, leading to biofilm inhibition and dispersal. The WTA-targeting co-β-peptide is a promising lead for the future development of broad-spectrum anti-biofilm strategies against Gram-positive bacteria.
Collapse
Affiliation(s)
- Kaixi Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Centre for Antimicrobial Bioengineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459
| | - Cheerlavancha Raju
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Centre for Antimicrobial Bioengineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459
| | - Wenbin Zhong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Centre for Antimicrobial Bioengineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459
| | - Kevin Pethe
- Centre for Antimicrobial Bioengineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921.,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551
| | - Angelika Gründling
- Faculty of Medicine, Department of Infectious Disease, Imperial College London, Flowers Building London, London SW7 2AZ, United Kingdom
| | - Mary B Chan-Park
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Centre for Antimicrobial Bioengineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459.,Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore 636921
| |
Collapse
|
53
|
Bamunuarachchi NI, Khan F, Kim YM. Inhibition of Virulence Factors and Biofilm Formation of Acinetobacter Baumannii by Naturally-derived and Synthetic Drugs. Curr Drug Targets 2021; 22:734-759. [PMID: 33100201 DOI: 10.2174/1389450121666201023122355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 11/22/2022]
Abstract
Acinetobacter baumannii is a gram-negative, aerobic, non-motile, and pleomorphic bacillus. A. baumannii is also a highly-infectious pathogen causing high mortality and morbidity rates in intensive care units. The discovery of novel agents against A. baumannii infections is urgently needed due to the emergence of drug-resistant A. baumannii strains and the limited number of efficacious antibiotics available for treatment. In addition to the production of several virulence factors, A. baumannii forms biofilms on the host cell surface as well. Formation of biofilms occurs through initial surface attachment, microcolony formation, biofilm maturation, and detachment stages, and is one of the major drug resistance mechanisms employed by A. baumannii. Several studies have previously reported the efficacy of naturally-derived and synthetic compounds as anti- biofilm and anti-virulence agents against A. baumannii. Here, inhibition of biofilm formation and virulence factors of A. baumannii using naturally-derived and synthetic compounds are reviewed.
Collapse
Affiliation(s)
| | - Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan 48513, South Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan 48513, South Korea
| |
Collapse
|
54
|
4-4-(Anilinomethyl)-3-[4-(trifluoromethyl)phenyl]-1H-pyrazol-1-ylbenzoic acid derivatives as potent anti-gram-positive bacterial agents. Eur J Med Chem 2021; 219:113402. [PMID: 33845234 DOI: 10.1016/j.ejmech.2021.113402] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/18/2021] [Accepted: 03/20/2021] [Indexed: 12/11/2022]
Abstract
A collection of potent antimicrobials consisting of novel 1,3-bis-benzoic acid and trifluoromethyl phenyl derived pyrazoles has been synthesized and tested for antibacterial activity. The majority of trifluoromethyl phenyl derivatives are highly potent growth inhibitors of Gram-positive bacteria and showed low toxicity to human cultured cells. In particular, two compounds (59 and 74) were selected for additional studies. These compounds are highly effective against Staphylococcus aureus as shown by a low minimum inhibitory concentration (MIC), a bactericidal effect in time-kill assays, moderate inhibition of biofilm formation as well as biofilm destruction, and a bactericidal effect against stationary phase cells representing non-growing persister cells. Multistep resistance assays showed a very low tendency for S. aureus and Enterococcus faecalis to develop resistance through mutation. Additionally, in vivo mouse model studies showed no harmful effects at doses up to 50 mg/kg using 14 blood plasma organ toxicity markers or TUNEL assay in liver and kidney. Investigations into the mode of action by performing macromolecular synthesis inhibition studies showed a broad range of inhibitory effects, suggesting targets that have a global effect on bacterial cell function.
Collapse
|
55
|
Lyu X, Li C, Zhang J, Wang L, Jiang Q, Shui Y, Chen L, Luo Y, Xu X. A Novel Small Molecule, LCG-N25, Inhibits Oral Streptococcal Biofilm. Front Microbiol 2021; 12:654692. [PMID: 33868212 PMCID: PMC8044806 DOI: 10.3389/fmicb.2021.654692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/05/2021] [Indexed: 02/05/2023] Open
Abstract
Dental caries is a chronic oral infectious disease caused by cariogenic biofilm adhered on the tooth surface. Our previous study demonstrated that a repurposed natural compound napabucasin (NAP) showed good antimicrobial activity against oral streptococcal biofilms. The current study designed a novel small molecule, namely LCG-N25, using NAP as a lead compound, and aimed to investigate its potential as an antimicrobial agent in the control of dental caries. LCG-N25 was designed and synthesized with reference to the structure of NAP. The minimal inhibitory concentrations and the minimal bactericidal concentrations of LCG-N25 against Streptococcus mutans, Streptococcus sanguinis, and Streptococcus gordonii were evaluated by microdilution method. The antimicrobial activity of LCG-N25 was further evaluated by crystal violet staining, colony forming units counting, biofilm metabolism assay, dead/live fluorescent staining, and scanning electron microscopy. The effect of LCG-N25 on the extracellular polysaccharides of biofilms was determined by both anthrone-sulfuric acid method and fluorescent staining. The microbial composition of streptococcal biofilms after LCG-N25 treatment was further visualized and quantified by fluorescence in situ hybridization. Besides, the cytotoxicity of LCG-N25 was evaluated by Cell Counting Kit-8 assay, and repeated exposure of S. mutans to LCG-N25 treatment was performed to assess if this novel compound could induce drug resistance of this cariogenic bacterium. We found that LCG-N25 exhibited a good antibacterial activity, low-cytotoxicity, and did not induce drug resistance of cariogenic S. mutans. These findings suggest that LCG-N25 may represent a promising antimicrobial agent that can be used as an adjuvant to the management of dental caries.
Collapse
Affiliation(s)
- Xiaoying Lyu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chungen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Jin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liang Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qingsong Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yusen Shui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
56
|
Atkinson AJ, Armstrong MD, Eskew JT, Coronell O. 2-Aminoimidazole Reduces Fouling and Improves Membrane Performance. J Memb Sci 2021; 629. [PMID: 34366551 DOI: 10.1016/j.memsci.2021.119262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biofouling is difficult to control and hinders the performance of membranes in all applications but is of particular concern when natural waters are purified. Fouling, via multiple mechanisms (organic-only, biofouling-only, cell-deposition-only, and organic+biofouling), of a commercially available membrane (control) and a corresponding membrane coated with an anti-biofouling 2-aminoimidazole (2-AI membrane) was monitored and characterized during the purification of a natural water. Results show that the amount of bacterial cell deposition and organic fouling was not significantly different between control and 2-AI membranes; however, biofilm formation, concurrent or not with other fouling mechanisms, was significantly inhibited (95-98%, p<0.001) by the 2-AI membrane. The limited biofilm that formed on the 2-AI membrane was weaker (as indicated by the polysaccharide to protein ratio) and thus presumably easier to remove. The conductivity rejection by the 2-AI and control membranes was not significantly different throughout the 75-hour experiments, but the rejection of dissolved organic carbon by biofouled (biofouling-only, cell-deposition-only, and organic+biofouling) 2-AI membranes was statistically higher (10-12%, p=0.003-0.07). When biofouled, the water permeance of the 2-AI membranes decreased significantly less (p<0.05) over 75 hours than that of the control membranes, whether or not other additional types of fouling occurred concurrently. Despite the initially lower water permeances of 2-AI membranes (11% lower on average than controls), the 2-AI membranes outperformed the controls (10-11% higher average water permeance) after biofilm formation occurred. Overall, 2-AI membranes fouled less than controls without detriment to water productivity and solute rejection.
Collapse
Affiliation(s)
- Ariel J Atkinson
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mikayla D Armstrong
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - John T Eskew
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Orlando Coronell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
57
|
Henly EL, Norris K, Rawson K, Zoulias N, Jaques L, Chirila PG, Parkin KL, Kadirvel M, Whiteoak C, Lacey MM, Smith TJ, Forbes S. Impact of long-term quorum sensing inhibition on uropathogenic Escherichia coli. J Antimicrob Chemother 2021; 76:909-919. [PMID: 33406232 DOI: 10.1093/jac/dkaa517] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/16/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Quorum sensing is an extracellular bacterial communication system used in the density-dependent regulation of gene expression and development of biofilms. Biofilm formation has been implicated in the establishment of catheter-associated urinary tract infections and therefore quorum sensing inhibitors (QSIs) have been suggested as anti-biofilm catheter coating agents. The long-term effects of QSIs in uropathogens is, however, not clearly understood. OBJECTIVES We evaluated the effects of repeated exposure to the QSIs cinnamaldehyde, (Z)-4-bromo-5(bromomethylene)-2(5H)-furanone-C30 (furanone-C30) and 4-fluoro-5-hydroxypentane-2,3-dione (F-DPD) on antimicrobial susceptibility, biofilm formation and relative pathogenicity in eight uropathogenic Escherichia coli (UPEC) isolates. METHODS MICs, MBCs and minimum biofilm eradication concentrations and antibiotic susceptibility were determined. Biofilm formation was quantified using crystal violet. Relative pathogenicity was assessed in a Galleria mellonella model. To correlate changes in phenotype to gene expression, transcriptomic profiles were created through RNA sequencing and variant analysis of genomes was performed in strain EC958. RESULTS Cinnamaldehyde and furanone-C30 led to increases in susceptibility in planktonic and biofilm-associated UPEC. Relative pathogenicity increased after cinnamaldehyde exposure (4/8 isolates), decreased after furanone-C30 exposure (6/8 isolates) and varied after F-DPD exposure (one increased and one decreased). A total of 9/96 cases of putative antibiotic cross-resistance were generated. Exposure to cinnamaldehyde or F-DPD reduced expression of genes associated with locomotion, whilst cinnamaldehyde caused an increase in genes encoding fimbrial and afimbrial-like adhesins. Furanone-C30 caused a reduction in genes involved in cellular biosynthetic processes, likely though impaired ribonucleoprotein assembly. CONCLUSIONS The multiple phenotypic adaptations induced during QSI exposure in UPEC should be considered when selecting an anti-infective catheter coating agent.
Collapse
Affiliation(s)
- E L Henly
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - K Norris
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - K Rawson
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - N Zoulias
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - L Jaques
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - P G Chirila
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - K L Parkin
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - M Kadirvel
- Manchester Pharmacy School, University of Manchester, Manchester, UK
| | - C Whiteoak
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - M M Lacey
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - T J Smith
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - S Forbes
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| |
Collapse
|
58
|
Kaizerman-Kane D, Hadar M, Joseph R, Logviniuk D, Zafrani Y, Fridman M, Cohen Y. Design Guidelines for Cationic Pillar[n]arenes that Prevent Biofilm Formation by Gram-Positive Pathogens. ACS Infect Dis 2021; 7:579-585. [PMID: 33657813 PMCID: PMC8041275 DOI: 10.1021/acsinfecdis.0c00662] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
![]()
Bacterial biofilms are a major threat
to human health, causing
persistent infections that lead to millions of fatalities worldwide
every year. Biofilms also cause billions of dollars of damage annually
by interfering with industrial processes. Recently, cationic pillararenes
were found to be potent inhibitors of biofilm formation in Gram-positive
bacteria. To identify the structural features of pillararenes that
result in antibiofilm activity, we evaluated the activity of 16 cationic
pillar[5]arene derivatives including that of the first cationic water-soluble
pillar[5]arene-based rotaxane. Twelve of the derivatives were potent
inhibitors of biofilm formation by Gram-positive pathogens. Structure
activity analyses of our pillararene derivatives indicated that positively
charged head groups are critical for the observed antibiofilm activity.
Although certain changes in the lipophilicity of the substituents
on the positively charged head groups are tolerated, dramatic elevation
in the hydrophobicity of the substituents or an increase in steric
bulk on these positive charges abolishes the antibiofilm activity.
An increase in the overall positive charge from 10 to 20 did not affect
the activity significantly, but pillararenes with 5 positive charges
and 5 long alkyl chains had reduced activity. Surprisingly, the cavity
of the pillar[n]arene is not essential for the observed activity,
although the macrocyclic structure of the pillar[n]arene core, which
facilitates the clustering of the positive charges, appears important.
Interestingly, the compounds found to be efficient inhibitors of biofilm
formation were nonhemolytic at concentrations that are ∼100-fold
of their MBIC50 (the minimal concentration of a compound
at which at least 50% inhibition of biofilm formation was observed
compared to untreated cells). The structure–activity relationship
guidelines established here pave the way for a rational design of
potent cationic pillar[n]arene-based antibiofilm agents.
Collapse
Affiliation(s)
- Dana Kaizerman-Kane
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Maya Hadar
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Roymon Joseph
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Dana Logviniuk
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Yossi Zafrani
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
- Department of Organic Chemistry, Israel Institute for Biological Research, Ness-Ziona 74000, Israel
| | - Micha Fridman
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| | - Yoram Cohen
- School of Chemistry, Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
59
|
Awadh AA, Le Gresley A, Forster-Wilkins G, Kelly AF, Fielder MD. Determination of metabolic activity in planktonic and biofilm cells of Mycoplasma fermentans and Mycoplasma pneumoniae by nuclear magnetic resonance. Sci Rep 2021; 11:5650. [PMID: 33707544 PMCID: PMC7952918 DOI: 10.1038/s41598-021-84326-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023] Open
Abstract
Mycoplasmas are fastidious microorganisms, typically characterised by their restricted metabolism and minimalist genome. Although there is reported evidence that some mycoplasmas can develop biofilms little is known about any differences in metabolism in these organisms between the growth states. A systematic metabolomics approach may help clarify differences associated between planktonic and biofilm associated mycoplasmas. In the current study, the metabolomics of two different mycoplasmas of clinical importance (Mycoplasma pneumoniae and Mycoplasma fermentans) were examined using a novel approach involving nuclear magnetic resonance spectroscopy and principle component analysis. Characterisation of metabolic changes was facilitated through the generation of high-density metabolite data and diffusion-ordered spectroscopy that provided the size and structural information of the molecules under examination. This enabled the discrimination between biofilms and planktonic states for the metabolomic profiles of both organisms. This work identified clear biofilm/planktonic differences in metabolite composition for both clinical mycoplasmas and the outcomes serve to establish a baseline understanding of the changes in metabolism observed in these pathogens in their different growth states. This may offer insight into how these organisms are capable of exploiting and persisting in different niches and so facilitate their survival in the clinical setting.
Collapse
Affiliation(s)
- Ammar A. Awadh
- grid.15538.3a0000 0001 0536 3773School of Life Sciences, Pharmacy and Chemistry, SEC Faculty, Kingston University London, Kingston Upon Thames, UK
| | - Adam Le Gresley
- grid.15538.3a0000 0001 0536 3773School of Life Sciences, Pharmacy and Chemistry, SEC Faculty, Kingston University London, Kingston Upon Thames, UK
| | - Gary Forster-Wilkins
- grid.15538.3a0000 0001 0536 3773School of Life Sciences, Pharmacy and Chemistry, SEC Faculty, Kingston University London, Kingston Upon Thames, UK
| | - Alison F. Kelly
- grid.15538.3a0000 0001 0536 3773School of Life Sciences, Pharmacy and Chemistry, SEC Faculty, Kingston University London, Kingston Upon Thames, UK
| | - Mark D. Fielder
- grid.15538.3a0000 0001 0536 3773School of Life Sciences, Pharmacy and Chemistry, SEC Faculty, Kingston University London, Kingston Upon Thames, UK
| |
Collapse
|
60
|
Jacobs L, Meesters J, Parijs I, Hooyberghs G, Van der Eycken EV, Lories B, Steenackers HP. 2-Aminoimidazoles as potent inhibitors of contaminating brewery biofilms. BIOFOULING 2021; 37:61-77. [PMID: 33573402 DOI: 10.1080/08927014.2021.1874366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
Cleaning and disinfection protocols are not always able to remove biofilm microbes present in breweries, indicating that novel anti-biofilm strategies are needed. The preventive activities of three in-house synthesized members of the 2-aminoimidazole class of anti-biofilm molecules were studied against 17 natural brewery biofilms and benchmarked against 18 known inhibitors. Two 2-aminoimidazoles belonged to the top six inhibitors, which were retested against 12 defined brewery biofilm models. For the three best inhibitors, tannic acid (n° 1), 2-aminoimidazole imi-AAC-5 (n° 2), and baicalein (n° 3), the effect on the microbial metabolic activity was evaluated. Here, the top three inhibitors showed similar effectiveness, with baicalein possessing a slightly higher efficacy. Even though the 2-aminoimidazole was the second-best inhibitor, it showed a lower biocidal activity than tannic acid, making it less prone to resistance evolution. Overall, this study supports the potential of 2-aminoimidazoles as a preventive anti-biofilm strategy.
Collapse
Affiliation(s)
- Lene Jacobs
- Centrum of Microbial and Plant Genetics, KU Leuven, Belgium
| | | | - Ilse Parijs
- Centrum of Microbial and Plant Genetics, KU Leuven, Belgium
| | - Geert Hooyberghs
- Laboratory for Organic and Microwave-Assisted Chemistry - LOMAC, KU Leuven, Belgium
| | - Erik V Van der Eycken
- Laboratory for Organic and Microwave-Assisted Chemistry - LOMAC, KU Leuven, Belgium
- Peoples' Friendship University of Russia (RUDN University), Moscow, Russia
| | - Bram Lories
- Centrum of Microbial and Plant Genetics, KU Leuven, Belgium
| | | |
Collapse
|
61
|
Martins FG, Melo A, Sousa SF. Databases for the study of biofilms: current status and potential applications. BIOFOULING 2021; 37:96-108. [PMID: 33508968 DOI: 10.1080/08927014.2021.1876849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Biofilms play an important role in health, being associated with >80% of all microbial infections in the body and in the development of antibiotic resistance. Research in this field has continuously produced large volumes of data. Being able to handle all this information will be paramount for progress in this field. However, this places a heavy burden on the development of strategies to gather, organize and make this information available in a way that can be readily and effectively used by those requiring it. Lately, efforts towards this goal have been reported, particularly with the development of Quorumpeps, BiofOmics, BaAMPs, QSPpred, dPABBs, aBiofilm and the Biofilms Structural Database. This work reviews these databases and highlights their applicability and potential, while stressing some of the challenges for the coming years in database development and usage brought about by the use of big data and machine learning.
Collapse
Affiliation(s)
- Fábio G Martins
- UCIBIO/REQUIMTE, BioSIM - Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Porto, Portugal
| | - André Melo
- LAQV/REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Porto, Portugal
| | - Sérgio F Sousa
- UCIBIO/REQUIMTE, BioSIM - Departamento de Biomedicina, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| |
Collapse
|
62
|
|
63
|
Melander RJ, Basak AK, Melander C. Natural products as inspiration for the development of bacterial antibiofilm agents. Nat Prod Rep 2020; 37:1454-1477. [PMID: 32608431 PMCID: PMC7677205 DOI: 10.1039/d0np00022a] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Natural products have historically been a rich source of diverse chemical matter with numerous biological activities, and have played an important role in drug discovery in many areas including infectious disease. Synthetic and medicinal chemistry have been, and continue to be, important tools to realize the potential of natural products as therapeutics and as chemical probes. The formation of biofilms by bacteria in an infection setting is a significant factor in the recalcitrance of many bacterial infections, conferring increased tolerance to many antibiotics and to the host immune response, and as yet there are no approved therapeutics for combatting biofilm-based bacterial infections. Small molecules that interfere with the ability of bacteria to form and maintain biofilms can overcome antibiotic tolerance conferred by the biofilm phenotype, and have the potential to form combination therapies with conventional antibiotics. Many natural products with anti-biofilm activity have been identified from plants, microbes, and marine life, including: elligic acid glycosides, hamamelitannin, carolacton, skyllamycins, promysalin, phenazines, bromoageliferin, flustramine C, meridianin D, and brominated furanones. Total synthesis and medicinal chemistry programs have facilitated structure confirmation, identification of critical structural motifs, better understanding of mechanistic pathways, and the development of more potent, more accessible, or more pharmacologically favorable derivatives of anti-biofilm natural products.
Collapse
Affiliation(s)
- Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA.
| | | | | |
Collapse
|
64
|
Babushkina IV, Ulyanov VY, Mamonova IA, Shpinyak SP. The Effect of Azithromycin on Biofilms Formation by Pathogens of Implant-Associated Infection in Large Joints. Bull Exp Biol Med 2020; 169:798-801. [PMID: 33108560 DOI: 10.1007/s10517-020-04982-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Indexed: 11/28/2022]
Abstract
We studied the effect of subbacteriostatic azithromycin concentrations on the formation of microbial biofilms by Pseudomonas aeruginosa strains that caused implant-associated infection of large joints. Azithromycin in subinhibitory for planktonic cells concentrations 0.01-0.02 μg/ml stimulated biofilm formation by both clinical and reference P. aeruginosa strains, while in concentrations of 1 μg/ml and higher completely inhibited the growth of both reference and clinical plankton P. aeruginosa strains, but stimulated biofilm formation. Increasing azithromycin concentration to 10 μg/ml led to inhibition of P. aeruginosa biofilm growth.
Collapse
Affiliation(s)
- I V Babushkina
- Research Institute of Traumatology, Orthopedics, and Neurosurgery, V. I. Razumovsky Saratov State Medical University, Ministry of Health of the Russian Federation, Saratov, Russia.
| | - V Yu Ulyanov
- Research Institute of Traumatology, Orthopedics, and Neurosurgery, V. I. Razumovsky Saratov State Medical University, Ministry of Health of the Russian Federation, Saratov, Russia
| | - I A Mamonova
- Research Institute of Traumatology, Orthopedics, and Neurosurgery, V. I. Razumovsky Saratov State Medical University, Ministry of Health of the Russian Federation, Saratov, Russia
| | - S P Shpinyak
- Research Institute of Traumatology, Orthopedics, and Neurosurgery, V. I. Razumovsky Saratov State Medical University, Ministry of Health of the Russian Federation, Saratov, Russia
| |
Collapse
|
65
|
Moore RE, Craft KM, Xu LL, Chambers SA, Nguyen JM, Marion KC, Gaddy JA, Townsend SD. Leveraging Stereoelectronic Effects in Biofilm Eradication: Synthetic β-Amino Human Milk Oligosaccharides Impede Microbial Adhesion As Observed by Scanning Electron Microscopy. J Org Chem 2020; 85:16128-16135. [PMID: 32996317 DOI: 10.1021/acs.joc.0c01958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Alongside Edward, Lemieux was among the earliest researchers studying negative hyperconjugation (i.e., the anomeric effect) or the preference for gauche conformations about the C1-O5 bond in carbohydrates. Lemieux also studied an esoteric, if not controversial, theory known as the reverse anomeric effect (RAE). This theory is used to rationalize scenarios where predicted anomeric stabilization does not occur. One such example is the Kochetkov amination where reducing end amines exist solely as the β-anomer. Herein, we provide a brief account of Lemieux's contributions to the field of stereoelectronics and apply this knowledge toward the synthesis of β-amino human milk oligosaccharides (βΑ-HMOs). These molecules were evaluated for their ability to inhibit growth and biofilm production in Group B Streptococcus (GBS) and Staphylococcus aureus. While the parent HMOs lacked antimicrobial and antibiofilm activity, their β-amino derivatives significantly inhibited biofilm formation in both species. Field emission gun-scanning single electron microscopy (FEG-SEM) revealed that treatment with β-amino HMOs significantly inhibits bacterial adherence and eliminates the ability of both microbes to form biofilms.
Collapse
Affiliation(s)
- Rebecca E Moore
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Kelly M Craft
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Lianyan L Xu
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Schuyler A Chambers
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Johny M Nguyen
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Keevan C Marion
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, 1161 21st Avenue South, D-3100 Medical Center North, Nashville, Tennessee 37232, United States.,Tennessee Valley Healthcare Systems, Department of Veterans Affairs, 1310 24th Avenue South, Nashville, Tennessee 37212, United States
| | - Steven D Townsend
- Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| |
Collapse
|
66
|
Afthab J, Khatoon N, Zhou L, Yao T, Shi S. Hepatoprotective Angelica sinensis silver nanoformulation against multidrug resistant bacteria and the integration of a multicomponent logic gate system. NANOSCALE 2020; 12:19149-19158. [PMID: 32936174 DOI: 10.1039/d0nr04744a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The rampant usage of antibiotics has led to the emergence of toxicity, especially hepatotoxicity and the emergence of microbial drug resistance. Hence, a series of novel hepatoprotective, biocompatible, antibacterial silver nanoformulations (AS-AgNPs) were developed by using the important Chinese medicinal plant Angelica sinensis. The different size of AS-AgNPs were characterized by UV-Visible spectroscopy, transmission electron microscopy (TEM), scanning electron microscopy (SEM), X-ray diffraction (XRD) and Fourier transform infrared spectroscopy (FT-IR). The size-dependent antibacterial properties of AS-AgNPs were investigated against Gram-positive, Gram-negative and multi-drug resistant bacteria. The minimum inhibitory concentration (MIC) of AS-AgNPs with different size against six bacteria was found to be in the range of 5-100 μg mL-1 with no resistance till 12 cycles. TEM and SEM results of bacteria after the treatment suggested that AS-AgNPs disrupted the cell membrane by creating pores. The cytocompatibility and cytoprotective effect of AS-AgNPs were evaluated against HepG2 cell lines, which showed that 85% of cells were viable up to 100 μg mL-1 of the concentration with almost no change in AST and ALT levels. Further, a logic combinatorial library, including basic logic gates (AND, OR, NOR, INHIBIT, IMPLICATION, and YES), three input logic gates (OR, and NOR) and combinatorial gates (INH-OR, INH-YES, INH-INH, AND-NOR, and NOT-AND-NOR) were designed by integrating multi-components based on the interaction between AS-AgNP1 and bacteria, where DiSC3(5) was used as the signal reporter. This system clearly demonstrates the ability of simple logic circuits to perform sophisticated analysis for the detection of multiple bacteria.
Collapse
Affiliation(s)
- Jouharsha Afthab
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Centre, Shanghai East Hospital, Tongji University, Shanghai, 200092, P. R. China.
| | - Nafeesa Khatoon
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Centre, Shanghai East Hospital, Tongji University, Shanghai, 200092, P. R. China.
| | - Lulu Zhou
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Centre, Shanghai East Hospital, Tongji University, Shanghai, 200092, P. R. China.
| | - Tianming Yao
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Centre, Shanghai East Hospital, Tongji University, Shanghai, 200092, P. R. China.
| | - Shuo Shi
- Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Breast Cancer Centre, Shanghai East Hospital, Tongji University, Shanghai, 200092, P. R. China.
| |
Collapse
|
67
|
Abdel Razek MMM, Moussa AY, El-Shanawany MA, Singab ANB. A New Phenolic Alkaloid from Halocnemum strobilaceum Endophytes: Antimicrobial, Antioxidant and Biofilm Inhibitory Activities. Chem Biodivers 2020; 17:e2000496. [PMID: 32840051 DOI: 10.1002/cbdv.202000496] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 07/21/2020] [Indexed: 01/27/2023]
Abstract
Human infections caused by microbial biofilm formation represent a growing major health threat. A new alkaloid, 3-amino-5-(3-hydroxybutan-2-yl)-4-methylphenol, was isolated from the corn grit culture of the endophytic isolate Penicillium citrinum-314 associated with Halocnemum strobilaceum, a halophyte growing in the Egyptian marshes. The new alkaloid was identified by 1D, 2D-NMR and HR-ESI-MS-MS and given the trivial name halociline. The antioxidant, antimicrobial and antibiofilm activities were recorded. Furthermore, another known compound, 1,3,6-trihydroxy-7-methoxy-9H-xanthen-9-one, was obtained in smaller amounts and revealed a non-microbicidal 100 % reduction in biofilm formation, with an MBIC value of 62.5 μg/mL (228 μM) against Pseudomonas aeruginosa (Ferm-BAM), a FRAP value of 447.941±37.876 mM/L as well as a marked safety profile against three cancer cell lines. Through in silico molecular docking study, in the binding sites of Pseudomonas enzymes, key ligand enzyme interactions were determined to support the in vitro results.
Collapse
Affiliation(s)
| | - Ashaimaa Y Moussa
- Faculty of Pharmacy, Ain Shams University, African Union Street, Abbassia, 11566, Cairo, Egypt
| | | | - Abdel Nasser B Singab
- Faculty of Pharmacy, Ain Shams University, African Union Street, Abbassia, 11566, Cairo, Egypt.,Center for Drug Discovery Research and Development, Ain Shams University, African Union Street, Abbassia, 11566, Cairo, Egypt
| |
Collapse
|
68
|
Zeiler MJ, Melander RJ, Melander C. Second-Generation Meridianin Analogues Inhibit the Formation of Mycobacterium smegmatis Biofilms and Sensitize Polymyxin-Resistant Gram-Negative Bacteria to Colistin. ChemMedChem 2020; 15:1672-1679. [PMID: 32662926 DOI: 10.1002/cmdc.202000438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 11/10/2022]
Abstract
Drug-resistant bacteria are rapidly becoming a significant problem across the globe. One element that factors into this crisis is the role played by bacterial biofilms in the recalcitrance of some infections to the effects of conventional antibiotics. Bacteria within a biofilm are highly tolerant of both antibiotic treatment and host immune responses. Biofilms are implicated in many chronic infections, including tuberculosis, in which they can act as bacterial reservoirs, requiring an arduous antibiotic regimen to eradicate the infection. A separate, compounding problem is that antibiotics once seen as last-resort drugs, such as the polymyxin colistin, are now seeing more frequent usage as resistance to front-line drugs in Gram-negative bacteria becomes more prevalent. The increased use of such antibiotics inevitably leads to an increased frequency of resistance. Drugs that inhibit biofilms and/or act as adjuvants to overcome resistance to existing antibiotics will potentially be an important component of future approaches to antibacterial treatment. We have previously demonstrated that analogues of the meridianin natural product family possess adjuvant and antibiofilm activities. In this study, we explore structural variation of the lead molecule from previous studies, and identify compounds showing both improved biofilm inhibition potency and synergy with colistin.
Collapse
Affiliation(s)
- Michael J Zeiler
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Roberta J Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Christian Melander
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| |
Collapse
|
69
|
Khan F, Tabassum N, Pham DTN, Oloketuyi SF, Kim YM. Molecules involved in motility regulation in Escherichia coli cells: a review. BIOFOULING 2020; 36:889-908. [PMID: 33028083 DOI: 10.1080/08927014.2020.1826939] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
The initial colonization of the host organism by commensal, probiotic, and pathogenic Escherichia coli strains is an important step in the development of infections and biofilms. Sensing and colonization of host cell surfaces are governed by flagellar and fimbriae/pili appendages, respectively. Biofilm formation confers great advantages on pathogenic E. coli cells such as protection against the host immune system, antimicrobial agents, and several environmental stress factors. The transition from planktonic to sessile physiological states involves several signaling cascades and factors responsible for the regulation of flagellar motility in E. coli cells. These regulatory factors have thus become important targets to control pathogenicity. Hence, attenuation of flagellar motility is considered a potential therapy against pathogenic E. coli. The present review describes signaling pathways and proteins involved in direct or indirect regulation of flagellar motility. Furthermore, application strategies for antimotility natural or synthetic compounds are discussed also.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan, Republic of Korea
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, Republic of Korea
| | - Dung Thuy Nguyen Pham
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| | | | - Young-Mog Kim
- Institute of Food Science, Pukyong National University, Busan, Republic of Korea
- Department of Food Science and Technology, Pukyong National University, Busan, Republic of Korea
| |
Collapse
|
70
|
Zhou S, Huang G. Retracted Article: The synthesis and biological activity of marine alkaloid derivatives and analogues. RSC Adv 2020; 10:31909-31935. [PMID: 35518151 PMCID: PMC9056551 DOI: 10.1039/d0ra05856d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
The ocean is the origin of life, with a unique ecological environment, which has given birth to a wealth of marine organisms. The ocean is an important source of biological resources and tens of thousands of monomeric compounds have been separated from marine organisms using modern separation technology. Most of these monomeric compounds have some kind of biological activity that has attracted extensive attention from researchers. Marine alkaloids are a kind of compound that can be separated from marine organisms. They have complex and special chemical structures, but at the same time, they can show diversity in biological activities. The biological activities of marine alkaloids mainly manifest in the form of anti-tumor, anti-fungus, anti-viral, anti-malaria, and anti-osteoporosis properties. Many marine alkaloids have good medicinal prospects and can possibly be used as anti-tumor, anti-viral, and anti-fungal clinical drugs or as lead compounds. The limited amounts of marine alkaloids that can be obtained by separation, coupled with the high cytotoxicity and low selectivity of these lead compounds, has restricted the clinical research and industrial development of marine alkaloids. Marine alkaloid derivatives and analogues have been obtained via rational drug design and chemical synthesis, to make up for the shortcomings of marine alkaloids; this has become an urgent subject for research and development. This work systematically reviews the recent developments relating to marine alkaloid derivatives and analogues in the field of medical chemistry over the last 10 years (2010-2019). We divide marine alkaloid derivatives and analogues into five types from the point-of-view of biological activity and elaborated on these activities. We also briefly discuss the optimization process, chemical synthesis, biological activity evaluation, and structure-activity relationship (SAR) of each of these compounds. The abundant SAR data provides reasonable approaches for the design and development of new biologically active marine alkaloid derivatives and analogues.
Collapse
Affiliation(s)
- Shiyang Zhou
- Chongqing Key Laboratory of Green Synthesis and Application, Active Carbohydrate Research Institute, College of Chemistry, Chongqing Normal University Chongqing 401331 China
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University Haikou Hainan 571158 China
| | - Gangliang Huang
- Chongqing Key Laboratory of Green Synthesis and Application, Active Carbohydrate Research Institute, College of Chemistry, Chongqing Normal University Chongqing 401331 China
| |
Collapse
|
71
|
Campana R, Mangiaterra G, Tiboni M, Frangipani E, Biavasco F, Lucarini S, Citterio B. A Fluorinated Analogue of Marine Bisindole Alkaloid 2,2-Bis(6-bromo-1 H-indol-3-yl)ethanamine as Potential Anti-Biofilm Agent and Antibiotic Adjuvant Against Staphylococcus aureus. Pharmaceuticals (Basel) 2020; 13:ph13090210. [PMID: 32859056 PMCID: PMC7557854 DOI: 10.3390/ph13090210] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/23/2022] Open
Abstract
Methicillin resistant Staphylococcus aureus (MRSA) infections represent a major global healthcare problem. Therapeutic options are often limited by the ability of MRSA strains to grow as biofilms on medical devices, where antibiotic persistence and resistance is positively selected, leading to recurrent and chronic implant-associated infections. One strategy to circumvent these problems is the co-administration of adjuvants, which may prolong the efficacy of antibiotic treatments, by broadening their spectrum and lowering the required dosage. The marine bisindole alkaloid 2,2-bis(6-bromo-1H-indol-3-yl)ethanamine (1) and its fluorinated analogue (2) were tested for their potential use as antibiotic adjuvants and antibiofilm agents against S. aureus CH 10850 (MRSA) and S. aureus ATCC 29213 (MSSA). Both compounds showed antimicrobial activity and bisindole 2 enabled 256-fold reduction (ΣFICs = 0.5) in the minimum inhibitory concentration (MIC) of oxacillin for the clinical MRSA strain. In addition, these molecules inhibited biofilm formation of S. aureus strains, and compound 2 showed greater eradicating activity on preformed biofilm compared to 1. None of the tested molecules exerted a viable but non-culturable cells (VBNC) inducing effect at their MIC values. Moreover, both compounds exhibited no hemolytic activity and a good stability in plasma, indicating a non-toxic profile, hence, in particular compound 2, a potential for in vivo applications to restore antibiotic treatment against MRSA infections.
Collapse
Affiliation(s)
- Raffaella Campana
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (R.C.); (M.T.); (E.F.)
| | - Gianmarco Mangiaterra
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (G.M.); (F.B.)
| | - Mattia Tiboni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (R.C.); (M.T.); (E.F.)
| | - Emanuela Frangipani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (R.C.); (M.T.); (E.F.)
| | - Francesca Biavasco
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy; (G.M.); (F.B.)
| | - Simone Lucarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (R.C.); (M.T.); (E.F.)
- Correspondence: (S.L.); (B.C.); Tel.: +39-0722-303-333 (S.L.); +39-0722-304-962 (B.C.)
| | - Barbara Citterio
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy; (R.C.); (M.T.); (E.F.)
- Correspondence: (S.L.); (B.C.); Tel.: +39-0722-303-333 (S.L.); +39-0722-304-962 (B.C.)
| |
Collapse
|
72
|
Khan F, Tabassum N, Anand R, Kim YM. Motility of Vibrio spp.: regulation and controlling strategies. Appl Microbiol Biotechnol 2020; 104:8187-8208. [PMID: 32816086 DOI: 10.1007/s00253-020-10794-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/15/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022]
Abstract
Flagellar motility in bacteria is a highly regulated and complex cellular process that requires high energy investment for movement and host colonization. Motility plays an important role in the lifestyle of Vibrio spp. in the aquatic environment and during host colonization. Flagellar motility in vibrios is associated with several cellular processes, such as movement, colonization, adhesion, biofilm formation, and virulence. The transcription of all flagella-related genes occurs hierarchically and is regulated positively or negatively by several transcription factors and regulatory proteins. The flagellar regulatory hierarchy is well studied in Vibrio cholerae and Vibrio parahaemolyticus. Here, we compared the regulatory cascade and molecules involved in the flagellar motility of V. cholerae and V. parahaemolyticus in detail. The evolutionary relatedness of the master regulator of the polar and lateral flagella in different Vibrio species is also discussed. Although they can form symbiotic associations of some Vibrio species with humans and aquatic organisms can be harmed by several species of Vibrio as a result of surface contact, characterized by flagellar movement. Thus, targeting flagellar motility in pathogenic Vibrio species is considered a promising approach to control Vibrio infections. This approach, along with the strategies for controlling flagellar motility in different species of Vibrio using naturally derived and chemically synthesized compounds, is discussed in this review. KEY POINTS: • Vibrio species are ubiquitous and distributed across the aquatic environments. • The flagellar motility is responsible for the chemotactic movement and initial colonization to the host. • The transition from the motile into the biofilm stage is one of the crucial events in the infection. • Several signaling pathways are involved in the motility and formation of biofilm. • Attenuation of motility by naturally derived or chemically synthesized compounds could be a potential treatment for preventing Vibrio biofilm-associated infections.
Collapse
Affiliation(s)
- Fazlurrahman Khan
- Institute of Food Science, Pukyong National University, Busan, 48513, South Korea.
| | - Nazia Tabassum
- Industrial Convergence Bionix Engineering, Pukyong National University, Busan, 48513, Republic of Korea
| | - Raksha Anand
- Department of Life Science, School of Basic Science and Research, Sharda University, 201306, Greater Noida, U.P., India
| | - Young-Mog Kim
- Institute of Food Science, Pukyong National University, Busan, 48513, South Korea. .,Department of Food Science and Technology, Pukyong National University, Busan, 48513, South Korea.
| |
Collapse
|
73
|
Targeting a bacterial DNABII protein with a chimeric peptide immunogen or humanised monoclonal antibody to prevent or treat recalcitrant biofilm-mediated infections. EBioMedicine 2020; 59:102867. [PMID: 32651162 PMCID: PMC7502671 DOI: 10.1016/j.ebiom.2020.102867] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/02/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Abstract
Background: Chronic and recurrent bacterial diseases are recalcitrant to treatment due to the ability of the causative agents to establish biofilms, thus development of means to prevent or resolve these structures are greatly needed. Our approach targets the DNABII family of bacterial DNA-binding proteins, which serve as critical structural components within the extracellular DNA scaffold of biofilms formed by all bacterial species tested to date. DNABII-directed antibodies rapidly disrupt biofilms and release the resident bacteria which promote their subsequent clearance by either host immune effectors or antibiotics that are now effective at a notably reduced concentration. Methods: First, as a therapeutic approach, we used intact IgG or Fab fragments against a chimeric peptide immunogen designed to target protective epitopes within the DNA-binding tip domains of integration host factor to disrupt established biofilms in vitro and to mediate resolution of existing disease in vivo. Second, we performed preventative active immunisation with the chimeric peptide to induce the formation of antibody that blocks biofilm formation and disease development in a model of viral-bacterial superinfection. Further, toward the path for clinical use, we humanised a monoclonal antibody against the chimeric peptide immunogen, then characterised and validated that it maintained therapeutic efficacy. Findings: We demonstrated efficacy of each approach in two well-established pre-clinical models of otitis media induced by the prevalent respiratory tract pathogen nontypeable Haemophilus influenzae, a common biofilm disease. Interpretation: Collectively, our data revealed two approaches with substantive efficacy and potential for broad application to combat diseases with a biofilm component. Funding Supported by R01 DC011818 to LOB and SDG.
Collapse
|
74
|
Srivastava GN, Malwe AS, Sharma AK, Shastri V, Hibare K, Sharma VK. Molib: A machine learning based classification tool for the prediction of biofilm inhibitory molecules. Genomics 2020; 112:2823-2832. [DOI: 10.1016/j.ygeno.2020.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 03/14/2020] [Accepted: 03/22/2020] [Indexed: 10/24/2022]
|
75
|
Design and synthesis of 4-[4-formyl-3-(2-naphthyl)pyrazol-1-yl]benzoic acid derivatives as potent growth inhibitors of drug-resistant Staphylococcus aureus. J Antibiot (Tokyo) 2020; 73:818-827. [PMID: 32601342 PMCID: PMC7655718 DOI: 10.1038/s41429-020-0341-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/30/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022]
Abstract
We report the synthesis and antimicrobial studies of a new series of naphthyl-substituted pyrazole-derived hydrazones. Many of these novel compounds are potent growth inhibitors of several strains of drug-resistant bacteria. These potent compounds have inclined growth inhibitory properties for planktonic Staphylococcus aureus and Acinetobacter baumannii, and its drug-resistant variants with minimum inhibitory concentration (MIC) as low as 0.78 and 1.56 μg/mL respectively. These compounds also show potent activity against S. aureus and A. baumannii biofilm formation and eradication properties. Time Kill Assay shows that these compounds are bactericidal for S. aureus and bacteriostatic for A. baumannii. The probable mode of action is the disruption of the bacterial cell membrane. Furthermore, potent compounds are nontoxic to human cell lines at several fold higher concentrations than the MICs.
Collapse
|
76
|
Ran HH, Cheng X, Bao YW, Hua XW, Gao G, Zhang X, Jiang YW, Zhu YX, Wu FG. Multifunctional quaternized carbon dots with enhanced biofilm penetration and eradication efficiencies. J Mater Chem B 2020; 7:5104-5114. [PMID: 31432881 DOI: 10.1039/c9tb00681h] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biofilm formation can lead to the treatment failure of persistent bacterial infections. Although a variety of antibacterial agents have been developed, the restricted drug penetration and the embedded bacteria's potentiated recalcitrance to these agents synergistically lead to the unsatisfactory anti-biofilm effect. Herein, we report the applications of metal-free quaternized carbon dots (CDs) in imaging and eliminating bacterial biofilms. The CDs prepared by the solvothermal treatment of dimethyloctadecyl[3-(trimethoxysilyl)propyl]ammonium chloride (abbreviated as Si-QAC) and glycerol possess ultrasmall size (ca. 3.3 ± 0.4 nm) and strong positively charged (zeta potential: ca. +33.1 ± 2.5 mV) surfaces with long alkyl chain-linked quaternary ammonium groups. The small size of the CDs endows them with the penetration ability into the interior of Gram-negative and Gram-positive bacterial biofilms, which enables excellent fluorescence imaging of the biofilms. Due to the different surfaces of the two types of bacteria, the positively charged CDs selectively interact with the more negatively charged Gram-positive bacteria via electrostatic and hydrophobic interactions, which inactivates the Gram-positive bacteria and ultimately eradicates the Gram-positive bacterial biofilms. In addition, we synthesize a new type of quaternized CDs without long alkyl chains (termed TTPAC CDs), and validate that the long alkyl chains potentiate the hydrophobic adhesion between CDs and Gram-positive bacteria. Meanwhile, the crystal violet staining results reveal that the cationic CDs inhibit the formation of Gram-positive bacterial biofilms. Collectively, our work highlights the feasibility of using cationic and ultrasmall metal-free CDs to eliminate and inhibit Gram-positive bacterial biofilms, which represents a highly effective strategy to cope with refractory biofilm-associated infections.
Collapse
Affiliation(s)
- Huan-Huan Ran
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Xiaotong Cheng
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Yan-Wen Bao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Xian-Wu Hua
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Ge Gao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Xiaodong Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Yao-Wen Jiang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Ya-Xuan Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China.
| |
Collapse
|
77
|
Ashrafudoulla M, Mizan MFR, Park SH, Ha SD. Current and future perspectives for controlling Vibrio biofilms in the seafood industry: a comprehensive review. Crit Rev Food Sci Nutr 2020; 61:1827-1851. [PMID: 32436440 DOI: 10.1080/10408398.2020.1767031] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The contamination of seafood with Vibrio species can have severe repercussions in the seafood industry. Vibrio species can form mature biofilms and persist on the surface of several seafoods such as crabs, oysters, mussels, and shrimp, for extended duration. Several conventional approaches have been employed to inhibit the growth of planktonic cells and prevent the formation of Vibrio biofilms. Since Vibrio biofilms are mostly resistant to these control measures, novel alternative methods need to be urgently developed. In this review, we propose environmentally friendly approaches to suppress Vibrio biofilm formation using a hypothesized mechanism of action.
Collapse
Affiliation(s)
- Md Ashrafudoulla
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, Anseong, Gyunggi-do, Republic of Korea
| | - Md Furkanur Rahaman Mizan
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, Anseong, Gyunggi-do, Republic of Korea
| | - Si Hong Park
- Food Science and Technology, Oregon State University, Corvallis, Oregon, USA
| | - Sang-Do Ha
- Department of Food Science and Technology, Advanced Food Safety Research Group, Chung-Ang University, Anseong, Gyunggi-do, Republic of Korea
| |
Collapse
|
78
|
Bijle MN, Neelakantan P, Ekambaram M, Lo ECM, Yiu CKY. Effect of a novel synbiotic on Streptococcus mutans. Sci Rep 2020; 10:7951. [PMID: 32409686 PMCID: PMC7224275 DOI: 10.1038/s41598-020-64956-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
We examined the effect of L-arginine - (i) on the growth of L. rhamnosus GG (LrG) and (ii) combined LrG synbiotic on the growth of cariogenic S. mutans. Viability of LrG was assessed using MTT/XTT assays, confocal imaging with ADS activity measurement. The effect of L-arginine (0.5%/1%/2%) (2×/24 h) with LrG on S. mutans was evaluated by measuring the colony forming units, biofilm biomass, real-time qPCR and confocal imaging. The pH of the spent media was measured immediately and 24 h post-treatment with assessment of lactic acid. The LrG viability was highest with 2% L-arginine (p < 0.001). Confocal imaging showed that 2% L-arginine increased biofilm thickness of LrG. The 2% L-arginine and LrG synbiotic significantly inhibited the growth of S. mutans (p < 0.001) reducing the viable counts (p = 0.002) and biofilm biomass (p < 0.001). The pH of spent media was the highest when treated with 2% L-arginine and LrG synbiotic (p < 0.001) with no difference between post-treatment and 24 h post-treatment (p > 0.05). Conversely, the 2% L-arginine and LrG synbiotic showed the lowest lactic acid production (p < 0.001). This study demonstrated that L-arginine enhanced the growth of LrG. The 2% L-arginine and LrG synbiotic synergistically inhibits the growth of S. mutans with significant potential to develop as an anti-caries regimen.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Prasanna Neelakantan
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Manikandan Ekambaram
- Paediatric Dentistry, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Edward C M Lo
- Dental Public Health, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
79
|
Taner B, Sevgi F, Göver T. Synthesis and Anti-Biofilm Activity of New Ferrocene Schiff
Bases. RUSS J GEN CHEM+ 2020. [DOI: 10.1134/s1070363220050254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
80
|
Leclercq L, Tessier J, Douyère G, Nardello-Rataj V, Schmitzer AR. Phytochemical- and Cyclodextrin-Based Pickering Emulsions: Natural Potentiators of Antibacterial, Antifungal, and Antibiofilm Activity. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:4317-4323. [PMID: 32271592 DOI: 10.1021/acs.langmuir.0c00314] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We present self-assembled Pickering emulsions containing biocidal phytochemical oils (carvacrol and terpinen-4-ol) and β-cyclodextrin able to potentiate the antimicrobial and antibiofilm activity of miconazoctylium bromide. The carvacrol-containing emulsion is 2-fold more sensitive against C. albicans and S. aureus and highly active against E. coli, compared to the commercial cream containing miconazole nitrate. Moreover, this emulsion shows a synergistic effect against fungi, additive responses against bacteria, and remarkable staphylococcal biofilm eradication. These results are associated with membrane permeabilization, enzymes inhibition, and the accumulation of reactive oxygen species in microorganisms.
Collapse
Affiliation(s)
- Loïc Leclercq
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181, UCCS - Unité de Catalyse et Chimie du Solide, 59000 Lille, France
| | - Jérémie Tessier
- Université de Montréal, Département de Chimie, CP 6128 Succursale Centre-Ville, H3C3J7 Montréal, Québec, Canada
| | - Grégory Douyère
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181, UCCS - Unité de Catalyse et Chimie du Solide, 59000 Lille, France
| | - Véronique Nardello-Rataj
- Univ. Lille, CNRS, Centrale Lille, Univ. Artois, UMR 8181, UCCS - Unité de Catalyse et Chimie du Solide, 59000 Lille, France
| | - Andreea R Schmitzer
- Université de Montréal, Département de Chimie, CP 6128 Succursale Centre-Ville, H3C3J7 Montréal, Québec, Canada
| |
Collapse
|
81
|
Liu K, Huigens RW. Instructive Advances in Chemical Microbiology Inspired by Nature's Diverse Inventory of Molecules. ACS Infect Dis 2020; 6:541-562. [PMID: 31842540 PMCID: PMC7346871 DOI: 10.1021/acsinfecdis.9b00413] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Natural product antibiotics have played an essential role in the treatment of bacterial infection in addition to serving as useful tools to explore the intricate biology of bacteria. Our current arsenal of antibiotics operate through the inhibition of well-defined bacterial targets critical for replication and growth. Pathogenic bacteria effectively utilize a diversity of mechanisms that lead to acquired resistance and/or innate tolerance toward antibiotic therapies, which can result in devastating consequences to human life. Several research groups have established innovative programs that work at the chemistry-biology interface to develop new molecules that aim to define and address concerns related to antibiotic resistance and tolerance. In this Review, we present recent progress by select research groups that highlight a diversity of integrated chemical biology and medicinal chemistry approaches aimed at the development and utilization of chemical tools that have led to promising new microbiological insights that may lead to significant clinical advances regarding the treatment of pathogenic bacteria.
Collapse
Affiliation(s)
- Ke Liu
- 1345 Center Drive, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| | - Robert W. Huigens
- 1345 Center Drive, Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
82
|
Walsh DJ, Livinghouse T, Durling GM, Chase-Bayless Y, Arnold AD, Stewart PS. Sulfenate Esters of Simple Phenols Exhibit Enhanced Activity against Biofilms. ACS OMEGA 2020; 5:6010-6020. [PMID: 32226882 PMCID: PMC7098047 DOI: 10.1021/acsomega.9b04392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 02/27/2020] [Indexed: 06/10/2023]
Abstract
The recalcitrance exhibited by microbial biofilms to conventional disinfectants has motivated the development of new chemical strategies to control and eradicate biofilms. The activities of several small phenolic compounds and their trichloromethylsulfenyl ester derivatives were evaluated against planktonic cells and mature biofilms of Staphylococcus epidermidis and Pseudomonas aeruginosa. Some of the phenolic parent compounds are well-studied constituents of plant essential oils, for example, eugenol, menthol, carvacrol, and thymol. The potency of sulfenate ester derivatives was markedly and consistently increased toward both planktonic cells and biofilms. The mean fold difference between the parent and derivative minimum inhibitory concentration against planktonic cells was 44 for S. epidermidis and 16 for P. aeruginosa. The mean fold difference between the parent and derivative biofilm eradication concentration for 22 tested compounds against both S. epidermidis and P. aeruginosa was 3. This work demonstrates the possibilities of a new class of biofilm-targeting disinfectants deploying a sulfenate ester functional group to increase the antimicrobial potency toward microorganisms in biofilms.
Collapse
Affiliation(s)
- Danica J Walsh
- Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, United States
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana 59717, United States
| | - Tom Livinghouse
- Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, United States
| | - Greg M Durling
- Chemistry and Biochemistry, Montana State University, Bozeman, Montana 59717, United States
| | - Yenny Chase-Bayless
- Fish and Wildlife, Montana State University, Bozeman, Montana 59717, United States
| | - Adrienne D Arnold
- Microbiology and Immunology, Montana State University, Bozeman, Montana 59717, United States
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana 59717, United States
| |
Collapse
|
83
|
Strategies to Prevent Biofilm Infections on Biomaterials: Effect of Novel Naturally-Derived Biofilm Inhibitors on a Competitive Colonization Model of Titanium by Staphylococcus aureus and SaOS-2 Cells. Microorganisms 2020; 8:microorganisms8030345. [PMID: 32121332 PMCID: PMC7143544 DOI: 10.3390/microorganisms8030345] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Biofilm-mediated infection is a major cause of bone prosthesis failure. The lack of molecules able to act in biofilms has driven research aimed at identifying new anti-biofilm agents via chemical screens. However, to be able to accommodate a large number of compounds, the testing conditions of these screenings end up being typically far from the clinical scenario. In this study, we assess the potential applicability of three previously discovered anti-biofilm compounds to be part of implanted medical devices by testing them on in vitro systems that more closely resemble the clinical scenario. To that end, we used a competition model based on the co-culture of SaOS-2 mammalian cells and Staphylococcus aureus (collection and clinical strains) on a titanium surface, as well as titanium pre-conditioned with high serum protein concentration. Additionally, we studied whether these compounds enhance the previously proven protective effect of pre-incubating titanium with SaOS-2 cells. Out of the three, DHA1 was the one with the highest potential, showing a preventive effect on bacterial adherence in all tested conditions, making it the most promising agent for incorporation into bone implants. This study emphasizes and demonstrates the importance of using meaningful experimental models, where potential antimicrobials ought to be tested for the protection of biomaterials in translational applications.
Collapse
|
84
|
de Oliveira Júnior NG, Franco OL. Promising strategies for future treatment of Klebsiella pneumoniae biofilms. Future Microbiol 2020; 15:63-79. [PMID: 32048525 DOI: 10.2217/fmb-2019-0180] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Klebsiella pneumoniae is a Gram-negative pathogenic bacterium that has the ability to aggregate as biofilm, representing one of the main agents in hospital infections, showing high rates of resistance to antibiotics. The K. pneumoniae biofilm aggregates are composed mainly of extracellular polysaccharides, eDNA and proteins. Besides, biofilms can attach to medical devices, such as endotracheal tubes and catheters, but are most dangerous on body surfaces. Here, we discuss the recent findings about the resistance mechanisms of K. pneumoniae biofilms, including genes and protein involved in 'classic', multidrug-resistant and hypervirulent strains, and also virulence factors. In addition, we also explore new strategies for possible treatment of these biofilms, and recently discovered molecules which may lead to future treatments.
Collapse
Affiliation(s)
- Nelson G de Oliveira Júnior
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil.,S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117-900, Brazil
| | - Octávio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, DF, Brazil.,S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, MS 79117-900, Brazil
| |
Collapse
|
85
|
Gu L, Chen Q, Guo A, Liu W, Ruan Y, Zhang X, Nou X. Differential Effects of Growth Medium Salinity on Biofilm Formation of Two Salmonella enterica Strains. J Food Prot 2020; 83:196-203. [PMID: 31895006 DOI: 10.4315/0362-028x.jfp-19-418] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 12/18/2022]
Abstract
ABSTRACT Salmonella enterica is a prominent foodborne pathogen, including diverse serotypes that are prolific biofilm formers. Its ability to form biofilm can be affected by multiple environmental factors. In this study, the effect of salinity on biofilm formation by S. enterica was evaluated by using two recently isolated strains of Salmonella serotypes Enteritidis and Newport. Although supplementing the growth medium with a low concentration (0.5 to 2%) of sodium chloride (NaCl) slightly enhanced biofilm formation for the strain S. enterica serovar Enteritidis 110, it sharply reduced or abolished biofilm formation by the strain S. enterica serovar Newport 193. This differential effect of salinity on S. enterica strains of different serotypes was poorly correlated with inhibition of planktonic growth but strongly correlated with cell motility. Examining genes known to affect biofilm formation showed that the expression of adrA, csgD, and fliC, which encode proteins required for surface adhesion and cell motility, was significantly downregulated with salinity increase in Salmonella Newport 193 but not in Salmonella Enteritidis 110. Therefore, it is plausible that the differential effect of salinity on biofilm formation by Salmonella Enteritidis 110 and Salmonella Newport 193 resulted from the differential regulation to genes required for cell adherence and motility. HIGHLIGHTS
Collapse
Affiliation(s)
- Lihong Gu
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Qian Chen
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Ailing Guo
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Wukang Liu
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Yao Ruan
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xinshuai Zhang
- National Research and Development Center for Egg Processing, College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| | - Xiangwu Nou
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, Maryland 20705, USA (ORCID: https://orcid.org/0000-0002-0886-460X [X.N.])
| |
Collapse
|
86
|
Huang Q, Wang S, Sun Y, Shi C, Yang H, Lu Z. Effects of Ag/ZnO nanocomposite at sub-minimum inhibitory concentrations on virulence factors of Streptococcus mutans. Arch Oral Biol 2019; 111:104640. [PMID: 31884336 DOI: 10.1016/j.archoralbio.2019.104640] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/09/2019] [Accepted: 12/23/2019] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Streptococcus mutans (S. mutans), the main pathogen of dental caries, could be well killed by Ag/ZnO nanocomposite. However, effects on virulence factors remain to be elucidated. This study investigated effects of Ag/ZnO at sub-minimum inhibitory concentrations (sub-MICs) on virulence factors of S. mutans and related genes expressions. DESIGN Effects of Ag/ZnO on the growth of S. mutans was investigated by growth curves and MTT staining method. The influence of Ag/ZnO at sub-MICs on biofilm formation was measured by the crystal violet staining method and observed by a scanning electron microscopy. Adherence, cell-surface hydrophobicity, acidogenicity and extracellular polysaccharides (EPS) of S. mutans after treatment by Ag/ZnO at sub-MICs were also investigated. Virulence factors related genes expressions after treated by Ag/ZnO at 1/2 MIC was conducted by the quantitative real-time PCR (qRT-PCR) method. RESULTS Sub-MICs of Ag/ZnO exhibited a dose-dependent inhibition on the virulence factors of S. mutans. Specially, Ag/ZnO at 1/2 MIC decreased 69.00 % biofilm formation, 31.78 % sucrose-independent and 48.08 % sucrose-dependent adherence, 69.44 % cell-surface hydrophobicity and 72.45 % water-soluble and 90.60 % water-insoluble EPS. Furthermore, the expression of virulence factors related genes was significantly suppressed by Ag/ZnO at 1/2 MIC. CONCLUSIONS Ag/ZnO at sub-MICs inhibited multiple virulence factors of S. mutans through downregulating the related genes. Ag/ZnO nanocomposite could be used for prevention of dental caries at low dosage.
Collapse
Affiliation(s)
- Qiaomu Huang
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China
| | - Shilei Wang
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China
| | - Yujun Sun
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China
| | - Cheng Shi
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China
| | - Hao Yang
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China
| | - Zhong Lu
- Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, No.206, Guanggu First Road, Wuhan, 430073, PR China.
| |
Collapse
|
87
|
Deepika MS, Thangam R, Sundarraj S, Sheena TS, Sivasubramanian S, Kulandaivel J, Thirumurugan R. Co-delivery of Diverse Therapeutic Compounds Using PEG–PLGA Nanoparticle Cargo against Drug-Resistant Bacteria: An Improved Anti-biofilm Strategy. ACS APPLIED BIO MATERIALS 2019; 3:385-399. [DOI: 10.1021/acsabm.9b00850] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Murugesan Sathiya Deepika
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| | - Ramar Thangam
- Centre for Academic and Research Excellence (CARE), CSIR-Central Leather Research Institute, Chennai, Tamil Nadu 600 020, India
| | - Shenbagamoorthy Sundarraj
- Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology (Deemed to be University), Jeppiaar Nagar, Chennai, Tamil Nadu 600 119, India
| | - Thankaraj Salammal Sheena
- Centre for Nanoscience and Nanotechnology, Department of Physics, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| | | | - Jeganathan Kulandaivel
- Centre for Nanoscience and Nanotechnology, Department of Physics, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| | - Ramasamy Thirumurugan
- Laboratory of Aquabiotics/Nanoscience, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, Tamil Nadu 620 024, India
| |
Collapse
|
88
|
Huigens RW, Abouelhassan Y, Yang H. Phenazine Antibiotic-Inspired Discovery of Bacterial Biofilm-Eradicating Agents. Chembiochem 2019; 20:2885-2902. [PMID: 30811834 PMCID: PMC7325843 DOI: 10.1002/cbic.201900116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Indexed: 12/19/2022]
Abstract
Bacterial biofilms are surface-attached communities of slow-growing and non-replicating persister cells that demonstrate high levels of antibiotic tolerance. Biofilms occur in nearly 80 % of infections and present unique challenges to our current arsenal of antibiotic therapies, all of which were initially discovered for their abilities to target rapidly dividing, free-floating planktonic bacteria. Bacterial biofilms are credited as the underlying cause of chronic and recurring bacterial infections. Innovative approaches are required to identify new small molecules that operate through bacterial growth-independent mechanisms to effectively eradicate biofilms. One source of inspiration comes from within the lungs of young cystic fibrosis (CF) patients, who often endure persistent Staphylococcus aureus infections. As these CF patients age, Pseudomonas aeruginosa co-infects the lungs and utilizes phenazine antibiotics to eradicate the established S. aureus infection. Our group has taken a special interest in this microbial competition strategy and we are investigating the potential of phenazine antibiotic-inspired compounds and synthetic analogues thereof to eradicate persistent bacterial biofilms. To discover new biofilm-eradicating agents, we have established an interdisciplinary research program involving synthetic medicinal chemistry, microbiology and molecular biology. From these efforts, we have identified a series of halogenated phenazines (HPs) that potently eradicate bacterial biofilms, and future work aims to translate these preliminary findings into ground-breaking clinical advances for the treatment of persistent biofilm infections.
Collapse
Affiliation(s)
- Robert W. Huigens
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| | - Yasmeen Abouelhassan
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| | - Hongfen Yang
- Department of Medicinal Chemistry; Center for Natural Products Drug Discovery and Development (CNPD3); University of Florida, Gainesville, FL, USA
| |
Collapse
|
89
|
Verderosa AD, Totsika M, Fairfull-Smith KE. Bacterial Biofilm Eradication Agents: A Current Review. Front Chem 2019; 7:824. [PMID: 31850313 PMCID: PMC6893625 DOI: 10.3389/fchem.2019.00824] [Citation(s) in RCA: 302] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 11/12/2019] [Indexed: 12/19/2022] Open
Abstract
Most free-living bacteria can attach to surfaces and aggregate to grow into multicellular communities encased in extracellular polymeric substances called biofilms. Biofilms are recalcitrant to antibiotic therapy and a major cause of persistent and recurrent infections by clinically important pathogens worldwide (e.g., Pseudomonas aeruginosa, Escherichia coli, and Staphylococcus aureus). Currently, most biofilm remediation strategies involve the development of biofilm-inhibition agents, aimed at preventing the early stages of biofilm formation, or biofilm-dispersal agents, aimed at disrupting the biofilm cell community. While both strategies offer some clinical promise, neither represents a direct treatment and eradication strategy for established biofilms. Consequently, the discovery and development of biofilm eradication agents as comprehensive, stand-alone biofilm treatment options has become a fundamental area of research. Here we review our current understanding of biofilm antibiotic tolerance mechanisms and provide an overview of biofilm remediation strategies, focusing primarily on the most promising biofilm eradication agents and approaches. Many of these offer exciting prospects for the future of biofilm therapeutics for a large number of infections that are currently refractory to conventional antibiotics.
Collapse
Affiliation(s)
- Anthony D Verderosa
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.,School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Makrina Totsika
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia.,School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kathryn E Fairfull-Smith
- School of Chemistry, Physics, and Mechanical Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
90
|
Deveaux W, Selvarajoo K. Searching for simple rules in Pseudomonas aeruginosa biofilm formation. BMC Res Notes 2019; 12:763. [PMID: 31752996 PMCID: PMC6873713 DOI: 10.1186/s13104-019-4795-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/11/2019] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE Living cells display complex and non-linear behaviors, especially when posed to environmental threats. Here, to understand the self-organizing cooperative behavior of a microorganism Pseudomonas aeruginosa, we developed a discrete spatiotemporal cellular automata model based on simple physical rules, similar to Conway's game of life. RESULTS The time evolution model simulations were experimentally verified for P. aeruginosa biofilm for both control and antibiotic azithromycin (AZM) treated condition. Our model suggests that AZM regulates the single cell motility, thereby resulting in delayed, but not abolished, biofilm formation. In addition, the model highlights the importance of reproduction by cell to cell interaction is key for biofilm formation. Overall, this work highlights another example where biological evolutionary complexity may be interpreted using rules taken from theoretical disciplines.
Collapse
Affiliation(s)
| | - Kumar Selvarajoo
- Biotransformation Innovation Platform (BioTrans), Agency for Science, Technology and Research A*STAR, Biopolis, Singapore.
| |
Collapse
|
91
|
Gondil VS, Kalaiyarasan T, Bharti VK, Chhibber S. Antibiofilm potential of Seabuckthorn silver nanoparticles (SBT@AgNPs) against Pseudomonas aeruginosa. 3 Biotech 2019; 9:402. [PMID: 31681523 PMCID: PMC6800877 DOI: 10.1007/s13205-019-1947-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/10/2019] [Indexed: 10/25/2022] Open
Abstract
In era of antibiotic resistance, antibacterial silver nanoparticles are considered as potential alternative therapeutic agent to combat drug resistant pathogens. The aim of present study was to evaluate the antibacterial, antibiofilm and biocompatible potential of green synthesized Seabuckthorn silver nanoparticles (SBT@AgNPs). In the study, antibacterial efficiency of SBT@AgNPs was studied against Pseudomonas aeruginosa, Klebsiella pneumoniae, Escherichia coli and methicillin resistant Staphylococcus aureus. SBT@AgNPs were found to possess high antibacterial activity which was indicated in terms of low minimum inhibitory and bactericidal concentrations (2-4 µg/ml) obtained against test pathogens. Anti-biofilm activity of SBT@AgNPs on young as well as mature P. aeruginosa biofilms was also evaluated. SBT@AgNPs were able to eradicate the P. aeruginosa biofilms, which was further confirmed by field emission scanning electron microscopy and confocal laser scanning microscopy. Quorum sensing assay also revealed the quorum quenching activity of SBT@AgNPs. Biocompatibility and cytocompatibility results demonstrated SBT@AgNPs to exhibit first-rate non-toxicity as no membrane damage on RBCs or detrimental morphology variation was seen in human dermal fibroblast. LC-MS analysis was also carried out to analyze the potential antibacterial chemical compounds present in aqueous extract of Seabuckthorn leaves. To the best of our knowledge this is first study in which green synthesized silver nanoparticles were exploited to eradicate young as well as mature biofilms of P. aeruginosa. Results showed that SBT@AgNPs are highly antibacterial, antibiofilm, nontoxic in nature and consequently can aid in biomedical applications.
Collapse
Affiliation(s)
- Vijay Singh Gondil
- Department of Microbiology, Basic Medical Sciences, Panjab University, Chandigarh, 160014 India
| | | | - Vijay K. Bharti
- Defence Institute of High Altitude Research (DIHAR), DRDO, Leh-Ladakh, J&K 194101 India
| | - Sanjay Chhibber
- Department of Microbiology, Basic Medical Sciences, Panjab University, Chandigarh, 160014 India
| |
Collapse
|
92
|
Li X, Yin L, Ramage G, Li B, Tao Y, Zhi Q, Lin H, Zhou Y. Assessing the impact of curcumin on dual-species biofilms formed by Streptococcus mutans and Candida albicans. Microbiologyopen 2019; 8:e937. [PMID: 31560838 PMCID: PMC6925172 DOI: 10.1002/mbo3.937] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 01/27/2023] Open
Abstract
Streptococcus mutans and Candida albicans are often isolated from plaques associated with early childhood caries. However, there are limited studies examining how these microorganisms interact with one another and how best to manage them. Recent studies have shown that curcumin (CUR), a natural compound, has the potential to independently control both of these microorganisms. The purpose of this study was to investigate how S. mutans and C. albicans respond in mono‐ and dual‐species biofilms challenged with CUR. Quantitative biofilm biomass and viability were first evaluated and supported by live–dead PCR to assess biofilm composition. Confocal laser scanning microscopy (CLSM) was used to evaluate the exopolysaccharide (EPS) content and thickness of the biofilms, and the structure of the biofilms and morphology of the cells were observed by scanning electron microscopy (SEM). Quantitative real‐time PCR (qRT‐PCR) was applied to assess relative gene expression. The 50% minimum biofilm eradication concentration (MBEC50) of CUR against S. mutans and C. albicans was 0.5 mM. The biomass and viability decreased after treatment with CUR both in dual‐species biofilms and in mono‐species biofilm. CUR inhibited S. mutans and C. albicans in both mono‐ and dual‐species biofilms. Streptococcus mutans was more sensitive to CUR in dual‐species biofilm than in mono‐species biofilms, whereas C. albicans was less sensitive in dual‐species biofilms. EPS production was decreased by CUR in both mono‐ and dual‐species biofilms, which coincided with the downregulation of glucosyltransferase and quorum sensing‐related gene expression of S. mutans. In C. albicans, the agglutinin‐like sequence family of C. albicans was also downregulated in dual‐species biofilms. Collectively, these data show the potential benefit of using a natural antimicrobial, CUR, to control caries‐related dual‐species plaque biofilms.
Collapse
Affiliation(s)
- Xinlong Li
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Luoping Yin
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bingchun Li
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ye Tao
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qinghui Zhi
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Huancai Lin
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yan Zhou
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
93
|
Cattò C, de Vincenti L, Borgonovo G, Bassoli A, Marai S, Villa F, Cappitelli F, Saracchi M. Sub-lethal concentrations of Perilla frutescens essential oils affect phytopathogenic fungal biofilms. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2019; 245:264-272. [PMID: 31158678 DOI: 10.1016/j.jenvman.2019.05.096] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 01/07/2019] [Accepted: 05/23/2019] [Indexed: 06/09/2023]
Abstract
The lack of deep knowledge of plant pathogenic fungal biofilms is reflected in the few existing environmental-friendly options for controlling fungal plant disease. Indeed, chemical fungicides still dominate the market but present-day concerns about their real efficacy, increasing awareness of the risk they pose to human health and the environment, and the incidence of fungicide resistance have all led to the current trend of near zero-market-tolerance for pesticide residues in fruit and vegetables. Here, essential oils (PK and PK-IK) from the edible leaves of two cultivars of Perilla frutescens are proposed as new, effective, non-toxic, eco-friendly pesticide-free options suitable for a preventive or integrative approach for sustainable crop protection and product preservation. PK and PK-IK were extracted and characterized, and their ability to affect the biofilm formation of the phytopathogenic model fungi Colletotrichum musae, Fusarium dimerum and Fusarium oxysporum was studied at non-lethal doses. Both essential oils at 1000 and 2000 mg l-1 showed excellent anti-biofilm performance: i) reducing conidia adhesion up to 80.3 ± 16.2%; ii) inhibiting conidia germination up to 100.0 ± 0.0%; iii) affecting biofilm structural development, with a reduction in dry weight of up to 100.0 ± 0.0% and extracellular polysaccharides and proteins up to 81.4 ± 8.0% and 51.0 ± 6.1% respectively. In all cases PK-IK showed better activity than PK.
Collapse
Affiliation(s)
- Cristina Cattò
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Luca de Vincenti
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Gigliola Borgonovo
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Angela Bassoli
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Simone Marai
- Department of Agricultural and Environmental Sciences - Production, Landscape, Agroenergy, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Federica Villa
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| | - Francesca Cappitelli
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy.
| | - Marco Saracchi
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milano, Italy
| |
Collapse
|
94
|
In Vitro Anti-Biofilm Activity of Curcumin Nanoparticles in Acinetobacter baumannii: A Culture-Based and Molecular Approach. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2019. [DOI: 10.5812/archcid.83263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
95
|
Craft KM, Nguyen JM, Berg LJ, Townsend SD. Methicillin-resistant Staphylococcus aureus (MRSA): antibiotic-resistance and the biofilm phenotype. MEDCHEMCOMM 2019; 10:1231-1241. [PMID: 31534648 PMCID: PMC6748282 DOI: 10.1039/c9md00044e] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/12/2019] [Indexed: 12/18/2022]
Abstract
Staphylococcus aureus (S. aureus) is an asymptomatic colonizer of 30% of all human beings. While generally benign, antibiotic resistance contributes to the success of S. aureus as a human pathogen. Resistance is rapidly evolved through a wide portfolio of mechanisms including horizontal gene transfer and chromosomal mutation. In addition to traditional resistance mechanisms, a special feature of S. aureus pathogenesis is its ability to survive on both biotic and abiotic surfaces in the biofilm state. Due to this characteristic, S. aureus is a leading cause of human infection. Methicillin-resistant S. aureus (MRSA) in particular has emerged as a widespread cause of both community- and hospital-acquired infections. Currently, MRSA is responsible for 10-fold more infections than all multi-drug resistant (MDR) Gram-negative pathogens combined. Recently, MRSA was classified by the World Health Organization (WHO) as one of twelve priority pathogens that threaten human health. In this targeted mini-review, we discuss MRSA biofilm production, the relationship of biofilm production to antibiotic resistance, and front-line techniques to defeat the biofilm-resistance system.
Collapse
Affiliation(s)
- Kelly M Craft
- Department of Chemistry , Vanderbilt University , 7300 Stevenson Science Center , Nashville , TN 37235 , USA .
| | - Johny M Nguyen
- Department of Chemistry , Vanderbilt University , 7300 Stevenson Science Center , Nashville , TN 37235 , USA .
| | - Lawrence J Berg
- Department of Chemistry , Vanderbilt University , 7300 Stevenson Science Center , Nashville , TN 37235 , USA .
| | - Steven D Townsend
- Department of Chemistry , Vanderbilt University , 7300 Stevenson Science Center , Nashville , TN 37235 , USA .
| |
Collapse
|
96
|
Jain N, Mansuri A. Stopping the Unstoppable: Unconventional Methods to Prevent the Biofilm Growth. Curr Drug Discov Technol 2019; 17:515-522. [PMID: 31362660 DOI: 10.2174/1570163816666190726153441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/11/2019] [Accepted: 06/03/2019] [Indexed: 12/18/2022]
Abstract
Biofilms are consortia of microorganisms encased in extracellular matrix that protect cells from adverse conditions. A biofilm matrix is typically composed of extracellular DNA, cellulose and proteinaceous amyloid fibers. The matrix aids in adhesion to abiotic and biotic surface including medical devices and host tissues. The presence of biofilm makes bacteria more resilient and non-responsive to most current treatment regimes at disposal. Therefore, biofilm-associated infections are serious threat in hospital settings and pose a huge burden on economy. Inhibition of matrix components (cellulose and/or amyloid formation) has emerged as a lucrative alternative strategy to cure biofilm-related infections and combat antibiotic resistance. Here we review the current and emerging therapeutic interventions to mitigate persistent infections due to biofilms. The successful implementation of these interventions will have a huge impact on alleviating the current financial burden on healthcare services.
Collapse
Affiliation(s)
- Neha Jain
- Department of Bioscience and Bioengineering, Indian Institute of Technology (IIT) Jodhpur, NH 65, Nagaur Road, Karwar, Jodhpur (Rajasthan), India
| | - Abdulkhalik Mansuri
- Department of Bioscience and Bioengineering, Indian Institute of Technology (IIT) Jodhpur, NH 65, Nagaur Road, Karwar, Jodhpur (Rajasthan), India
| |
Collapse
|
97
|
Scharnow AM, Solinski AE, Wuest WM. Targeting S. mutans biofilms: a perspective on preventing dental caries. MEDCHEMCOMM 2019; 10:1057-1067. [PMID: 31391878 PMCID: PMC6644389 DOI: 10.1039/c9md00015a] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
The prevalence of biofilm diseases, and dental caries in particular, have encouraged extensive research on S. mutans biofilms, including methods of preventing its formation. Numerous small molecules with specific anti-biofilm activity against this pathogen have been isolated and synthesized. Generally, these molecules can be characterized into three categories: sucrose-dependent anti-adhesion, sucrose-independent anti-adhesion and cellular signaling interference. This review aims to provide an overview of the current small molecule strategies used for targeting S. mutans biofilms, and a perspective of the future for the field.
Collapse
Affiliation(s)
- Amber M Scharnow
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| | - Amy E Solinski
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| | - William M Wuest
- Emory University , Chemistry Department , 1515 Dickey Dr , Atlanta , GA 30322 , USA .
| |
Collapse
|
98
|
Manna S, Ghosh AK, Mandal SM. Curd-Peptide Based Novel Hydrogel Inhibits Biofilm Formation, Quorum Sensing, Swimming Mortility of Multi-Antibiotic Resistant Clinical Isolates and Accelerates Wound Healing Activity. Front Microbiol 2019; 10:951. [PMID: 31139155 PMCID: PMC6527846 DOI: 10.3389/fmicb.2019.00951] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 04/15/2019] [Indexed: 01/12/2023] Open
Abstract
The search for a bioactive natural antibacterial agent with wound healing properties is a common practice for the development of new-generation molecules. Antimicrobial peptides are a good alternative to antibiotics and easy-to-form hydrogels under self-assembled conditions without pH adjustment. With this in mind, the peptide pool was extracted from a formulated curd composed of a blend of probiotic bacteria such as Streptococcus thermophilus, Lactobacillus casei, and Bifidobacterium bifidum at an optimized ratio of 7:1:2. The water content of curd was collected by the drainage column, centrifuged, filtered through a 0.45-μM filter, and used for hydrogel preparation. Matrix-assisted laser desorption/ionization time of flight (MALDI-TOF) mass spectrometry (MS) analysis confirmed the presence of peptide pool in the extracted water. The prepared hydrogel was freeze dried, and its effect on biofilm formation, swarming mortality, antimicrobials, wound healing, and biocompatibility was subsequently verified. Transmission electron microscope (TEM) and scanning electron microscope (SEM) images revealed the fibrous network of peptides after self-assembly with non-polar n-hexane solvent and a porous structure after drying, respectively. The observed biocompatibility, antimicrobial activity, and strong wound healing activity of the developed curd-based hydrogel have opened a new platform for antibacterial ointment formulation.
Collapse
Affiliation(s)
- Sounik Manna
- Central Research Facility, Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Ananta K Ghosh
- Central Research Facility, Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Santi M Mandal
- Central Research Facility, Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
99
|
A Novel Small Molecule, ZY354, Inhibits Dental Caries-Associated Oral Biofilms. Antimicrob Agents Chemother 2019; 63:AAC.02414-18. [PMID: 30858201 DOI: 10.1128/aac.02414-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 03/06/2019] [Indexed: 02/05/2023] Open
Abstract
Biofilm control is a critical approach to the better management of dental caries. Antimicrobial small molecules have shown their potential in the disruption of oral biofilm and control of dental caries. The objectives of this study were to examine the antimicrobial activity and cytotoxicity of a newly designed small-molecule compound, ZY354. ZY354 was synthesized, and its cytotoxicity was evaluated in human oral keratinocytes (HOK), human gingival epithelial cells (HGE), and macrophages (RAW) by CCK-8 assays. Minimal inhibitory concentrations (MICs), minimum bactericidal concentrations (MBCs), minimum biofilm inhibition concentrations (MBICs), and minimum biofilm reduction concentrations (MBRCs) of ZY354 against common oral streptococci (i.e., Streptococcus mutans, Streptococcus gordonii, and Streptococcus sanguinis) were determined by microdilution method. The exopolysaccharide (EPS)/bacterium ratio and the dead/live bacterium ratio in the ZY354-treated multispecies biofilms were determined by confocal laser scanning microscopy, and the microbial composition was visualized and quantified by fluorescent in situ hybridization and quantitative PCR (qPCR). The demineralizing activity of ZY354-treated biofilms was evaluated by transverse microradiography. The results showed that ZY354 exhibited low cytotoxicity in HOK, HGE, and RAW cells and exhibited potent antimicrobial activity against common oral streptococci. The EPS and the abundance of S. mutans were significantly reduced after ZY354 treatment, along with an increased dead/live microbial ratio in multispecies biofilms compared to the level with the nontreated control. The ZY354-treated multispecies biofilms exhibited reduced demineralizing activity at the biofilm/enamel interface. In conclusion, the small-molecule compound ZY354 exhibits low cytotoxicity and remarkable antimicrobial activity against oral streptococci, and it may have a great potential in anticaries clinical applications.
Collapse
|
100
|
Abouelhassan Y, Garrison AT, Yang H, Chávez-Riveros A, Burch GM, Huigens RW. Recent Progress in Natural-Product-Inspired Programs Aimed To Address Antibiotic Resistance and Tolerance. J Med Chem 2019; 62:7618-7642. [PMID: 30951303 DOI: 10.1021/acs.jmedchem.9b00370] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bacteria utilize multiple mechanisms that enable them to gain or acquire resistance to antibiotic therapies during the treatment of infections. In addition, bacteria form biofilms which are surface-attached communities of enriched populations containing persister cells encased within a protective extracellular matrix of biomolecules, leading to chronic and recurring antibiotic-tolerant infections. Antibiotic resistance and tolerance are major global problems that require innovative therapeutic strategies to address the challenges associated with pathogenic bacteria. Historically, natural products have played a critical role in bringing new therapies to the clinic to treat life-threatening bacterial infections. This Perspective provides an overview of antibiotic resistance and tolerance and highlights recent advances (chemistry, biology, drug discovery, and development) from various research programs involved in the discovery of new antibacterial agents inspired by a diverse series of natural product antibiotics.
Collapse
Affiliation(s)
- Yasmeen Abouelhassan
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| | - Aaron T Garrison
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| | - Hongfen Yang
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| | - Alejandra Chávez-Riveros
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| | - Gena M Burch
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| | - Robert W Huigens
- Department of Medicinal Chemistry, Center for Natural Products, Drug Discovery and Development (CNPD3), College of Pharmacy , University of Florida , Gainesville , Florida 32610 , United States
| |
Collapse
|