1
|
Tao J, Sun Y, Wang G, Sun J, Dong S, Ding J. Advanced biomaterials for targeting mature biofilms in periodontitis therapy. Bioact Mater 2025; 48:474-492. [PMID: 40093304 PMCID: PMC11910363 DOI: 10.1016/j.bioactmat.2025.02.026] [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: 11/22/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 03/19/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease primarily caused by bacteria, leading to inflamed and bleeding gums, periodontal pocket formation, and bone loss. Affecting 70%-90% of adults over 65, periodontitis is a leading cause of tooth loss and significantly impacts quality of life. Standard treatments, including subgingival scraping and antibiotics, have limitations, and antibiotic resistance among periodontal pathogens is an increasing concern. Biofilms are barriers to drugs and immune responses, contributing to bacterial resistance and reducing antibiotic effectiveness. Due to their adjustable physicochemical properties, bioactive materials potentially eliminate bacterial biofilms, presenting a promising alternative for periodontitis therapy. In this review, the recent innovations in biomaterials for removing mature biofilms in periodontitis are examined, and their broader potential is discussed. Additionally, the compositions of bacterial biofilms, formation pathways, and intrinsic drug resistance mechanisms are discussed. Finally, the strategies for optimizing subgingival biofilm removal in periodontitis are highlighted, such as targeting biofilms-embedded bacteria, disrupting the extracellular polymeric substances, and utilizing combined approaches. A comprehensive understanding of the properties of biomaterials guides the rational design of highly targeted and effective therapies for periodontitis.
Collapse
Affiliation(s)
- Jiawen Tao
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun, 130021, PR China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Yirong Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Guoliang Wang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Jingru Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
| | - Shujun Dong
- The First Outpatient Department, Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 6822 Jinhu Road, Changchun, 130021, PR China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, PR China
| |
Collapse
|
2
|
Subramani T, Saravanan H, David H, Solanke J, Rajaramon S, Dandela R, Solomon AP. Bioorganic compounds in quorum sensing disruption: strategies, Mechanisms, and future prospects. Bioorg Chem 2025; 156:108192. [PMID: 39874908 DOI: 10.1016/j.bioorg.2025.108192] [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: 08/16/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 01/30/2025]
Abstract
Recent research has shed light on the complex world of bacterial communication through quorum sensing. This sophisticated intercellular signalling mechanism, driven by auto-inducers, regulates crucial bacterial community behaviours such as biofilm formation, expression of virulence factors, and resistance mechanisms. The increasing threat of antibiotic resistance, coupled with quorum sensing mediated response, necessitates alternative strategies to combat bacterial infections. Quorum quenching has emerged as a promising approach, utilizing quorum quenching enzymes and quorum sensing inhibitors to disrupt quorum sensing signalling pathways, thus reducing virulence and biofilm formation. This review focuses on natural and synthetic bioorganic compounds that act as quorum-sensing inhibitors, providing insights into their mechanisms, structure-activity relationships, and potential as anti-virulence agents. The review also explores the communication languages of bacteria, including AHLs in gram-negative bacteria, oligopeptides in gram-positive bacteria, and LuxS, a universal microbial language. By highlighting recent advancements and prospects in bioorganic QSIs, this article underscores their crucial role in developing effective anti-virulence therapies and combating the growing threat of antimicrobial resistance.
Collapse
Affiliation(s)
- Tarunkarthick Subramani
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Harish Saravanan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Helma David
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India
| | - Jayshree Solanke
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India
| | - Shobana Rajaramon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| | - Rambabu Dandela
- Department of Industrial and Engineering Chemistry, Institute of Chemical Technology, Bhubaneswar, Odisha, India.
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur 613401, India.
| |
Collapse
|
3
|
Wang Y, Ma J, Wang H, Yi J, Bai Y, Hu M, Yan J. Mesalazine: a novel therapeutic agent for periodontitis via regulation of periodontal microbiota and inhibiting Porphyromonas gingivalis. Front Microbiol 2025; 16:1531258. [PMID: 39911249 PMCID: PMC11794529 DOI: 10.3389/fmicb.2025.1531258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025] Open
Abstract
Introduction Periodontitis and inflammatory bowel disease are chronic inflammatory diseases with shared epidemiological, biological, and therapeutic associations. Given the similarities in their pathogenic factors, this study hypothesized that mesalazine, a key treatment agent for inflammatory bowel disease, could also be effective in managing periodontitis. Methods The antimicrobial effect of mesalazine on Porphyromonas gingivalis was investigated in vitro, including observations of morphological changes on the surface of P. gingivalis. Additionally, the impact of mesalazine on both the formation and established plaque biofilms was examined. The antimicrobial mechanism was elucidated by assessing the expression of P. gingivalis virulence genes and by determining the disruptive effect on P. gingivalis cell membranes. An in vivo rat model of periodontitis was constructed to evaluate mesalazine's efficacy and its influence on the periodontal bacterial flora in the context of periodontitis. Results and discussion Our results demonstrated that mesalazine concentrations ranging from 0.5 to 2 mg/mL significantly inhibited P. gingivalis proliferation over 72 h. Flow cytometry revealed a marked reduction in the number of viable cells following mesalazine treatment. At the nanometer scale, mesalazine induced crumpling and rupture of the P. gingivalis surface, compromising cell membrane integrity. Mesalazine not only suppressed the formation of plaque biofilms by P. gingivalis and polymicrobial communities but also disrupted pre-existing biofilms. The data also suggested that mesalazine could disrupt the integrity of the P. gingivalis cell membrane and inhibit the expression of virulence factors. An animal model of periodontitis in rats was successfully constructed in vivo. Mesalazine treatment inhibited alveolar bone resorption, alleviated inflammation of periodontal tissues, and improved the composition of the periodontal flora to a healthier state. This study establishes that mesalazine can treat periodontitis through modulation of the periodontal flora and its anti-inflammatory properties, thus broadening its classical therapeutic applications.
Collapse
Affiliation(s)
- Yuqi Wang
- School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jun Ma
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Haoyu Wang
- School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | | | - Yuxin Bai
- School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Min Hu
- School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jiaqing Yan
- School and Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
4
|
Yang L, Wang G, Ma Y, Zhao Q, Zhao H, Wang Q, Zhong C, Zhang C, Yang Y. TRPML1 acts as a predisposing factor in lymphedema development by regulating the subcellular localization of aquaporin-3, -5. PLoS One 2024; 19:e0310653. [PMID: 39637010 PMCID: PMC11620549 DOI: 10.1371/journal.pone.0310653] [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: 04/27/2024] [Accepted: 09/04/2024] [Indexed: 12/07/2024] Open
Abstract
An imbalance in lymphatic fluid, whether it is caused by generation, transport, outflow, or dysfunctional vessels, can lead to lymphedema; however, the exact pathogenesis of this disease remains unclear. To explore the mechanism, we focused on the association among TRPML1, aquaporin-3 (AQP3), and aquaporin-5 (AQP5) in human lymphatic endothelial cells (HLECs). We explored the role of TRPML1 in altering the permeability of HLECs in lymphedema. Meanwhile, we constructed a disease model using gene-knockout mice to observe the effect of TRPML1 on inflammation and fibrosis in lymphedema sites. Our results indicate that TRPML1 not only regulates the localization of AQP3, -5 to the cell membrane but also increases HLEC permeability, disrupts lymphatic fluid transport, and mediates the development of chronic inflammation at the site of lymphedema. Our study suggests that TRPML1 is a precipitating factor in lymphedema. Our findings improve the understanding of TRPML1 and aquaporins in secondary lymphedema, providing valuable insights for future research.
Collapse
Affiliation(s)
- Lijie Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Guanzheng Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yuan Ma
- College of Basic Medicine, Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Qiancheng Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - He Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Qi Wang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chonghua Zhong
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Chunmei Zhang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| | - Yiming Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
- College of Basic Medical Sciences, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
5
|
Mitra A. Combatting biofilm-mediated infections in clinical settings by targeting quorum sensing. Cell Surf 2024; 12:100133. [PMID: 39634722 PMCID: PMC11615143 DOI: 10.1016/j.tcsw.2024.100133] [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: 09/08/2024] [Revised: 11/05/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024] Open
Abstract
Biofilm-associated infections constitute a significant challenge in managing infectious diseases due to their high resistance to antibiotics and host immune responses. Biofilms are responsible for various infections, including urinary tract infections, cystic fibrosis, dental plaque, bone infections, and chronic wounds. Quorum sensing (QS) is a process of cell-to-cell communication that bacteria use to coordinate gene expression in response to cell density, which is crucial for biofilm formation and maintenance.. Its disruption has been proposed as a potential strategy to prevent or treat biofilm-associated infections leading to improved treatment outcomes for infectious diseases. This review article aims to provide a comprehensive overview of the literature on QS-mediated disruption of biofilms for treating infectious diseases. It will discuss the mechanisms of QS disruption and the various approaches that have been developed to disrupt QS in reference to multiple clinical pathogens. In particular, numerous studies have demonstrated the efficacy of QS disruption in reducing biofilm formation in various pathogens, including Pseudomonas aeruginosa and Staphylococcus aureus. Finally, the review will discuss the challenges and future directions for developing QS disruption as a clinical therapy for biofilm-associated infections. This includes the development of effective delivery systems and the identification of suitable targets for QS disruption. Overall, the literature suggests that QS disruption is a promising alternative to traditional antibiotic treatment for biofilm-associated infections and warrants further investigation.
Collapse
Affiliation(s)
- Arindam Mitra
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, India
| |
Collapse
|
6
|
Markowska K, Szymanek-Majchrzak K, Pituch H, Majewska A. Understanding Quorum-Sensing and Biofilm Forming in Anaerobic Bacterial Communities. Int J Mol Sci 2024; 25:12808. [PMID: 39684519 DOI: 10.3390/ijms252312808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Biofilms are complex, highly organized structures formed by microorganisms, with functional cell arrangements that allow for intricate communication. Severe clinical challenges occur when anaerobic bacterial species establish long-lasting infections, especially those involving biofilms. These infections can occur in device-related settings (e.g., implants) as well as in non-device-related conditions (e.g., inflammatory bowel disease). Within biofilms, bacterial cells communicate by producing and detecting extracellular signals, particularly through specific small signaling molecules known as autoinducers. These quorum-sensing signals are crucial in all steps of biofilm formation: initial adhesion, maturation, and dispersion, triggering gene expression that coordinates bacterial virulence factors, stimulates immune responses in host tissues, and contributes to antibiotic resistance development. Within anaerobic biofilms, bacteria communicate via quorum-sensing molecules such as N-Acyl homoserine lactones (AHLs), autoinducer-2 (AI-2), and antimicrobial molecules (autoinducing peptides, AIPs). To effectively combat pathogenic biofilms, understanding biofilm formation mechanisms and bacterial interactions is essential. The strategy to disrupt quorum sensing, termed quorum quenching, involves methods like inactivating or enzymatically degrading signaling molecules, competing with signaling molecules for binding sites, or noncompetitively binding to receptors, and blocking signal transduction pathways. In this review, we comprehensively analyzed the fundamental molecular mechanisms of quorum sensing in biofilms formed by anaerobic bacteria. We also highlight quorum quenching as a promising strategy to manage bacterial infections associated with anaerobic bacterial biofilms.
Collapse
Affiliation(s)
- Kinga Markowska
- Department of Medical Microbiology, Medical University of Warsaw, 5 Chalubinski Str., 02-004 Warsaw, Poland
| | - Ksenia Szymanek-Majchrzak
- Department of Medical Microbiology, Medical University of Warsaw, 5 Chalubinski Str., 02-004 Warsaw, Poland
| | - Hanna Pituch
- Department of Medical Microbiology, Medical University of Warsaw, 5 Chalubinski Str., 02-004 Warsaw, Poland
| | - Anna Majewska
- Department of Medical Microbiology, Medical University of Warsaw, 5 Chalubinski Str., 02-004 Warsaw, Poland
| |
Collapse
|
7
|
Eltarahony M, El-Deeb N, Abu-Serie M, El-Shall H. Biovalorization of whey waste as economic nutriment for mycogenic production of single cell oils with promising antibiofilm and anticancer potentiality. J Biol Eng 2024; 18:62. [PMID: 39497156 PMCID: PMC11533293 DOI: 10.1186/s13036-024-00455-y] [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: 07/06/2024] [Accepted: 10/05/2024] [Indexed: 11/06/2024] Open
Abstract
The production of value-added bio-compounds from rejuvenated sources and their recruitment for healthcare services are paramount objectives in the agenda of white biotechnology. Hereupon, the current study focused on economic production of single cell oils (SCOs) from oleaginous fungi Alternaria sp. (A-OS) and Drechslera sp. (D-OS) using cheese whey waste stream, followed by their evaluation as antibiofilm and anticancer agents, for the first time. As a sole substrate for growth, the whey aided in lipid accumulation by 3.22 and 4.33 g/L, which representing 45.3 and 48.2% lipid content in Drechslera sp. (D-OS) and Alternaria sp. (A-OS), respectively. Meanwhile, a higher unsaturation degree was detected in A-OS by 62.18% comparing to 53.15% of D-OS, with advantageous presence of omega-6 poly unsaturated fatty acid by 22.67% and 15.04% for A-OS and D-OD, respectively, as revealed by GC-MS and FTIR characterization analysis. Interestingly, an eminent and significant (P ≤ 0.05) antibiofilm potency was observed in a dose-dependent modality upon employing both SCOs as antibiofilm agents. Whereas, 100 µg/mL of A-OS recorded superior inhibition of P. aeruginosa, S. aureus and C. albicans biofilms development by 84.10 ± 0.445, 90.37 ± 0.065 and 94.96 ± 0.21%, respectively. Whereas, D-OS (100 µg/mL) thwarted the biofilms of P. aeruginosa, S. aureus and C. albicans by 47.41 ± 2.83, 62.63 ± 5.82 and 78.67 ± 0.23%, correspondingly. Besides, the metabolic performance of cells within biofilm matrix, protein, carbohydrate contents and hydrophobicity of examined biofilms were also curtailed in a significant correlation with biofilm biomass (r ≥ 0.9). Further, as anticancer agents, D-OS recorded higher potency against A549 and CaCo-2 cell lines with IC50 values of 2.55 and 3.425% and SI values of 10.1 and 7.5, respectively. However, A-OS recorded 8.275% and 2.88 for IC50 and SI of Caco-2 cells, respectively. Additionally, A-OS activated caspase 3 by 64.23 ± 1.18% and 53.77 ± 0.995% more than D-OS (52.09 ± 0.222% and 49.72 ± 0.952%) in A549 and Caco-2 cells, respectively. Furthermore, the enzymes, which associated with cancer invasion, metastasis, and angiogenesis (i.e., MMP2 and MMP9) were strongly inhibited by A-OS with 18.58% and 8.295%, respectively as IC50 values; while D-OS results recorded 23.61% and 13.16%, respectively, which could be ascribed to the higher ω-6/ω-3 contents of A-OS. The promising results of the current study opens up the vision to employ SCOs as anti-infective nutraceuticals and in complementary/alternative therapy and prophylactic programs as well.
Collapse
Affiliation(s)
- Marwa Eltarahony
- Environmental Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El‑Arab City, Alexandria, 21934, Egypt.
| | - Nehal El-Deeb
- Pharmaceutical Bioproducts Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria, 21934, Egypt
| | - Marwa Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El‑Arab City, Alexandria, 21934, Egypt
| | - Hadeel El-Shall
- Environmental Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El‑Arab City, Alexandria, 21934, Egypt.
| |
Collapse
|
8
|
Tavares SJS, Pereira CR, Fortes RAM, Alves BES, Fonteles CSR, Wong DVT, Lima-Júnior RCP, Moraes MO, Lima V. Umbelliferone reduces inflammation and ligature-induced osteoclastic alveolar bone resorption in mice. J Periodontal Res 2024; 59:982-992. [PMID: 38742802 DOI: 10.1111/jre.13277] [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: 07/18/2023] [Revised: 04/08/2024] [Accepted: 04/12/2024] [Indexed: 05/16/2024]
Abstract
AIMS This study aimed to investigate the effects of Umbelliferone (UMB) on the inflammation underlying alveolar bone resorption in mouse periodontitis. METHODS Male Swiss mice subjected to a ligature of molars were grouped as non-treated (NT), received UMB (15, 45, or 135 mg/kg) or saline daily for 7 days, respectively, and were compared with naïve mice as control. Gingival tissues were evaluated by myeloperoxidase (MPO) activity and interleukin-1β level by ELISA. The bone resorption was directly assessed on the region between the cement-enamel junction and the alveolar bone crest. Microscopically, histomorphometry of the furcation region, immunofluorescence for nuclear factor-kappa B (NF-ĸB), and immunohistochemistry for tartrate-resistant acid phosphatase (TRAP), and cathepsin K (CTSK) were performed. Systemically, body mass variation and leukogram were analyzed. RESULTS Periodontitis significantly increased MPO activity, interleukin-1β level, and NF-ĸB+ immunofluorescence, and induced severe alveolar bone and furcation resorptions, besides increased TRAP+ and CTSK+ cells compared with naïve. UMB significantly prevented the inflammation by reducing MPO activity, interleukin-1β level, and NF-ĸB+ intensity, besides reduction of resorption of alveolar bone and furcation area, and TRAP+ and CTSK+ cells compared with the NT group. Periodontitis or UMB treatment did not affect the animals systemically. CONCLUSION UMB improved periodontitis by reducing inflammation and bone markers.
Collapse
Affiliation(s)
- Samia Jessica Silva Tavares
- Faculty of Pharmacy, Dentistry and Nursing, Course of Dentistry, Federal University of Ceará, Fortaleza, Brazil
| | - Camila Rodrigues Pereira
- Faculty of Pharmacy, Dentistry and Nursing, Course of Dentistry, Federal University of Ceará, Fortaleza, Brazil
| | | | - Bianca Elen Souza Alves
- Faculty of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Center for Drug Research and Development, Federal University of Ceará, Fortaleza, Brazil
| | - Cristiane Sá Roriz Fonteles
- Faculty of Pharmacy, Dentistry and Nursing, Course of Dentistry, Federal University of Ceará, Fortaleza, Brazil
| | - Deysi Viviana Tenazoa Wong
- Faculty of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Center for Drug Research and Development, Federal University of Ceará, Fortaleza, Brazil
| | - Roberto César Pereira Lima-Júnior
- Faculty of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Center for Drug Research and Development, Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico Moraes
- Faculty of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Center for Drug Research and Development, Federal University of Ceará, Fortaleza, Brazil
| | - Vilma Lima
- Faculty of Medicine, Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Center for Drug Research and Development, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
9
|
Conte R, Valentino A, De Luca I, Soares Pontes G, Calarco A, Cerruti P. Thermo-Responsive Hydrogel Containing Microfluidic Chitosan Nanoparticles Loaded with Opuntia ficus-indica Extract for Periodontitis Treatment. Int J Mol Sci 2024; 25:9374. [PMID: 39273327 PMCID: PMC11395269 DOI: 10.3390/ijms25179374] [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: 07/30/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease resulting from the dysbiosis of periodontal bacteria and the host's immune response, leading to tissue degradation and sustained inflammation. Traditional treatments, such as mechanical debridement and antimicrobial agents, often fail to fully eradicate pathogenic bacteria, especially in deep periodontal pockets. Consequently, the need for novel therapeutic approaches has increased the interest in bioactive natural extracts, such as that of Opuntia ficus-indica, known for its anti-inflammatory, antioxidant, and antimicrobial properties. This study investigates the encapsulation of Opuntia ficus-indica extract in OFI-loaded chitosan nanoparticles (OFI-NPs) via ionotropic gelation using a microfluidic system, allowing precise control over nanoparticle characteristics and enhancing protection against enzymatic degradation. To achieve localized and sustained release in periodontal pockets, a thermo-responsive hydrogel comprising hyaluronic acid and Pluronic F127 (OFI@tgels) was developed. The transition of OFI@tgels from a solution at low temperatures to a solid at body temperature enables prolonged drug release at inflammation sites. The in vitro application of the optimized formulation eradicated biofilms of S. mutans, P. aeruginosa (PAO1), and P. gingivalis over 36 h and disrupted extracellular polymeric substance formation. Additionally, OFI@tgel modulated immune responses by inhibiting M1 macrophage polarization and promoting a shift to the M2 phenotype. These findings suggest that OFI@tgel is a promising alternative treatment for periodontitis, effectively reducing biofilm formation and modulating the immune response.
Collapse
Affiliation(s)
- Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Anna Valentino
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Ilenia De Luca
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Gemilson Soares Pontes
- Laboratory of Virology and Immunology, National Institute of Amazonian Research (INPA), Manaus 69067-375, AM, Brazil
- Post-Graduate Program in Basic and Applied Immunology, Institute of Biological Science, Federal University of Amazonas, Manaus 69077-000, AM, Brazil
| | - Anna Calarco
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Pierfrancesco Cerruti
- Institute for Polymers, Composites, and Biomaterials (IPCB-CNR), Via Campi Flegrei 34, 80078 Pozzuoli, Italy
| |
Collapse
|
10
|
Chen Z, Lang G, Xu X, Liang X, Han Y, Han Y. The role of NF-kappaB in the inflammatory processes related to dental caries, pulpitis, apical periodontitis, and periodontitis-a narrative review. PeerJ 2024; 12:e17953. [PMID: 39221277 PMCID: PMC11366231 DOI: 10.7717/peerj.17953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Tooth-related inflammatory disorders, including caries, pulpitis, apical periodontitis (AP), and periodontitis (PD), are primarily caused by resident oral microorganisms. Although these dental inflammatory conditions are typically not life-threatening, neglecting them can result in significant complications and greatly reduce an individual's quality of life. Nuclear factor κB (NF-κB), a family formed by various combinations of Rel proteins, is extensively involved in inflammatory diseases and even cancer. This study reviews recent data on NF-κB signaling and its role in dental pulp stem cells (DPSCs), dental pulp fibroblasts (DPFs), odontoblasts, human periodontal ligament cells (hPDLCs), and various experimental animal models. The findings indicate that NF-κB signaling is abnormally activated in caries, pulpitis, AP, and PD, leading to changes in related cellular differentiation. Under specific conditions, NF-κB signaling occasionally interacts with other signaling pathways, affecting inflammation, bone metabolism, and tissue regeneration processes. In summary, data collected over recent years confirm the central role of NF-κB in dental inflammatory diseases, potentially providing new insights for drug development targeting NF-κB signaling pathways in the treatment of these conditions. Keywords: NF-κB, dental caries, pulpitis, apical periodontitis, periodontitis.
Collapse
Affiliation(s)
- Zhonglan Chen
- Zunyi Medical University, Special Key Laboratory of Oral Diseases Research, Hospital/School of Stomatology, Zunyi, Guizhou, China
| | - Guangping Lang
- Zunyi Medical University, Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi, Guizhou, China
| | - Xi Xu
- Zunyi Medical University, Special Key Laboratory of Oral Diseases Research, Hospital/School of Stomatology, Zunyi, Guizhou, China
| | - Xinghua Liang
- Zunyi Medical University, Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi, Guizhou, China
| | - Yalin Han
- Zunyi Medical University, Special Key Laboratory of Oral Diseases Research, Hospital/School of Stomatology, Zunyi, Guizhou, China
| | - Yingying Han
- Zunyi Medical University, Special Key Laboratory of Oral Diseases Research, Hospital/School of Stomatology, Zunyi, Guizhou, China
| |
Collapse
|
11
|
Nadaf R, Kumbar VM, Ghagane S. Unravelling the intricacies of Porphyromonas gingivalis: virulence factors, lifecycle dynamics and phytochemical interventions for periodontal disease management. APMIS 2024. [PMID: 39030947 DOI: 10.1111/apm.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024]
Abstract
Porphyromonas gingivalis is a gram-negative anaerobic bacterium recognized for its pivotal role in the pathogenesis of periodontal diseases. This review covers an overview of the virulence factors and lifecycle stages of P. gingivalis, with a specific focus on attachment and colonization, biofilm formation, growth and multiplication, dormancy survival and dissemination. Additionally, we explore the significance of inter-bacterial cross-feeding within biofilms. Furthermore, we discuss potential phytochemical-based strategies to target P. gingivalis, including the use of curcumin, apigenin, quercetin and resveratrol. Understanding the virulence factors and lifecycle stages of P. gingivalis, along with the promising phytochemical-based interventions, holds promise for advancing strategies in periodontal disease management and oral health promotion.
Collapse
Affiliation(s)
- Rubeen Nadaf
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Vijay M Kumbar
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Shridhar Ghagane
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| |
Collapse
|
12
|
Li M, Cruz CD, Ilina P, Tammela P. High-throughput combination assay for studying biofilm formation of uropathogenic Escherichia coli. Arch Microbiol 2024; 206:344. [PMID: 38967798 PMCID: PMC11226472 DOI: 10.1007/s00203-024-04029-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: 05/23/2024] [Revised: 05/23/2024] [Accepted: 06/03/2024] [Indexed: 07/06/2024]
Abstract
Uropathogenic Escherichia coli, the most common cause for urinary tract infections, forms biofilm enhancing its antibiotic resistance. To assess the effects of compounds on biofilm formation of uropathogenic Escherichia coli UMN026 strain, a high-throughput combination assay using resazurin followed by crystal violet staining was optimized for 384-well microplate. Optimized assay parameters included, for example, resazurin and crystal violet concentrations, and incubation time for readouts. For the assay validation, quality parameters Z' factor, coefficient of variation, signal-to-noise, and signal-to-background were calculated. Microplate uniformity, signal variability, edge well effects, and fold shift were also assessed. Finally, a screening with known antibacterial compounds was conducted to evaluate the assay performance. The best conditions found were achieved by using 12 µg/mL resazurin for 150 min and 0.023% crystal violet. This assay was able to detect compounds displaying antibiofilm activity against UMN026 strain at sub-inhibitory concentrations, in terms of metabolic activity and/or biomass.
Collapse
Affiliation(s)
- M Li
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki, FI-00014, Finland
| | - C D Cruz
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki, FI-00014, Finland
| | - P Ilina
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki, FI-00014, Finland
| | - P Tammela
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, Helsinki, FI-00014, Finland.
| |
Collapse
|
13
|
D’Aquila P, De Rose E, Sena G, Scorza A, Cretella B, Passarino G, Bellizzi D. Quorum Quenching Approaches against Bacterial-Biofilm-Induced Antibiotic Resistance. Antibiotics (Basel) 2024; 13:619. [PMID: 39061301 PMCID: PMC11273524 DOI: 10.3390/antibiotics13070619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
With the widespread phenomenon of antibiotic resistance and the diffusion of multiple drug-resistant bacterial strains, enormous efforts are being conducted to identify suitable alternative agents against pathogenic microorganisms. Since an association between biofilm formation and antibiotic resistance phenotype has been observed, a promising strategy pursued in recent years focuses on controlling and preventing this formation by targeting and inhibiting the Quorum Sensing (QS) system, whose central role in biofilm has been extensively demonstrated. Therefore, the research and development of Quorum Quenching (QQ) compounds, which inhibit QS, has gradually attracted the attention of researchers and has become a new strategy for controlling harmful microorganisms. Among these, a number of both natural and synthetic compounds have been progressively identified as able to interrupt the intercellular communication within a microbial community and the adhesion to a surface, thus disintegrating mature/preformed biofilms. This review describes the role played by QS in the formation of bacterial biofilms and then focuses on the mechanisms of different natural and synthetic QS inhibitors (QSIs) exhibiting promising antibiofilm ability against Gram-positive and Gram-negative bacterial pathogens and on their applications as biocontrol strategies in various fields.
Collapse
Affiliation(s)
- Patrizia D’Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Elisabetta De Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Giada Sena
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Angelo Scorza
- Villa Ermelinda, Progetto Terza Età, 88842 Cutro, Italy; (A.S.); (B.C.)
| | | | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| |
Collapse
|
14
|
Fang L, Zhang Y, Cheng L, Zheng H, Wang Y, Qin L, Cai Y, Cheng L, Zhou W, Liu F, Wang S. Silica nanoparticles containing nano-silver and chlorhexidine to suppress Porphyromonas gingivalis biofilm and modulate multispecies biofilms toward healthy tendency. J Oral Microbiol 2024; 16:2361403. [PMID: 38847000 PMCID: PMC11155433 DOI: 10.1080/20002297.2024.2361403] [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: 09/19/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Objectives This research first investigated the effect of mesoporous silica nanoparticles (nMS) carrying chlorhexidine and silver (nMS-nAg-Chx) on periodontitis-related biofilms. This study aimed to investigate (1) the antibacterial activity on Porphyromonas gingivalis (P. gingivalis) biofilm; (2) the suppressing effect on virulence of P. gingivalis biofilm; (3) the regulating effect on periodontitis-related multispecies biofilm. Methods Silver nanoparticles (nAg) and chlorhexidine (Chx) were co-loaded into nMS to form nMS-nAg-Chx. Inhibitory zone test and minimum inhibitory concentration (MIC) against P. gingivalis were tested. Growth curves, crystal violet (CV) staining, live/dead staining and scanning electron microscopy (SEM) observation were performed. Biofilm virulence was assessed. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and Quantitative Real Time-PCR (qPCR) were performed to validate the activity and composition changes of multispecies biofilm (P. gingivalis, Streptococcus gordonii and Streptococcus sanguinis). Results nMS-nAg-Chx inhibited P. gingivalis biofilm dose-dependently (p<0.05), with MIC of 18.75 µg/mL. There were fewer live bacteria, less biomass and less virulence in nMS-nAg-Chx groups (p<0.05). nMS-nAg-Chx inhibited and modified periodontitis-related biofilms. The proportion of pathogenic bacteria decreased from 16.08 to 1.07% and that of helpful bacteria increased from 82.65 to 94.31% in 25 μg/mL nMS-nAg-Chx group for 72 h. Conclusions nMS-nAg-Chx inhibited P. gingivalis growth, decreased biofilm virulence and modulated periodontitis-related multispecies biofilms toward healthy tendency. pH-sensitive nMS-nAg-Chx inhibit the pathogens and regulate oral microecology, showing great potential in periodontitis adjunctive therapy.
Collapse
Affiliation(s)
- Lixin Fang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yishuang Zhang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Long Cheng
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hao Zheng
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yiyi Wang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lu Qin
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yingchun Cai
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Wen Zhou
- School and Hospital of Stomatology, Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, Fuzhou, China
| | - Fei Liu
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Suping Wang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Ahmed GE, Elshahid ZA, El-Sawy ER, Abdel-Aziz MS, Abdel-Aziem A. Synthesis, biofilm formation inhibitory, and inflammation inhibitory activities of new coumarin derivatives. Sci Rep 2024; 14:9106. [PMID: 38643226 PMCID: PMC11032357 DOI: 10.1038/s41598-024-59072-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: 07/23/2023] [Accepted: 04/07/2024] [Indexed: 04/22/2024] Open
Abstract
Coumarins are heterocycles of great interest in the development of valuable active structures in chemistry and biological domains. The ability of coumarins to inhibit biofilm formation of Gram positive bacterium (Staphylococcus aureus), Gram negative bacterium (Escherichia coli) as well as the methicillin-resistant S. aureus (MRSA) has been previously described. In the present work, new hybrid coumarin-heterocycles have been synthesized via the reaction of coumarin-6-sulfonyl chloride and 6-aminocoumarin with different small heterocycle moieties. The biological efficacy of the new compounds was evaluated towards their ability to inhibit biofilm formation and their anti-inflammatory properties. The antimicrobial activities of the newly synthesized compounds were tested against Gram positive bacterium (S. aureus ATCC 6538), Gram negative bacterium (E. coli ATCC 25922), yeast (Candida albicans ATCC 10231) and the fungus (Aspergillus niger NRRL-A326). Compounds 4d, 4e, 4f, 6a and 9 showed significant MIC and MBC values against S. aureus, E. coli, C. albicans, and methicillin-resistant S. aureus (MRSA) with especial incidence on compound 9 which surpasses all the other compounds giving MIC and MBC values of (4.88 and 9.76 µg/mL for S. aureus), (78.13 and 312.5 µg/mL for E. coli), (9.77 and 78.13 µg/mL for C. albicans), and (39.06 and 76.7 µg/mL for MRSA), respectively. With reference to the antibiofilm activity, compound 9 exhibited potent antibiofilm activity with IC50 of 60, 133.32, and 19.67 µg/mL against S. aureus, E. coli, and MRSA, (respectively) considering the reference drug (neomycin). Out of all studied compounds, the anti-inflammatory results indicated that compound 4d effectively inhibited nitric oxide production in lipopolysaccharide-(LPS-) stimulated RAW264.7 macrophage cells, giving NO% inhibition of 70% compared to Sulindac (55.2%).
Collapse
Affiliation(s)
- Ghada E Ahmed
- High Canal Institute of Engineering & Technology, Suez, Egypt
| | - Zeinab A Elshahid
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Giza, Egypt
| | - Eslam Reda El-Sawy
- Chemistry of Natural Compounds Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Mohamed S Abdel-Aziz
- Microbial Chemistry Department, Genetic Engineering and Biotechnology Institute, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Anhar Abdel-Aziem
- Chemistry Department, Faculty of Science (Girl's Branch), Al-Azhar University, Cairo, 11754, Egypt.
| |
Collapse
|
16
|
Ustianowska K, Ustianowski Ł, Bakinowska E, Kiełbowski K, Szostak J, Murawka M, Szostak B, Pawlik A. The Genetic Aspects of Periodontitis Pathogenesis and the Regenerative Properties of Stem Cells. Cells 2024; 13:117. [PMID: 38247810 PMCID: PMC10814055 DOI: 10.3390/cells13020117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/23/2024] Open
Abstract
Periodontitis (PD) is a prevalent and chronic inflammatory disease with a complex pathogenesis, and it is associated with the presence of specific pathogens, such as Porphyromonas gingivalis. Dysbiosis and dysregulated immune responses ultimately lead to chronic inflammation as well as tooth and alveolar bone loss. Multiple studies have demonstrated that genetic polymorphisms may increase the susceptibility to PD. Furthermore, gene expression is modulated by various epigenetic mechanisms, such as DNA methylation, histone modifications, or the activity of non-coding RNA. These processes can also be induced by PD-associated pathogens. In this review, we try to summarize the genetic processes that are implicated in the pathogenesis of PD. Furthermore, we discuss the use of these mechanisms in diagnosis and therapeutic purposes. Importantly, novel treatment methods that could promote tissue regeneration are greatly needed in PD. In this paper, we also demonstrate current evidence on the potential use of stem cells and extracellular vesicles to stimulate tissue regeneration and suppress inflammation. The understanding of the molecular mechanisms involved in the pathogenesis of PD, as well as the impact of PD-associated bacteria and stem cells in these processes, may enhance future research and ultimately improve long-term treatment outcomes.
Collapse
Affiliation(s)
- Klaudia Ustianowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Łukasz Ustianowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Joanna Szostak
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Martyna Murawka
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Bartosz Szostak
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.U.); (Ł.U.); (E.B.); (K.K.); (M.M.); (B.S.)
| |
Collapse
|
17
|
Kalia VC, Patel SKS, Lee JK. Bacterial biofilm inhibitors: An overview. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115389. [PMID: 37634478 DOI: 10.1016/j.ecoenv.2023.115389] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/05/2023] [Accepted: 08/17/2023] [Indexed: 08/29/2023]
Abstract
Bacteria that cause infectious diseases adopt biofilms as one of their most prevalent lifestyles. Biofilms enable bacteria to tolerate environmental stress and evade antibacterial agents. This bacterial defense mechanism has rendered the use of antibiotics ineffective for the treatment of infectious diseases. However, many highly drug-resistant microbes have rapidly emerged owing to such treatments. Different signaling mechanisms regulate bacterial biofilm formation, including cyclic dinucleotide (c-di-GMP), small non-coding RNAs, and quorum sensing (QS). A cell density-dependent phenomenon, QS is associated with c-di-GMP (a global messenger), which regulates gene expression related to adhesion, extracellular matrix production, the transition from the planktonic to biofilm stage, stability, pathogenicity, virulence, and acquisition of nutrients. The article aims to provide information on inhibiting biofilm formation and disintegrating mature/preformed biofilms. This treatment enables antimicrobials to target the free-living/exposed bacterial cells at lower concentrations than those needed to treat bacteria within the biofilm. Therefore, a complementary action of antibiofilm and antimicrobial agents can be a robust strategic approach to dealing with infectious diseases. Taken together, these molecules have broad implications for human health.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sanjay K S Patel
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
18
|
Zhang Z, Sun Y, Yi Y, Bai X, Zhu L, Zhu J, Gu M, Zhu Y, Jiang L. Screening and Identification of a Streptomyces Strain with Quorum-Sensing Inhibitory Activity and Effect of the Crude Extracts on Virulence Factors of Pseudomonas aeruginosa. Microorganisms 2023; 11:2079. [PMID: 37630639 PMCID: PMC10458028 DOI: 10.3390/microorganisms11082079] [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: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Quorum-sensing (QS) is involved in numerous physiological processes in bacteria, such as biofilm formation, sporulation, and virulence formation. Therefore, the search for new quorum-sensing inhibitors (QSI) is a promising strategy that opens up a new perspective for controlling QS-mediated bacterial pathogens. To explore new QSIs, a strain named Streptomyces sp. D67 with QS inhibitory activity was isolated from the soil of the arid zone around the Kumutag Desert in Xinjiang. Phylogenetic analyses demonstrated that strain D67 shared the highest similarity with Streptomyces ardesiacus NBRC 15402T (98.39%), which indicated it represented a potential novel species in the Streptomyces genus. The fermentation crude extracts of strain D67 can effectively reduce the violacein production produced by Chromobacterium violaceum CV026 and the swarming and swimming abilities of Pseudomonas aeruginosa. It also has significant inhibitory activity on the production of virulence factors such as biofilm, pyocyanin, and rhamnolipids of P. aeruginosa in a significant concentration-dependent manner, but not on protease activity. A total of 618 compounds were identified from the fermentation crude extracts of strain D67 by LC-MS, and 19 compounds with significant QS inhibitory activity were observed. Overall, the strain with QS inhibitory activity was screened from Kumutag Desert in Xinjiang for the first time, which provided a basis for further research and development of new QSI.
Collapse
Affiliation(s)
- Zhidong Zhang
- Xinjiang Key Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; (Z.Z.); (Y.Y.); (J.Z.)
- College of Life Sciences, Xinjiang Normal University, Urumqi 830054, China;
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China;
| | - Yang Sun
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China;
| | - Yuanyang Yi
- Xinjiang Key Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; (Z.Z.); (Y.Y.); (J.Z.)
- College of Life Sciences, Xinjiang Normal University, Urumqi 830054, China;
| | - Xiaoyu Bai
- Xinjiang Key Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; (Z.Z.); (Y.Y.); (J.Z.)
- College of Life Sciences, Xinjiang Normal University, Urumqi 830054, China;
| | - Liying Zhu
- College of Chemical and Molecular Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Jing Zhu
- Xinjiang Key Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; (Z.Z.); (Y.Y.); (J.Z.)
| | - Meiying Gu
- Xinjiang Key Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; (Z.Z.); (Y.Y.); (J.Z.)
| | - Yanlei Zhu
- College of Life Sciences, Xinjiang Normal University, Urumqi 830054, China;
| | - Ling Jiang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing 211816, China;
| |
Collapse
|
19
|
Li F, Wang C, Xu J, Wang X, Cao M, Wang S, Zhang T, Xu Y, Wang J, Pan S, Hu W. Evaluation of the antibacterial activity of Elsholtzia ciliate essential oil against halitosis-related Fusobacterium nucleatum and Porphyromonas gingivalis. Front Microbiol 2023; 14:1219004. [PMID: 37608950 PMCID: PMC10440386 DOI: 10.3389/fmicb.2023.1219004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023] Open
Abstract
The broad-spectrum antimicrobial activity of Elsholtzia ciliate essential oil (ECO) has been previously reported, but its effectiveness against halitosis-causing bacteria such as Fusobacterium nucleatum and Porphyromonas gingivalis is not well understood. In this study, we investigated the bacteriostatic activity of ECO against planktonic cells and biofilms of F. nucleatum and P. gingivalis, as well as its ability to inhibit bacterial metabolism and production of volatile sulfur compounds (VSCs) at sub-lethal concentrations. Our findings revealed that ECO exhibited comparable activities to chlorhexidine against these oral bacteria. Treatment with ECO significantly reduced the production of VSCs, including hydrogen sulfide, dimethyl disulfide, and methanethiol, which are major contributors to bad breath. As the major chemical components of ECO, carvacrol, p-cymene, and phellandrene, were demonstrated in vitro inhibitory effects on F. nucleatum and P. gingivalis, and their combined use showed synergistic and additive effects, suggesting that the overall activity of ECO is derived from the cumulative or synergistic effect of multiple active components. ECO was found to have a destructive effect on the bacterial cell membrane by examining the cell morphology and permeability. Furthermore, the application of ECO induced significant changes in the bacterial composition of saliva-derived biofilm, resulting in the elimination of bacterial species that contribute to halitosis, including Fusobacterium, Porphyromonas, and Prevotella. These results provide experimental evidence for the potential clinical applications of ECOs in the prevention and treatment of halitosis.
Collapse
Affiliation(s)
- Fengjiao Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chuandong Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Jing Xu
- Shenzhen RELX Technology Co., Ltd., Shenzhen, China
| | - Xiaoyu Wang
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Meng Cao
- Shandong Aobo Biotechnology Co., Ltd., Liaocheng, China
| | - Shuhua Wang
- Shandong Aobo Biotechnology Co., Ltd., Liaocheng, China
| | | | - Yanyong Xu
- Beijing Xinyue Technology Co., Ltd., Beijing, China
| | - Jing Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shaobin Pan
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Hu
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| |
Collapse
|
20
|
El-Naggar NEA, Dalal SR, Zweil AM, Eltarahony M. Artificial intelligence-based optimization for chitosan nanoparticles biosynthesis, characterization and in‑vitro assessment of its anti-biofilm potentiality. Sci Rep 2023; 13:4401. [PMID: 36928367 PMCID: PMC10019797 DOI: 10.1038/s41598-023-30911-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Chitosan nanoparticles (CNPs) are promising biopolymeric nanoparticles with excellent physicochemical, antimicrobial, and biological properties. CNPs have a wide range of applications due to their unique characteristics, including plant growth promotion and protection, drug delivery, antimicrobials, and encapsulation. The current study describes an alternative, biologically-based strategy for CNPs biosynthesis using Olea europaea leaves extract. Face centered central composite design (FCCCD), with 50 experiments was used for optimization of CNPs biosynthesis. The artificial neural network (ANN) was employed for analyzing, validating, and predicting CNPs biosynthesis using Olea europaea leaves extract. Using the desirability function, the optimum conditions for maximum CNPs biosynthesis were determined theoretically and verified experimentally. The highest experimental yield of CNPs (21.15 mg CNPs/mL) was obtained using chitosan solution of 1%, leaves extract solution of 100%, initial pH 4.47, and incubation time of 60 min at 53.83°C. The SEM and TEM images revealed that CNPs had a spherical form and varied in size between 6.91 and 11.14 nm. X-ray diffraction demonstrates the crystalline nature of CNPs. The surface of the CNPs is positively charged, having a Zeta potential of 33.1 mV. FTIR analysis revealed various functional groups including C-H, C-O, CONH2, NH2, C-OH and C-O-C. The thermogravimetric investigation indicated that CNPs are thermally stable. The CNPs were able to suppress biofilm formation by P. aeruginosa, S. aureus and C. albicans at concentrations ranging from 10 to 1500 µg/mL in a dose-dependent manner. Inhibition of biofilm formation was associated with suppression of metabolic activity, protein/exopolysaccharide moieties, and hydrophobicity of biofilm encased cells (r ˃ 0.9, P = 0.00). Due to their small size, in the range of 6.91 to 11.14 nm, CNPs produced using Olea europaea leaves extract are promising for applications in the medical and pharmaceutical industries, in addition to their potential application in controlling multidrug-resistant microorganisms, especially those associated with post COVID-19 pneumonia in immunosuppressed patients.
Collapse
Affiliation(s)
- Noura El-Ahmady El-Naggar
- Department of Bioprocess Development, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt.
| | - Shimaa R Dalal
- Botany Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Amal M Zweil
- Plant Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Marwa Eltarahony
- Environmental Biotechnology Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt
| |
Collapse
|
21
|
Fan R, Zhou Y, Chen X, Zhong X, He F, Peng W, Li L, Wang X, Xu Y. Porphyromonas gingivalis Outer Membrane Vesicles Promote Apoptosis via msRNA-Regulated DNA Methylation in Periodontitis. Microbiol Spectr 2023; 11:e0328822. [PMID: 36629433 PMCID: PMC9927323 DOI: 10.1128/spectrum.03288-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 12/07/2022] [Indexed: 01/12/2023] Open
Abstract
The outer membrane vesicles (OMVs) produced by Porphyromonas gingivalis contain a variety of bioactive molecules that may be involved in the progression of periodontitis. However, the participation of P. gingivalis OMVs in the development of periodontitis has not been elucidated. Here, we isolated P. gingivalis OMVs and confirmed their participation in periodontitis both in vivo and in vitro. Microcomputed tomography (micro-CT) and histological analysis showed that under stimulation with P. gingivalis OMVs, the alveolar bone of rats was significantly resorbed in vivo. We found that P. gingivalis OMVs were taken up by human periodontal ligament cells ([hPDLCs]) in vitro, which subsequently resulted in apoptosis and inflammatory cytokine release, which was accomplished by the microRNA-size small RNA (msRNA) sRNA45033 in the P. gingivalis OMVs. Through bioinformatics analysis and screening of target genes, chromobox 5 (CBX5) was identified as the downstream target of screened-out sRNA45033. Using a dual-luciferase reporter assay, overexpression, and knockdown methods, sRNA45033 was confirmed to target CBX5 to regulate hPDLC apoptosis. In addition, CUT&Tag (cleavage under targets and tagmentation) analysis confirmed the mechanism that CBX5 regulates apoptosis through the methylation of p53 DNA. Collectively, these findings indicate that the role of P. gingivalis OMVs is immunologically relevant and related to bacterial virulence during the development of periodontitis. IMPORTANCE P. gingivalis is a bacterium often associated with periodontitis. This study demonstrates that (i) sRNA45033 in P. gingivalis OMVs targets CBX5, (ii) CBX5 regulates the methylation of p53 DNA and its expression, which is associated with apoptosis, and (iii) a novel mechanism of interaction between hosts and pathogens is mediated by OMVs in the occurrence of periodontitis.
Collapse
Affiliation(s)
- Ruyi Fan
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yi Zhou
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xu Chen
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xianmei Zhong
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Periodontics, Taizhou Stomatological Hospital, Taizhou, China
| | - Fanzhen He
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Wenzao Peng
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Lu Li
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xiaoqian Wang
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yan Xu
- Department of Periodontics, the Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
22
|
Bessa LJ, Botelho J, Machado V, Alves R, Mendes JJ. Managing Oral Health in the Context of Antimicrobial Resistance. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:16448. [PMID: 36554332 PMCID: PMC9778414 DOI: 10.3390/ijerph192416448] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/05/2022] [Accepted: 12/05/2022] [Indexed: 05/25/2023]
Abstract
The oral microbiome plays a major role in shaping oral health/disease state; thus, a main challenge for dental practitioners is to preserve or restore a balanced oral microbiome. Nonetheless, when pathogenic microorganisms install in the oral cavity and are incorporated into the oral biofilm, oral infections, such as gingivitis, dental caries, periodontitis, and peri-implantitis, can arise. Several prophylactic and treatment approaches are available nowadays, but most of them have been antibiotic-based. Given the actual context of antimicrobial resistance (AMR), antibiotic stewardship in dentistry would be a beneficial approach to optimize and avoid inappropriate or even unnecessary antibiotic use, representing a step towards precision medicine. Furthermore, the development of new effective treatment options to replace the need for antibiotics is being pursued, including the application of photodynamic therapy and the use of probiotics. In this review, we highlight the advances undergoing towards a better understanding of the oral microbiome and oral resistome. We also provide an updated overview of how dentists are adapting to better manage the treatment of oral infections given the problem of AMR.
Collapse
Affiliation(s)
- Lucinda J. Bessa
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
| | - João Botelho
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Clinical Research Unit (CRU), CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Evidence-Based Hub, CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
| | - Vanessa Machado
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Clinical Research Unit (CRU), CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Evidence-Based Hub, CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
| | - Ricardo Alves
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Clinical Research Unit (CRU), CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
| | - José João Mendes
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Clinical Research Unit (CRU), CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
- Evidence-Based Hub, CiiEM, Egas Moniz—Cooperativa de Ensino Superior, Caparica, 2829-511 Almada, Portugal
| |
Collapse
|