1
|
Xu Z, Wang Y, Li S, Li Y, Chang L, Yao Y, Peng Q. Advances of functional nanomaterials as either therapeutic agents or delivery systems in the treatment of periodontitis. BIOMATERIALS ADVANCES 2025; 175:214326. [PMID: 40300444 DOI: 10.1016/j.bioadv.2025.214326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Periodontitis is a common chronic inflammatory disease primarily caused by pathogenic microorganisms in the oral cavity. Without appropriate treatments, it may lead to the gradual destruction of the supporting tissues of the teeth. While current treatments can alleviate symptoms, they still have limitations, particularly in eliminating pathogenic bacteria, promoting periodontal tissue regeneration, and avoiding antibiotic resistance. In recent years, functional nanomaterials have shown great potential in the treatment of periodontitis due to their unique physicochemical and biological properties. This review summarizes various functionalization strategies of nanomaterials and explores their potential applications in periodontitis treatment, including metal-based nanoparticles, carbon nanomaterials, polymeric nanoparticles, and exosomes. The mechanisms and advances in antibacterial effects, immune regulation, reactive oxygen species (ROS) scavenging, and bone tissue regeneration are discussed in detail. In addition, the challenges and future directions of applying nanomaterials in periodontitis therapy are also discussed.
Collapse
Affiliation(s)
- Ziyi Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shuoshun Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yuanhong Li
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, China
| | - Lili Chang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qiang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Iwasaki A, Hatakeyama M, Liu Q, Orimoto A, Fukuda T, Kitaoka T. Proliferation and differentiation of human dental pulp stem cells on phosphorylated cellulose nanofiber scaffolds. Carbohydr Polym 2025; 359:123593. [PMID: 40306767 DOI: 10.1016/j.carbpol.2025.123593] [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: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025]
Abstract
Human dental pulp stem cells (hDPSCs) are a promising cell source for tooth regeneration therapies. However, conventional culture scaffold materials are often animal-derived, leading to immunogenicity concerns and limited availability. In this study, we explored phosphorylated cellulose nanofibers (P-CNFs), which have a fine fiber morphology and phosphate groups, as a novel scaffold material for cell culture. Immortalized hDPSCs were cultured on P-CNF scaffolds with different phosphate contents (0-1.42 mmol g-1) prepared by varying the molar ratio of urea and diammonium hydrogen phosphate and the reaction time. Cells cultured on unmodified CNFs exhibited poor adhesion and formed spheroids, indicating low bioadaptability. In contrast, P-CNF scaffolds with moderate phosphate content (0.54-0.78 mmol g-1) significantly improved cell adhesion; further increases in phosphate content decreased cell adhesion, indicating a strong dependence on phosphate content. Intriguingly, even in the absence of differentiation inducers, hDPSCs on P-CNF scaffolds with an optimal phosphate content of 0.78 mmol g-1 showed equal or higher expression of hard tissue marker genes compared to collagen scaffolds with differentiation inducers, suggesting that P-CNFs can directly promote hard tissue differentiation. These findings highlight plant-derived, animal-free P-CNFs as a promising biomaterial for advanced dental tissue engineering.
Collapse
Affiliation(s)
- Akihiro Iwasaki
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Mayumi Hatakeyama
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Qimei Liu
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Ai Orimoto
- Division of Endodontics and Restorative Dentistry, Kyushu Dental University, Kitakyushu 803-8580, Japan
| | - Tomokazu Fukuda
- Graduate School of Science and Engineering, Iwate University, Morioka, Iwate 020-8551, Japan
| | - Takuya Kitaoka
- Department of Agro-Environmental Sciences, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka 819-0395, Japan.
| |
Collapse
|
3
|
Yang N, Xia Y, Gao H, Wang C, Jiang Y, Song W, Yu JF, Liang L. Regulatory T Cells promote osteogenic differentiation of periodontal ligament stem cells through the Jagged1-Notch2 signaling Axis. J Dent 2025; 158:105772. [PMID: 40287047 DOI: 10.1016/j.jdent.2025.105772] [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: 11/11/2024] [Revised: 04/13/2025] [Accepted: 04/20/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVES This study aimed to elucidate the role of regulatory T cells (Tregs) in promoting the osteogenic differentiation of periodontal ligament stem cells (PDLSCs) and to investigate the underlying mechanisms involving Notch signaling. METHOD Tregs were isolated via fluorescence-activated cell sorting (FACS) and co-cultured with PDLSCs. Osteogenic differentiation was assessed through in vitro assays and in vivo transplantation experiments. Gene expression profiles were quantified using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analysis. A murine periodontitis model was used to validated therapeutic outcomes, with bone remodeling quantified by micro-CT and histology (H&E, Masson's staining). Immunophenotypic analysis of Jagged1 expression in Tregs and Notch2 receptor localization in PDLSCs were performed using flow cytometry and immunofluorescence microscopy, respectively. The role of immobilized Jagged1 in osteogenic differentiation was further evaluated, while Notch pathway inhibition was achieved via γ-secretase inhibitor DAPT in vitro. RESULTS Elevated levels of Th17 cells and Tregs were observed in peripheral blood samples from periodontitis patients, with a significantly increased Th17/Treg ratio (p < 0.01). In vitro, co-culturing Tregs with PDLSCs significantly enhanced PDLSC osteogenesis, as evidenced by increased ALP activity (p < 0.01), elevated expression of osteogenesis-related genes (Runx2 and Osterix; p < 0.01), and enhanced mineralization (Alizarin Red staining) (p < 0.01). In vivo, intravenous infusion of Tregs into a periodontitis mouse model reduced periodontal damage and promoted bone regeneration, as demonstrated by reduced CEJ-ABC distance and increased BV/TV ratio (p < 0.01). Mechanistically, Tregs expressed the Notch ligand Jagged1 and upregulated Notch2 receptor expression in PDLSCs, indicating activation of the Notch signaling pathway. Jagged1 promoted osteogenic differentiation of PDLSCs in a dose- and time-dependent manner. Inhibition of Notch signaling using DAPT reduced Tregs-mediated enhancement of PDLSC osteogenesis (p < 0.05). CONCLUSION These findings suggest that Tregs promote PDLSC osteogenic differentiation via Jagged1-Notch2 signaling, highlighting the therapeutic potential of modulating Tregs and Notch signaling for periodontal regeneration and bone tissue engineering. CLINICAL SIGNIFICANCE This study provides new insights into the complex interplay between immune modulation and stem cell differentiation, laying the foundation for potential Tregs-based therapeutic strategies for periodontal and bone tissue regeneration.
Collapse
Affiliation(s)
- Nan Yang
- Department of Stomatology, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| | - Yu Xia
- Department of Stomatology, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| | - Hui Gao
- Department of Stomatology, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| | - Chen Wang
- Department of Stomatology, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| | - Ying Jiang
- Clinical Laboratory, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| | - Wei Song
- Department of Information, Medical Supplies Center of PLA General Hospital, Beijing, PR China.
| | - Ji-Feng Yu
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, PR China.
| | - Li Liang
- Department of Stomatology, Eighth Medical Center of Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
4
|
Song R, Wan Z, Yuan X, Wang N, Gao Y, Zhang L, Ren H, Jin Y, Liu X, Sang J, Yuan Z, Zhao Y. Macrophage membrane functionalized composite microspheres promote bone regeneration in periodontitis via manipulating inflammation reversing-osteogenesis coupling. Mater Today Bio 2025; 32:101789. [PMID: 40331151 PMCID: PMC12054120 DOI: 10.1016/j.mtbio.2025.101789] [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: 01/20/2025] [Revised: 04/02/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Periodontitis is characterized by inflammation and alveolar bone loss, primarily caused by immune cells activated by oral bacteria, leading to an imbalance between osteogenesis and bone resorption. Traditional treatments have limited efficacy, which has led to the exploration of regulating the immune microenvironment and utilizing tissue engineering methods as new research directions. Our study demonstrates that macrophage membranes, activated by LPS and IFN-γ, can effectively neutralize inflammatory factors. By coating the poly-L-lysine (PLL) modified poly (lactic-co-glycolic acid) (PLGA)/β-TCP microspheres with such macrophage membrane vesicles, the MM@PPT microspheres regulate intercellular responses by inhibiting macrophage M1 polarization and osteoclast differentiation, promoting M2 polarization, and enhancing osteogenic differentiation of bone marrow stromal cells (BMSCs) even in an inflammatory environment. By injecting the MM@PPT into sites of periodontitis induced bone resorption, it is found that they can effectively promote bone regeneration by modulating the immune-regeneration microenvironment. This work not only highlights the potential of MM@PPT microspheres in promoting alveolar bone regeneration but also provides insights into how these microspheres modulate cell behavior and interactions. The findings of this study offer novel therapeutic strategies for promoting alveolar bone repair in periodontitis.
Collapse
Affiliation(s)
- Rui Song
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced Technology, College of Engineering, Peking University, Beijing, 100871, PR China
| | - Xiaojing Yuan
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Nan Wang
- Department of Stomatology, Peking University Third Hospital, Beijing, 100191, PR China
| | - Yike Gao
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Linxue Zhang
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Huihui Ren
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Yu Jin
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Xiya Liu
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Jingyi Sang
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| | - Zuoying Yuan
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, PR China
| | - Yuming Zhao
- Department of Pediatrics, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing Key Laboratory of Digital Stomatology, Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health, NMPA Key Laboratory for Dental Materials, Beijing, 100081, PR China
| |
Collapse
|
5
|
Huangfu Y, Zhao Z, Liu X, Wang J, Zhang Y, Lan T, Wang Y, Wu C, Zhang J, Huang P, Zhang C, Dong A, Feng Z, Kong D, Wang W. An Off-the-Shelf Artificial Blood Clot Hydrogel Neutralizing Multiple Proinflammatory Mediators for Pro-Regenerative Periodontitis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04106. [PMID: 40433947 DOI: 10.1002/advs.202504106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 05/12/2025] [Indexed: 05/29/2025]
Abstract
Periodontitis is a destructive and chronic inflammatory disease initiated and sustained by multiple proinflammatory mediators. Current therapies mainly deal with bacteria elimination, but directly addressing the over-accumulated multiple inflammatory mediators in the periodontal microenvironment still remains a substantial challenge for regenerative periodontitis treatment. Herein, inspired by blood coagulation, an off-the-shelf artificial blood clot hydrogel encapsulated with platelet-rich plasma (PRP) is reported to mitigate the deteriorative inflammatory environment in periodontitis. The hydrogel (CCS-RSF@PRP) with a hierarchical fibers-interwoven network structure, in which the activated platelets are enriched, is structurally similar to the native blood clot. Functionally, in addition to the function of enriching and releasing growth factors, CCS-RSF@PRP can remarkably scavenge reactive oxygen species (ROS), neutralize endotoxin lipopolysaccharide (LPS), and proinflammatory cytokines (TNF-α, IFN-γ and IL-1β), inhibit M1 macrophage polarization and induce M2 macrophage polarization, thus blocking the chronic inflammatory feedback loop in the periodontitis. In rat periodontitis model, CCS-RSF@PRP hydrogel significantly expedits the repair of periodontium by normalizing the periodontal immune-environment. The work highlights the importance of local immunomodulation in the treatment of periodontitis, and the engineered PRP-derived hydrogel can mimic and expand the structure and function of native blood clot, holding great promise in treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Yini Huangfu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Zhezhe Zhao
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Xiang Liu
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Jingrong Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Yufeng Zhang
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Tingting Lan
- Research Institute of Transplant Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, 300190, P. R. China
| | - Yonglan Wang
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Chenxuan Wu
- Department of Periodontology, School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Ju Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Pingsheng Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Chuangnian Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Anjie Dong
- Department of Polymer Science and Engineering, Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Zujian Feng
- State Key Laboratory of Advanced Medical Materials and Devices, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300192, P. R. China
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, P. R. China
| | - Deling Kong
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, P. R. China
| | - Weiwei Wang
- College of Life Sciences, Key Laboratory of Bioactive Materials (Ministry of Education), State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
6
|
Huang X, Shen H, Li Z, Fu T, Wei X, Qiu S. Isochlorogenic Acid A Attenuated Periodontitis by Inhibiting Inflammation via ACE/PGK1/STAT1 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12702-12722. [PMID: 40368646 DOI: 10.1021/acs.jafc.4c13299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
The purpose of the present study was to evaluate the effect of isochlorogenic acid A (IAA) on periodontitis and explore its potential mechanism. A ligature/LPS procedure was employed to induce periodontitis in rats. LPS-induced HPDLF was used as the in vitro model. Bioinformatic analyses indicated that angiotensin-converting enzyme (ACE) might be the target of IAA in periodontitis. PGK1/STAT1 might be the related molecules of ACE. Treatment with IAA inhibited inflammatory cytokines, inflammatory molecule transcriptions in gingival tissues and HPDLFs. MicroCT, H&E, TRAP and Runx2 staining showed that IAA relieved periodontitis. Immunofluorescence observation and WB revealed that IAA inhibited ACE/PGK1/STAT1 in dental papilla, parodontium, gingival tissues and HPDLFs. IAA suppressed glycolysis, mitochondrial fission, oxidative stress and promoted oxidative phosphorylation, mitochondrial fusion. The application of the ACE-overexpression plasmid, PGK1 SiRNA, the ACE inhibitor captopril, the PGK1 inhibitor NG52 suggested that ACE and PGK1 were involved in IAA-mediated anti-inflammatory response. Molecular docking, molecular dynamics, DARTS and CETSA indicated that IAA might combine with ACE. It was also found that MYC governed ACE transcription. The Y195 residue of PGK1 was conservative and critical for the combination between PGK1 and ACE. ACE and PGK1 were also upregulated in gingival tissues of periodontitis patients. In conclusion, IAA ameliorated periodontitis by inhibiting inflammation via ACE/PGK1/STAT1 signaling.
Collapse
Affiliation(s)
- Xin Huang
- Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Hong Shen
- Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Zirui Li
- Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Tingting Fu
- Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Xin Wei
- Department of Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| | - Sainan Qiu
- Department of Pediatric and Preventive Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
7
|
Bostanci N, Manoil D, Van Holm W, Belibasakis GN, Teughels W. Microbial Markers for Diagnosis and Risk Assessment for Periodontal Diseases: A Systematic Literature Search and Narrative Synthesis. J Clin Periodontol 2025. [PMID: 40414257 DOI: 10.1111/jcpe.14183] [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: 12/21/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/27/2025]
Abstract
AIM To examine the accuracy of microbiological biomarkers in diagnosing periodontal diseases, specifically addressing three focus questions: (FQ1) distinguishing health from disease; (FQ2) predicting disease progression; and (FQ3) assessing treatment outcomes. MATERIALS AND METHODS A PRISMA-guided search in MEDLINE, EMBAS and WEB OF SCIENCE included cross-sectional and longitudinal studies (e.g., randomised controlled trials, cohort studies) with ≥ 20 participants per group. Eligible studies involved individuals diagnosed with periodontal health, gingivitis or periodontitis, based on well-defined clinical criteria, and utilised microbiological analyses of oral fluids and/or dental plaque. Diagnostic accuracy had to be evaluated using sensitivity, specificity or area under the receiver operating characteristic (ROC) curve (AUC), or alternatively, data for their computation had to be provided. RESULTS Thirty-one studies were included, mostly cross-sectional or case-control, with significant variability in sampling sites, microbial analyses and diagnostic definitions, complicating direct comparisons. Frequently investigated biomarkers included Aggregatibacter actinomycetemcomitans (JP2 genotype), Porphyromonas gingivalis, Tannerella forsythia and Treponema denticola. The highest diagnostic accuracy (AUC > 0.95) was achieved through composite microbiome-based metrics such as the subgingival microbial dysbiosis index. However, methodological heterogeneity and inconsistent criteria limited reliability. CONCLUSION Although microbiological biomarkers hold promise for periodontal disease diagnostics and monitoring, current evidence is insufficient for clinical implementation. Future research should standardise methodologies, sampling protocols and diagnostic criteria to ensure robust validation and facilitate integration into precision dentistry.
Collapse
Affiliation(s)
- Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel Manoil
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Cariology and Endodontics, Faculty of Medicine, University Clinics of Dental Medicine, University of Geneva, Geneva, Switzerland
| | - Wannes Van Holm
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Georgios N Belibasakis
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wim Teughels
- Department of Oral Health Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
- University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Lowpradit P, Janmanee R, Tansriratanawong K. Performance Validation of Fabricated Nanomaterial-Based Biosensor for Matrix Metalloproteinase-8 Protein Detection. Eur J Dent 2025. [PMID: 40398657 DOI: 10.1055/s-0045-1809182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2025] Open
Abstract
Matrix metalloproteinase-8 (MMP-8) is a crucial collagenase enzyme that primarily degrades type I collagen and extracellular glycoproteins, playing a significant role in the pathological processes of periodontal disease. It can serve as a biomarker for early detection and screening of the disease through advanced biosensor technology. The aim of this study was to fabricate and validate the performance of a nanomaterial-based biosensor for detecting MMP-8 protein.The screen-printed gold electrode was modified with a thin film of 11-mercaptoundecanoic acid using the self-assembled monolayer technique. A biosensor was then created with N-hydroxysuccinimide and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide to form ester bonds, followed by the immobilization of the antibody MMP-8 and blocking of nonspecific binding. The performance characteristics of a biosensor for detecting MMP-8 concentrations, ranging from 1 to 50 ng/mL, were evaluated using electrochemical techniques with data analysis performed using the NOVA software. Enzyme-linked immunosorbent assay (ELISA) was used as a control.The results were expressed as mean values ± standard deviation. The coefficient of determination (R 2) was calculated based on the obtained calibration curve.Electrochemical measurements revealed that the peak current after modifying the thin film on the electrode was lower than the bare electrode. Characterization of biosensors showed an increase in response compared to the previous step. Differential pulse voltammetry measurements indicated that the peak current for MMP-8 concentrations ranging from 1 to 50 ng/mL increased proportionally with concentration. The biosensor demonstrated high sensitivity, with a correlation coefficient of R 2 = 0.953 when compared to ELISA (R 2 = 1).A biosensor utilizing nanomaterials has been successfully fabricated for the detection of the MMP-8 protein with high sensitivity. Subsequent research should prioritize the evaluation of its performance in clinical patients alongside an assessment of its specificity and stability. The objective was to advance this biosensor as a reliable diagnostic tool for the screening of periodontitis.
Collapse
Affiliation(s)
- Pimsunee Lowpradit
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Rapiphun Janmanee
- Department of Chemistry, Faculty of Science and Technology, Pibulsongkram Rajabhat University, Phitsanulok, Thailand
| | - Kallapat Tansriratanawong
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
Yuan Z, Ostrowska‐Podhorodecka Z, Cox T, Norouzi M, Wang Y, Robaszkiewicz K, Siatkowska M, Xia K, Ali A, Abovsky M, Jurisica I, Smith P, McCulloch CA. Annexin A2 Contributes to Release of Extracellular Vimentin in Response to Inflammation. FASEB J 2025; 39:e70621. [PMID: 40346842 PMCID: PMC12065020 DOI: 10.1096/fj.202500793r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/21/2025] [Accepted: 04/29/2025] [Indexed: 05/12/2025]
Abstract
Vimentin, an abundant intracellular cytoskeletal protein, is secreted into the extracellular space, where it can amplify tissue destruction in inflammatory diseases. The mechanisms by which inflammation promotes the release of extracellular vimentin (ECV) are not defined. In human subjects, we found > twofold higher levels of ECV in gingival crevicular fluid from periodontitis sites with inflammation compared with healthy sites. In cultures of human gingival fibroblasts (hGFs) treated with 1% serum or IL-1β (10 ng/mL) to model tissue injury or inflammation, respectively, we found that 1% serum increased ECV release > 11-fold while IL-1β further enhanced release 17-fold. Mass spectrometry of vimentin immunoprecipitates identified Annexin A2 (AnxA2), a Ca2+-dependent phospholipid-binding protein, as a potential binding protein of ECV, which was confirmed by immunoprecipitation of cultured hGFs and immunostaining of inflamed human gingiva. IL-1β treatment enhanced the abundance of AnxA2 and vimentin in membrane fractions prepared by sucrose gradients of hGF lysates. IL-1β increased colocalization of ECV and AnxA2 at the outer aspect of the plasma membrane of intact hGFs. Knockdown of AnxA2 with siRNA or inhibition of the unconventional secretory pathway reduced ECV release from hGFs. These findings indicate that the production of ECV by hGFs in response to inflammation is mediated by an AnxA2-dependent, unconventional secretory pathway that may play a role in amplification of the inflammatory response.
Collapse
Affiliation(s)
- Zhiyao Yuan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Research Institute of StomatologyNanjing UniversityNanjingChina
| | | | - T. Cox
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| | - M. Norouzi
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| | - Y. Wang
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| | - K. Robaszkiewicz
- Department of Biochemistry and Cell Biology, Faculty of Natural SciencesKazimierz Wielki University in BydgoszczBydgoszczPoland
| | - M. Siatkowska
- Department of Biochemistry and Cell Biology, Faculty of Natural SciencesKazimierz Wielki University in BydgoszczBydgoszczPoland
- Laboratory of Molecular and Nanostructural BiophysicsBionanoparkLodzPoland
| | - K. Xia
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| | - A. Ali
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| | - M. Abovsky
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research InstituteUniversity Health NetworkTorontoOntarioCanada
| | - I. Jurisica
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
- Osteoarthritis Research Program, Division of Orthopedic Surgery, Schroeder Arthritis Institute and Data Science Discovery Centre for Chronic Diseases, Krembil Research InstituteUniversity Health NetworkTorontoOntarioCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
- Department of Computer ScienceUniversity of TorontoTorontoOntarioCanada
- Institute of NeuroimmunologySlovak Academy of SciencesBratislavaSlovakia
| | - P. Smith
- Faculty of Medicine, School of DentistryPontificia Universidad Católica de ChileSantiagoChile
| | - C. A. McCulloch
- Faculty of DentistryUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
10
|
Chiu CJ, Chiu E, Chang ML. Interaction between Infection of Porphyromonas gingivalis, A Keystone Microbe of Oral Microbiome, and Serum Levels of Lutein/Zeaxanthin Is Associated with Risk for Age-related Macular Degeneration. RESEARCH SQUARE 2025:rs.3.rs-6188207. [PMID: 40386394 PMCID: PMC12083656 DOI: 10.21203/rs.3.rs-6188207/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Porphyromonas gingivalis (P. gingivalis) functions as a catalyst bacterium in the development of periodontitis, and the serum antibody level against P. gingivalis is considered a surrogate marker for the activity level of periodontopathic microbiome. The chronic systemic inflammation induced by P. gingivalis elevates the risk of various systemic and neurodegenerative disorders, including atherosclerosis, diabetes, and Alzheimer's disease. Although the connection between human microbiome and age-related macular degeneration (AMD) remains relatively unexplored, it is noteworthy that AMD shares risk factors and etiological mechanisms with diseases related to P. gingivalis. To investigate the potential association between periodontopathic microbiome and AMD occurrence, we conducted a candidate microbe approach case-control study. Our hypothesis was tested by examining the correlation between serum P. gingivalis immunoglobulin G (IgG) levels and AMD. Comparing the lowest IgG category (≤ 57 enzyme-linked immunosorbent assay units (EU)) with higher categories revealed escalating risks: the second higher category (58-65 EU) conferred almost a 30% increased risk (odds ratio (OR) = 1.28, 95% confidence interval (CI): 1.17 to 1.4), the third higher category (66-119 EU) conferred nearly a 60% increase (OR = 1.58, 95% CI: 1.46 to 1.72), and the highest category (> 119 EU) conveyed over a two-fold risk (OR = 2.04, 95% CI: 1.62 to 2.58) of early AMD. Aligning with the notion that the microbiome composition is significantly shaped by the host's diet, our analysis indicates that sustaining elevated serum levels of lutein/zeaxanthin (≥ 0.35 μmol/L or ≥ 20 μg/dL) might potentially mitigate the P. gingivalis-related AMD risk by as much as 35% (P for interaction < 0.0001). Although the precise mechanism requires additional exploration, these findings suggest a connection between nutrition and oral microbiome, emphasizing their collective role in maintaining eye health. SIGNIFICANCE STATEMENT While our oral microbiome may impact eye health, nutritional factors could play a modulatory role in mitigating the associated risk.
Collapse
|
11
|
Coats SR, Su TH, Luderman Miller Z, King AJ, Ortiz J, Reddy A, Alaei SR, Jain S. Porphyromonas gingivalis outer membrane vesicles divert host innate immunity and promote inflammation via C4' monophosphorylated lipid A. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:1008-1021. [PMID: 40131356 PMCID: PMC12123218 DOI: 10.1093/jimmun/vkae050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/05/2024] [Indexed: 03/27/2025]
Abstract
Porphyromonas gingivalis (Pg) is a prevalent pathogen that promotes human periodontal disease (PD) and exacerbates systemic comorbidities such as atherosclerosis, rheumatoid arthritis, and Alzheimer's disease. Pg produces nonphosphorylated tetra-acylated lipid A (NPLA) in its outer membrane (OM) that evades host Toll-like receptor 4 (TLR4), inflammasome pathways, and cationic peptides, enhancing bacterial survival. Here, we show that Pg also releases outer membrane vesicles (OMVs) that engage and divert host cell TLR4, inflammasome, and LL-37 responses away from the microbe. We determined that Pg OMVs are enriched for C4' monophosphoryl lipid A (C4'-MPLA), an established agonist for TLR4-TRIF-IFNβ and inflammasome-IL-1β responses. Comparisons of Pg 381 and Pg 33277 stationary phase cultures revealed higher OMV production by Pg 381, which correlates with its higher proinflammatory pathogenicity. The cationic peptide, polymyxin B (PMB), which selectively binds lipid A C4'-phosphate, reduces OMV-stimulated HEK cell TLR4 activation and THP-1 cell IL-1β production, confirming the proinflammatory role for OMV-C4'-MPLA. Similar to PMB, the host defense peptide, LL-37, inhibits OMV-C4'-MPLA-dependent HEK cell TLR4 activation. PMB and LL-37 also blocked OMV-C4'-MPLA-driven TLR4 activation in human umbilical vein endothelial cells. Finally, wild-type Pg-containing OM-NPLA is highly resistant to LL-37 antimicrobial activity, whereas the ΔlpxF mutant bacterium, retaining OM-C4'-MPLA, is killed by the peptide. In summary, Pg escapes host TLR4 signaling, inflammasome activation, and LL-37 interaction by retaining immunoevasive OM-NPLA. Moreover, Pg dispenses proinflammatory OMV-C4'-MPLA, which engages and redirects those host defenses. We suggest that OMV-C4'-MPLA triggers elevated IFNβ and IL-1β cytokines, which typify PD comorbidities, and drive PD-related alveolar bone loss.
Collapse
Affiliation(s)
- Stephen R Coats
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Thet Hnin Su
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Zoe Luderman Miller
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| | - Alisa J King
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Joshua Ortiz
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Angel Reddy
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sarah R Alaei
- Sciences and Mathematics Division, School of Interdisciplinary Arts and Sciences, University of Washington Tacoma, Tacoma, WA, United States
| | - Sumita Jain
- Department of Periodontics, University of Washington School of Dentistry, Seattle, WA, United States
| |
Collapse
|
12
|
Samal A, Kripfgans OD, Wang IC, Betancourt ABR, Webber L, Quesada C, Mazzocco J, Wishart JD, Chan HL. High-Frequency Ultrasound Characterization of Periodontal Soft Tissues Pre- and Post-Bacterial Inoculation. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:860-869. [PMID: 39947944 PMCID: PMC11996069 DOI: 10.1016/j.ultrasmedbio.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVE Current diagnostic methods of inflammatory periodontal diseases, e.g., visual evaluation, periodontal probing, and radiographs, are either subjective or insensitive. Intra-oral high-frequency ultrasound was investigated to quantify inflammation by detecting tissue dimensional and perfusion changes. METHODS A cohort of 15-month-old mini-pigs, 4 female/male each, was analyzed. Pre-molars (PM) 3 and 4, as well as first molars (M1), were scanned. In bi-weekly time intervals all 4 quadrants were randomly enrolled and bacterial injection followed each quadrant scan in a weekly fashion. Soft tissue dimensions were obtained from B-mode images and statistically analyzed to identify correlations to inoculation time, i.e., response to bacterial loading, tooth type and sex, using analysis of variance and regression analysis. Color flow velocity and power-weighted color pixel density was obtained and statistically analyzed analogous to soft tissue. RESULTS Soft tissue thickness increased significantly post-inoculation at 1 and 2 mm below the free gingival margin for both genders and all observed teeth. The significance lasted for weeks 2, 4 and 6, except for female M1s (4 weeks). Color flow velocity was significantly higher compared with baseline for 6 weeks, except for male PM4 (2 weeks). Color flow power did not show significance for PM3 and 4, only in M1 (except male week 4). Significance also extended to tooth type and sex. CONCLUSION Periodontal tissue dimension and color flow velocity increased in correlation to bacterial inoculation. Further studies are needed to obtain an understanding of the underlying biology observed here. Eruption of dentition may have been a confounding factor for inflammation interpretation.
Collapse
Affiliation(s)
- Ankita Samal
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Periodontics, University of Iowa College of Dentistry & Dental Clinics, Iowa City, IA, USA; Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA
| | - Oliver D Kripfgans
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| | - I-Ching Wang
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Periodontics, University of Iowa College of Dentistry & Dental Clinics, Iowa City, IA, USA; Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA
| | - Amanda B Rodriguez Betancourt
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA; Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA; Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - Liana Webber
- Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA
| | - Carole Quesada
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - John Mazzocco
- Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA
| | - James D Wishart
- Department of Radiology, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Hsun-Liang Chan
- Department of Periodontology and Oral Medicine, School of Dentistry, Ann Arbor, MI, USA; Division of Periodontology, College of Dentistry, Ohio State University, Columbus, OH, USA
| |
Collapse
|
13
|
Ye Z, Gao L, Guo Z, Wang Q. Oral and intestinal flora translocation and tumor development. J Cancer Res Ther 2025; 21:323-333. [PMID: 40317136 DOI: 10.4103/jcrt.jcrt_50_24] [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: 01/07/2024] [Accepted: 02/07/2025] [Indexed: 05/07/2025]
Abstract
ABSTRACT Cancer metastasis is the leading cause of death in patients. In recent years, there has been a growing recognition of the role of tumor-associated microflora in tumor metastasis. The connection between oral and gut microflora and the tumor microenvironment has also been extensively studied. The migration of oral and gut microflora is closely associated with tumor development. Although there is awareness regarding the significant impact of microbial communities on human health, the focus on their relationship with host organisms, particularly those related to tumor-associated microflora, remains inadequate. As an integral part of the body, the host microflora is crucial for regulating the cancer risk and preventing tumor recurrence. The oral-gut axis plays an indispensable role in human immunity, and many types of cancers, such as colorectal, pancreatic, and breast, are significantly influenced by their internal microbial communities. However, further exploration into the mechanisms underlying the role of the intratumoral microflora in cancer is necessary to achieve a comprehensive understanding. We have summarized and analyzed related articles in PubMed. This article reviews the impact of the oral-gut axis on the human immune system, explores the relationship between the translocation of the oral and intestinal flora and the tumor microenvironment, analyzes the specific mechanisms involved in the translocation of the oral and intestinal microflora during the evolution and progression of tumors, and elaborates on the correlations between the occurrence and development of tumors and the changes in the microflora. Finally, a summary of these abovementioned points is provided.
Collapse
Affiliation(s)
- Zhiyuan Ye
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Linglin Gao
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Guo
- Department of Hematology, The 6 Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Wang Y, Li Y, Cai Y, Yang X, Li H, Wang Q, Huang D, Liu L, Fan Z, Yuan Q, Wang Y. Dimethyl Citraconate Alleviates Periodontitis via Activating the NRF2 Cascade. J Dent Res 2025:220345251319249. [PMID: 40289519 DOI: 10.1177/00220345251319249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a pivotal transcription factor that regulates redox signaling, playing a protective role in inflammation. Citraconate is verified as the strongest NRF2 agonist among its isomers. Dimethyl citraconate (DMC), an esterified derivative of citraconate, holds the potential for activating NRF2 and relieving inflammation. Here, we show that DMC is a strong NRF2-activating compound, stabilizing the intracellular NRF2 level and its nuclear translocation. DMC increases the expression levels of NRF2 downstream genes, thereby restricting the accumulation of reactive oxygen species and performing anti-inflammatory functions. The local administration of DMC effectively alleviates periodontal destruction in a ligation-induced periodontitis mouse model, elevating the NRF2 levels and downstream antioxidant enzymes. Moreover, the protective effect of DMC against periodontitis is absent in Nfe2l2-/- mice. Mechanically, DMC prolongs the half-life of NRF2 and facilitates its dissociation from KEAP1 (Kelch-like ECH-associated protein 1), which suggests that DMC interrupts the crosstalk between KEAP1 and NRF2. Collectively, our findings illustrate the role of DMC in activating NRF2 and ameliorating periodontal inflammation, suggesting its therapeutic potential for inflammation-related diseases.
Collapse
Affiliation(s)
- Y Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Cai
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - D Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Liu
- 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
| | - Z Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
- Research Unit of Tooth Development and Regeneration, Chinese Academy of Medical Sciences, Beijing, China
| | - Q Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Beklen A, Yavuz MB, Uckan D. Interleukin-37 reduces lipopolysaccharide induced matrix metalloproteinase-9 in gingival epithelial cells. BMC Oral Health 2025; 25:637. [PMID: 40281482 PMCID: PMC12023668 DOI: 10.1186/s12903-025-06016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND In periodontal diseases, the recognition of pathogen-associated molecular patterns (PAMPs) triggers signaling cascades that lead to the release of matrix metalloproteinases (MMPs). Interleukin-37 (IL-37) is recognized as a key suppressor of the immune response. This study aimed to detect the expression and distribution of IL-37 in gingival tissues and analyze its suppressor role in MMP-9 in response to lipopolysaccharide (LPS)-stimulated gingival epithelial cells. METHODS Immunohistochemistry localized IL-37 in gingival tissues from periodontitis patients and healthy controls (N = 10). The induction of IL-37 expression by LPS was analyzed using the conditioned medium of gingival epithelial cells through enzyme-linked immunosorbent assay (ELISA). To determine the relevant MMP-9 levels in epithelial cells following exposure to LPS alone or in combination with IL-37, both quantitative PCR (qPCR) and enzyme-linked immunosorbent assay (ELISA) were performed. RESULTS Cultured epithelial cells secreted significantly higher levels of IL-37 when stimulated with LPS compared to unstimulated controls. Both ELISA and qPCR showed that LPS stimulation significantly increased MMP-9 levels. However, co-culture with IL-37 markedly reduced LPS-induced MMP-9 expression at both the protein and mRNA levels. Furthermore, immunohistochemistry revealed increased IL-37 expression in periodontitis tissues, both in epithelial cells and connective tissue. CONCLUSIONS Gingival epithelial cells may contribute to tissue responses in periodontitis through the secretion of MMP-9 in response to PAMPs. Furthermore, IL-37 appears to have a potential role in modulating and reducing this response, as observed in the decreased MMP-9 expression following IL-37 co-stimulation.
Collapse
Affiliation(s)
- Arzu Beklen
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), University of Helsinki, Helsinki, Finland.
- Department of Periodontology, Faculty of Dentistry, Eskisehir Osmangazi University, Eskisehir, Turkey.
| | - Muhammet Burak Yavuz
- Department of Periodontology, Faculty of Dentistry, Eskisehir Osmangazi University, Eskisehir, Turkey
| | - Deniz Uckan
- Bogazici University, Medico-Social Dental Clinic, Istanbul, Turkey
| |
Collapse
|
16
|
Chen X, Lin WY, Zhang FW, Guo LQ, Ge H, Ge DZ, Tan JJ, Liu BC, Wang RR, Zhang L. Investigation of oral microbiome composition in elderly Chinese patients with hypertension: a cross-sectional study. J Oral Microbiol 2025; 17:2489603. [PMID: 40270620 PMCID: PMC12016255 DOI: 10.1080/20002297.2025.2489603] [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: 10/15/2024] [Revised: 02/10/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hypertension is a prevalent metabolic disorder in the elderly, with its pathogenesis linked to gut microbiota dysbiosis. Recent studies suggested that oral microbiota may also play a role in hypertension development, yet its relationship with hypertension in the elderly remains underexplored. Objective This cross-sectional study aimed to examine the structure of the oral microbiota and its association with hypertension in elderly patients, providing insights into hypertension prevention and treatment. Methods A total of 206 subjects (60-89 years) were categorized into normal (CON) and hypertensive (HTN) groups, based on the Chinese Hypertension Guidelines. Saliva samples were analyzed using 16S rRNA gene sequencing. Results Oral microbiota composition was significantly influenced by blood pressure. At the phylum level, Synergistetes and Spirochaetes were more significantly abundant in the HTN group, while at the genus level Treponema and Leptothrix was higher, Actinomyces and Capnocytophaga were lower in HTN. Random Forest analysis identified 15 key microbiota as strong discriminators of HTN (AUC 0.74). Blood pressure was negatively correlated with Actinomycetes and positively correlated with Leptothrix. PICRUST2 analysis revealed elevated chlorinated compound degradation in HTN patients. Conclusions This study identified distinct oral microbiota in elderly hypertensive patients, highlighting the role of the oral microbiome in hypertension pathogenesis.
Collapse
Affiliation(s)
- Xin Chen
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Yong Lin
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feng-Wei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Qiang Guo
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Han Ge
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ding-Zuo Ge
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Juan-Juan Tan
- Institute of Integrative Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
- Institute of Integrative Medicine, Shaanxi Key Laboratory of Integrated Traditional and Western Medicine for Prevention and Treatment of Cardiovascular Diseases, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Bao-Cheng Liu
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rui-Rui Wang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhang
- Shanghai Innovation Center of Traditional Chinese Medicine Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
17
|
Matsuoka M, Soria SA, Pires JR, Sant'Ana ACP, Freire M. Natural and induced immune responses in oral cavity and saliva. BMC Immunol 2025; 26:34. [PMID: 40251519 PMCID: PMC12007159 DOI: 10.1186/s12865-025-00713-8] [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: 11/22/2024] [Accepted: 04/07/2025] [Indexed: 04/20/2025] Open
Abstract
This review comprehensively explores the intricate immune responses within the oral cavity, emphasizing the pivotal role of saliva in maintaining both oral and systemic health. Saliva, a complex biofluid, functions as a dynamic barrier against pathogens, housing diverse cellular components including epithelial cells, neutrophils, monocytes, dendritic cells, and lymphocytes, which collectively contribute to robust innate and adaptive immune responses. It acts as a physical and immunological barrier, providing the first line of defense against pathogens. The multifaceted protective mechanisms of salivary proteins, cytokines, and immunoglobulins, particularly secretory IgA (SIgA), are elucidated. We explore the natural and induced immune responses in saliva, focusing on its cellular and molecular composition. In addition to saliva, we highlight the significance of a serum-like fluid, the gingival crevicular fluid (GCF), in periodontal health and disease, and its potential as a diagnostic tool. Additionally, the review delves into the impact of diseases such as periodontitis, oral cancer, type 2 diabetes, and lupus on salivary immune responses, highlighting the potential of saliva as a non-invasive diagnostic tool for both oral and systemic conditions. We describe how oral tissue and the biofluid responds to diseases, including considerations to periodontal tissue health and in disease periodontitis. By examining the interplay between oral and systemic health through the oral-systemic axis, this review underscores the significance of salivary immune mechanisms in overall well-being and disease pathogenesis, emphasizing the importance of salivary mechanisms across the body.
Collapse
Affiliation(s)
- Michele Matsuoka
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Salim Abraham Soria
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA
| | - Julien Rodrigues Pires
- Department of Periodontology, Bauru School of Dentistry, University of São Paulo, Bauru, 17012-901, Brazil
| | | | - Marcelo Freire
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA, 92037, USA.
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
18
|
Papantonaki AI, Georgakopoulou E, Barda C, Loumou P, Sfiniadakis I, Anastassopoulou J, Vitsos A, Rallis MC. Exploring the Non-Toxic Therapeutic Potential of Dioscorea communis in Combating Oral Pathogenic Bacteria and Their Effects on Hard and Soft Oral Tissues. Biomedicines 2025; 13:983. [PMID: 40299662 PMCID: PMC12024764 DOI: 10.3390/biomedicines13040983] [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: 02/23/2025] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 05/01/2025] Open
Abstract
Background/Objectives: Gingivitis and dental caries are oral diseases resulting from bacterial accumulation in dental plaque, leading to inflammation, tissue destruction and the demineralization of tooth structures. Dioscorea communis, due to its anti-inflammatory and antimicrobial properties, could be a new treatment candidate. Methods: This study evaluated the preventive and therapeutic effect of a D. communis berry juice paste, formulated at 3% and 7% concentrations, on gingivitis and dental caries, in 55 male SKH-hr2 hairless mice. Gingivitis and dental caries were induced by ligation of the upper left incisor and the paste was applied topically three times daily, five days a week. Treatment efficacy was assessed through clinical examinations, photo-documentation, histopathological analysis and FT-IR spectroscopy. Results/Conclusions: Preventive administration of D. communis 7% significantly delayed disease onset, while therapeutic effects on established conditions were limited. Both concentrations were non-toxic to gingival tissues and dental structures.
Collapse
Affiliation(s)
- Anastasia-Ioanna Papantonaki
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | - Eleni Georgakopoulou
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | - Christina Barda
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | - Panagiota Loumou
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | | | - Jane Anastassopoulou
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | - Andreas Vitsos
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| | - Michail Christou Rallis
- Section of Pharmaceutical Technology, Department of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15784 Athens, Greece; (A.-I.P.); (E.G.); (C.B.); (P.L.); (J.A.); (A.V.)
| |
Collapse
|
19
|
Lopes MES, Marcantonio CC, Salmon CR, Mofatto LS, Nociti Junior FH, Eick S, Deschner J, Cirelli JA, Nogueira AVB. Effects of periodontal disease on the proteomic profile of the periodontal ligament. J Proteomics 2025; 314:105384. [PMID: 39800186 DOI: 10.1016/j.jprot.2025.105384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/20/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Periodontal disease affects over 1 billion people globally. This study investigated how periodontitis affects the protein profile of the periodontal ligament (PDL) in rats. Eight Holtzman rats were divided into control and experimental periodontitis groups. The PDL was isolated using laser capture microdissection and protein extracts were analyzed by mass spectrometry. Data analysis utilized specialized software, and Gene Ontology enrichment analysis identified significant protein functions. The data are available via ProteomeXchange with identifier PXD055817. Proteins such as SerpinB1, C5, and Lgals3 were validated through immunohistochemistry, and their gene expression was examined in an in vitro human PDL cell line. This study identified 1326 proteins, with 156 unique to the control group, 294 unique to the periodontitis group, and 876 common to both groups. Enrichment analysis revealed that proteins associated with the regulation of enzyme activity and RNA binding were significantly represented in the periodontitis group. There were increased levels of SerpinB1, C5, and Lgals3 in the periodontitis group based on proteomic and immunohistochemical analyses. Furthermore, these targets showed increased gene expression in stimulated human PDL cells. This study provides insights into the periodontitis-related alterations in the protein composition of the PDL and PDL cells, identifying both novel and previously known disease-associated proteins. SIGNIFICANCE: The periodontal ligament plays a crucial role in oral functions by providing structural support to the tooth. Due to the presence of undifferentiated mesenchymal cells, research into its regenerative capacity is ongoing. Pathological conditions can affect these functions and protein composition. Currently, there is a lack of comprehensive research specifically focusing on evaluating the periodontal ligament in both healthy and diseased states. This pioneering study screened for protein alterations and the mechanisms related to periodontitis. The possibility of using proteomic analysis to evaluate the protein alterations that occur in periodontitis, a disease with a high global incidence, could provide therapeutic targets and new biomarkers for future clinical studies.
Collapse
Affiliation(s)
- Maria Eduarda Scordamaia Lopes
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Camila Chierici Marcantonio
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Cristiane Ribeiro Salmon
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, São Paulo, Brazil; Dental School, Centro Universitário N. Sra do Patrocínio - CEUNSP, Itu, São Paulo, Brazil
| | - Luciana Souto Mofatto
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas - UNICAMP, Campinas, São Paulo, Brazil
| | - Francisco Humberto Nociti Junior
- ADA Forsyth Institute, Cambridge, MA, USA; Dental School, São Leopoldo Mandic, Department of Research, Campinas, São Paulo, Brazil
| | - Sigrun Eick
- Laboratory of Oral Microbiology, Department of Periodontology, University of Bern, Bern, Switzerland
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Joni Augusto Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil.
| | - Andressa Vilas Boas Nogueira
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil; Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
20
|
Hsu CW, Okano T, Niinuma Y, Leewananthawet A, Iida T, Onsoi P, Boonyaleka K, Ashida H, Suzuki T. A complex of NLRP3 with caspase-4 is essential for inflammasome activation by Tannerella forsythia infection. Int Immunol 2025; 37:261-271. [PMID: 39673522 DOI: 10.1093/intimm/dxae071] [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: 09/24/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024] Open
Abstract
Periodontitis, a chronic inflammatory disease of periodontal tissue, is often associated with a group of pathogenic bacteria known as the "red complex", including Tannerella forsythia. Previous papers showed that T. forsythia induces many kinds of inflammatory cytokines including interleukin (IL)-1β regulated by inflammasome activation. However, the physiological function of periodontitis and the mechanism to induce inflammasome activation by T. forsythia infection are poorly understood. In this study, we demonstrate that the Nod-like receptor pyrin domain containing 3 (NLRP3) and caspase-4 are essential for inflammasome activation by T. forsythia infection, playing a crucial role in IL-1β maturation in THP-1 cells. We also showed that the knockout of ASC or Gasdermin D suppresses pyroptotic cell death. Moreover, co-immunoprecipitation assays confirmed the formation of a complex involving caspase-4, NLRP3, and ASC following T. forsythia infection. Additionally, reactive oxygen species production was identified as a key factor in caspase-4-mediated NLRP3 inflammasome activation by T. forsythia infection. These results enhance our understanding of inflammasome activation in response to T. forsythia infection and provide new insights into the pathogenic mechanisms of periodontitis.
Collapse
Affiliation(s)
- Chen-Wei Hsu
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Tokuju Okano
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Yuiko Niinuma
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Anongwee Leewananthawet
- Specialized Dental Center, Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Tamako Iida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Poramed Onsoi
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Kotchakorn Boonyaleka
- Division of Periodontology, Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hiroshi Ashida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| |
Collapse
|
21
|
Anselmi NK, Vanyo ST, Visser MB. Emerging oral Treponema membrane proteins disorder neutrophil phosphoinositide signaling via phosphatidylinositol-4-phosphate 5-kinase. FRONTIERS IN ORAL HEALTH 2025; 6:1568983. [PMID: 40248422 PMCID: PMC12003349 DOI: 10.3389/froh.2025.1568983] [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: 01/31/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Background Periodontitis (PD) is a group of inflammatory pathologies characterized by destruction of the tooth-supporting tissues. During PD, dysbiosis of the oral biofilm disrupts the host immune response and supports growth of pathogenic bacteria including the spirochetes Treponema denticola (Td), T. maltophilum (Tm), and T. lecithinolyticum (Tl). The outer membrane protein of Td, Msp, perturbs the function of neutrophils by modulating phosphoinositide (PIP) signaling. While Tm and Tl have similar outer membrane proteins, MspA and MspTL respectively, little is known of how these proteins affect neutrophil function. Methods This study examines putative mechanisms by which T. maltophilum MspA and T. lecithinolyticum MspTL inhibit neutrophil chemotaxis. Murine bone marrow neutrophils were treated with recombinant MspA or MspTL protein. Protein phosphorylation was assessed via immunoblot, phosphate release by malachite green assay, and PTEN and SHIP phosphatase activity through immunoprecipitation, enzymatic assays, and chemical inhibition. PIP quantification was assessed by immunofluorescence microscopy and Mass ELISAs, while small GTPase activity was measured with G-Protein Activation Assays. Neutrophil F-actin localization was determined through immunofluorescence. Results MspA and MspTL increase phosphate release in neutrophils, but unlike Msp, they do not affect PTEN or SHIP activity, despite modulating cellular levels of multiple PIP species [PI(3,4)P2, PI(4,5)P2, and PIP3]. Overall, MspA and MspTL differentially affected the metabolism of individual PIP species, but both increased PI(4,5)P2 levels in a PIP5K-dependent manner. Downstream effects of disrupted PIP signaling included inhibition of Akt and Rac1 activation and increased cortical F-actin localization. Conclusions Understanding distinct mechanistic relationships between novel Msp proteins and neutrophils provides important insight into how these understudied bacteria promote periodontitis progression.
Collapse
Affiliation(s)
| | | | - Michelle B. Visser
- Department of Oral Biology, The State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
22
|
Wu H, Li Y, Shi L, Liu Y, Shen J. New Advances in Periodontal Functional Materials Based on Antibacterial, Anti-Inflammatory, and Tissue Regeneration Strategies. Adv Healthc Mater 2025; 14:e2403206. [PMID: 39895157 DOI: 10.1002/adhm.202403206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/17/2025] [Indexed: 02/04/2025]
Abstract
With the global population aging, awareness of oral health is rising. Periodontitis, a widespread bacterial infectious disease, is gaining attention. Current novel biomaterials address key clinical issues like bacterial infection, gum inflammation, tooth loosening, and loss, focusing on antibacterial, anti-inflammatory, and tissue regeneration properties. However, strategies that integrate the advantages of these biomaterials to achieve synergistic therapeutic effects by clearing oral biofilms, inhibiting inflammation activation, and restoring periodontal soft and hard tissue functions remain very limited. Recent studies highlight the link between periodontitis and systemic diseases, underscoring the complexity of the periodontal disease. There is an urgent need to find comprehensive treatment plans that address clinical requirements. Whether by integrating new biomaterials to enhance existing periodontal treatments or by developing novel approaches to replace traditional therapies, these efforts will drive advancements in periodontitis treatment. Therefore, this review compares novel biomaterials with traditional treatments. It highlights the design concepts and mechanisms of these functional materials, focusing on their antibacterial, anti-inflammatory, and tissue regeneration properties, and discusses the importance of developing comprehensive treatment strategies. This review aims to provide guidance for emerging periodontitis research and to promote the development of precise and efficient treatment strategies.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| |
Collapse
|
23
|
Xu M, Gan D, Zhang X, He X, Wu RX, Yin Y, Jin R, Li L, Tan Y, Chen F, Li X, Tian B. SLC30A4-AS1 Mediates the Senescence of Periodontal Ligament Stem Cells in Inflammatory Environments via the Alternative Splicing of TP53BP1. Cell Prolif 2025; 58:e13778. [PMID: 39572253 PMCID: PMC11969240 DOI: 10.1111/cpr.13778] [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: 09/03/2024] [Revised: 10/25/2024] [Accepted: 11/09/2024] [Indexed: 04/05/2025] Open
Abstract
Periodontal ligament stem cells (PDLSCs) are key cells that suppress periodontal damage during both the progression and recovery stages of periodontitis. Although substantial evidence has demonstrated that incubation under an inflammatory condition may accelerate senescence of PDLSCs, whether cellular senescence in response to inflammatory incubation contributes to cell dysfunction remain unexplored. In this study, we first observed inflammation-caused PDLSC senescence in periodontitis based on comparisons of matched patients, and this cellular senescence was demonstrated in healthy cells that were subjected to inflammatory conditions. We subsequently designed further experiments to investigate the possible mechanism underlying inflammation-induced PDLSC senescence with a particular focus on the role of long noncoding RNAs (lncRNAs). LncRNA microarray analysis and functional gain/loss studies revealed SLC30A4-AS1 as a regulator of inflammation-mediated PDLSC senescence. By full-length transcriptome sequencing, we found that SLC30A4-AS1 interacted with SRSF3 to affect the alternative splicing (AS) of TP53BP1 and alter the expression of TP53BP1-204. Further functional studies showed that decreased expression of TP53BP1-204 reversed PDLSC senescence, and SLC30A4-AS1 overexpression-induced PDLSC senescence was abolished by TP53BP1-204 knockdown. Our data suggest for the first time that SLC30A4-AS1 plays a key role in regulating PDLSC senescence in inflammatory environments by modulating the AS of TP53BP1.
Collapse
Affiliation(s)
- Mei Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Dian Gan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xi‐Yu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xiao‐Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Xin Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yuan Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Rui Jin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Lin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Yu‐Jie Tan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Fa‐Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| | - Bei‐Min Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of StomatologyThe Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
24
|
Wen J, Li J, Wu Z. Neutrophil extracellular traps induced by diabetes aggravate periodontitis by inhibiting janus kinase/signal transducers and activators of transcription signaling in macrophages. J Dent Sci 2025; 20:869-876. [PMID: 40224106 PMCID: PMC11993069 DOI: 10.1016/j.jds.2024.09.006] [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: 07/13/2024] [Revised: 09/14/2024] [Indexed: 04/15/2025] Open
Abstract
Background/purpose Diabetes, which is a systemic disease, increases susceptibility to destructive periodontal diseases, which are characterized by infectious susceptibility, but the potential mechanisms remain unknown. The aim of this study was to investigate the mechanism of high glucose environment promoting the occurrence and development of local periodontal inflammation. Materials and methods In this study, the effects of neutrophil extracellular traps (NETs) on macrophage polarization and the mechanism were designed to verify whether this course plays a role in periodontal tissue impairment associated with diabetes. Here, we examined the impact of NETs on macrophages in vitro. NETs were isolated from cultures of neutrophils exposed to hyperglycemia. Mouse models of diabetic periodontitis (DP) and macrophage polarization were developed, and the degrees of NET formation in the periodontal tissue of DP mice were assessed. Furthermore, western blotting was performed to analyze the related mechanisms. Results The results revealed that hyperglycemia induced the formation of NETs, and abundant NET formation led to proinflammatory cytokine secretion by macrophages and low expression of JAK-2 and STAT-3 in vitro and in vivo. NETs regulated macrophage polarization through the JAK/STAT pathway. Conclusion These results suggest that NETs target proinflammatory cytokine secretion via the JAK/STAT pathway and may play important roles in DP progression and macrophage polarization, which indicates that therapeutically referring to this regulatory pathway might be a promising method for treating diabetes-associated inflammatory diseases.
Collapse
Affiliation(s)
- Jing Wen
- Changsha Stomatology Hosipital, Changsha, China
| | - Jingru Li
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha, China
| | - Zhenhuan Wu
- Central South University Xiangya Stomatological Hospital, Central South University, Changsha, China
| |
Collapse
|
25
|
Burra Anand D, Ramamurthy J, Kannan B, Jayaseelan VP, Arumugam P. N6-methyladenosine-mediated overexpression of TREM-1 is associated with periodontal disease. Odontology 2025; 113:834-843. [PMID: 39327319 DOI: 10.1007/s10266-024-01009-w] [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: 05/13/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024]
Abstract
Periodontitis, a prevalent inflammatory disease, involves the destruction of tooth-supporting tissues. N6-methyladenosine (m6A) is a type of post-transcriptional modification that significantly influences gene and protein expression. It is involved in the regulation of various diseases, including those with an inflammatory component. This study investigates the potential role of m6A-mediated TREM-1 expression in the development of periodontitis. Clinical features and TREM-1 expression were assessed in periodontitis patients and healthy controls. LPS-stimulated human gingival fibroblasts (HGFs) were used to investigate m6A levels, m6A regulator METTL3, TREM-1, and inflammatory gene expression. In silico functional analysis explored TREM-1 interactions and functionalities. Periodontitis patients showed significantly elevated TREM-1 expression at both mRNA and protein levels. Predicted m6A motifs were present within the TREM-1 transcript. LPS stimulation of HGFs increased m6A content, METTL3, and TREM-1 expression, suggesting a potential link between m6A modification and TREM-1 regulation. Bioinformatic analysis revealed TREM-1 interaction with genes associated with periodontitis and its association with inflammatory pathways. This study suggests a potential role for METTL3-mediated m6A modification in regulating TREM-1 expression in periodontitis. Further investigation is needed to solidify this link and translate findings into clinical applications for improved periodontal health.
Collapse
Affiliation(s)
- Deepika Burra Anand
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Jaiganesh Ramamurthy
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Balachander Kannan
- Molecular Biology Lab, Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, 600077, India
| | - Vijayashree Priyadharsini Jayaseelan
- Clinical Genetics Lab, Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Paramasivam Arumugam
- Molecular Biology Lab, Centre for Cellular and Molecular Research, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, 600077, India.
| |
Collapse
|
26
|
Demirel KJ, Neves Guimaraes A, Demirel I. The Role of Caspase-1 and Caspase-4 in Modulating Gingival Epithelial Cell Responses to Aggregatibacter actinomycetemcomitans Infection. Pathogens 2025; 14:295. [PMID: 40137780 PMCID: PMC11945752 DOI: 10.3390/pathogens14030295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by bacterial infection and immune dysregulation. Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) is a key pathogen linked to disease progression. Caspase-1 and caspase-4 regulate inflammasome activation and cytokine release, yet their roles in gingival epithelial immunity remain unclear. The aim of this study was to elucidate the involvement of caspase-1 and caspase-4 in regulating the immune response to A. actinomycetemcomitans infection in gingival epithelial cells. Human gingival epithelial cells (Ca9-22) and caspase-1- and caspase-4-deficient cells were infected with A. actinomycetemcomitans for 24 h. Inflammatory mediator release was analyzed using Olink proteomics. Bacterial colonization and invasion were assessed using fluorescence-based assays and gentamicin protection assays. Caspase-1- and caspase-4-deficient cells showed significantly altered cytokine and chemokine profiles after infection with A. actinomycetemcomitans, showing reduced IL-17C and IL-18 release. We also found an increased release of TGF-α and LIF from caspase-4-deficient cells, along with elevated levels of the chemokines IL-8, CXCL9, and CXCL10. Additionally, both caspase-1- and caspase-4-deficient cells showed increased bacterial colonization and invasion, particularly in caspase-4-deficient cells. These findings suggest that caspase-1 and caspase-4 play distinct yet essential roles in gingival epithelial immunity, regulating cytokine release, barrier integrity, and defense against A. actinomycetemcomitans colonization.
Collapse
Affiliation(s)
- Kartheyaene Jayaprakash Demirel
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden;
| | - Alessandra Neves Guimaraes
- Department of Odontological Research, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden;
- Department of Periodontology and Implantology, Public Dental Service, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Isak Demirel
- School of Medical Sciences, Örebro University, 701 82 Örebro, Sweden;
| |
Collapse
|
27
|
Pardiñas López S, García-Caro ME, Vallejo JA, Aja-Macaya P, Conde-Pérez K, Nión-Cabeza P, Khouly I, Bou G, Cendal AIR, Díaz-Prado S, Poza M. Anti-inflammatory and antimicrobial efficacy of coconut oil for periodontal pathogens: a triple-blind randomized clinical trial. Clin Oral Investig 2025; 29:182. [PMID: 40085302 PMCID: PMC11909057 DOI: 10.1007/s00784-025-06267-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
OBJECTIVES To evaluate the effect of coconut oil on the oral bacteriome and inflammatory response in patients with periodontitis by integrating next-generation sequencing analyses of pathogenic bacterial shifts and quantification of inflammatory markers, thereby assessing its potential as a natural adjunct to standard nonsurgical periodontal therapy. MATERIALS AND METHODS A triple-blind clinical trial was conducted with 30 participants diagnosed with periodontitis, randomized into 3 groups: (1) coconut oil, (2) chlorhexidine and (3) placebo. Saliva and gingival crevicular fluid (GCF) samples were collected before treatment, one month after treatment, and one month post-non-surgical periodontal therapy. Bacterial DNA was extracted, and the V3-V4 region of the 16 S rRNA gene was PCR-amplified and sequenced using Illumina MiSeq technologies. Inflammatory biomarkers, including Interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), were quantified from GCF samples. RESULTS Coconut oil treatment significantly reduced pathogenic bacterial families such as Spirochaetaceae and Tannerellaceae while promoting beneficial bacteria such as Streptococcaceae. At the genus and species levels, coconut oil reduced pathogens such as Tannerella forsythia and Treponema denticola along with increase in beneficial bacteria such as Streptococcus. The subgingival microbial dysbiosis index improved significantly in both coconut oil and chlorhexidine groups. Furthermore, the coconut oil demonstrated a reduction in IL-6 and TNF-α levels, indicating decreased local inflammation. CONCLUSIONS Coconut oil treatment significantly modulated the oral microbiome and reduced inflammatory markers in patients with periodontitis, suggesting its potential as a natural and effective adjunct in periodontal therapy. CLINICAL RELEVANCE This study highlights coconut oil's potential as a natural adjunct in periodontal therapy, effectively reducing pathogenic bacteria and inflammatory markers (IL-6, TNF-α). It offers a safe alternative to chlorhexidine, promoting microbiome balance and improved periodontal health.
Collapse
Affiliation(s)
- Simón Pardiñas López
- Periodontology and Oral Surgery, Clínica Médico Dental Pardiñas, Real 66, 3, A Coruña, 15003, Spain.
- Grupo de Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, 15003, Spain.
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud-Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña, A Coruña, 15701, Spain.
- Department of Oral and Maxillofacial Surgery, College of Dentistry, New York University, New York, NY, 10010, USA.
| | - Mónica E García-Caro
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
| | - Juan A Vallejo
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain.
| | - Pablo Aja-Macaya
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
| | - Kelly Conde-Pérez
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
| | - Paula Nión-Cabeza
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
| | - Ismael Khouly
- Department of Oral and Maxillofacial Surgery, College of Dentistry, New York University, New York, NY, 10010, USA
- Multidisciplinary Implant and Aesthetic Miami Institute (M.I.A.M.I.), Miami, FL, 33137, USA
| | - Germán Bou
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
| | - Ana Isabel Rodríguez Cendal
- Grupo de Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, 15003, Spain
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud-Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña, A Coruña, 15701, Spain
| | - Silvia Díaz-Prado
- Grupo de Terapia Celular y Medicina Regenerativa, Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Complexo Hospitalario Universitario de A Coruña (CHUAC), A Coruña, 15003, Spain
- Grupo de Terapia Celular y Medicina Regenerativa, Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Ciencias de la Salud-Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña, A Coruña, 15701, Spain
| | - Margarita Poza
- Grupo de Investigación en Microbiología, Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC)- Hospital Universitario de A Coruña (CHUAC)-Universidade da Coruña (UDC)-CIBER de Enfermedades Infecciosas (CIBERINFEC, ISCIII), Hospital Universitario, Coruña, 15006 A, Spain
- Grupo Microbioma y Salud, Facultad de Ciencias- Centro Interdisciplinar de Química y Biología (CICA), Universidade da Coruña, A Coruña, 15071, Spain
| |
Collapse
|
28
|
Yang Y, Yang Z, Liu H, Zhou Y. Aptamers in dentistry: diagnosis, therapeutics, and future perspectives. Biomater Sci 2025; 13:1368-1378. [PMID: 39523847 DOI: 10.1039/d4bm01233j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Oral health is essential to general health. The diagnosis of dental diseases and treatment planning of dental care need to be straightforward and accurate. Recent studies have reported the use of aptamers in dentistry to achieve a simple diagnosis and facilitate therapy. Aptamers comprise nucleic acid sequences that possess a strong affinity for their target. Synthesized chemically, aptamers have several advantages, including smaller size, higher stability, and lower immunogenicity compared with monoclonal antibodies. They can be used to detect biomarkers in saliva and the presence of various pathogens, or can be used as a targeted drug delivery system for disease treatment. This review highlights current research on aptamers for dental care, especially the recent progress in oral disease diagnosis and therapeutics. The challenges and unresolved problems faced by the clinical use of aptamers are also discussed. In the future, the clinical applications of aptamers will be further extended to include, for example, dental indications and regenerative dentistry.
Collapse
Affiliation(s)
- Yang Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, China
| | - Zhen Yang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, China
| | - Hao Liu
- Central Laboratory, Peking University School and Hospital of Stomatology, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, China.
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, China
| |
Collapse
|
29
|
Huang S, Xia C, Zuo W, Wang R, Xu N, Ye W, Li X, Chen Y, Zhu X. Minocycline-Loaded Titanium Dioxide Nanoparticles for Augmented Synergistic Periodontal Sonodynamic Chemotherapy. Int J Nanomedicine 2025; 20:2727-2741. [PMID: 40066323 PMCID: PMC11892737 DOI: 10.2147/ijn.s501964] [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: 10/20/2024] [Accepted: 02/07/2025] [Indexed: 05/13/2025] Open
Abstract
Background The current clinical treatment of periodontitis usually involves mechanical removal of pathogenic bacteria through ultrasonic scaling and root planing, supplemented with antibacterial medications to inhibit microbial overgrowth. However, the therapeutic efficiency remains unsatisfactory due to complicated periodontal anatomy, limited plaque removal, short retention of antibiotics, and related side effects. Methods and Results To address these issues, we successfully synthesized mesoporous titanium dioxide nanoparticles (MTN) via a sol-gel method, which were modified with hemoglobin (Hb) and loaded with minocycline (MINO). The resulting Hb-MTN/MINO nanoparticles had a size of 215 nm, zeta potential of -19.8 mV±0.9 mV, and uniform shape with a PDI index of 0.176. The modification with hemoglobin (Hb) provided sufficient oxygen for antimicrobial sonodynamic therapy (aSDT), contributing to improved generation of reactive oxygen species (ROS) under low ultrasound intensity. After MINO loading, the system exhibited notable antibacterial efficacy, with a 6 log reduction of bacterial counts compared to the control group. Hb-MTN/MINO was evaluated in vivo in terms of oral index, soft and hard tissues, along with biosafety evaluation in periodontal disease model rats. Hb-MTN/MINO demonstrated a satisfactory therapeutic effect, whereby the periodontal condition of the rats exhibited a greater improvement than the control group, and measurement of the serum levels of inflammatory factors revealed that both IL-6 and MMP-9 were significantly downregulated. Conclusion These findings confirm the potential of Hb-MTN/MINO nanoparticles as a promising treatment option for periodontitis.
Collapse
Affiliation(s)
- Shuying Huang
- Xiamen University School of Public Health, Xiamen, People’s Republic of China
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Chengyao Xia
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People’s Republic of China
| | - Wenbao Zuo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People’s Republic of China
| | - Rui Wang
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Nuo Xu
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Wenxin Ye
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Xiao Li
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Yong Chen
- Department of Stomatology, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Xuan Zhu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, People’s Republic of China
| |
Collapse
|
30
|
Guo J, Han J, Li F, Ma Q, He J, You F, Ren Y, Fu X. 16S rRNA sequencing reveals relationships among enrichment of oral microbiota in the lower respiratory tract and pulmonary nodules malignant progression. Microbiol Spectr 2025; 13:e0128424. [PMID: 39907436 PMCID: PMC11878090 DOI: 10.1128/spectrum.01284-24] [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: 05/26/2024] [Accepted: 12/17/2024] [Indexed: 02/06/2025] Open
Abstract
Micro-aspiration of oral microorganisms results in considerable enrichment within the lower respiratory tract (LRT), constituting an early event in lung cancer pathogenesis. To explore the correlation between malignant risk of pulmonary nodules (PNs) and oral commensals enrichment in LRT, oral saliva and bronchial alveolar lavage fluid samples from 22 low-risk PN patients, 17 intermediate-risk PN patients, and 11 high-risk PN patients were analyzed using 16S rRNA gene sequencing. Alpha and beta diversity analyses reveal minimal variation in oral microbial diversity and abundance among patients with different risks of PN. In contrast, a significant reduction in the diversity of LRT microbiota is observed in patients at high risk of PN. Based on multigroup comparative analysis of species differences and the linear discriminant analysis effect size method, Synergistes and Tannerella were identified as the dominant bacterial genera in the oral and LRT of high-risk PN patients, respectively. The study found that the LRT microbiota of PN patients seemed to originate from the oral, and the high enrichment of oral microbiota in the lower respiratory tract was most common in high-risk PN patients. The predominant bacterial genera present in the oral cavity and LRT of patients with PN were identified through abundance variance analysis. Eight key microbial genera were found in both the oral cavity and LRT: Streptococcus, Granulicatella, Porphyromonas, Bacillus, Neisseria, Alloprevotella, Prevotella, and Leptotrichia. Notably, receiver operating characteristic analysis identified Streptococcus, Granulicatella, and Leptotrichia as reliable biomarkers to differentiate high-risk PN. Spearman correlation analysis confirmed that the accumulation of oral microorganisms in the LRT played an important role in the process of PN cancerization. The co-occurrence network showed that the coexistence of Veillonella and Streptococcus in the oral and LRT may be involved in the occurrence of PN, while the LRT cluster of Rothia occurred in high-risk PN patients. Correlation analysis among species identified microbial communities predominantly composed of Veillonella, which may facilitate pulmonary carcinogenesis. IMPORTANCE This study is the first to elucidate the composition and interrelationships of oral and lower respiratory tract (LRT) microbiota in patients with pulmonary nodule (PN) across varying malignancy risk levels. We conducted an analysis to investigate the correlation between the malignant potential of PNs and the enrichment of oral microbiota within the LRT. Additionally, we explored the feasibility of utilizing oral-lower respiratory commensal microbiota as biomarkers to assess the benign and malignant nature of pulmonary nodules. This study aims to provide evidence supporting early diagnosis and intervention strategies for lung cancer.
Collapse
Affiliation(s)
- Jing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Jierong Han
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Fang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Qiong Ma
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Jiawei He
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
- Cancer Institute, Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Yifeng Ren
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
- Tumor Teaching and Research Office, Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| | - Xi Fu
- Hospital of Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
- Tumor Teaching and Research Office, Chengdu University of Traditional Chinese Medicine, Jinniu District, Chengdu, Sichuan, China
| |
Collapse
|
31
|
Lin J, Li M, Wang L, Lu X, Xu Q, Chen H, Dai D. Plant and animal-derived fusion nanovesicles rescue inflammation-compromised osteogenic potential of periodontal ligament stem cells. Front Cell Dev Biol 2025; 13:1512238. [PMID: 40083664 PMCID: PMC11903722 DOI: 10.3389/fcell.2025.1512238] [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: 10/16/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Periodontitis is a chronic inflammatory disease affecting the supporting tissues of the teeth and has emerged as a global public health issue. Current therapies primarily address pathogenic factors and alleviate symptoms, with limited options available for complete restoration and reconstruction of already absorbed periodontal bone tissue. In this study, we developed a nanotherapeutic strategy utilizing fusion nanovesicles (FVs) to modulate the inflammatory microenvironment and create a regenerative niche for periodontal ligament stem cells (PDLSCs), which play a crucial role in periodontal tissue repair. The FVs are composed of Scutellaria baicalensis nanovesicles (SBNVs) with anti-Porphyromonas gingivalis (P. gingivalis) and anti-inflammatory properties, combined with PDLSC membrane-derived nanovesicles genetically engineered to express TNFR1. These FVs preserved the biological activity of SBNVs and the immunomodulatory function of PDLSCs. Additionally, FVs effectively captured and cleared TNF-α from the microenvironment through TNFR1. Moreover, FVs alleviated the inflammatory response of PDLSCs induced by P. gingivalis-LPS (Pg-LPS) and TNF-α, restoring their proliferation, migration, and osteogenic differentiation capabilities. Hence, this nanotherapeutic strategy holds great potential for treating periodontitis.
Collapse
Affiliation(s)
- Jingxiong Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Manchun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Xingyu Lu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Quanle Xu
- College of Life Sciences, Northwest A&F University, Xianyang, Shaanxi, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Dongling Dai
- Endoscopy Center and Gastroenterology Department, Key Laboratory for Precision Diagnosis and Treatment of Pediatric Digestive System Diseases, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
32
|
Dai W, Ye Y, Mao B, Tang X, Cui S, Zhao J, Feng C, Zhang Q. Microbiological and Metabolomic Analysis of Biomarkers for Grades A and B in Stage II Periodontitis. Inflammation 2025:10.1007/s10753-025-02260-1. [PMID: 40011293 DOI: 10.1007/s10753-025-02260-1] [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: 12/11/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
Periodontitis is a chronic inflammatory disease characterized by inflammation of the periodontal soft tissues and loss of alveolar bone. In the oral environment, subgingival microorganisms and salivary metabolites reflect the host's health status. This study aimed to understand periodontitis severity and progression rate by analyzing subgingival microflora and salivary metabolites to identify potential biomarkers. Fifty-three volunteers with stage II periodontitis were graded using the bone loss (%)/age index into two grades: 33 in grade A (< 0.25) and 20 in grade B (0.25-1.00). Using a case-control study, simultaneously analyzed biomarkers associated with the severity and rate of progression of periodontitis. The red complex, the orange complex, Campylobacter spp., uncultured Candidatus Saccharibacteria and metabolites such as 5-Aminovaleric acid, N1-Acetylspermine showed a significant positive correlation with periodontal clinical parameters. Furthermore, we identified four of the salivary differential metabolites (DL-Leucineamide, Dodecanedioic acid, L-Tyrosine methyl ester and Phenylpyruvic acid) that may serve as potential biomarkers for predicting the rate of periodontitis progression. These results showed that the red complex significantly correlated with periodontitis severity and influenced changes in salivary metabolites. Additionally, biomarkers indicating the progression rate were predominantly amino acid derivatives, confirming that interactions between microorganisms and metabolites may exacerbate periodontitis development.
Collapse
Affiliation(s)
- Wenjie Dai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yuhan Ye
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Chenchen Feng
- Wuxi Stomatological Hospital, Wuxi, 214000, Jiangsu, China.
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, 214122, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
33
|
Makkar H, Sriram G. Advances in modeling periodontal host-microbe interactions: insights from organotypic and organ-on-chip systems. LAB ON A CHIP 2025; 25:1342-1371. [PMID: 39963082 PMCID: PMC11833442 DOI: 10.1039/d4lc00871e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Periodontal disease, a chronic inflammatory condition affecting the supporting structures of teeth, is driven by an imbalanced interaction between the periodontal microbiota and the host inflammatory response. Beyond its local impact, periodontal disease is associated with systemic conditions such as diabetes mellitus, cardiovascular disease, and inflammatory bowel disease, emphasizing the importance of understanding its mechanisms. Traditional pre-clinical models, such as monolayer cultures and animal studies, have provided foundational insights but are limited by their physiological relevance and ethical concerns. Recent advancements in tissue engineering and microfluidic technologies have led to the development of three-dimensional (3D) organotypic culture models and organ-on-chip systems that more closely mimic native tissue microenvironments. This review provides an overview of the evolution of methods to study periodontal host-microbe interactions, from simple 2D monolayer cultures to complex 3D organotypic and microfluidic organ-on-chip (OoC) models. We discuss various fabrication strategies, host-microbe co-culture techniques, and methods for evaluating outcomes in these advanced models. Additionally, we highlight insights gained from gut-on-chip platforms and their potential applications in periodontal research and understanding oral-systemic links of periodontal disease. Through a comprehensive overview of current advancements and future directions, this review provides insights on the transformative potential of OoC technology in periodontal research, offering new avenues for studying disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| |
Collapse
|
34
|
Zhao W, Liu C, Cui X, Chen Q. Immunological landscape of periodontitis and rheumatoid arthritis and their molecular crosstalk. Eur J Med Res 2025; 30:124. [PMID: 39987090 PMCID: PMC11847375 DOI: 10.1186/s40001-025-02376-y] [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: 10/30/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The association between periodontitis (PT) and rheumatoid arthritis (RA) is well-established; however, the molecular mechanisms underlying this relationship remain poorly understood. This study aims to delineate shared genetic and molecular features between PT and RA to uncover potential common pathways involved in their pathogenesis. METHODS Gene expression data sets for PT and RA were retrieved from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) and co-expressed gene modules were identified using weighted gene co-expression network analysis (WGCNA) and the DESeq2 package. Enrichment analyses, including KEGG and Gene Ontology (GO) pathways, as well as immune cell infiltration profiling, were performed to explore shared biological pathways. A protein-protein interaction (PPI) network was constructed to pinpoint key genes linking PT and RA. Functional assays were conducted by overexpressing the identified core gene, PTPRC, in MH7A cells via lentiviral transfection, followed by cell viability (CCK-8), migration, and invasion assays. In addition, transcription factor enrichment and connectivity map (cMAP) analyses were employed to identify common transcriptional regulators and potential therapeutic targets for both conditions. RESULTS WGCNA and DESeq2 analyses revealed 154 shared DEGs between PT and RA, predominantly enriched in immune and inflammatory response pathways. PTPRC emerged as a pivotal shared gene, exhibiting significantly higher expression in PT patients compared to controls. In vitro assays confirmed that PTPRC overexpression enhanced fibroblast proliferation, migration, and invasion. Furthermore, transcription factor enrichment analysis and cMAP identified overlapping transcriptional regulators and potential pharmacological agents for both diseases. CONCLUSIONS This study provides novel insights into shared gene expression profiles and molecular mechanisms linking PT and RA, identifying PTPRC as a potential key regulator. These findings suggest that targeting PTPRC could offer therapeutic opportunities for RA driven by PT.
Collapse
Affiliation(s)
- Weimin Zhao
- The Seventh People's Hospital of Zhengzhou City, Zhengzhou, China.
| | - Chenxu Liu
- School of Basic Medical Science, Zhengzhou University, Zhengzhou, China
| | - Xiangzhi Cui
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qianjiang Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
35
|
Chandra Nayak S, Latha PB, Kandanattu B, Pympallil U, Kumar A, Kumar Banga H. The Oral Microbiome and Systemic Health: Bridging the Gap Between Dentistry and Medicine. Cureus 2025; 17:e78918. [PMID: 40091996 PMCID: PMC11909285 DOI: 10.7759/cureus.78918] [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] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
The oral microbiome, consisting of a mixture of bacteria, fungi, and viruses, is an important contributor to oral and systemic health. Microbial balance disruptions are associated with oral pathologies like dental caries and periodontitis as well as systemic diseases such as cardiovascular diseases, adverse pregnancy outcomes, and respiratory diseases. This review explores the mechanistic pathways linking oral dysbiosis to systemic inflammation, endothelial dysfunction, and immune modulation. The roles of key microbial species in health and disease are analyzed, with an emphasis on how hematogenous dissemination leads to systemic pathologies through inflammatory signaling. Also, advances in high throughput sequencing are discussed, as well as microbial diversity and its implications for diagnostics and therapeutics. The review highlights the potential of oral microbiota-targeted interventions to mitigate systemic diseases through dentistry and medicine integration, by throwing light on interdisciplinary strategies. Future work should focus on the evaluation of the mechanisms by which the oral microbiome plays a role in systemic diseases through the integration of multi-omics approaches such as metagenomics, transcriptomics, and metabolomics. Furthermore, clinical trials need to be designed in a way to evaluate the efficacy of microbiome-targeted therapies in the prevention of cardiovascular diseases, adverse pregnancy outcomes, and autoimmune disorders.
Collapse
Affiliation(s)
- Subash Chandra Nayak
- Department of Conservative Dentistry and Endodontics, Hi-Tech Dental College and Hospital, Bhubaneshwar, IND
| | - P Bhagya Latha
- Department of Zoology, SIR C R Reddy College, Eluru, IND
| | - Bharath Kandanattu
- Pediatric and Preventive Dentistry, Institute of Dental Studies and Technologies, Modinagar, Ghaziabad, IND
| | - Unni Pympallil
- Department of Prosthodontics, Mahe Institute of Dental Sciences & Hospital, Mahe, IND
| | - Ankit Kumar
- Dentistry, Mithila Minority Dental College and Hospital, Darbhanga, IND
| | - Harish Kumar Banga
- Fashion and Lifestyle Accessory Design, National Institute of Fashion Technology, Kangra, IND
| |
Collapse
|
36
|
Peng Y, Iwasaki K, Taguchi Y, Ishikawa I, Umeda M. Mesenchymal stem cell-derived protein extract induces periodontal regeneration. Cytotherapy 2025; 27:201-212. [PMID: 39545910 DOI: 10.1016/j.jcyt.2024.10.003] [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: 07/11/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Periodontal disease is characterized by chronic inflammation and destruction of supporting periodontal tissues, ultimately leading to tooth loss. In recent years, "cell-free treatment" without stem cell transplantation has attracted considerable attention for tissue regeneration. This study investigated the effects of extracts of mesenchymal stem cells (MSC-extract) and their protein components (MSC-protein) on the proliferation and migration of periodontal ligament (PDL) cells and whether MSC-protein can induce periodontal regeneration. METHODS MSC-extract and MSC-protein were obtained by subjecting mesenchymal stem cells (MSCs) to freeze-thaw cycles and acetone precipitation. Cell proliferation was examined using a WST-8 assay and Ki67 immunostaining, and cell migration was examined using Boyden chambers. The MSC-protein content was analyzed using liquid chromatography-mass spectrometry, protein arrays, and enzyme-linked immunosorbent assays (ELISAs). Gene expression in MSC-protein-treated PDL cells was examined using RNA-sequencing and Gene Ontology analyses. The regenerative potential of MSC-protein was examined using micro-computer tomography (CT) and histological analyses after transplantation into a rat periodontal defect model. RESULTS MSC-extract and MSC-protein promoted the proliferation and migration of PDL cells. Protein array and ELISA revealed that MSC-protein contained high concentrations of basic fibroblast growth factor (bFGF) and hepatocyte growth factor (HGF). Exogenous bFGF promoted the proliferation and migration of PDL cells. Furthermore, the transplantation of MSC-protein enhanced periodontal tissue regeneration with the formation of new alveolar bone and PDLs. CONCLUSIONS These results indicate that the MSC-protein promotes the proliferation and migration of PDL cells and induces significant periodontal tissue regeneration, suggesting that the MSC-protein could be used as a new cell-free treatment for periodontal disease.
Collapse
Affiliation(s)
- Yihao Peng
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | - Kengo Iwasaki
- Division of Creative and Integrated Medicine, Advanced Medicine Research Center, Translational Research Institute for Medical Innovation (TRIMI), Osaka Dental University, Osaka, Japan.
| | - Yoichiro Taguchi
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| | | | - Makoto Umeda
- Department of Periodontology, Osaka Dental University, Osaka, Japan
| |
Collapse
|
37
|
Li K, Li B, Li J, Wu X, Zhao Y, Yu J, Guo J, Huang C. Chairside live biotherapeutic hydrogel for comprehensive periodontitis therapy. Trends Biotechnol 2025; 43:408-432. [PMID: 39505614 DOI: 10.1016/j.tibtech.2024.10.001] [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: 05/30/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 11/08/2024]
Abstract
Periodontitis, characterized by microbial dysbiosis and immune dysregulation, destroys tooth-supporting tissues and negatively affects overall health. Current strategies face significant challenges in restoring damaged tissues while halting periodontitis progression. In this study, we introduce a live biotherapeutic product (LBP) in an engineered living hydrogel for comprehensive periodontitis therapy. A dental blue light-responsive hydrogel (LRG) was fabricated to deliver and confine live Lactobacillus rhamnosus GG (LGG) in periodontal pockets, endowing the LRG with sustained antibacterial and immunomodulatory effects. The LRG was engineered through peptide modification to also promote tissue regeneration. Both in vitro and in vivo evaluations confirmed the effectiveness of this integrated therapeutic strategy, which combines antibacterial, anti-inflammatory, and regenerative properties with an underlying immunomodulatory mechanism that involves suppressor of cytokine signaling (SOCS)3 upregulation and the Janus kinase/signal transducer and activator of transcription (JAK-STAT) pathway suppression in macrophages. Demonstrating a new paradigm, this proof of concept highlights the synergistic integration of live organisms and synthetic material engineering in a chairside treatment to address the multifaceted etiology of periodontitis.
Collapse
Affiliation(s)
- Kaifeng Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Boyi Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jiyun Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jian Yu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Jingmei Guo
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Cui Huang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
38
|
Toraman A, Sağlam E, Savran L, Köseoğlu S. Evaluation of Salivary Il-38 Levels in Periodontitis: A Cross-Sectional Study. J Interferon Cytokine Res 2025; 45:76-82. [PMID: 38497769 DOI: 10.1089/jir.2023.0233] [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] [Indexed: 03/19/2024] Open
Abstract
The goal of the current study was to assess levels of salivary interleukin (IL)-38, IL-1β, and IL-10 in various periodontal clinical conditions. In total, 60 (20 healthy, 20 gingivitis, and 20 stage II-III, grade A-B periodontitis) subjects were included in the study. Demographic and clinical periodontal parameters were recorded. Samples were examined for IL-38, IL-1β, and IL-10 levels by means of enzyme-linked immunosorbent assay. Results demonstrated that the periodontitis group had significantly lower salivary IL-38 levels (P < 0.05) than the healthy group. Salivary IL-10 levels did not differ significantly between the groups (P > 0.05). The salivary IL-1β levels of gingivitis (P < 0.001) and periodontitis groups (P < 0.01) were significantly higher than those of the healthy group. The present study indicated that IL-38 level is decreased in periodontal disease. The results suggested a possible role of IL-38 in the periodontal inflammation process. Clarifying the mechanisms of IL-38 in the inflammatory process may contribute to the development of novel treatment strategies in periodontal diseases.
Collapse
Affiliation(s)
- Ayşe Toraman
- Department of Periodontology, Hamidiye Faculty of Dentistry, Health Sciences University, Istanbul, Türkiye
| | - Ebru Sağlam
- Department of Periodontology, Faculty of Dentistry, İstanbul Medeniyet University, Istanbul, Türkiye
| | - Levent Savran
- Department of Periodontology, Faculty of Dentistry, İzmir Katip Çelebi University, Izmir, Türkiye
| | - Serhat Köseoğlu
- Department of Periodontology, Faculty of Dentistry, İstanbul Medeniyet University, Istanbul, Türkiye
| |
Collapse
|
39
|
Anselmi NK, Vanyo ST, Clark ND, Leyva Rodriguez DM, Jones MM, Rosenthal S, Patel D, Marconi RT, Visser MB. Topology and functional characterization of major outer membrane proteins of Treponema maltophilum and Treponema lecithinolyticum. Mol Oral Microbiol 2025; 40:17-36. [PMID: 39263909 PMCID: PMC11752107 DOI: 10.1111/omi.12484] [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: 05/09/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/13/2024]
Abstract
Numerous Treponema species are prevalent in the dysbiotic subgingival microbial community during periodontitis. The major outer sheath protein is a highly expressed virulence factor of the well-characterized species Treponema denticola. Msp forms an oligomeric membrane protein complex with adhesin and porin properties and contributes to host-microbial interaction. Treponema maltophilum and Treponema lecithinolyticum species are also prominent during periodontitis but are relatively understudied. Msp-like membrane surface proteins exist in T. maltophilum (MspA) and T. lecithinolyticum (MspTL), but limited information exists regarding their structural features or functionality. Protein profiling reveals numerous differences between these species, but minimal differences between strains of the same species. Using protein modeling tools, we predict MspA and MspTL monomeric forms to be large β-barrel structures composed of 20 all-next-neighbor antiparallel β strands which most likely adopt a homotrimer formation. Using cell fractionation, Triton X-114 phase partitioning, heat modifiability, and chemical and detergent release assays, we found evidence of amphiphilic integral membrane-associated oligomerization for both native MspA and MspTL in intact spirochetes. Proteinase K accessibility and immunofluorescence assays demonstrate surface exposure of MspA and MspTL. Functionally, purified recombinant MspA or MspTL monomer proteins can impair neutrophil chemotaxis. Expressions of MspA or MspTL with a PelB leader sequence in Escherichia coli also demonstrate surface exposure and can impair neutrophil chemotaxis in an in vivo air pouch model of inflammation. Collectively, our data demonstrate that MspA and MspTL membrane proteins can contribute to pathogenesis of these understudied oral spirochete species.
Collapse
Affiliation(s)
- Natalie K. Anselmi
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Stephen T. Vanyo
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Nicholas D. Clark
- Department of Structural Biology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Dayron M. Leyva Rodriguez
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Megan M. Jones
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Sara Rosenthal
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| | - Dhara Patel
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Richard T. Marconi
- Department of Microbiology and Immunology, Virginia Commonwealth University Medical Center, Richmond, Virginia, USA
| | - Michelle B. Visser
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, The State University of New York, Buffalo, New York, USA
| |
Collapse
|
40
|
de Souza Araújo IJ, Bottino MC. Biofabrication - Revolutionizing the future of regenerative periodontics. Dent Mater 2025; 41:179-193. [PMID: 39632205 DOI: 10.1016/j.dental.2024.11.006] [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: 10/04/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Periodontium is a compartmentalized and highly specialized tissue responsible for tooth stability. Loss of tooth attachment due to periodontitis and trauma is a complex clinical burden affecting a large parcel of the adult and elderly population worldwide, and regenerative strategies to reestablish the native conditions of the periodontium are paramount. Biofabrication of scaffolds, through various techniques and materials, for regenerative periodontics has significantly evolved in the last decades. From the basics of occlusive membranes and graft materials to the complexity of converging 3D printing and Bioprinting using image-based models, biofabrication opens many possibilities for patient-specific scaffolds that recapitulate the anatomical and physiological conditions of periodontal tissues and interfaces. Thus, this review presents fundamental concepts related to the native characteristics of the periodontal tissues, the key to designing personalized strategies, and the latest trends of biofabrication in regenerative periodontics with a critical overview of how these emerging technologies have the potential to shift the one-size-fits-all paradigm.
Collapse
Affiliation(s)
- Isaac J de Souza Araújo
- Department of Bioscience Research, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
41
|
Choi HY, Kim HJ, Lee JY, Joo JY. Adjunctive Treatment Effect of Non-Thermal Atmospheric Pressure Plasma in Periodontitis-Induced Rats. J Clin Med 2025; 14:896. [PMID: 39941567 PMCID: PMC11818203 DOI: 10.3390/jcm14030896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: As non-thermal atmospheric pressure plasma (NTP) is known to have advantages in application in the medical field, we consider its applicability to periodontitis, a representative chronic inflammatory disease. The purpose of this study was to evaluate the effect of NTP in inhibiting the progression of periodontitis in a rat model when additionally used in scaling and root planing (SRP). Methods: To induce experimental periodontitis in 20 rats, ligatures were placed in the maxillary second molar and lipopolysaccharide from Porphyromonas gingivalis was injected around the teeth. Then, NTP treatment was performed for 2 or 5 min, together with scaling and root planing (SRP). To evaluate alveolar bone loss, micro-computed tomography (micro-CT) analysis and hematoxylin-eosin (H-E) staining were performed. Tartrate-resistant acid phosphatase (TRAP) analysis was performed to compare the number of osteoclasts, while immunohistochemistry (IHC) analysis was performed to determine the expression levels of receptor activator of nuclear factor-κB ligand (RANKL) and osteoprotegerin (OPG). Enzyme-linked immunosorbent assay (ELISA) analysis was performed for the detection of cytokines (TNF-α, IL-1β, and IL-10) in tissues and sera. Results: When SRP was combined with NTP, alveolar bone loss was decreased, the number of osteoclasts and RANKL expression were decreased, OPG expression was increased, and pro-inflammatory cytokine (TNF-α and IL-1β) levels were significantly decreased. Compared with the NTP treatment for 2 min, when treated for 5 min, less alveolar bone loss, fewer osteoclasts, a lower RANKL expression level, and a higher OPG expression level were observed. Conclusions: This study evaluated the adjunctive treatment effect of NTP in periodontitis-induced rats. Based on the results of this study, we suggest that supplemental NTP treatment may be a good option for non-surgical periodontal treatment; however, further studies are needed to elucidate the mechanism through which NTP suppresses periodontal inflammation.
Collapse
Affiliation(s)
- Hee-Young Choi
- Department of Periodontology, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (H.-Y.C.); (H.-J.K.); (J.-Y.L.)
| | - Hyun-Joo Kim
- Department of Periodontology, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (H.-Y.C.); (H.-J.K.); (J.-Y.L.)
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea
| | - Ju-Youn Lee
- Department of Periodontology, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (H.-Y.C.); (H.-J.K.); (J.-Y.L.)
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea
| | - Ji-Young Joo
- Department of Periodontology, Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea; (H.-Y.C.); (H.-J.K.); (J.-Y.L.)
- Department of Periodontology, Dental Research Institute, Pusan National University Dental Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
42
|
Huamán-Mendoza AA, Pantigozo-Morán FL, da Silva JC, Chuquimez-Ventura CV, Holzhausen M. Hotspots and global trends in research of host immune response in periodontitis: A bibliometric analysis. J Periodontol 2025. [PMID: 39869002 DOI: 10.1002/jper.24-0473] [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: 07/25/2024] [Revised: 11/21/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The host immune response plays a major role in the pathogenesis of periodontitis. A bibliometric study can be crucial to understanding the different processes involved in this area; however, to our knowledge, it has not been published until now. Therefore, a bibliometric analysis was conducted to assess research hotspots and global trends in scientific articles about the immune response in periodontitis published between 1952 and 2023. METHODS The search strategy was defined using keywords and Boolean operators. The Web of Science Core Collection database was used. In addition, the bibliometric analysis was performed using four tools: Python 3.12, VOSviewer 1.6.19, R-Bibliometrix, and CiteSpace. RESULTS A total of 7696 articles were included, comprising 6691 regular articles and 1005 review articles. Data analysis revealed an increasing trend in the number of publications over the years in this field, with the most cited article written by Preshaw et al. (2012). Leading countries in article production include the United States, China, Japan, and Brazil. Regarding institutions, the University of Helsinki had the highest number of publications (N = 339), with one of its researchers being the author with the most publications (Dr. Sorsa, N = 143, H-index = 52). Concerning journals, the Journal of Periodontology was the most influential journal. Finally, recent trending topics in research were related to the influence of inflammation on dysbiosis, immune biomarkers, immunomodulation, and the impact of aging on immunocompetence. CONCLUSION This bibliometric study demonstrated a growing interest and the emergence of new trends in research on host immune response in periodontitis.
Collapse
Affiliation(s)
- Aldrin André Huamán-Mendoza
- Department of Stomatology, School of Dentistry, University of São Paulo, Av. Prof. Lineu Prestes, São Paulo, SP, Brazil
| | - Fabiola Lucía Pantigozo-Morán
- Department of Stomatology, School of Dentistry, University of São Paulo, Av. Prof. Lineu Prestes, São Paulo, SP, Brazil
| | - José Carlos da Silva
- Department of Stomatology, School of Dentistry, University of São Paulo, Av. Prof. Lineu Prestes, São Paulo, SP, Brazil
| | | | - Marinella Holzhausen
- Department of Stomatology, School of Dentistry, University of São Paulo, Av. Prof. Lineu Prestes, São Paulo, SP, Brazil
| |
Collapse
|
43
|
Razooqi Z, Khzam N, L’Hostis M, Belibasakis GN, Johansson A, Oscarsson J. Prevalence of the oral pathogen Filifactor alocis and its FtxA toxin related to clinical parameters and presence of Aggregatibacter actinomycetemcomitans. Front Cell Infect Microbiol 2025; 14:1501028. [PMID: 39911492 PMCID: PMC11794325 DOI: 10.3389/fcimb.2024.1501028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/19/2024] [Indexed: 02/07/2025] Open
Abstract
The Gram-positive organism Filifactor alocis is implicated in multiple oral diseases including periodontitis, and approximately 50% of known strains encode and produce a recently identified repeat-in-toxin (RTX) protein, FtxA, partly homologous to the Aggregatibacter actinomycetemcomitans leukotoxin. By assessing a longitudinal Ghanaian study population of adolescents, we recently identified a possible correlation between F. alocis levels, ftxA gene carriage, and progression of clinical attachment loss (CAL). To extend knowledge on the possible significance of F. alocis and its FtxA in periodontal disease, we have in the present work analyzed saliva samples in an independent cohort of periodontitis (n=156), collected at two private periodontal specialist practices in Perth, Western Australia. The present results corroborate that high loads of F. alocis and the presence of its ftxA gene together are associated with parameters of periodontal tissue destruction and severity. Moreover, among the individuals carrying A. actinomycetemcomitans, a majority also exhibited an ftxA-positive F. alocis, supporting the notion of the synergistic behavior of these two species. This emphasizes that F. alocis and its ftxA are involved in the pathogenesis of periodontitis and may have ecological roles, with diagnostic and prognostic implications for the disease.
Collapse
Affiliation(s)
| | - Nabil Khzam
- Oral Health Centre of Western Australia, Dental School, The University of Western Australia, Perth, WA, Australia
- NK Periodontics, Perth, WA, Australia
| | | | - Georgios N. Belibasakis
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Oscarsson
- Department of Odontology, Umeå University, Umea, Sweden
| |
Collapse
|
44
|
Shawkatova I, Durmanova V, Javor J. Alzheimer's Disease and Porphyromonas gingivalis: Exploring the Links. Life (Basel) 2025; 15:96. [PMID: 39860036 PMCID: PMC11766648 DOI: 10.3390/life15010096] [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: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Recent research highlights compelling links between oral health, particularly periodontitis, and systemic diseases, including Alzheimer's disease (AD). Although the biological mechanisms underlying these associations remain unclear, the role of periodontal pathogens, particularly Porphyromonas gingivalis, has garnered significant attention. P. gingivalis, a major driver of periodontitis, is recognized for its potential systemic effects and its putative role in AD pathogenesis. This review examines evidence connecting P. gingivalis to hallmark AD features, such as amyloid β accumulation, tau hyperphosphorylation, neuroinflammation, and other neuropathological features consistent with AD. Virulence factors, such as gingipains and lipopolysaccharides, were shown to be implicated in blood-brain barrier disruption, neuroinflammation, and neuronal damage. P. gingivalis-derived outer membrane vesicles may serve to disseminate virulence factors to brain tissues. Indirect mechanisms, including systemic inflammation triggered by chronic periodontal infections, are also supposed to exacerbate neurodegenerative processes. While the exact pathways remain uncertain, studies detecting P. gingivalis virulence factors and its other components in AD-affected brains support their possible role in disease pathogenesis. This review underscores the need for further investigation into P. gingivalis-mediated mechanisms and their interplay with host responses. Understanding these interactions could provide critical insights into novel strategies for reducing AD risk through periodontal disease management.
Collapse
Affiliation(s)
- Ivana Shawkatova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske nam. 14, 811 08 Bratislava, Slovakia; (V.D.); (J.J.)
| | | | | |
Collapse
|
45
|
Huang P, Li W, Guan J, Jia Y, Wang D, Chen Y, Xiao N, Ou S, Wang Y, Yang B. Synthetic Vesicle-Based Drug Delivery Systems for Oral Disease Therapy: Current Applications and Future Directions. J Funct Biomater 2025; 16:25. [PMID: 39852581 PMCID: PMC11766321 DOI: 10.3390/jfb16010025] [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: 12/10/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
Oral diseases such as dental caries, periodontitis, and oral cancer are prevalent and present significant challenges to global public health. Although these diseases are typically treated through procedures like dental preparation and resin filling, scaling and root planning, or surgical excision, these interventions are often not entirely effective, and postoperative drug therapy is usually required. Traditional drug treatments, however, are limited by factors such as poor drug penetration, significant side effects, and the development of drug resistance. As a result, there is a growing need for novel drug delivery systems that can enhance therapeutic efficacy, reduce side effects, and improve treatment outcomes. In recent years, drug-loaded vesicles, such as liposomes, polymersomes, and extracellular vesicles (EVs), have emerged as promising drug delivery platforms due to their high drug encapsulation efficiency, controlled release properties, and excellent biocompatibility. This review provides an in-depth examination of the characteristics, advantages, and limitations of liposomes, polymersomes, and extracellular vesicles in the context of oral disease treatment. It further explores the reasons for their advantages and limitations and discusses the specific applications, development prospects, and strategies for optimizing these vesicle-based systems for improved clinical outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yan Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (P.H.); (W.L.); (J.G.); (Y.J.); (D.W.); (Y.C.); (N.X.); (S.O.)
| | - Bo Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China; (P.H.); (W.L.); (J.G.); (Y.J.); (D.W.); (Y.C.); (N.X.); (S.O.)
| |
Collapse
|
46
|
Amato M, Polizzi A, Viglianisi G, Leonforte F, Mascitti M, Isola G. Impact of Periodontitis and Oral Dysbiosis Metabolites in the Modulation of Accelerating Ageing and Human Senescence. Metabolites 2025; 15:35. [PMID: 39852378 PMCID: PMC11767177 DOI: 10.3390/metabo15010035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Periodontitis, a chronic multifactorial inflammatory condition of the periodontium, is originated by a dysbiotic oral microbiota and is negatively correlated with several systemic diseases. The low-chronic burden of gingival inflammation not only exacerbates periodontitis but also predisposes individuals to a spectrum of age-related conditions, including cardiovascular diseases, neurodegenerative disorders, and metabolic dysfunction, especially related to ageing. In this regard, over the local periodontal treatment, lifestyle modifications and adjunctive therapies may offer synergistic benefits in ameliorating both oral and systemic health in ageing populations. Elucidating the intricate connections between periodontitis and senescence is important for understanding oral health's systemic implications for ageing and age-related diseases. Effective management strategies targeting the oral microbiota and senescent pathways may offer novel avenues for promoting healthy ageing and preventing age-related morbidities. This review will analyze the current literature about the intricate interplay between periodontitis, oral dysbiosis, and the processes of senescence, shedding light on their collective impact on the modulation and accelerated ageing and age-related diseases. Lastly, therapeutic strategies targeting periodontitis and oral dysbiosis to mitigate senescence and its associated morbidities will be discussed.
Collapse
Affiliation(s)
- Mariacristina Amato
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Gaia Viglianisi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Francesco Leonforte
- Hygiene Unit, Department of Medical and Surgical Sciences and Advanced Technologies, University of Catania, 95124 Catania, Italy
| | - Marco Mascitti
- Department of Clinical Specialistic and Dental Sciences, Marche Polytechnic University, 60121 Ancona, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| |
Collapse
|
47
|
Sriram G, Makkar H. Microfluidic organ-on-chip systems for periodontal research: advances and future directions. Front Bioeng Biotechnol 2025; 12:1490453. [PMID: 39840127 PMCID: PMC11747509 DOI: 10.3389/fbioe.2024.1490453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Advances in tissue engineering and microfluidic technologies have enabled the development of sophisticated in vitro models known as organ-on-a-chip (OoC) or microphysiological systems. These systems enable to potential to simulate the dynamic interactions between host tissues and their microenvironment including microbes, biomaterials, mechanical forces, pharmaceutical, and consumer-care products. These fluidic technologies are increasingly being utilized to investigate host-microbe and host-material interactions in oral health and disease. Of interest is their application in understanding periodontal disease, a chronic inflammatory condition marked by the progressive destruction of periodontal tissues, including gingiva, periodontal ligament, and alveolar bone. The pathogenesis of periodontal disease involves a complex interplay between microbial dysbiosis and host immune responses, which can lead to a loss of dental support structures and contribute to systemic conditions such as cardiovascular disease, diabetes, and inflammatory bowel disease. This provides a comprehensive overview of the latest developments in millifluidic and microfluidic systems designed to emulate periodontal host-microbe and host-material interactions. We discuss the critical engineering and biological considerations in designing these platforms, their applications in studying oral biofilms, periodontal tissue responses, and their potential to unravel disease mechanisms and therapeutic targets in periodontal disease.
Collapse
Affiliation(s)
- Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
48
|
Yan N, Zhou H, Jin P, Li T, Liu Q, Ning H, Ma Z, Feng L, Jin T, Deng Y, Wu Z. A Multifunctional Cobalt-Containing Implant for Treating Biofilm Infections and Promoting Osteointegration in Infected Bone Defects Through Macrophage-Mediated Immunomodulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409200. [PMID: 39587976 PMCID: PMC11744729 DOI: 10.1002/advs.202409200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/26/2024] [Indexed: 11/27/2024]
Abstract
Treating bone infections and ensuring bone recovery is one of the major global problems facing modern orthopedics. Prolonged antibiotic use may increase the risk of antimicrobial resistance, and inflammation caused by biofilms can obstruct tissue healing, making bone infection treatment even more challenging. The optimal treatment strategy combines immune response modification to promote osteogenesis with effective bacterial infection removal that does not require long-term antibiotic use. A one-step plasma immersion ion implantation approach is used to create titanium alloy implants incorporating cobalt. According to experimental findings, cobalt-containing titanium implants exhibit improved antibacterial activity by efficiently disrupting biofilm formations and reducing Methicillin-resistant Staphylococcus aureus adherence by over 80%. Additionally, the implants exhibit superior anti-inflammatory and osseointegration properties. RNA sequencing analysis reveals the potential mechanism of Co2+ in regulating the polarization of macrophages toward the anti-inflammatory M2 phenotype, which is crucial for creating an immune environment conducive to bone healing. Concurrently, these implants promote osteogenic differentiation while suppressing osteoclast activity, further supporting bone repair. Overall, without exogenous recombinant proteins or antibiotics, the implants effectively eradicate infections and expedite bone repair, offering a novel therapeutic strategy for complex skeletal diseases with clinical promise.
Collapse
Affiliation(s)
- Nongyang Yan
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
| | - Hao Zhou
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Penghe Jin
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Tengfei Li
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
| | - Qi Liu
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| | - Hao Ning
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Zhixin Ma
- Comprehensive supervision officeAnhui provincial Health Commission435 Tunbrook RoadHefei230032China
| | - Linfei Feng
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
- Department of Oral and Maxillofacial SurgeryThe First Affiliated Hospital of Anhui Medical UniversityNo. 218 Jixi AvenueHeifeiAnhui230032China
| | - Tao Jin
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| | - Youwen Deng
- Department of Spine SurgeryThe Third Xiangya HospitalCentral South UniversityNo. 138 Tongzi RoadChangshaHunan410013China
| | - Zhengwei Wu
- Institute of Advanced TechnologyUniversity of Science and Technology of ChinaNo. 5089 Wangjiang West RoadHefeiAnhui230031China
- School of Nuclear Science and TechnologyUniversity of Science and Technology of ChinaNo. 96 Jinzhai RoadHefeiAnhui230026China
| |
Collapse
|
49
|
Qu C, Luo T, Han R, Mo D, Shi K, Li X, Bei Z, Pan M, Tan S, Liu J, Qian Z. NIR-driven GOx-like nanozyme-modified injectable thermoresponsive hydrogel for antibacterial therapy and bone regeneration in periodontitis. CHEMICAL ENGINEERING JOURNAL 2025; 506:160108. [DOI: 10.1016/j.cej.2025.160108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2025]
|
50
|
Vijayakumar A, Vasudevan S, John S, Ozbun MA, Bartee E, Palanisamy V. Navigating a complex dance: the interplay between RNA-binding proteins and T cells in oral epithelial plasticity. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00054. [PMID: 39816132 PMCID: PMC11731067 DOI: 10.1097/in9.0000000000000054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
The oral epithelium, a dynamic interface constantly facing environmental challenges, relies on intricate molecular pathways to maintain its homeostasis. This comprehensive review delves into the nuanced interplay between T-lymphocytic cells (T cells) and RNA-binding proteins (RBPs) within the oral epithelium, elucidating their roles in orchestrating immune responses and influencing tissue plasticity. By synthesizing current knowledge, we aim to unravel the molecular intricacies that govern this interplay, with a focus on potential therapeutic implications for oral health and diseases. Understanding the regulatory networks shaped by T cells and RBPs in the oral epithelial microenvironment holds promise for innovative strategies in managing conditions associated with epithelial dysfunction.
Collapse
Affiliation(s)
- Anitha Vijayakumar
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Sekar Vasudevan
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Samu John
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Michelle A. Ozbun
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Eric Bartee
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Viswanathan Palanisamy
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|