1
|
Mulvaney R, Pan Y, Zhao N, Teles F, Lu J, Platz EA, Kelsey KT, Michaud DS. Blood Leukocyte DNA Methylation Markers of Periodontal Disease and Risk of Lung Cancer in a Case-Control Study Nested in the CLUE II Cohort. Cancer Epidemiol Biomarkers Prev 2024; 33:1339-1346. [PMID: 39093033 PMCID: PMC11446649 DOI: 10.1158/1055-9965.epi-24-0279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/20/2024] [Accepted: 07/31/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Periodontal disease and DNA methylation markers have separately been associated with lung cancer risk. Examining methylation levels at genomic regions previously linked to periodontal disease may provide insights on the link between periodontal disease and lung cancer. METHODS In a nested case-control study drawn from the CLUE II cohort, we measured DNA methylation levels in 208 lung cancer cases and 208 controls. We examined the association between 37 DNA-methylated 5'-C-phosphate-G-3' (CpG) sites at three genomic regions, homeobox 4 (HOXA4), zinc finger protein (ZFP57), and a long noncoding RNA gene located in Chr10 (ENSG00000231601), and lung cancer risk. RESULTS Statistically significant associations with lung cancer risk were observed for all 14 CpG sites from HOXA4 (OR ranging 1.41-1.62 for 1 SD increase in the DNA methylation level, especially within 15 years) and 1 CpG site on gene ENSG00000231601 (OR = 1.34 for 1 SD increase in the DNA methylation level). Although CpG sites on gene ZFP57 were not associated with lung cancer risk overall, statistically significant inverse associations were noted for six CpG sites when restricting follow-up to 15 years (OR = 0.73-0.77 for 1 SD increase in the DNA methylation level). CONCLUSIONS Key methylation levels associated with periodontal disease are also associated with lung cancer risk. For both HOXA4 and ZFP57, the associations were stronger within 15 years of follow-up, which suggest that, if causal, the impact of methylation is acting late in the natural history of lung cancer. IMPACT Identifying biological pathways that link periodontal disease and lung cancer could provide new opportunities for lung cancer detection and prevention.
Collapse
Affiliation(s)
- Rachel Mulvaney
- Department of Epidemiology, Brown University, Providence, RI
| | - Yongyi Pan
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA
| | - Naisi Zhao
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA
| | - Flavia Teles
- Department of Basic & Translational Sciences, University of Pennsylvania, Philadelphia, PA
| | - Jiayun Lu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Karl T. Kelsey
- Department of Epidemiology, Brown University, Providence, RI
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI
| | - Dominique S. Michaud
- Department of Epidemiology, Brown University, Providence, RI
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Tufts University, Boston, MA
| |
Collapse
|
2
|
Hernandez Martinez CDJ, Glessner J, Finoti LS, Silva PF, Messora M, Coletta RD, Hakonarson H, Palioto DB. Methylome-wide analysis in systemic microbial-induced experimental periodontal disease in mice with different susceptibility. Front Cell Infect Microbiol 2024; 14:1369226. [PMID: 39086605 PMCID: PMC11289848 DOI: 10.3389/fcimb.2024.1369226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Objective The study delved into the epigenetic factors associated with periodontal disease in two lineages of mice, namely C57bl/6 and Balb/c. Its primary objective was to elucidate alterations in the methylome of mice with distinct genetic backgrounds following systemic microbial challenge, employing high-throughput DNA methylation analysis as the investigative tool. Methods Porphyromonas gingivalis (Pg)was orally administered to induce periodontitis in both Balb/c and C57bl/6 lineage. After euthanasia, genomic DNA from both maxilla and blood were subjected to bisulfite conversion, PCR amplification and genome-wide DNA methylation analysis using the Ovation RRBS Methyl-Seq System coupled with the Illumina Infinium Mouse Methylation BeadChip. Results Of particular significance was the distinct methylation profile observed within the Pg-induced group of the Balb/c lineage, contrasting with both the control and Pg-induced groups of the C57bl/6 lineage. Utilizing rigorous filtering criteria, we successfully identified a substantial number of differentially methylated regions (DMRs) across various tissues and comparison groups, shedding light on the prevailing hypermethylation in non-induced cohorts and hypomethylation in induced groups. The comparison between blood and maxilla samples underscored the unique methylation patterns specific to the jaw tissue. Our comprehensive methylome analysis further unveiled statistically significant disparities, particularly within promoter regions, in several comparison groups. Conclusion The differential DNA methylation patterns observed between C57bl/6 and Balb/c mouse lines suggest that epigenetic factors contribute to the variations in disease susceptibility. The identified differentially methylated regions associated with immune regulation and inflammatory response provide potential targets for further investigation. These findings emphasize the importance of considering epigenetic mechanisms in the development and progression of periodontitis.
Collapse
Affiliation(s)
- Cristhiam de Jesus Hernandez Martinez
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Joseph Glessner
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Livia Sertori Finoti
- Laboratory of Rebecca Ahrens-Nicklas,Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Pedro Felix Silva
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Michel Messora
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| | - Ricardo Della Coletta
- Department of Oral Diagnosis and Graduate Program in Oral Biology, Piracicaba Dental School, University of Campinas, Piracicaba, Brazil
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Division of Pulmonary Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Daniela Bazan Palioto
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental School, University of São Paulo - USP, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
3
|
Altamura S, Del Pinto R, Pietropaoli D, Ferri C. Oral health as a modifiable risk factor for cardiovascular diseases. Trends Cardiovasc Med 2024; 34:267-275. [PMID: 36963476 PMCID: PMC10517086 DOI: 10.1016/j.tcm.2023.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/26/2023]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality worldwide with a high socioeconomic burden. Increasing evidence supports a convincing connection with increased cardiovascular risk of periodontal diseases (PD), a group of widespread, debilitating, and costly dysbiotic relapsing-remitting inflammatory diseases of the tissues supporting the teeth. Herein, we ensembled the best available evidence on the connection between CVDs and PD to review the recently emerging concept of the latter as a non-traditional risk factor for CVDs. We focused on oral dysbiosis, inflammation-associated molecular and cellular mechanisms, and epigenetic changes as potential causative links between PD and CVDs. The available evidence on the effects of periodontal treatment on cardiovascular risk factors and diseases was also described.
Collapse
Affiliation(s)
- Serena Altamura
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Italy; Center of Oral Diseases, Prevention and Translational Research - Dental Clinic, L'Aquila, Italy; Oral Diseases and Systemic Interactions Study Group (ODISSY Group), L'Aquila, Italy; PhD School in Medicine and Public Health
| | - Rita Del Pinto
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Italy; Oral Diseases and Systemic Interactions Study Group (ODISSY Group), L'Aquila, Italy; Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, L'Aquila, Italy
| | - Davide Pietropaoli
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Italy; Center of Oral Diseases, Prevention and Translational Research - Dental Clinic, L'Aquila, Italy; Oral Diseases and Systemic Interactions Study Group (ODISSY Group), L'Aquila, Italy.
| | - Claudio Ferri
- Department of Life, Health & Environmental Sciences, University of L'Aquila, Italy; Oral Diseases and Systemic Interactions Study Group (ODISSY Group), L'Aquila, Italy; Unit of Internal Medicine and Nephrology, Center for Hypertension and Cardiovascular Prevention, San Salvatore Hospital, L'Aquila, Italy
| |
Collapse
|
4
|
Carneiro BT, de Castro FNAM, Benetti F, Nima G, Suzuki TYU, André CB. Flavonoids effects against bacteria associated to periodontal disease and dental caries: a scoping review. BIOFOULING 2024; 40:99-113. [PMID: 38425046 DOI: 10.1080/08927014.2024.2321965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
This scoping review focused on exploring the efficacy of flavonoids against bacteria associated with dental caries and periodontal diseases. Inclusion criteria comprise studies investigating the antibacterial effects of flavonoids against bacteria linked to caries or periodontal diseases, both pure or diluted in vehicle forms. The search, conducted in August 2023, in databases including PubMed/MEDLINE, Scopus, Web of Science, Embase, LILACS, and Gray Literature. Out of the initial 1125 studies, 79 met the inclusion criteria, majority in vitro studies. Prominent flavonoids tested included epigallocatechin-gallate, apigenin, quercetin, and myricetin. Predominant findings consistently pointed to bacteriostatic, bactericidal, and antibiofilm activities. The study primarily investigated bacteria associated with dental caries, followed by periodontopathogens. A higher number of publications presented positive antibacterial results against Streptococcus mutans in comparison to Porphyromonas gingivalis. These encouraging findings underline the potential applicability of commercially available flavonoids in materials or therapies, underscoring the need for further exploration in this field.
Collapse
Affiliation(s)
- Bruna Tavares Carneiro
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Francine Benetti
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Gabriel Nima
- Departament of Biomaterials, School of Dentistry, Universidad de los Andes, Santiago, Chile
| | - Thais Yumi Umeda Suzuki
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carolina Bosso André
- Departament of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
5
|
Anestino TA, Queiroz-Junior CM, Cruz AMF, Souza DG, Madeira MFM. The impact of arthritogenic viruses in oral tissues. J Appl Microbiol 2024; 135:lxae029. [PMID: 38323434 DOI: 10.1093/jambio/lxae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/14/2023] [Accepted: 02/05/2024] [Indexed: 02/08/2024]
Abstract
Arthritis and periodontitis are inflammatory diseases that share several immunopathogenic features. The expansion in the study of virus-induced arthritis has shed light on how this condition could impact other parts of the human body, including the mouth. Viral arthritis is an inflammatory joint disease caused by several viruses, most notably the alphaviruses Chikungunya virus (CHIKV), Sindbis virus (SINV), Ross River virus (RRV), Mayaro virus (MAYV), and O'nyong'nyong virus (ONNV). These viruses can induce an upsurge of matrix metalloproteinases and immune-inflammatory mediators such as Interleukin-6 (IL6), IL-1β, tumor necrosis factor, chemokine ligand 2, and receptor activator of nuclear factor kappa-B ligand in the joint and serum of infected individuals. This can lead to the influx of inflammatory cells to the joints and associated muscles as well as osteoclast activation and differentiation, culminating in clinical signs of swelling, pain, and bone resorption. Moreover, several data indicate that these viral infections can affect other sites of the body, including the mouth. The human oral cavity is a rich and diverse microbial ecosystem, and viral infection can disrupt the balance of microbial species, causing local dysbiosis. Such events can result in oral mucosal damage and gingival bleeding, which are indicative of periodontitis. Additionally, infection by RRV, CHIKV, SINV, MAYV, or ONNV can trigger the formation of osteoclasts and upregulate pro-osteoclastogenic inflammatory mediators, interfering with osteoclast activation. As a result, these viruses may be linked to systemic conditions, including oral manifestations. Therefore, this review focuses on the involvement of alphavirus infections in joint and oral health, acting as potential agents associated with oral mucosal inflammation and alveolar bone loss. The findings of this review demonstrate how alphavirus infections could be linked to the comorbidity between arthritis and periodontitis and may provide a better understanding of potential therapeutic management for both conditions.
Collapse
Affiliation(s)
- Thales Augusto Anestino
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP: 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP: 31270-901, Brazil
| | - Amanda Medeiros Frota Cruz
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP: 31270-901, Brazil
| | - Daniele G Souza
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP: 31270-901, Brazil
| | - Mila Fernandes Moreira Madeira
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, CEP: 31270-901, Brazil
- Department of Oral Biology, Biomedical Research Institute, University at Buffalo, Buffalo, NY, 14203, United States
| |
Collapse
|
6
|
Barczak K, Droździk A, Bosiacki M, Łagocka R, Cenariu D, Uriciuc WA, Baranowska-Bosiacka I. CCL5's Role in Periodontal Disease: A Narrative Review. Int J Mol Sci 2023; 24:17332. [PMID: 38139161 PMCID: PMC10744061 DOI: 10.3390/ijms242417332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Persistent host inflammatory and immune responses to biofilm play a critical role in the mechanisms that govern soft and hard tissue destruction in periodontal disease. Among the less explored facets of these mechanisms are chemokines, including CCL5 (C-C motif chemokine ligand 5), also known as RANTES (regulated on activation, normal T cell expressed and secreted), a proinflammatory CC subfamily chemokine synthesized by T lymphocytes. Despite its importance, there is currently no comprehensive review of the role of CCL5 in periodontitis in the literature. Therefore, this paper aims to fill this gap by summarizing the existing knowledge on the involvement of CCL5 in the onset and progression of periodontitis. In addition, we aim to stimulate interest in this relatively overlooked factor among periodontitis researchers, potentially accelerating the development of drugs targeting CCL5 or its receptors. The review examines the association of CCL5 with periodontitis risk factors, including aging, cigarette smoking, diabetes, and obesity. It discusses the involvement of CCL5 in pathological processes during periodontitis, such as connective tissue and bone destruction. The data show that CCL5 expression is observed in affected gums and gingival crevicular fluid of periodontitis patients, with bacterial activity contributing significantly to this increase, but the reviewed studies of the association between CCL5 expression and periodontal disease have yielded inconclusive results. Although CCL5 has been implicated in the pathomechanism of periodontitis, a comprehensive understanding of its molecular mechanisms and significance remains elusive, hindering the development of drugs targeting this chemokine or its receptors.
Collapse
Affiliation(s)
- Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Agnieszka Droździk
- Laboratory of Preclinical Periodontology, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (I.B.-B.)
| | - Ryta Łagocka
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Diana Cenariu
- MEDFUTURE—Research Center for Advanced Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania;
| | - Willi Andrei Uriciuc
- Faculty of Dental Medicine, “Iuliu-Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania;
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.); (I.B.-B.)
| |
Collapse
|
7
|
Hernández Martínez CDJ, Felix Silva P, Salvador SL, Messora M, Palioto DB. Chronological analysis of periodontal bone loss in experimental periodontitis in mice. Clin Exp Dent Res 2023; 9:1009-1020. [PMID: 37997536 PMCID: PMC10728515 DOI: 10.1002/cre2.806] [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: 03/12/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023] Open
Abstract
OBJECTIVES Periodontal disease is understood to be a result of dysbiotic interactions between the host and the biofilm, causing a unique reaction for each individual, which in turn characterizes their susceptibility. The objective of this study was to chronologically evaluate periodontal tissue destruction induced by systemic bacterial challenge in known susceptible (BALB/c) and resistant (C57BL/6) mouse lineages. MATERIAL AND METHODS Animals, 6-8 weeks old, were allocated into three experimental groups: Negative control (C), Gavage with sterile carboxymethyl cellulose 2%-without bacteria (Sham), and Gavage with carboxymethyl cellulose 2% + Porphyromonas gingivalis (Pg-W83). Before infection, all animals received antibiotic treatment (sulfamethoxazole/trimethoprim, 400/80 mg/5 mL) for 7 days, followed by 3 days of rest. Microbial challenge was performed 3 times per week for 1, 2, or 3 weeks. After that, the animals were kept until the completion of 42 days of experiments, when they were euthanized. The alveolar bone microarchitecture was assessed by computed microtomography. RESULTS Both C57BL/6 and BALB/c mice exhibited significant bone volume loss and lower trabecular thickness as well as greater bone porosity compared to the (C) and (Sham) groups after 1 week of microbial challenge (p < .001). When comparing only the gavage groups regarding disease implantation, time and lineage, it was possible to observe that within 1 week of induction the disease was more established in BALB/c than in C57BL/6 (p < .05). CONCLUSIONS Our results reflected that after 1 week of microbial challenge, there was evidence of alveolar bone loss for both lineages, with the loss observed in BALB/c mice being more pronounced.
Collapse
Affiliation(s)
- Cristhiam de J. Hernández Martínez
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental SchoolUniversity of Sao Paulo—USPRibeirão Preto SPBrazil
| | - Pedro Felix Silva
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental SchoolUniversity of Sao Paulo—USPRibeirão Preto SPBrazil
| | - Sergio L. Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirao PretoUniversity of Sao Paulo—USPRibeirão Preto SPBrazil
| | - Michel Messora
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental SchoolUniversity of Sao Paulo—USPRibeirão Preto SPBrazil
| | - Daniela B. Palioto
- Department of Oral & Maxillofacial Surgery and Periodontology, Ribeirão Preto Dental SchoolUniversity of Sao Paulo—USPRibeirão Preto SPBrazil
| |
Collapse
|
8
|
Pacheco-Yanes J, Reynolds E, Li J, Mariño E. Microbiome-targeted interventions for the control of oral-gut dysbiosis and chronic systemic inflammation. Trends Mol Med 2023; 29:912-925. [PMID: 37730461 DOI: 10.1016/j.molmed.2023.08.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/22/2023]
Abstract
Recent research has confirmed the strong connection between imbalances in the oral and gut microbiome (oral-gut dysbiosis), periodontitis, and inflammatory conditions such as diabetes, Alzheimer's disease, and cardiovascular diseases. Microbiome modulation is crucial for preventing and treating several autoimmune and inflammatory diseases, including periodontitis. However, the causal relationships between the microbiome and its derived metabolites that mediate periodontitis and chronic inflammation constitute a notable knowledge gap. Here we review the mechanisms involved in the microbiome-host crosstalk, and describe novel precision medicine for the control of systemic inflammation. As microbiome-targeted therapies begin to enter clinical trials, the success of these approaches relies upon understanding these reciprocal microbiome-host interactions, and it may provide new therapeutic avenues to reduce the risk of periodontitis-associated diseases.
Collapse
Affiliation(s)
- Juan Pacheco-Yanes
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Eric Reynolds
- Oral Health Collaborative Research Centre, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Eliana Mariño
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; ImmunoBiota Therapeutics Pty Ltd, Melbourne, Australia.
| |
Collapse
|
9
|
Li S, Zeng W, Liu G, Zang J, Yu X. Evaluation of morphological, histological, and immune-related cellular changes in ligature-induced experimental periodontitis in mice. J Dent Sci 2023; 18:1716-1722. [PMID: 37799858 PMCID: PMC10547956 DOI: 10.1016/j.jds.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/02/2023] [Indexed: 01/13/2023] Open
Abstract
Background/purpose The ligature-induced periodontitis model is an effective approach to induce inflammation and bone loss similar to that of human periodontitis. Previous clinical and in vitro studies have shown the involvement of lymphocytes in periodontitis, while, the local and systemic profile of immune cells associated with periodontitis in the ligature-induced periodontitis model in mice remains unclear. Materials and methods Experimental periodontitis was constructed in mice by ligating around the maxillary second molars for 14 and 28 days, respectively. Alveolar bone loss was assessed by micro-computed tomography (micro-CT). Hematoxylin and eosin (H&E) and tartrate-resistant acid phosphatase (TRAP) staining were used to evaluate the histological changes in the periodontal tissues. B and T cells in the cervical lymph nodes, spleen, and peripheral blood were analyzed by flow cytometry. Results The 14-day ligation effectively induced significant periodontal inflammation and alveolar bone loss in C57BL/6J mice, which were progressive and maintained for a relatively long-term period until day 28. In addition, CD3+ T cells and CD19+ B cells were the dominant population in both health and disease, and the B cell population within the cervical lymph nodes (LN) increased significantly under periodontitis condition, while, no significant differences of the T and B cell population among the spleen and peripheral blood were observed. Conclusion The ligature-induced periodontitis mice model was established to perform a longitudinal assessment of changes in periodontal tissues morphologically and histologically, meanwhile, explore the local and systemic changes of the predominant immune-associated cells.
Collapse
Affiliation(s)
- Shiyi Li
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Wenmin Zeng
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Guojing Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Jing Zang
- Department of Periodontology, Peking University Third Hospital, Beijing, China
| | - Xiaoqian Yu
- Department of Periodontology, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
10
|
Baima G, Ribaldone DG, Romano F, Aimetti M, Romandini M. The Gum-Gut Axis: Periodontitis and the Risk of Gastrointestinal Cancers. Cancers (Basel) 2023; 15:4594. [PMID: 37760563 PMCID: PMC10526746 DOI: 10.3390/cancers15184594] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Periodontitis has been linked to an increased risk of various chronic non-communicable diseases, including gastrointestinal cancers. Indeed, dysbiosis of the oral microbiome and immune-inflammatory pathways related to periodontitis may impact the pathophysiology of the gastrointestinal tract and its accessory organs through the so-called "gum-gut axis". In addition to the hematogenous spread of periodontal pathogens and inflammatory cytokines, recent research suggests that oral pathobionts may translocate to the gastrointestinal tract through saliva, possibly impacting neoplastic processes in the gastrointestinal, liver, and pancreatic systems. The exact mechanisms by which oral pathogens contribute to the development of digestive tract cancers are not fully understood but may involve dysbiosis of the gut microbiome, chronic inflammation, and immune modulation/evasion, mainly through the interaction with T-helper and monocytic cells. Specifically, keystone periodontal pathogens, including Porphyromonas gingivalis and Fusobacterium nucleatum, are known to interact with the molecular hallmarks of gastrointestinal cancers, inducing genomic mutations, and promote a permissive immune microenvironment by impairing anti-tumor checkpoints. The evidence gathered here suggests a possible role of periodontitis and oral dysbiosis in the carcinogenesis of the enteral tract. The "gum-gut axis" may therefore represent a promising target for the development of strategies for the prevention and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Giacomo Baima
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | | | - Federica Romano
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Aimetti
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0313 Oslo, Norway
| |
Collapse
|
11
|
Laberge S, Akoum D, Wlodarczyk P, Massé JD, Fournier D, Semlali A. The Potential Role of Epigenetic Modifications on Different Facets in the Periodontal Pathogenesis. Genes (Basel) 2023; 14:1202. [PMID: 37372382 DOI: 10.3390/genes14061202] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Periodontitis is a chronic inflammatory disease that affects the supporting structures of teeth. In the literature, the association between the pathogenicity of bacteria and environmental factors in this regard have been extensively examined. In the present study, we will shed light on the potential role that epigenetic change can play on different facets of its process, more particularly the modifications concerning the genes involved in inflammation, defense, and immune systems. Since the 1960s, the role of genetic variants in the onset and severity of periodontal disease has been widely demonstrated. These make some people more susceptible to developing it than others. It has been documented that the wide variation in its frequency for various racial and ethnic populations is due primarily to the complex interplay among genetic factors with those affecting the environment and the demography. In molecular biology, epigenetic modifications are defined as any change in the promoter for the CpG islands, in the structure of the histone protein, as well as post-translational regulation by microRNAs (miRNAs), being known to contribute to the alteration in gene expression for complex multifactorial diseases such as periodontitis. The key role of epigenetic modification is to understand the mechanism involved in the gene-environment interaction, and the development of periodontitis is now the subject of more and more studies that attempt to identify which factors are stimulating it, but also affect the reduced response to therapy.
Collapse
Affiliation(s)
- Samuel Laberge
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Daniel Akoum
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Piotr Wlodarczyk
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Jean-Daniel Massé
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | | | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
12
|
Suzuki S, Yamada S. Epigenetics in susceptibility, progression, and diagnosis of periodontitis. JAPANESE DENTAL SCIENCE REVIEW 2022; 58:183-192. [PMID: 35754944 PMCID: PMC9218144 DOI: 10.1016/j.jdsr.2022.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/14/2022] [Accepted: 06/01/2022] [Indexed: 12/12/2022] Open
Abstract
Periodontitis is characterized by irreversible destruction of periodontal tissue. At present, the accepted etiology of periodontitis is based on a three-factor theory including pathogenic bacteria, host factors, and acquired factors. Periodontitis development usually takes a decade or longer and is therefore called chronic periodontitis (CP). To search for genetic factors associated with CP, several genome-wide association study (GWAS) analyses were conducted; however, polymorphisms associated with CP have not been identified. Epigenetics, on the other hand, involves acquired transcriptional regulatory mechanisms due to reversibly altered chromatin accessibility. Epigenetic status is a condition specific to each tissue and cell, mostly determined by the responses of host cells to stimulations by local factors, like bacterial inflammation, and systemic factors such as nutrition status, metabolic diseases, and health conditions. Significantly, epigenetic status has been linked with the onset and progression of several acquired diseases. Thus, epigenetic factors in periodontal tissues are attractive targets for periodontitis diagnosis and treatments. In this review, we introduce accumulating evidence to reveal the epigenetic background effects related to periodontitis caused by genetic factors, systemic diseases, and local environmental factors, such as smoking, and clarify the underlying mechanisms by which epigenetic alteration influences the susceptibility of periodontitis.
Collapse
Key Words
- 5mC, 5-methylcytocine
- AP, aggressive periodontitis
- ATAC-seq, assay for transposase-accessible chromatin sequencing
- CP, chronic periodontitis
- DNA methylation
- ECM, extracellular matrix
- Epigenetics
- Epigenome
- GWAS, genome-wide association study
- H3K27ac, acetylation of histone H3 lysine 27
- H3K27me3, trimethylation of histone H3 lysine 27
- H3K4me3, trimethylation of histone H3 lysine 4
- H3K9ac, histone H3 lysine 9
- HATs, histone acetyltransferases
- HDACs, histone deacetylases
- Histone modifications
- LPS, lipopolysaccharide
- PDL, periodontal ligament
- Periodontal ligament
- Periodontitis
- ceRNA, competing endogenous RNA
- lncRNAs, long ncRNAs
- m6A, N6-methyladenosine
- ncRNAs, non-coding RNAs
- sEV, small extracellular vesicles
Collapse
Affiliation(s)
- Shigeki Suzuki
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Satoru Yamada
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| |
Collapse
|
13
|
Epigenetic Regulation of Methylation in Determining the Fate of Dental Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:5015856. [PMID: 36187229 PMCID: PMC9522499 DOI: 10.1155/2022/5015856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Dental mesenchymal stem cells (DMSCs) are crucial in tooth development and periodontal health, and their multipotential differentiation and self-renewal ability play a critical role in tissue engineering and regenerative medicine. Methylation modifications could promote the appropriate biological behavior by postsynthetic modification of DNA or protein and make the organism adapt to developmental and environmental prompts by regulating gene expression without changing the DNA sequence. Methylation modifications involved in DMSC fate include DNA methylation, RNA methylation, and histone modifications, which have been proven to exert a significant effect on the regulation of the fate of DMSCs, such as proliferation, self-renewal, and differentiation potential. Understanding the regulation of methylation modifications on the behavior and the immunoinflammatory responses involved in DMSCs contributes to further study of the mechanism of methylation on tissue regeneration and inflammation. In this review, we briefly summarize the key functions of histone methylation, RNA methylation, and DNA methylation in the differentiation potential and self-renewal of DMSCs as well as the opportunities and challenges for their application in tissue regeneration and disease therapy.
Collapse
|
14
|
Local and systemic effects produced in different models of experimental periodontitis in mice: a systematic review. Arch Oral Biol 2022; 143:105528. [DOI: 10.1016/j.archoralbio.2022.105528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 12/09/2022]
|
15
|
Starzyńska A, Wychowański P, Nowak M, Sobocki BK, Jereczek-Fossa BA, Słupecka-Ziemilska M. Association between Maternal Periodontitis and Development of Systematic Diseases in Offspring. Int J Mol Sci 2022; 23:2473. [PMID: 35269617 PMCID: PMC8910384 DOI: 10.3390/ijms23052473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/18/2022] [Accepted: 02/20/2022] [Indexed: 12/24/2022] Open
Abstract
Periodontal disease (PD) is one of the most common oral conditions affecting both youths and adults. There are some research works suggesting a high incidence of PD in pregnant women. As an inflammatory disease of bacterial origin, PD may result in the activation of the pathways affecting the course and the pregnancy outcome. The authors, based on the literature review, try to answer the PICO question: Does maternal periodontitis (exposure) influence the incidence of complications rates in pregnancy and the development of systemic diseases in childhood and adult offspring (outcome) in the humans of any race (population) compared to the offspring of mothers with healthy periodontium (comparison)? The authors try to describe the molecular pathways and mechanisms of these interdependencies. There is some evidence that maternal periodontitis may affect the pregnancy course and outcome, resulting in preeclampsia, preterm delivery, vulvovaginitis and low birth weight. It can be suggested that maternal periodontitis may affect offspring epigenome and result in some health consequences in their adult life.
Collapse
Affiliation(s)
- Anna Starzyńska
- Department of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland;
| | - Piotr Wychowański
- Department of Oral Surgery, Medical University of Warsaw, 6 Binieckiego Street, 02-097 Warsaw, Poland;
- Specialized Private Implantology Clinic Wychowanski Stomatologia, 9/33 Rakowiecka Street, 02-517 Warsaw, Poland
| | - Maciej Nowak
- Department of Periodontology and Oral Diseases, Medical University of Warsaw, 6 Binieckiego Street, 02-097 Warsaw, Poland;
| | - Bartosz Kamil Sobocki
- Department of Oral Surgery, Medical University of Gdańsk, 7 Dębinki Street, 80-211 Gdańsk, Poland;
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, 7 Festa del Perdono Street, 20-112 Milan, Italy;
- Division of Radiotherapy, IEO European Institute of Oncology, IRCCS, 435 Ripamonti Street, 20-141 Milan, Italy
| | - Monika Słupecka-Ziemilska
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 5 Pawińskiego Street, 02-106 Warsaw, Poland;
| |
Collapse
|
16
|
Sansores-España LD, Melgar-Rodríguez S, Olivares-Sagredo K, Cafferata EA, Martínez-Aguilar VM, Vernal R, Paula-Lima AC, Díaz-Zúñiga J. Oral-Gut-Brain Axis in Experimental Models of Periodontitis: Associating Gut Dysbiosis With Neurodegenerative Diseases. FRONTIERS IN AGING 2021; 2:781582. [PMID: 35822001 PMCID: PMC9261337 DOI: 10.3389/fragi.2021.781582] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
Periodontitis is considered a non-communicable chronic disease caused by a dysbiotic microbiota, which generates a low-grade systemic inflammation that chronically damages the organism. Several studies have associated periodontitis with other chronic non-communicable diseases, such as cardiovascular or neurodegenerative diseases. Besides, the oral bacteria considered a keystone pathogen, Porphyromonas gingivalis, has been detected in the hippocampus and brain cortex. Likewise, gut microbiota dysbiosis triggers a low-grade systemic inflammation, which also favors the risk for both cardiovascular and neurodegenerative diseases. Recently, the existence of an axis of Oral-Gut communication has been proposed, whose possible involvement in the development of neurodegenerative diseases has not been uncovered yet. The present review aims to compile evidence that the dysbiosis of the oral microbiota triggers changes in the gut microbiota, which creates a higher predisposition for the development of neuroinflammatory or neurodegenerative diseases.The Oral-Gut-Brain axis could be defined based on anatomical communications, where the mouth and the intestine are in constant communication. The oral-brain axis is mainly established from the trigeminal nerve and the gut-brain axis from the vagus nerve. The oral-gut communication is defined from an anatomical relation and the constant swallowing of oral bacteria. The gut-brain communication is more complex and due to bacteria-cells, immune and nervous system interactions. Thus, the gut-brain and oral-brain axis are in a bi-directional relationship. Through the qualitative analysis of the selected papers, we conclude that experimental periodontitis could produce both neurodegenerative pathologies and intestinal dysbiosis, and that periodontitis is likely to induce both conditions simultaneously. The severity of the neurodegenerative disease could depend, at least in part, on the effects of periodontitis in the gut microbiota, which could strengthen the immune response and create an injurious inflammatory and dysbiotic cycle. Thus, dementias would have their onset in dysbiotic phenomena that affect the oral cavity or the intestine. The selected studies allow us to speculate that oral-gut-brain communication exists, and bacteria probably get to the brain via trigeminal and vagus nerves.
Collapse
Affiliation(s)
- Luis Daniel Sansores-España
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
- Faculty of Dentistry, Autonomous University of Yucatán, Mérida, México
| | | | | | - Emilio A. Cafferata
- Department of Periodontology, School of Dentistry, Universidad Científica Del Sur, Lima, Perú
| | | | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Andrea Cristina Paula-Lima
- Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
- Institute for Research in Dental Sciences, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Jaime Díaz-Zúñiga
- Periodontal Biology Laboratory, Faculty of Dentistry, University of Chile, Santiago, Chile
- Department of Medicine, Faculty of Medicine, University of Atacama, Copiapó, Chile
- *Correspondence: Jaime Díaz-Zúñiga, ,
| |
Collapse
|
17
|
Jia X, Yang R, Li J, Zhao L, Zhou X, Xu X. Gut-Bone Axis: A Non-Negligible Contributor to Periodontitis. Front Cell Infect Microbiol 2021; 11:752708. [PMID: 34869062 PMCID: PMC8637199 DOI: 10.3389/fcimb.2021.752708] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/26/2021] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is a polymicrobial infectious disease characterized by alveolar bone loss. Systemic diseases or local infections, such as diabetes, postmenopausal osteoporosis, obesity, and inflammatory bowel disease, promote the development and progression of periodontitis. Accumulating evidences have revealed the pivotal effects of gut microbiota on bone health via gut-alveolar-bone axis. Gut pathogens or metabolites may translocate to distant alveolar bone via circulation and regulate bone homeostasis. In addition, gut pathogens can induce aberrant gut immune responses and subsequent homing of immunocytes to distant organs, contributing to pathological bone loss. Gut microbial translocation also enhances systemic inflammation and induces trained myelopoiesis in the bone marrow, which potentially aggravates periodontitis. Furthermore, gut microbiota possibly affects bone health via regulating the production of hormone or hormone-like substances. In this review, we discussed the links between gut microbiota and periodontitis, with a particular focus on the underlying mechanisms of gut-bone axis by which systemic diseases or local infections contribute to the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Xiaoyue Jia
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ran Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Martínez-García M, Hernández-Lemus E. Periodontal Inflammation and Systemic Diseases: An Overview. Front Physiol 2021; 12:709438. [PMID: 34776994 PMCID: PMC8578868 DOI: 10.3389/fphys.2021.709438] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Periodontitis is a common inflammatory disease of infectious origins that often evolves into a chronic condition. Aside from its importance as a stomatologic ailment, chronic periodontitis has gained relevance since it has been shown that it can develop into a systemic condition characterized by unresolved hyper-inflammation, disruption of the innate and adaptive immune system, dysbiosis of the oral, gut and other location's microbiota and other system-wide alterations that may cause, coexist or aggravate other health issues associated to elevated morbi-mortality. The relationships between the infectious, immune, inflammatory, and systemic features of periodontitis and its many related diseases are far from being fully understood and are indeed still debated. However, to date, a large body of evidence on the different biological, clinical, and policy-enabling sources of information, is available. The aim of the present work is to summarize many of these sources of information and contextualize them under a systemic inflammation framework that may set the basis to an integral vision, useful for basic, clinical, and therapeutic goals.
Collapse
Affiliation(s)
- Mireya Martínez-García
- Sociomedical Research Unit, National Institute of Cardiology "Ignacio Chávez", Mexico City, Mexico
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine (INMEGEN), Mexico City, Mexico.,Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de Mèxico, Mexico City, Mexico
| |
Collapse
|
19
|
Haas AN, Furlaneto F, Gaio EJ, Gomes SC, Palioto DB, Castilho RM, Sanz M, Messora MR. New tendencies in non-surgical periodontal therapy. Braz Oral Res 2021; 35:e095. [PMID: 34586209 DOI: 10.1590/1807-3107bor-2021.vol35.0095] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review was to update the evidence of new approaches to non-surgical therapy (NSPT) in the treatment of periodontitis. Preclinical and clinical studies addressing the benefits of adjunctive antimicrobial photodynamic therapy, probiotics, prebiotics/synbiotics, statins, pro-resolving mediators, omega-6 and -3, ozone, and epigenetic therapy were scrutinized and discussed. Currently, the outcomes of these nine new approaches, when compared with subgingival debridement alone, did not demonstrate a significant added clinical benefit. However, some of these new alternative interventions may have the potential to improve the outcomes of NSPT alone. Future evidence based on randomized controlled clinical trials would help clinicians and patients in the selection of different adjunctive therapies.
Collapse
Affiliation(s)
- Alex Nogueira Haas
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Flavia Furlaneto
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| | - Eduardo José Gaio
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Sabrina Carvalho Gomes
- Universidade Federal do Rio Grande do Sul - UFRGS, School of Dentistry, Department of Periodontology, Porto Alegre, RS, Brazil
| | - Daniela Bazan Palioto
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| | - Rogerio Moraes Castilho
- Michigan University, School of Dentistry, Department of Periodontics and Oral Medicine, Ann Arbor, MI, USA
| | - Mariano Sanz
- Complutense University of Madrid, Etiology and Therapy of Periodontal and Peri-implant Diseases Research Group, Madrid, Spain
| | - Michel Reis Messora
- Universidade de São Paulo - USP, School of Dentistry of Ribeirão Preto, Department of Oral Surgery and Periodontology, Ribeirão Preto, SP, Brazil
| |
Collapse
|
20
|
Lin P, Niimi H, Ohsugi Y, Tsuchiya Y, Shimohira T, Komatsu K, Liu A, Shiba T, Aoki A, Iwata T, Katagiri S. Application of Ligature-Induced Periodontitis in Mice to Explore the Molecular Mechanism of Periodontal Disease. Int J Mol Sci 2021; 22:ijms22168900. [PMID: 34445604 PMCID: PMC8396362 DOI: 10.3390/ijms22168900] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is an inflammatory disease characterized by the destruction of the periodontium. In the last decade, a new murine model of periodontitis has been widely used to simulate alveolar bone resorption and periodontal soft tissue destruction by ligation. Typically, 3-0 to 9-0 silks are selected for ligation around the molars in mice, and significant bone loss and inflammatory infiltration are observed within a week. The ligature-maintained period can vary according to specific aims. We reviewed the findings on the interaction of systemic diseases with periodontitis, periodontal tissue destruction, the immunological and bacteriological responses, and new treatments. In these studies, the activation of osteoclasts, upregulation of pro-inflammatory factors, and excessive immune response have been considered as major factors in periodontal disruption. Multiple genes identified in periodontal tissues partly reflect the complexity of the pathogenesis of periodontitis. The effects of novel treatment methods on periodontitis have also been evaluated in a ligature-induced periodontitis model in mice. This model cannot completely represent all aspects of periodontitis in humans but is considered an effective method for the exploration of its mechanisms. Through this review, we aimed to provide evidence and enlightenment for future studies planning to use this model.
Collapse
Affiliation(s)
- Peiya Lin
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Hiromi Niimi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
- Correspondence: (H.N.); (Y.O.); Tel.: +81-3-5803-5488 (H.N. & Y.O.)
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
- Correspondence: (H.N.); (Y.O.); Tel.: +81-3-5803-5488 (H.N. & Y.O.)
| | - Yosuke Tsuchiya
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Tsuyoshi Shimohira
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Keiji Komatsu
- Department of Lifetime Oral Health Care Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan;
| | - Anhao Liu
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Takahiko Shiba
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Akira Aoki
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8549, Japan; (P.L.); (Y.T.); (T.S.); (A.L.); (T.S.); (A.A.); (T.I.); (S.K.)
| |
Collapse
|
21
|
Liu Y, Huang W, Wang J, Ma J, Zhang M, Lu X, Liu J, Kou Y. Multifaceted Impacts of Periodontal Pathogens in Disorders of the Intestinal Barrier. Front Immunol 2021; 12:693479. [PMID: 34386004 PMCID: PMC8353228 DOI: 10.3389/fimmu.2021.693479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Periodontal disease, a common inflammatory disease, is considered a hazardous factor that contributes to the development of diseases of the digestive system as well as other systems. The bridge between periodontitis and systemic diseases is believed to be periodontal pathogens. The intestine, as part of the lower gastrointestinal tract, has a close connection with the oral cavity. Within the intestine, the intestinal barrier acts as a multifunctional system including microbial, mucous, physical and immune barrier. The intestinal barrier forms the body's first line of defense against external pathogens; its breakdown can lead to pathological changes in the gut and other organs or systems. Reports in the literature have described how oral periodontal pathogens and pathobiont-reactive immune cells can transmigrate to the intestinal mucosa, causing the destruction of intestinal barrier homeostasis. Such findings might lead to novel ideas for investigating the relationship between periodontal disease and other systemic diseases. This review summarizes studies on the effects of periodontal pathogens on the intestinal barrier, which might contribute to understanding the link between periodontitis and gastrointestinal diseases.
Collapse
Affiliation(s)
- Yingman Liu
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Wenxuan Huang
- School of Stomatology, Shenyang Medical College, Shenyang, China
| | - Jiaqi Wang
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Jiaojiao Ma
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Manman Zhang
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xiaoying Lu
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Jie Liu
- Science Experiment Center, China Medical University, Shenyang, China
| | - Yurong Kou
- Department of Periodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
- Department of Oral Biology, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
22
|
Rojas C, García MP, Polanco AF, González-Osuna L, Sierra-Cristancho A, Melgar-Rodríguez S, Cafferata EA, Vernal R. Humanized Mouse Models for the Study of Periodontitis: An Opportunity to Elucidate Unresolved Aspects of Its Immunopathogenesis and Analyze New Immunotherapeutic Strategies. Front Immunol 2021; 12:663328. [PMID: 34220811 PMCID: PMC8248545 DOI: 10.3389/fimmu.2021.663328] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/10/2021] [Indexed: 12/17/2022] Open
Abstract
Periodontitis is an oral inflammatory disease in which the polymicrobial synergy and dysbiosis of the subgingival microbiota trigger a deregulated host immune response, that leads to the breakdown of tooth-supporting tissues and finally tooth loss. Periodontitis is characterized by the increased pathogenic activity of T helper type 17 (Th17) lymphocytes and defective immunoregulation mediated by phenotypically unstable T regulatory (Treg), lymphocytes, incapable of resolving the bone-resorbing inflammatory milieu. In this context, the complexity of the immune response orchestrated against the microbial challenge during periodontitis has made the study of its pathogenesis and therapy difficult and limited. Indeed, the ethical limitations that accompany human studies can lead to an insufficient etiopathogenic understanding of the disease and consequently, biased treatment decision-making. Alternatively, animal models allow us to manage these difficulties and give us the opportunity to partially emulate the etiopathogenesis of periodontitis by inoculating periodontopathogenic bacteria or by placing bacteria-accumulating ligatures around the teeth; however, these models still have limited translational application in humans. Accordingly, humanized animal models are able to emulate human-like complex networks of immune responses by engrafting human cells or tissues into specific strains of immunodeficient mice. Their characteristics enable a viable time window for the study of the establishment of a specific human immune response pattern in an in vivo setting and could be exploited for a wider study of the etiopathogenesis and/or treatment of periodontitis. For instance, the antigen-specific response of human dendritic cells against the periodontopathogen Porphyromonas gingivalis favoring the Th17/Treg response has already been tested in humanized mice models. Hypothetically, the proper emulation of periodontal dysbiosis in a humanized animal could give insights into the subtle molecular characteristics of a human-like local and systemic immune response during periodontitis and support the design of novel immunotherapeutic strategies. Therefore, the aims of this review are: To elucidate how the microbiota-elicited immunopathogenesis of periodontitis can be potentially emulated in humanized mouse models, to highlight their advantages and limitations in comparison with the already available experimental periodontitis non-humanized animal models, and to discuss the potential translational application of using these models for periodontitis immunotherapeutics.
Collapse
Affiliation(s)
- Carolina Rojas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Michelle P García
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alan F Polanco
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Luis González-Osuna
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Alfredo Sierra-Cristancho
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Faculty of Dentistry, Universidad Andres Bello, Santiago, Chile
| | - Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| | - Emilio A Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima, Perú
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago, Chile.,Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago, Chile
| |
Collapse
|
23
|
Mata K, Nobre AVV, Felix Silva PH, Oliezer RS, Fernandes C, Amaral J, Ramos J, Constante Gabriel Del-Arco M, Messora MR, Tanus-Santos JE, Gerlach RF, Salvador SL. A new mixed model of periodontitis-induced preeclampsia: A pilot study. J Periodontal Res 2021; 56:726-734. [PMID: 33686671 DOI: 10.1111/jre.12869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023]
Abstract
OBJECTIVES/BACKGROUND Recent studies have shown that periodontal disease is strongly related to gestational complications such as preeclampsia (PE). PE is responsible for 42% of maternal deaths worldwide and kills approximately 76 000 women a year. In addition, children born under PE conditions are at increased risk of hospitalization due to metabolic disorders, epilepsy, and other complications. Numerous reviews and clinical studies on PE have been published, but the mechanisms underlying the relationship between periodontal disease and PE and the way periodontopathogens alter vascular response in pregnant women remain unclear. METHODS This study aims to verify whether periodontal disease induces PE by using the association of two periodontitis (PD) models: ligature and oral Porphyromonas gingivalis (P. gingivalis) W83 inoculation in Wistar rats. At gestational day 5, the ligature was placed on each mandibular first molar, which was followed by daily oral P. gingivalis inoculation for 15 days. At gestational day 19, urine was collected, and invasive arterial pressure was measured. The animals were euthanized, and plasma and tissues were collected. RESULTS After 15 days of the association of ligature and P. gingivalis inoculation, the animals presented the characteristic symptoms of PE: altered blood pressure, proteinuria, and change in litter size (number of pups) and pup weight when compared to the control group (p < .005). The PE animals also presented greater bone porosity, trabecular separation, and reduced bone volume in the hemimandibles, as well as altered inflammatory response. The level of cytokine IL-6 was higher in the PE group than in the control group (p < .005). CONCLUSION The association of two PD models effectively induced PE. To our knowledge, this is the first study on the oral use of P. gingivalis for PE induction. Our results support the importance of PD as a possible cause for PE development, opening an important new avenue to study cause and consequence relationships in inflammation and PE due to exposure to periodontal infection.
Collapse
Affiliation(s)
- Karina Mata
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil.,University of Rio Verde, UniRV, Formosa, GO, Brazil
| | - Atila Vinícius Vitor Nobre
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Pedro Henrique Felix Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Rene Seabra Oliezer
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Cleverson Fernandes
- University of Rio Verde, UniRV, Formosa, GO, Brazil.,Department of Pathology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Jefferson Amaral
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Junia Ramos
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Marina Constante Gabriel Del-Arco
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - Michel Reis Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, Ribeirao Preto Dental School, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| | - José Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Raquel Fernanda Gerlach
- Department of Basic and Oral Biology, Dental School of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Sergio Luiz Salvador
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo-USP, Ribeirao Preto, SP, Brazil
| |
Collapse
|
24
|
Hernández HG, Hernández-Castañeda AA, Pieschacón MP, Arboleda H. ZNF718, HOXA4, and ZFP57 are differentially methylated in periodontitis in comparison with periodontal health: Epigenome-wide DNA methylation pilot study. J Periodontal Res 2021; 56:710-725. [PMID: 33660869 DOI: 10.1111/jre.12868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 02/07/2021] [Accepted: 02/14/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To investigate the differences in the epigenomic patterns of DNA methylation in peripheral leukocytes between patients with periodontitis and gingivally healthy controls evaluating its functional meaning by functional enrichment analysis. BACKGROUND The DNA methylation profiling of peripheral leukocytes as immune-related tissue potentially relevant as a source of biomarkers between periodontitis patients and gingivally healthy subjects has not been investigated. METHODS A DNA methylation epigenome-wide study of peripheral leukocytes was conducted using the Illumina MethylationEPIC platform in sixteen subjects, eight diagnosed with periodontitis patients and eight age-matched and sex-matched periodontally healthy controls. A trained periodontist performed the clinical evaluation. Global DNA methylation was estimated using methylation-sensitive high-resolution melting in LINE1. Routine cell count cytometry and metabolic laboratory tests were also performed. The analysis of differentially methylated positions (DMPs) and differentially methylated regions (DMRs) was made using R/Bioconductor environment considering leukocyte populations assessed in both routine cell counts and using the FlowSorted.Blood.EPIC package. Finally, a DMP and DMR intersection analysis was performed. Functional enrichment analysis was carried out with the differentially methylated genes found in DMP. RESULTS DMP analysis identified 81 differentially hypermethylated genes and 21 differentially hypomethylated genes. Importantly, the intersection analysis showed that zinc finger protein 718 (ZNF718) and homeobox A4 (HOXA4) were differentially hypermethylated and zinc finger protein 57 (ZFP57) was differentially hypomethylated in periodontitis. The functional enrichment analysis found clearly immune-related ontologies such as "detection of bacterium" and "antigen processing and presentation." CONCLUSION The results of this study propose three new periodontitis-related genes: ZNF718, HOXA4, and ZFP57 but also evidence the suitability and relevance of studying leukocytes' DNA methylome for biological interpretation of systemic immune-related epigenetic patterns in periodontitis.
Collapse
Affiliation(s)
- Hernán G Hernández
- Faculty of Dentistry, Division of Health Sciences, Universidad Santo Tomás, Bucaramanga, Colombia
| | | | - Maria P Pieschacón
- Faculty of Dentistry, Division of Health Sciences, Universidad Santo Tomás, Bucaramanga, Colombia
| | - Humberto Arboleda
- Neurosciences Research Group, Faculty of Medicine and Genetic Institute, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
25
|
Decitabine Inhibits Bone Resorption in Periodontitis by Upregulating Anti-Inflammatory Cytokines and Suppressing Osteoclastogenesis. Biomedicines 2021; 9:biomedicines9020199. [PMID: 33671221 PMCID: PMC7922804 DOI: 10.3390/biomedicines9020199] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/12/2021] [Accepted: 02/13/2021] [Indexed: 12/14/2022] Open
Abstract
DNA methylation controls several inflammatory genes affecting bone homeostasis. Hitherto, inhibition of DNA methylation in vivo in the context of periodontitis and osteoclastogenesis has not been attempted. Ligature-induced periodontitis in C57BL/6J mice was induced by placing ligature for five days with Decitabine (5-aza-2′-deoxycytidine) (1 mg/kg/day) or vehicle treatment. We evaluated bone resorption, osteoclast differentiation by tartrate-resistant acid phosphatase (TRAP) and mRNA expression of anti-inflammatory molecules using cluster differentiation 14 positive (CD14+) monocytes from human peripheral blood. Our data showed that decitabine inhibited bone loss and osteoclast differentiation experimental periodontitis, and suppressed osteoclast CD14+ human monocytes; and conversely, that it increased bone mineralization in osteoblastic cell line MC3T3-E1 in a concentration-dependent manner. In addition to increasing IL10 (interleukin-10), TGFB (transforming growth factor beta-1) in CD14+ monocytes, decitabine upregulated KLF2 (Krüppel-like factor-2) expression. Overexpression of KLF2 protein enhanced the transcription of IL10 and TGFB. On the contrary, site-directed mutagenesis of KLF2 binding site in IL10 and TFGB abrogated luciferase activity in HEK293T cells. Decitabine reduces bone loss in a mouse model of periodontitis by inhibiting osteoclastogenesis through the upregulation of anti-inflammatory cytokines via KLF2 dependent mechanisms. DNA methyltransferase inhibitors merit further investigation as a possible novel therapy for periodontitis.
Collapse
|
26
|
Ramos-Lopez O, Milagro FI, Riezu-Boj JI, Martinez JA. Epigenetic signatures underlying inflammation: an interplay of nutrition, physical activity, metabolic diseases, and environmental factors for personalized nutrition. Inflamm Res 2021; 70:29-49. [PMID: 33231704 PMCID: PMC7684853 DOI: 10.1007/s00011-020-01425-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
AIM AND OBJECTIVE Emerging translational evidence suggests that epigenetic alterations (DNA methylation, miRNA expression, and histone modifications) occur after external stimuli and may contribute to exacerbated inflammation and the risk of suffering several diseases including diabetes, cardiovascular diseases, cancer, and neurological disorders. This review summarizes the current knowledge about the harmful effects of high-fat/high-sugar diets, micronutrient deficiencies (folate, manganese, and carotenoids), obesity and associated complications, bacterial/viral infections, smoking, excessive alcohol consumption, sleep deprivation, chronic stress, air pollution, and chemical exposure on inflammation through epigenetic mechanisms. Additionally, the epigenetic phenomena underlying the anti-inflammatory potential of caloric restriction, n-3 PUFA, Mediterranean diet, vitamin D, zinc, polyphenols (i.e., resveratrol, gallic acid, epicatechin, luteolin, curcumin), and the role of systematic exercise are discussed. METHODS Original and review articles encompassing epigenetics and inflammation were screened from major databases (including PubMed, Medline, Science Direct, Scopus, etc.) and analyzed for the writing of the review paper. CONCLUSION Although caution should be exercised, research on epigenetic mechanisms is contributing to understand pathological processes involving inflammatory responses, the prediction of disease risk based on the epigenotype, as well as the putative design of therapeutic interventions targeting the epigenome.
Collapse
Affiliation(s)
- Omar Ramos-Lopez
- Medicine and Psychology School, Autonomous University of Baja California, Tijuana, Baja California, Mexico
| | - Fermin I Milagro
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain.
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.
| | - Jose I Riezu-Boj
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - J Alfredo Martinez
- Department of Nutrition, Food Science and Physiology, Center for Nutrition Research, University of Navarra, 1 Irunlarrea Street, 31008, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain
- Precision Nutrition and Cardiometabolic Health, IMDEA-Food Institute (Madrid Institute for Advanced Studies), Madrid, Spain
| |
Collapse
|
27
|
Suárez LJ, Garzón H, Arboleda S, Rodríguez A. Oral Dysbiosis and Autoimmunity: From Local Periodontal Responses to an Imbalanced Systemic Immunity. A Review. Front Immunol 2020; 11:591255. [PMID: 33363538 PMCID: PMC7754713 DOI: 10.3389/fimmu.2020.591255] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022] Open
Abstract
The current paradigm of onset and progression of periodontitis includes oral dysbiosis directed by inflammophilic bacteria, leading to altered resolution of inflammation and lack of regulation of the inflammatory responses. In the construction of explanatory models of the etiopathogenesis of periodontal disease, autoimmune mechanisms were among the first to be explored and historically, for more than five decades, they have been described in an isolated manner as part of the tissue damage process observed in periodontitis, however direct participation of these mechanisms in the tissue damage is still controversial. Autoimmunity is affected by genetic and environmental factors, leading to an imbalance between the effector and regulatory responses, mostly associated with failed resolution mechanisms. However, dysbiosis/infection and chronic inflammation could trigger autoimmunity by several mechanisms including bystander activation, dysregulation of toll-like receptors, amplification of autoimmunity by cytokines, epitope spreading, autoantigens complementarity, autoantigens overproduction, microbial translocation, molecular mimicry, superantigens, and activation or inhibition of receptors related to autoimmunity by microorganisms. Even though autoreactivity in periodontitis is biologically plausible, the associated mechanisms could be related to non-pathologic responses which could even explain non-recognized physiological functions. In this review we shall discuss from a descriptive point of view, the autoimmune mechanisms related to periodontitis physio-pathogenesis and the participation of oral dysbiosis on local periodontal autoimmune responses as well as on different systemic inflammatory diseases.
Collapse
Affiliation(s)
- Lina J. Suárez
- Departamento de Ciencias Básicas y Medicina Oral, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Hernan Garzón
- Grupo de Investigación en Salud Oral, Universidad Antonio Nariño, Bogotá, Colombia
| | - Silie Arboleda
- Unidad de Investigación en Epidemiologia Clínica Oral (UNIECLO), Universidad El Bosque, Bogotá, Colombia
| | - Adriana Rodríguez
- Centro de Investigaciones Odontológicas, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
28
|
Jurdziński KT, Potempa J, Grabiec AM. Epigenetic regulation of inflammation in periodontitis: cellular mechanisms and therapeutic potential. Clin Epigenetics 2020; 12:186. [PMID: 33256844 PMCID: PMC7706209 DOI: 10.1186/s13148-020-00982-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epigenetic mechanisms, namely DNA and histone modifications, are critical regulators of immunity and inflammation which have emerged as potential targets for immunomodulating therapies. The prevalence and significant morbidity of periodontitis, in combination with accumulating evidence that genetic, environmental and lifestyle factors cannot fully explain the susceptibility of individuals to disease development, have driven interest in epigenetic regulation as an important factor in periodontitis pathogenesis. Aberrant promoter methylation profiles of genes involved in inflammatory activation, including TLR2, PTGS2, IFNG, IL6, IL8, and TNF, have been observed in the gingival tissue, peripheral blood or buccal mucosa from patients with periodontitis, correlating with changes in expression and disease severity. The expression of enzymes that regulate histone acetylation, in particular histone deacetylases (HDACs), is also dysregulated in periodontitis-affected gingival tissue. Infection of gingival epithelial cells, gingival fibroblasts and periodontal ligament cells with the oral pathogens Porphyromonas gingivalis or Treponema denticola induces alterations in expression and activity of chromatin-modifying enzymes, as well as site-specific and global changes in DNA methylation profiles and in histone acetylation and methylation marks. These epigenetic changes are associated with excessive production of inflammatory cytokines, chemokines, and matrix-degrading enzymes that can be suppressed by small molecule inhibitors of HDACs (HDACi) or DNA methyltransferases. HDACi and inhibitors of bromodomain-containing BET proteins ameliorate inflammation, osteoclastogenesis, and alveolar bone resorption in animal models of periodontitis, suggesting their clinical potential as host modulation therapeutic agents. However, broader application of epigenomic methods will be required to create a comprehensive map of epigenetic changes in periodontitis. The integration of functional studies with global analyses of the epigenetic landscape will provide critical information on the therapeutic and diagnostic potential of epigenetics in periodontal disease.
Collapse
Affiliation(s)
- Krzysztof T Jurdziński
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.,Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Aleksander M Grabiec
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
29
|
Qiao Y, Wang Z, Li Y, Han Y, Zhou Y, Cao X. Rheumatoid arthritis risk in periodontitis patients: A systematic review and meta-analysis. Joint Bone Spine 2020; 87:556-564. [PMID: 32593704 DOI: 10.1016/j.jbspin.2020.04.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 04/10/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Many clinical studies have been carried out to investigate the relationship between periodontitis and rheumatoid arthritis (RA). Owing to limited evidence and inconsistent findings among these studies, it is unclear whether periodontitis would increase the risk for RA. This meta-analysis was performed to evaluate whether periodontitis represents a risk factor for RA. METHODS PubMed, Cochrane Library, Embase, Web of Science, and Wanfang were searched for eligible studies that compared periodontitis patients with controls. A pooled odds ratio (OR) and 95% confidence interval (CI) were calculated to assess the association between periodontitis and RA. RESULTS Thirteen studies including a total of 706611 periodontitis patients and 349983 control subjects were included. The pooled OR of RA risk between periodontitis and controls was (OR: 1.69; 95% CI: 1.31-2.17; P<0.0001), indicating that the patients in periodontitis group had a 69% greater risk for RA than people in control group. When stratified by disease type, the pooled results showed periodontitis represents a risk factor for incident RA (OR=1.70, 95%CI: 0.75-3.85, P<0.001) and mixed RA (OR=1.61, 95%CI: 1.26-2.06; P<0.001). When stratified by disease duration, the pooled results showed periodontitis represents a risk factor for RA disease duration>5 years (OR=2.88, 95%CI: 0.66-12.62, P=0.018), disease duration<5 years (OR=2.59, 95%CI: 0.83-8.11, P<0.001), mixed disease duration (OR=1.53; 95%CI: 1.05-2.22, P<0.001). CONCLUSION Our meta-analysis revealed an increased risk of RA in patients with periodontitis compared to healthy controls. Moreover, when stratified by disease type, there was a higher risk between incident RA and periodontitis. When stratified by disease duration, the patients with periodontitis might be more closely associated with the RA patients with disease duration >5 years.
Collapse
Affiliation(s)
- Yiqiang Qiao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China
| | - Zao Wang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China; Zhengzhou Stomotology Hospital, Zhengzhou, 450000, P. R. China
| | - Yafang Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China; Zhengzhou Stomotology Hospital, Zhengzhou, 450000, P. R. China
| | - Yafei Han
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China
| | - Yanheng Zhou
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China
| | - Xuanping Cao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, P. R. China.
| |
Collapse
|
30
|
Gu Y, Han X. Toll-Like Receptor Signaling and Immune Regulatory Lymphocytes in Periodontal Disease. Int J Mol Sci 2020; 21:ijms21093329. [PMID: 32397173 PMCID: PMC7247565 DOI: 10.3390/ijms21093329] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/27/2022] Open
Abstract
Periodontitis is known to be initiated by periodontal microbiota derived from biofilm formation. The microbial dysbiotic changes in the biofilm trigger the host immune and inflammatory responses that can be both beneficial for the protection of the host from infection, and detrimental to the host, causing tissue destruction. During this process, recognition of Pathogen-Associated Molecular Patterns (PAMPs) by the host Pattern Recognition Receptors (PRRs) such as Toll-like receptors (TLRs) play an essential role in the host–microbe interaction and the subsequent innate as well as adaptive responses. If persistent, the adverse interaction triggered by the host immune response to the microorganisms associated with periodontal biofilms is a direct cause of periodontal inflammation and bone loss. A large number of T and B lymphocytes are infiltrated in the diseased gingival tissues, which can secrete inflammatory mediators and activate the osteolytic pathways, promoting periodontal inflammation and bone resorption. On the other hand, there is evidence showing that immune regulatory T and B cells are present in the diseased tissue and can be induced for the enhancement of their anti-inflammatory effects. Changes and distribution of the T/B lymphocytes phenotype seem to be a key determinant of the periodontal disease outcome, as the functional activities of these cells not only shape up the overall immune response pattern, but may directly regulate the osteoimmunological balance. Therefore, interventional strategies targeting TLR signaling and immune regulatory T/B cells may be a promising approach to rebalance the immune response and alleviate bone loss in periodontal disease. In this review, we will examine the etiological role of TLR signaling and immune cell osteoclastogenic activity in the pathogenesis of periodontitis. More importantly, the protective effects of immune regulatory lymphocytes, particularly the activation and functional role of IL-10 expressing regulatory B cells, will be discussed.
Collapse
Affiliation(s)
- Yingzhi Gu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA;
| | - Xiaozhe Han
- Department of Immunology and Infectious Diseases, The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA;
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
31
|
Coêlho MC, Queiroz IC, Viana JMC, de Aquino SG, Persuhn DC, de Oliveira NFP. miR-9-1 gene methylation and DNMT3B (rs2424913) polymorphism may contribute to periodontitis. J Appl Oral Sci 2020; 28:e20190583. [PMID: 32267380 PMCID: PMC7137733 DOI: 10.1590/1678-7757-2019-0583] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/11/2019] [Accepted: 11/27/2019] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Genetic and epigenetic changes have been associated with periodontitis in various genes; however, little is known about genes involved in epigenetic mechanisms and in oxidative stress. OBJECTIVE This study aims to investigate the association of polymorphisms C677T in MTHFR (rs1801133) and -149C→T in DNMT3B (rs2424913), as well as the methylation profiles of MTHFR, miR-9-1, miR-9-3, SOD1, and CAT with periodontitis. The association between polymorphisms and DNA methylation profiles was also analyzed. METHODOLOGY The population studied was composed of 100 nonsmokers of both sexes, divided into healthy and periodontitis groups. Genomic DNA was extracted from the epithelial buccal cells, which were collected through a mouthwash. Polymorphism analysis was performed through polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), while methylation-specific PCR (MSP) or combined bisulfite restriction analysis techniques were applied for methylation analysis. RESULTS For DNMT3B, the T allele and the TT genotype were detected more frequently in the periodontitis group, as well as the methylated profile on the miR-9-1 promoter region. There was also a tendency towards promoter region methylation on the CAT sequence of individuals with periodontal disease. CONCLUSION The polymorphism -149C→T in DNMT3B (rs2424913) and the methylated profile of the miR-9-1 promoter region are associated with periodontitis.
Collapse
Affiliation(s)
- Marina Castro Coêlho
- Universidade Federal da ParaíbaCentro de Ciências da SaúdePrograma de Pós Graduação em OdontologiaJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós Graduação em Odontologia, João Pessoa, Paraíba, Brasil.
| | - Ingrid Costa Queiroz
- Universidade Federal da ParaíbaCentro de Ciências da SaúdeJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, João Pessoa, Paraíba, Brasil.
| | - José Maria Chagas Viana
- Universidade Federal da ParaíbaCentro de Ciências da SaúdePrograma de Pós Graduação em OdontologiaJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós Graduação em Odontologia, João Pessoa, Paraíba, Brasil.
| | - Sabrina Garcia de Aquino
- Universidade Federal da ParaíbaCentro de Ciências da SaúdePrograma de Pós Graduação em OdontologiaJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós Graduação em Odontologia, João Pessoa, Paraíba, Brasil.
- Universidade Federal da ParaíbaCentro de Ciências da SaúdeDepartamento de Odontologia Clínica e SocialJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, Departamento de Odontologia Clínica e Social, João Pessoa, Paraíba, Brasil.
| | - Darlene Camati Persuhn
- Universidade Federal da ParaíbaCentro de Ciências Exatas e da NaturezaDepartamento de Biologia MolecularJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências Exatas e da Natureza, Departamento de Biologia Molecular, João Pessoa, Paraíba, Brasil.
| | - Naila Francis Paulo de Oliveira
- Universidade Federal da ParaíbaCentro de Ciências da SaúdePrograma de Pós Graduação em OdontologiaJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós Graduação em Odontologia, João Pessoa, Paraíba, Brasil.
- Universidade Federal da ParaíbaCentro de Ciências Exatas e da NaturezaDepartamento de Biologia MolecularJoão PessoaParaíbaBrasilUniversidade Federal da Paraíba, Centro de Ciências Exatas e da Natureza, Departamento de Biologia Molecular, João Pessoa, Paraíba, Brasil.
| |
Collapse
|
32
|
Rath-Deschner B, Memmert S, Damanaki A, Nokhbehsaim M, Eick S, Cirelli JA, Götz W, Deschner J, Jäger A, Nogueira AVB. CXCL1, CCL2, and CCL5 modulation by microbial and biomechanical signals in periodontal cells and tissues-in vitro and in vivo studies. Clin Oral Investig 2020; 24:3661-3670. [PMID: 32124070 DOI: 10.1007/s00784-020-03244-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/12/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVES This study was established to investigate whether the chemokines CXCL1, CCL2, and CCL5 are produced in periodontal cells and tissues and, if so, whether their levels are regulated by microbial and/or mechanical signals. MATERIALS AND METHODS The chemokine expression and protein levels in gingival biopsies from patients with and without periodontitis were analyzed by RT-PCR and immunohistochemistry. The chemokines were also analyzed in gingival biopsies from rats subjected to experimental periodontitis and/or orthodontic tooth movement. Additionally, chemokine levels were determined in periodontal fibroblasts exposed to the periodontopathogen Fusobacterium nucleatum and mechanical forces by RT-PCR and ELISA. RESULTS Higher CXCL1, CCL2, and CCL5 levels were found in human and rat gingiva from sites of periodontitis as compared with periodontally healthy sites. In the rat experimental periodontitis model, the bacteria-induced upregulation of these chemokines was significantly counteracted by orthodontic forces. In vitro, F. nucleatum caused a significant upregulation of all chemokines at 1 day. When the cells were subjected simultaneously to F. nucleatum and mechanical forces, the upregulation of chemokines was significantly inhibited. The transcriptional findings were paralleled at protein level. CONCLUSIONS This study provides original evidence in vitro and in vivo that the chemokines CXCL1, CCL2, and CCL5 are regulated by both microbial and mechanical signals in periodontal cells and tissues. Furthermore, our study revealed that biomechanical forces can counteract the stimulatory actions of F. nucleatum on these chemokines. CLINICAL RELEVANCE Mechanical loading might aggravate periodontal infection by compromising the recruitment of immunoinflammatory cells.
Collapse
Affiliation(s)
- Birgit Rath-Deschner
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstrasse 17, 53111, Bonn, Germany.
| | - Svenja Memmert
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstrasse 17, 53111, Bonn, Germany.,Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
| | - Anna Damanaki
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Marjan Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Bonn, Germany
| | - Sigrun Eick
- Department of Periodontology, Laboratory for Oral Microbiology, University of Bern, Bern, Switzerland
| | - Joni A Cirelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, Sao Paulo State University, UNESP, Araraquara, Brazil
| | - Werner Götz
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstrasse 17, 53111, Bonn, Germany
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Andreas Jäger
- Department of Orthodontics, Center of Dento-Maxillo-Facial Medicine, University of Bonn, Welschnonnenstrasse 17, 53111, Bonn, Germany
| | - Andressa V B Nogueira
- Department of Periodontology and Operative Dentistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
33
|
Olsen I, Singhrao SK. Is there a link between genetic defects in the complement cascade and Porphyromonas gingivalis in Alzheimer's disease? J Oral Microbiol 2019; 12:1676486. [PMID: 31893014 PMCID: PMC6818111 DOI: 10.1080/20002297.2019.1676486] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
Defects, as determined by Genome-Wide Association Studies (GWAS), in the complement cascade of innate immunity have been suggested to play a key role in Alzheimer's disease (AD). These defective genes encode sub-component 1s (C1s), complement receptor 1, complement component 9, and clusterin, a fluid-phase regulatory protein. A dysregulated complement cascade has been shown to relate to cell activation, defective complement mediated clearance and possible cognitive decline in AD patients. Porphyromonas gingivalis, a putative keystone pathogen of periodontal disease, has been reported to be associated with human AD. The inflammatory burden following experimental oral infection in mice and putative entry of this bacterium into the brain appears to drive the formation of amyloid-beta plaques and neurofibrillary tangles with loss of cognition. P. gingivalis is a master of immune subversion in this inflammatory cascade and may establish microbial dysbiosis where it is located. Here we discuss if P. gingivalis may enhance the detrimental effects of the defective GWAS complement cascade protein genes.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Sim K Singhrao
- Dementia and Neurodegenerative Diseases Research Group, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| |
Collapse
|
34
|
Linkage of Periodontitis and Rheumatoid Arthritis: Current Evidence and Potential Biological Interactions. Int J Mol Sci 2019; 20:ijms20184541. [PMID: 31540277 PMCID: PMC6769683 DOI: 10.3390/ijms20184541] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 08/07/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
The association between rheumatoid arthritis (RA) and periodontal disease (PD) has been the focus of numerous investigations driven by their common pathological features. RA is an autoimmune disease characterized by chronic inflammation, the production of anti-citrullinated proteins antibodies (ACPA) leading to synovial joint inflammation and destruction. PD is a chronic inflammatory condition associated with a dysbiotic microbial biofilm affecting the supporting tissues around the teeth leading to the destruction of mineralized and non-mineralized connective tissues. Chronic inflammation associated with both RA and PD is similar in the predominant adaptive immune phenotype, in the imbalance between pro- and anti-inflammatory cytokines and in the role of smoking and genetic background as risk factors. Structural damage that occurs in consequence of chronic inflammation is the ultimate cause of loss of function and disability observed with the progression of RA and PD. Interestingly, the periodontal pathogen Porphyromonas gingivalis has been implicated in the generation of ACPA in RA patients, suggesting a direct biological intersection between PD and RA. However, more studies are warranted to confirm this link, elucidate potential mechanisms involved, and ascertain temporal associations between RA and PD. This review is mainly focused on recent clinical and translational research intends to discuss and provide an overview of the relationship between RA and PD, exploring the similarities in the immune-pathological aspects and the possible mechanisms linking the development and progression of both diseases. In addition, the current available treatments targeting both RA and PD were revised.
Collapse
|