1
|
Atta L, Siddiqui AR, Mushtaq M, Munsif S, Nur-E-Alam M, Ahmed A, Ul-Haq Z. Molecular insights into antibiofilm inhibitors of Streptococcus mutans glucosyltransferases through in silico approaches. Sci Rep 2025; 15:14160. [PMID: 40269071 PMCID: PMC12018939 DOI: 10.1038/s41598-025-98927-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 04/15/2025] [Indexed: 04/25/2025] Open
Abstract
Streptococcus mutans, a primary cariogenic bacterium, plays a central role in dental caries, one of the most widespread chronic diseases globally. Glucosyltransferases (GTFs) are key virulence factors in this process, as they synthesize extracellular polysaccharides that contribute to biofilm formation and pathogenicity. Targeting GTFs has emerged as a promising strategy for preventing dental caries, with previous studies demonstrating its potential efficacy. This study builds on our prior work by providing detailed molecular insights into the binding modes of previously identified GTF inhibitors. Using computational tools, including density functional theory, molecular docking, and molecular dynamics simulations, we examined the binding interactions and structural stability of selected inhibitors. All investigated candidates demonstrated superior binding behavior compared to the reference ligand, acarbose, as indicated by multiple structural parameters. Structural dynamics analysis revealed significant stability in the binding interactions of Complex III and V, with average deviations of 2.06 ± 0.38 and 2.07 ± 0.30 Å, respectively. Similarly, a trend in structural compactness was observed, with gyration values of 32.98 ± 0.23 and 33.01 ± 0.24 Å, respectively. Principal component analysis indicated that the constructed pattern approaches zero with the achievement of a global energy minimum, particularly for Complex III and V. Furthermore, MM/PBSA free energy calculations identified Compound V as the most favorable binder, with a binding free energy of -24.20 kcal/mol. Our findings provide valuable molecular-level insights into the inhibitory mechanisms of GTF-targeting compounds, strengthening their potential as anti-cariogenic agents. By elucidating key binding interactions, this study contributes to the ongoing search for improved scaffolds that may hinder biofilm-mediated infections and advance therapeutic strategies against dental caries.
Collapse
Affiliation(s)
- Lubna Atta
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Ali Raza Siddiqui
- International Center for Chemical and Biological Sciences, H. E. J. Research Institute of Chemistry, University of Karachi, Karachi, 75270, Pakistan
| | - Mamona Mushtaq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sajida Munsif
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Mohammad Nur-E-Alam
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box. 2457, 11451, Riyadh, Kingdom of Saudi Arabia
| | - Aftab Ahmed
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA, 92618, USA
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
2
|
Santos AM, Vieira EM, de Jesus JR, Santana Júnior CC, Nascimento Júnior JAC, Oliveira AMS, Araújo AADS, Picot L, Alves IA, Serafini MR. Development and characterization of farnesol complexed in β- and hydroxypropyl-β-cyclodextrin and their antibacterial activity. Carbohydr Res 2025; 550:109406. [PMID: 39864120 DOI: 10.1016/j.carres.2025.109406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Farnesol (FAR) belongs to terpenes group and is a sesquiterpene alcohol and a hydrophobic compound, which can be extracted from natural sources or obtained by organic chemical or biological synthesis. Recent advances in the field of nanotechnology allow the drawbacks of low drug solubility, which can improve the drug therapeutic index. Therefore, this study aimed to prepare the FAR inclusion complexes with β-cyclodextrin (β-CD) and hydroxypropyl-β-cyclodextrin (HP-β-CD) through freeze-drying method, proposing their physicochemical characterization, comparing their toxicity, and evaluating their in vitro antibacterial activity. Initially, physical mixture and freeze-dried inclusion complexes of FAR/β-CD and FAR/HP-β-CD were obtained in the molar ratio (1:1). The samples were characterized by DSC, TG/DTG, FTIR, PXRD, SEM, pHPZC, and the complexation efficiency were performed by HPLC. In vivo toxicity assay was performed using Tenebrio molitor larvae to determine the LD50 and toxic dose of the samples. Also, it was proposed that the evaluation of the fluorescence suppression of Bovine Serum Albumin and the antibacterial activity. The complexation of FAR was evidenced with β-CD and HP-β-CD by the characterization techniques analyzed. The complexation efficiency of FAR/β-CD and FAR/HP-β-CD were 73,53 % and 74.12 %, respectively. The inclusion complexes demonstrated a reduction in toxicity, as evidenced by lower toxic and LD50 doses compared to the free FAR. The inclusion complexes induced conformational changes in BSA, suggesting that they reached the subdomains containing tryptophan residues. In terms of antibacterial activity, FAR/β-CD and FAR/HP-β-CD did not exhibit significant MIC results compared to free FAR, except for FAR/HP-β-CD against S. aureus ATCC 25923.
Collapse
Affiliation(s)
- Anamaria Mendonça Santos
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil.
| | - Edileuza Marcelo Vieira
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Jemmyson Romário de Jesus
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Ana Maria Santos Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Laurent Picot
- La Rochelle Université, UMR CNRS 7266 LIENSs, La Rochelle, France
| | - Izabel Almeida Alves
- Postgraduate Program in Pharmaceutical Sciences, University of the State of Bahia and Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil; Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
| |
Collapse
|
3
|
Kashi M, Varseh M, Hariri Y, Chegini Z, Shariati A. Natural compounds: new therapeutic approach for inhibition of Streptococcus mutans and dental caries. Front Pharmacol 2025; 16:1548117. [PMID: 40235544 PMCID: PMC11996897 DOI: 10.3389/fphar.2025.1548117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/12/2025] [Indexed: 04/17/2025] Open
Abstract
Streptococcus mutans is recognized as one of the leading causes of dental caries, and biofilm formation by this bacterium plays a key role in dental plaque development and caries progression. Given the increasing resistance of bacteria to antibiotics and the adverse effects of some synthetic antimicrobials, the search for natural alternatives has received increasing attention. The recently published studies have demonstrated that natural compounds (NCs) such as curcumin, cinnamaldehyde, eugenol, thymol, carvacrol, epigallocatechin gallate, farnesol, catechin, inulin, menthol, apigenin, myricetin, oleanolic acid, and resveratrol, have notable antimicrobial properties and can effectively inhibit the growth of Streptococcus mutans. NCs can disrupt bacterial membrane integrity, leading to cell death, and possess the capability to inhibit acid production, which is a key factor in caries development. NCs can also interfere with bacterial adhesion to surfaces, including teeth. The attachment inhibition is achieved by decreasing the expression of adhesion factors such as gtfs, ftf, fruA, and gbpB. NCs can disrupt bacterial metabolism, inhibit biofilm formation, disperse existing biofilm, and interfere with quorum sensing and two-component signal transduction systems. Moreover, novel drug delivery platforms were used to enhance the bioavailability and stability of NCs. Studies have also indicated that NCs exhibit significant efficacy in combination therapies. Notably, curcumin has shown promising results in photodynamic therapy against S. mutans. The current review article analyzes the mechanisms of action of various NCs against S. mutans and investigates their potential as alternative or complementary therapeutic options for managing this bacterium and dental caries.
Collapse
Affiliation(s)
- Milad Kashi
- Student research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Mahdieh Varseh
- Student Research Committee, Khomein University of Medical Sciences, Khomein, Iran
| | - Yasaman Hariri
- Student research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Aref Shariati
- Infectious Diseases Research Center (IDRC), Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
4
|
Zhou L, Liu Q, Fang Z, Li QL, Wong HM. Targeted antimicrobial self-assembly peptide hydrogel with in situ bio-mimic remineralization for caries management. Bioact Mater 2025; 44:428-446. [PMID: 39534787 PMCID: PMC11555604 DOI: 10.1016/j.bioactmat.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
The single-function agents with wide-spectrum activity which tend to disturb the ecological balance of oral cavity cannot satisfy dental treatment need. A multi-functional agent with specifically targeted killing property and in situ remineralization is warranted for caries management. A novel multi-functional agent (8DSS-C8-P-113) consisting of three domains, i.e., a non-specific antimicrobial peptide (AMP) (P-113), a competence stimulating peptide (C8), and an enhancing remineralization domain (8DSS), is fabricated and evaluated in this study. The findings demonstrates that 2 μM mL-1 of 8DSS-C8-P-113 eliminates planktonic Streptococcus mutans (S. mutans) without disrupting the oral normal flora. At a concentration of 8 μM mL-1, it exhibits the ability to prevent S. mutans' adhesion. Furthermore, 8DSS-C8-P-113 self-assembles a hydrogel state at the higher concentration of 16 μM mL-1. This hydrogel self-adheres on the tooth surface, resisting acid attack, eradicating S. mutans' biofilm, and inducing mineralization in order to facilitate the repair of demineralized dental hard tissue. Its significant effectiveness in reducing the severity of dental caries is also demonstrated in vivo in a rat model. This study suggests that the multi-functional bioactive AMP 8DSS-C8-P-113 is a promising agent to specifically target pathogen, prevent tooth demineralization, and effectively induce in situ bio-mimic remineralization for the management of dental caries.
Collapse
Affiliation(s)
- Li Zhou
- Faculty of Dentistry, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| | - Qing Liu
- Faculty of Dentistry, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| | - Zehui Fang
- Faculty of Dentistry, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| | - Quan Li Li
- Institute of Oral Science, Department of Stomatology, Longgang Otorhinolaryngology Hospital, No. 3004L Longgang Avenue, Shenzhen, 518172, China
- Key Lab. of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Meishan Road, Hefei, 230000, China
| | - Hai Ming Wong
- Faculty of Dentistry, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, China
| |
Collapse
|
5
|
Aragão MGB, Aires CP, Corona SAM, He X. Effects of epigallocatechin gallate on the development of matrix-rich Streptococcus mutans biofilm. BIOFOULING 2025; 41:171-180. [PMID: 39745056 DOI: 10.1080/08927014.2024.2446932] [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: 02/08/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 02/12/2025]
Abstract
In this study, we evaluated the impact of Epigalocatechin-3-gallate (EGCG) on S. mutans biofilm development for 24 and 46 h using high-resolution confocal laser scanning microscopy. EGCG treatment led to the formation of interspaced exopolysaccharide (EPS)-microcolony complexes unevenly distributed on the surface of hydroxyapatite disc, forming a thinner and less complex biofilm structure with significantly reduced biomass, matrix volume, and thickness compared to the NaCl treated group (negative control). At 46 h, the biofilm of the EGCG-treatment group failed to form the bacterial-EPS superstructures which is characteristic of the biofilm in the negative control group. EGCG treatment seems to significantly delay biofilm development, with the 46 h biofilm in the EGCG treatment group resembling the negative control group at 24 h. EGCG topical treatments impaired S. mutans biofilm initial growth and maturation, suggesting its potential to be used as a preventive agent against dental caries.
Collapse
Affiliation(s)
- Maria Gerusa Brito Aragão
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Microbiology, American Dental Association Forsyth Institute, Cambridge, Massachusetts, USA
| | - Carolina Patricia Aires
- Department of Biomolecular Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Silmara Aparecida Milori Corona
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Xuesong He
- Department of Microbiology, American Dental Association Forsyth Institute, Cambridge, Massachusetts, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Haj-Yahya F, Steinberg D, Sionov RV. Trans, Trans-Farnesol Enhances the Anti-Bacterial and Anti-Biofilm Effect of Arachidonic Acid on the Cariogenic Bacteria Streptococcus mutans and Streptococcus sobrinus. Int J Mol Sci 2024; 25:11770. [PMID: 39519322 PMCID: PMC11546208 DOI: 10.3390/ijms252111770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Streptococcus mutans and Streptococcus sobrinus are Gram-positive bacteria involved in the development of dental caries, as they are able to form biofilms on tooth enamel, ferment sugars into acids, and survive under acidic conditions. This ultimately leads to a local lowering of the pH value on the tooth surface, which causes enamel cavities. HYPOTHESIS One measure to reduce caries is to limit the growth of cariogenic bacteria by using two anti-bacterial agents with different mechanisms of action. The hypothesis of this study was that the anti-bacterial activity of ω-6 polyunsaturated arachidonic acid (AA) against S. mutans and S. sobrinus can be enhanced by the sesquiterpene alcohol trans, trans-farnesol (t,t-farnesol). METHODS The anti-bacterial activity of single and combined treatment was determined by the checkerboard assay. Bacterial viability was assessed by live/dead SYTO 9/propidium iodide (PI) staining on flow cytometry. Anti-biofilm activity was determined by MTT metabolic assay, crystal violet staining of biofilm biomass, SYTO 9/PI staining by spinning disk confocal microscopy (SDCM) and high-resolution scanning electron microscopy (HR-SEM). RESULTS t,t-Farnesol lowered the minimum inhibitory concentration (MIC) and the minimum biofilm inhibitory concentration (MBIC) of AA at sub-MICs. AA reduced the metabolic activity of preformed mature biofilms, while t,t-farnesol had no significant effect. The enhanced anti-bacterial effect of the combined t,t-farnesol/AA treatment was further evidenced by increased PI uptake, indicating membrane perforation. The enhanced anti-biofilm effect was further verified by SDCM and HR-SEM. Gene expression studies showed reduced expression of some biofilm-related genes. CONCLUSIONS Altogether, our study suggests a potential use of the two naturally occurring compounds arachidonic acid and t,t-farnesol for preventing biofilm formation by the cariogenic bacteria S. mutans and S. sobrinus. These findings have implications for caries prevention.
Collapse
|
7
|
Goc A, Sumera W, Rath M, Niedzwiecki A. Antibacterial and Antibiofilm Effects of L-Carnitine-Fumarate on Oral Streptococcal Strains Streptococcus mutans and Streptococcus sobrinus. Microorganisms 2024; 12:1613. [PMID: 39203455 PMCID: PMC11356751 DOI: 10.3390/microorganisms12081613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Streptococcus mutans is a major pathogenic habitant of oral caries. Owing to its physiological and biochemical features, it prevails in the form of plaque biofilm together with another important mutans streptococci species, Streptococcus sobrinus. Both species are considered as initiators of cavity lesions, and biofilm is essential to the dental caries process. Compared with the planktonic populations, the biofilm form has higher resistance to environmental conditions and antibiotics. Dental plaques also secure the long-term survival of microorganisms and protection from any stress conditions. To address the need for new antibiofilm agents, we have focused on L-carnitine-fumarate, a fumarate-conjugated quaternary ammonium compound. Using the macro-broth susceptibility testing method, we established its MIC value as 6.0 mg/mL. The MBC value, determined from the broth dilution minimum inhibitory concentration test by sub-culturing it to BHI agar plates, was established as 7.0 mg/mL. Antibiofilm efficacy was tested in 96-well plates coated with saliva using BHI broth supplemented with 1% sucrose as a standard approach. The obtained results allowed us to assess the MIBC as 7.5 mg/mL and the MBBC value as 10.0 mg/mL. The latter concentration also caused approximately 20% eradication of pre-existing biofilm. EPS-rich matrix, forming the core of the biofilm and enabling a confined acidic microenvironment, was also examined and confirmed the effectiveness of 10.0 mg/mL L-carnitine-fumarate concentration in inhibiting EPS formation. Furthermore, the anti-adherent and anti-aciduric impacts of L-carnitine-fumarate were investigated and revealed significant inhibitory effects at sub-MIC concentrations. The influence of L-carnitine-fumarate on the phosphotransferase system was investigated as well. Our results provide a new insight into the antibacterial potential of L-carnitine-fumarate as a valuable compound to be considered for alternative or adjunct anti-caries and antibiofilm preventive approaches.
Collapse
Affiliation(s)
- Anna Goc
- Dr. Rath Research Institute, 5941 Optical Ct., San Jose, CA 95138, USA; (W.S.); (M.R.)
| | | | | | | |
Collapse
|
8
|
Sang H, Jin H, Song P, Xu W, Wang F. Gallic acid exerts antibiofilm activity by inhibiting methicillin-resistant Staphylococcus aureus adhesion. Sci Rep 2024; 14:17220. [PMID: 39060363 PMCID: PMC11282228 DOI: 10.1038/s41598-024-68279-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a serious threat to patients with nosocomial infections, and infection is strongly associated with biofilm formation. Gallic acid (GA) is a natural bioactive compound found in traditional Chinese medicines that exerts potent antimicrobial activity. However, the anti-MRSA biofilm efficacy of GA remained to be determined. This study investigated the antimicrobial activities of GA against MRSA and the mechanisms involved. The results revealed the significant antibacterial and antibiofilm activities of GA. The minimal inhibitory concentration of GA against MRSA was 32 μg/mL and a growth curve assay confirmed the significant inhibitory effect of GA on planktonic MRSA. Crystal violet and XTT assays showed that 8 µg/mL GA effectively inhibited the formation of new biofilms and disrupted existing biofilms by reducing both biofilm biomass and metabolic activities. Alkaline phosphatase and β-galactosidase leakage assays and live/dead staining provided evidence that GA disrupted the integrity of bacterial cell walls and membranes within the biofilm. Scanning electron microscopy observations showed that GA significantly inhibited bacterial adhesion and aggregation, affecting the overall structure of the biofilm. Bacterial adhesion, polysaccharide intercellular adhesion (PIA) production and real-time quantitative PCR assay confirmed that GA inhibited bacterial adhesion, PIA synthesis, and the expression of icaAD and sarA. These results suggested that GA inhibited biofilm formation by inhibiting the expression of sarA, then downregulating the expression of icaA and icaD, thereby reducing the synthesis of PIA to attenuate the adhesion capacity of MRSA. GA is therefore a promising candidate for development as a pharmaceutical agent for the prevention and treatment of bacterial infections caused by MRSA.
Collapse
Affiliation(s)
- He Sang
- School of Life Science, Liaocheng University, Liaocheng, 252059, China
| | - Han Jin
- School of Life Science, Liaocheng University, Liaocheng, 252059, China
| | - Peng Song
- School of Life Science, Liaocheng University, Liaocheng, 252059, China
| | - Wei Xu
- School of Life Science, Liaocheng University, Liaocheng, 252059, China
| | - Fei Wang
- School of Life Science, Liaocheng University, Liaocheng, 252059, China.
| |
Collapse
|
9
|
Si B, Yang Y, Naveed M, Wang F, Chan MWH. Characterizations of biogenic selenium nanoparticles and their anti-biofilm potential against Streptococcus mutans ATCC 25175. J Trace Elem Med Biol 2024; 84:127448. [PMID: 38626650 DOI: 10.1016/j.jtemb.2024.127448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/18/2024]
Abstract
INTRODUCTION S. mutans has been identified as the primary pathogenic bacterium in biofilm-mediated dental caries. The biogenic selenium nanoparticles (SeNPs) produced by L. plantarum KNF-5 were used in this study against S. mutans ATCC 25175. OBJECTIVES The aims of this study were: (1) the biosynthesis of SeNPs by L. plantarum KNF-5, (2) the characterization of SeNPs, (3) the investigation of the inhibitory effect of biogenic SeNPs against S. mutans ATCC 25175, and (4) the determination of the anti-biofilm potential of SeNPS against S. mutans ATCC 25175. METHODOLOGY 3 mL of the culture was added to 100 mL of MRS medium and incubated. After 4 h, Na2SeO3 solution (concentration 100 μg/mL) was added and incubated at 37 °C for 36 h. The color of the culture solution changed from brownish-yellow to reddish, indicating the formation of SeNPs. The characterization of SeNPs was confirmed by UV-Vis spectrophotometry, FTIR, SEM-EDS and a particle size analyzer. The antibacterial activity was determined by the disk diffusion method, the MIC by the micro-double dilution method, and the biofilm inhibitory potential by the crystal violet method and the MTT assay. The effect of SeNPs on S. mutans ATCC 25175 was determined using SEM and CLSM spectrometry techniques. The sulfate-anthrone method was used to analyze the effect of SeNPs on insoluble extracellular polysaccharides. The expression of genes in S. mutans ATCC 25175 was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). PREPARATION OF NANOPARTICLES SeNPs produced by probiotic bacteria are considered a safe method. In this study, L. plantarum KNF-5 (probiotic strain) was used for the production of SeNPs. RESULTS The biogenic SeNPs were spherical and coated with proteins and polysaccharides and had a diameter of about 270 nm. The MIC of the SeNPs against S. mutans ATCC 25175 was 3.125 mg/mL. Biofilm growth was also significantly suppressed at this concentration. The expression of genes responsible for biofilm formation (GtfB, GtfC, BrpA and GbpB,) was reduced when S. mutans ATCC 25175 was treated with SeNPs. CONCLUSION It was concluded that the biogenic SeNPs produced by L. plantarum KNF-5 was highly effective to inhibit the growth of S. mutans ATCC 25175. NOVELTY STATEMENT The application of biogenic SeNPs, a natural anti-biofilm agent against S. mutans ATCC 25175. In the future, this study will provide a new option for the prevention and treatment of dental caries.
Collapse
Affiliation(s)
- Binbin Si
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Yang Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Muhammad Naveed
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Fenghuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Malik Wajid Hussain Chan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
10
|
Allemailem KS, Almatroudi A, Alharbi HOA, AlSuhaymi N, Alsugoor MH, Aldakheel FM, Khan AA, Rahmani AH. Apigenin: A Bioflavonoid with a Promising Role in Disease Prevention and Treatment. Biomedicines 2024; 12:1353. [PMID: 38927560 PMCID: PMC11202028 DOI: 10.3390/biomedicines12061353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/05/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Apigenin is a powerful flavone compound found in numerous fruits and vegetables, and it offers numerous health-promoting benefits. Many studies have evidenced that this compound has a potential role as an anti-inflammatory and antioxidant compound, making it a promising candidate for reducing the risk of pathogenesis. It has also been found to positively affect various systems in the body, such as the respiratory, digestive, immune, and reproductive systems. Apigenin is effective in treating liver, lung, heart, kidney, neurological diseases, diabetes, and maintaining good oral and skin health. Multiple studies have reported that this compound is capable of suppressing various types of cancer through the induction of apoptosis and cell-cycle arrest, suppressing cell migration and invasion, reduction of inflammation, and inhibiting angiogenesis. When used in combination with other drugs, apigenin increases their efficacy, reduces the risk of side effects, and improves the response to chemotherapy. This review broadly analyzes apigenin's potential in disease management by modulating various biological activities. In addition, this review also described apigenin's interaction with other compounds or drugs and the potential role of nanoformulation in different pathogeneses. Further extensive research is needed to explore the mechanism of action, safety, and efficacy of this compound in disease prevention and treatment.
Collapse
Affiliation(s)
- Khaled S. Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.A.); (H.O.A.A.)
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.A.); (H.O.A.A.)
| | - Hajed Obaid A. Alharbi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.A.); (H.O.A.A.)
| | - Naif AlSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia (M.H.A.)
| | - Mahdi H. Alsugoor
- Department of Emergency Medical Services, Faculty of Health Sciences, AlQunfudah, Umm Al-Qura University, Makkah 21912, Saudi Arabia (M.H.A.)
| | - Fahad M. Aldakheel
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia; (K.S.A.); (A.A.); (H.O.A.A.)
| |
Collapse
|
11
|
Lange A, Matuszewski A, Kutwin M, Ostrowska A, Jaworski S. Farnesol and Selected Nanoparticles (Silver, Gold, Copper, and Zinc Oxide) as Effective Agents Against Biofilms Formed by Pathogenic Microorganisms. Nanotechnol Sci Appl 2024; 17:107-125. [PMID: 38645468 PMCID: PMC11032122 DOI: 10.2147/nsa.s457124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024] Open
Abstract
Purpose Biofilms, which are created by most microorganisms, are known for their widely developed drug resistance, even more than planktonic forms of microorganisms. The aim of the study was to assess the effectiveness of agents composed of farnesol and nanoparticles (silver, gold, copper, and zinc oxide) in the degradation of biofilms produced by pathogenic microorganisms. Methods Escherichia coli, Enterococcus faecalis, Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans were used to create the biofilm structure. Colloidal suspensions of silver, gold, copper, and zinc oxide (Ag, Au, Cu, ZnO) with the addition of farnesol (F) were used as the treatment factor. The size distribution of those composites was analyzed, their zeta potential was measured, and their structure was visualized by transmission electron microscopy. The viability of the microorganism strains was assessed by an XTT assay, the ability to form biofilms was analyzed by confocal microscopy, and the changes in biofilm structure were evaluated by scanning electron microscopy. The general toxicity toward the HFFF2 cell line was determined by a neutral red assay and a human inflammation antibody array. Results The link between the two components (farnesol and nanoparticles) caused mutual stability of both components. Planktonic forms of the microorganisms were the most sensitive when exposed to AgF and CuF; however, the biofilm structure of all microorganism strains was the most disrupted (both inhibition of formation and changes within the structure) after AgF treatment. Composites were not toxic toward the HFFF2 cell line, although the expression of several cytokines was higher than in the not-treated group. Conclusion The in vitro studies demonstrated antibiofilm properties of composites based on farnesol and nanoparticles. The greatest changes in biofilm structure were triggered by AgF, causing an alteration in the biofilm formation process as well as in the biofilm structure.
Collapse
Affiliation(s)
- Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Arkadiusz Matuszewski
- Department of Animal Environment Biology, Institute of Animal Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
12
|
Dos Santos Ré AC, Cury JA, Sassaki GL, Aires CP. Structure of rhamnoglucan, an unexpected alkali-stable polysaccharide extracted from Streptococcus mutans cell wall. Int J Biol Macromol 2024; 262:130121. [PMID: 38350588 DOI: 10.1016/j.ijbiomac.2024.130121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/07/2024] [Accepted: 02/09/2024] [Indexed: 02/15/2024]
Abstract
This study identified a rhamnose-containing cell wall polysaccharide (RhaCWP) in an alkaline extract prepared to analyze intracellular polysaccharides (IPS) from Streptococcus mutans biofilm. IPS was an 1,4-α-D-glucan with branchpoints introduced by 1,6-α-glucan while RhaCWP presented 1,2-α-L-and 1,3-α-L rhamnose backbone and side chains connected by 1,2-α-D-glucans, as identified by nuclear magnetic resonance (NMR) spectroscopy and methylation analyses. The MW of IPS and RhaCWP was 11,298 Da, as determined by diffusion-ordered NMR spectroscopy. Therefore, this study analyzed the chemical structure of RhaCWP and IPS from biofilm in a single fraction prepared via a convenient hot-alkali extraction method. This method could be a feasible approach to obtain such molecules and improve the comprehension of the structure-function relationships in polymers from S. mutans in future studies.
Collapse
Affiliation(s)
- Ana Carolina Dos Santos Ré
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, CEP 14040-903 Ribeirão Preto, SP, Brazil.
| | - Jaime Aparecido Cury
- Department of Biosciences, Piracicaba Dental School, UNICAMP, CP 52, 13414-903 Piracicaba, SP, Brazil.
| | - Guilherme Lanzi Sassaki
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, CEP: 81531-980 Curitiba, PR, Brazil.
| | - Carolina Patrícia Aires
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, CEP 14040-903 Ribeirão Preto, SP, Brazil.
| |
Collapse
|
13
|
Carneiro BT, de Castro FNAM, Benetti F, Nima G, Suzuki TYU, André CB. Flavonoids effects against bacteria associated to periodontal disease and dental caries: a scoping review. BIOFOULING 2024; 40:99-113. [PMID: 38425046 DOI: 10.1080/08927014.2024.2321965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
This scoping review focused on exploring the efficacy of flavonoids against bacteria associated with dental caries and periodontal diseases. Inclusion criteria comprise studies investigating the antibacterial effects of flavonoids against bacteria linked to caries or periodontal diseases, both pure or diluted in vehicle forms. The search, conducted in August 2023, in databases including PubMed/MEDLINE, Scopus, Web of Science, Embase, LILACS, and Gray Literature. Out of the initial 1125 studies, 79 met the inclusion criteria, majority in vitro studies. Prominent flavonoids tested included epigallocatechin-gallate, apigenin, quercetin, and myricetin. Predominant findings consistently pointed to bacteriostatic, bactericidal, and antibiofilm activities. The study primarily investigated bacteria associated with dental caries, followed by periodontopathogens. A higher number of publications presented positive antibacterial results against Streptococcus mutans in comparison to Porphyromonas gingivalis. These encouraging findings underline the potential applicability of commercially available flavonoids in materials or therapies, underscoring the need for further exploration in this field.
Collapse
Affiliation(s)
- Bruna Tavares Carneiro
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Francine Benetti
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriel Nima
- Departament of Biomaterials, School of Dentistry, Universidad de los Andes, Santiago, Chile
| | - Thais Yumi Umeda Suzuki
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carolina Bosso André
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
14
|
Landa G, Miranda-Calderon LG, Gomez A, Perez M, Sebastian V, Arruebo M, Lamarche I, Tewes F, Irusta S, Mendoza G. Real-time in vivo monitoring of the antimicrobial action of combination therapies in the management of infected topical wounds. Int J Pharm 2023; 646:123502. [PMID: 37827392 DOI: 10.1016/j.ijpharm.2023.123502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/27/2023] [Accepted: 10/09/2023] [Indexed: 10/14/2023]
Abstract
The increasing prevalence of non-healing infected wounds has become a serious concern in the clinical practice, being associated to population aging and to the rising prevalence of several chronic conditions such as diabetes. Herein, the evaluation of the bactericidal and antibiofilm effects of the natural antiseptic terpenes thymol and farnesol standing alone or in combination with the standard care antiseptic chlorhexidine was carried out both in vitro and in vivo. The in vitro combinatorial treatment of chlorhexidine associated with those terpenes against Staphylococcus aureus in its planktonic and sessile forms demonstrated a superior antibacterial activity than that of chlorhexidine alone. Real-time in vivo monitoring of infection progression and antimicrobial treatment outcomes were evaluated using the bioluminescent S. aureus strain Xen36. In vivo studies on infected wound splinting murine models corroborated the superior bactericidal effects of the combinatorial treatments here proposed. Moreover, the encapsulation of thymol in electrospun Eudragit® S100 (i.e., a synthetic anionic copolymer of methacrylic acid and ethyl acrylate)-based wound dressings was also carried out in order to design efficient antimicrobial wound dressings.
Collapse
Affiliation(s)
- Guillermo Landa
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering. University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain
| | - Laura G Miranda-Calderon
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering. University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain
| | - Alex Gomez
- Department of Animal Pathology, University of Zaragoza, 177 Miguel Servet Street, 50013 Zaragoza, Spain; Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2), University of Zaragoza, 50013 Zaragoza, Spain
| | - Marta Perez
- Instituto Universitario de Investigación Mixto Agroalimentario de Aragón (IA2), University of Zaragoza, 50013 Zaragoza, Spain; Department of Anatomy, Embriology and Animal Genetics, University of Zaragoza, 177 Miguel Servet Street, 50013 Zaragoza, Spain
| | - Victor Sebastian
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering. University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Manuel Arruebo
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering. University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain.
| | - Isabelle Lamarche
- INSERM U1070 "Pharmacology of anti-infective agents", 1 rue Georges Bonnet, Pôle Biologie Santé, 86022 Poitiers, France
| | - Frederic Tewes
- INSERM U1070 "Pharmacology of anti-infective agents", 1 rue Georges Bonnet, Pôle Biologie Santé, 86022 Poitiers, France.
| | - Silvia Irusta
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC-Universidad de Zaragoza, 50009 Zaragoza, Spain; Department of Chemical and Environmental Engineering. University of Zaragoza, Campus Río Ebro-Edificio I+D, C/ Poeta Mariano Esquillor S/N, 50018 Zaragoza, Spain; Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| | - Gracia Mendoza
- Aragon Health Research Institute (IIS Aragon), 50009 Zaragoza, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, CIBER-BBN, 28029 Madrid, Spain
| |
Collapse
|
15
|
Dini C, Costa RC, Bertolini M, Shibli JA, Feres M, Klein MI, de Avila ÉD, Souza JGS, Barão VAR. In-vitro polymicrobial oral biofilm model represents clinical microbial profile and disease progression during implant-related infections. J Appl Microbiol 2023; 134:lxad265. [PMID: 37951291 DOI: 10.1093/jambio/lxad265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/13/2023]
Abstract
AIM Clinically relevant in-vitro biofilm models are essential and valuable tools for mechanistically dissecting the etiopathogenesis of infectious diseases and test new antimicrobial therapies. Thus, the aim of this study was to develop and test a clinically relevant in-vitro oral polymicrobial biofilm model that mimics implant-related infections in terms of microbial profile. METHODS AND RESULTS For this purpose, 24-well plate system was used to model oral biofilms, using three different microbial inoculums to grow in-vitro biofilms: (1) human saliva from periodontally healthy patients; (2) saliva as in inoculum 1 + Porphyromonas gingivalis strain; and (3) supra and subgingival biofilm collected from peri-implant sites of patients diagnosed with peri-implantitis. Biofilms were grown to represent the dynamic transition from an aerobic to anaerobic community profile. Subsequently, biofilms were collected after each phase and evaluated for microbiological composition, microbial counts, biofilm biomass, structure, and susceptibility to chlorhexidine (CHX). Results showed higher live cell count (P < .05) for biofilms developed from patients' biofilm inoculum, but biomass volume, dry weight, and microbiological composition were similar among groups (P > .05). Interestingly, according to the checkerboard DNA-DNA hybridization results, the biofilm developed from stimulated human saliva exhibited a microbial composition more similar to the clinical subgingival biofilm of patients with peri-implantitis, with proportions of the main pathogens closer to those found in the disease. In addition, biofilm developed using saliva as inoculum was shown to be susceptible to CHX with significant reduction in bacteria compared with biofilms without exposure to CHX (P < .05). CONCLUSION The findings suggested that the in-vitro polymicrobial biofilm developed from human saliva as inoculum is a suitable model and clinically relevant tool for mimicking the microbial composition of implant-related infections.
Collapse
Affiliation(s)
- Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, SP 13414-903, Brazil
| | - Raphael Cavalcante Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, SP 13414-903, Brazil
| | - Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jamil Awad Shibli
- Dental Research Division, Guarulhos University, Guarulhos, SP 07011-010, Brazil
| | - Magda Feres
- Dental Research Division, Guarulhos University, Guarulhos, SP 07011-010, Brazil
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States
| | - Marlise Inêz Klein
- Department of Oral Diagnosis, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, SP 13414-903, Brazil
| | - Érica Dorigatti de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, São Paulo State University (UNESP), Araraquara, SP 14801-385, Brazil
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, SP 16015-050, Brazil
| | | | - Valentim Adelino Ricardo Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, SP 13414-903, Brazil
| |
Collapse
|
16
|
Zhou Y, Zhang B, Wang Y, Hu R. Effects of Sulforaphene on the Cariogenic Properties of Streptococcus Mutans In Vitro and Dental Caries Development In Vivo. Antibiotics (Basel) 2023; 12:1359. [PMID: 37760656 PMCID: PMC10525627 DOI: 10.3390/antibiotics12091359] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Sulforaphene (SFE) is a common nutritional supplement with antibacterial, anti-cancer, and anti-inflammatory effects. However, the effects of SFE on the cariogenicity of Streptococcus mutans and dental caries have not been reported. The objectives of this study were to investigate the caries-controlling potential of SFE. The effects of SFE on S. mutans were investigated using the broth microdilution method, crystal violet staining, SEM observation, acid tolerance assays, lactic acid quantification, and polysaccharide measurements. A rat caries model was established to evaluate the caries-controlling effects and biocompatibility of SFE in vivo. SFE inhibited S. mutans growth and biofilm formation. Furthermore, SFE restrained the cariogenic properties of S. mutans, including its acid production, acid tolerance, and extracellular polysaccharide production, without affecting the bacterial viability at sub-inhibitory levels. In the rat caries model, SFE significantly arrested the onset and development of dental caries. Moreover, no visible hemolytic phenomenon or cytotoxicity was detected in the SFE groups. After four weeks of SFE treatment, all rats remained in apparent good health with no significant differences in weight gain; their hemogram and biochemical parameters were normal; no pathological changes were observed in the oral mucosa, liver, or kidneys. In conclusion, SFE was safe and inhibited the development of caries effectively.
Collapse
Affiliation(s)
- Yuehong Zhou
- The College of Renji, Wenzhou Medical University, Wenzhou 325000, China
| | - Binhan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- The College of Life Science, Sichuan University, Chengdu 610041, China
| | - Yufei Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Rongdang Hu
- The College of Renji, Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
17
|
Bem JSP, Lacerda NGS, Polizello ACM, Cabral H, da Rosa-Garzon NG, Aires CP. Mutanase from Trichoderma harzianum inductively Produced by Mutan: Short-Term Treatment to Degrade Mature Streptococcus mutans Biofilm. Curr Microbiol 2023; 80:312. [PMID: 37542660 DOI: 10.1007/s00284-023-03417-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 07/14/2023] [Indexed: 08/07/2023]
Abstract
This study aimed to evaluate the disruptive effect of fungal mutanase against cariogenic biofilm after short-term treatment. For that, mature Streptococcus mutans biofilms (n = 9) were exposed to active or inactivated enzymes produced by Trichoderma harzianum for 1 min, two times per day. Biofilms were analyzed by amount of matrix water-insoluble polysaccharides, bacterial viability, acidogenicity, and morphology by scanning electron microscopy (SEM). The group treated with active enzymes (AE) had a significantly lower amount of insoluble polysaccharides (893.30 ± 293.69) when compared to the negative control group (NaCl, 2192.59 ± 361.96), yet no significant difference was found when comparing to the positive control group (CHX, 436.82 ± 151.07). Also, there was no significant effect on bacteria metabolism and viability (P-value < 0.05). Data generated by the quantitative analysis were confirmed through scanning electron microscopy images. Thus, fungal mutanase degraded the biofilm after a short-term treatment without interfering with bacterial viability and metabolism. Such findings offer insight to the development of routine oral care products containing this input.
Collapse
Affiliation(s)
- Jéssica Silva Peixoto Bem
- Department of Children's Clinic, Ribeirão Preto School of Dentistry, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-904, Brazil
| | - Nayanna Gomes Silva Lacerda
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Ana Cristina Morseli Polizello
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Hamilton Cabral
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Nathalia Gonsales da Rosa-Garzon
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil
| | - Carolina Patrícia Aires
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Café Avenue s/n, Ribeirão Preto, São Paulo, 14040-903, Brazil.
| |
Collapse
|
18
|
Rudin L, Bornstein MM, Shyp V. Inhibition of biofilm formation and virulence factors of cariogenic oral pathogen Streptococcus mutans by natural flavonoid phloretin. J Oral Microbiol 2023; 15:2230711. [PMID: 37416858 PMCID: PMC10321187 DOI: 10.1080/20002297.2023.2230711] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Objectives To evaluate the effect and mechanism of action of the flavonoid phloretin on the growth and sucrose-dependent biofilm formation of Streptococcus mutans. Methods Minimum inhibitory concentration, viability, and biofilm susceptibility assays were conducted to assess antimicrobial and antibiofilm effect of phloretin. Biofilm composition and structure were analysed with scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). Water-soluble (WSG) and water-insoluble glucan (WIG) were determined using anthrone method. Lactic acid measurements and acid tolerance assay were performed to assess acidogenicity and aciduricity. Reverse transcription quantitative PCR (RT-qPCR) was used to measure the expression of virulence genes essential for surface attachment, biofilm formation, and quorum sensing. Results Phloretin inhibited S. mutans growth and viability in a dose-dependent manner. Furthermore, it reduced gtfB and gtfC gene expression, correlating with the reduction of extracellular polysaccharides (EPS)/bacteria and WIG/WSG ratio. Inhibition of comED and luxS gene expression, involved in stress tolerance, was associated with compromised acidogenicity and aciduricity of S. mutans. Conclusions Phloretin exhibits antibacterial properties against S. mutans, modulates acid production and tolerance, and reduces biofilm formation. Clinical significance Phloretin is a promising natural compound with pronounced inhibitory effect on key virulence factors of the cariogenic pathogen, S. mutans.
Collapse
Affiliation(s)
- Lucille Rudin
- Department Research, University Center for Dental Medicine Basel UZB, University of Basel, Basel, Switzerland
| | - Michael M. Bornstein
- Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of BaselBaselSwitzerland
- Head of the Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel. Mattenstrasse 40, Basel, Switzerland
| | - Viktoriya Shyp
- Postdoctoral Researcher. Department Research, University Center for Dental Medicine Basel UZB
- Department of Oral Health & Medicine, University Center for Dental Medicine Basel UZB, University of Basel. Mattenstrasse 40, Basel, Switzerland
| |
Collapse
|
19
|
Souza LVS, Pavanello L, Picolo MZD, Kury M, Matos ICRT, Cogo-Müller K, Esteban Florez FL, Cavalli V. Mechanical and antibacterial properties of an experimental flowable composite containing Nb 2O 5 and NF_TiO 2 nanoparticles. J Mech Behav Biomed Mater 2023; 143:105919. [PMID: 37279637 DOI: 10.1016/j.jmbbm.2023.105919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 06/08/2023]
Abstract
This study developed an experimental flowable composite incorporated with niobium pentoxide (Nb2O5) combined or not with titanium dioxide co-doped with fluorine and nitrogen (NF_TiO2) and evaluated the mechanical and antibacterial properties. The experimental flowable composite (TEGDMA + BisGMA 1:1 + 60%wt - inorganic filler - borosilicate 0.7 μm) was formulated according to the type and concentration of Nb2O5 and NF_TiO2 (0.5, 1, 1.5 and 2 wt%) or NF_TiO2 + Nb2O5 (0.25, 0.5, 0.75 and 1 wt% - 1:1). The control groups were formed by the experimental composite without the incorporation of Nb2O5 and/or NF_TiO2 (GC-E) and by a commercial flowable composite (GC). The characterization of the surface of the composite and its particles was carried out using scanning electron microscopy (SEM) and energy dispersive x-rays (EDX). Specimens were manufactured and subjected to mechanical tests of flexural strength (FS) (n = 12), flexural modulus (FM) (n = 12), roughness (Ra) (n = 10), microhardness (n = 10), and contact angle (n = 10); and, to evaluate the antibacterial activity, they were submitted to tests of biofilm formation against S. mutans (CFU/mL) (n = 5), biofilm biomass by dry weight (n = 5) and confocal laser microscopy (%LIVE/DEAD) (n = 5). Data were submitted to one-way ANOVA and Tukey's post-hoc and, those that were not homoscedastic, but with normality, were submitted to Welch's ANOVA and Games-Howell's post-hoc. Dunnet's test was used to compare the controls with the other experimental groups (α = 5). The Nb2O5 particles had an average size of 32.4 μm and the nanoparticles (NPs) of NF_TiO2, 10 nm. EDX analysis identified isolated peaks of N, F, Ti, and Nb confirming the presence of these particles in the resin matrix. The 1.5% NF_TiO2 group had a higher FS and FM than the controls (p < 0.05). GC showed higher microhardness between groups (p < 0.05). There was no difference between the experimental groups regarding contact angle and roughness (p > 0.05), except for GC, which had the highest Ra values and the lowest contact angle between groups (p < 0.05). Composites containing 0.5%, 1%, 1.5%, and 2% Nb2O5, 1%, 1.5%, and 2% NF_TiO2 and 2% Nb2O5 + NF_TiO2 showed lower biofilm formation (p < 0.05), lower total biofilm biomass (p < 0.05), and a higher percentage of dead cells (44%, 52%, 52%, 79%, 42% 43%, 62%, 65%, respectively) than GC and GC-E (5% and 1%, respectively). It is concluded that the incorporation of 1.5% NF_TiO2 promoted a greater FS and FM among the experimental composites and that the addition of Nb2O5 particles (0.5%, 1%, 1.5%, and 2%), NF_TiO2 (1%, 1.5% and 2%) and the combination Nb2O5 + NF_TiO2 (2%) showed significant antibacterial effects.
Collapse
Affiliation(s)
| | - Larissa Pavanello
- Department of Biosciences, University of Campinas, Piracicaba Dental School, Piracicaba, SP, Brazil
| | - Mayara Zaghi Dal Picolo
- Department of Restorative Dentistry, University of Campinas - Piracicaba Dental School, Piracicaba, SP, Brazil
| | - Matheus Kury
- Department of Restorative Dentistry, University of Campinas - Piracicaba Dental School, Piracicaba, SP, Brazil
| | | | - Karina Cogo-Müller
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Fernando Luis Esteban Florez
- Division of Dental Biomaterials, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Vanessa Cavalli
- Department of Restorative Dentistry, University of Campinas - Piracicaba Dental School, Piracicaba, SP, Brazil.
| |
Collapse
|
20
|
Giacaman RA, Umaña R, Nuñez MJ, Díaz-Garrido N, Echeverría C, García-Manríquez N, Mira A, Fernández CE, Gambetta-Tessini K, Lozano CP. Saliva Decreases Sucrose-Induced Cariogenicity in an Experimental Biological Caries Model. Microorganisms 2023; 11:1426. [PMID: 37374928 DOI: 10.3390/microorganisms11061426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVE Whether a minimum quantity of saliva inhibit the caries process remains uncertain. This study aimed to investigate the impact of saliva dilutions on an in vitro caries model using Streptococcus mutans (S. mutans) biofilms. METHODS S. mutans biofilms were cultivated on enamel and root dentin slabs, in culture media containing different proportions of saliva (v/v): 0%, 5%, 10%, 25%, 50%, 75%, and 100% saliva, and exposed to a 10% sucrose solution (5 min, 3x/day), with appropriate controls. After 5 (enamel) and 4 (dentin) days, demineralization, biomass, viable bacteria, and polysaccharide formation were analyzed. The acidogenicity of the spent media was monitored overtime. Each assay was performed in triplicate across two independent experiments (n = 6). RESULTS In both enamel and dentin, an inverse relationship was observed between acidogenicity, demineralization, and the proportion of saliva. Even small quantities of saliva incorporated into the media led to a noticeable reduction in enamel and dentin demineralization. Saliva presence resulted in significant reductions in biomass, viable S. mutans cells, and polysaccharides, with the effects being concentration-dependent for both tissues. CONCLUSIONS High quantities of saliva can almost completely inhibit sucrose-induced cariogenicity, while even small amounts exhibit a dose-dependent caries-protective effect.
Collapse
Affiliation(s)
- Rodrigo A Giacaman
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
- Interuniversity Center for Healthy Aging, Consortium of Chilean State Universities, Chilecito 03825, Chile
| | - Rodrigo Umaña
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - María José Nuñez
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Natalia Díaz-Garrido
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Constanza Echeverría
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Natalia García-Manríquez
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Alex Mira
- Genomics and Health Department, Foundation for the Promotion of Health and Biomedical Research (FISABIO), 46020 Valencia, Spain
| | - Constanza E Fernández
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Karla Gambetta-Tessini
- Cariology Unit, Department of Oral Rehabilitation, Faculty of Dentistry, University of Talca, Talca 3462227, Chile
| | - Carla P Lozano
- Laboratory of Biochemistry and Oral Biology, Institute for Research in Dental Sciences, Faculty of Dentistry, University of Chile, Santiago 8330111, Chile
| |
Collapse
|
21
|
Kang CE, Park YJ, Kim JH, Lee NK, Paik HD. Probiotic Weissella cibaria displays antibacterial and anti-biofilm effect against cavity-causing Streptococcus mutans. Microb Pathog 2023; 180:106151. [PMID: 37172659 DOI: 10.1016/j.micpath.2023.106151] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/24/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Streptococcus mutans is a significant contributor to dental caries and causes functional and aesthetic discomfort. Weissella cibaria strains were isolated from kimchi, and their functional properties were determined. In this study, the antibacterial and antibiofilm effects of four W. cibaria strains (D29, D30, D31, and B22) were evaluated against three S. mutans strains using culture fluid and cell-free supernatants. The results showed that W. cibaria reduced the exopolysaccharides production and auto-aggregation, increased co-aggregation, and downregulated virulence factors, leading to the inhibition of bacterial growth and biofilm formation. These findings were confirmed using scanning electron microscopy and confocal laser scanning microscopy. These results indicate that oral health can be potentially improved by W. cibaria.
Collapse
Affiliation(s)
- Cho Eun Kang
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Yeong Jin Park
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Ji Hun Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Wijesinghe GK, Nobbs AH, Bandara HMHN. Cross-kingdom Microbial Interactions Within the Oral Cavity and Their Implications for Oral Disease. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023. [DOI: 10.1007/s40588-023-00191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Abstract
Purpose of Review
This review serves to highlight the cross-kingdom interactions that can occur within the human oral cavity between fungus Candida albicans and oral bacteria, and their impact on the delicate balance between oral health and disease.
Recent Findings
A growing number of physical, chemical, and metabolic networks have been identified that underpin these cross-kingdom interactions. Moreover, these partnerships are often synergistic and can modulate microbial burden or virulence. This, in turn, can drive the onset or progression of oral diseases such as dental caries, periodontitis, denture-associated stomatitis, and oral cancer.
Summary
The impact of cross-kingdom interactions on the cellular, biochemical, and communal composition of oral microbial biofilms is increasingly clear. With growing insight into these processes at the molecular level, so this knowledge can be used to better inform the development of novel strategies to manipulate the oral microbiota to promote oral health and combat oral disease.
Collapse
|
23
|
Abstract
Candida albicans, a fungus typically found in the mucosal niche, is frequently detected in biofilms formed on teeth (dental plaque) of toddlers with severe childhood caries, a global public health problem that causes rampant tooth decay. However, knowledge about fungal traits on the tooth surface remains limited. Here, we assess the phylogeny, phenotype, and interkingdom interactions of C. albicans isolated from plaque of diseased toddlers and compare their properties to reference strains, including 529L (mucosal isolate). C. albicans isolates exhibit broad phenotypic variations, but all display cariogenic traits, including high proteinase activity, acidogenicity, and acid tolerance. Unexpectedly, we find distinctive variations in filamentous growth, ranging from hyphal defective to hyperfilamentous. We then investigate the ability of tooth isolates to form interkingdom biofilms with Streptococcus mutans (cariogenic partner) and Streptococcus gordonii (mucosal partner). The hyphal-defective isolate lacks cobinding with S. gordonii, but all C. albicans isolates develop robust biofilms with S. mutans irrespective of their filamentation state. Moreover, either type of C. albicans (hyphae defective or hyperfilamentous) enhances sucrose metabolism and biofilm acidogenicity, creating highly acidic environmental pH (<5.5). Notably, C. albicans isolates show altered transcriptomes associated with pH, adhesion, and cell wall composition (versus reference strains), further supporting niche-associated traits. Our data reveal that C. albicans displays distinctive adaptive mechanisms on the tooth surface and develops interactions with pathogenic bacteria while creating an acidogenic state regardless of fungal morphology, contrasting with interkingdom partnerships in mucosal infections. Human tooth may provide new insights into fungal colonization/adaptation, interkingdom biofilms, and contributions to disease pathogenesis. IMPORTANCE Severe early childhood caries is a widespread global public health problem causing extensive tooth decay and systemic complications. Candida albicans, a fungus typically found in mucosal surfaces, is frequently detected in dental plaque formed on teeth of diseased toddlers. However, the clinical traits of C. albicans isolated from tooth remain underexplored. Here, we find that C. albicans tooth isolates exhibit unique biological and transcriptomic traits. Notably, interkingdom biofilms with S. mutans can be formed irrespective of their filamentation state. Furthermore, tooth isolates commonly share dental caries-promoting functions, including acidogenesis, proteolytic activity, and enhanced sugar metabolism, while displaying increased expression of pH-responsive and adhesion genes. Our findings reveal that C. albicans colonizing human teeth displays distinctive adaptive mechanisms to mediate interkingdom interactions associated with a disease-causing state on a mineralized surface, providing new insights into Candida pathobiology and its role in a costly pediatric disease.
Collapse
|
24
|
Gaálová-Radochová B, Kendra S, Jordao L, Kursawe L, Kikhney J, Moter A, Bujdáková H. Effect of Quorum Sensing Molecule Farnesol on Mixed Biofilms of Candida albicans and Staphylococcus aureus. Antibiotics (Basel) 2023; 12:antibiotics12030441. [PMID: 36978309 PMCID: PMC10044556 DOI: 10.3390/antibiotics12030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The natural bioactive molecule farnesol (FAR) is widely studied mainly for its antibiofilm and antimicrobial properties. In addition, it increases the effectiveness of some antimicrobial substances, which makes it interesting for the development of combined therapy. In the present work, the effect of FAR either alone or in combination with oxacillin (OXA) on mixed biofilms formed by clinically relevant pathogens, Candida albicans and Staphylococcus aureus, was studied. S. aureus isolates used for biofilm formation originated from blood cultures and central venous catheters (CVC) were characterized in terms of antimicrobial resistance. The minimal biofilm inhibitory concentration (MBIC50) for FAR of 48 h mixed biofilms formed by the C. albicans and methicillin-sensitive S. aureus (MSSA) was determined to be 125 μM, and for the mixed biofilms with methicillin-resistant S. aureus (MRSA) was determined to be 250 μM. Treatment of mixed biofilms with OXA (2 mg/mL) showed ≤4% inhibition; however, the combination of OXA (2 mg/mL) and FAR (300 μM) resulted in 80% inhibition of biofilms. In addition, planktonic cells of S. aureus exhibited an increased susceptibility to OXA, cefoxitin and kanamycin in the presence of FAR (150 and 300 μM). Scanning electron microscopy (SEM) micrographs confirmed patchy biofilm and lack of candidal hyphae in the samples treated with FAR and FAR/OXA in comparison to control and mixed biofilms treated only with OXA. Intriguingly, in a pilot experiment using fluorescence in situ hybridization (FISH), considerable differences in activity (as indicated by ribosome content) of staphylococcal cells were detected. While the activity rate of the staphylococci in mixed biofilms treated with FAR was high, no FISH-positive signal for staphylococcal cells was found in the biofilm treated with FAR/OXA.
Collapse
Affiliation(s)
- Barbora Gaálová-Radochová
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-2-9014-9480
| | - Samuel Kendra
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| | - Luisa Jordao
- Department of Environmental Health, Research and Development Unit, National Institute of Health Dr. Ricardo Jorge (INSA), Av. Padre Cruz, 1649-016 Lisboa, Portugal
| | - Laura Kursawe
- Biofilmcenter, Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- MoKi Analytics GmbH, Charité-Universitätsmedizin Berlin, Hindenburdamm 30, 12203 Berlin, Germany
| | - Judith Kikhney
- Biofilmcenter, Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- MoKi Analytics GmbH, Charité-Universitätsmedizin Berlin, Hindenburdamm 30, 12203 Berlin, Germany
| | - Annette Moter
- Biofilmcenter, Institute of Microbiology, Infectious Diseases and Immunology, Charité—Universitätsmedizin Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- MoKi Analytics GmbH, Charité-Universitätsmedizin Berlin, Hindenburdamm 30, 12203 Berlin, Germany
- Moter Diagnostics, Marienplatz 9, 12207 Berlin, Germany
| | - Helena Bujdáková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, 842 15 Bratislava, Slovakia
| |
Collapse
|
25
|
Gu M, Cho JH, Suh JW, Cheng J. Potential oral probiotic Lactobacillus pentosus MJM60383 inhibits Streptococcus mutans biofilm formation by inhibiting sucrose decomposition. J Oral Microbiol 2022; 15:2161179. [PMID: 36605406 PMCID: PMC9809368 DOI: 10.1080/20002297.2022.2161179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
Streptococcus mutans is known as a contributor to dental caries. In this work, Lactobacillus pentosus MJM60383 was selected for its strong antagonistic activity against S. mutans and was characterized by good oral probiotic properties including lysozyme tolerance, adhesive ability to oral cells, good aggregation (auto-aggregation, co-aggregation) ability, hydrogen peroxide production and inhibition of biofilm formation of S. mutans. L. pentosus MJM60383 also exhibited safety as a probiotic characterized by no hemolytic activity, no D-lactate production, no biogenic amine production, and susceptibility to antibiotics. Furthermore, the biofilm formation of S. mutans was also significantly inhibited by the supernatant of L. pentosus MJM60383. An anti-biofilm mechanism study revealed that sucrose decomposition and the production of water-insoluble exopolysaccharides by S. mutans were inhibited by the treatment with L. pentosus MJM60383 supernatant. Real-time PCR analysis indicated that the supernatant of L. pentosus MJM60383 significantly inhibited the mRNA expression of S. mutans glycosyltransferases, which synthesize glucan to construct biofilm architecture and mediate bacterial adherence. Our study demonstrated L. pentosus MJM60383 as a potential oral probiotic and revealed its anti-biofilm mechanism.
Collapse
Affiliation(s)
- Mingkun Gu
- Interdisciplinary Program of Biomodulation, Myongji University, Yongin, Republic of Korea
| | - Joo-Hyung Cho
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| | - Joo-Won Suh
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| | - Jinhua Cheng
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| |
Collapse
|
26
|
Kováč J, Slobodníková L, Trajčíková E, Rendeková K, Mučaji P, Sychrová A, Bittner Fialová S. Therapeutic Potential of Flavonoids and Tannins in Management of Oral Infectious Diseases-A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 28:molecules28010158. [PMID: 36615352 PMCID: PMC9821998 DOI: 10.3390/molecules28010158] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/18/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022]
Abstract
Medicinal plants are rich sources of valuable molecules with various profitable biological effects, including antimicrobial activity. The advantages of herbal products are their effectiveness, relative safety based on research or extended traditional use, and accessibility without prescription. Extensive and irrational usage of antibiotics since their discovery in 1928 has led to the increasing expiration of their effectiveness due to antibacterial resistance. Now, medical research is facing a big and challenging mission to find effective and safe antimicrobial therapies to replace inactive drugs. Over the years, one of the research fields that remained the most available is the area of natural products: medicinal plants and their metabolites, which could serve as active substances to fight against microbes or be considered as models in drug design. This review presents selected flavonoids (such as apigenin, quercetin, kaempferol, kurarinone, and morin) and tannins (including oligomeric proanthocyanidins, gallotannins, ellagitannins, catechins, and epigallocatechin gallate), but also medicinal plants rich in these compounds as potential therapeutic agents in oral infectious diseases based on traditional usages such as Agrimonia eupatoria L., Hamamelis virginiana L., Matricaria chamomilla L., Vaccinium myrtillus L., Quercus robur L., Rosa gallica L., Rubus idaeus L., or Potentilla erecta (L.). Some of the presented compounds and extracts are already successfully used to maintain oral health, as the main or additive ingredient of toothpastes or mouthwashes. Others are promising for further research or future applications.
Collapse
Affiliation(s)
- Ján Kováč
- Department of Stomatology and Maxillofacial Surgery, Faculty of Medicine, Comenius University in Bratislava, Heydukova 10, 812 50 Bratislava, Slovakia
- Department of Stomatology and Maxillofacial Surgery, St. Elizabeth’s Hospital, Heydukova 10, 812 50 Bratislava, Slovakia
| | - Lívia Slobodníková
- Institute of Microbiology, Faculty of Medicine and the University Hospital in Bratislava, Comenius University in Bratislava, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Eva Trajčíková
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
| | - Katarína Rendeková
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
| | - Pavel Mučaji
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
| | - Alice Sychrová
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého 1946/1, 612 00 Brno, Czech Republic
| | - Silvia Bittner Fialová
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University in Bratislava, Odbojárov 10, 832 32 Bratislava, Slovakia
- Correspondence: ; Tel.: +421-250-117-206
| |
Collapse
|
27
|
Negrini TDC, Ren Z, Miao Y, Kim D, Simon-Soro Á, Liu Y, Koo H, Arthur RA. Dietary sugars modulate bacterial-fungal interactions in saliva and inter-kingdom biofilm formation on apatitic surface. Front Cell Infect Microbiol 2022; 12:993640. [PMID: 36439211 PMCID: PMC9681999 DOI: 10.3389/fcimb.2022.993640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Bacteria and fungi can interact to form inter-kingdom biofilms in the oral cavity. Streptococcus mutans and Candida albicans are frequently detected in saliva and in dental biofilms associated with early childhood caries (tooth-decay), a prevalent oral disease induced by dietary sugars. However, how different sugars influence this bacterial-fungal interaction remains unclear. Here, we investigate whether specific sugars affect the inter-kingdom interaction in saliva and subsequent biofilm formation on tooth-mimetic surfaces. The microbes were incubated in saliva containing common dietary sugars (glucose and fructose, sucrose, starch, and combinations) and analyzed via fluorescence imaging and quantitative computational analyses. The bacterial and fungal cells in saliva were then transferred to hydroxyapatite discs (tooth mimic) to allow microbial binding and biofilm development. We found diverse bacterial-fungal aggregates which varied in size, structure, and spatial organization depending on the type of sugars. Sucrose and starch+sucrose induced the formation of large mixed-species aggregates characterized by bacterial clusters co-bound with fungal cells, whereas mostly single-cells were found in the absence of sugar or in the presence of glucose and fructose. Notably, both colonization and further growth on the apatitic surface were dependent on sugar-mediated aggregation, leading to biofilms with distinctive spatial organizations and 3D architectures. Starch+sucrose and sucrose-mediated aggregates developed into large and highly acidogenic biofilms with complex network of bacterial and fungal cells (yeast and hyphae) surrounded by an intricate matrix of extracellular glucans. In contrast, biofilms originated from glucose and fructose-mediated consortia (or without sugar) were sparsely distributed on the surface without structural integration, growing predominantly as individual species with reduced acidogenicity. These findings reveal the impact of dietary sugars on inter-kingdom interactions in saliva and how they mediate biofilm formation with distinctive structural organization and varying acidogenicity implicated with human tooth-decay.
Collapse
Affiliation(s)
- Thais de Cássia Negrini
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Araraquara, Brazil
| | - Zhi Ren
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yilan Miao
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dongyeop Kim
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Preventive Dentistry, School of Dentistry, and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, South Korea
| | - Áurea Simon-Soro
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Stomatology, School of Dentistry, University of Seville, Seville, Spain
| | - Yuan Liu
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Preventive & Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hyun Koo
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rodrigo Alex Arthur
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Preventive and Community Dentistry, Dental School, Federal University of Rio Grande do Sul, Porto Alegre, Brazil,*Correspondence: Rodrigo Alex Arthur,
| |
Collapse
|
28
|
Ito T, Sims KR, Liu Y, Xiang Z, Arthur RA, Hara AT, Koo H, Benoit DSW, Klein MI. Farnesol delivery via polymeric nanoparticle carriers inhibits cariogenic cross-kingdom biofilms and prevents enamel demineralization. Mol Oral Microbiol 2022; 37:218-228. [PMID: 35859523 PMCID: PMC9529802 DOI: 10.1111/omi.12379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 07/01/2022] [Accepted: 07/14/2022] [Indexed: 11/29/2022]
Abstract
Streptococcus mutans and Candida albicans are frequently detected together in the plaque from patients with early childhood caries (ECC) and synergistically interact to form a cariogenic cross-kingdom biofilm. However, this biofilm is difficult to control. Thus, to achieve maximal efficacy within the complex biofilm microenvironment, nanoparticle carriers have shown increased interest in treating oral biofilms in recent years. Here, we assessed the anti-biofilm efficacy of farnesol (Far), a hydrophobic antibacterial drug and repressor of Candida filamentous forms, against cross-kingdom biofilms employing drug delivery via polymeric nanoparticle carriers (NPCs). We also evaluated the effect of the strategy on teeth enamel demineralization. The farnesol-loaded NPCs (NPC+Far) resulted in a 2-log CFU/mL reduction of S. mutans and C. albicans (hydroxyapatite disc biofilm model). High-resolution confocal images further confirmed a significant reduction in exopolysaccharides, smaller microcolonies of S. mutans, and no hyphal form of C. albicans after treatment with NPC+Far on human tooth enamel (HT) slabs, altering the biofilm 3D structure. Furthermore, NPC+Far treatment was highly effective in preventing enamel demineralization on HT, reducing lesion depth (79% reduction) and mineral loss (85% reduction) versus vehicle PBS-treated HT, while NPC or Far alone had no differences with the PBS. The drug delivery via polymeric NPCs has the potential for targeting bacterial-fungal biofilms associated with a prevalent and costly pediatric oral disease, such as ECC.
Collapse
Affiliation(s)
- Tatsuro Ito
- Department of Pediatric Dentistry, Nihon University School of Dentistry at Matsudo, Chiba, Japan
- Biofilm Research Labs, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth R. Sims
- Department of Translational Biomedical Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Yuan Liu
- Biofilm Research Labs, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenting Xiang
- Biofilm Research Labs, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rodrigo A. Arthur
- Preventive and Community Dentistry Department, Dental School, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Anderson T. Hara
- Department of Cariology, Operative Dentistry and Dental Public Health, Oral Health Research Institute, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Hyun Koo
- Biofilm Research Labs, Levy Center for Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Danielle S. W. Benoit
- Department of Biomedical Engineering, Department of Chemical Engineering, Materials Science Program, University of Rochester, Rochester, NY, USA
| | - Marlise I. Klein
- Department of Dental Materials and Prosthodontics, São Paulo State University (UNESP), School of Dentistry, Araraquara, São Paulo, Brazil
| |
Collapse
|
29
|
Li H, Miao MX, Jia CL, Cao YB, Yan TH, Jiang YY, Yang F. Interactions between Candida albicans and the resident microbiota. Front Microbiol 2022; 13:930495. [PMID: 36204612 PMCID: PMC9531752 DOI: 10.3389/fmicb.2022.930495] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/31/2022] [Indexed: 01/09/2023] Open
Abstract
Candida albicans is a prevalent, opportunistic human fungal pathogen. It usually dwells in the human body as a commensal, however, once in its pathogenic state, it causes diseases ranging from debilitating superficial to life-threatening systemic infections. The switch from harmless colonizer to virulent pathogen is, in most cases, due to perturbation of the fungus-host-microbiota interplay. In this review, we focused on the interactions between C. albicans and the host microbiota in the mouth, gut, blood, and vagina. We also highlighted important future research directions. We expect that the evaluation of these interplays will help better our understanding of the etiology of fungal infections and shed new light on the therapeutic approaches.
Collapse
Affiliation(s)
- Hao Li
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ming-xing Miao
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Cheng-lin Jia
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-bing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-hua Yan
- Department of Physiology and Pharmacology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China,*Correspondence: Tian-hua Yan,
| | - Yuan-ying Jiang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Yuan-ying Jiang,
| | - Feng Yang
- Department of Pharmacy, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China,Feng Yang,
| |
Collapse
|
30
|
Díaz-Garrido N, Lozano CP, Kreth J, Giacaman RA. Extended biofilm formation time by Streptococcus sanguinis modifies its non-cariogenic behavior, in vitro. Braz Oral Res 2022; 36:e107. [PMID: 35946735 DOI: 10.1590/1807-3107bor-2022.vol36.0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
Abstract
Although the commensal Streptococcus sanguinis [ S. sanguinis] is isolated from caries-free people, it can ferment carbohydrates producing acids. We aimed to characterize S. sanguinis cariogenic potential as a function of different enamel biofilm formation periods, in vitro. Saliva-coated enamel slabs were inoculated with S. sanguinis to form initial biofilms for 8, 12 or 16 h in presence of sucrose and followed by a period in medium with glucose for 16, 12 or 8 h, respectively, until completion of 24 h. To simulate cariogenic challenges, S. sanguinis biofilms were exposed to 10% sucrose for 5 minutes, 3x/day for 5 days. Biofilm biomass, viable cells, total proteins, intracellular and extracellular polysaccharides production, acidogenicity and enamel demineralization were determined. Biofilms of Streptococcus mutans [ S. mutans ] served as caries-positive control. Biofilms of S. sanguinis forming on enamel for 12 and 16 h showed higher demineralization than those formed during 8 h, but lower than S. mutans biofilms, regardless of the initial biofilm formation time. No differences were detected in the biofilm properties among the different biofilm formation times tested for S. sanguinis . Increased enamel initial biofilm formation time by S. sanguinis appears to induce a cariogenic potential, but lower than S. mutans .
Collapse
Affiliation(s)
- Natalia Díaz-Garrido
- University of Talca , Cariology Unit , Department of Oral Rehabilitation , Talca , Chile
| | - Carla Paola Lozano
- Universidad de Chile , Institute for Research in Dental Sciences , Faculty of Dentistry , Laboratory of Biochemistry and Oral Biology, Santiago , Chile
| | - Jens Kreth
- Oregon Health & Science University , Department of Restorative Dentistry , Portland , OR , USA
| | - Rodrigo Andrés Giacaman
- Interuniversity Center on Healthy Aging , Chilean State Universities Consortium , Santiago , Chile
| |
Collapse
|
31
|
Ivanova A, Ivanova K, Fiandra L, Mantecca P, Catelani T, Natan M, Banin E, Jacobi G, Tzanov T. Antibacterial, Antibiofilm, and Antiviral Farnesol-Containing Nanoparticles Prevent Staphylococcus aureus from Drug Resistance Development. Int J Mol Sci 2022; 23:ijms23147527. [PMID: 35886883 PMCID: PMC9321328 DOI: 10.3390/ijms23147527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022] Open
Abstract
Multidrug antimicrobial resistance is a constantly growing health care issue associated with increased mortality and morbidity, and huge financial burden. Bacteria frequently form biofilm communities responsible for numerous persistent infections resistant to conventional antibiotics. Herein, novel nanoparticles (NPs) loaded with the natural bactericide farnesol (FSL NPs) are generated using high-intensity ultrasound. The nanoformulation of farnesol improved its antibacterial properties and demonstrated complete eradication of Staphylococcus aureus within less than 3 h, without inducing resistance development, and was able to 100% inhibit the establishment of a drug-resistant S. aureus biofilm. These antibiotic-free nano-antimicrobials also reduced the mature biofilm at a very low concentration of the active agent. In addition to the outstanding antibacterial properties, the engineered nano-entities demonstrated strong antiviral properties and inhibited the spike proteins of SARS-CoV-2 by up to 83%. The novel FSL NPs did not cause skin tissue irritation and did not induce the secretion of anti-inflammatory cytokines in a 3D skin tissue model. These results support the potential of these bio-based nano-actives to replace the existing antibiotics and they may be used for the development of topical pharmaceutic products for controlling microbial skin infections, without inducing resistance development.
Collapse
Affiliation(s)
- Aleksandra Ivanova
- Group of Molecular and Industrial Biotechnology, Chemical Engineering, Universitat Politécnica de Catalunya, 08222 Terrassa, Spain; (A.I.); (K.I.)
| | - Kristina Ivanova
- Group of Molecular and Industrial Biotechnology, Chemical Engineering, Universitat Politécnica de Catalunya, 08222 Terrassa, Spain; (A.I.); (K.I.)
| | - Luisa Fiandra
- Department of Earth and Environmental Sciences, Research Center POLARIA, Universita degli Studi di Milano-Bicocca, 20900 Milano, Italy; (L.F.); (P.M.)
| | - Paride Mantecca
- Department of Earth and Environmental Sciences, Research Center POLARIA, Universita degli Studi di Milano-Bicocca, 20900 Milano, Italy; (L.F.); (P.M.)
| | - Tiziano Catelani
- Interdepartmental Microscopy Platform, University of Milano-Bicocca, 20126 Milano, Italy;
| | - Michal Natan
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (M.N.); (E.B.); (G.J.)
| | - Ehud Banin
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (M.N.); (E.B.); (G.J.)
| | - Gila Jacobi
- The Institute for Advanced Materials and Nanotechnology, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (M.N.); (E.B.); (G.J.)
| | - Tzanko Tzanov
- Group of Molecular and Industrial Biotechnology, Chemical Engineering, Universitat Politécnica de Catalunya, 08222 Terrassa, Spain; (A.I.); (K.I.)
- Correspondence:
| |
Collapse
|
32
|
Bezerra SJC, Viana ÍEL, Aoki IV, Duarte S, Hara AT, Scaramucci T. In-vitro evaluation of the anti-cariogenic effect of a hybrid coating associated with encapsulated sodium fluoride and stannous chloride in nanoclays on enamel. J Appl Oral Sci 2022; 30:e20210643. [PMID: 35507984 PMCID: PMC9064272 DOI: 10.1590/1678-7757-2021-0643] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/21/2022] [Accepted: 03/02/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The aim of this study is to test, in vitro, the anti-cariogenic effect of experimental hybrid coatings, with nano clays of halloysite or bentonite, loaded with sodium fluoride or with a combination of sodium fluoride and stannous chloride, respectively. METHODOLOGY The varnish Fluor Protector (1,000 ppm of F-) was used as positive control and no treatment was the negative control. Enamel specimens (5 mm × 5 mm) were obtained from bovine teeth. The specimens (n=10) had their surfaces divided into two halves (5 mm × 2.5 mm each), in which one half received one of the treatments (Hybrid; Hybrid + NaF; Hybrid + NaF + SnCl2; Hybrid + NaF Loaded; Hybrid + NaF + SnCl2 Loaded). The specimens were submitted to a cariogenic challenge using a biofilm model (S. mutans UA159, for 5 days). Enamel surfaces both under and adjacent to the treated area were analyzed for mineral loss and lesion depth, by transverse microradiography. The pH of the medium was measured twice a day, and the fluoride release was analyzed. Additional specimens were submitted to confocal analysis. RESULTS Data were statistically analyzed by two-way ANOVA followed by Tukey test (α=0.05). None of hybrid groups were able to reduce the lesion depth; the Hybrid + NaF group, however, was able to reduce mineral loss differing from the negative control (p=0.008). The groups showed no significant difference in the pH measurement and fluoride release. Confocal analysis confirmed that for all groups the biofilm growth was similar. CONCLUSION None of the hybrid groups reduced lesion depth, but the Hybrid + NaF group was able to promote protection against mineral loss.
Collapse
Affiliation(s)
- Sávio José Cardoso Bezerra
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Dentística, São Paulo, Brasil
- Indiana University School of Dentistry, Operative Dentistry and Dental Public Health, Department of Cariology, Indianapolis, United States
| | - Ítallo Emídio Lira Viana
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Dentística, São Paulo, Brasil
| | - Idalina Vieira Aoki
- Universidade de São Paulo, Escola Politécnica, Departamento de Engenharia Química, São Paulo, Brasil
| | - Simone Duarte
- Indiana University School of Dentistry, Operative Dentistry and Dental Public Health, Department of Cariology, Indianapolis, United States
| | - Anderson Takeo Hara
- Indiana University School of Dentistry, Operative Dentistry and Dental Public Health, Department of Cariology, Indianapolis, United States
| | - Taís Scaramucci
- Universidade de São Paulo, Faculdade de Odontologia, Departamento de Dentística, São Paulo, Brasil
| |
Collapse
|
33
|
Singla RK, Dhir V, Madaan R, Kumar D, Singh Bola S, Bansal M, Kumar S, Dubey AK, Singla S, Shen B. The Genus Alternanthera: Phytochemical and Ethnopharmacological Perspectives. Front Pharmacol 2022; 13:769111. [PMID: 35479320 PMCID: PMC9036189 DOI: 10.3389/fphar.2022.769111] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Ethnopharmacological relevance: The genus Alternanthera (Amaranthaceae) comprises 139 species including 14 species used traditionally for the treatment of various ailments such as hypertension, pain, inflammation, diabetes, cancer, microbial and mental disorders. Aim of the review: To search research gaps through critical assessment of pharmacological activities not performed to validate traditional claims of various species of Alternanthera. This review will aid natural product researchers in identifying Alternanthera species with therapeutic potential for future investigation. Materials and methods: Scattered raw data on ethnopharmacological, morphological, phytochemical, pharmacological, toxicological, and clinical studies of various species of the genus Alternanthera have been compiled utilizing search engines like SciFinder, Google Scholar, PubMed, Science Direct, and Open J-Gate for 100 years up to April 2021. Results: Few species of Alternanthera genus have been exhaustively investigated phytochemically, and about 129 chemical constituents related to different classes such as flavonoids, steroids, saponins, alkaloids, triterpenoids, glycosides, and phenolic compounds have been isolated from 9 species. Anticancer, antioxidant, antibacterial, CNS depressive, antidiabetic, analgesic, anti-inflammatory, and immunomodulator effects have been explored in the twelve species of the genus. A toxicity study has been conducted on 3 species and a clinical study on 2 species. Conclusions: The available literature on pharmacological studies of Alternanthera species reveals that few species have been selected based on ethnobotanical surveys for scientific validation of their traditional claims. But most of these studies have been conducted on uncharacterized and non-standardized crude extracts. A roadmap of research needs to be developed for the isolation of new bioactive compounds from Alternanthera species, which can emerge out as clinically potential medicines.
Collapse
Affiliation(s)
- Rajeev K. Singla
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Vivek Dhir
- Chitkara College of Pharmacy, Chitkara University Punjab, Rajpura, India
| | - Reecha Madaan
- Chitkara College of Pharmacy, Chitkara University Punjab, Rajpura, India
- *Correspondence: Bairong Shen, ; Reecha Madaan,
| | - Deepak Kumar
- Department of Health and Family Welfare, Civil Hospital, Rampura Phul, India
| | - Simranjit Singh Bola
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, India
| | - Monika Bansal
- Akal College of Pharmacy and Technical Education, Mastuana Sahib, Sangrur, India
| | - Suresh Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | - Shailja Singla
- iGlobal Research and Publishing Foundation, New Delhi, India
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Bairong Shen, ; Reecha Madaan,
| |
Collapse
|
34
|
Ghosh S, Nag M, Lahiri D, Sarkar T, Pati S, Joshi S, Ray RR. New holistic approach for the management of biofilm‐associated infections by myco‐metabolites. J Basic Microbiol 2022; 62:1291-1306. [PMID: 35373364 DOI: 10.1002/jobm.202200047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/23/2022] [Accepted: 03/05/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Sreejita Ghosh
- Department of Biotechnology Maulana Abul Kalam Azad University of Technology Haringhata West Bengal India
| | - Moupriya Nag
- Department of Biotechnology University of Engineering & Management Kolkata West Bengal India
| | - Dibyajit Lahiri
- Department of Biotechnology University of Engineering & Management Kolkata West Bengal India
| | - Tanmay Sarkar
- Department of Food Processing Technology Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal Malda India
| | - Siddhartha Pati
- Skills innovation & Academic network (SIAN) Institute‐ABC Balasore Odisha India
- NatNov Bioscience Private Limited Balasore Odisha India
| | - Sanket Joshi
- Oil & Gas Research Center, Central Analytical and Applied Research Unit Sultan Qaboos University Maskat Oman
| | - Rina R. Ray
- Department of Biotechnology Maulana Abul Kalam Azad University of Technology Haringhata West Bengal India
| |
Collapse
|
35
|
Wieczorek PP, Hudz N, Yezerska O, Horčinová-Sedláčková V, Shanaida M, Korytniuk O, Jasicka-Misiak I. Chemical Variability and Pharmacological Potential of Propolis as a Source for the Development of New Pharmaceutical Products. Molecules 2022; 27:1600. [PMID: 35268700 PMCID: PMC8911684 DOI: 10.3390/molecules27051600] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 11/23/2022] Open
Abstract
This review aims to analyze propolis as a potential raw material for the development and manufacture of new health-promoting products. Many scientific publications were retrieved from the Scopus, PubMed, and Google Scholar databases via searching the word "propolis". The different extraction procedures, key biologically active compounds, biological properties, and therapeutic potential of propolis were analyzed. It was concluded that propolis possesses a variety of biological properties because of a very complex chemical composition that mainly depends on the plant species visited by bees and species of bees. Numerous studies found versatile pharmacological activities of propolis: antimicrobial, antifungal, antiviral, antioxidant, anticancer, anti-inflammatory, immunomodulatory, etc. In this review, the composition and biological activities of propolis are presented from a point of view of the origin and standardization of propolis for the purpose of the development of new pharmaceutical products on its base. It was revealed that some types of propolis, especially European propolis, contain flavonoids and phenolic acids, which could be markers for the standardization and quality evaluation of propolis and its preparations. One more focus of this paper was the overview of microorganisms' sensitivity to propolis for further development of antimicrobial and antioxidant products for the treatment of various infectious diseases with an emphasis on the illnesses of the oral cavity. It was established that the antimicrobial activity of different types of propolis is quite significant, especially to Gram-negative bacteria and lipophilic viruses. The present study could be also of interest to the pharmaceutical industry as a review for the appropriate design of standardized propolis preparations such as mouthwashes, toothpastes, oral drops, sprays, creams, ointments, suppositories, tablets, and capsules, etc. Moreover, propolis could be regarded as a source for the isolation of biologically active substances. Furthermore, this review can facilitate partially overcoming the problem of the standardization of propolis preparations, which is a principal obstacle to the broader use of propolis in the pharmaceutical industry. Finally, this study could be of interest in the area of the food industry for the development of nutritionally well-balanced products. The results of this review indicate that propolis deserves to be better studied for its promising therapeutic effects from the point of view of the connection of its chemical composition with the locality of its collection, vegetation, appropriate extraction methods, and standardization.
Collapse
Affiliation(s)
| | - Nataliia Hudz
- Department of Drug Technology and Biopharmacy, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (N.H.); (O.Y.)
- Department of Pharmacy and Ecological Chemistry, University of Opole, 45-052 Opole, Poland;
| | - Oksana Yezerska
- Department of Drug Technology and Biopharmacy, Danylo Halytsky Lviv National Medical University, 79010 Lviv, Ukraine; (N.H.); (O.Y.)
| | | | - Mariia Shanaida
- Department of Pharmacognosy and Medical Botany, I. Horbachevsky Ternopil National Medical University, 46001 Ternopil, Ukraine;
| | - Oleksii Korytniuk
- Department of Occupational and Facial Surgery and Dentistry, Ukrainian Military Medical Academy, 01015 Kyiv, Ukraine;
| | - Iza Jasicka-Misiak
- Department of Pharmacy and Ecological Chemistry, University of Opole, 45-052 Opole, Poland;
| |
Collapse
|
36
|
Yang S, Lyu X, Zhang J, Shui Y, Yang R, Xu X. The Application of Small Molecules to the Control of Typical Species Associated With Oral Infectious Diseases. Front Cell Infect Microbiol 2022; 12:816386. [PMID: 35265531 PMCID: PMC8899129 DOI: 10.3389/fcimb.2022.816386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/28/2022] [Indexed: 12/12/2022] Open
Abstract
Oral microbial dysbiosis is the major causative factor for common oral infectious diseases including dental caries and periodontal diseases. Interventions that can lessen the microbial virulence and reconstitute microbial ecology have drawn increasing attention in the development of novel therapeutics for oral diseases. Antimicrobial small molecules are a series of natural or synthetic bioactive compounds that have shown inhibitory effect on oral microbiota associated with oral infectious diseases. Novel small molecules, which can either selectively inhibit keystone microbes that drive dysbiosis of oral microbiota or inhibit the key virulence of the microbial community without necessarily killing the microbes, are promising for the ecological management of oral diseases. Here we discussed the research progress in the development of antimicrobial small molecules and delivery systems, with a particular focus on their antimicrobial activity against typical species associated with oral infectious diseases and the underlying mechanisms.
Collapse
Affiliation(s)
- Sirui Yang
- 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
| | - Xiaoying Lyu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, 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
| | - Yusen Shui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ran Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - 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
|
37
|
Oliveira JMDS, Cavalcanti TFS, Leite IF, Dos Santos DMRC, Porto ICCDM, de Aquino FLT, Sonsin AF, Lins RML, Vitti RP, de Freitas JD, Barreto EDO, de Souza ST, Kamiya RU, do Nascimento TG, Tonholo J. Propolis in Oral Healthcare: Antibacterial Activity of a Composite Resin Enriched With Brazilian Red Propolis. Front Pharmacol 2021; 12:787633. [PMID: 34912230 PMCID: PMC8667603 DOI: 10.3389/fphar.2021.787633] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/03/2021] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to obtain a Brazilian red propolis (BRP) enriched composite resin and to perform the characterization of its antibacterial activity, mechanical, and physical-chemical properties. Brazilian red propolis ethyl acetate extract (EABRP) was characterized by LC-ESI-Orbitrap-FTMS, UPLC-DAD, antibacterial activity, total flavonoids content, and radical scavenging capacity. BRP was incorporated to a commercial composite resin (RC) to obtain BRP enriched composite at 0.1, 0.15 and 0.25% (RP10, RP15 and RP25, respectively). The antibacterial activity RPs was evaluated against Streptococcus mutans by contact direct test and expressed by antibacterial ratio. The RPs were characterized as its cytotoxicity against 3T3 fibroblasts, flexural strength (FS), Knoop microhardness (KHN), post-cure depth (CD), degree of conversion (DC%), water sorption (Wsp), water solubility (Wsl), average roughness (Ra), and thermal analysis. Were identified 50 chemical compounds from BRP extract by LC-ESI-Orbitrap-FTMS. EABRP was bacteriostatic and bactericide at 125 and 500 μg/ml, respectively. The RP25 exhibited antibacterial ratio of 90.76% after 1 h of direct contact with S. mutans (p < 0.0001) while RC no showed significative antibacterial activity (p = 0.1865), both compared with cell control group. RPs and RC no showed cytotoxicity. RPs exhibited CD from 2.74 to 4.48 mm, DC% from 80.70 to 83.96%, Wsp from 17.15 to 21.67 μg/mm3, Wsl from 3.66 to 4.20 μg/mm3, Ra from 14.48 to 20.76 nm. RPs showed thermal resistance between 448–455°C. The results support that propolis can be used on development of modified composite resins that show antibacterial activity and that have compatible mechanical and physical-chemical properties to the indicate for composite resins.
Collapse
Affiliation(s)
- José Marcos Dos Santos Oliveira
- Postgraduate Program of Chemistry and Biotechnology, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil.,Postgraduate Program in Health Research, Cesmac University Center, Maceió, Brazil
| | - Théo Fortes Silveira Cavalcanti
- Postgraduate Program in Materials, Center of Technology, Federal University of Alagoas, Maceió, Brazil.,Faculty of Dentistry, Federal University of Alagoas, Maceió, Brazil
| | | | | | - Isabel Cristina Celerino de Moraes Porto
- Faculty of Dentistry, Federal University of Alagoas, Maceió, Brazil.,Postgraduate Program in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Fernanda Lima Torres de Aquino
- Postgraduate Program in Health Sciences, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Artur Falqueto Sonsin
- Postgraduate Program in Physics, Institute of Physics, Federal University of Alagoas, Maceió, Brazil
| | | | | | | | - Emiliano de Oliveira Barreto
- Postgraduate Program in Health Sciences, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Samuel Teixeira de Souza
- Postgraduate Program in Physics, Institute of Physics, Federal University of Alagoas, Maceió, Brazil
| | - Regianne Umeko Kamiya
- Postgraduate Program in Health Sciences, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Ticiano Gomes do Nascimento
- Postgraduate Program in Pharmaceutical Sciences, Institute of Pharmaceutical Sciences, Federal University of Alagoas, Maceió, Brazil
| | - Josealdo Tonholo
- Postgraduate Program of Chemistry and Biotechnology, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil
| |
Collapse
|
38
|
Liu D, Ma X, Ji Y, Chen R, Zhou S, Yao H, Zhang Z, Ye M, Xu Z, Du M. Bioresponsive nanotherapy for preventing dental caries by inhibiting multispecies cariogenic biofilms. Bioact Mater 2021; 14:1-14. [PMID: 35310362 PMCID: PMC8891616 DOI: 10.1016/j.bioactmat.2021.12.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/15/2022] Open
Abstract
Early childhood caries (ECC) is a public healthcare concern that greatly reduces the quality of life of young children. As a leading factor of ECC, cariogenic biofilms are composed of acidogenic/aciduric pathogens and extracellular polysaccharides (EPSs), creating an acidic and protected microenvironment. Antimicrobial photodynamic therapy (aPDT) is a noninvasive, painless, and efficient therapeutic approach that is suitable for treating ECC. However, due to the hyperfine structure of cariogenic biofilms, most photosensitizers (PSs) could not access and penetrate deeply in biofilms, which dramatically hamper their efficiency in the clinic. Herein, bioresponsive nanoparticle loaded with chlorin e6 (MPP-Ce6) is developed, which largely increases the penetration depth (by over 75%) and retention (by over 100%) of PS in the biofilm compared with free Ce6. Furthermore, MPP-Ce6-mediated aPDT not only kills the bacteria in preformed biofilms but also inhibits multispecies biofilm formation. A rampant caries model is established to mimic ECC in vivo, where the population of cariogenic bacteria is decreased to 10% after MPP-Ce6-mediated aPDT. Importantly, the number and severity of carious lesions are efficiently reduced via Keyes’ scoring and micro-CT analysis. This simple but effective strategy can serve as a promising approach for daily oral hygiene in preventing ECC. A pH-responsive nano-system is developed for biofilm-targeted drug delivery. The nano-system could overcome biological barriers and penetrate deeply in biofilms. This nano-system facilitates aPDT to kill bacteria in deep cariogenic biofilm. This strategy prevents the progression of early childhood caries in a rat model.
Collapse
Affiliation(s)
- Danfeng Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Xianbin Ma
- School of Materials and Energy & Chongqing Engineering Research Center for MicroNano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Yaoting Ji
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Rourong Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Shuhui Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Hantao Yao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Zichen Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Mengjie Ye
- School of Materials and Energy & Chongqing Engineering Research Center for MicroNano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Zhigang Xu
- School of Materials and Energy & Chongqing Engineering Research Center for MicroNano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
- Corresponding author.
| | - Minquan Du
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
- Corresponding author.,
| |
Collapse
|
39
|
Roncari Rocha G, Sims KR, Xiao B, Klein MI, Benoit DS. Nanoparticle carrier co-delivery of complementary antibiofilm drugs abrogates dual species cariogenic biofilm formation in vitro. J Oral Microbiol 2021; 14:1997230. [PMID: 34868474 PMCID: PMC8635615 DOI: 10.1080/20002297.2021.1997230] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Dental caries is a multifactorial disease caused by pathogenic biofilm. In particular, Streptococcus mutans synthesizes biofilm exopolysaccharides, while Candida albicans is associated with the development of severe carious lesions. AIM This study aimed to prevent the formation of S. mutans and C. albicans biofilms by exploiting pH-sensitive nanoparticle carriers (NPCs) with high affinity to exopolysaccharides to increase the substantivity of multi-targeted antibiofilm drugs introduced topically in vitro. METHODS Dual-species biofilms were grown on saliva-coated hydroxyapatite discs with sucrose. Twice-daily, 1.5 min topical treatment regimens of unloaded and drug-loaded NPC were used. Drugs included combinations of two or three compounds with distinct, complementary antibiofilm targets: tt-farnesol (terpenoid; bacterial acid tolerance, fungal quorum sensing), myricetin (flavonoid; exopolysaccharides inhibitor), and 1771 (lipoteichoic acid inhibitor; bacterial adhesion and co-aggregation). Biofilms were evaluated for biomass, microbial population, and architecture. RESULTS NPC delivering tt-farnesol and 1771 with or without myricetin completely prevented biofilm formation by impeding biomass accumulation, bacterial and fungal population growth, and exopolysaccharide matrix deposition (vs. control unloaded NPC). Both formulations hindered acid production, maintaining the pH of spent media above the threshold for enamel demineralization. However, treatments had no effect on pre-established dual-species biofilms. CONCLUSION Complementary antibiofilm drug-NPC treatments prevented biofilm formation by targeting critical virulence factors of acidogenicity and exopolysaccharides synthesis.
Collapse
Affiliation(s)
- Guilherme Roncari Rocha
- Department of Dental Materials and Prosthodontics, São Paulo State University, São Paulo, Brazil
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Kenneth R. Sims
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Marlise I. Klein
- Department of Dental Materials and Prosthodontics, São Paulo State University, São Paulo, Brazil
| | - Danielle S.W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Materials Science Program, University of Rochester, Ny, USA
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester, Ny, USA
- Center for Oral Biology, University of Rochester, NY, USA
- Department of Chemical Engineering, University of Rochester, Ny, USA
| |
Collapse
|
40
|
Yang S, Zhang J, Yang R, Xu X. Small Molecule Compounds, A Novel Strategy against Streptococcus mutans. Pathogens 2021; 10:pathogens10121540. [PMID: 34959495 PMCID: PMC8708136 DOI: 10.3390/pathogens10121540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 02/05/2023] Open
Abstract
Dental caries, as a common oral infectious disease, is a worldwide public health issue. Oral biofilms are the main cause of dental caries. Streptococcus mutans (S. mutans) is well recognized as the major causative factor of dental caries within oral biofilms. In addition to mechanical removal such as tooth brushing and flossing, the topical application of antimicrobial agents is necessarily adjuvant to the control of caries particularly for high-risk populations. The mainstay antimicrobial agents for caries such as chlorhexidine have limitations including taste confusions, mucosal soreness, tooth discoloration, and disruption of an oral microbial equilibrium. Antimicrobial small molecules are promising in the control of S. mutans due to good antimicrobial activity, good selectivity, and low toxicity. In this paper, we discussed the application of antimicrobial small molecules to the control of S. mutans, with a particular focus on the identification and development of active compounds and their modes of action against the growth and virulence of S. mutans.
Collapse
Affiliation(s)
- Sirui Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ran Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (R.Y.); (X.X.)
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chengdu 610041, China; (S.Y.); (J.Z.)
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Correspondence: (R.Y.); (X.X.)
| |
Collapse
|
41
|
Singh R, Ren Z, Shi Y, Lin S, Kwon K, Balamurugan S, Rai V, Mante F, Koo H, Daniell H. Affordable oral health care: dental biofilm disruption using chloroplast made enzymes with chewing gum delivery. PLANT BIOTECHNOLOGY JOURNAL 2021; 19:2113-2125. [PMID: 34076337 PMCID: PMC8486246 DOI: 10.1111/pbi.13643] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 05/09/2023]
Abstract
Current approaches for oral health care rely on procedures that are unaffordable to impoverished populations, whereas aerosolized droplets in the dental clinic and poor oral hygiene may contribute to spread of several infectious diseases including COVID-19, requiring new solutions for dental biofilm/plaque treatment at home. Plant cells have been used to produce monoclonal antibodies or antimicrobial peptides for topical applications to decrease colonization of pathogenic microbes on dental surface. Therefore, we investigated an affordable method for dental biofilm disruption by expressing lipase, dextranase or mutanase in plant cells via the chloroplast genome. Antibiotic resistance gene used to engineer foreign genes into the chloroplast genome were subsequently removed using direct repeats flanking the aadA gene and enzymes were successfully expressed in marker-free lettuce transplastomic lines. Equivalent enzyme units of plant-derived lipase performed better than purified commercial enzymes against biofilms, specifically targeting fungal hyphae formation. Combination of lipase with dextranase and mutanase suppressed biofilm development by degrading the biofilm matrix, with concomitant reduction of bacterial and fungal accumulation. In chewing gum tablets formulated with freeze-dried plant cells, expressed protein was stable up to 3 years at ambient temperature and was efficiently released in a time-dependent manner using a mechanical chewing simulator device. Development of edible plant cells expressing enzymes eliminates the need for purification and cold-chain transportation, providing a potential translatable therapeutic approach. Biofilm disruption through plant enzymes and chewing gum-based delivery offers an effective and affordable dental biofilm control at home particularly for populations with minimal oral care access.
Collapse
Affiliation(s)
- Rahul Singh
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Zhi Ren
- Divisions of Community Oral Health & Pediatric DentistryDepartment of OrthodonticsSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Yao Shi
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Shina Lin
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Kwang‐Chul Kwon
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Shanmugaraj Balamurugan
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Vineeta Rai
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Francis Mante
- Department of Preventive and Restorative DentistrySchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Hyun Koo
- Divisions of Community Oral Health & Pediatric DentistryDepartment of OrthodonticsSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Center for Innovation & Precision DentistrySchool of Dental Medicine and School of Engineering & Applied SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Henry Daniell
- Department of Basic and Translational SciencesSchool of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- Center for Innovation & Precision DentistrySchool of Dental Medicine and School of Engineering & Applied SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| |
Collapse
|
42
|
Amissah F, Andey T, Ahlschwede KM. Nanotechnology-based therapies for the prevention and treatment of Streptococcus mutans-derived dental caries. J Oral Biosci 2021; 63:327-336. [PMID: 34536629 DOI: 10.1016/j.job.2021.09.002] [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] [Received: 04/28/2021] [Revised: 09/03/2021] [Accepted: 09/07/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Dental caries results from long-term acid production when sugar is metabolized by a bacterial biofilm, resulting in a loss of calcium and phosphate from the enamel. Streptococcus mutans is a type of acid-producing bacteria and a virulent contributor to oral biofilms. Conventional treatment options, such as cefazolin and ampicillin, have significant levels of bacterial resistance. Other topical agents, such as fluoride, tend to be washed away by saliva, resulting in low therapeutic efficacy. HIGHLIGHT This review aims to highlight the solubility issues that plague poorly water-soluble therapeutic agents, various novel polymeric, and lipid-based nanotechnology systems that aim to improve the retention of therapeutic agents in the oral cavity. CONCLUSION In this review, different formulation types demonstrated improved therapeutic outcomes by enhancing drug solubility, promoting penetration into the deep layers of the biofilm, facilitating prolonged residence time in the buccal cavity, and reducing the emergence of drug-resistant phenotypes. These formulations have a strong potential to give new life to therapeutic agents that have limited physicochemical characteristics.
Collapse
Affiliation(s)
- Felix Amissah
- Department of Pharmaceutical Sciences, College of Pharmacy, Ferris State University, Big Rapids, MI, USA
| | - Terrick Andey
- Department of Pharmaceutical Sciences, School of Pharmacy, MCPHS University, Worcester, MA, USA
| | - Kristen M Ahlschwede
- Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA.
| |
Collapse
|
43
|
Gross M, Ashqar F, Sionov RV, Friedman M, Eliashar R, Zaks B, Gati I, Duanis-Assaf D, Feldman M, Steinberg D. Sustained release varnish containing chlorhexidine for prevention of Streptococcus mutans biofilm formation on voice prosthesis surface: an in vitro study. Int Microbiol 2021; 25:177-187. [PMID: 34505216 DOI: 10.1007/s10123-021-00205-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 08/13/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES In this study, we aimed to develop a novel, sustained release varnish (SRV) for voice prostheses (VP) releasing chlorhexidine (CHX), for the prevention of biofilm formation caused by the common oral bacteria Streptococcus mutans on VP surfaces. METHODS This study was performed in an in vitro model as a step towards future in vivo trials. VPs were coated with a SRV containing CHX (SRV-CHX) or SRV alone (placebo-SRV) that were daily exposed to S. mutans. The polymeric materials of SRV were composed of ethylcellulose and PEG-400. Biofilm formation was assessed by DNA quantification (qPCR), crystal violet staining, confocal laser scanning microscopy (CLSM), scanning electron microscopy (SEM), and kinetics experiments. RESULTS The amount of DNA in the biofilms formed by S. mutans on VP surfaces coated once with SRV-CHX (1.024 ± 0.218 ng DNA/piece) was 58.5 ± 8.8% lower than that of placebo-SRV-coated VPs (2.465 ± 0.198 ng DNA/piece) after a 48-h exposure to S. mutans (p = 0.038). Reduced biofilm mass on SRV-CHX-coated VPs was visually confirmed by CLSM and SEM. CV staining of SRV-CHX single-coated VPs that have been exposed to S. mutans nine times showed a 98.1 ± 0.2% reduction in biofilm mass compared to placebo-SRV-coated VPs (p = 0.003). Kinetic experiments revealed that SRV-CHX triple-coated VPs could delay bacterial growth for 23 days. CONCLUSIONS Coating VPs with SRV-CHX has an inhibitory effect on biofilm formation and prevents bacterial growth in their vicinities. This study is a proof-of-principle that paves the way for developing new clinical means for reducing both VPs' bacterial biofilm formation and device failure.
Collapse
Affiliation(s)
- Menachem Gross
- Department of Otolaryngology - Head and Neck Surgery, Hadassah Hebrew University Medical Center, P.O.Box 12000, 91120, Jerusalem, Israel. .,Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel. .,School of Medicine, Hebrew University, Jerusalem, Israel.
| | - Fadi Ashqar
- Department of Otolaryngology - Head and Neck Surgery, Hadassah Hebrew University Medical Center, P.O.Box 12000, 91120, Jerusalem, Israel
| | - Ronit Vogt Sionov
- Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel
| | - Michael Friedman
- School of Medicine, Hebrew University, Jerusalem, Israel.,School of Pharmacy, Hebrew University, Jerusalem, Israel
| | - Ron Eliashar
- Department of Otolaryngology - Head and Neck Surgery, Hadassah Hebrew University Medical Center, P.O.Box 12000, 91120, Jerusalem, Israel.,School of Medicine, Hebrew University, Jerusalem, Israel
| | - Batya Zaks
- Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel
| | - Irith Gati
- School of Pharmacy, Hebrew University, Jerusalem, Israel
| | - Danielle Duanis-Assaf
- Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel
| | - Mark Feldman
- Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel
| | - Doron Steinberg
- Biofilm Research Laboratory, Institute of Dental Sciences, Hebrew University, 91120, Jerusalem, Israel
| |
Collapse
|
44
|
The Current Strategies in Controlling Oral Diseases by Herbal and Chemical Materials. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:3423001. [PMID: 34471415 PMCID: PMC8405301 DOI: 10.1155/2021/3423001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/26/2021] [Indexed: 01/23/2023]
Abstract
Dental plaque is a biofilm composed of complex microbial communities. It is the main cause of major dental diseases such as caries and periodontal diseases. In a healthy state, there is a delicate balance between the dental biofilm and host tissues. Nevertheless, due to the oral cavity changes, this biofilm can become pathogenic. The pathogenic biofilm shifts the balance from demineralization-remineralization to demineralization and results in dental caries. Dentists should consider caries as a result of biological processes of dental plaque and seek treatments for the etiologic factors, not merely look for the treatment of the outcome caused by biofilm, i.e., dental caries. Caries prevention strategies can be classified into three groups based on the role and responsibility of the individuals doing them: (1) community-based strategy, (2) dental professionals-based strategy, and (3) individual-based strategy. The community-based methods include fluoridation of water, salt, and milk. The dental professionals-based methods include professional tooth cleaning and use of varnish, fluoride gel and foam, fissure sealant, and antimicrobial agents. The individual-based (self-care) methods include the use of fluoride toothpaste, fluoride supplements, fluoride mouthwashes, fluoride gels, chlorhexidine gels and mouthwashes, slow-release fluoride devices, oral hygiene, diet control, and noncariogenic sweeteners such as xylitol. This study aimed to study the research in the recent five years (2015–2020) to identify the characteristics of dental biofilm and its role in dental caries and explore the employed approaches to prevent the related infections.
Collapse
|
45
|
Villar CC, Dongari-Bagtzoglou A. Fungal diseases: Oral dysbiosis in susceptible hosts. Periodontol 2000 2021; 87:166-180. [PMID: 34463992 DOI: 10.1111/prd.12378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The oral cavity is colonized by a large number of microorganisms that are referred to collectively as the oral microbiota. These indigenous microorganisms have evolved in symbiotic relationships with the oral mucosal immune system and are involved in maintaining homeostasis in the oral cavity. Although Candida species are commonly found in the healthy oral cavity without causing infection, these fungi can become pathogenic. Recents advances indicate that the development of oral candidiasis is driven both by Candida albicans overgrowth in a dysbiotic microbiome and by disturbances in the host's immune system. Perturbation of the oral microbiota triggered by host-extrinsic (ie, medications), host-intrinsic (ie, host genetics), and microbiome-intrinsic (ie, microbial interactions) factors may increase the risk of oral candidiasis. In this review, we provide an overview of the oral mycobiome, with a particular focus on the interactions of Candida albicans with some of the most common oral bacteria and the oral mucosal immune system. Also, we present a summary of our current knowledge of the host-intrinsic and host-extrinsic factors that can predispose to oral candidiasis.
Collapse
Affiliation(s)
- Cristina Cunha Villar
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| |
Collapse
|
46
|
Fasiku V, Omolo CA, Devnarain N, Ibrahim UH, Rambharose S, Faya M, Mocktar C, Singh SD, Govender T. Chitosan-Based Hydrogel for the Dual Delivery of Antimicrobial Agents Against Bacterial Methicillin-Resistant Staphylococcus aureus Biofilm-Infected Wounds. ACS OMEGA 2021; 6:21994-22010. [PMID: 34497894 PMCID: PMC8412894 DOI: 10.1021/acsomega.1c02547] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/04/2021] [Indexed: 06/13/2023]
Abstract
Chronic wound infections caused by antibiotic-resistant bacteria have become a global health concern. This is attributed to the biofilm-forming ability of bacteria on wound surfaces, thus enabling their persistent growth. In most cases, it leads to morbidity and in severe cases mortality. Current conventional approaches used in the treatment of biofilm wounds are proving to be ineffective due to limitations such as the inability to penetrate the biofilm matrix; hence, biofilm-related wounds remain a challenge. Therefore, there is a need for more efficient alternate therapeutic interventions. Hydrogen peroxide (HP) is a known antibacterial/antibiofilm agent; however, prolonged delivery has been challenging due to its short half-life. In this study, we developed a hydrogel for the codelivery of HP and antimicrobial peptides (Ps) against bacteria, biofilms, and wound infection associated with biofilms. The hydrogel was prepared via the Michael addition technique, and the physiochemical properties were characterized. The safety, in vitro, and in vivo antibacterial/antibiofilm activity of the hydrogel was also investigated. Results showed that the hydrogel is biosafe. A greater antibacterial effect was observed with HP-loaded hydrogels (CS-HP; hydrogel loaded with HP and CS-HP-P; hydrogel loaded with HP and peptide) when compared to HP as seen in an approximately twofold and threefold decrease in minimum inhibitory concentration values against methicillin-resistant Staphylococcus aureus (MRSA) bacteria, respectively. Similarly, both the HP-releasing hydrogels showed enhanced antibiofilm activity in the in vivo study in mice models as seen in greater wound closure and enhanced wound healing in histomorphological analysis. Interestingly, the results revealed a synergistic antibacterial/antibiofilm effect between HP and P in both in vitro and in vivo studies. The successfully prepared HP-releasing hydrogels showed the potential to combat bacterial biofilm-related infections and enhance wound healing in mice models. These results suggest that the HP-releasing hydrogels may be a superior platform for eliminating bacterial biofilms without using antibiotics in the treatment of chronic MRSA wound infections, thus improving the quality of human health.
Collapse
Affiliation(s)
- Victoria
O. Fasiku
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Calvin A. Omolo
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
- School
of Pharmacy and Health Sciences, Department of Pharmaceutics, United States International University-Africa, P.O. Box 14634, Nairobi 00800, Kenya
| | - Nikita Devnarain
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Usri H. Ibrahim
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Sanjeev Rambharose
- Department
of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch 7602, South Africa
| | - Mbuso Faya
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Chunderika Mocktar
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Sanil D. Singh
- Biomedical
Research Unit, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| | - Thirumala Govender
- Discipline
of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South
Africa
| |
Collapse
|
47
|
Wang Y, Hoffmann JP, Baker SM, Bentrup KHZ, Wimley WC, Fuselier JA, Bitoun JP, Morici LA. Inhibition of Streptococcus mutans biofilms with bacterial-derived outer membrane vesicles. BMC Microbiol 2021; 21:234. [PMID: 34429066 PMCID: PMC8386047 DOI: 10.1186/s12866-021-02296-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Biofilms are microbial communities surrounded by a self-produced extracellular matrix which protects them from environmental stress. Bacteria within biofilms are 10- to 1000-fold more resistant to antibiotics, making it challenging but imperative to develop new therapeutics that can disperse biofilms and eradicate infection. Gram-negative bacteria produce outer membrane vesicles (OMV) that play critical roles in communication, genetic exchange, cargo delivery, and pathogenesis. We have previously shown that OMVs derived from Burkholderia thailandensis inhibit the growth of drug-sensitive and drug-resistant bacteria and fungi. RESULTS Here, we examine the antibiofilm activity of Burkholderia thailandensis OMVs against the oral biofilm-forming pathogen Streptococcus mutans. We demonstrate that OMV treatment reduces biofilm biomass, biofilm integrity, and bacterial cell viability. Both heat-labile and heat-stable components, including 4-hydroxy-3-methyl-2-(2-non-enyl)-quinoline and long-chain rhamnolipid, contribute to the antibiofilm activity of OMVs. When OMVs are co-administered with gentamicin, the efficacy of the antibiotic against S. mutans biofilms is enhanced. CONCLUSION These studies indicate that bacterial-derived OMVs are highly effective biological nanoparticles that can inhibit and potentially eradicate biofilms.
Collapse
Affiliation(s)
- Yihui Wang
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Joseph P. Hoffmann
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Sarah M. Baker
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Kerstin Höner zu Bentrup
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - William C. Wimley
- grid.265219.b0000 0001 2217 8588Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA USA
| | - Joseph A. Fuselier
- grid.265219.b0000 0001 2217 8588Department of Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Jacob P. Bitoun
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Lisa A. Morici
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| |
Collapse
|
48
|
Lahiri D, Nag M, Garai S, Ray RR. The Chemistry of Antibiofilm Phytocompounds. Mini Rev Med Chem 2021; 21:1034-1047. [PMID: 32767942 DOI: 10.2174/1389557520666200807135243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/20/2020] [Accepted: 06/23/2020] [Indexed: 11/22/2022]
Abstract
Phytocompounds are long known for their therapeutic uses due to their competence as antimicrobial agents. The antimicrobial activity of these bioactive compounds manifests their ability as an antibiofilm agent and is thereby proved to be competent to treat the widespread biofilm-associated chronic infections. The rapid development of antibiotic resistance in bacteria has made the treatment of these infections almost impossible by conventional antibiotic therapy, which forced a switch-over to the use of phytocompounds. The present overview deals with the classification of a huge array of phytocompounds according to their chemical nature, detection of their target pathogen, and elucidation of their mode of action.
Collapse
Affiliation(s)
- Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Sayantani Garai
- Department of Biotechnology, University of Engineering & Management, Kolkata, India
| | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, West Bengal, India
| |
Collapse
|
49
|
Cai S, Meng K, Liu P, Cao X, Wang G. Suppressive effects of gecko cathelicidin on biofilm formation and cariogenic virulence factors of Streptococcus mutans. Arch Oral Biol 2021; 129:105205. [PMID: 34237581 DOI: 10.1016/j.archoralbio.2021.105205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/17/2021] [Accepted: 06/26/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The aims of this study were to investigate the effectivity of gecko cathelicidin Gj-CATH2 on biofilm formation and cariogenic virulence factors of S. mutans, and preliminary explore its function mechanisms. DESIGN Minimum inhibitory concentration and bacterial killing kinetics assays were performed to assess the antimicrobial effect of Gj-CATH2.The influence of Gj-CATH2 on S. mutans biofilm formation was determined by crystal violet staining method and observed by SEM. The effects of Gj-CATH2 on exopolysaccharides (EPS) synthesis, bacterial aggregation, acidogenicity and aciduricity of S. mutans were also investigated. Quantitative real-time PCR was conducted to acquire the expression profile of related genes. RESULTS Gj-CATH2 showed strong bactericidal and anti-biofilm effects on S. mutans. SEM confirmed the reduction of the dense structure in S. mutans biofilm in Gj-CATH2-treated groups. Gj-CATH2 significantly inhibited EPS synthesis, cell aggregation, acid production of S. mutans, but showed no influence on its acid proof. Furthermore, the expression of genes related to biofilm formation (gtfB/C/D, gbpB/D), quorum sensing system (luxS and comD/E) and acidogenicity (ldh) was significantly suppressed by Gj-CATH2. Gj-CATH2 also displayed advantageous resistance in human saliva and exhibited negligible toxicity against mammalian cells. CONCLUSIONS Gj-CATH2 inhibited S. mutans biofilm formation by targeting the bacterial adhesion and the biofilm maturation stages. Gj-CATH2 significantly suppressed virulence factors production of S. mutans, resulting in decreased EPS synthesis and reduced acidogenicity of bacteria. These findings suggest Gj-CATH2 might be a promising agent for clinical application in prevention of dental caries.
Collapse
Affiliation(s)
- Shasha Cai
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China.
| | - Kai Meng
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, 272067, China
| | - Peng Liu
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China
| | - Xianting Cao
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China
| | - Guannan Wang
- College of Medical Engineering & the Key Laboratory for Medical Functional Nanomaterials, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
50
|
Passos MR, Almeida RS, Lima BO, Rodrigues JZDS, Macêdo Neres NSD, Pita LS, Marinho PDF, Santos IA, da Silva JP, Oliveira MC, Oliveira MA, Pessoa SMB, Silva MML, Silveira PHS, Reis MM, Santos IP, Ricardo LDON, Andrade LOSB, Soares AB, Correia TML, Souza ÉPD, Pires PN, Cruz MP, Marques LM, Uetanabaro APT, Yatsuda R. Anticariogenic activities of Libidibia ferrea, gallic acid and ethyl gallate against Streptococcus mutans in biofilm model. JOURNAL OF ETHNOPHARMACOLOGY 2021; 274:114059. [PMID: 33794333 DOI: 10.1016/j.jep.2021.114059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Brazil, ethnopharmacological studies show that Libidibia ferrea (Mart. ex Tul.) L. P. Queiroz is commonly used in folk medicine as an antifungal, antimicrobial and anti-inflammatory. In the Amazon region, the dried fruit powder of L. ferrea are widely used empirically by the population in an alcoholic tincture as an antimicrobial mouthwash in oral infections and the infusion is also recommended for healing oral wounds. However, there are few articles that have evaluated the antimicrobial activity against oral pathogens in a biofilm model, identifying active compounds and mechanisms of action. AIM OF THE STUDY The aim of this study was to evaluate the antimicrobial and anti-adherence activities of the ethanolic extract, fractions and isolated compounds (gallic acid and ethyl gallate) of the fruit and seed of L. ferrea against Streptococcus mutans. The inhibition of acidicity/acidogenicity and the expression of the S. mutans GTF genes in biofilms were also evaluated. MATERIALS AND METHODS Minimal Inhibitory Concentration (MIC), Minimum Bactericidal Concentration (MBC) and Minimum Inhibitory Concentration of Cell Adhesion (MICA) were evaluated with ethanolic extract (EELF), fractions, gallic acid (GA) and ethyl gallate (EG) against S. mutans. Inhibition of biofilm formation, pH drop and proton permeability tests were conducted with EELF, GA and EG, and also evaluated the expression of the GTF genes in biofilms. The compounds of dichloromethane fraction were identified by GC-MS. RESULTS This is the first report of shikimic, pyroglutamic, malic and protocatechuic acids identified in L. ferrea. EELF, GA and EG showed MIC at 250 μg/mL, and MBC at 1000 μg/mL by EELF. EELF biofilms showed reduced dry weight and acidogenicity of S. mutans in biofilms. GA and EG reduced viable cells, glucans soluble in alkali, acidogenicity, aciduricity and downregulated expression of gtfB, gtfC and gtfD genes in biofilms. SEM images of GA and EG biofilms showed a reduction of biomass, exopolysaccharide and microcolonies of S. mutans. CONCLUSIONS The ethanolic extract of fruit and seed of L. ferrea, gallic acid and ethyl gallate showed great antimicrobial activity and inhibition of adhesion, reduction of acidogenicity and aciduricity in S. mutans biofilms. The results obtained in vitro validate the use of this plant in ethnopharmacology, and open opportunities for the development of new oral anticariogenic agents, originated by plants that can inhibit pathogenic biofilm that leads to the development of caries.
Collapse
Affiliation(s)
- Manuela Ribeiro Passos
- State University of Santa Cruz (UESC), Soane Nazaré de Andrade Campus, Jorge Amado Highway, 16 Km, Salobrinho, Ilhéus, Bahia, Brazil
| | - Rafael Silva Almeida
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Brenda Oliveira Lima
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Jeisa Zielle de Souza Rodrigues
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Nayara Silva de Macêdo Neres
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Louise Soares Pita
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | | | - Iago Almeida Santos
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Jamile Pinheiro da Silva
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Milena Cardoso Oliveira
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Mariana Araújo Oliveira
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Stela Mares Brasileiro Pessoa
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Marlon Mário Leles Silva
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Paulo Henrique Santana Silveira
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Mariane Mares Reis
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Isabella Porto Santos
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | | | | | - Ananda Brito Soares
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Thiago Macêdo Lopes Correia
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Érika Pereira de Souza
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Polyane Novais Pires
- State University of Santa Cruz (UESC), Soane Nazaré de Andrade Campus, Jorge Amado Highway, 16 Km, Salobrinho, Ilhéus, Bahia, Brazil
| | - Mariluze Peixoto Cruz
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Lucas Miranda Marques
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil
| | - Ana Paula Trovatti Uetanabaro
- State University of Santa Cruz (UESC), Soane Nazaré de Andrade Campus, Jorge Amado Highway, 16 Km, Salobrinho, Ilhéus, Bahia, Brazil
| | - Regiane Yatsuda
- Multidisciplinary Health Institute, Federal University of Bahia, 58 Hormindo Barros Street, Vitória da Conquista, Bahia, Brazil.
| |
Collapse
|