1
|
Konkel JE, Cox JR, Wemyss K. Bite-sized immunology; damage and microbes educating immunity at the gingiva. Mucosal Immunol 2024; 17:1141-1150. [PMID: 39038755 DOI: 10.1016/j.mucimm.2024.07.004] [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: 06/14/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Immune cells residing at the gingiva experience diverse and unique signals, tailoring their functions to enable them to appropriately respond to immunological challenges and maintain tissue integrity. The gingiva, defined as the mucosal barrier that surrounds and supports the teeth, is the only barrier site completely transected by a hard structure, the tooth. The tissue is damaged in early life during tooth eruption and chronically throughout life by the process of mastication. This occurs alongside challenges typical of barrier sites, including exposure to invading pathogens, the local commensal microbial community and environmental antigens. This review will focus on the immune network safeguarding gingival integrity, which is far less understood than that resident at other barrier sites. A detailed understanding of the gingiva-resident immune network is vital as it is the site of the inflammatory disease periodontitis, the most common chronic inflammatory condition in humans which has well-known detrimental systemic effects. Furthering our understanding of how the immune populations within the gingiva develop, are tailored in health, and how this is dysregulated in disease would further the development of effective therapies for periodontitis.
Collapse
Affiliation(s)
- Joanne E Konkel
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.
| | - Joshua R Cox
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Kelly Wemyss
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| |
Collapse
|
2
|
Bittner-Eddy PD, Fischer LA, Parachuru PV, Costalonga M. MHC-II presentation by oral Langerhans cells impacts intraepithelial Tc17 abundance and Candida albicans oral infection via CD4 T cells. FRONTIERS IN ORAL HEALTH 2024; 5:1408255. [PMID: 38872986 PMCID: PMC11169704 DOI: 10.3389/froh.2024.1408255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 05/13/2024] [Indexed: 06/15/2024] Open
Abstract
In a murine model (LCΔMHC-II) designed to abolish MHC-II expression in Langerhans cells (LCs), ∼18% of oral LCs retain MHC-II, yet oral mucosal CD4 T cells numbers are unaffected. In LCΔMHC-II mice, we now show that oral intraepithelial conventional CD8αβ T cell numbers expand 30-fold. Antibody-mediated ablation of CD4 T cells in wild-type mice also resulted in CD8αβ T cell expansion in the oral mucosa. Therefore, we hypothesize that MHC class II molecules uniquely expressed on Langerhans cells mediate the suppression of intraepithelial resident-memory CD8 T cell numbers via a CD4 T cell-dependent mechanism. The expanded oral CD8 T cells co-expressed CD69 and CD103 and the majority produced IL-17A [CD8 T cytotoxic (Tc)17 cells] with a minority expressing IFN-γ (Tc1 cells). These oral CD8 T cells showed broad T cell receptor Vβ gene usage indicating responsiveness to diverse oral antigens. Generally supporting Tc17 cells, transforming growth factor-β1 (TGF-β1) increased 4-fold in the oral mucosa. Surprisingly, blocking TGF-β1 signaling with the TGF-R1 kinase inhibitor, LY364947, did not reduce Tc17 or Tc1 numbers. Nonetheless, LY364947 increased γδ T cell numbers and decreased CD49a expression on Tc1 cells. Although IL-17A-expressing γδ T cells were reduced by 30%, LCΔMHC-II mice displayed greater resistance to Candida albicans in early stages of oral infection. These findings suggest that modulating MHC-II expression in oral LC may be an effective strategy against fungal infections at mucosal surfaces counteracted by IL-17A-dependent mechanisms.
Collapse
Affiliation(s)
- Peter D. Bittner-Eddy
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Lori A. Fischer
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Praveen Venkata Parachuru
- Division of Periodontology, Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| | - Massimo Costalonga
- Division of Basic Sciences, Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
3
|
Kondo T, Gleason A, Okawa H, Hokugo A, Nishimura I. Mouse gingival single-cell transcriptomic atlas identified a novel fibroblast subpopulation activated to guide oral barrier immunity in periodontitis. eLife 2023; 12:RP88183. [PMID: 38015204 PMCID: PMC10684155 DOI: 10.7554/elife.88183] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023] Open
Abstract
Periodontitis, one of the most common non-communicable diseases, is characterized by chronic oral inflammation and uncontrolled tooth supporting alveolar bone resorption. Its underlying mechanism to initiate aberrant oral barrier immunity has yet to be delineated. Here, we report a unique fibroblast subpopulation activated to guide oral inflammation (AG fibroblasts) identified in a single-cell RNA sequencing gingival cell atlas constructed from the mouse periodontitis models. AG fibroblasts localized beneath the gingival epithelium and in the cervical periodontal ligament responded to the ligature placement and to the discrete topical application of Toll-like receptor stimulants to mouse maxillary tissue. The upregulated chemokines and ligands of AG fibroblasts linked to the putative receptors of neutrophils in the early stages of periodontitis. In the established chronic inflammation, neutrophils, together with AG fibroblasts, appeared to induce type 3 innate lymphoid cells (ILC3s) that were the primary source of interleukin-17 cytokines. The comparative analysis of Rag2-/- and Rag2-/-Il2rg-/- mice suggested that ILC3 contributed to the cervical alveolar bone resorption interfacing the gingival inflammation. We propose the AG fibroblast-neutrophil-ILC3 axis as a previously unrecognized mechanism which could be involved in the complex interplay between oral barrier immune cells contributing to pathological inflammation in periodontitis.
Collapse
Affiliation(s)
- Takeru Kondo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of DentistryLos AngelesUnited States
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of DentistrySendaiJapan
| | - Annie Gleason
- Weintraub Center for Reconstructive Biotechnology, UCLA School of DentistryLos AngelesUnited States
- UCLA Bruin in Genomics Summer ProgramLos AngelesUnited States
| | - Hiroko Okawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of DentistryLos AngelesUnited States
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of DentistrySendaiJapan
| | - Akishige Hokugo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of DentistryLos AngelesUnited States
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLALos AngelesUnited States
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of DentistryLos AngelesUnited States
| |
Collapse
|
4
|
Kondo T, Gleason A, Okawa H, Hokugo A, Nishimura I. Mouse gingival single-cell transcriptomic atlas: An activated fibroblast subpopulation guides oral barrier immunity in periodontitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.13.536751. [PMID: 37546811 PMCID: PMC10401928 DOI: 10.1101/2023.04.13.536751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Periodontitis, one of the most common non-communicable diseases, is characterized by chronic oral inflammation and uncontrolled tooth supporting alveolar bone resorption. Its underlying mechanism to initiate aberrant oral barrier immunity has yet to be delineated. Here, we report a unique fibroblast subpopulation activated to guide oral inflammation (AG fibroblasts) identified in a single-cell RNA sequencing gingival cell atlas constructed from the mouse periodontitis models. AG fibroblasts localized beneath the gingival epithelium and in the cervical periodontal ligament responded to the ligature placement and to the discrete application of Toll-like receptor stimulants to mouse maxillary tissue. The upregulated chemokines and ligands of AG fibroblasts linked to the putative receptors of neutrophils in the early stages of periodontitis. In the established chronic inflammation, neutrophils together with AG fibroblasts appeared to induce type 3 innate lymphoid cells (ILC3s) that were the primary source of interleukin-17 cytokines. The comparative analysis of Rag2-/- and Rag2γc-/- mice suggested that ILC3 contributed to the cervical alveolar bone resorption interfacing the gingival inflammation. We propose that AG fibroblasts function as a previously unrecognized surveillant to initiate gingival inflammation leading to periodontitis through the AG fibroblast-neutrophil-ILC3 axis.
Collapse
Affiliation(s)
- Takeru Kondo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi 980-8575, Japan
| | - Annie Gleason
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- UCLA Bruin in Genomics Summer Program
| | - Hiroko Okawa
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi 980-8575, Japan
| | - Akishige Hokugo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
- Regenerative Bioengineering and Repair Laboratory, Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Chen D, Li Z, Li Z, Sun Y, Liu Q, Yang J, Song J, Cai H, Feng Z, Chen Z, Huang B. Transcriptome analysis of human peri-implant soft tissue and periodontal gingiva: a paired design study. Clin Oral Investig 2023:10.1007/s00784-023-05017-y. [PMID: 37060358 DOI: 10.1007/s00784-023-05017-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 04/02/2023] [Indexed: 04/16/2023]
Abstract
OBJECTIVES Limited information is available about the biological characterization of peri-implant soft tissue at the transcriptional level. The aim of this study was to investigate the effect of dental implant on the soft tissue in vivo by using paired samples and compare the differences between peri-implant soft tissue and periodontal gingiva at the transcriptional level. METHODS Paired peri-implant soft tissue and periodontal gingiva tissue from 6 patients were obtained, and the pooled RNAs were analyzed by deep sequencing. Venn diagram was used to further screen out differentially expressed genes in every pair of samples. Annotation and enrichment analysis was performed. Further verification was done by quantitative real-time PCR. RESULTS Totally 3549 differentially expressed genes (DEGs) were found between peri-implant and periodontal groups. The Venn diagram further identified 185 DEGs in every pair of samples, of which the enrichment analysis identified significant enrichment for cellular component was associated with external side of plasma membrane, for molecular function was protein binding, for biological process was immune system process, and for KEGG pathway was cytokine-cytokine receptor interaction. Among the DEGs, CST1, SPP1, AQP9, and SFRP2 were verified to be upregulated in peri-implant soft tissue. CONCLUSIONS Peri-implant soft tissue showed altered expressions of several genes related to the cell-ECM interaction compared to periodontal gingiva. CLINICAL RELEVANCE Compared to periodontal gingiva, altered cell-ECM interactions in peri-implant may contribute to the susceptibility of peri-implant diseases. At the transcriptional level, periodontal gingiva is generally considered the appropriate control for peri-implantitis, except regarding the cell-ECM interactions.
Collapse
Affiliation(s)
- Danying Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Zhixin Li
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Zhipeng Li
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Yue Sun
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Qifan Liu
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jieting Yang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jiaying Song
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Huaxiong Cai
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Zhicai Feng
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Zhuofan Chen
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Baoxin Huang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
6
|
Chen H, Feng X, Yang Q, Yang K, Man S. Expression Pattern and Value of Brain-Derived Neurotrophic Factor in Periodontitis. Int Dent J 2023:S0020-6539(23)00055-2. [PMID: 36997422 PMCID: PMC10390664 DOI: 10.1016/j.identj.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/14/2022] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Periodontitis is a common human disease with an increasing incidence. Brain-derived neurotrophic factor (BDNF) is known to play a crucial role in the regeneration of periodontal tissue; however, the expression, methylation level, molecular function, and clinical value of BDNF in periodontitis require further investigation. This study aimed to investigate the expression and potential functions of BDNF in periodontitis. METHODS RNA expression and methylation data were obtained from the Gene Expression Omnibus (GEO) database, and the expression and methylation levels of BDNF were compared between periodontitis and normal tissues. In addition, bioinformatics analysis was performed to investigate the downstream molecular functions of BDNF. Finally, Reverse transcription Quantitative real-time polymerase chain reaction was performed to determine the level of BDNF expression in periodontitis and normal tissues. RESULTS GEO database analysis revealed that BDNF was hypermethylated in periodontitis tissues and that its expression was downregulated. Reverse transcription Quantitative real-time polymerase chain reaction confirmed that BDNF expression was downregulated in periodontitis tissues. Several genes that interact with BDNF were determined using a protein-protein interaction network. Functional analysis of BDNF revealed that it was enriched in the Gene Ontology terms cytoplasmic dynein complex, glutathione transferase activity, and glycoside metabolic process. Kyoto Encyclopedia of Genes and Genomes analysis suggested that BDNF was associated with the mechanistic target of rapamycin signaling pathway, fatty acid metabolism, the Janus kinase-signal transducer and activator of transcription signaling pathway, glutathione metabolism, and others. Furthermore, the level of BDNF expression was correlated with the immune infiltration degree of B cells and CD4+ T cells. CONCLUSIONS This study shown that BDNF was hypermethylated and downregulated in periodontitis tissues, which could be a biomarker and treatment target of periodontitis.
Collapse
|
7
|
Sheibak N, Heidari Z, Mahmoudzadeh-Sagheb H. Immunoexpression of interferon-gamma in the interdental gingiva of chronic periodontitis patients with interferon-gamma (+874A/T) rs62559044 polymorphism. J Oral Biol Craniofac Res 2022; 12:727-732. [DOI: 10.1016/j.jobcr.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/25/2022] [Accepted: 08/20/2022] [Indexed: 10/15/2022] Open
|
8
|
Polarization Profiles of T Lymphocytes and Macrophages Responses in Periodontitis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:195-208. [PMID: 35612799 DOI: 10.1007/978-3-030-96881-6_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Periodontitis is a multifactorial, chronic inflammatory disease affecting the supporting structures of teeth triggered by the complex interactions between a dysbiotic bacterial biofilm and the host's immune response that results in the characteristic loss of periodontal attachment and alveolar bone. The differential phenotypic presentations of periodontitis emerge from inter-individual differences in immune response regulatory mechanisms. The monocyte-macrophage system has a crucial role in innate immunity and the initiation of the T and B lymphocyte adaptive immune responses. Macrophages involve a heterogeneous cell population that shows wide plasticity and differentiation dynamics. In response to the inflammatory milieu, they can skew at the time of TLR ligation to predominant M1 -pro-inflammatory- or M2 -anti-inflammatory/healing- functional phenotypes. The perpetuation of inflammation by M1 macrophages leads to the recruitment of the adaptive immune response, promoting Th1, Th17, and Th22 differentiation, which are directly associated with periodontal breakdown. In contrast, M2 macrophages induce Th2 and Treg responses which are associated with periodontal homeostasis. In this article, we review the recent advances comprising the role of macrophages and lymphocyte polarization profiles and their reprogramming as potential therapeutic strategies. For this purpose, we reviewed the available literature targeting periodontitis, macrophage, and lymphocyte subpopulations with an emphasis in the later 5 years. The active reprogramming of macrophages and lymphocytes polarization crosstalk opens a promising area for therapeutic development.
Collapse
|
9
|
Jiang Q, Huang X, Yu W, Huang R, Zhao X, Chen C. mTOR Signaling in the Regulation of CD4+ T Cell Subsets in Periodontal Diseases. Front Immunol 2022; 13:827461. [PMID: 35222410 PMCID: PMC8866697 DOI: 10.3389/fimmu.2022.827461] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/20/2022] [Indexed: 02/05/2023] Open
Abstract
Periodontal disease results from the inflammatory infiltration by the microbial community which is marked through tooth mobility and alveolar bone resorption. The inflammation in periodontal disease is mediated by CD4+ T cells through cytokine secretion and osteoclastogenetic activity. Historically, the inflammatory model in periodontal disease is described through disruption of the balance between two subsets of T helper cells which are T-helper type 1 (Th1) and T-helper type 2 (Th2). However, more and more studies have found that apart from subsets of helper T cells, regulatory T-cells and Th17 cells are also involved in the pathogenesis of periodontal diseases. Growing evidence proves that helper T cells differentiation, activation, and subset determination are under the strong impact of mTOR signaling. mTOR signaling could promote Th1 and Th17 cell differentiation and inhibit Treg commitment through different mTOR complexes, therefore we anticipate a regulation effect of mTOR signaling on periodontal diseases by regulating CD4+ T cell subsets. This review aims to integrate the topical researches about the role of different types of Th cells in the pathogenesis of periodontal diseases, as well as the regulation of mTOR signaling in the specification and selection of Th cell commitment.
Collapse
Affiliation(s)
- Qian Jiang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xiaobin Huang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wenjing Yu
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ranran Huang
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Xuefeng Zhao
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States.,Center of Innovation and Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
10
|
Berthelot JM, Bandiaky ON, Le Goff B, Amador G, Chaux AG, Soueidan A, Denis F. Another Look at the Contribution of Oral Microbiota to the Pathogenesis of Rheumatoid Arthritis: A Narrative Review. Microorganisms 2021; 10:59. [PMID: 35056507 PMCID: PMC8778040 DOI: 10.3390/microorganisms10010059] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/17/2022] Open
Abstract
Although autoimmunity contributes to rheumatoid arthritis (RA), several lines of evidence challenge the dogma that it is mainly an autoimmune disorder. As RA-associated human leukocyte antigens shape microbiomes and increase the risk of dysbiosis in mucosae, RA might rather be induced by epigenetic changes in long-lived synovial presenting cells, stressed by excessive translocations into joints of bacteria from the poorly cultivable gut, lung, or oral microbiota (in the same way as more pathogenic bacteria can lead to "reactive arthritis"). This narrative review (i) lists evidence supporting this scenario, including the identification of DNA from oral and gut microbiota in the RA synovium (but in also healthy synovia), and the possibility of translocation through blood, from mucosae to joints, of microbiota, either directly from the oral cavity or from the gut, following an increase of gut permeability worsened by migration within the gut of oral bacteria such as Porphyromonas gingivalis; (ii) suggests other methodologies for future works other than cross-sectional studies of periodontal microbiota in cohorts of patients with RA versus controls, namely, longitudinal studies of oral, gut, blood, and synovial microbiota combined with transcriptomic analyses of immune cells in individual patients at risk of RA, and in overt RA, before, during, and following flares of RA.
Collapse
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Unit, Nantes University Hospital, Place Alexis Ricordeau, CEDEX 01, 44093 Nantes, France; (J.-M.B.); (B.L.G.)
| | - Octave Nadile Bandiaky
- Division of Fixed Prosthodontics, University of Nantes, 1 Place Alexis Ricordeau, 44042 Nantes, France;
| | - Benoit Le Goff
- Rheumatology Unit, Nantes University Hospital, Place Alexis Ricordeau, CEDEX 01, 44093 Nantes, France; (J.-M.B.); (B.L.G.)
| | - Gilles Amador
- Department of Dental Public Health, Faculty of Dental Surgery, University of Nantes, 44093 Nantes, France;
- Nantes Teaching Hospital, 44000 Nantes, France;
| | - Anne-Gaelle Chaux
- Nantes Teaching Hospital, 44000 Nantes, France;
- Department of Oral Surgery, Faculty of Dental Surgery, University of Nantes, 44000 Nantes, France
| | - Assem Soueidan
- Department of Periodontology, Faculty of Dental Surgery, UIC 11, Rmes U1229, CHU de Nantes, 44000 Nantes, France;
| | - Frederic Denis
- Department of Dental Public Health, Faculty of Dental Surgery, University of Nantes, 44093 Nantes, France;
- Tours Teaching Hospital, 37000 Tours, France
| |
Collapse
|
11
|
Sun X, Gao J, Meng X, Lu X, Zhang L, Chen R. Polarized Macrophages in Periodontitis: Characteristics, Function, and Molecular Signaling. Front Immunol 2021; 12:763334. [PMID: 34950140 PMCID: PMC8688840 DOI: 10.3389/fimmu.2021.763334] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/04/2021] [Indexed: 12/23/2022] Open
Abstract
Periodontitis (PD) is a common chronic infectious disease. The local inflammatory response in the host may cause the destruction of supporting periodontal tissue. Macrophages play a variety of roles in PD, including regulatory and phagocytosis. Moreover, under the induction of different factors, macrophages polarize and form different functional phenotypes. Among them, M1-type macrophages with proinflammatory functions and M2-type macrophages with anti-inflammatory functions are the most representative, and both of them can regulate the tendency of the immune system to exert proinflammatory or anti-inflammatory functions. M1 and M2 macrophages are involved in the destructive and reparative stages of PD. Due to the complex microenvironment of PD, the dynamic development of PD, and various local mediators, increasing attention has been given to the study of macrophage polarization in PD. This review summarizes the role of macrophage polarization in the development of PD and its research progress.
Collapse
Affiliation(s)
- Xiaoyu Sun
- *Correspondence: Lei Zhang, ; Xiaoyu Sun,
| | | | | | | | - Lei Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, Department of Periodontology, Stomatologic Hospital & College, Anhui Medical University, Hefei, China
| | | |
Collapse
|
12
|
Yang B, Pang X, Li Z, Chen Z, Wang Y. Immunomodulation in the Treatment of Periodontitis: Progress and Perspectives. Front Immunol 2021; 12:781378. [PMID: 34868054 PMCID: PMC8640126 DOI: 10.3389/fimmu.2021.781378] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Periodontitis is one of the most common dental diseases. Compared with healthy periodontal tissues, the immune microenvironment plays the key role in periodontitis by allowing the invasion of pathogens. It is possible that modulating the immune microenvironment can supplement traditional treatments and may even promote periodontal regeneration by using stem cells, bacteria, etc. New anti-inflammatory therapies can enhance the generation of a viable local immune microenvironment and promote cell homing and tissue formation, thereby achieving higher levels of immune regulation and tissue repair. We screened recent studies to summarize the advances of the immunomodulatory treatments for periodontitis in the aspects of drug therapy, microbial therapy, stem cell therapy, gene therapy and other therapies. In addition, we included the changes of immune cells and cytokines in the immune microenvironment of periodontitis in the section of drug therapy so as to make it clearer how the treatments took effects accordingly. In the future, more research needs to be done to improve immunotherapy methods and understand the risks and long-term efficacy of these methods in periodontitis.
Collapse
Affiliation(s)
- Bo Yang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xuefei Pang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhipeng Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhuofan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yan Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Luan X, Zhou X, Fallah P, Pandya M, Lyu H, Foyle D, Burch D, Diekwisch TGH. MicroRNAs: Harbingers and shapers of periodontal inflammation. Semin Cell Dev Biol 2021; 124:85-98. [PMID: 34120836 DOI: 10.1016/j.semcdb.2021.05.030] [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: 02/02/2021] [Revised: 05/03/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023]
Abstract
Periodontal disease is an inflammatory reaction of the periodontal tissues to oral pathogens. In the present review we discuss the intricate effects of a regulatory network of gene expression modulators, microRNAs (miRNAs), as they affect periodontal morphology, function and gene expression during periodontal disease. These miRNAs are small RNAs involved in RNA silencing and post-transcriptional regulation and affect all stages of periodontal disease, from the earliest signs of gingivitis to the regulation of periodontal homeostasis and immunity and to the involvement in periodontal tissue destruction. MiRNAs coordinate periodontal disease progression not only directly but also through long non-coding RNAs (lncRNAs), which have been demonstrated to act as endogenous sponges or decoys that regulate the expression and function of miRNAs, and which in turn suppress the targeting of mRNAs involved in the inflammatory response, cell proliferation, migration and differentiation. While the integrity of miRNA function is essential for periodontal health and immunity, miRNA sequence variations (genetic polymorphisms) contribute toward an enhanced risk for periodontal disease progression and severity. Several polymorphisms in miRNA genes have been linked to an increased risk of periodontitis, and among those, miR-146a, miR-196, and miR-499 polymorphisms have been identified as risk factors for periodontal disease. The role of miRNAs in periodontal disease progression is not limited to the host tissues but also extends to the viruses that reside in periodontal lesions, such as herpesviruses (human herpesvirus, HHV). In advanced periodontal lesions, HHV infections result in the release of cytokines from periodontal tissues and impair antibacterial immune mechanisms that promote bacterial overgrowth. In turn, controlling the exacerbation of periodontal disease by minimizing the effect of periodontal HHV in periodontal lesions may provide novel avenues for therapeutic intervention. In summary, this review highlights multiple levels of miRNA-mediated control of periodontal disease progression, (i) through their role in periodontal inflammation and the dysregulation of homeostasis, (ii) as a regulatory target of lncRNAs, (iii) by contributing toward periodontal disease susceptibility through miRNA polymorphism, and (iv) as periodontal microflora modulators via viral miRNAs.
Collapse
Affiliation(s)
- Xianghong Luan
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA
| | - Xiaofeng Zhou
- Department of Periodontics, College of Dentistry, University of Illinois at Chicago, 801 South Paulina Street, Chicago, IL 60612, USA
| | - Pooria Fallah
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA
| | - Mirali Pandya
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA
| | - Huling Lyu
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA; Key Laboratory of Oral Medicine, Guangzhou Institute of Oral Disease, Stomatology Hospital of Guangzhou Medical University, Guangzhou 510140, China
| | - Deborah Foyle
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA
| | - Dan Burch
- Department of Pedodontics, TAMU College of Dentistry, 75246 Dallas, TX, USA
| | - Thomas G H Diekwisch
- Texas A&M Center for Craniofacial Research and Diagnosis and Department of Periodontics, TAMU College of Dentistry, 75246 Dallas, TX USA.
| |
Collapse
|
14
|
Zubeidat K, Hovav AH. Shaped by the epithelium - postnatal immune mechanisms of oral homeostasis. Trends Immunol 2021; 42:622-634. [PMID: 34083119 DOI: 10.1016/j.it.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
The first encounter of mucosal barriers with the microbiota initiates host-microbiota feedback loops instructing the tailored development of both the immune system and microbiota at each mucosal site. Once established, balanced immunological interactions enable symbiotic relationships with the microbiota in adult life. This process has been extensively investigated in the mammalian monolayer epithelium-covered intestine and lung mucosae; however, the postnatal mechanisms engaged by the oral mucosa to establish homeostasis are currently being discovered. Here, we discuss the early life dialogue between the oral mucosa and the microbiota, with particular emphasis on the central role the multilayer epithelium plays to protect the oral mucosa. These intricate and unique postnatal immunological processes shape oral homeostasis, which can potentially affect buccal and systemic health in adult life.
Collapse
Affiliation(s)
- Khaled Zubeidat
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Avi-Hai Hovav
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
15
|
Wongtim K, Ikeda E, Ohno T, Nagai S, Okuhara S, Kure K, Azuma M. Overexpression of PD-L1 in gingival basal keratinocytes reduces periodontal inflammation in a ligature-induced periodontitis model. J Periodontol 2021; 93:146-155. [PMID: 34021604 PMCID: PMC9292381 DOI: 10.1002/jper.21-0017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 01/29/2023]
Abstract
Background The immune checkpoint programmed cell death 1 (PD‐1): PD‐1 ligand 1 (PD‐L1) pathway plays a crucial role in maintaining immune tolerance and preventing tissue damages by excessive immune responses. PD‐L1 is physiologically expressed and upregulated in keratinocytes (KCs) in the oral cavity. We here investigated the contribution of PD‐L1 that was overexpressed in gingival basal KCs in a ligature‐induced periodontitis model. Methods Wild‐type (WT) BALB/c and K14/PD‐L1 transgenic (tg) mice, in which PD‐L1 was overexpressed in basal KCs under control of the keratin 14 promoter, were used. To induce periodontitis, a 9‐0 silk ligature was placed around the upper right second molar, and lipopolysaccharide from Porphyromonas gingivalis was applied on the suture. Gingival tissues were collected on day 7, after which histological analyses were performed, including by hematoxylin and eosin and tartrate‐resistant acid phosphate staining (TRAP) and quantitative PCR for proinflammatory cytokines and bone metabolism‐related genes. Alveolar bone loss at 7 weeks after ligature placement was assessed by micro‐computed tomography analysis. Results PD‐L1 was overexpressed in the basal KCs of all gingival epithelia in K14/PD‐L1tg mice. Early ligature‐induced periodontal inflammation, as assessed based on histological changes, elevation of proinflammatory cytokine (IL‐1β, IL‐6, TNF‐α) expression, periodontal ligament degeneration, and osteoclastogenesis as assessed by Rankl and Opg expression and TRAP+ cells, was markedly impaired in K14/PD‐L1tg mice. Alveolar bone resorption at a late time point was also clearly minimized in K14/PD‐L1tg mice. Conclusion Overexpression of PD‐L1 in gingival basal keratinocytes in K14/PD‐L1tg mice reduces periodontal inflammation and alveolar bone resorption in a ligature‐induced periodontitis model.
Collapse
Affiliation(s)
- Keeratika Wongtim
- Department of Molecular ImmunologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Eri Ikeda
- Department of PeriodontologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Tatsukuni Ohno
- Department of Molecular ImmunologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shigenori Nagai
- Department of Molecular ImmunologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shigeru Okuhara
- Department of Molecular Craniofacial EmbryologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Keitetsu Kure
- Department of PeriodontologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Miyuki Azuma
- Department of Molecular ImmunologyGraduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
16
|
Wald S, Leibowitz A, Aizenbud Y, Saba Y, Zubeidat K, Barel O, Koren N, Heyman O, Wilharm A, Sandrock I, Fleissig O, Tal Y, Prinz I, Aizenbud D, Chaushu S, Hovav AH. γδT Cells Are Essential for Orthodontic Tooth Movement. J Dent Res 2021; 100:731-738. [PMID: 33478315 DOI: 10.1177/0022034520984774] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sustained mechanical forces applied to tissue are known to shape local immunity. In the oral mucosa, mechanical stress, either naturally induced by masticatory forces or externally via mechanical loading during orthodontic tooth movement (OTM), is translated, in part, by T cells to alveolar bone resorption. Nevertheless, despite being considered critical for OTM, depletion of CD4+ and CD8+ T cells is reported to have no impact on tooth movement, thus questioning the function of αβT cells in OTM-associated bone resorption. To further address the role of T cells in OTM, we first characterized the leukocytes residing in the periodontal ligament (PDL), the tissue of interest during OTM, and compared it to the neighboring gingiva. Unlike the gingiva, monocytes and neutrophils represent the major leukocytes of the PDL. These myeloid cells were also the main leukocytes in the PDL of germ-free mice, although at lower levels than SPF mice. T lymphocytes were more enriched in the gingiva than the PDL, yet in both tissues, the relative fraction of the γδT cells was higher than the αβ T cells. We thus sought to examine the role of γδT cells in OTM. γδT cells residing in the PDL were mainly Vγ6+ and produced interleukin (IL)-17A but not interferon-γ. Using Tcrd-GDL mice allowing conditional ablation of γδT cells in vivo, we demonstrate that OTM was greatly diminished in the absence of γδT cells. Further analysis revealed that ablation of γδT cells decreased early IL-17A expression, monocyte and neutrophil recruitment, and the expression of the osteoclastogenic molecule receptor activator of nuclear factor-κβ ligand. This, eventually, resulted in reduced numbers of osteoclasts in the pressure site during OTM. Collectively, our data suggest that γδT cells are essential in OTM for translating orthodontic mechanical forces to bone resorption, required for relocating the tooth in the alveolar bone.
Collapse
Affiliation(s)
- S Wald
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.,Department of Orthodontics, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - A Leibowitz
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Y Aizenbud
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Y Saba
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - K Zubeidat
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - O Barel
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - N Koren
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - O Heyman
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - A Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - I Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - O Fleissig
- Department of Orthodontics, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - Y Tal
- Allergy and Clinical Immunology Unit and Internal Medicine Division, Hadassah Medical Center, Jerusalem, Israel
| | - I Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - D Aizenbud
- Department of Orthodontics and Craniofacial Anomalies, School of Graduate Dentistry, Rambam Health Care Campus and Technion Faculty of Medicine, Haifa, Israel
| | - S Chaushu
- Department of Orthodontics, Hebrew University-Hadassah Faculty of Dental Medicine, Jerusalem, Israel
| | - A H Hovav
- Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| |
Collapse
|
17
|
Freire M, Nelson KE, Edlund A. The Oral Host-Microbial Interactome: An Ecological Chronometer of Health? Trends Microbiol 2020; 29:551-561. [PMID: 33279381 DOI: 10.1016/j.tim.2020.11.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
An increasing number of studies reveal that host-microbial interactome networks are coordinated, impacting human health and disease. Recently, several lines of evidence have revealed associations between the acquisition of a complex microbiota and adaptive immunity, supporting that host-microbiota symbiotic relationships have evolved as a means to maintain homeostasis where the role of the microbiota is to promote and educate the immune system. Here, we hypothesize an oral host-microbial interactome that could serve as an ecological chronometer of health and disease, with specific focus on caries, periodontal diseases, and cancer. We also review the current state of the art on the human oral microbiome and its correlations with host innate immunity, and host cytokine control, with the goal of using this information for disease prediction and designing novel treatments for local and systemic dysbiosis. In addition, we discuss new insights into the role of novel host-microbial signals as potential biomarkers, and their relevance for the future of precision dentistry and medicine.
Collapse
Affiliation(s)
- M Freire
- Genomic Medicine group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA; Department of Infectious Diseases and Global Health, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - K E Nelson
- Genomic Medicine group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA; Genomic Medicine group, J. Craig Venter Institute, 9605 Medical Center Drive, Suite 150, Rockville, MD 20850, USA
| | - A Edlund
- Genomic Medicine group, J. Craig Venter Institute, 4120 Capricorn Lane, La Jolla, CA 92037, USA; Department of Pediatrics, University of California at San Diego, La Jolla, CA 92023, USA.
| |
Collapse
|
18
|
Medara N, Lenzo JC, Walsh KA, Holden JA, Reynolds EC, Darby IB, O'Brien-Simpson NM. Peripheral memory T-cell profile is modified in patients undergoing periodontal management. J Clin Periodontol 2020; 48:249-262. [PMID: 33131124 DOI: 10.1111/jcpe.13399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 09/23/2020] [Accepted: 10/25/2020] [Indexed: 11/28/2022]
Abstract
AIMS T-cells are known to have a role in periodontitis, however, the effect of periodontal therapy on peripheral memory T-cells is unclear. This study evaluated variation in peripheral memory T-cells and red complex bacteria in sub-gingival plaque in patients undergoing periodontal management. METHODS Peripheral blood mononuclear cells and sub-gingival plaque were collected from 54 periodontitis patients at baseline, 3-, 6- and 12-months post-therapy and 40 healthy controls. Periodontitis patients were divided into treatment outcome (TxO) groups based on prevalence of sites with probing depth ≥5 mm as good (<10% of sites), moderate (10-20%) or poor (>20%) at study conclusion. Naïve (TN -CCR7+ CD45RA+ ), central memory (TCM -CCR7+ CD45RA- ), effector memory (TEM -CCR7- CD45RA- ) and effector memory T-cells re-expressing CD45RA (TEMRA -CCR7- CD45RA+ ) were phenotyped using flow cytometry in CD4+ , CD8+ , CD4+ CD8+ and CD4- CD8- T-cells and red complex bacteria were quantified using qPCR. RESULTS At baseline, periodontitis subjects had significantly greater mean probing depths and Porphyromonas gingivalis proportions, lower TN but higher CD4+ TCM , CD8+ TCM , CD4+ CD8+ TEM and CD4- CD8- TEM cell proportions compared to health. Periodontal therapy decreased mean probing depths, P. gingivalis proportions, TEM and CD4+ and CD8+ TCM cells, but increased TN and CD4+ and CD8+ TEMRA cells. The T-cell profile in the good TxO group showed therapy-related changes in CD4+ TEM , and CD8+ TN and TEM cells, whereas, no changes were observed in the poor TxO group. CONCLUSION Management and the reduction in red complex bacteria were associated with changes in peripheral memory T-cells in periodontitis.
Collapse
Affiliation(s)
- Nidhi Medara
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia
| | - Jason C Lenzo
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia.,The Centre for Oral Health Research, The University of Melbourne, Carlton, Vic., Australia
| | | | - James A Holden
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia.,The Centre for Oral Health Research, The University of Melbourne, Carlton, Vic., Australia
| | - Eric C Reynolds
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia.,The Centre for Oral Health Research, The University of Melbourne, Carlton, Vic., Australia
| | - Ivan B Darby
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia
| | - Neil M O'Brien-Simpson
- Melbourne Dental School, The University of Melbourne, Carlton, Vic., Australia.,The Centre for Oral Health Research, The University of Melbourne, Carlton, Vic., Australia
| |
Collapse
|
19
|
Li W, Zhang Z, Wang ZM. Differential immune cell infiltrations between healthy periodontal and chronic periodontitis tissues. BMC Oral Health 2020; 20:293. [PMID: 33109155 PMCID: PMC7590666 DOI: 10.1186/s12903-020-01287-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022] Open
Abstract
Background Host immunity plays an important role against oral microorganisms in periodontitis. Methods This study assessed the infiltrating immune cell subtypes in 133 healthy periodontal and 210 chronic periodontitis tissues from Gene Expression Omnibus (GEO) datasets using the CIBERSORT gene signature files. Results Plasma cells, naive B cells and neutrophils were all elevated in periodontitis tissues, when compared to those in healthy controls. In contrast, memory B cells, resting dendritic, mast cells and CD4 memory cells, as well as activated mast cells, M1 and M2 macrophages, and follicular helper T cells, were mainly present in healthy periodontal tissues. Furthermore, these periodontitis tissues generally contained a higher proportion of activated CD4 memory T cells, while the other subtypes of T cells, including resting CD4 memory T cells, CD8 T cells, follicular helper T cells (TFH) and regulatory T cells (Tregs), were relatively lower in periodontitis tissues, when compared to healthy tissues. The ratio of dendritic and mast cells and macrophages was lower in periodontitis tissues, when compared to healthy tissues. In addition, there was a significant negative association of plasma cells with most of the other immune cells, such as plasma cells vs. memory B cells (γ = − 0.84), plasma cells vs. resting dendritic cells (γ = − 0.64), plasma cells vs. resting CD4 memory T cells (γ = 0.50), plasma cells versus activated dendritic cells (γ = − 0.46), plasma cells versus TFH (γ = − 0.46), plasma cells versus macrophage M2 cells (γ = − 0.43), or plasma cells versus macrophage M1 cells (γ = − 0.40), between healthy control and periodontitis tissues. Conclusion Plasma cells, naive B cells and neutrophils were all elevated in periodontitis tissues. The infiltration of different immune cell subtypes in the periodontitis site could lead the host immunity against periodontitis.
Collapse
Affiliation(s)
- Wei Li
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China
| | - Zheng Zhang
- Department of Periodontology, Tianjin Stomatological Hospital, Tianjin Key Laboratory of Oral Function Reconstruction, Hospital of Stomatology, Nankai University, 75th Dagu North Road, Tianjin, 300000, China.
| | - Zuo-Min Wang
- Department of Stomatology, Beijing Chao-Yang Hospital, Capital Medical University, 8th Gongti South Road, Beijing, 100020, China.
| |
Collapse
|
20
|
Möller B, Kollert F, Sculean A, Villiger PM. Infectious Triggers in Periodontitis and the Gut in Rheumatoid Arthritis (RA): A Complex Story About Association and Causality. Front Immunol 2020; 11:1108. [PMID: 32582191 PMCID: PMC7283532 DOI: 10.3389/fimmu.2020.01108] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune mediated inflammatory disease of unknown origin, which is predominantly affecting the joints. Antibodies against citrullinated peptides are a rather specific immunological hallmark of this heterogeneous entity. Furthermore, certain sequences of the third hypervariable region of human leukocyte antigen (HLA)-DR class II major histocompatibility (MHC) molecules, the so called "shared epitope" sequences, appear to promote autoantibody positive types of RA. However, MHC-II molecule and other genetic associations with RA could not be linked to immune responses against specific citrullinated peptides, nor do genetic factors fully explain the origin of RA. Consequently, non-genetic factors must play an important role in the complex interaction of endogenous and exogenous disease factors. Tobacco smoking was the first environmental factor that was associated with onset and severity of RA. Notably, smoking is also an established risk factor for oral diseases. Furthermore, smoking is associated with extra-articular RA manifestations such as interstitial lung disease in anatomical proximity to the airway mucosa, but also with subcutaneous rheumatoid nodules. In the mouth, Porphyromonas gingivalis is a periodontal pathogen with unique citrullinating capacity of foreign microbial antigens as well as candidate RA autoantigens. Although the original hypothesis that this single pathogen is causative for RA remained unproven, epidemiological as well as experimental evidence linking periodontitis (PD) with RA is rapidly accumulating. Other periopathogens such as Aggregatibacter actinomycetemcomitans and Prevotella intermedia were also proposed to play a specific immunodominant role in context of RA. However, demonstration of T cell reactivity against citrullinated, MHC-II presented autoantigens from RA synovium coinciding with immunity against Prevotella copri (Pc.), a gut microbe attracted attention to another mucosal site, the intestine. Pc. was accumulated in the feces of clinically healthy subjects with citrulline directed immune responses and was correlated with RA onset. In conclusion, we retrieved more than one line of evidence for mucosal sites and different microbial taxa to be potentially involved in the development of RA. This review gives an overview of infectious agents and mucosal pathologies, and discusses the current evidence for causality between different exogenous or mucosal factors and systemic inflammation in RA.
Collapse
Affiliation(s)
- Burkhard Möller
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Florian Kollert
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Peter M Villiger
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
21
|
Deli F, Romano F, Gualini G, Mariani GM, Sala I, Veneziano F, Bertero L, Cassoni P, Aimetti M. Resident memory T cells: Possible players in periodontal disease recurrence. J Periodontal Res 2020; 55:324-330. [DOI: 10.1111/jre.12709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 09/12/2019] [Accepted: 10/10/2019] [Indexed: 01/12/2023]
Affiliation(s)
- Federico Deli
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Federica Romano
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Giacomo Gualini
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Giulia Maria Mariani
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Irene Sala
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| | - Francesca Veneziano
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Luca Bertero
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Paola Cassoni
- Department of Medical Sciences Pathology Unit University of Turin Turin Italy
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School Section of Periodontology University of Turin Turin Italy
| |
Collapse
|
22
|
Autophagy negative-regulating Wnt signaling enhanced inflammatory osteoclastogenesis from Pre-OCs in vitro. Biomed Pharmacother 2020; 126:110093. [PMID: 32199225 DOI: 10.1016/j.biopha.2020.110093] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/24/2022] Open
Abstract
Periodontitis thereby the alveolar bone loss induced by inflammation, is a wide-spread phenomenon around the world. It is an ongoing challenge faced by clinicians worldwide. This study aimed to identify the effects of lipopolysaccharide (LPS) on osteoclasts (OCs) differentiation in vitro and to investigate its molecular mechanism. For bone marrow derived macrophages (considered as Pro-OCs), LPS impaired their differentiation into OCs in a dose-dependent manner. In contrast, it promoted Pre-OCs (referred to receptor activator of nuclear factor-κB ligand (RANKL) pretreated Pro-OCs) and differentiated to OCs with increased maximum diameter, quantity, the covering area and the fusion index in vitro. It also facilitated OCs proliferation, bone resorption and OCs related genes expression. Furthermore, it was revealed that LPS enhanced OCs genesis from Pre-OCs via activating autophagy pathway consequently elevated the accumulation of TRAP, Cts K and NFATC1, specific genes of OCs. The members of Wnt signaling were expressed as at lower states during the LPS induced OCs formation, but they could be rescued in the presence of autophagy inhibitor. The most promising observation was the direct interaction of LC3B and Dvl2, indicating that the crosstalk between above pathways existed in OCs. Taken together, we consider that LPS activates autophagy which negatively regulates Wnt signaling via autophagic degradation of Dvl2 is significant for osteoclastogenesis from Pre-OCs in vitro. Our study sheds light on the fact that autophagy inhibitors will become a new, potentially applicable therapeutic option in the treatment of periodontal bone loss.
Collapse
|
23
|
Aggregatibacter actinomycetemcomitans LtxA Hijacks Endocytic Trafficking Pathways in Human Lymphocytes. Pathogens 2020; 9:pathogens9020074. [PMID: 31973183 PMCID: PMC7168647 DOI: 10.3390/pathogens9020074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Leukotoxin (LtxA), from oral pathogen Aggregatibacter actinomycetemcomitans, is a secreted membrane-damaging protein. LtxA is internalized by β2 integrin LFA-1 (CD11a/CD18)-expressing leukocytes and ultimately causes cell death; however, toxin localization in the host cell is poorly understood and these studies fill this void. We investigated LtxA trafficking using multi-fluor confocal imaging, flow cytometry and Rab5a knockdown in human T lymphocyte Jurkat cells. Planar lipid bilayers were used to characterize LtxA pore-forming activity at different pHs. Our results demonstrate that the LtxA/LFA-1 complex gains access to the cytosol of Jurkat cells without evidence of plasma membrane damage, utilizing dynamin-dependent and presumably clathrin-independent mechanisms. Upon internalization, LtxA follows the LFA-1 endocytic trafficking pathways, as identified by co-localization experiments with endosomal and lysosomal markers (Rab5, Rab11A, Rab7, and Lamp1) and CD11a. Knockdown of Rab5a resulted in the loss of susceptibility of Jurkat cells to LtxA cytotoxicity, suggesting that late events of LtxA endocytic trafficking are required for toxicity. Toxin trafficking via the degradative endocytic pathway may culminate in the delivery of the protein to lysosomes or its accumulation in Rab11A-dependent recycling endosomes. The ability of LtxA to form pores at acidic pH may result in permeabilization of the endosomal and lysosomal membranes.
Collapse
|
24
|
Pandiyan P, Bhaskaran N, Zou M, Schneider E, Jayaraman S, Huehn J. Microbiome Dependent Regulation of T regs and Th17 Cells in Mucosa. Front Immunol 2019; 10:426. [PMID: 30906299 PMCID: PMC6419713 DOI: 10.3389/fimmu.2019.00426] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/18/2019] [Indexed: 12/19/2022] Open
Abstract
Mammals co-exist with resident microbial ecosystem that is composed of an incredible number and diversity of bacteria, viruses and fungi. Owing to direct contact between resident microbes and mucosal surfaces, both parties are in continuous and complex interactions resulting in important functional consequences. These interactions govern immune homeostasis, host response to infection, vaccination and cancer, as well as predisposition to metabolic, inflammatory and neurological disorders. Here, we discuss recent studies on direct and indirect effects of resident microbiota on regulatory T cells (Tregs) and Th17 cells at the cellular and molecular level. We review mechanisms by which commensal microbes influence mucosa in the context of bioactive molecules derived from resident bacteria, immune senescence, chronic inflammation and cancer. Lastly, we discuss potential therapeutic applications of microbiota alterations and microbial derivatives, for improving resilience of mucosal immunity and combating immunopathology.
Collapse
Affiliation(s)
- Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Natarajan Bhaskaran
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Elizabeth Schneider
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sangeetha Jayaraman
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Hamburg, Germany.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|