1
|
Benavides-Reyes C, Cabello I, Magán-Fernández A, Rodríguez-Barranco M, Usta SN, Mesa F. Clinical effects of probiotics on the treatment of gingivitis and periodontitis: a systematic review and meta-analysis. BMC Oral Health 2025; 25:490. [PMID: 40186219 PMCID: PMC11971800 DOI: 10.1186/s12903-025-05888-5] [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: 03/13/2023] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
AIMS This study aimed to evaluate the impact of probiotics as an adjunct to periodontal therapy on clinical outcomes in patients with gingivitis and periodontitis through a meta-analysis of available evidence. MATERIALS AND METHODS A detailed bibliographic search on four databases (PubMed, Scopus, Cochrane and EMBASE) was conducted with a language restriction. The collected data were assessed according to the predefined eligibility criteria and randomized clinical trials reporting the effects of probiotics on plaque index (PI), bleeding on probing (BOP) and pocket probing depth (PPD) compared to control or placebo groups were selected and analysed. The risk of bias assessment was conducted using SYRCLE's RoB- 2 tool. The GRADEpro tool was used to determine the overall quality of evidence. RESULTS Twenty-four studies (10 about gingivitis and 14 about periodontitis) were included in the meta-analysis. In the gingivitis studies, lower but non-significant PI and BOP were found in the probiotic group. In periodontitis, lower PI (95%-CI [- 0.54; - 0.15], p = 0.001) were reported in the probiotic group, and this difference was greater in studies with longer follow-up. Lower BOP (95%-CI [- 0.58; - 0.05], p = 0.021) was also reported, but this difference was only significant in studies with a shorter follow-up (95%-CI [- 0.86; - 0.11], p = 0.012). Meta-analysis for PPD showed lower, but non-significant, values (95%-CI [- 0.53; + 0.03], p = 0.077). However, this difference became significant when assessing studies with shorter follow-up (95% CI [- 0.77; - 0.07], p = 0.019). CONCLUSIONS The meta-analysis provides evidence suggested that probiotics can serve as a beneficial adjunct to periodontal treatment in patients with periodontitis, particularly in improving clinical outcomes such as plaque index and bleeding on probing. The results from gingivitis studies highlight the need for further investigation to better understand the impact of probiotics in the early stages of periodontal disease. These findings emphasize the importance of future research with standardized protocols and longer follow-up periods to confirm and expand on the clinical utility of probiotics in periodontal therapy.
Collapse
Affiliation(s)
- Cristina Benavides-Reyes
- Department of Operative Dentistry, School of Dentistry, University of Granada, Granada, 18071, Spain
| | - Inmaculada Cabello
- Department of Integral Paediatric Dentistry, School of Dentistry, University of Granada, Granada, 18071, Spain.
| | - Antonio Magán-Fernández
- Department of Periodontics, School of Dentistry, University of Granada, Granada, 18071, Spain
| | - Miguel Rodríguez-Barranco
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, 28028, Spain
- Andalusian School of Public Health (EASP), Granada, 18011, Spain
- Instituto de Investigación Biosanitaria Ibs.GRANADA, 18012, Granada, Spain
| | - Sıla Nur Usta
- Department of Endodontics, Gulhane Faculty of Dentistry, University of Health Sciences, Ankara, Etlik, Keçiören, 06018, Turkey
| | - Francisco Mesa
- Department of Periodontics, School of Dentistry, University of Granada, Granada, 18071, Spain
| |
Collapse
|
2
|
Man Y, Li X, Cui L, Song J, Cheng C, Zhang X, Niu F. Dydrogesterone alleviates periodontitis in perimenopausal women undergoing periodontal therapy by decreasing inflammation and mediating oral microbiota. Microb Pathog 2025; 201:107380. [PMID: 39956343 DOI: 10.1016/j.micpath.2025.107380] [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: 09/25/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
OBJECTIVE Dydrogesterone (DG), a synthetic isomer of progesterone, plays a potential regulatory role in the periodontal environment. The aim of this study was to investigate the potential effects of DG on periodontitis under periodontal therapy (PT) and the underlying mechanisms related to oral microbiota. METHODS As a cohort study, perimenopausal women with periodontitis and abnormal uterine bleeding associated with ovulatory dysfunction were screened. A total of 30 women received PT (PT group) and 30 women received PT and oral DG 10 mg twice/day for 10 days/month (PT + DG group). At baseline and 3 months after treatment, pocket probing depth (PPD), bleeding index (BI), bleeding on probing (BOP), plaque index, CRP, IL-6, and TNF-α were measured. Additionally, 16S rDNA sequencing was performed to determine the characteristics of oral microbiota, mainly in terms of abundance, diversity, composition, and community structure. RESULTS Three months after treatment, the levels of PPD, BI, and BOP, as well as the levels of CRP, IL-6, and TNF-α in gingival crevicular fluid were significantly lower in the PT + DG group than those in the PT group. After treatment, a relatively lower microbial abundance, and some differences in microbial composition were revealed between the PT and PT + DG groups. At the genus level, significantly fewer Escherichia-Shigella, Porphyromonas, and Absconditabacteriales (SR1), and more Lactobacillus, Gordonia, Bifidobacterium, and Oribacterium were found in the PT group than in the PT + DG group. CONCLUSIONS DG enhances the effect of PT on inhibiting inflammatory response in women with periodontitis by mediating oral microbiota.
Collapse
Affiliation(s)
- Ying Man
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Xiaofei Li
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Liyun Cui
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Jiajia Song
- Department of Stomatology, Shengli Oilfield Central Hospital, China
| | - Cheng Cheng
- Dongying District Hospital of Traditional Chinese Medicine, China
| | - Xinyue Zhang
- Department of Stomatology, Shengli Oilfield Central Hospital, China.
| | - Feifei Niu
- Department of Gynaecology, Shengli Oilfield Central Hospital, China.
| |
Collapse
|
3
|
Naja JR, Desparois L, Hebert EM, Nader MEF, Saavedra L, Minahk CJ, Houde VP. In vitro modulation of proinflammatory and proteolytic activities of Porphyromonas gingivalis by selected lactobacilli. J Oral Microbiol 2025; 17:2469894. [PMID: 40013015 PMCID: PMC11864006 DOI: 10.1080/20002297.2025.2469894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025] Open
Abstract
Objective The aim of the present study was to characterize the antimicrobial and anti-inflammatory activities of postbiotics from lactic acid bacteria against Porphyromonas gingivalis. Material and methods The anti-P. gingivalis activity of postbiotics from the CERELA culture collection was assessed by measuring changes in the expression of key host proteins by ELISA and qPCR, the proteolytic activity by a fluorescence and a spectrophotometric method and virulence factors from P. gingivalis by qPCR. Results Even though Lacticaseibacillus (L.) rhamnosus CRL1522 and Lactiplantibacillus (L.) plantarum CRL1363 exhibit only a discrete antibacterial activity against P. gingivalis, the cell-free supernatants of these strains significantly reduced P. gingivalis-induced secretion of interleukins IL-6 and IL-8 by keratinocytes and TNF-α and IL-6 by U937 macrophage-like cells. More importantly, P. gingivalis arginine-gingipain (Rgp) protease activity was markedly reduced by both lactic acid bacteria (LAB) strains. This finding is particularly interesting because it means that both LAB might prevent the ulterior citrullination of peptides and the consequent generation of autoantibodies. The expression of COX2 and TLR2 was also significantly downregulated in macrophages. Conclusion Postbiotics from L. rhamnosus CRL1522 and L. plantarum CRL1363 rise as suitable candidates for antagonizing the periodontopathogen P. gingivalis, since they were able to reduce the expression of proinflammatory cytokines and the protein degradation induced by this pathogen. We propose that postbiotics from these LAB could potentially halt the progression of periodontitis based on this in vitro study.
Collapse
Affiliation(s)
- Johana R. Naja
- Laboratorio de Genética y Biología Molecular, Centro de Referencia para Lactobacilos (CERELA), San Miguel de Tucumán, Argentina
- Oral Ecology Research Group (GREB), Faculty of Dental Medicine, Université Laval, Québec, QC, Canada
| | - Leyla Desparois
- Oral Ecology Research Group (GREB), Faculty of Dental Medicine, Université Laval, Québec, QC, Canada
| | - Elvira M. Hebert
- Laboratorio de Genética y Biología Molecular, Centro de Referencia para Lactobacilos (CERELA), San Miguel de Tucumán, Argentina
| | - Maria Elena Fátima Nader
- Laboratorio de Genética y Biología Molecular, Centro de Referencia para Lactobacilos (CERELA), San Miguel de Tucumán, Argentina
| | - Lucila Saavedra
- Laboratorio de Genética y Biología Molecular, Centro de Referencia para Lactobacilos (CERELA), San Miguel de Tucumán, Argentina
| | - Carlos J. Minahk
- Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, Instituto de Química Biológica “Dr. Bernabé Bloj”, Facultad de Bioquímica, Química y Farmacia, UNT, San Miguel de Tucumán, Argentina
| | - Vanessa P. Houde
- Oral Ecology Research Group (GREB), Faculty of Dental Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
4
|
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
|
5
|
Xia M, Lei L, Zhao L, Xu W, Zhang H, Li M, Hu J, Cheng R, Hu T. The dynamic oral-gastric microbial axis connects oral and gastric health: current evidence and disputes. NPJ Biofilms Microbiomes 2025; 11:1. [PMID: 39747247 PMCID: PMC11696714 DOI: 10.1038/s41522-024-00623-4] [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: 04/02/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025] Open
Abstract
Emerging evidence indicates that oral microbes are closely related to gastric microbes and gastric lesions, including gastric atrophy, intestinal metaplasia and gastric cancer (GC). Helicobacter pylori is a key pathogen involved in GC. However, the increasing prevalence of H. pylori-negative GC and gastric dysbiosis in GC patients emphasize the potential role of other microbial factors. In this review, we discussed the current evidence about the relationship between the oral-gastric microbial axis and oral and gastric health. Epidemiologic evidence indicates that poor oral hygiene is related to greater GC risk. Multiple oral-associated microbes are enriched in the stomach of GC patients. Once colonizing the stomach, oral-associated microbes Streptococcus anginosus and Prevotella melaninogenica, are involved in gastric inflammation or carcinogenesis. Microbial metabolites such as lactate, nitrite, and acetaldehyde promote malignant transformation. The stomach, as a checkpoint of microbial transmission in the digestive tract, is of great importance since the link between oral microbes and intestinal diseases has been emphasized. Still, new technologies and standardized metrics are necessary to identify potential pathogenetic microbes for GC and the core microbiota, interactions, richness, colonization, location and effect (CIRCLE). In the future, oral microbes could be candidates for noninvasive indicators to predict gastric diseases.
Collapse
Affiliation(s)
- Mengying Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Microbiology, ADA Forsyth Institute, Cambridge, USA
| | - Linyong Zhao
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Wenqing Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Faculty of Dentistry, University of Hong Kong, Hong Kong, China
| | - Hongyu Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Mingming Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiankun Hu
- Gastric Cancer Center and Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Ran Cheng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Tao Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Frontier Innovation Center for Dental Medicine Plus & Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
6
|
Dharmarathne G, Kazi S, King S, Jayasinghe TN. The Bidirectional Relationship Between Cardiovascular Medications and Oral and Gut Microbiome Health: A Comprehensive Review. Microorganisms 2024; 12:2246. [PMID: 39597635 PMCID: PMC11596509 DOI: 10.3390/microorganisms12112246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of widespread morbidity and mortality. It has been found that the gut and oral microbiomes differ in individuals with CVDs compared to healthy individuals. Patients with CVDs often require long-term pharmacological interventions. While these medications have been extensively studied for their cardiovascular benefits, emerging research indicates that they may also impact the diversity and composition of the oral and gut microbiomes. However, our understanding of how these factors influence the compositions of the oral and gut microbiomes in individuals remains limited. Studies have shown that statins and beta-blockers, in particular, cause gut and oral microbial dysbiosis, impacting the metabolism and absorption of these medications. These alterations can lead to variations in drug responses, highlighting the need for personalized treatment approaches. The microbiome's role in drug metabolism and the impact of CVD medications on the microbiome are crucial in understanding these variations. However, there are very few studies in this area, and not all medications have been studied, emphasizing the necessity for further research to conclusively establish cause-and-effect relationships and determine the clinical significance of these interactions. This review will provide evidence of how the oral and gut microbiomes in patients with cardiovascular diseases (CVDs) interact with specific drugs used in CVD treatment.
Collapse
Affiliation(s)
- Gangani Dharmarathne
- Australian Laboratory Services Global, Water and Hydrographic, Hume, ACT 2620, Australia
| | - Samia Kazi
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Sydney, NSW 2145, Australia
| | - Shalinie King
- Westmead Applied Research Centre, The University of Sydney, Sydney, NSW 2145, Australia
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Thilini N. Jayasinghe
- The Sydney Dental School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- The Charles Perkins Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Hashim NT, Babiker R, Priya SP, Mohammed R, Chaitanya NCSK, Padmanabhan V, El Bahra S, Rahman MM, Gismalla BG. Microbial Dynamics in Periodontal Regeneration: Understanding Microbiome Shifts and the Role of Antifouling and Bactericidal Materials: A Narrative Review. Curr Issues Mol Biol 2024; 46:12196-12213. [PMID: 39590318 PMCID: PMC11592519 DOI: 10.3390/cimb46110724] [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: 10/06/2024] [Revised: 10/21/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Periodontal regeneration is a multifaceted therapeutic approach to restore the tooth-supporting structures lost due to periodontal diseases. This manuscript explores the intricate interactions between regenerative therapies and the oral microbiome, emphasizing the critical role of microbial balance in achieving long-term success. While guided tissue regeneration (GTR), bone grafting, and soft tissue grafting offer promising outcomes in terms of tissue regeneration, these procedures can inadvertently alter the oral microbial ecosystem, potentially leading to dysbiosis or pathogenic recolonization. Different grafting materials, including autografts, allografts, xenografts, and alloplasts, influence microbial shifts, with variations in the healing timeline and microbial stabilization. Biologics and antimicrobials, such as enamel matrix derivatives (EMD) and sub-antimicrobial dose doxycycline (SDD), play a key role in promoting microbial homeostasis by supporting tissue repair and reducing pathogenic bacteria. Emerging strategies, such as enzyme-based therapies and antifouling materials, aim to disrupt biofilm formation and enhance the effectiveness of periodontal treatments. Understanding these microbial dynamics is essential for optimizing regenerative therapies and improving patient outcomes. The future of periodontal therapy lies in the development of advanced materials and strategies that not only restore lost tissues but also stabilize the oral microbiome, ultimately leading to long-term periodontal health.
Collapse
Affiliation(s)
- Nada Tawfig Hashim
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Rasha Babiker
- RAK College of Medical Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 11172, United Arab Emirates;
| | - Sivan Padma Priya
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Riham Mohammed
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Nallan CSK Chaitanya
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Vivek Padmanabhan
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Shadi El Bahra
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | - Muhammed Mustahsen Rahman
- RAK College of Dental Sciences, RAK Medical & Health Sciences University, Ras Al Khaimah P.O. Box 12973, United Arab Emirates; (S.P.P.); (R.M.); (N.C.C.); (V.P.); (S.E.B.); (M.M.R.)
| | | |
Collapse
|
8
|
Janson TM, Ramenzoni LL, Hatz CR, Schlagenhauf U, Attin T, Schmidlin PR. Limosilactobacillus reuteri supernatant attenuates inflammatory responses of human gingival fibroblasts to LPS but not to elevated glucose levels. J Periodontal Res 2024; 59:974-981. [PMID: 38764133 DOI: 10.1111/jre.13290] [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: 11/07/2023] [Revised: 05/04/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024]
Abstract
AIM We investigated the in vitro effect of Limosilactobacillus reuteri DSM 17938 supernatant on the inflammatory response of human gingival fibroblasts (HGF) challenged by lipopolysaccharide (LPS) or elevated glucose levels. METHODS HGF were exposed to LPS (1 μg/mL), glucose (5, 12 mM or 25 mM), and dilutions of supernatant prepared from L. reuteri DSM 17938 (0.5 × 107, 1.0 × 107, 2.5 × 107, and 5.0 × 107 CFU/mL). After 24 h cell viability and levels of cytokines (IL-1β, IL-6 and IL-8) and TLR-2 were determined. RESULTS None of the tested L. reuteri (DSM 17938) supernatant concentrations reduced the viability of HGF. Supernatant concentrations (2.5 × 107 and 5 × 107 CFU/mL) significantly (p < .05) decreased the production of IL-1β, IL-6, IL-8, and TLR-2 in the presence of LPS. In contrast, inflammatory markers were not reduced by L. reuteri supernatant in the presence of glucose. Glucose concentrations of 12 mM and 24 mM still lead to an elevated production of the investigated biochemical mediators. CONCLUSION While L. reuteri (DSM 17938) supernatant attenuates the inflammatory response of HGF to LPS in a dose-dependent manner, elevated glucose levels suppress this action. These in vitro results support the overall anti-inflammatory efficacy of L. reuteri supplementation in plaque-associated periodontal inflammations.
Collapse
Affiliation(s)
- T M Janson
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - L L Ramenzoni
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - C R Hatz
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - U Schlagenhauf
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
- Department of Conservative Dentistry and Periodontology, Center for Oral Health, University Hospital Wuerzburg, Wuerzburg, Germany
| | - T Attin
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| | - P R Schmidlin
- Division of Periodontology and Peri-implant Diseases, Clinic of Conservative and Preventive Dentistry, Center for Dental Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Mohammed S, Çon AH. Postbiotic nanoparticles (postbiotics-NPs): a novel strategy for providing probiotics' health advantages through food consumption. Food Sci Biotechnol 2024; 33:2729-2736. [PMID: 39184983 PMCID: PMC11339192 DOI: 10.1007/s10068-024-01629-6] [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: 02/08/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, the term "postbiotics" has become increasingly popular in food microbiology, food science, and commercial products. This importance has been raised due to the safety issues associated with live bacterial cells. Postbiotics are defined as bioactive substances of probiotics. It confers unique health-promoting functions with its chemical structure, safe profile, and long shelf life. Meanwhile, postbiotics nanoparticles (postbiotics-NPs) can be considered novel postbiotic delivery systems to deliver bioactive components with health benefits and therapeutic aims, promote the shelf-life of food products, and develop novel functional foods. The present scientific literature shows that nanotechnology approaches are not yet mature enough to be used in postbiotic delivery systems. For all of that, the potential applications of postbiotics-NPs in the food industry and biomedical fields will be a new trend in the future.
Collapse
Affiliation(s)
- Sarhan Mohammed
- Department of Food Engineering, Faculty of Engineering, Ondokuz Mayis University, 55139 Atakum, Samsun Turkey
| | - Ahmet Hilmi Çon
- Department of Food Engineering, Faculty of Engineering, Ondokuz Mayis University, 55139 Atakum, Samsun Turkey
| |
Collapse
|
10
|
Beibei L, Mengying W, Xiao H, Yuzi J, Lijin M, Ke Z, Shengjie Y, Li L. Dysbiosis and interactions of the mycobiome and bacteriome in mucosal lesions of erosive and non-erosive oral lichen planus patients. J Oral Microbiol 2024; 16:2374639. [PMID: 38979477 PMCID: PMC11229720 DOI: 10.1080/20002297.2024.2374639] [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: 04/19/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
Background Oral lichen planus (OLP) is a common oral mucosal disease, clinically categorized into erosive OLP (EOLP) and non-erosive OLP (NEOLP) based on symptoms, but its pathogenic mechanism remains unclear. This study aims to explore the relationship between OLP and the oral microbiome. Methods We collected oral mucosal samples from 49 patients and 10 healthy individuals and conducted 16S rRNA and ITS gene sequencing to explore the oral fungal and bacterial communities. Results We observed significantly lower α diversity of fungi in the EOLP group, with Candida being significantly enriched as the main dominant genus. In the NEOLP group, Aspergillaceae were significantly enriched. The EOLP group showed significant enrichment of Aggregatibacter and Lactobacillus, but the relative abundance of Streptococcus was notably lower than in the other two groups. In the NEOLP group, two species including Prevotella intermedia were significantly enriched. The microbial co-occurrence and co-exclusion networks display distinct characteristics across the three groups, with Lactobacillus assuming a significant bridging role in the ELOP group. Conclusions Our study indicates that EOLP and NEOLP experience varying degrees of dysbiosis at both the fungal and bacterial levels. Therefore, the pathogenic mechanisms and interactive relationships of these microbiota associated with OLP merit further in-depth investigation.
Collapse
Affiliation(s)
- Liang Beibei
- Hebei Key Laboratory of Stomatology, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Wei Mengying
- Department of Dental Pulp, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Huo Xiao
- Department of Oral Mucosa, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Jing Yuzi
- School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Mi Lijin
- Department of Oral Mucosa, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Zhang Ke
- Department of Oral Mucosa, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Yi Shengjie
- School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| | - Liu Li
- Department of Oral Mucosa, School and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
11
|
Puzhankara L, Banerjee A, Chopra A, Venkitachalam R, Kedlaya MN. Effectiveness of probiotics compared to antibiotics to treat periodontal disease: Systematic review. Oral Dis 2024; 30:2820-2837. [PMID: 37964394 DOI: 10.1111/odi.14781] [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: 07/11/2023] [Revised: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVES Probiotics are promising adjuncts to non-surgical periodontal therapy (scaling and root planing (SRP)) for managing gingival and periodontal diseases. Probiotics are considered alternatives to antibiotics, especially with the emergence of antimicrobial resistance. Hence, the present systematic review aims to generate evidence on the role of probiotics compared to antimicrobial agents for managing periodontal diseases (gingivitis and periodontitis). METHOD Six electronic databases (PubMed, SCOPUS, Web of Science, EBSCO, Cochrane, Clinical Trial Registry) were searched to collect studies comparing the effect of probiotics with antibiotics for periodontal disease. In total, 5530 articles were retrieved from all databases, of which 1891 were included for title and abstract screening. After screening, a total of ten clinical studies were included for data extraction and analysis. Probing pocket depth (PPD), Clinical attachment loss (CAL), bleeding on probing (BOP), plaque index (PI), gingival index (GI), and microbial profile were recorded. RESULTS Probiotics showed a significant reduction in the PPD and CAL compared to antibiotics. Antibiotics were more effective in reducing the PI and GI. A combination of probiotics and antibiotics superior compared to probiotics and antibiotics alone. CONCLUSION Probiotics can be used as an alternative to antibiotics, however, a combination is more effective for managing periodontal disease.
Collapse
Affiliation(s)
- Lakshmi Puzhankara
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Avishikta Banerjee
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Aditi Chopra
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, India
| | | | - Madhurya N Kedlaya
- Department of Periodontology, Manipal College of Dental Sciences, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
12
|
Lee Y, Jung BH, Yoo KY, Lim HJ, Shin KJ, Lee JK. Lactobacillus fermentum attenuates the alveolar bone loss in ligature-induced periodontitis in mice. Oral Dis 2024; 30:3328-3335. [PMID: 37724481 DOI: 10.1111/odi.14739] [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: 05/01/2023] [Revised: 08/14/2023] [Accepted: 08/29/2023] [Indexed: 09/20/2023]
Abstract
OBJECTIVE This study investigated the effects of Lactobacillus fermentum BELF11 on periodontitis in mice (LIP). METHODS Sixty mice were randomly assigned to a control group (CTL), LIP/PBS group (LIP and PBS applied), or LIP/BELF11 group (LIP and L. fermentum BELF11 applied). For 14 days, PBS or L. fermentum BELF11 was applied twice daily to the mice in the LIP/PBS or LIP/BELF11 group, respectively. After 14 days, radiographic, histological, and pro-inflammatory cytokine assessments were conducted. RESULTS The LIP/PBS and LIP/BELF11 groups demonstrated greater alveolar bone loss than the CTL group (p < 0.05). The LIP/BELF11 group showed significantly reduced alveolar bone loss on the mesial side compared to the LIP/PBS group. Histologically, the LIP/BELF11 group showed consistent patterns of connective tissue fiber arrangement, lower levels of inflammatory infiltration, less alveolar bone loss, and higher alveolar bone density than the LIP/PBS group, despite showing more signs of destruction than the CTL group. The LIP/BELF11 group also exhibited significantly lower levels of pro-inflammatory cytokines than the LIP/PBS group. CONCLUSIONS L. fermentum BELF11 inhibits alveolar bone loss and periodontitis progression by regulating pro-inflammatory cytokine production. These findings suggest that L. fermentum BELF11 may be a potential adjunctive therapy in periodontal treatment.
Collapse
Affiliation(s)
- Yuni Lee
- Department of Periodontology and Research Institute of Oral Sciences, Gangneung-Wonju National University College of Dentistry, Gangneung, Republic of Korea
| | - Bo Hyun Jung
- Department of Anatomy and Research Institute of Oral Sciences, Gangneung-Wonju National University College of Dentistry, Gangneung, Republic of Korea
| | - Ki-Yeon Yoo
- Department of Anatomy and Research Institute of Oral Sciences, Gangneung-Wonju National University College of Dentistry, Gangneung, Republic of Korea
| | - Hye Ji Lim
- R&D Center, Hecto Healthcare Co., Ltd, Seoul, Republic of Korea
| | - Kum-Joo Shin
- R&D Center, Hecto Healthcare Co., Ltd, Seoul, Republic of Korea
| | - Jae-Kwan Lee
- Department of Periodontology and Research Institute of Oral Sciences, Gangneung-Wonju National University College of Dentistry, Gangneung, Republic of Korea
| |
Collapse
|
13
|
Shi Q, Sun L, Gao J, Li F, Chen D, Shi T, Tan Y, Chang H, Liu X, Kang J, Lu F, Huang Z, Zhao H. Effects of sodium lauryl sulfate and postbiotic toothpaste on oral microecology. J Oral Microbiol 2024; 16:2372224. [PMID: 38939048 PMCID: PMC11210412 DOI: 10.1080/20002297.2024.2372224] [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: 01/29/2024] [Accepted: 06/18/2024] [Indexed: 06/29/2024] Open
Abstract
The diversity and delicate balance of the oral microbiome contribute to oral health, with its disruption leading to oral and systemic diseases. Toothpaste includes elements like traditional additives such as sodium lauryl sulfate (SLS) as well as novel postbiotics derived from probiotics, which are commonly employed for maintaining oral hygiene and a healthy oral cavity. However, the response of the oral microbiota to these treatments remains poorly understood. In this study, we systematically investigated the impact of SLS, and toothpaste containing postbiotics (hereafter, postbiotic toothpaste) across three systems: biofilms, animal models, and clinical populations. SLS was found to kill bacteria in both preformed biofilms (mature biofilms) and developing biofilms (immature biofilms), and disturbed the microbial community structure by increasing the number of pathogenic bacteria. SLS also destroyed periodontal tissue, promoted alveolar bone resorption, and enhanced the extent of inflammatory response level. The postbiotic toothpaste favored bacterial homeostasis and the normal development of the two types of biofilms in vitro, and attenuated periodontitis and gingivitis in vivo via modulation of oral microecology. Importantly, the postbiotic toothpaste mitigated the adverse effects of SLS when used in combination, both in vitro and in vivo. Overall, the findings of this study describe the impact of toothpaste components on oral microflora and stress the necessity for obtaining a comprehensive understanding of oral microbial ecology by considering multiple aspects.
Collapse
Affiliation(s)
- Qingying Shi
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
| | - Lianlian Sun
- Stomatology Department, Binhai Hospital of Peking University, Tianjin, China
| | - Jing Gao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Fengzhu Li
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Dongxiao Chen
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Tingting Shi
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Youlan Tan
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Huimin Chang
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Xiaozhi Liu
- Central Laboratory, Binhai Hospital of Peking University, Tianjin, China
| | - Jian Kang
- Periodontal Disease Department, Tianjin Stomatological Hospital, Tianjin, China
| | - Fuping Lu
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
| | - Zhengmei Huang
- Oral and Skin Microecology Institute of Tust & Benzhen, Science and Technology Park of Tianjin University of Science and Technology, Tianjin, China
| | - Huabing Zhao
- College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, Tianjin, China
- Oral and Skin Microecology Institute of Tust & Benzhen, Science and Technology Park of Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
14
|
Manoil D, Parga A, Bostanci N, Belibasakis GN. Microbial diagnostics in periodontal diseases. Periodontol 2000 2024; 95:176-193. [PMID: 38797888 DOI: 10.1111/prd.12571] [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: 02/06/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
Microbial analytical methods have been instrumental in elucidating the complex microbial etiology of periodontal diseases, by shaping our understanding of subgingival community dynamics. Certain pathobionts can orchestrate the establishment of dysbiotic communities that can subvert the host immune system, triggering inflammation and tissue destruction. Yet, diagnosis and management of periodontal conditions still rely on clinical and radiographic examinations, overlooking the well-established microbial etiology. This review summarizes the chronological emergence of periodontal etiological models and the co-evolution with technological advances in microbial detection. We additionally review the microbial analytical approaches currently accessible to clinicians, highlighting their value in broadening the periodontal assessment. The epidemiological importance of obtaining culture-based antimicrobial susceptibility profiles of periodontal taxa for antibiotic resistance surveillance is also underscored, together with clinically relevant analytical approaches to guide antibiotherapy choices, when necessary. Furthermore, the importance of 16S-based community and shotgun metagenomic profiling is discussed in outlining dysbiotic microbial signatures. Because dysbiosis precedes periodontal damage, biomarker identification offers early diagnostic possibilities to forestall disease relapses during maintenance. Altogether, this review highlights the underutilized potential of clinical microbiology in periodontology, spotlighting the clinical areas most conductive to its diagnostic implementation for enhancing prevention, treatment predictability, and addressing global antibiotic resistance.
Collapse
Affiliation(s)
- Daniel Manoil
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Ana Parga
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Microbiology and Parasitology, CIBUS-Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Georgios N Belibasakis
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| |
Collapse
|
15
|
Van Holm W, Zayed N, Lauwens K, Saghi M, Axelsson J, Aktan MK, Braem A, Simoens K, Vanbrabant L, Proost P, Van Holm B, Maes P, Boon N, Bernaerts K, Teughels W. Oral Biofilm Composition, Dissemination to Keratinocytes, and Inflammatory Attenuation Depend on Probiotic and Synbiotic Strain Specificity. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10253-z. [PMID: 38619794 DOI: 10.1007/s12602-024-10253-z] [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] [Accepted: 04/01/2024] [Indexed: 04/16/2024]
Abstract
Several inflammatory diseases are characterized by a disruption in the equilibrium between the host and its microbiome. Due to the increase in resistance, the use of antibiotics for the widespread, nonspecific killing of microorganisms is at risk. Pro-microbial approaches focused on stimulating or introducing beneficial species antagonistic toward pathobionts may be a viable alternative for restoring the host-microbiome equilibrium. Unfortunately, not all potential probiotic or synbiotic species and even subspecies (to strain level) are equally effective for the designated pathology, leading to conflicting accounts of their efficacy. To assess the extent of these species- and strain-specific effects, 13 probiotic candidates were evaluated for their probiotic and synbiotic potential with glycerol on in vitro oral biofilms, dissemination from biofilms to keratinocytes, and anti-inflammatory activity. Species- and strain-specific effects and efficacies were observed in how they functioned as probiotics or synbiotics by influencing oral pathobionts and commensals within biofilms and affected the dissemination of pathobionts to keratinocytes, ranging from ineffective strains to strains that reduced pathobionts by 3 + log. In addition, a minority of the candidates exhibited the ability to mitigate the inflammatory response of LPS-stimulated monocytes. For a comprehensive assessment of probiotic therapy for oral health, a judicious selection of fully characterized probiotic strains that are specifically tailored to the designated pathology is required. This approach aims to challenge the prevailing perception of probiotics, shifting the focus away from "form over function." Rather than using unproven, hypothetical probiotic strains from known genera or species, one should choose strains that are actually functional in resolving the desired pathology before labelling them probiotics.
Collapse
Affiliation(s)
- Wannes Van Holm
- KU Leuven, Department of Oral Health Sciences, Periodontology and Oral Microbiology, B-3000, Leuven, Belgium
- Ghent University (UGent), Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Naiera Zayed
- KU Leuven, Department of Oral Health Sciences, Periodontology and Oral Microbiology, B-3000, Leuven, Belgium
- Ghent University (UGent), Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
- Faculty of Pharmacy, Menoufia University, Shebeen El-Kom, Egypt
| | - Katalina Lauwens
- KU Leuven, Department of Oral Health Sciences, Periodontology and Oral Microbiology, B-3000, Leuven, Belgium
| | - Mehraveh Saghi
- KU Leuven, Department of Oral Health Sciences, Periodontology and Oral Microbiology, B-3000, Leuven, Belgium
| | | | - Merve Kübra Aktan
- KU Leuven, Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering, B-3000, Leuven, Belgium
| | - Annabel Braem
- KU Leuven, Department of Materials Engineering (MTM), Biomaterials and Tissue Engineering, B-3000, Leuven, Belgium
| | - Kenneth Simoens
- KU Leuven, Department of Chemical Engineering, Bio- and Chemical Systems Technology, B-3000, Leuven, Belgium
| | - Lotte Vanbrabant
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Research Group Immunity and Inflammation, B-3000, Leuven, Belgium
| | - Paul Proost
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Research Group Immunity and Inflammation, B-3000, Leuven, Belgium
| | - Bram Van Holm
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, B-3000, Leuven, Belgium
| | - Piet Maes
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Clinical and Epidemiological Virology, B-3000, Leuven, Belgium
| | - Nico Boon
- Ghent University (UGent), Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Kristel Bernaerts
- KU Leuven, Department of Chemical Engineering, Bio- and Chemical Systems Technology, B-3000, Leuven, Belgium
| | - Wim Teughels
- KU Leuven, Department of Oral Health Sciences, Periodontology and Oral Microbiology, B-3000, Leuven, Belgium.
| |
Collapse
|
16
|
Zhao D, Li MH, Pan T, Guo J, Li J, Shi C, Wang N, Huang H, Wang C, Yang G. Preventive and Therapeutic Potential of Streptococcus cristatus CA119 in Experimental Periodontitis in Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10254-y. [PMID: 38607584 DOI: 10.1007/s12602-024-10254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Periodontitis is an inflammatory condition of the oral cavity caused by a mixed infection of various bacteria, which not only severely affects the alveolar bone and connective tissues but also displays potential correlations with distal intestinal inflammation. In this study, we aimed to elucidate the therapeutic effects of Streptococcus cristatus CA119 on experimental periodontitis in rats and its impact on intestinal morphology. The results demonstrate that CA119 is capable of colonizing the oral cavity and exerting antagonistic effects on Porphyromonas gingivalis and Fusobacterium nucleatum, thus leading to a significant reduction in the oral pathogen load. Following CA119 intervention, there was a significant alleviation of weight loss in rats induced by periodontitis (P < 0.001). CA119 also regulated the expression of IL-6 (P < 0.05), IL-1β (P < 0.001), IL-18 (P < 0.001), COX-2 (P < 0.001), iNOS (P < 0.001), and MCP-1 (P < 0.01) in the gingival tissue. Additionally, CA119 reduced oxidative stress levels in rats and enhanced their antioxidant capacity. Microcomputed tomography (micro-CT) and histological analysis revealed that CA119 significantly reduced alveolar bone loss and reversed the downregulation of OPG/RANKL (P < 0.001). Furthermore, CA119 exhibited a significant protective effect against intestinal inflammation induced by periodontal disease and improved the colonic morphology in rats. In conclusion, this study demonstrates the role of CA119 as a potential oral probiotic in the prevention and treatment of experimental periodontitis, underscoring the potential of probiotics as a complementary approach to traditional periodontal care.
Collapse
Affiliation(s)
- Dongyu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming-Han Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tianxu Pan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jialin Guo
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Junyi Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
17
|
Wang Y, Yang F, Wang Y, Deng S, Zhu R. Alterations and correlations in dental plaque microbial communities and metabolome characteristics in patients with caries, periodontitis, and comorbid diseases. BMC Oral Health 2024; 24:132. [PMID: 38273329 PMCID: PMC10811826 DOI: 10.1186/s12903-023-03785-3] [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: 05/12/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUNDS The pathogenic microorganisms and clinical manifestations of caries and periodontitis are different, caries and periodontitis are usually discussed separately, and the relationship between them is ignored. Clinically, patients prone to dental caries generally have a healthier periodontal status, whereas patients with periodontitis generally have a lower incidence of dental caries. The relationship between dental caries and periodontitis remains unclear. OBJECTIVES This study aimed to explain the clinical phenomenon of antagonism between dental caries and periodontitis by exploring the ecological chain and bacterial interactions in dental caries, periodontitis, and other comorbid diseases. METHODS The dental plaque microbiomes of 30 patients with oral diseases (10 each with caries, periodontitis, and comorbid diseases) were sequenced and analysed using 16 S rRNA gene sequencing. The Kyoto Encyclopaedia of Genes and Genomes (KEGG) database was used for a differential functional analysis of dental plaque microbial communities in caries, periodontitis, and comorbid diseases. RESULTS The coinfection group had the greatest bacterial richness in dental plaque. The principal coordinate analysis showed that caries and periodontitis were separate from each other, and comorbid diseases were located at the overlap of caries and periodontitis, with most of them being periodontitis. Simultaneously, we compared the microbiomes with significant differences among the three groups and the correlations between the microbiome samples. In addition, KEGG pathway analysis revealed significant differences in functional changes among the three groups. CONCLUSIONS This study revealed the composition of the dental plaque microbial communities in caries, periodontitis, and comorbidities and the differences among the three. Additionally, we identified a possible antagonism between periodontitis and caries. We identified a new treatment strategy for the prediction and diagnosis of caries and periodontitis.
Collapse
Affiliation(s)
- Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Fei Yang
- Department of Nutrition and Food Hygiene, School of Public Health, School of Medicine, Zhejiang University, 866 Yu-hang-tang Road, Hangzhou, Zhejiang Province, 310058, China
| | - Yuan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, China.
| | - Rui Zhu
- Department of Stomatology, Affiliated XiaoShan Hospital, Hangzhou Normal University, Hangzhou, 310000, China.
| |
Collapse
|
18
|
Petrariu OA, Barbu IC, Niculescu AG, Constantin M, Grigore GA, Cristian RE, Mihaescu G, Vrancianu CO. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front Microbiol 2024; 14:1296447. [PMID: 38249451 PMCID: PMC10797027 DOI: 10.3389/fmicb.2023.1296447] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Collapse
Affiliation(s)
- Oana-Alina Petrariu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ilda Czobor Barbu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Georgiana Alexandra Grigore
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
19
|
Ghaffarpour M, Karami‐Zarandi M, Rahdar HA, Feyisa SG, Taki E. Periodontal disease in down syndrome: Predisposing factors and potential non-surgical therapeutic approaches. J Clin Lab Anal 2024; 38:e25002. [PMID: 38254289 PMCID: PMC10829694 DOI: 10.1002/jcla.25002] [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: 03/18/2023] [Revised: 11/06/2023] [Accepted: 12/31/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Periodontal diseases (PDs) have been documented to be significantly more prevalent and severe in patients with Down syndrome (DS). Different immunological and microbiological factors contributed to predisposing these patients to progressive and recurrent PDs. AIM The aim of this review was to investigate the altered immunological responses and oral microbiota disorders as well as focus on adjunctive non-surgical methods for the treatment of PDs and its applicability in patients with DS. MATERIAL AND METHODS A literature review was conducted addressing the following topics: (1) the altered immunological responses, (2) orofacial disorders related to DS patients, (3) oral microbiota changing, and (4) adjunctive non-surgical treatment and its efficacy in patients with DS. RESULTS Due to the early onset of PDs in children with DS, the need for prompt and effective treatment in these patients is essential. DISCUSSION AND CONCLUSION So, investigating underlying factors may open a new window to better understand the pathology of PDs in DS people and thus, find better strategies for treatment in such group. Although non-surgical treatments such as photodynamic therapy and probiotic consumption represented acceptable outcomes in different examined patients without DS, data about the application of these convenience and no need for local anesthesia methods in patients with DS is limited.
Collapse
Affiliation(s)
- Mahdie Ghaffarpour
- Department of Oral Medicine, School of DentistryTehran University of Medical SciencesTehranIran
| | - Morteza Karami‐Zarandi
- Department of Microbiology, School of MedicineZanjan University of Medical SciencesZanjanIran
| | - Hossein Ali Rahdar
- Department of Microbiology, School of MedicineIranshahr University of Medical SciencesIranshahrIran
| | - Seifu Gizaw Feyisa
- Department of Medical LaboratorySalale University College of Health SciencesFicheEthiopia
| | - Elahe Taki
- Department of Microbiology, School of MedicineKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
20
|
Kim D, Choi H, Oh H, Lee J, Hwang Y, Kang SS. Mutanolysin-Digested Peptidoglycan of Lactobacillus reuteri Promotes the Inhibition of Porphyromonas gingivalis Lipopolysaccharide-Induced Inflammatory Responses through the Regulation of Signaling Cascades via TLR4 Suppression. Int J Mol Sci 2023; 25:42. [PMID: 38203215 PMCID: PMC10779245 DOI: 10.3390/ijms25010042] [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: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Periodontitis is an oral infectious disease caused by various pathogenic bacteria, such as Porphyromonas gingivalis. Although probiotics and their cellular components have demonstrated positive effects on periodontitis, the beneficial impact of peptidoglycan (PGN) from probiotic Lactobacillus remains unclear. Therefore, our study sought to investigate the inhibitory effect of PGN isolated from L. reuteri (LrPGN) on P. gingivalis-induced inflammatory responses. Pretreatment with LrPGN significantly inhibited the production of interleukin (IL)-1β, IL-6, and CCL20 in RAW 264.7 cells induced by P. gingivalis lipopolysaccharide (LPS). LrPGN reduced the phosphorylation of PI3K/Akt and MAPKs, as well as NF-κB activation, which were induced by P. gingivalis LPS. Furthermore, LrPGN dose-dependently reduced the expression of Toll-like receptor 4 (TLR4), indicating that LrPGN inhibits periodontal inflammation by regulating cellular signaling cascades through TLR4 suppression. Notably, LrPGN exhibited stronger inhibition of P. gingivalis LPS-induced production of inflammatory mediators compared to insoluble LrPGN and proteinase K-treated LrPGN. Moreover, MDP, a minimal bioactive PGN motif, also dose-dependently inhibited P. gingivalis LPS-induced inflammatory mediators, suggesting that MDP-like molecules present in the LrPGN structure may play a crucial role in the inhibition of inflammatory responses. Collectively, these findings suggest that LrPGN can mitigate periodontal inflammation and could be a useful agent for the prevention and treatment of periodontitis.
Collapse
Affiliation(s)
- Donghan Kim
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| | - Hanhee Choi
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| | - Hyeonjun Oh
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| | - Jiyeon Lee
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| | - Yongjin Hwang
- Novalacto Co., Ltd., Daejon 34016, Republic of Korea
| | - Seok-Seong Kang
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University, Goyang 10326, Republic of Korea
| |
Collapse
|
21
|
Mayta-Tovalino F, Espinoza-Carhuancho F, Alvitez-Temoche D, Calderon I, Munive-Degregori A, Barja-Ore J, Diaz-Arocutipa C. A 10-year Scientometric Study of the Emerging Patterns and Spatial-Temporal Trends of Probiotics in Dentistry. J Contemp Dent Pract 2023; 24:981-986. [PMID: 38317396 DOI: 10.5005/jp-journals-10024-3617] [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: 02/07/2024]
Abstract
AIM To perform a bibliometric analysis of the scientific production related to the use of probiotics in oral health. MATERIALS AND METHODS A retrospective, descriptive, observational study with a scientometric approach was conducted. Metadata from scientific articles were retrieved from the Web of Science (WoS) Core Collection and analyzed using Medical Subject Headings (MeSH) and Emtree terms. The search strategy included the terms related to probiotics and oral health, and articles published between 2013 and 2022 were analyzed. Different scientometric indicators were used to analyze the production, impact, and network collaboration. RESULTS A total of 485 articles were retrieved from 241 sources, with an average annual growth rate of 10.72%. The manuscripts received an average citation per doc of 15.2. Authors collaborated on an average of 5.76 coauthors per paper. The number of articles published increased progressively from 2013 to 2022. The year 2022 had the highest number of articles published (90), while in 2013 only 36 articles were published. There was a progressive decrease in the average number of citations per article, from 32.44 in 2013 to 2.41 in 2022. CONCLUSION The production of articles on probiotics in dentistry has grown steadily, with an annual growth rate of 10.72%. The most productive countries were the United States, Italy, and China. The importance of international scientific collaborations and the need for more research in this field is emphasized. CLINICAL SIGNIFICANCE The findings of this study may help researchers identify areas that require further study and lead to more rapid and efficient advances in the use of probiotics to improve oral health. Its clinical importance lies in its value and ability to inform and guide future research to advance this field of science. How to cite this article: Mayta-Tovalino F, Espinoza-Carhuancho F, Alvitez-Temoche D, et al. A 10-year Scientometric Study of the Emerging Patterns and Spatial-Temporal Trends of Probiotics in Dentistry. J Contemp Dent Pract 2023;24(12):981-986.
Collapse
Affiliation(s)
- Frank Mayta-Tovalino
- Department of Clinical Epidemiology and Biostatistics, School of Dentistry, Universidad Cientifica del Sur, Lima, Peru, Phone: +51 12142500, e-mail:
| | - Fran Espinoza-Carhuancho
- Grupo de Bibliometría, Evaluación de evidencia y Revisiones Sistemáticas (BEERS), Human Medicine Career, Universidad Cientifica del Sur, Lima, Peru
| | - Daniel Alvitez-Temoche
- Academic Department, Faculty of Dentistry, Universidad Nacional Federico Villarreal, Lima, Peru
| | - Ivan Calderon
- Academic Department, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | | | - John Barja-Ore
- Department of Research, Universidad Privada del Norte, Lima, Peru
| | | |
Collapse
|
22
|
Wu YH, Wang CW, Kapila Y. EVIDENCE SUPPORTS SHORT-TERM CLINICAL BENEFITS OF ADJUNCTIVE ORAL PROBIOTICS FOLLOWING SCALING AND ROOT PLANING TREATMENT. J Evid Based Dent Pract 2023; 23:101916. [PMID: 38035893 DOI: 10.1016/j.jebdp.2023.101916] [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: 12/02/2023]
Abstract
ARTICLE TITLE AND BIBLIOGRAPHIC INFORMATION Hu D, Zhong T, Dai Q. Clinical efficacy of probiotics as an adjunctive therapy to scaling and root planning in the management of periodontitis: a systematic review and meta-analysis of randomized controlled trails. J Evid Based Dent Pract. 2021;21(2):101547. doi:10.1016/j.jebdp.2021.101547. SOURCE OF FUNDING Self-funded. TYPE OF STUDY/DESIGN Systematic review with meta-analysis of data.
Collapse
|
23
|
Chen S, Zhang Y. Mechanism and application of Lactobacillus in type 2 diabetes-associated periodontitis. Front Public Health 2023; 11:1248518. [PMID: 38098816 PMCID: PMC10720667 DOI: 10.3389/fpubh.2023.1248518] [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: 06/27/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) accelerates the progression of periodontitis through diverse pathways. Abnormal immune responses, excessive activation of inflammation, increased levels of advanced glycation end products, and oxidative stress have defined roles in the pathophysiological process of T2DM-associated periodontitis. Furthermore, in the periodontium of diabetic individuals, there are high levels of advanced glycation end-products and glucose. Meanwhile, progress in microbiomics has revealed that dysbacteriosis caused by T2DM also contributes to the progression of periodontitis. Lactobacillus, owing to its fine-tuning function in the local microbiota, has sparked tremendous interest in this field. Accumulating research on Lactobacillus has detailed its beneficial role in both diabetes and oral diseases. In this study, we summarize the newly discovered mechanisms underlying Lactobacillus-mediated improvement of T2DM-associated periodontitis and propose the application of Lactobacillus in the clinic.
Collapse
Affiliation(s)
- Sisi Chen
- Chongqing Three Gorges Medical College, Chongqing, China
- Chongqing Medical University, Chongqing, China
| | - Yuhan Zhang
- Chongqing Three Gorges Medical College, Chongqing, China
- Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Peng R, Zhang Z, Qu Y, Chen W. The impact of Helicobacter pylori eradication with vonoprazan-amoxicillin dual therapy combined with probiotics on oral microbiota: a randomized double-blind placebo-controlled trial. Front Microbiol 2023; 14:1273709. [PMID: 37849923 PMCID: PMC10577438 DOI: 10.3389/fmicb.2023.1273709] [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: 08/08/2023] [Accepted: 09/11/2023] [Indexed: 10/19/2023] Open
Abstract
Background Helicobacter pylori infection and eradication have been reported to cause dysbiosis of the oral microbiota. Probiotics are increasingly being used to maintain the balance of the oral microbiota. We aimed to investigate the effects of H. pylori infection, H. pylori eradication with vonoprazan-amoxicillin dual therapy, and probiotics supplementation on the oral microbiota. Methods H. pylori positive patients were randomly assigned to a vonoprazan-amoxicillin regimen plus probiotics (BtT group) or the placebo (PT group) for 14 days. H. pylori negative population served as normal controls. Tongue coating samples were collected from 60 H. pylori positive patients at three time points (before H. pylori eradication, after H. pylori eradication, and at confirmation of H. pylori infection cure) and 20 H. pylori negative subjects. 16S rRNA gene sequencing was used to analyze the oral microbiota. Results H. pylori was detected in the oral cavity in positive (34/60), negative (7/20), and eradicated (1/60) subjects using high-throughput sequencing. Compared with normal controls, H. pylori positive patients exhibited higher richness (p = 0.012) and comparable diversity (p = 0.075) of oral microbiota. Beta diversity and KEGG analysis showed oral flora composition and function differences in H. pylori positive and negative subjects. Alpha diversity dramatically decreased after H. pylori eradication and modestly increased with confirmation of H. pylori eradication. Beta diversity and LEfSe analysis revealed distinct structures, and KEGG analysis showed distinct signaling pathways of tongue coating flora at three time points. There was a significant reduction of Firmicutes and Lactobacillus after H. pylori erdication. The PT group and BtT group had identical compositional and functional differences of oral microbiota at three time points. Conclusion No substantial link existed between oral and stomach H. pylori, while removing gastric H. pylori helped eliminate oral H. pylori. H. pylori infection and vonoprazan-amoxicillin dual therapy affected oral microbiota diversity, structure, and function. H. pylori eradication demonstrated a suppressive impact on the proliferation of oral pathogens, specifically Firmicutes and Lactobacillus. Nevertheless, probiotics supplementation did not reduce the oral microbial disturbance caused by H. pylori eradication. Clinical trial registration https://www.chictr.org.cn/, identifiers CHICTR2200060023.
Collapse
Affiliation(s)
| | - Zhenyu Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | | | | |
Collapse
|
25
|
Irie K, Azuma T, Tomofuji T, Yamamoto T. Exploring the Role of IL-17A in Oral Dysbiosis-Associated Periodontitis and Its Correlation with Systemic Inflammatory Disease. Dent J (Basel) 2023; 11:194. [PMID: 37623290 PMCID: PMC10453731 DOI: 10.3390/dj11080194] [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: 05/16/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Oral microbiota play a pivotal role in maintaining homeostasis, safeguarding the oral cavity, and preventing the onset of disease. Oral dysbiosis has the potential to trigger pro-inflammatory effects and immune dysregulation, which can have a negative impact on systemic health. It is regarded as a key etiological factor for periodontitis. The emergence and persistence of oral dysbiosis have been demonstrated to mediate inflammatory pathology locally and at distant sites. The heightened inflammation observed in oral dysbiosis is dependent upon the secretion of interleukin-17A (IL-17A) by various innate and adaptive immune cells. IL-17A has been found to play a significant role in host defense mechanisms by inducing antibacterial peptides, recruiting neutrophils, and promoting local inflammation via cytokines and chemokines. This review seeks to present the current knowledge on oral dysbiosis and its prevention, as well as the underlying role of IL-17A in periodontitis induced by oral dysbiosis and its impact on systemic inflammatory disease.
Collapse
Affiliation(s)
- Koichiro Irie
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| | - Tetsuji Azuma
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Takaaki Tomofuji
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Tatsuo Yamamoto
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| |
Collapse
|
26
|
López-Valverde N, López-Valverde A, Montero J, Rodríguez C, Macedo de Sousa B, Aragoneses JM. Antioxidant, anti-inflammatory and antimicrobial activity of natural products in periodontal disease: a comprehensive review. Front Bioeng Biotechnol 2023; 11:1226907. [PMID: 37600299 PMCID: PMC10435350 DOI: 10.3389/fbioe.2023.1226907] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Periodontal diseases (PD) are common chronic inflammatory oral pathologies that are strongly linked to others not found in the mouth cavity. The immune system mediates the host response, which includes the upregulation of proinflammatory cytokines, metalloproteinases, and reactive oxygen species (ROS); the latter may play an important role in the establishment and progression of inflammatory diseases, particularly periodontal disease, via the development of oxidative stress (OS). Natural antioxidants have powerful anti-inflammatory properties, and some can reduce serum levels of key PD indicators such tumor necrosis factor (TNF) and interleukin IL-1. This review compiles, through a thorough literature analysis, the antioxidant, anti-inflammatory, and antibacterial effects of a variety of natural products, as well as their therapeutic potential in the treatment of PD.
Collapse
Affiliation(s)
- Nansi López-Valverde
- Department of Medicine and Medical Specialties, Faculty of Health Sciences, Universidad Alcalá de Henares, Madrid, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Madrid, Spain
| | - Antonio López-Valverde
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Javier Montero
- Department of Surgery, Instituto de Investigación Biomédica de Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | - Cinthia Rodríguez
- Department of Dentistry, Universidad Federico Henríquez y Carvajal, Santo Domingo, Dominican Republic
| | - Bruno Macedo de Sousa
- Institute for Occlusion and Orofacial Pain Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | |
Collapse
|
27
|
Sulistiowati CP, Suhartono M, Rahmawati DF, Ulfah N, Supandi SK, Wijaksana IKE, Abullais SS, Dhadse P. In-Vitro Inhibitory Efficacy of 3 Types of Probiotics on the Growth of Aggregatibacter actinomycetemcomitans Bacteria. FRONT BIOSCI-LANDMRK 2023; 28:106. [PMID: 37258475 DOI: 10.31083/j.fbl2805106] [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: 02/12/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Amongst the specific plaque pathogen Aggregatibacter actinomycetemcomitans (Aa) ATCC 43718 serotype b is one of the highly virulent bacteria that causes periodontitis. Probiotic therapy is a treatment in which the lactic acid bacteria in are utilized to impede the colonization and growth of the pathogenic bacteria to prevent the further formation of dental plaque. OBJECTIVE The present research aimed to evaluate inhibiting effect of purified bacteria from various commercially available yogurt product containing bacteria named (Lactobacillus casei strain Shirota; Lactobacillus bulgaricus and Streptococcus thermophilus; Lactobacillus reuteri Prodentis) on the growth of Aa. METHODS The research made use of the diffusion method by fixing Aa on BHIB (brain heart infusion broth) medium, incubated at 37 °C and 24 hours later planted on MHA (Mueller-Hinton agar) media. Aa were divided into four subgroups each with a paper disk; group 1 consists of untreated bacteria (i.e., control group), group 2 with purified bacteria from Yakult 0.5 μL, group 3 with purified bacteria from Cimory Yogurt Drink 0.5 μL and group 4 with purified bacteria from BioGaia Prodentis 0.5 μL. All commercially available yogurt were treated to get the purified probiotic. Additionally, it was incubated for 24 hours at 37 °C and later the inhibition zone diameter was observed. RESULTS In the research, it was found that the average impeding ability, so-called inhibition zone, in group 1 indicated 0 mm, group 2 indicated 12.70 mm, group 3 indicated 16.60 mm and group 4 indicated 19.60 mm. The statistical test outcomes showed a significance of 0.000 (p < 0.05). CONCLUSIONS The purified bacteria from three probiotics indeed inhibit the growth of the Aa bacteria and a substantial difference in the diameter of the inhibition zone were found among the three probiotics.
Collapse
Affiliation(s)
| | - Michelle Suhartono
- Department of Periodontology, Universitas Airlangga, 160132 Ulfah, Indonesia
| | - Diana F Rahmawati
- Department of Periodontology, Universitas Airlangga, 160132 Ulfah, Indonesia
| | - Noer Ulfah
- Department of Periodontology, Universitas Airlangga, 160132 Ulfah, Indonesia
| | - Shafira K Supandi
- Department of Periodontology, Universitas Airlangga, 160132 Ulfah, Indonesia
| | | | - Shahabe Saquib Abullais
- Department of Periodontics and Community Dental Sciences, College of Dentistry, King Khalid University, 62529 Abha, Saudi Arabia
- Department of Periodontics, Datta Meghe Institute of Higher Education and Research, Deemed to be University, 442001 Wardha, India
| | - Prasad Dhadse
- Department of Periodontics, Datta Meghe Institute of Higher Education and Research, Deemed to be University, 442001 Wardha, India
| |
Collapse
|
28
|
Park DY, Hwang J, Kim Y, Lee D, Kim YY, Kim HS, Hwang I. Antimicrobial activity of Limosilactobacillus fermentum strains isolated from the human oral cavity against Streptococcus mutans. Sci Rep 2023; 13:7969. [PMID: 37198248 DOI: 10.1038/s41598-023-35168-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 05/13/2023] [Indexed: 05/19/2023] Open
Abstract
Oral probiotics have been recently gaining much attention owing to their potential to inhibit the progression of dental caries by controlling the cariogenic effects of Streptococcus mutans. We isolated and genotypically identified 77 lactic acid bacteria including 12 Limosilactobacillus fermentum probiotic candidates from the oral cavity of healthy volunteers. Among the 12 L. fermentum isolates, nine isolates effectively inhibited the growth of S. mutans via hydrogen peroxide (H2O2) production. The others neither suppressed the growth of S. mutans nor produced H2O2. Eight out of the nine H2O2-producing L. fermentum isolates exhibited strong adherence to oral epithelial KB cells while inhibiting the adherence of S. mutans to KB cells. The eight H2O2-producing isolates were neither haemolytic based on a blood-agar test, cytotoxic according to lactate dehydrogenase assay, nor resistant to eight antibiotics represented by the European Food Safety Authority guideline, indicating that the isolates have potential to suppress the cariogenesis driven by S. mutans while providing general probiotic benefits.
Collapse
Affiliation(s)
| | | | - Yunji Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-Si, South Korea
| | - Dahye Lee
- Apple Tree Dental Hospital, Apple Tree Medical Foundation, Goyang-Si, South Korea
| | - Young-Youn Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-Si, South Korea
- Apple Tree Dental Hospital, Apple Tree Medical Foundation, Goyang-Si, South Korea
| | - Hye-Sung Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-Si, South Korea
- Apple Tree Dental Hospital, Apple Tree Medical Foundation, Goyang-Si, South Korea
| | | |
Collapse
|
29
|
Li Y, Huang S, Du J, Wu M, Huang X. Current and prospective therapeutic strategies: tackling Candida albicans and Streptococcus mutans cross-kingdom biofilm. Front Cell Infect Microbiol 2023; 13:1106231. [PMID: 37249973 PMCID: PMC10213903 DOI: 10.3389/fcimb.2023.1106231] [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: 11/23/2022] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
Candida albicans (C. albicans) is the most frequent strain associated with cross-kingdom infections in the oral cavity. Clinical evidence shows the co-existence of Streptococcus mutans (S. mutans) and C. albicans in the carious lesions especially in children with early childhood caries (ECC) and demonstrates the close interaction between them. During the interaction, both S. mutans and C. albicans have evolved a complex network of regulatory mechanisms to boost cariogenic virulence and modulate tolerance upon stress changes in the external environment. The intricate relationship and unpredictable consequences pose great therapeutic challenges in clinics, which indicate the demand for de novo emergence of potential antimicrobial therapy with multi-targets or combinatorial therapies. In this article, we present an overview of the clinical significance, and cooperative network of the cross-kingdom interaction between S. mutans and C. albicans. Furthermore, we also summarize the current strategies for targeting cross-kingdom biofilm.
Collapse
Affiliation(s)
- Yijun Li
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen, China
| | - Shan Huang
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jingyun Du
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Minjing Wu
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases and Fujian Provincial Engineering Research Center of Oral Biomaterial and Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
30
|
Minervini G, Franco R, Marrapodi MM, Fiorillo L, Badnjević A, Cervino G, Cicciù M. Probiotics in the Treatment of Radiotherapy-Induced Oral Mucositis: Systematic Review with Meta-Analysis. Pharmaceuticals (Basel) 2023; 16:ph16050654. [PMID: 37242437 DOI: 10.3390/ph16050654] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The inflammatory injury of the mucous membranes lining the digestive tract, from the mouth to the anus, is called mucositis. One of the intriguing and compelling new therapeutic modalities that has emerged in recent decades due to advances in our understanding of this condition's pathophysiology is probiotics. The purpose of this meta-analysis is to evaluate the efficiency of probiotics in the treatment of chemotherapy-induced mucositis for head and neck malignancies; a literature search was performed on PubMed, Lilacs, and Web of Science, and articles published from 2000 to 31 January 2023 were considered, according to the keywords entered. The term "Probiotics" was combined with "oral mucositis" using the Boolean connector AND; at the end of the research, 189 studies were identified from the search on the three engines. Only three were used to draw up the present systematic study and metanalysis; this meta-analysis showed that the treatment of mucositis with probiotics is an effective method, and the analysis of the results of these studies showed that the use of probiotics promoted a decrease in the severity of mucositis symptoms.
Collapse
Affiliation(s)
- Giuseppe Minervini
- Multidisciplinary Department of Medical-Surgical and Odontostomatological Specialties, University of Campania "Luigi Vanvitelli", 80121 Naples, Italy
| | - Rocco Franco
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", 00100 Rome, Italy
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", 80121 Naples, Italy
| | - Luca Fiorillo
- School of Dentistry, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, via Consolare Valeria, 1, 98125 Messina, Italy
| | - Almir Badnjević
- Verlab Research Institute for Biomedical Engineering, Medical Devices and Artificial Intelligence, 71000 Sarajevo, Bosnia and Herzegovina
| | - Gabriele Cervino
- School of Dentistry, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, via Consolare Valeria, 1, 98125 Messina, Italy
| | - Marco Cicciù
- Department of Biomedical and Surgical and Biomedical Sciences, Catania University, 95123 Catania, Italy
| |
Collapse
|
31
|
Tagg JR, Harold LK, Jain R, Hale JDF. Beneficial modulation of human health in the oral cavity and beyond using bacteriocin-like inhibitory substance-producing streptococcal probiotics. Front Microbiol 2023; 14:1161155. [PMID: 37056747 PMCID: PMC10086258 DOI: 10.3389/fmicb.2023.1161155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
The human oral cavity contains a diversity of microbial habitats that have been adopted and adapted to as homeland by an amazingly heterogeneous population of microorganisms collectively referred to as the oral microbiota. These microbes generally co-habit in harmonious homeostasis. However, under conditions of imposed stress, as with changes to the host’s physiology or nutritional status, or as a response to foreign microbial or antimicrobial incursions, some components of the oral “microbiome” (viz. the in situ microbiota) may enter a dysbiotic state. This microbiome dysbiosis can manifest in a variety of guises including streptococcal sore throats, dental caries, oral thrush, halitosis and periodontal disease. Most of the strategies currently available for the management or treatment of microbial diseases of the oral cavity focus on the repetitive “broad sweep” and short-term culling of oral microbe populations, hopefully including the perceived principal pathogens. Both physical and chemical techniques are used. However, the application of more focused approaches to the harnessing or elimination of key oral cavity pathogens is now feasible through the use of probiotic strains that are naturally adapted for oral cavity colonization and also are equipped to produce anti-competitor molecules such as the bacteriocins and bacteriocin-like inhibitory substances (viz BLIS). Some of these probiotics are capable of suppressing the proliferation of a variety of recognized microbial pathogens of the human mouth, thereby assisting with the restoration of oral microbiome homeostasis. BLIS K12 and BLIS M18, the progenitors of the BLIS-producing oral probiotics, are members of the human oral cavity commensal species Streptococcus salivarius. More recently however, a number of other streptococcal and some non-streptococcal candidate oral probiotics have also been promoted. What is becoming increasingly apparent is that the future for oral probiotic applications will probably extend well beyond the attempted limitation of the direct pathological consequences of oral microbiome dysbiosis to also encompass a plethora of systemic diseases and disorders of the human host. The background to and the evolving prospects for the beneficial modulation of the oral microbiome via the application of BLIS-producing S. salivarius probiotics comprises the principal focus of the present review.
Collapse
|
32
|
Deandra FA, Ketherin K, Rachmasari R, Sulijaya B, Takahashi N. Probiotics and metabolites regulate the oral and gut microbiome composition as host modulation agents in periodontitis: A narrative review. Heliyon 2023; 9:e13475. [PMID: 36820037 PMCID: PMC9937986 DOI: 10.1016/j.heliyon.2023.e13475] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is defined as an oral bacterial dysbiosis-induced persistent inflammation on dental supporting tissue resulting in periodontal tissue breakdown and alveolar bone destruction. The disease is initiated by the interaction between periodontopathogens and the host immune system. Its development and severity can be associated with several systemic diseases, such as cardiovascular disease (CVD), diabetes mellitus, and rheumatoid arthritis (RA). Moreover, the latest research has suggested that the oral and gut microbiome hypothesis lays the oral and systemic connection mechanism. Bacterial homeostasis and restoration in the oral cavity and intestine become therapeutics concepts. Concerning the treatment of periodontitis, a local inflammatory condition, prolonged systemic administration of antibiotics is no longer recommended due to bacterial resistance issues. Probiotics and several bioactive metabolites have been widely investigated to address the needs of host modulation therapy in periodontitis. Evidence suggests that the use of probiotics helps downregulate the inflammation process through the regulation of toll-like receptor 4 (TLR4) and the production of fatty acid, targeting reactive oxygen species (ROS). In brief, several herbals have anti-inflammatory properties by inhibiting pro-inflammatory cytokines and mediators, including mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB). Consistently, improvement of periodontal pocket depth (PPD) and gingival index (GI) was seen in a group given melatonin as an adjunct treatment. In all, this review will highlight host modulation agents regarding periodontitis therapy, plausible mechanisms on how probiotics and metabolites work on periodontal restoration, and their reported studies. Limitations given by published studies will be elaborated, while future directions will be proposed.
Collapse
Affiliation(s)
- Fathia Agzarine Deandra
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Ketherin Ketherin
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Rieska Rachmasari
- Postgraduate Program in Periodontology, Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia
| | - Benso Sulijaya
- Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia,Dental Division, Universitas Indonesia Hospital, Depok, West Java, Indonesia,Corresponding author. Department of Periodontology, Universitas Indonesia, Jakarta, Indonesia.
| | - Naoki Takahashi
- Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| |
Collapse
|
33
|
Van Holm W, Carvalho R, Delanghe L, Eilers T, Zayed N, Mermans F, Bernaerts K, Boon N, Claes I, Lebeer S, Teughels W. Antimicrobial potential of known and novel probiotics on in vitro periodontitis biofilms. NPJ Biofilms Microbiomes 2023; 9:3. [PMID: 36681674 PMCID: PMC9867767 DOI: 10.1038/s41522-023-00370-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
Several oral diseases are characterized by a shift within the oral microbiome towards a pathogenic, dysbiotic composition. Broad-spectrum antimicrobials are often part of patient care. However, because of the rising antibiotic resistance, alternatives are increasingly desirable. Alternatively, supplying beneficial species through probiotics is increasingly showing favorable results. Unfortunately, these probiotics are rarely evaluated comparatively. In this study, the in vitro effects of three known and three novel Lactobacillus strains, together with four novel Streptococcus salivarius strains were comparatively evaluated for antagonistic effects on proximal agar growth, antimicrobial properties of probiotic supernatant and the probiotic's effects on in vitro periodontal biofilms. Strain-specific effects were observed as differences in efficacy between genera and differences within genera. While some of the Lactobacillus candidates were able to reduce the periodontal pathobiont A. actinomycetemcomitans, the S. salivarius strains were not. However, the S. salivarius strains were more effective against periodontal pathobionts P. intermedia, P. gingivalis, and F. nucleatum. Vexingly, most of the Lactobacillus strains also negatively affected the prevalence of commensal species within the biofilms, while this was lower for S. salivarius strains. Both within lactobacilli and streptococci, some strains showed significantly more inhibition of the pathobionts, indicating the importance of proper strain selection. Additionally, some species showed reductions in non-target species, which can result in unexpected and unexplored effects on the whole microbiome.
Collapse
Affiliation(s)
- Wannes Van Holm
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium ,grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Rita Carvalho
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| | - Lize Delanghe
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Tom Eilers
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Naiera Zayed
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium ,grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium ,grid.411775.10000 0004 0621 4712Faculty of Pharmacy, Menoufia University, Shibin el Kom, Egypt
| | - Fabian Mermans
- grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Kristel Bernaerts
- grid.5596.f0000 0001 0668 7884Bio- and Chemical Systems Technology, Reactor Engineering and Safety, Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - Nico Boon
- grid.5342.00000 0001 2069 7798Centre for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | | | - Sarah Lebeer
- grid.5284.b0000 0001 0790 3681Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Wim Teughels
- grid.5596.f0000 0001 0668 7884Department of Oral Health Sciences, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
34
|
Tonelli A, Lumngwena EN, Ntusi NAB. The oral microbiome in the pathophysiology of cardiovascular disease. Nat Rev Cardiol 2023; 20:386-403. [PMID: 36624275 DOI: 10.1038/s41569-022-00825-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
Despite advances in our understanding of the pathophysiology of many cardiovascular diseases (CVDs) and expansion of available therapies, the global burden of CVD-associated morbidity and mortality remains unacceptably high. Important gaps remain in our understanding of the mechanisms of CVD and determinants of disease progression. In the past decade, much research has been conducted on the human microbiome and its potential role in modulating CVD. With the advent of high-throughput technologies and multiomics analyses, the complex and dynamic relationship between the microbiota, their 'theatre of activity' and the host is gradually being elucidated. The relationship between the gut microbiome and CVD is well established. Much less is known about the role of disruption (dysbiosis) of the oral microbiome; however, interest in the field is growing, as is the body of literature from basic science and animal and human investigations. In this Review, we examine the link between the oral microbiome and CVD, specifically coronary artery disease, stroke, peripheral artery disease, heart failure, infective endocarditis and rheumatic heart disease. We discuss the various mechanisms by which oral dysbiosis contributes to CVD pathogenesis and potential strategies for prevention and treatment.
Collapse
Affiliation(s)
- Andrea Tonelli
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, Department of Surgery, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa
| | - Evelyn N Lumngwena
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.,Centre for the Study of Emerging and Re-emerging Infections, Institute for Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa. .,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa. .,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Disease Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
35
|
Neculae E, Gosav EM, Valasciuc E, Dima N, Floria M, Tanase DM. The Oral Microbiota in Valvular Heart Disease: Current Knowledge and Future Directions. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010182. [PMID: 36676130 PMCID: PMC9862471 DOI: 10.3390/life13010182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Oral microbiota formation begins from birth, and everything from genetic components to the environment, alongside the host's behavior (such as diet, smoking, oral hygiene, and even physical activity), contributes to oral microbiota structure. Even though recent studies have focused on the gut microbiota's role in systemic diseases, the oral microbiome represents the second largest community of microorganisms, making it a new promising therapeutic target. Periodontitis and dental caries are considered the two main consequences of oral bacterial imbalance. Studies have shown that oral dysbiosis effects are not limited locally. Due to technological advancement, research identified oral bacterial species in heart valves. This evidence links oral dysbiosis with the development of valvular heart disease (VHD). This review focuses on describing the mechanism behind prolonged local inflammation and dysbiosis, that can induce bacteriemia by direct or immune-mediated mechanisms and finally VHD. Additionally, we highlight emerging therapies based on controlling oral dysbiosis, periodontal disease, and inflammation with immunological and systemic effects, that exert beneficial effects in VHD management.
Collapse
Affiliation(s)
- Ecaterina Neculae
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Correspondence:
| | - Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| |
Collapse
|
36
|
Effects of oral administration of Bifidobacterium animalis subsp. lactis HN019 on the treatment of plaque-induced generalized gingivitis. Clin Oral Investig 2023; 27:387-398. [PMID: 36305963 PMCID: PMC9614197 DOI: 10.1007/s00784-022-04744-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 10/02/2022] [Indexed: 01/28/2023]
Abstract
OBJECTIVES This double-blind, randomized, placebo-controlled clinical trial evaluated the adjuvant effects of Bifidobacterium lactis HN019 on the treatment of plaque-induced generalized gingivitis. MATERIALS AND METHODS Sixty patients were submitted to professional supragingival scaling and prophylaxis. They were randomly assigned to test (probiotic lozenges containing B. lactis HN019, n = 30) or control (placebo lozenges, n = 30) groups. Lozenges were consumed twice a day for 8 weeks. Bleeding on probing (BoP), Gingival Index (GI), Plaque Index (PI), probing depth (PD), and clinical attachment level (CAL) were evaluated at baseline and after 2 and 8 weeks. Gingival crevicular fluid (GCF) was collected at baseline and at 8 weeks for analysis of the inflammatory mediators IL-1β, IL-1α, IL-8, MCP-1, and MIP-1β. Data were statistically analyzed (p < 0.05). RESULTS After 8 weeks, both groups showed reduction in the percentage of PI, with no significant difference between groups (p = 0.7423). The test group presented a lower percentage of BoP and a higher percentage of sites with GI ≤ 1 when compared with the control group at the end of the study (p < 0.0001). At 8 weeks, the test group had a greater number of patients without generalized gingivitis than the control group (20 and 11 patients, respectively; p < 0.05). The test group presented significantly lower levels of IL-1α, IL-1β, and MCP-1 in GCF than the control group at the end of the study (p < 0.05). CONCLUSION The adjunct use of B. lactis HN019 promotes additional clinical and immunological benefits in the treatment of generalized gingivitis. CLINICAL RELEVANCE B. lactis HN019 can be an efficient and side-effect-free adjunct strategy in the treatment of generalized gingivitis.
Collapse
|
37
|
Choi YH, Kim BS, Kang SS. Inhibitory Effect of Genomic DNA Extracted from Pediococcus acidilactici on Porphyromonas gingivalis Lipopolysaccharide-Induced Inflammatory Responses. Food Sci Anim Resour 2023; 43:101-112. [PMID: 36789204 PMCID: PMC9890371 DOI: 10.5851/kosfa.2022.e62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
This study aimed to assess whether genomic DNA (gDNA) extracted from Pediococcus acidilactici inhibits Porphyromonas gingivalis lipopolysaccharide (LPS)-induced inflammatory responses in RAW 264.7 cells. Pretreatment with gDNA of P. acidilactici K10 or P. acidilactici HW01 for 15 h effectively inhibited P. gingivalis LPS-induced mRNA expression of interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein (MCP)-1. Although both gDNAs did not dose-dependently inhibit P. gingivalis LPS-induced mRNA expression of IL-6 and MCP-1, they inhibited IL-1β mRNA expression in a dose-dependent manner. Moreover, pretreatment with both gDNAs inhibited the secretion of IL-1β, IL-6, and MCP-1. When RAW 264.7 cells were stimulated with P. gingivalis LPS alone, the phosphorylation of mitogen-activated protein kinases (MAPKs) was increased. However, the phosphorylation of MAPKs was reduced in the presence of gDNAs. Furthermore, both gDNAs restored IκBα degradation induced by P. gingivalis LPS, indicating that both gDNAs suppressed the activation of nuclear factor-κB (NF-κB). In summary, P. acidilactici gDNA could inhibit P. gingivalis LPS-induced inflammatory responses through the suppression of MAPKs and NF-κB, suggesting that P. acidilactici gDNA could be effective in preventing periodontitis.
Collapse
Affiliation(s)
- Young Hyeon Choi
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| | - Bong Sun Kim
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| | - Seok-Seong Kang
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| |
Collapse
|
38
|
Colamarino AN, Johnson TM, Boudreaux DM, Dutner JM, Stancoven BW, Lincicum AR, Akers JA. Influence of Lactobacillus reuteri, Bifidobacterium animalis subsp. lactis, and prebiotic inulin on dysbiotic dental biofilm composition ex vivo. J Periodontol 2022. [PMID: 36542391 DOI: 10.1002/jper.22-0505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Probiotic bacterial supplementation has shown promising results in the treatment of periodontitis and the maintenance of periodontal health. The purpose of this investigation was to evaluate the influence of Lactobacillus reuteri or Bifidobacterium animalis subsp. lactis supplementation with and without prebiotic inulin on biofilm composition using an ex vivo biofilm model. METHODS Subgingival plaque specimens from three periodontitis-affected human donors were used to grow biofilms on hydroxyapatite disks in media supplemented with varying combinations of prebiotic inulin, Lactobacillus reuteri, and Bifidobacterium animalis subsp. lactis. Relative abundances of bacterial genera present in mature biofilms were evaluated using 16S rRNA next-generation sequencing. Diversity metrics of microbial communities were evaluated using a next-generation microbiome bioinformatics platform. RESULTS Inulin supplementation produced statistically significant dose-dependent increases in relative abundances of Lactobacillus and Bifidobacterium species (p < 0.001) with concomitant decreases in relative abundances of Streptococcus, Veillonella, Fusobacterium, Parvimonas, and Prevotella species (p < 0.001). Inoculation with L. reuteri or B. animalis subsp. lactis increased the relative abundance of only the supplemented probiotic genera (p < 0.05). Supplemental inulin led to a statistically significant decrease in biofilm alpha diversity (p < 0.001). CONCLUSIONS The described ex vivo model appears suitable for investigating the effects of probiotic bacteria, prebiotic oligosaccharides, and combinations thereof on biofilm composition and complexity. Within the limitations imposed by this model, results from the present study underscore the potential for prebiotic inulin to modify biofilm composition favorably. Additional research further elucidating biologic rationale and controlled clinical research defining therapeutic benefits is warranted.
Collapse
Affiliation(s)
- Aaron N Colamarino
- Department of Periodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| | - Thomas M Johnson
- Department of Periodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| | | | - Joseph M Dutner
- Department of Endodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| | - Brian W Stancoven
- Department of Periodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| | - Adam R Lincicum
- Department of Periodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| | - Joshua A Akers
- Department of Periodontics, Army Postgraduate Dental School, Uniformed Services University of the Health Sciences, Fort Gordon, Georgia, USA
| |
Collapse
|
39
|
Matsubara VH, Fakhruddin KS, Ngo H, Samaranayake LP. Probiotic Bifidobacteria in Managing Periodontal Disease: A Systematic Review. Int Dent J 2022; 73:11-20. [PMID: 36535806 PMCID: PMC9875235 DOI: 10.1016/j.identj.2022.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022] Open
Abstract
Although various probiotic organisms have been evaluated for their utility in the management of periodontitis, their strain-specific mechanisms of action are still unclear. We aimed to systematically review the effect of bifidobacterial probiotics on periodontopathogens and host immune responses in periodontal diseases. An electronic search of articles published until June 2022 in Medline, PubMed, Web of Science, and Cochrane Library databases was performed. Randomised controlled trials (RCTs) and in vitro and animal studies were assessed, and the data regarding antimicrobial properties, immunomodulation, and clinical outcomes were analysed. A total of 304 studies were screened, but only 3 RCTs and 6 animal and in vitro studies met the inclusion criteria. The use of different strains of bifidobacteria led to (1) a reduction of key players of the red complex periodontopathogens; (2) reduced levels of pro-inflammatory cytokines (eg, interleukin [IL]1-β and IL-8) and higher levels of anti-inflammatory cytokines (IL-10); (3) enhanced levels of osteoprotegerin and reduced levels of receptor activator of nuclear factor kappa-B ligand; and (4) a reduction of the dental plaque, bleeding on probing, alveolar bone loss, and clinical attachment loss. Bifidobacterial probiotic adjuvant supplementation, especially with Bifidobacterium animalis subspecies lactis, appears to help improve clinical periodontal parameters and develop a healthy plaque microbiome through microbiological and immunomodulatory pathways. Further human and animal studies are warranted prior to the therapeutic use of bifidobacteria in the routine management of periodontal infections.
Collapse
Affiliation(s)
- Victor Haruo Matsubara
- UWA Dental School, University of Western Australia, Perth, Western Australia, Australia,Corresponding author. Dental School, University of Western Australia, 17 Monash Avenue, Nedlands, Perth, WA 6009, Australia.
| | - Kausar Sadia Fakhruddin
- Department of Preventive and Restorative Dentistry, University of Sharjah, Sharjah, United Arab Emirates
| | - Hien Ngo
- UWA Dental School, University of Western Australia, Perth, Western Australia, Australia
| | - Lakshman P. Samaranayake
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| |
Collapse
|
40
|
Complete Genome Sequence of Lacticaseibacillus rhamnosus DM065, Isolated from the Human Oral Cavity. Microbiol Resour Announc 2022; 11:e0089922. [PMID: 36321910 PMCID: PMC9753688 DOI: 10.1128/mra.00899-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The 3.0-Mb complete genome of Lacticaseibacillus rhamnosus strain DM065, which was isolated from the oral cavity of healthy volunteers in South Korea, was sequenced using a combination of PacBio and Illumina technologies. The genome consists of one circular chromosome and two plasmids and lacks antimicrobial resistance genes.
Collapse
|
41
|
Polizzi A, Donzella M, Nicolosi G, Santonocito S, Pesce P, Isola G. Drugs for the Quorum Sensing Inhibition of Oral Biofilm: New Frontiers and Insights in the Treatment of Periodontitis. Pharmaceutics 2022; 14:2740. [PMID: 36559234 PMCID: PMC9781207 DOI: 10.3390/pharmaceutics14122740] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Chemical molecules are used by microorganisms to communicate with each other. Quorum sensing is the mechanism through which microorganisms regulate their population density and activity with chemical signaling. The inhibition of quorum sensing, called quorum quenching, may disrupt oral biofilm formation, which is the main etiological factor of oral diseases, including periodontitis. Periodontitis is a chronic inflammatory disorder of infectious etiology involving the hard and soft periodontal tissues and which is related to various systemic disorders, including cardiovascular diseases, diabetes and obesity. The employment of adjuvant therapies to traditional scaling and root planing is currently being studied to further reduce the impact of periodontitis. In this sense, using antibiotics and antiseptics involves non-negligible risks, such as antibiotic resistance phenomena and hinders the re-establishment of eubiosis. Different quorum sensing signal molecules have been identified in periodontal pathogenic oral bacteria. In this regard, quorum sensing inhibitors are emerging as some interesting solutions for the management of periodontitis. Therefore, the aim of this review is to summarize the current state of knowledge on the mechanisms of quorum sensing signal molecules produced by oral biofilm and to analyze the potential of quorum sensing inhibitors for the management of periodontitis.
Collapse
Affiliation(s)
- Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
- Department of Surgical Sciences (DISC), University of Genova, 16132 Genoa, Italy
| | - Martina Donzella
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Giada Nicolosi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| | - Paolo Pesce
- Department of Surgical Sciences (DISC), University of Genova, 16132 Genoa, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, Via Sofia 78, 95125 Catania, Italy
| |
Collapse
|
42
|
Functional biomaterials for comprehensive periodontitis therapy. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
43
|
Nannan M, Xiaoping L, Ying J. Periodontal disease in pregnancy and adverse pregnancy outcomes: Progress in related mechanisms and management strategies. Front Med (Lausanne) 2022; 9:963956. [PMID: 36388896 PMCID: PMC9640773 DOI: 10.3389/fmed.2022.963956] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/10/2022] [Indexed: 10/28/2023] Open
Abstract
Periodontal disease is an inflammatory and destructive disease of tissues supporting the tooth. A large number of studies have confirmed that periodontal pathogens and their metabolites can lead to adverse pregnancy outcomes in direct or indirect ways. Adverse pregnancy outcomes, such as preterm birth, low birth weight, and pre-eclampsia, have a serious impact on human reproductive health. In recent years, although the level of global medical technology has gradually improved, the incidence of adverse pregnancy outcomes has not declined and is still a global public health problem. The purpose of this review is to summarize the current data on periodontal disease in pregnancy and adverse pregnancy outcomes, including the association between periodontal disease and adverse pregnancy outcomes, the pathogenic mechanism related to this association, the efficacy of different nutrition supplements for both periodontal disease and adverse pregnancy outcomes and the effect of providing periodontal treatment on the occurrence of adverse pregnancy outcomes, to provide guidance for the prevention and treatment of adverse pregnancy outcomes in clinical practice.
Collapse
Affiliation(s)
| | | | - Jin Ying
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
44
|
Chen YL, Huang KC, Wu JH, Liu T, Chen JW, Xie JY, Chen MY, Wu LW, Tung CL. Microbiome dysbiosis inhibits carcinogen-induced murine oral tumorigenesis. J Cancer 2022; 13:3051-3060. [PMID: 36046649 PMCID: PMC9414028 DOI: 10.7150/jca.75947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/26/2022] [Indexed: 11/08/2022] Open
Abstract
Oral cancer is one of the most common cancers worldwide and ranks fourth for the mortality rate of cancers in males in Taiwan. The oral microbiota is the microbial community in the oral cavity, which is essential for maintaining oral health, but the relationship between oral tumorigenesis and the oral microbiota remains to be clarified. This study evaluated the effect of microbiome dysbiosis on oral carcinogenesis in mice, and the impact of the microbiome and its metabolic pathways on regulating oral carcinogenesis. We found that antibiotics treatment decreases carcinogen-induced oral epithelial malignant transformation. Microbiome analysis based on 16S rRNA gene sequencing revealed that the species richness of fecal specimens was significantly reduced in antibiotic-treated mice, while that in the salivary specimens was not decreased accordingly. Differences in bacterial composition, including Lactobacillus animalis abundance, in the salivary samples of cancer-bearing mice was dramatically decreased. L. animalis was the bacterial species that increased the most in the saliva of antibiotic-treated mice, suggesting that L. animalis may be negatively associated with oral carcinogenesis. In functional analysis, the microbiome in the saliva of the tumor-bearing group showed greater potential for polyamine biosynthesis. Immunochemical staining proved that spermine oxidase, an effective polyamine oxidase, was upregulated in mouse oral cancer lesions. In conclusion, oral microbiome dysbiosis may alter polyamine metabolic pathways and reduce carcinogen-induced malignant transformation of the oral epithelium.
Collapse
Affiliation(s)
- Yuh-Ling Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Chih Huang
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jer-Horng Wu
- Department of Environmental Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Tsunglin Liu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Jiung-Wen Chen
- Department of Environmental Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Yan Xie
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Yen Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Liang Tung
- Department of Oral Maxillo-Facial Surgery, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60080, Taiwan
| |
Collapse
|
45
|
Effects of extracellular vesicles derived from oral bacteria on osteoclast differentiation and activation. Sci Rep 2022; 12:14239. [PMID: 35987920 PMCID: PMC9396627 DOI: 10.1038/s41598-022-18412-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Dysbiosis of the oral microbiota plays an important role in the progression of periodontitis, which is characterized by chronic inflammation and alveolar bone loss, and associated with systemic diseases. Bacterial extracellular vesicles (EVs) contain various bioactive molecules and show diverse effects on host environments depending on the bacterial species. Recently, we reported that EVs derived from Filifactor alocis, a Gram-positive periodontal pathogen, had osteoclastogenic activity. In the present study, we analysed the osteoclastogenic potency and immunostimulatory activity of EVs derived from the Gram-negative periodontal pathogens Porphyromonas gingivalis and Tannerella forsythia, the oral commensal bacterium Streptococcus oralis, and the gut probiotic strain Lactobacillus reuteri. Bacterial EVs were purified by density gradient ultracentrifugation using OptiPrep (iodixanol) reagent. EVs from P. gingivalis, T. forsythia, and S. oralis increased osteoclast differentiation and osteoclstogenic cytokine expression in osteoclast precursors, whereas EVs from L. reuteri did not. EVs from P. gingivalis, T. forsythia, and S. oralis preferentially activated Toll-like receptor 2 (TLR2) rather than TLR4 or TLR9, and induced osteoclastogenesis mainly through TLR2. The osteoclastogenic effects of EVs from P. gingivalis and T. forsythia were reduced by both lipoprotein lipase and polymyxin B, an inhibitor of lipopolysaccharide (LPS), while the osteoclastogenic effects of EVs from S. oralis were reduced by lipoprotein lipase alone. These results demonstrate that EVs from periodontal pathogens and oral commensal have osteoclastogenic activity through TLR2 activation by lipoproteins and/or LPS.
Collapse
|
46
|
Use of the Probiotic Bifidobacterium animalis subsp. lactis HN019 in Oral Diseases. Int J Mol Sci 2022; 23:ijms23169334. [PMID: 36012597 PMCID: PMC9409207 DOI: 10.3390/ijms23169334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
The oral cavity is one of the environments on the human body with the highest concentrations of microorganisms that coexist harmoniously and maintain homeostasis related to oral health. Several local factors can shift the microbiome to a pathogenic state of dysbiosis. Existing treatments for infections caused by changes in the oral cavity aim to control biofilm dysbiosis and restore microbial balance. Studies have used probiotics as treatments for oral diseases, due to their ability to reduce the pathogenicity of the microbiota and immunoinflammatory changes. This review investigates the role of the probiotic Bifidobacterium animalis subsp. lactis (B. lactis) HN019 in oral health, and its mechanism of action in pre-clinical and clinical studies. This probiotic strain is a lactic acid bacterium that is safe for human consumption. It mediates bacterial co-aggregation with pathogens and modulates the immune response. Studies using B. lactis HN019 in periodontitis and peri-implant mucositis have shown it to be a potential adjuvant treatment with beneficial microbiological and immunological effects. Studies evaluating its oral effects and mechanism of action show that this probiotic strain has the potential to be used in several dental applications because of its benefit to the host.
Collapse
|
47
|
Sufaru IG, Teslaru S, Pasarin L, Iovan G, Stoleriu S, Solomon SM. Host Response Modulation Therapy in the Diabetes Mellitus—Periodontitis Conjuncture: A Narrative Review. Pharmaceutics 2022; 14:pharmaceutics14081728. [PMID: 36015357 PMCID: PMC9414216 DOI: 10.3390/pharmaceutics14081728] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022] Open
Abstract
The inflammatory response of the host in periodontitis is the phenomenon that underlies the onset and evolution of periodontal destructive phenomena. A number of systemic factors, such as diabetes mellitus (DM), can negatively affect the patient with periodontitis, just as the periodontal disease can aggravate the status of the DM patient. Host response modulation therapy involves the use of anti-inflammatory and anti-oxidant products aimed at resolving inflammation, stopping destructive processes, and promoting periodontal healing, all important aspects in patients with high tissue loss rates, such as diabetic patients. This paper reviews the data available in the literature on the relationship between DM and periodontitis, the main substances modulating the inflammatory response (nonsteroidal anti-inflammatory drugs, sub-antimicrobial doses of doxycycline, or omega-3 fatty acids and their products, specialized pro-resolving mediators), as well as their application in diabetic patients.
Collapse
Affiliation(s)
- Irina-Georgeta Sufaru
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Silvia Teslaru
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
- Correspondence: (S.T.); (L.P.)
| | - Liliana Pasarin
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
- Correspondence: (S.T.); (L.P.)
| | - Gianina Iovan
- Department of Cariology and Restorative Dental Therapy, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Simona Stoleriu
- Department of Cariology and Restorative Dental Therapy, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| | - Sorina Mihaela Solomon
- Department of Periodontology, Grigore T. Popa University of Medicine and Pharmacy, Universitatii Street 16, 700115 Iasi, Romania
| |
Collapse
|
48
|
Characteristics of Probiotic Preparations and Their Applications. Foods 2022; 11:foods11162472. [PMID: 36010472 PMCID: PMC9407510 DOI: 10.3390/foods11162472] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/03/2022] [Accepted: 08/09/2022] [Indexed: 12/17/2022] Open
Abstract
The probiotics market is one of the fastest growing segments of the food industry as there is growing scientific evidence of the positive health effects of probiotics on consumers. Currently, there are various forms of probiotic products and they can be categorized according to dosage form and the site of action. To increase the effectiveness of probiotic preparations, they need to be specifically designed so they can target different sites, such as the oral, upper respiratory or gastrointestinal tracts. Here we review the characteristics of different dosage forms of probiotics and discuss methods to improve their bioavailability in detail, in the hope that this article will provide a reference for the development of probiotic products.
Collapse
|
49
|
Radaic A, Brody H, Contreras F, Hajfathalian M, Lucido L, Kamarajan P, Kapila YL. Nisin and Nisin Probiotic Disrupt Oral Pathogenic Biofilms and Restore Their Microbiome Composition towards Healthy Control Levels in a Peri-Implantitis Setting. Microorganisms 2022; 10:1336. [PMID: 35889055 PMCID: PMC9324437 DOI: 10.3390/microorganisms10071336] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/06/2023] Open
Abstract
Peri-implantitis is characterized by chronic inflammation of the peri-implant supporting tissues that progressively and irreversibly leads to bone loss and, consequently, implant loss. Similar to periodontal disease, oral dysbiosis is thought to be a driver of peri-implantitis. However, managing peri-implantitis with traditional treatment methods, such as nonsurgical debridement or surgery, is not always successful. Thus, novel strategies have been proposed to address these shortcomings. One strategy is the use of probiotics as antimicrobial agents since they are considered safe for humans and the environment. Specifically, the probiotic Lactococcus lactis produces nisin, which has been used worldwide for food preservation. The objective of this study was to determine whether nisin and the wild-type (WT) nisin-producing L. lactis probiotic can disrupt oral pathogenic biofilms and promote a healthier oral microbiome within these oral biofilms on titanium discs. Using confocal imaging and 16S rRNA sequencing, this study revealed that nisin and WT L. lactis probiotic disrupt oral pathogenic biofilms in a peri-implantitis setting in vitro. More specifically, nisin decreased the viability of the pathogen-spiked biofilms dose-dependently from 62.53 ± 3.69% to 54.26 ± 3.35% and 44.88 ± 2.98%, respectively. Similarly, 105 CFU/mL of WT L. lactis significantly decreased biofilm viability to 52.45 ± 3.41%. Further, both treatments shift the composition, relative abundance, and diversity levels of these biofilms towards healthy control levels. A total of 1 µg/mL of nisin and 103 CFU/mL of WT L. lactis were able to revert the pathogen-mediated changes in the Proteobacteria (from 80.5 ± 2.9% to 75.6 ± 2.0%, 78.0 ± 2.8%, and 75.1 ± 5.3%, respectively) and Firmicutes (from 11.6 ± 1.6% to 15.4 ± 1.3%, 13.8 ± 1.8%, and 13.7 ± 2.6%, respectively) phyla back towards control levels. Thus, nisin and its nisin-producing L. lactis probiotic may be useful in treating peri-implantitis by promoting healthier oral biofilms, which may be useful for improving patient oral health.
Collapse
Affiliation(s)
- Allan Radaic
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Hanna Brody
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Fernando Contreras
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Maryam Hajfathalian
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luke Lucido
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
| | - Yvonne L. Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, CA 94143, USA; (A.R.); (H.B.); (F.C.); (M.H.); (L.L.); (P.K.)
- Division of Oral and Systemic Health Sciences, Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
50
|
Qin H, Li G, Xu X, Zhang C, Zhong W, Xu S, Yin Y, Song J. The role of oral microbiome in periodontitis under diabetes mellitus. J Oral Microbiol 2022; 14:2078031. [PMID: 35694215 PMCID: PMC9176325 DOI: 10.1080/20002297.2022.2078031] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Periodontitis is among most common human inflammatory diseases and characterized by destruction of tooth-supporting tissues that will eventually lead to tooth loss. Diabetes mellitus (DM) is a group of metabolic disorders characterized by chronic hyperglycemia which results from defects in insulin secretion and/or insulin resistance. Numerous studies have provided evidence for the inter-relationship between DM and periodontitis that has been considered as the sixth most frequent complication of DM. However, the mechanisms are not fully understood yet. The impact of DM on periodontitis through hyperglycemia and inflammatory pathways is well described, but the effects of DM on oral microbiota remain controversial according to previous studies. Recent studies using next-generation sequencing technology indicate that DM can alter the biodiversity and composition of oral microbiome especially subgingival microbiome. This may be another mechanism by which DM risks or aggravates periodontitis. Thus, to understand the role of oral microbiome in periodontitis of diabetics and the mechanism of shifts of oral microbiome under DM would be valuable for making specific therapeutic regimens for treating periodontitis patients with DM or preventing diabetic patients from periodontitis. This article reviews the role of oral microbiome in periodontal health (symbiosis) and disease (dysbiosis), highlights the oral microbial shifts under DM and summarizes the mechanism of the shifts.
Collapse
Affiliation(s)
- Han Qin
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Guangyue Li
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Xiaohui Xu
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Chuangwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Shihan Xu
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Yuanyuan Yin
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, Unknown, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, Unknown, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, Unknown, China
| |
Collapse
|