1
|
Condor AM, Kui A, Condor DC, Negucioiu M, Buduru SD, Lucaciu PO. Metabolomics Applications for Diagnosing Peri-Implantitis: A Systematic Review of In Vivo Studies. Diagnostics (Basel) 2025; 15:990. [PMID: 40310396 PMCID: PMC12025503 DOI: 10.3390/diagnostics15080990] [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: 03/24/2025] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Background/Objectives: Peri-implantitis is a prevalent inflammatory condition affecting dental implants, leading to increased treatment costs, patient dissatisfaction, and potential implant failure. Novel biomarker-based approaches may contribute to early detection, thereby decreasing the burden of the disease. The aim of this review was to assess in vivo studies using metabolomics to identify the metabolic profiles and potential biomarkers of peri-implantitis. Methods: The protocol for this study was registered with PROSPERO (CRD42025634865). Five databases and grey literature sources (PubMed, Scopus, Web of Science, ProQuest, and Google Scholar) were searched using keywords related to metabolomics and peri-implantitis. Studies were selected by independent, inter-calibrated researchers. Data were extracted using predefined, custom forms. The risk of bias was assessed using the ROBINS-I tool. Results: An electronic literature search retrieved 543 articles, of which five were selected. All studies were published within the last five years of the search. All but one study used untargeted metabolomics, and all studies identified metabolites associated with peri-implantitis or distinct metabolomic profiles of peri-implantitis. SCFAs and lysine metabolites were recurring in the results, confirming the findings of previous metabolomic studies on periodontal disease. Conclusions: Metabolomics has not been widely used to study peri-implantitis. Evidence from existing studies confirms the findings of metabolomics studies on periodontitis. Several metabolites related to PI are associated with immune response, tissue degradation, and cellular energy pathways. Integrating -omics technologies into peri-implantitis diagnosis may facilitate biomarker discovery and improve early detection strategies.
Collapse
Affiliation(s)
- Ana-Maria Condor
- Oral Health Discipline, Department 3—Oral Rehabilitation, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.-M.C.)
- Cluj County Emergency Clinical Hospital, 400006 Cluj-Napoca, Romania
- Prosthodontics Discipline, Department 4—Prosthodontics and Dental Materials, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Andreea Kui
- Prosthodontics Discipline, Department 4—Prosthodontics and Dental Materials, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Daniela Cornelia Condor
- Periodontology Discipline, Department 3—Oral Rehabilitation, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Marius Negucioiu
- Prosthodontics Discipline, Department 4—Prosthodontics and Dental Materials, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Smaranda Dana Buduru
- Prosthodontics Discipline, Department 4—Prosthodontics and Dental Materials, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Patricia Ondine Lucaciu
- Oral Health Discipline, Department 3—Oral Rehabilitation, Faculty of Dental Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.-M.C.)
| |
Collapse
|
2
|
Zayed N, Munjaković H, Aktan MK, Simoens K, Bernaerts K, Boon N, Braem A, Pamuk F, Saghi M, Van Holm W, Fidler A, Gašperšič R, Teughels W. Electrolyzed Saline Targets Biofilm Periodontal Pathogens In Vitro. J Dent Res 2024; 103:243-252. [PMID: 38185942 DOI: 10.1177/00220345231216660] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024] Open
Abstract
Preventing the development and recurrence of periodontal diseases often includes antimicrobial mouthrinses to control the growth of the periodontal pathogens. Most antimicrobials are nonselective, targeting the symbiotic oral species as well as the dysbiosis-inducing ones. This affects the overall microbial composition and metabolic activity and consequently the host-microbe interactions, which can be detrimental (associated with inflammation) or beneficial (health-associated). Consequently, guiding the antimicrobial effect for modulating the microbial composition to a health-associated one should be considered. For such an approach, this study investigated electrolyzed saline as a novel rinse. Electrolyzed saline was prepared from sterile saline using a portable electrolysis device. Multispecies oral homeostatic and dysbiotic biofilms were grown on hydroxyapatite discs and rinsed daily with electrolyzed saline (EOS). Corresponding positive (NaOCl) and negative (phosphate-buffered saline) controls were included. After 3 rinses, biofilms were analyzed with viability quantitative polymerase chain reaction and scanning electron microscopy. Supernatants of rinsed biofilms were used for metabolic activity analysis (high-performance liquid chromatography) through measuring organic acid content. In addition, human oral keratinocytes (HOKs) were exposed to EOS to test biocompatibility (cytotoxicity and inflammation induction) and also to rinsed biofilms to assess their immunogenicity after rinsing. Rinsing the dysbiotic biofilms with EOS could reduce the counts of the pathobionts (>3 log10 Geq/mm2 reduction) and avert biofilm dysbiosis (≤1% pathobiont abundance), leading to the dominance of commensal species (≥99%), which altered both biofilm metabolism and interleukin 8 (IL-8) induction in HOKs. EOS had no harmful effects on homeostatic biofilms. The scanning electron micrographs confirmed the same. In addition, tested concentrations of EOS did not have any cytotoxic effects and did not induce IL-8 production in HOKs. EOS showed promising results for diverting dysbiosis in in vitro rinsed biofilms and controlling key periopathogens, with no toxic effects on commensal species or human cells. This novel rinsing should be considered for clinical applications.
Collapse
Affiliation(s)
- N Zayed
- Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - H Munjaković
- Department of Oral Medicine and Periodontology, University Clinical Centre Ljubljana, Ljubljana, Slovenia
| | - M K Aktan
- Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Leuven, Belgium
| | - K Simoens
- Chemical and Biochemical Reactor Engineering and Safety, Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - K Bernaerts
- Chemical and Biochemical Reactor Engineering and Safety, Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - N Boon
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - A Braem
- Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering Research Group, Leuven, Belgium
| | - F Pamuk
- Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| | - M Saghi
- Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| | - W Van Holm
- Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - A Fidler
- Department of Endodontic and Restorative Dentistry, University Clinical Centre Ljubljana, Ljubljana, Slovenia
| | - R Gašperšič
- Department of Oral Medicine and Periodontology, University Clinical Centre Ljubljana, Ljubljana, Slovenia
| | - W Teughels
- Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
3
|
Li R, Hou M, Yu L, Luo W, Kong J, Yu R, Liu R, Li Q, Tan L, Pan C, Wang H. Anti-biofilm effect of salivary histatin 5 on Porphyromonas gingivalis. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12664-4. [PMID: 37395749 DOI: 10.1007/s00253-023-12664-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/14/2023] [Accepted: 06/24/2023] [Indexed: 07/04/2023]
Abstract
This study aimed to investigate the effects of salivary histatin 5 (Hst5) on Porphyromonas gingivalis (P. gingivalis) biofilms in vitro and in vivo and the possible mechanisms. In in vitro experiments, P. gingivalis biomass was determined by crystal violet staining. Polymerase chain reaction, scanning electron microscopy, and confocal laser scanning microscopy were used to determine the Hst5 concentration. A search for potential targets was performed using transcriptomic and proteomic analyses. In vivo experimental periodontitis was established in rats to evaluate the effects of Hst5 on periodontal tissues. Experimental results showed that 25 µg/mL Hst5 effectively inhibited biofilm formation, and increased concentrations of Hst5 increased the inhibitive effect. Hst5 might bind to the outer membrane protein RagAB. A combination of transcriptomic and proteomic analyses revealed that Hst5 could regulate membrane function and metabolic processes in P. gingivalis, in which RpoD and FeoB proteins were involved. In the rat periodontitis model, alveolar bone resorption and inflammation levels in periodontal tissues were reduced by 100 µg/mL Hst5. This study showed that 25 µg/mL Hst5 inhibited P. gingivalis biofilm formation in vitro by changing membrane function and metabolic process, and RpoD and FeoB proteins might play important roles in this process. Moreover, 100 µg/mL Hst5 inhibited periodontal inflammation and alveolar bone loss in rat periodontitis via its antibacterial and anti-inflammatory effects. KEY POINTS: • Anti-biofilm activity of histatin 5 on Porphyromonas gingivalis was investigated. • Histatin 5 inhibited Porphyromonas gingivalis biofilm formation. • Histatin 5 showed inhibitory effects on the occurrence of rat periodontitis.
Collapse
Affiliation(s)
- Rui Li
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Mengjie Hou
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Liying Yu
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Wen Luo
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Jie Kong
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Renmei Yu
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Ruihan Liu
- Clinical Medicine, Shenyang Medical College, Huanghe North Street 146, Shenyang, 110034, Liaoning Province, China
| | - Qian Li
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Lisi Tan
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Chunling Pan
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China
| | - Hongyan Wang
- Department of Periodontology, School of Stomatology, China Medical University, Nanjing North Street 117, Shenyang, 110000, Liaoning Province, China.
| |
Collapse
|
4
|
Copling A, Akantibila M, Kumaresan R, Fleischer G, Cortes D, Tripathi RS, Carabetta VJ, Vega SL. Recent Advances in Antimicrobial Peptide Hydrogels. Int J Mol Sci 2023; 24:7563. [PMID: 37108725 PMCID: PMC10139150 DOI: 10.3390/ijms24087563] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Advances in the number and type of available biomaterials have improved medical devices such as catheters, stents, pacemakers, prosthetic joints, and orthopedic devices. The introduction of a foreign material into the body comes with a risk of microbial colonization and subsequent infection. Infections of surgically implanted devices often lead to device failure, which leads to increased patient morbidity and mortality. The overuse and improper use of antimicrobials has led to an alarming rise and spread of drug-resistant infections. To overcome the problem of drug-resistant infections, novel antimicrobial biomaterials are increasingly being researched and developed. Hydrogels are a class of 3D biomaterials consisting of a hydrated polymer network with tunable functionality. As hydrogels are customizable, many different antimicrobial agents, such as inorganic molecules, metals, and antibiotics have been incorporated or tethered to them. Due to the increased prevalence of antibiotic resistance, antimicrobial peptides (AMPs) are being increasingly explored as alternative agents. AMP-tethered hydrogels are being increasingly examined for antimicrobial properties and practical applications, such as wound-healing. Here, we provide a recent update, from the last 5 years of innovations and discoveries made in the development of photopolymerizable, self-assembling, and AMP-releasing hydrogels.
Collapse
Affiliation(s)
- Aryanna Copling
- Department of Molecular & Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Maxwell Akantibila
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Raaha Kumaresan
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA;
| | - Gilbert Fleischer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Dennise Cortes
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Rahul S. Tripathi
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA;
- Department of Orthopedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
5
|
Promising Application of D-Amino Acids toward Clinical Therapy. Int J Mol Sci 2022; 23:ijms231810794. [PMID: 36142706 PMCID: PMC9503604 DOI: 10.3390/ijms231810794] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/25/2022] Open
Abstract
The versatile roles of D-amino acids (D-AAs) in foods, diseases, and organisms, etc., have been widely reported. They have been regarded, not only as biomarkers of diseases but also as regulators of the physiological function of organisms. Over the past few decades, increasing data has revealed that D-AAs have great potential in treating disease. D-AAs also showed overwhelming success in disengaging biofilm, which might provide promise to inhibit microbial infection. Moreover, it can effectively restrain the growth of cancer cells. Herein, we reviewed recent reports on the potential of D-AAs as therapeutic agents for treating neurological disease or tissue/organ injury, ameliorating reproduction function, preventing biofilm infection, and inhibiting cancer cell growth. Additionally, we also reviewed the potential application of D-AAs in drug modification, such as improving biostability and efficiency, which has a better effect on therapy or diagnosis.
Collapse
|
6
|
The effect of surface material, roughness and wettability on the adhesion and proliferation of Streptococcus gordonii, Fusobacterium nucleatum and Porphyromonas gingivalis. J Dent Sci 2022; 18:517-525. [PMID: 37123448 PMCID: PMC10131180 DOI: 10.1016/j.jds.2022.09.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/12/2022] [Indexed: 11/24/2022] Open
Abstract
Background/purpose Dental implants are inevitably exposed to bacteria in oral cavity. Understanding the colonization of bacteria on implant surface is necessary to prevent bacteria-related inflammation surrounding dental implants. The purpose of this study was to investigate the effect of surface properties on biofilm formation on the implant surface. Materials and methods One early colonizer, Streptococcus gordonii (S. gordonii), and two late colonizers, Fusobacterium nucleatum (F. nucleatum) and Porphyromonas gingivalis (P. gingivalis), were grown on the titanium and zirconia surfaces with two types of surface roughness for 24 and 72 h. Each bacterial biofilm on specimens was quantified using crystal violet assay and observed by scanning electron microscopy. Results S. gordonii formed more biofilm on the titanium surface than zirconia at the same roughness and more biofilm on the rough surface than smooth one of the same materials at 24 and 72 h of incubation. F. nucleatum adhered on all the surfaces at 24 h and proliferated actively on the surfaces except smooth zirconia at 72 h. P. gingivalis proliferated vigorously on the surfaces at 72 h while it scarcely adhered at 24 h. There was no consistent correlation between contact angle and biofilm formation of the three bacteria. Conclusion The three bacteria proliferated most on the rough titanium surface and least on the smooth zirconia surface. In addition, the proliferation was affected by the bacterial species as well as the surface properties.
Collapse
|
7
|
Vahdati SN, Behboudi H, Navasatli SA, Tavakoli S, Safavi M. New insights into the inhibitory roles and mechanisms of D-amino acids in bacterial biofilms in medicine, industry, and agriculture. Microbiol Res 2022; 263:127107. [PMID: 35843196 DOI: 10.1016/j.micres.2022.127107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022]
Abstract
Biofilms are complex aggregates of microbes that are tightly protected by an extracellular matrix (ECM) and may attach to a surface or adhere together. A higher persistence of bacteria on biofilms makes them resistant not only to harsh conditions but also to various antibiotics which led to the emergence of problems in different applications. Recently, it has been discovered that many bacteria produce and release various D-amino acids (D-AAs) to inhibit biofilm formation, which made a great deal of interest in research into the control of bacterial biofilms in diverse fields, such as human health, industrial settings, and medical devices. D-AAs have various mechanisms to inhibit bacterial biofilms such as: (i) interfering with protein synthesis (ii) Inhibition of extracellular polymeric materials (EPS) productions (protein, eDNA, and polysaccharide) (iii) Inhibition of quorum sensing (autoinducers), and (iv) interfere with peptidoglycan synthesis, these various modes of action, enables these small molecules to inhibit both Gram-negative and Gram-positive bacterial biofilms. Since most biofilms are multi-species, D-AAs in combination with other antimicrobial agents are good choices to combat a variety of bacterial biofilms without displaying toxicity on human cells. This review article addressed the role of D-AAs in controlling several bacterial biofilms and described the possible or definite mechanisms involved in this process.
Collapse
Affiliation(s)
- Saeed Niazi Vahdati
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Hossein Behboudi
- Department of Biology, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran.
| | - Sepideh Aliniaye Navasatli
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Sara Tavakoli
- Department of Biotechnology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| |
Collapse
|
8
|
Du X, Li B, Cai Q, Qiao S, Wang Z, Li Z, Li Y, Meng W. D-aspartic acid protects against gingival fibroblasts inflammation by suppressing pyroptosis. Mol Biol Rep 2022; 49:5821-5829. [PMID: 35716284 DOI: 10.1007/s11033-022-07335-y] [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: 12/13/2021] [Revised: 02/19/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Peri-implantitis is the main cause of dental implant failure, which is associated with pyroptosis. The roles of D-aspartic acid (D-Asp) on pyroptosis and the mechanism of the protective effect of D-Asp on human gingival fibroblasts (HGFs) remain unknown. This study investigated the effects of D-Asp on the pyroptosis of HGFs induced by high mobility group box 1 protein (HMGB1). METHODS The cytotoxic effects of D-Asp on HGFs was detected by Cell Counting Kit-8 assay, the membrane permeability was investigated by propidium iodide/ Hoechst 33,342 double staining, flow cytometry analysis, and lactate dehydrogenase releasing, The gene and protein expression levels were detected by real-time quantitative PCR, enzyme-linked immunosorbent assay, and Western blot, respectively. RESULTS Cell viability analysis showed that D-Asp ≤ 30 mM had no cytotoxicity to HGFs. HMGB1 drastically raised the membrane permeability of HGFs, while 1/10/30 mM D-Asp suppressed the permeability and remained the integrity of the membrane. HMGB1 promoted the mRNA expression of NLRP3, caspase-1, GSDMD, IL-1β, and IL-18, and the protein expression of IL-1β, IL-18, caspase-1, GSDMD, and NLRP3. CONCLUSIONS With the pretreatment of HGFs with D-Asp of 1/10/30 mM for 24 h, the cell membrane permeability was reduced and the expression of NLRP3, caspase-1, GSDMD, IL-1β, and IL-18 was significantly decreased compared with the HMGB1 group, indicating the competitive antagonism of D-Asp against HMGB1 on the binding with toll-like receptors. Hence, this study may provide a novel insight into preventing pyroptosis and propose a new strategy for the treatment of peri-implantitis.
Collapse
Affiliation(s)
- Xuechun Du
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, Jilin, China
| | - Baosheng Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China
| | - Qing Cai
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China
| | - Shuwei Qiao
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, Jilin, China
| | - Zixuan Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, Jilin, China
| | - Zhen Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.,Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, Jilin, China
| | - Yuyang Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.,Department of Cariology and Endodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Weiyan Meng
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, 130021, Changchun, Jilin, China.
| |
Collapse
|
9
|
Wang TY, Guo R, Hu LL, Liu JJ, Lu HT. Mass Spectrometry-Based Targeted Metabolomics Revealed the Regulatory Roles of Magnesium on Biofilm Formation in Escherichia coli by Targeting Functional Metabolites. JOURNAL OF ANALYSIS AND TESTING 2022. [DOI: 10.1007/s41664-021-00208-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Gong M, Zhang R, Qi J, Wang J, Liu Q, Zhou H, Song Y, Song X, Mei Y. In vitro evaluation of the antibacterial effect of colloidal bismuth subcitrate on Porphyromonas gingivalis and its biofilm. Arch Oral Biol 2021; 133:105300. [PMID: 34742000 DOI: 10.1016/j.archoralbio.2021.105300] [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: 09/05/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To investigate the antibacterial and anti-biofilm effects of colloidal bismuth subcitrate (CBS) on Porphyromonas gingivalis (P. gingivalis) in its planktonic and biofilm forms and also compare it with that of 0.2% chlorhexidine (CHX). DESIGN The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of CBS were determined by the microdilution method; the bacteriostatic rate of CBS was determined by the MTT assay; the effect of CBS on the membrane integrity of P. gingivalis was investigated by the flow cytometric methods. The effects of CBS on the biomass and bacterial activity of biofilm were investigated. Confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM) were used to investigate the activity and structure of biofilms. RESULTS The MIC and MBC values were 18.75 µg/mL and 37.5 µg/mL. CBS could damage the cell membrane of P. gingivalis. CBS effectively inhibited biofilm formation and promoted dissociation at higher concentrations of 37.5 µg/mL and 75 µg/mL, respectively. The results also indicated an altered biofilm structure and reduced biofilm thickness and bacterial aggregation. CONCLUSIONS CBS affected the metabolic and physiological processes of P. gingivalis, inhibited the formation of biofilm, and disrupted the mature biofilm.
Collapse
Affiliation(s)
- Min Gong
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Rui Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jianyan Qi
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jue Wang
- Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Qian Liu
- Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hongyan Zhou
- Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yumeng Song
- Nanjing Stomatological Hospital Medical School of Nanjing University, Nanjing, China
| | - Xiaomeng Song
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yufeng Mei
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China; Department of Pediatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Antibacterial Activity of Boswellia sacra Flueck. Oleoresin Extract against Porphyromonas gingivalis Periodontal Pathogen. Antibiotics (Basel) 2021; 10:antibiotics10070859. [PMID: 34356781 PMCID: PMC8300764 DOI: 10.3390/antibiotics10070859] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/07/2021] [Accepted: 07/10/2021] [Indexed: 11/17/2022] Open
Abstract
Boswellia sacra Flueck. oleoresin extract (frankincense) has traditionally been used in the treatment of different diseases, but there are no sufficient studies on its potential activity against periodontal pathogens. Therefore, antibacterial and antibiofilm activity of frankincense extract against Porphyromonas gingivalis clinical isolates were studied. The phytochemical composition of the volatile components of the extract was identified by GC-MS analysis revealing 49 compounds as trans-nerolidyl formate, cycloartenol acetate, ursenoic acid 3-oxomethyl ester, bisabolene epoxide, and kaur-16-ene. It decreased the growth and increased the leakage of nucleotides in 58.3% and 33.3% of isolates, respectively. Additionally, it reduced the extracellular polysaccharide production and the cell surface hydrophobicity in 41.67% and 50% of the isolates, respectively. Crystal violet assay revealed inhibition of biofilm formation by the tested isolates. Light microscope and scanning electron microscope were used to examine the biofilms and they confirmed the reduction of biofilm formation by frankincense extract. Downregulation of the genes linked to biofilm formation (fimA, hagA, and hagB) was observed using qRT-PCR after treatment with the frankincense extract. This study suggested that the frankincense extract could exhibit antibacterial and antibiofilm activity against P. gingivalis isolates. Thus, the frankincense extract could be used as a treatment approach for periodontitis.
Collapse
|
12
|
Abstract
Biofilms are widely involved in human lives, such as in medical infection, environmental remediation, and industrial processes. However, the control of the biofilm has still been a challenge because of its strong drug resistance. Here, we designed and synthesized an amphipathic antimicrobial peptide (Ac-DKDHDHDQDKDLDVDFDFDADK-NH2 (KKd-11)) that was composed of d-amino acids (DAAs). KKd-11 was found to self-assemble into a hydrogel with an improved long-term antimicrobial ability and a better antiprotease activity as compared to the hydrogel formed by Ac-LKLHLHLQLKLLLVLFLFLALK-NH2 (KK-11). Our results indicated that KKd-11 was not only able to inhibit the formation of biofilms but also could effectively damage preformed mature biofilms and kill the bacteria within the biofilms. Besides, cell viability assays indicated that the KKd-11 peptide had very good biocompatibility. We think d-peptide hydrogels may have great potential in the treatment of biofilm-induced infections.
Collapse
Affiliation(s)
- Zhen Guo
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yujiao Wang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingyuan Tan
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuwen Ji
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Hu
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Yi Zhang
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.,Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| |
Collapse
|
13
|
Zhang Z, Li B, Cai Q, Qiao S, Wang D, Wang H, Zhang H, Yang Y, Meng W. Synergistic effects of D-arginine, D-methionine and D-histidine against Porphyromonas gingivalis biofilms. BIOFOULING 2021; 37:222-234. [PMID: 33682548 DOI: 10.1080/08927014.2021.1893309] [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: 12/06/2020] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Porphyromonas gingivalis biofilms are implicated in the pathology of peri-implantitis and periodontitis. In this study, D-arginine (R), D-methionine (M), D-histidine (H), and a mixture of these D-amino acids (D-AAs) were investigated as an effective therapeutic strategy against P. gingivalis biofilms. The bacterial growth activity and minimum inhibitory concentrations were determined for each D-AA, along with the effects of the D-AAs mixture on biofilm development, morphology, structure, extracellular polysaccharides (EPS), cytotoxicity towards commensals, and bacterial structure. The D-AA mixture delayed the proliferation of P. gingivalis, changed its membrane structure, and decreased biofilm thickness and integrity, as compared with individual D-AAs. The EPS content increased with the concentration of D-AAs. The present study shows that a 4 mM RMH, triple D-AA mixture, enhanced deleterious effects on P. gingivalis biofilms without any cytotoxicity compared with individual D-AAs, thus providing a new strategy for the treatment of peri-implantitis and periodontitis.
Collapse
Affiliation(s)
- Zhenyang Zhang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Baosheng Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Qing Cai
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shuwei Qiao
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Dan Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
| | - Heling Wang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Huiyan Zhang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yalan Yang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, China
- Department of Pediatric Dentistry, Hospital of Stomatology, Jinlin University, Changchun, China
| | - Weiyan Meng
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
14
|
Li YY, Li BS, Liu WW, Cai Q, Wang HY, Liu YQ, Liu YJ, Meng WY. Effects of D-arginine on <i>Porphyromonas gingivalis</i> biofilm. J Oral Sci 2020; 62:57-61. [DOI: 10.2334/josnusd.19-0075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Yu-Yang Li
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling
| | - Bao-Sheng Li
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
| | - Wei-Wei Liu
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Jilin University
| | - Qing Cai
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
| | - Hao-Yang Wang
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling
| | - Yan-Qun Liu
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling
| | - Yu-Jie Liu
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling
| | - Wei-Yan Meng
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University
| |
Collapse
|
15
|
Simon G, Bérubé C, Voyer N, Grenier D. Anti-biofilm and anti-adherence properties of novel cyclic dipeptides against oral pathogens. Bioorg Med Chem 2018; 27:2323-2331. [PMID: 30528685 DOI: 10.1016/j.bmc.2018.11.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/14/2018] [Accepted: 11/28/2018] [Indexed: 12/19/2022]
Abstract
Microorganisms embedded in a biofilm are significantly more resistant to antimicrobial agents and the defences of the human immune system, than their planktonic counterpart. Consequently, compounds that can inhibit biofilm formation are of great interest for novel therapeutics. In this study, a screening approach was used to identify novel cyclic dipeptides that have anti-biofilm activity against oral pathogens. Five new active compounds were identified that prevent biofilm formation by the cariogenic bacterium Streptococcus mutans and the pathogenic fungus Candida albicans. These compounds also inhibit the adherence of microorganisms to a hydroxylapatite surface. Further investigations were conducted on these compounds to establish the structure-activity relationship, and it was deduced that the common cleft pattern is required for these molecules to act effectively against biofilms.
Collapse
Affiliation(s)
- Gaëlle Simon
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, 2420, rue de la Terrasse, Québec, QC G1V 0A6, Canada; Département de Chimie and PROTEO, Université Laval, Québec, QC G1V 0A6, Canada
| | - Christopher Bérubé
- Département de Chimie and PROTEO, Université Laval, Québec, QC G1V 0A6, Canada
| | - Normand Voyer
- Département de Chimie and PROTEO, Université Laval, Québec, QC G1V 0A6, Canada. normand.voyer.@chm.ulaval.ca
| | - Daniel Grenier
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, 2420, rue de la Terrasse, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
16
|
Binshabaib M, ALHarthi SS, Salehpoor D, Michelogiannakis D, Javed F. Contribution of herpesviruses in the progression of periodontal and peri-implant diseases in systemically healthy individuals. Rev Med Virol 2018; 28:e1996. [DOI: 10.1002/rmv.1996] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 06/18/2018] [Accepted: 06/21/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Munerah Binshabaib
- Department of Periodontology, College of Dentistry; Princess Nourah Bint Abdulrahman University; Riyadh Saudi Arabia
| | - Shatha Subhi ALHarthi
- Department of Periodontology, College of Dentistry; Princess Nourah Bint Abdulrahman University; Riyadh Saudi Arabia
| | - Danial Salehpoor
- Department of General Dentistry, Eastman Institute for Oral Health; University of Rochester; Rochester New York USA
| | - Dimitrios Michelogiannakis
- Departments of Community Dentistry and Oral Disease Prevention and Orthodontics and Dentofacial Orthopedics, Eastman Institute for Oral Health; University of Rochester; Rochester New York USA
| | - Fawad Javed
- Department of General Dentistry, Eastman Institute for Oral Health; University of Rochester; Rochester New York USA
| |
Collapse
|