1
|
Lam W, Yao Y, Tang C, Wang Y, Yuan Q, Peng L. Bifunctional mesoporous HMUiO-66-NH 2 nanoparticles for bone remodeling and ROS scavenging in periodontitis therapy. Biomaterials 2025; 314:122872. [PMID: 39383779 DOI: 10.1016/j.biomaterials.2024.122872] [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: 04/08/2024] [Revised: 09/17/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Periodontal bone defects represent an irreversible consequence of periodontitis associated with reactive oxygen species (ROS). However, indiscriminate removal of ROS proves to be counterproductive for tissue repair and insufficient for addressing existing bone defects. In the treatment of periodontitis, it is crucial to rationally alleviate local ROS while simultaneously promoting bone regeneration. In this study, Zr-based large-pore hierarchical mesoporous metal-organic framework (MOF) nanoparticles (NPs) HMUiO-66-NH2 were successfully proposed as bifunctional nanomaterials for bone regeneration and ROS scavenging in periodontitis therapy. HMUiO-66-NH2 NPs demonstrated outstanding biocompatibility both in vitro and in vivo. Significantly, these NPs enhanced the osteogenic differentiation of bone mesenchymal stem cells (BMSCs) under normal and high ROS conditions, upregulating osteogenic gene expression and mitigating oxidative stress. Furthermore, in vivo imaging revealed a gradual degradation of HMUiO-66-NH2 NPs in periodontal tissues. Local injection of HMUiO-66-NH2 effectively reduced bone defects and ROS levels in periodontitis-induced C57BL/6 mice. RNA sequencing highlighted that differentially expressed genes (DEGs) are predominantly involved in bone tissue development, with notable upregulation in Wnt and TGF-β signaling pathways. In conclusion, HMUiO-66-NH2 exhibits dual functionality in alleviating oxidative stress and promoting bone repair, positioning it as an effective strategy against bone resorption in oxidative stress-related periodontitis.
Collapse
Affiliation(s)
- Waishan Lam
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yufei Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Chenxi Tang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yue Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Oral Surgery, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lin Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
2
|
Dionísio T, Brandão P, Machado V, Mendes JJ, Fonte P, Botelho J. Drug delivery systems for mouth wound healing. DRUG DELIVERY SYSTEMS FOR WOUND HEALING 2025:173-196. [DOI: 10.1016/b978-0-323-85840-3.00006-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Lou J, Zhang B, Cai J, Zhang L, Zhao Y, Zhao Z. Diabetes exacerbates periodontitis by disrupting IL-33-mediated interaction between periodontal ligament fibroblasts and macrophages. Int Immunopharmacol 2024; 147:113896. [PMID: 39740505 DOI: 10.1016/j.intimp.2024.113896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
Tissue-resident fibroblasts with immunomodulatory properties have recently been identified as key players in inflammation. However, their roles within the periodontal niche in diabetes-associated periodontitis remain unclear. Interleukin (IL)-33, known as an "alarmin" in inflammatory responses, has recently emerged as a potential contributor to periodontitis. Herein, we show that IL-33 levels are reduced in periodontal ligament fibroblasts (PDLFs) in the in vivo models of diabetes-associated periodontitis and in vitro models of diabetic inflammation. In the in vitro co-culture model, overexpression of IL-33 in PDLFs promotes M2 macrophage polarization, while knockdown of IL-33 in PDLFs instigates M1 macrophage polarization. Notably, supplementation with IL-33 in vivosignificantly alleviates periodontal tissue destruction and enhances M2 macrophage infiltration, whereas targeting the IL-33/ST2 axis exacerbates tissue damage and promotes M1 macrophage polarization in diabetes-associated periodontitis. Additionally, theCUT&RUN assay confirms the direct regulation of IL-33 by Yes-associated protein (YAP). These findings demonstrate that IL-33 deficiency in PDLFs favors M1 macrophage polarization, thereby exacerbating the pathogenesis of diabetes-associated periodontitis. Our study underscores the essential immunomodulatory role of PDLFs in creating an inflammatory niche and unveils a novel interaction axis between PDLFs and macrophages in diabetes-associated periodontitis.
Collapse
Affiliation(s)
- Jingyang Lou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jingyi Cai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Linli Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yifan Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Jeon HH, Huang X, Rojas Cortez L, Sripinun P, Lee JM, Hong JJ, Graves DT. Inflammation and mechanical force-induced bone remodeling. Periodontol 2000 2024. [PMID: 39740162 DOI: 10.1111/prd.12619] [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: 01/25/2024] [Revised: 09/25/2024] [Accepted: 10/27/2024] [Indexed: 01/02/2025]
Abstract
Periodontitis arises from imbalanced host-microbe interactions, leading to dysbiosis and destructive inflammation. The host's innate and adaptive immune responses produce pro-inflammatory mediators that stimulate destructive events, which cause loss of alveolar bone and connective tissue attachment. There is no consensus on the factors that lead to a conversion from gingivitis to periodontitis, but one possibility is the proximity of the inflammation to the bone, which promotes bone resorption and inhibits subsequent bone formation during coupled bone formation. Conversely, orthodontic tooth movement is triggered by the mechanical force applied to the tooth, resulting in bone resorption on the compression side and new bone formation on the tension side. However, the environment around orthodontic brackets readily retains dental plaque and may contribute to inflammation and bone remodeling. The immune, epithelial, stromal, endothelial and bone cells of the host play an important role in setting the stage for bone remodeling that occurs in both periodontitis and orthodontic tooth movement. Recent advancements in single-cell RNA sequencing have provided new insights into the roles and interactions of different cell types in response to challenges. In this review, we meticulously examine the functions of key cell types such as keratinocytes, leukocytes, stromal cells, osteocytes, osteoblasts, and osteoclasts involved in inflammation- and mechanical force-driven bone remodeling. Moreover, we explore the combined effects of these two conditions: mechanical force-induced bone remodeling combined with periodontal disease (chronic inflammation) and periodontally accelerated osteogenic orthodontics (acute transient inflammation). This comprehensive review enhances our understanding of inflammation- and mechanical force-induced bone remodeling.
Collapse
Affiliation(s)
- Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xin Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leticia Rojas Cortez
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Puttipong Sripinun
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Muang, Chiang Mai, Thailand
| | - Jung-Me Lee
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA
| | - Julie J Hong
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Ma S, He H, Ren X. Single-Cell and Transcriptome Analysis of Periodontitis: Molecular Subtypes and Biomarkers Linked to Mitochondrial Dysfunction and Immunity. J Inflamm Res 2024; 17:11659-11678. [PMID: 39741754 PMCID: PMC11687296 DOI: 10.2147/jir.s498739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/10/2024] [Indexed: 01/03/2025] Open
Abstract
Background Periodontitis represents an inflammatory disease with multiple contributing factors, affecting both oral and systemic health. The mechanisms linking mitochondrial dysfunction to immune responses in periodontitis remain unclear, limiting the development of individualized diagnostic and therapeutic approaches. Objective This study aims to elucidate the roles of mitochondrial dysfunction and immune responses in the pathogenesis of periodontitis, identify distinct molecular subtypes, and discover robust diagnostic biomarkers to support precision medicine approaches. Methods Single-cell RNA sequencing and transcriptome data from periodontitis patients were analyzed to identify gene signatures linked to macrophages and mitochondria. Consensus clustering was applied to classify molecular subtypes. Potential biomarkers were identified using five machine learning algorithms and validated in clinical samples through qPCR and IHC. Results Four molecular subtypes were identified: quiescent, macrophage-dominant, mitochondria-dominant, and mixed, each exhibiting unique gene expression patterns. From 13 potential biomarkers, eight were shortlisted using machine learning, and five (BNIP3, FAHD1, UNG, CBR3, and SLC25A43) were validated in clinical samples. Among them, BNIP3, FAHD1, and UNG were significantly downregulated (p < 0.05). Conclusion This study identifies novel molecular subtypes and biomarkers that elucidate the interplay between immune responses and mitochondrial dysfunction in periodontitis. These findings provide insights into the disease's heterogeneity and lay the foundation for developing non-invasive diagnostic tools and personalized therapeutic strategies.
Collapse
Affiliation(s)
- Sijia Ma
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, People’s Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, People’s Republic of China
| | - Hongbing He
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, People’s Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, People’s Republic of China
| | - Xiaobin Ren
- Department of Periodontology, Kunming Medical University School and Hospital of Stomatology, Kunming, 650106, People’s Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, People’s Republic of China
| |
Collapse
|
6
|
Ci Z, Wang H, Luo J, Wei C, Chen J, Wang D, Zhou Y. Application of Nanomaterials Targeting Immune Cells in the Treatment of Chronic Inflammation. Int J Nanomedicine 2024; 19:13925-13946. [PMID: 39735324 PMCID: PMC11682674 DOI: 10.2147/ijn.s497590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024] Open
Abstract
Chronic inflammation is a common characteristic of all kinds of diseases, including autoimmune diseases, metabolic diseases, and tumors. It is distinguished by the presence of low concentrations of inflammatory factors stimulating the body for an extended period, resulting in a persistent state of infection. This condition is manifested by the aggregation and infiltration of mononuclear cells, lymphocytes, and other immune cells, leading to tissue hyperplasia and lesions. Although various anti-inflammatory medications, including glucocorticoids and non-steroidal anti-inflammatory drugs (NSAIDs), have shown strong therapeutic effects, they lack specificity and targeting ability, and require high dosages, which can lead to severe adverse reactions. Nanoparticle drug delivery mechanisms possess the capacity to minimize the effect on healthy cells or tissues due to their targeting capabilities and sustained drug release properties. However, most nanosystems can only target the inflammatory sites rather than specific types of immune cells, leaving room for further improvement in the therapeutic effects of nanomaterials. This article reviews the current research progress on the role of diverse immune cells in inflammation, focusing on the functions of neutrophils and macrophages during inflammation. It provides an overview of the domestic and international applications of nanomaterials in targeted therapy for inflammation, aiming to establish a conceptual foundation for the utilization of nanomaterials targeting immune cells in the treatment of chronic inflammation and offer new perspectives for the avoidance and management of inflammation.
Collapse
Affiliation(s)
- Zhen Ci
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Hanchi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jiaxin Luo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Chuqiao Wei
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Jingxia Chen
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Dongyang Wang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Department of Oral Biology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, 130021, People’s Republic of China
| |
Collapse
|
7
|
Wu H, Liu Y, Wang Y, Piao Y, Meng Z, Hu X, Shi L, Shen J, Li Y. Dynamic Covalent Prodrug Nanonetworks via Reaction-Induced Self-Assembly for Periodontitis Treatment. ACS NANO 2024; 18:34884-34901. [PMID: 39663546 DOI: 10.1021/acsnano.4c12580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Periodontitis is characterized by dysbiotic biofilms, gingival inflammation, and bone resorption, highlighting the urgent need for a comprehensive approach to drug combination therapy. In this study, we introduce dynamic covalent nanonetworks (dcNNWs) synthesized through a one-pot, four-component reaction-induced self-assembly method using polyamines, 2-formylphenylboronic acid, epigallocatechin gallate, and alendronate. The formation of iminoboronate bonds drives the creation of dcNNWs, allowing controlled release in the periodontitis microenvironment. The inclusion of catechol and bisphosphonate imparts exceptional bioadhesive properties to the dcNNWs, enhancing their efficacy in preventing pathogenic bacterial biofilm formation and eliminating mature biofilms. Moreover, the dcNNWs efficiently absorb pathogen-associated molecular patterns and scavenge excess reactive oxygen species, regulating the local immune response and demonstrating anti-inflammatory effects. Additionally, the released polyphenol and alendronate from the dcNNWs alleviated inflammation and enhanced osteogenesis significantly. The detailed synergistic effects of dcNNWs in biofilm eradication, anti-inflammation, and bone remodeling, with minimal impact on healthy tissues, are confirmed in a rat model of periodontitis. With a facile synthesis process, excellent synergistic effects in periodontitis treatment, and biocompatibility, our dcNNWs present a promising and translational solution for the effective management of periodontitis.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yumeng Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University institution, Wenzhou, Zhejiang 325035, China
| | - Yinzi Piao
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Zhuojun Meng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Xiaowen Hu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University institution, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
8
|
Zhou H, Li Y, Chen X, Miao D, Zhang L, Cao R, Li Q, Liu T. Association Between Neutrophil Percentage-to-Albumin Ratio and Periodontitis: A Cross-Sectional Study. Int Dent J 2024:S0020-6539(24)01607-1. [PMID: 39710554 DOI: 10.1016/j.identj.2024.10.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/21/2024] [Accepted: 10/02/2024] [Indexed: 12/24/2024] Open
Abstract
INTRODUCTION AND AIMS Neutrophil percentage-to-albumin ratio (NPAR) is a novel biomarker of systemic inflammation. The aim of this study was to explore the relationship between NPAR and periodontitis. METHODS Data from the National Health and Nutritional Examination Survey (NHANES) between 2009 and 2014 (N = 10,128) were utilized in this cross-sectional study. Periodontitis categories were defined according to the Centres for Disease Control and Prevention and American Academy of Periodontology (CDC/AAP) classification. The NPAR was calculated by dividing the neutrophil percentage by serum albumin. Covariates included age, sex, race, education level, annual household income, marital status, smoking status, BMI, recreational activity, work activity, diabetes mellitus, hypertension, and cardiovascular disease. Weighted logistic regression analysis was conducted to investigate the linkage between NPAR and moderate/severe periodontitis, and weighted linear regression analysis was performed to explore the relationship of NPAR with mean attachment loss (AL) and mean probing pocket depth (PPD). RESULTS Our analysis revealed a positive linear relationship between NPAR and periodontitis. Specifically, we found that the risk of moderate/severe periodontitis increased by 12% for each standard deviation increase in NPAR. Individuals in the highest tertile of NPAR were 28% more likely to have periodontitis compared to those in the lowest tertile (ORtertile3vs1 = 1.28, 95% CI: 1.10-1.49). These findings were consistent across different subgroups analysed. Furthermore, our study demonstrated that NPAR was also positively correlated with mean AL and PPD, which are key indicators of periodontal health. CONCLUSIONS Our results suggest that NPAR is significantly linked to poor periodontal health. However, owing to the cross-section design of this study, additional longitudinal studies are necessary to further enhance our comprehension of the impact of NPAR on periodontal status. CLINICAL RELEVANCE Elevated neutrophil counts and low albumin levels correlate with moderate/severe periodontitis. Monitoring these markers may aid in assessing periodontitis risk.
Collapse
Affiliation(s)
- Huan Zhou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yong Li
- Department of Stomatology, Hunan University of Medicine, Hunan, China
| | - Xin Chen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Di Miao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China; Department of Periodontology, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, Xi'an Children's Hospital, Xi'an, China
| | - Ruoyan Cao
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Qiulan Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, China.
| | - Tangsheng Liu
- Department of Stomatology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, Hubei, China.
| |
Collapse
|
9
|
Overmiller AM, Uchiyama A, Hope ED, Nayak S, O'Neill CG, Hasneen K, Chen YW, Naz F, Dell'Orso S, Brooks SR, Jiang K, Morasso MI. Reprogramming of epidermal keratinocytes by PITX1 transforms the cutaneous cellular landscape and promotes wound healing. JCI Insight 2024; 9:e182844. [PMID: 39480496 DOI: 10.1172/jci.insight.182844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
Cutaneous wound healing is a slow process that often terminates with permanent scarring while oral wounds, in contrast, regenerate after damage faster. Unique molecular networks in epidermal and oral epithelial keratinocytes contribute to the tissue-specific response to wounding, but key factors that establish those networks and how the keratinocytes interact with their cellular environment remain to be elucidated. The transcription factor PITX1 is highly expressed in the oral epithelium but is undetectable in cutaneous keratinocytes. To delineate if PITX1 contributes to oral keratinocyte identity, cell-cell interactions, and the improved wound healing capabilities, we ectopically expressed PITX1 in the epidermis of murine skin. Using comparative analysis of murine skin and oral (buccal) mucosa with single-cell RNA-Seq and spatial transcriptomics, we found that PITX1 expression enhances epidermal keratinocyte migration and proliferation and alters differentiation to a quasi-oral keratinocyte state. PITX1+ keratinocytes reprogrammed intercellular communication between skin-resident cells to mirror buccal tissue while stimulating the influx of neutrophils that establish a pro-inflammatory environment. Furthermore, PITX1+ skin healed significantly faster than control skin via increased keratinocyte activation and migration and a tunable inflammatory environment. These results illustrate that PITX1 programs oral keratinocyte identity and cellular interactions while revealing critical downstream networks that promote wound closure.
Collapse
Affiliation(s)
- Andrew M Overmiller
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Akihiko Uchiyama
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Emma D Hope
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Subhashree Nayak
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Christopher G O'Neill
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Kowser Hasneen
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Yi-Wen Chen
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC, USA
| | | | | | - Stephen R Brooks
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Kan Jiang
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Maria I Morasso
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Jing L, Wang HY, Zhang N, Zhang WJ, Chen Y, Deng DK, Li X, Chen FM, He XT. Critical roles of extracellular vesicles in periodontal disease and regeneration. Stem Cells Transl Med 2024:szae092. [PMID: 39703170 DOI: 10.1093/stcltm/szae092] [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: 09/22/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
Extracellular vesicles (EVs) are evolutionarily conserved communication mediators that play key roles in the development of periodontal disease as well as in regeneration processes. This concise review first outlines the pathogenic mechanisms through which EVs derived from bacteria lead to the progression of periodontitis, with a focus on the enrichment of virulence factors, the amplification of immune responses, and the induction of bone destruction as key aspects influenced by bacterial EVs. This review aims to elucidate the positive effects of EVs derived from mesenchymal stem cells (MSC-EVs) on periodontal tissue regeneration. In particular, the anti-inflammatory properties of MSC-EVs and their impact on the intricate interplay between MSCs and various immune cells, including macrophages, dendritic cells, and T cells, are described. Moreover, recent advancements regarding the repair-promoting functions of MSC-EVs are detailed, highlighting the mechanisms underlying their ability to promote osteogenesis, cementogenesis, angiogenesis, and the homing of stem cells, thus contributing significantly to periodontal tissue regeneration. Furthermore, this review provides insights into the therapeutic efficacy of MSC-EVs in treating periodontitis within a clinical context. By summarizing the current knowledge, this review aims to provide a comprehensive understanding of how MSC-EVs can be harnessed for the treatment of periodontal diseases. Finally, a discussion is presented on the challenges that lie ahead and the potential practical implications for translating EV-based therapies into clinical practices for the treatment of periodontitis.
Collapse
Affiliation(s)
- Lin Jing
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Hong-Yu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Ning Zhang
- Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi'an 710032, People's Republic of China
| | - Wen-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Yuzhe Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Dao-Kun Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Fa-Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | - Xiao-Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| |
Collapse
|
11
|
Hasuike A, Easter QT, Clark D, Byrd KM. Application of Single-Cell Genomics to Animal Models of Periodontitis and Peri-Implantitis. J Clin Periodontol 2024. [PMID: 39695834 DOI: 10.1111/jcpe.14093] [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: 09/06/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
AIMS This narrative review aims to synthesize current knowledge on integrating single-cell genomics technologies with animal models of periodontitis and peri-implantitis. REVIEW Single-cell RNA sequencing (scRNAseq) reveals cellular heterogeneity and specific cell roles in periodontitis and peri-implantitis, overcoming the limitations of bulk RNA sequencing. Under controlled conditions and genetic manipulation, animal models facilitate studying disease progression, gene functions and systemic disease links, aiding targeted therapy development. Knockout models have started to elucidate the impact of genetic mutations on periodontal disease and host responses. scRNAseq in animal models has been used to examine connections between periodontitis and systemic diseases, revealing altered immune environments and cellular interactions. Emerging studies are now applying these methods to animal models of peri-implantitis. Integrating these datasets into single-cell and spatially resolved atlases will enable future meta-analyses, providing deeper insights into disease mechanisms considering factors such as sex, strain, and age. CONCLUSIONS Integrating scRNAseq with animal models advances the understanding of periodontitis and peri-implantitis pathogenesis and precision therapies. The combined use of single-cell and spatial genomics and scRNAseq will further enhance data insights significantly for drug discovery and preclinical testing, making these technologies pivotal in validating animal models and translating findings into clinical practice.
Collapse
Affiliation(s)
- Akira Hasuike
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, Maryland, USA
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Quinn T Easter
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, Maryland, USA
- Department of Oral and Craniofacial Molecular Biology, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| | - Daniel Clark
- Department of Periodontics and Preventive Dentistry, University of Pittsburgh School of Dental Medicine, Pittsburgh, Pennsylvania, USA
| | - Kevin M Byrd
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, Maryland, USA
- Department of Oral and Craniofacial Molecular Biology, Virginia Commonwealth University School of Dentistry, Richmond, Virginia, USA
| |
Collapse
|
12
|
Li Z, Fan X, Liu Y, Yue M, Wu T, Wang X, Jiang W, Fan K. Engineering Mild-Photothermal Responsive and NO Donor Prussian Blue Nanozymes Using Mild Synthesis for Inflammation Regulation and Bacterial Eradication in Periodontal Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024:e2409840. [PMID: 39690880 DOI: 10.1002/adma.202409840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Periodontitis, an infectious disease of periodontal tissues caused by oral bacterial biofilms, is characterized by reactive oxygen species (ROS) accumulation and immune microenvironment imbalance. Multifunctional nanozymes, leveraging their physiochemical properties and enzymatic activities, offer promising antibacterial and anti-inflammatory strategies for managing periodontitis. In particular, Prussian blue nanozymes (PBzymes) exhibit exceptional ROS control due to their robust catalytic activity, diverse antioxidant functions, and high biocompatibility. However, the practical application of traditional high-temperature synthesis methods is limited. This study introduces a class of metal-engineered PBzymes synthesized at room temperature, identified for their potent antioxidative activity and excellent photothermal performance at mild temperatures. Nitric oxide (NO) gas therapy offers promising strategies for targeting deep infections in periodontal tissues. Thus, sodium nitroprusside is introduced into PBzyme to create SPBzyme via an in situ loading method. NO release by SPBzyme enhances antibacterial effects and overcomes resistance linked to bacterial biofilms, resulting in mild-photothermal antibacterial properties and synergistic antioxidant effects. In vitro antibacterial assays demonstrate the superior efficacy of SPBzyme under mild temperature conditions and near-infrared light exposure. Furthermore, SPBzyme effectively reduces inflammation and has positive therapeutic effects in periodontal animal models. Overall, mild-temperature photothermal NO release nanozyme therapy represents a novel approach for treating periodontitis.
Collapse
Affiliation(s)
- Zheng Li
- Department of Prosthodontics, National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, 22 Zhongguancun South Avenue, Beijing, Haidian District, 100081, P. R. China
| | - Xiaowan Fan
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, P. R. China
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, P. R. China
| | - Ying Liu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Muxin Yue
- Institute of Medical Technology, Peking University Health Science Center, 38 Xueyuan Road, Beijing, Haidian District, 100191, P. R. China
| | - Tingting Wu
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, P. R. China
| | - Wei Jiang
- Academy of Medical Sciences, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Kelong Fan
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Biomacromolecules (CAS), CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
- Nanozyme Laboratory in Zhongyuan, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, 451163, P. R. China
| |
Collapse
|
13
|
Irwandi RA, Marruganti C, Collins G, Carvalho JDS, Gilroy D, D’Aiuto F. The translational potential of inflammation-induced skin blister human models in exploring the pathogenesis of periodontitis and its systemic health implications. Front Immunol 2024; 15:1469828. [PMID: 39737182 PMCID: PMC11682961 DOI: 10.3389/fimmu.2024.1469828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Periodontitis is a highly prevalent chronic disease. Despite decades of extensive research on the topic, a complete understanding of its immunopathogenesis, especially when linked to other inflammatory comorbidities, is lacking. Ex vivo human and in vivo animal experiments have shown the host inflammatory response's crucial role in both the disease's onset and its systemic implications. These approaches, however, remain questionable when translating these findings into real-world scenarios linked to periodontitis. A clear need for new in vivo human models is discussed, especially within the context of understanding the host response to key pathogens linked to periodontitis, such as Porphyromonas gingivalis (P. gingivalis). Therefore, a skin blister model was employed to describe the stages of the host immune response in humans after challenges by microbial and/or sterile insults. A novel human challenge model using UV-killed P. gingivalis holds promise in producing new evidence and bridging the gap of the host response to periodontitis and its links with other common chronic diseases.
Collapse
Affiliation(s)
- Rizky Aditya Irwandi
- Periodontology Unit, UCL Eastman Dental Institute, University College London, London, United Kingdom
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Crystal Marruganti
- Periodontology Unit, UCL Eastman Dental Institute, University College London, London, United Kingdom
- Unit of Periodontology, Endodontology and Restorative Dentistry, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - George Collins
- Department of Ageing, Rheumatology and Regenerative Medicine, Division of Medicine, University College London, London, United Kingdom
- Department of Cardiology, St Bartholomew’s Hospital, Barts Health NHS Trust, London, United Kingdom
| | - Jhonatan de Souza Carvalho
- Department of Ageing, Rheumatology and Regenerative Medicine, Division of Medicine, University College London, London, United Kingdom
- Department of Diagnosis and Surgery, São Paulo State University (UNESP), School of Dentistry, Araraquara, Brazil
| | - Derek Gilroy
- Department of Ageing, Rheumatology and Regenerative Medicine, Division of Medicine, University College London, London, United Kingdom
| | - Francesco D’Aiuto
- Periodontology Unit, UCL Eastman Dental Institute, University College London, London, United Kingdom
| |
Collapse
|
14
|
Kang S, Liu S, Dong X, Li H, Qian Y, Dai A, He W, Li X, Chen Q, Wang H, Ding PH. USP4 depletion-driven RAB7A ubiquitylation impairs autophagosome-lysosome fusion and aggravates periodontitis. Autophagy 2024:1-18. [PMID: 39663592 DOI: 10.1080/15548627.2024.2429371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 11/02/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Periodontitis, a prevalent and chronic inflammatory disease, is intricately linked with macroautophagy/autophagy, which has a dual role in maintaining periodontal homeostasis. Despite its importance, the precise interplay between autophagy and periodontitis pathogenesis remains to be fully elucidated. In this study, our investigation revealed that the ubiquitination of RAB7A, mediated by reduced levels of the deubiquitinating enzyme USP4 (ubiquitin specific peptidase 4), disrupts normal lysosomal trafficking and autophagosome-lysosome fusion, thereby contributing significantly to periodontitis progression. Specifically, through genomic and histological analysis of clinical gingival samples, we observed a decreased RAB7A expression and impaired autophagic activity in periodontitis. This was further substantiated through experimental periodontitis mice, where RAB7A inactivation was shown to directly affect autophagy efficiency and drive periodontitis progression. Next, we explored the function of active RAB7A to promote lysosomal trafficking dynamics and autophagosome-lysosome fusion, which was inhibited by RAB7A ubiquitination in macrophages stimulated by Porphyromonas gingivalis (P. g.), one of the keystone pathogens of periodontitis. Last, by proteomics analysis, we revealed that the ubiquitination of RAB7A was mediated by USP4 and validated that upregulation of USP4 could attenuate periodontitis in vivo. In conclusion, these findings highlight the interaction between USP4 and RAB7A as a promising target for therapeutic intervention in managing periodontal diseases.Abbreviation: 3-MA: 3-methyladenine; Baf A1:bafilomycin A1; BECN1: beclin 1, autophagy related; CEJ-ABC: cementoenamel junctionto alveolar bone crest; IL1B/IL-1β: interleukin 1 beta; KD:knockdown; LPS: lipopolysaccharide; MOI: multiplicity of infection;OE: overexpression; P.g.: Porphyromonasgingivalis; RILP: Rabinteracting lysosomal protein; ScRNA-seq: single-cell RNA sequencing; SQSTM1/p62: sequestosome 1; S.s.: Streptococcus sanguinis; USP4:ubiquitin specific peptidase 4.
Collapse
Affiliation(s)
- Sen Kang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuxin Liu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xian Dong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyu Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuanyi Qian
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Anna Dai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wentao He
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Li
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qianming Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| | - Pei-Hui Ding
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
15
|
Seubert AC, Krafft M, Bopp S, Helal M, Bhandare P, Wolf E, Alemany A, Riedel A, Kretzschmar K. Spatial transcriptomics reveals molecular cues underlying the site specificity of the adult mouse oral mucosa and its stem cell niches. Stem Cell Reports 2024; 19:1706-1719. [PMID: 39547226 DOI: 10.1016/j.stemcr.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
The oral cavity is a multifunctional organ composed of structurally heterogeneous mucosal tissues that remain poorly characterized. Oral mucosal tissues are highly stratified and segmented along an epithelial-lamina propria axis. Here, we performed spatial transcriptomics (tomo-seq) on the tongue, cheeks, and palate of the adult mouse to understand the cues that maintain the oral mucosal sites. We define molecular markers of unique and shared cellular niches and differentiation programs across oral sites. Using a comparative approach, we identify fibroblast growth factor (FGF) pathway components as potential stem cell niche factors for oral epithelial stem cells. Using organoid-forming efficiency assays, we validated three FGF ligands (FGF1, FGF7, and FGF10) as site-specific niche factors in the dorsal and ventral tongue. Our dataset of the spatially resolved genes across major oral sites represents a comprehensive resource for unraveling the molecular mechanisms underlying the adult homeostasis of the oral mucosa and its stem cell niches.
Collapse
Affiliation(s)
- Anna C Seubert
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | - Marion Krafft
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany
| | - Sarah Bopp
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany
| | - Moutaz Helal
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | | | - Elmar Wolf
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Anna Alemany
- Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, the Netherlands; Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden, the Netherlands
| | - Angela Riedel
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany
| | - Kai Kretzschmar
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research Würzburg, IZKF/MSNZ, University Hospital Würzburg, Würzburg, Germany; Graduate School of Life Sciences (GSLS), University of Würzburg, Würzburg, Germany.
| |
Collapse
|
16
|
Ma Y, Jia R, Chen S, Ma J, Yin L, Pan X, He Y, Wu T, Zhao Z, Ma L, Wu S, Wang H, Liang G, Huang S, Sun X. Ubiquitin‐Proteasome System in Periodontitis: Mechanisms and Clinical Implications. Cell Prolif 2024. [DOI: 10.1111/cpr.13781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/09/2024] [Indexed: 01/03/2025] Open
Abstract
ABSTRACTThe progression of periodontitis, a bacteria‐driven inflammatory and bone‐destructive disease, involves myriad cellular and molecular mechanisms. Protein regulation significantly influences the pathogenesis and management of periodontitis. However, research regarding its regulatory role in periodontitis remains relatively limited. The ubiquitin‐proteasome system (UPS), which mainly involves ubiquitination by E3 ubiquitin ligases (E3s) and deubiquitination by deubiquitinating enzymes (DUBs), is the primary intracellular and non‐lysosomal mechanism of protein degradation. Recent studies have provided compelling evidence to support the involvement of UPS in periodontitis progression. Increasing evidence indicated that E3s, such as CUL3, Nedd4‐2, Synoviolin, FBXL19, PDLIM2, TRIMs and TRAFs, modulate inflammatory responses and bone resorption in periodontitis through multiple classical signalling pathways, including NLRP3, GSDMD, NF‐κB, Wnt/β‐catenin and Nrf2. Meanwhile, DUBs, including OTUD1, A20, CYLD, UCH‐L1 and USPs, also broadly modulate periodontitis progression by regulating signalling pathways such as NF‐κB, Wnt/β‐catenin, NLRP3, and BMP2. Therefore, the modulation of E3s and DUBs has proven to be an effective therapy against periodontitis. This review provides a comprehensive overview of the regulatory role of ubiquitinating and deubiquitinating enzymes in periodontitis progression and the underlying mechanisms. Finally, we summarise several chemical and genetic methods that regulate UPS enzymes and pave the way for the development of targeted therapies for periodontitis.
Collapse
Affiliation(s)
- Yilin Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Ruiwei Jia
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shuhong Chen
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Jun Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lei Yin
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xingbei Pan
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Yunuo He
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Tong Wu
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Lulu Ma
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Shengzhuang Wu
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Huining Wang
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Guang Liang
- Institute of Stomatology, School and Hospital of Stomatology Hangzhou Medical College Zhejiang Hangzhou China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Prosthodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| | - Xiaoyu Sun
- Institute of Stomatology, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
- Department of Periodontics, School and Hospital of Stomatology Wenzhou Medical University Wenzhou China
| |
Collapse
|
17
|
Martin K, Mianecki M, Maglaras V, Sheikh A, Saleh MHA, Decker AM, Decker JT. Neutrophils drive sexual dimorphism in experimental periodontitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625678. [PMID: 39677749 PMCID: PMC11642827 DOI: 10.1101/2024.11.27.625678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The motivating premise of this study is to improve the treatment of periodontal disease by elucidating sex-specific mechanisms of periodontal disease progression. Men and women experience inflammation in fundamentally different ways and understanding the sex-specific biology leading to inflammation and bone loss in the periodontium will inevitably improve patient outcomes. We therefore examined clinical and immunological differences in the progression of periodontal disease using the ligature-induced periodontitis model. Periodontitis was induced in male and female C57BL/6j mice by tying a 5-0 silk suture around the left maxillary second molar. The ligature was left on for 7 or 21 days at which point maxillae were characterized for bone loss by μCT or immune infiltrate by flow cytometry. Neutrophil depletion was accomplished through systemic administration of a Ly6G antibody. Conditions were compared using two-way ANOVA with Tukey's multiple comparison correction from n≥5 animals. Ligature-induced periodontitis led to alveolar bone loss at both 7 and 21 days in both female and male mice. Males and females had approximately the same amount of linear bone loss 7 days post-ligature placement, while male mice had significantly more linear bone loss by day 21. Male mice had significantly more immune cells in their maxillae 7 days post ligature placement compared to female mice. Both male and female mice showed a shift in immune populations towards neutrophils, with no significant difference between males and females. Neutrophil counts were significantly elevated in male mice on day 7 but not day 21, while female mice did not have any statistically significant changes in neutrophil counts. Neutrophil depletion using a Ly6G antibody limited bone loss in male but not female mice relative to isotype antibody-treated controls. Analysis of single-cell sequencing data from human patients with periodontitis showed differences in neutrophil phenotypes that were also observed in a mouse model of periodontitis. Together, these data suggest a mechanistic role for neutrophil inflammation in sexual dimorphism in periodontitis.
Collapse
Affiliation(s)
- Kelsey Martin
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Maxwell Mianecki
- Department of Cariology, Restorative Sciences, and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Victoria Maglaras
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
- Department of Cariology, Restorative Sciences, and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Asfandyar Sheikh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Muhammad H A Saleh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Ann M Decker
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| | - Joseph T Decker
- Department of Cariology, Restorative Sciences, and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, 48109
| |
Collapse
|
18
|
Wang Q, Lin W, Lei K, Wang H, Zhang X, Jiang S, Zhang D, Wang W, Cao S, Li Y, Yu B, Wang Y, Yin Q, Yuan Q. Hyperglycemia-Enhanced Neutrophil Extracellular Traps Drive Mucosal Immunopathology at the Oral Barrier. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407346. [PMID: 39499780 DOI: 10.1002/advs.202407346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/15/2024] [Indexed: 11/07/2024]
Abstract
Type 2 diabetes (T2D) is a risk factor for mucosal homeostasis and enhances the susceptibility to inflammation, in which neutrophils have been increasingly appreciated for their role. Here, barrier disruption and inflammation are observed at oral mucosa (gingiva) of T2D patients and mice. It is demonstrated that neutrophils infiltrate the gingival mucosa of T2D mice and expel obvious neutrophil extracellular traps (NETs), while removal of NETs alleviates the disruption of mucosal barrier. Mechanistically, gingival neutrophils released NETs are dependent of their metabolic reprogramming. Under hyperglycemic condition, neutrophils elevate both glucose incorporation and glycolysis via increased expression of GLUT1. Moreover, significantly increased levels of NETs are observed in local gingival lesions of patients, which are associated with clinical disease severity. This work elucidates a causative link between hyperglycemia and oral mucosal immunopathology, mediated by the altered immuno-metabolic axis in neutrophil, thereby suggesting a potential therapeutic strategy.
Collapse
Affiliation(s)
- Qian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Prosthodontics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Weimin Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Kexin Lei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Xiaohan Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shuang Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Danting Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wen Wang
- Hebei Key Laboratory of Stomatology, Hebei Clinical Research Center for Oral Diseases, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Shuqin Cao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yuyu Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Bo Yu
- Division of Preventive and Restorative Sciences, School of Dentistry, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuan Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qi Yin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
19
|
Guo S, Fu L, Yin C, Shao W, Sun Q, Chen L, Xia T, Wang M, Xia H. ROS-Induced Gingival Fibroblast Senescence: Implications in Exacerbating Inflammatory Responses in Periodontal Disease. Inflammation 2024; 47:1918-1935. [PMID: 38630168 DOI: 10.1007/s10753-024-02014-5] [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: 01/16/2024] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 11/30/2024]
Abstract
Periodontal disease is the pathological outcome of the overwhelming inflammation in periodontal tissue. Cellular senescence has been associated with chronic inflammation in several diseases. However, the role of cellular senescence in the pathogenesis of periodontal disease remained unclear. This study aimed to investigate the role and the mechanism of cellular senescence in periodontal disease. Using single-cell RNA sequencing, we first found the upregulated level of cellular senescence in fibroblasts and endothelial cells from inflamed gingival tissue. Subsequently, human gingival fibroblasts isolated from healthy and inflamed gingival tissues were labeled as H-GFs and I-GFs, respectively. Compared to H-GFs, I-GFs exhibited a distinct cellular senescence phenotype, including an increased proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells, enlarged cell morphology, and significant upregulation of p16INK4A expression. We further observed increased cellular reactive oxygen species (ROS) activity, mitochondrial ROS, and DNA damage of I-GFs. These phenotypes could be reversed by ROS scavenger NAC, which suggested the cause of cellular senescence in I-GFs. The migration and proliferation assay showed the decreased activity of I-GFs while the gene expression of senescence-associated secretory phenotype (SASP) factors such as IL-1β, IL-6, TGF-β, and IL-8 was all significantly increased. Finally, we found that supernatants of I-GF culture induced more neutrophil extracellular trap (NET) formation and drove macrophage polarization toward the CD86-positive M1 pro-inflammatory phenotype. Altogether, our findings implicate that, in the inflamed gingiva, human gingival fibroblasts acquire a senescent phenotype due to oxidative stress-induced DNA and mitochondrial damage, which in turn activate neutrophils and macrophages through the secretion of SASP factors.
Collapse
Affiliation(s)
- Shuling Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Liangliang Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Chenghu Yin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wenjun Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Quan Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Liangwen Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Ting Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Min Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| | - Haibin Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
20
|
Haller J, Abedi N, Hafedi A, Shehab O, Wietecha MS. Spatial Transcriptomics Unravel the Tissue Complexity of Oral Pathogenesis. J Dent Res 2024; 103:1331-1339. [PMID: 39382116 DOI: 10.1177/00220345241271934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024] Open
Abstract
Spatial transcriptomics (ST) is a cutting-edge methodology that enables the simultaneous profiling of global gene expression and spatial information within histological tissue sections. Traditional transcriptomic methods lack the spatial resolution required to sufficiently examine the complex interrelationships between cellular regions in diseased and healthy tissue states. We review the general workflows for ST, from specimen processing to ST data analysis and interpretations of the ST dataset using visualizations and cell deconvolution approaches. We show how recent studies used ST to explore the development or pathogenesis of specific craniofacial regions, including the cranium, palate, salivary glands, tongue, floor of mouth, oropharynx, and periodontium. Analyses of cranial suture patency and palatal fusion during development using ST identified spatial patterns of bone morphogenetic protein in sutures and osteogenic differentiation pathways in the palate, in addition to the discovery of several genes expressed at critical locations during craniofacial development. ST of salivary glands from patients with Sjögren's disease revealed co-localization of autoimmune antigens with ductal cells and a subpopulation of acinar cells that was specifically depleted by the dysregulated autoimmune response. ST of head and neck lesions, such as premalignant leukoplakia progressing to established oral squamous cell carcinomas, oral cancers with perineural invasions, and oropharyngeal lesions associated with HPV infection spatially profiled the complex tumor microenvironment, showing functionally important gene signatures of tumor cell differentiation, invasion, and nontumor cell dysregulation within patient biopsies. ST also enabled the localization of periodontal disease-associated gene expression signatures within gingival tissues, including genes involved in inflammation, and the discovery of a fibroblast subtype mediating the transition between innate and adaptive immune responses in periodontitis. The increased use of ST, especially in conjunction with single-cell analyses, promises to improve our understandings of craniofacial development and pathogenesis at unprecedented tissue-level resolution in both space and time.
Collapse
Affiliation(s)
- J Haller
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - N Abedi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - A Hafedi
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - O Shehab
- Department of Periodontics, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| | - M S Wietecha
- Department of Oral Biology, College of Dentistry, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
21
|
Li J, Ye LJ, Dai YW, Wang HW, Gao J, Shen YH, Wang F, Dai QG, Wu YQ. Single-cell analysis reveals a unique microenvironment in peri-implantitis. J Clin Periodontol 2024; 51:1665-1676. [PMID: 38566468 PMCID: PMC11651718 DOI: 10.1111/jcpe.13982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 01/31/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
AIM This study aimed to reveal the unique microenvironment of peri-implantitis through single-cell analysis. MATERIALS AND METHODS Herein, we performed single-cell RNA sequencing (scRNA-seq) of biopsies from patients with peri-implantitis (PI) and compared the results with healthy individuals (H) and patients with periodontitis (PD). RESULTS Decreased numbers of stromal cells and increased immune cells were found in the PI group, which implies a severe inflammatory infiltration. The fibroblasts were found to be heterogeneous and the specific pro-inflammatory CXCL13+ sub-cluster was more represented in the PI group, in contrast to the PD and H groups. Furthermore, more neutrophil infiltration was detected in the PI group than in the PD group, and cell-cell communication and ligand-receptor pairs revealed most neutrophils were recruited by CXCL13+ fibroblasts through CXCL8/CXCL6-CXCR2/CXCR1. Notably, our study demonstrated that the unique microenvironment of the PI group promoted the differentiation of monocyte/macrophage lineage cells into osteoclasts, which might explain the faster and more severe bone resorption in the progression of PI than PD. CONCLUSIONS Collectively, this study suggests a unique immune microenvironment of PI, which may explain the differences between PI and PD in the clinic. These outcomes will aid in finding new specific and effective treatments for PI.
Collapse
Affiliation(s)
- J. Li
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - L. J. Ye
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - Y. W. Dai
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - H. W. Wang
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - J. Gao
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - Y. H. Shen
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - F. Wang
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| | - Q. G. Dai
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
- Department of Stomatology, Zhang Zhiyuan Academician Work StationHainan, Western Central HospitalDanzhouHainanChina
| | - Y. Q. Wu
- Department of Second Dental Center, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong UniversityShanghaiChina
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of StomatologyShanghaiChina
| |
Collapse
|
22
|
Xin TY, Zhu ZL, Chen LJ, Han B, Yang RL, Shi J. Profile of Inflammatory Cytokines in Gingival Crevicular Fluid and Plasma in Patients With Grade C Periodontitis During Orthodontic Treatment: A Longitudinal Case Series Report. Orthod Craniofac Res 2024. [PMID: 39589230 DOI: 10.1111/ocr.12880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/27/2024]
Abstract
AIM To examine the immune responses in patients diagnosed as grade C periodontitis during orthodontic treatment. MATERIALS AND METHODS Our study included seven orthodontic patients with grade C periodontitis and measured their levels of inflammatory cytokines in gingival crevicular fluid and plasma before orthodontic treatment, during the alignment and levelling phase, and during the detailing and finishing phase. The key signal pathways in the orthodontic process of patients with periodontitis were detected by KEGG analysis. RESULTS Studies have shown that orthodontic treatment brings great improvement to patients with grade C periodontitis, and most of the local/systemic inflammatory cytokines can be reduced after orthodontic treatment. Simultaneously, orthodontic treatment can reduce the percentage of IFN-γ+ Th1 cells in patients with grade C periodontitis. Through KEGG analysis, the IL-17 signalling pathway and TNF signalling pathway are closely interrelated in the orthodontic treatment of patients diagnosed with grade C periodontitis (p-value < 0.001). CONCLUSION Orthodontic treatment can effectively control the local and system levels of inflammation in patients with grade C periodontitis, with IL-17A and TNF-α as potential distinctive inflammatory markers for orthodontic-periodontal combined treatment in individuals with periodontitis.
Collapse
Affiliation(s)
- Tian-Yi Xin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Zi-Lu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Liu-Jing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Bin Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Rui-Li Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| | - Jie Shi
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, People's Republic of China
- Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, People's Republic of China
| |
Collapse
|
23
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Kleb SS, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Fibroblast-Mediated Macrophage Recruitment Supports Acute Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02956-7. [PMID: 39581458 DOI: 10.1016/j.jid.2024.10.609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single-nuclei RNA sequencing, we defined changes in gene expression associated with inflammation 1 day after wounding in mouse skin. Compared with those in keratinocytes and myeloid cells, we detected enriched expression of proinflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL-33, compared with SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound-bed macrophages and monocytes during injury-induced inflammation, with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M Amuso
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - MaryEllen R Haas
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Paula O Cooper
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Sana Hafiz
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Shatha Salameh
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Miguel F Mazumder
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Violet Josephson
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Sarah S Kleb
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Khatereh Khorsandi
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Anelia Horvath
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Brett A Shook
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA; The Department of Dermatology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA.
| |
Collapse
|
24
|
Kim WS, Prasongyuenyong K, Ko A, Debnath R, Chen Z, Zhou JX, Shaaf E, Ko KI. ICAM1 + gingival fibroblasts modulate periodontal inflammation to mitigate bone loss. Front Immunol 2024; 15:1484483. [PMID: 39650645 PMCID: PMC11621011 DOI: 10.3389/fimmu.2024.1484483] [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: 08/21/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Tissue-resident fibroblasts are heterogeneous and provide an endogenous source of cytokines that regulate immunologic events in many osteolytic diseases. Identifying distinct inflammatory fibroblast subsets and conducting mechanistic in vivo studies are critical for understanding disease pathogenesis and precision therapeutics, which is poorly explored in periodontitis. Here, we surveyed published single-cell datasets for fibroblast-specific analysis and show that Intercellular Adhesion Molecule-1 (ICAM1) expression selectively defines a fibroblast subset that exhibits an inflammatory transcriptional profile associated with nuclear factor-κB (NF-κB) pathway. ICAM1+ fibroblasts expand in both human periodontitis and murine ligature-induced periodontitis model, which have upregulated expression of CCL2 and CXCL1 compared to other fibroblast populations. Using a mouse model to selectively target gingival stromal cells, we further show that disruption of an inflammatory pathway by inhibiting transcriptional activity of NF-κB in these cells accelerated periodontal bone loss. Mechanistically, this was linked to a reduction of CCL2 expression by the ICAM1+ fibroblasts, leading to impaired macrophage recruitment and efferocytosis that was associated with persistent neutrophilic inflammation. These results may have a significant therapeutic implication as ICAM1+ gingival fibroblasts exert a protective response by regulating innate immune responses that are needed for the controlled inflammatory events in early stages of periodontitis.
Collapse
Affiliation(s)
- William S. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kawintip Prasongyuenyong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Annette Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rahul Debnath
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhaoxu Chen
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan X. Zhou
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emon Shaaf
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kang I. Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
25
|
Hickman HD, Moutsopoulos NM. Viral infection and antiviral immunity in the oral cavity. Nat Rev Immunol 2024:10.1038/s41577-024-01100-x. [PMID: 39533045 DOI: 10.1038/s41577-024-01100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Individual tissues have distinct antiviral properties garnered through various mechanisms, including physical characteristics, tissue-resident immune cells and commensal organisms. Although the oral mucosa has long been appreciated as a critical barrier tissue that is exposed to a continuous barrage of pathogens, many fundamental aspects of the antiviral immune response in this tissue remain unknown. Several viral pathogens, such as herpesviruses and human papillomaviruses, have been acknowledged both historically and at present for infections in the oral cavity that result in substantial clinical burden. However, recent viral outbreaks, including those with SARS-CoV-2 and mpox, featured oral symptoms even though these viruses are not generally considered oral pathogens. Ensuing studies have shown that the oral cavity is an important locale for viral infection and potential transmission of newly emergent or re-emergent pathogens, highlighting the need for an increased understanding of the mechanisms of antiviral immunity at this site. In this Review, we provide a broad overview of antiviral immune responses in the oral cavity and discuss common viral infections and their manifestations in the oral mucosa. In addition, we present current mouse models for the study of oral viral infections.
Collapse
Affiliation(s)
- Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Pan L, Chen J, Lai Y, Du G, Wang H, Sun L, Tang G, Wang Y. HSP90 Complex From OLP Lesion Induces T-Cell Polarization via Activation of Dendritic Cells. Oral Dis 2024. [PMID: 39530299 DOI: 10.1111/odi.15195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/11/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES To investigate the effects of the heat shock protein 90 (HSP90) complex from oral lichen planus (OLP) lesion tissues on dendritic cell (DC) activation and the polarization of naïve T cells. METHODS Expression of HSP90 in OLP lesions and healthy control (HC) mucosa was evaluated by single-cell RNA sequence, IHC, qRT-PCR, and immunoblotting. HSP90 complex was extracted by immunoprecipitation from oral mucosa as the agonist of DCs. Expression of IFN-α and MHC was detected by flow cytometry. After cocultured with pre-stimulated DCs, polarization of naïve T cells was investigated by cytokine analysis. RESULTS HSP90 was significantly higher in the lamina propria of OLP lesion and closely related to lymphocyte infiltration. HSP90 complex of OLP lesion activated DCs via TLR9 and increased their IFN-α secretion and MHC II expression. Pre-stimulated DCs increased the proportion of Th17 cells. CONCLUSIONS HSP90 complex isolated from OLP lesion activated TLR9/IFN-α of DCs and further promoted the polarization of naïve T cells toward Th17 immunity.
Collapse
Affiliation(s)
- Lei Pan
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Junjun Chen
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yirao Lai
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanhuan Du
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Wang
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Sun
- Institute of Developmental Biology and Molecular Medicine, Fudan University, Shanghai, China
| | - Guoyao Tang
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Stomatology, Shanghai Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufeng Wang
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Oliver AJ, Huang N, Bartolome-Casado R, Li R, Koplev S, Nilsen HR, Moy M, Cakir B, Polanski K, Gudiño V, Melón-Ardanaz E, Sumanaweera D, Dimitrov D, Milchsack LM, FitzPatrick MEB, Provine NM, Boccacino JM, Dann E, Predeus AV, To K, Prete M, Chapman JA, Masi AC, Stephenson E, Engelbert J, Lobentanzer S, Perera S, Richardson L, Kapuge R, Wilbrey-Clark A, Semprich CI, Ellams S, Tudor C, Joseph P, Garrido-Trigo A, Corraliza AM, Oliver TRW, Hook CE, James KR, Mahbubani KT, Saeb-Parsy K, Zilbauer M, Saez-Rodriguez J, Høivik ML, Bækkevold ES, Stewart CJ, Berrington JE, Meyer KB, Klenerman P, Salas A, Haniffa M, Jahnsen FL, Elmentaite R, Teichmann SA. Single-cell integration reveals metaplasia in inflammatory gut diseases. Nature 2024; 635:699-707. [PMID: 39567783 PMCID: PMC11578898 DOI: 10.1038/s41586-024-07571-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/15/2024] [Indexed: 11/22/2024]
Abstract
The gastrointestinal tract is a multi-organ system crucial for efficient nutrient uptake and barrier immunity. Advances in genomics and a surge in gastrointestinal diseases1,2 has fuelled efforts to catalogue cells constituting gastrointestinal tissues in health and disease3. Here we present systematic integration of 25 single-cell RNA sequencing datasets spanning the entire healthy gastrointestinal tract in development and in adulthood. We uniformly processed 385 samples from 189 healthy controls using a newly developed automated quality control approach (scAutoQC), leading to a healthy reference atlas with approximately 1.1 million cells and 136 fine-grained cell states. We anchor 12 gastrointestinal disease datasets spanning gastrointestinal cancers, coeliac disease, ulcerative colitis and Crohn's disease to this reference. Utilizing this 1.6 million cell resource (gutcellatlas.org), we discover epithelial cell metaplasia originating from stem cells in intestinal inflammatory diseases with transcriptional similarity to cells found in pyloric and Brunner's glands. Although previously linked to mucosal healing4, we now implicate pyloric gland metaplastic cells in inflammation through recruitment of immune cells including T cells and neutrophils. Overall, we describe inflammation-induced changes in stem cells that alter mucosal tissue architecture and promote further inflammation, a concept applicable to other tissues and diseases.
Collapse
Affiliation(s)
- Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Raquel Bartolome-Casado
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, US
| | - Simon Koplev
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Hogne R Nilsen
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Madelyn Moy
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Victoria Gudiño
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Elisa Melón-Ardanaz
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | | | - Daniel Dimitrov
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | | | - Michael E B FitzPatrick
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicholas M Provine
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Emma Dann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Ken To
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Jonathan A Chapman
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Andrea C Masi
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Emily Stephenson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Justin Engelbert
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Sebastian Lobentanzer
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Laura Richardson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Rakeshlal Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | | | - Sophie Ellams
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Alba Garrido-Trigo
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Ana M Corraliza
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Thomas R W Oliver
- Department of Histopathology and Cytology, Cambridge University Hospitals, Cambridge, UK
| | | | - Kylie R James
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Haematology, Cambridge Stem Cell Institute, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Matthias Zilbauer
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- University Department of Paediatrics, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals, Cambridge, UK
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Marte Lie Høivik
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen S Bækkevold
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | | | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Azucena Salas
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and National Institute for Health Research (NIHR) Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Frode L Jahnsen
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK.
- Theory of Condensed Matter, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- CIFAR Macmillan Multi-scale Human Program, CIFAR, Toronto, Ontario, Canada.
| |
Collapse
|
28
|
Wen YH, Lin YX, Zhou L, Lin C, Zhang L. The immune landscape in apical periodontitis: From mechanism to therapy. Int Endod J 2024; 57:1526-1545. [PMID: 39087849 DOI: 10.1111/iej.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/04/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024]
Abstract
Apical periodontitis (AP) is featured by a persistent inflammatory response and alveolar bone resorption initiated by microorganisms, posing risks to both dental and systemic health. Nonsurgical endodontic treatment is the recommended treatment plan for AP with a high success rate, but in some cases, periapical lesions may persist despite standard endodontic treatment. Better comprehension of the AP inflammatory microenvironment can help develop adjunct therapies to improve the outcome of endodontic treatment. This review presents an overview of the immune landscape in AP, elucidating how microbial invasion triggers host immune activation and shapes the inflammatory microenvironment, ultimately impacting bone homeostasis. The destructive effect of excessive immune activation on periapical tissues is emphasized. This review aimed to systematically discuss the immunological basis of AP, the inflammatory bone resorption and the immune cell network in AP, thereby providing insights into potential immunotherapeutic strategies such as targeted therapy, antioxidant therapy, adoptive cell therapy and cytokine therapy to mitigate AP-associated tissue destruction.
Collapse
Affiliation(s)
- Yuan-Hao Wen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu-Xiu Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chen Lin
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen, China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Cariology and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
29
|
Hoytema van Konijnenburg DP, Nigrovic PA, Zanoni I. Regional specialization within the mammalian respiratory immune system. Trends Immunol 2024; 45:871-891. [PMID: 39438172 PMCID: PMC11560516 DOI: 10.1016/j.it.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The respiratory tract is exposed to infection from inhaled pathogens, including viruses, bacteria, and fungi. So far, a comprehensive assessment that integrates common and distinct aspects of the immune response along different areas of the respiratory tract has been lacking. Here, we discuss key recent findings regarding anatomical, functional, and microbial factors driving regional immune adaptation in the mammalian respiratory system, how they differ between mice and humans, and the similarities and differences with the gastrointestinal tract. We demonstrate that, under evolutionary pressure, mammals evolved spatially organized immune defenses that vary between the upper and lower respiratory tract. Overall, we propose that the functional specialization of the immune response along the respiratory tract has fundamental implications for the management of infectious or inflammatory diseases.
Collapse
Affiliation(s)
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
30
|
Li X, Chen X, Zhu Q, Zhang P, Nan S, Lv L, Qi S. D-mannose alleviates chronic periodontitis in rats by regulating the functions of neutrophils. BMC Oral Health 2024; 24:1336. [PMID: 39487474 PMCID: PMC11529006 DOI: 10.1186/s12903-024-05080-1] [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: 03/24/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease characterized by the destruction of the components of the periodontium. It significantly impacts oral health and has been linked to systemic conditions like cardiovascular disease and diabetes. The critical role of neutrophils in the occurrence and development of chronic periodontitis has been paid increasing attention. The study aimed to explore the protective effects of D-mannose on chronic periodontitis and determine whether its underlying mechanisms is related to neutrophils. METHODS To explore the protective effects of D-mannose on chronic periodontitis, the eight-week-old Sprague Dawley rat model of lipopolysaccharide (LPS)-induced periodontitis was established, followed by D-mannose treatment by oral gavage. To evaluate the protective effects of D-mannose against periodontal bone loss, methylene blue staining, hematoxylin and eosin (H&E) staining, and micro-CT scanning were utilized. Then, immunofluorescence (IF), Western Blot, and RT-PCR were applied to assess the expression levels of pro-inflammatory cytokines (IL-1β, IL-6, and IL-17), anti-inflammatory cytokine (IL-10), tumor necrosis factor-alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), ten-eleven translocation 2 (TET2), and key glycolytic enzymes (HK1, HK2, PFKFB3), and to examine D-mannose's impact on the recruitment and activation of neutrophils in the gingiva. Additionally, neutrophils isolated from the peripheral blood of healthy rats were treated with LPS and D-mannose, and changes in the expression levels of myeloperoxidase (MPO), IL-1β, IL-6, IL-17, IL-10, and TET2 were observed via IF. RESULTS In vivo, D-mannose inhibited LPS-induced alveolar bone resorption in rats. After D-mannose treatment, the expression levels of IL-17 (p<0.01) and TET2 (p<0.01) were suppressed by IF, and the expression levels of IL-1β (p<0.05), IL-17 (p<0.05) and TET2 (p<0.01) were downregulated by WB. The results of qPCR showed that D-mannose reduced the expression levels of IL-1β (p<0.05), IL-6 (p<0.01), IL-17 (p<0.01), TNF-α (p<0.01), G-CSF (p<0.01), GM-CSF (p<0.01), TET2 (p<0.01), HK1 (p<0.01), HK2 (p<0.01), and PFKFB3 (p<0.01). D-mannose also inhibited the recruitment and activation of neutrophils in LPS-treated rat gingival tissues. In vitro, the results of IF showed that D-mannose inhibited the activation of neutrophils stimulated by LPS, downregulated the expression of IL-1β (p < 0.05), IL-6, IL-17 (p < 0.01), and TET2 (p < 0.01), and upregulated the expression of IL-10 (p < 0.01). CONCLUSIONS D-mannose can alleviate chronic periodontitis in rats by regulating the functions of neutrophils, potentially associated with the expression of TET2 and glycolysis, providing new insights into the potential application of D-mannose to chronic periodontitis.
Collapse
Affiliation(s)
- Xue Li
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Xueting Chen
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Qingyu Zhu
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Pengye Zhang
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Shunxue Nan
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Lei Lv
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Shengcai Qi
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Yan Y, Yan W, Wu S, Zhao H, Chen Q, Wang J. Oral Patch/Film for Drug Delivery-Current Status and Future Prospects. Biopolymers 2024; 115:e23625. [PMID: 39230032 DOI: 10.1002/bip.23625] [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: 06/20/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
In recent years, there has been extensive research into drug delivery systems aimed at enhancing drug utilization while minimizing drug toxicities. Among these systems, oral patches/films have garnered significant attention due to their convenience, noninvasive administration, ability to bypass hepatic first-pass metabolism, thereby enhancing drug bioavailability, and their potential to ensure good compliance, particularly among special patient populations. In this review, from the perspective of the anatomical characteristics of the oral cavity and the advantages and difficulties of oral drug delivery, we illustrate the design ideas, manufacturing techniques, research methodologies, and the essential attributes of an ideal oral patch/film. Furthermore, the applications of oral patches/films in both localized and systemic drug delivery were discussed. Finally, we offer insights into the future prospects of the oral patch/film, aiming to provide valuable reference for the advancement of oral localized drug delivery systems.
Collapse
Affiliation(s)
- Yujie Yan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenjie Yan
- Inner Mongolia Medical University College of Traditional Chinese Medicine, Hohhot, China
| | - Sihua Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiongke Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management & Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Cheng J, Zhou C, Liu J, Geng Y, Liu L, Fan Y. Expression of neutrophil extracellular trap-related proteins and its correlation with IL-17 and TNF-α in patients with oral lichen planus. PeerJ 2024; 12:e18260. [PMID: 39430569 PMCID: PMC11488494 DOI: 10.7717/peerj.18260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Background Neutrophil extracellular traps (NETs) are produced by polymorphonuclear neutrophils (PMNs) stimulated by interleukin-17 (IL-17) and tumor necrosis factor α (TNF-α). However, the level and role of NETs in oral lichen planus (OLP) remain poorly understood. Objective This study aimed to investigate the expression of NETs in OLP and explore the correlation between NETs and the levels of IL-17 and TNF-α. Methods The expression and distribution of NET-related proteins in tissue samples from each group were assessed using hematoxylin-eosin (HE) staining and immunofluorescence (IF). Additionally, the expression of NET-related proteins in peripheral blood samples from each group was evaluated using cell IF technique and fluorescence spectrophotometry. The relative formation level of NETs in each group was determined by fluorescence spectrophotometry via plasma co-culture. Furthermore, the levels of inflammatory cytokines IL-17 and TNF-α in plasma and culture supernatant were measured using enzyme-linked immunosorbent assay (ELISA). Results NET-related proteins were located in the subepithelial and lamina propria layers of OLP lesions. OLP had significantly higher expression of NET-related proteins in lesion tissues and peripheral blood compared to the healthy control (HC) group (p < 0.05). The rate of NETs formation in the erosive-stage OLP (EOLP) group was significantly higher than that in the HC group (p < 0.05), in contrast, no significant increase was observed in the non-erosive OLP (NEOLP) group (p > 0.05). Furthermore, the levels of IL-17 and TNF-α in the EOLP group were significantly elevated compared to those in the NEOLP group and HC group (p < 0.05), while the levels in the NEOLP group did not significantly differ from those in the HC group (p > 0.05). The rate of NETs formation showed a positive correlation with the levels of IL-17 and TNF-α in plasma. Conclusion The expression of NET-related proteins was upregulated in OLP lesion tissues and peripheral blood. Elevated levels of IL-17 and TNF-α in peripheral blood plasma positively correlated with the rate of NETs formation, suggesting that IL-17 and TNF-α mediate the formation of NETs in OLP patients, and may thereby contribute to the development of OLP.
Collapse
Affiliation(s)
- Juehua Cheng
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Chenyu Zhou
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
- Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Liu
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yanlin Geng
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Lin Liu
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Yuan Fan
- Department of Oral Mucosal Diseases, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| |
Collapse
|
33
|
Easter QT, Alvarado-Martinez Z, Kunz M, Matuck BF, Rupp BT, Weaver T, Ren Z, Tata A, Caballero-Perez J, Oscarson N, Hasuike A, Ghodke AN, Kimple AJ, Tata PR, Randell SH, Koo H, Ko KI, Byrd KM. Polybacterial Intracellular Macromolecules Shape Single-Cell Epikine Profiles in Upper Airway Mucosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617279. [PMID: 39416216 PMCID: PMC11482982 DOI: 10.1101/2024.10.08.617279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The upper airway, particularly the nasal and oral mucosal epithelium, serves as a primary barrier for microbial interactions throughout life. Specialized niches like the anterior nares and the tooth are especially susceptible to dysbiosis and chronic inflammatory diseases. To investigate host-microbial interactions in mucosal epithelial cell types, we reanalyzed our single-cell RNA sequencing atlas of human oral mucosa, identifying polybacterial signatures (20% Gram-positive, 80% Gram-negative) within both epithelial- and stromal-resident cells. This analysis revealed unique responses of bacterial-associated epithelia when compared to two inflammatory disease states of mucosa. Single-cell RNA sequencing, in situ hybridization, and immunohistochemistry detected numerous persistent macromolecules from Gram-positive and Gram-negative bacteria within human oral keratinocytes (HOKs), including bacterial rRNA, mRNA and glycolipids. Epithelial cells with higher concentrations of 16S rRNA and glycolipids exhibited enhanced receptor-ligand signaling in vivo. HOKs with a spectrum of polybacterial intracellular macromolecular (PIM) concentrations were challenged with purified exogenous lipopolysaccharide, resulting in the synergistic upregulation of select innate (CXCL8, TNFSF15) and adaptive (CXCL17, CCL28) epikines. Notably, endogenous lipoteichoic acid, rather than lipopolysaccharide, directly correlated with epikine expression in vitro and in vivo. Application of the Drug2Cell algorithm to health and inflammatory disease data suggested altered drug efficacy predictions based on PIM detection. Our findings demonstrate that PIMs persist within mucosal epithelial cells at variable concentrations, linearly driving single-cell effector cytokine expression and influencing drug responses, underscoring the importance of understanding host-microbe interactions and the implications of PIMs on cell behavior in health and disease at single-cell resolution.
Collapse
Affiliation(s)
- Quinn T Easter
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Zabdiel Alvarado-Martinez
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Meik Kunz
- The Bioinformatics CRO, Orlando, FL, USA
| | - Bruno Fernandes Matuck
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Brittany T Rupp
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Theresa Weaver
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Zhi Ren
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | | | - Nick Oscarson
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Akira Hasuike
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, JP
| | - Ameer N Ghodke
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Adam J Kimple
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Purushothama R Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hyun Koo
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin M Byrd
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
- UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
34
|
Zhou J, Hu Z, Wang L, Hu Q, Chen Z, Lin T, Zhou R, Cai Y, Wu Z, Zhang Z, Yang Y, Zhang C, Li G, Zeng L, Su K, Li H, Su Q, Zeng G, Cheng B, Wu T. Tumor-colonized Streptococcus mutans metabolically reprograms tumor microenvironment and promotes oral squamous cell carcinoma. MICROBIOME 2024; 12:193. [PMID: 39369210 PMCID: PMC11452938 DOI: 10.1186/s40168-024-01907-9] [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: 12/21/2023] [Accepted: 08/13/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) remains a major death cause in head and neck cancers, but the exact pathogenesis mechanisms of OSCC are largely unclear. RESULTS Saliva derived from OSCC patients but not healthy controls (HCs) significantly promotes OSCC development and progression in rat models, and metabolomic analyses reveal saliva of OSCC patients but not HCs and OSCC tissues but not adjacent non-tumor tissues contain higher levels of kynurenic acid (KYNA). Furthermore, large amounts of Streptococcus mutans (S. mutans) colonize in OSCC tumor tissues, and such intratumoral S. mutans mediates KYNA overproductions via utilizing its protein antigen c (PAc). KYNA shifts the cellular types in the tumor microenvironment (TME) of OSCC and predominantly expedites the expansions of S100a8highS100a9high neutrophils to produce more interleukin 1β (IL-1β), which further expands neutrophils and induces CD8 + T cell exhaustion in TME and therefore promotes OSCC. Also, KYNA compromises the therapeutic effects of programmed cell death ligand 1 (PD-L1) and IL-1β blockades in oral carcinogenesis model. Moreover, KYNA-mediated immunosuppressive program and aryl hydrocarbon receptor (AHR) expression correlate with impaired anti-tumor immunity and poorer survival of OSCC patients. CONCLUSIONS Thus, aberration of oral microbiota and intratumoral colonization of specific oral bacterium such as S. mutans may increase the production of onco-metabolites, exacerbate the oral mucosal carcinogenesis, reprogram a highly immunosuppressive TME, and promote OSCC, highlighting the potential of interfering with oral microbiota and microbial metabolism for OSCC preventions and therapeutics. Video Abstract.
Collapse
Affiliation(s)
- Jiaying Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Zixuan Hu
- The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, 330006, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, 330006, China
| | - Lei Wang
- BGI Research, Chongqing, 401329, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI Research, Shenzhen, 518083, China
| | - Qinchao Hu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Zixu Chen
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Tao Lin
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Rui Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Yongjie Cai
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhiying Wu
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhiyi Zhang
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yi Yang
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | | | - Guibo Li
- BGI Research, Chongqing, 401329, China
- Guangdong Provincial Key Laboratory of Human Disease Genomics, BGI Research, Shenzhen, 518083, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI Research, Shenzhen, 518083, China
| | - Lingchan Zeng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
| | - Kai Su
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China
| | - Huan Li
- Department of Intensive Care Unit (ICU), State Key Laboratory of Oncology in South China, CollaborativeInnovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Gucheng Zeng
- Department of Microbiology, Key Laboratory for Tropical Diseases Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Bin Cheng
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China.
| | - Tong Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Lingyuan Road West, Guangzhou, 510055, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510080, China.
| |
Collapse
|
35
|
Cao H, Lin J, Yuan H, Yang Z, Nie M, Pathak JL, Yuan ZG, Yu M. The emerging role of Toxoplasma gondii in periodontal diseases and underlying mechanisms. Front Immunol 2024; 15:1464108. [PMID: 39430742 PMCID: PMC11487530 DOI: 10.3389/fimmu.2024.1464108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/17/2024] [Indexed: 10/22/2024] Open
Abstract
Toxoplasma gondii (T. gondii), an obligate intracellular protozoan parasite, is increasingly recognized for its role in various human diseases, including periodontal diseases. Periodontal diseases comprise a wide range of inflammatory conditions that not only affect the supporting structures of the teeth and oral health but also contribute to systemic diseases. The parasite's ability to modulate the host's immune response and induce chronic inflammation within the periodontium is a key factor in periodontal tissue damage. Through its virulence factors, T. gondii disrupts the balance of inflammatory cytokines, leading to dysregulated immune responses, and exacerbates oxidative stress in periodontal tissues. And T. gondii invasion could affect specific proteins in host cells including HSP70, BAGs, MICs, ROPs, SAGs, and GRAs leading to periodontal tissue damage. The indirect role of the host immune response to T. gondii via natural killer cells, monocytes, macrophages, neutrophils, dendritic cells, T cells, and B cells also contributes to periodontal diseases. Understanding these complex interactions of T. gondii with host cells could unravel disease mechanisms and therapeutic targets for periodontal diseases. This review delves into the pathogenic mechanisms of T. gondii in periodontal diseases, offering a detailed exploration of both direct and indirect pathways of its impact on periodontal health.
Collapse
Affiliation(s)
- Henglong Cao
- Department of Periodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Jianfeng Lin
- Department of Periodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Hao Yuan
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zipeng Yang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Min Nie
- Department of Periodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Janak L. Pathak
- Department of Periodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
| | - Zi-Guo Yuan
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Miao Yu
- Department of Periodontics, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, China
- Department of Oral Health Sciences-BIOMAT, KU Leuven and Dentistry, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
36
|
Chen S, Gao G, Shi J, Li N, Xie L, Zhang Y, Shan Z, Xie J, Xiao Y, Chen Z, Chen Z. Unveiling the governing role of 'remodeling triangle area' in soft-hard tissue interface equilibrium for metal implants advancement. Mater Today Bio 2024; 28:101170. [PMID: 39211290 PMCID: PMC11357867 DOI: 10.1016/j.mtbio.2024.101170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/13/2024] [Accepted: 07/27/2024] [Indexed: 09/04/2024] Open
Abstract
Metal implants holds significant promise for diverse fixed prostheses. However, their long-term reliability and broader application are hindered by challenges related to the disequilibrium at the soft-hard tissue interface. By using anti-inflammatory (PDA/IL4) and pro-inflammatory (PDA/LPS/IFNγ) coatings to modulate distinct immune characteristics, we discovered a dynamic bioactive structure at the soft-hard tissue interface around metal implant, which we have named the 'Remodeling Triangle Area' (RTA). We further demonstrate that the RTA can be influenced by the PDA/IL4 coating to favor a phenotype that enhances both innate and adaptive immunity. This leads to stronger epithelial adhesion, the formation of dense connective tissue via IGF1 secretion, and a more balanced soft-hard tissue interface through the OPG/RANKL axis. Conversely, the PDA/LPS/IFNγ coating shifts the RTA towards a phenotype that activates the innate immune response. This results in a less cohesive tissue structure and bone resorption, characterized by reduced IGF1 secretion and an imbalanced OPG/RANKL axis. Over all, our study introduces the novel concept termed the 'Remodeling Triangle Area' (RTA), an immune-rich anatomical region located at the nexus of the implant interface, epithelial, connective, and bone tissue, which becomes highly interactive post-implantation to modulate the soft-hard tissue interface equilibrium. We believe that an RTA-centric, immunomodulatory approach has the potential to revolutionize the design of next-generation metal implants, providing unparalleled soft-hard tissue interface equilibrium properties.
Collapse
Affiliation(s)
- Shoucheng Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Guangqi Gao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Jiamin Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Na Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Lv Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yingye Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zhengjie Shan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Jiaxin Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yin Xiao
- School of Medicine and Dentistry, Griffith University (GU), Gold Coast, QLD, 4222, Australia
| | - Zhuofan Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University and Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| |
Collapse
|
37
|
Xiang X, Zhang J, Yue Y. Pyroptosis: A major trigger of excessive immune response in the gingiva. Oral Dis 2024; 30:4152-4160. [PMID: 38852159 DOI: 10.1111/odi.15013] [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: 01/23/2024] [Revised: 04/21/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVES The gingival mucosal barrier, an important oral cavity barrier, plays a significant role in preventing pathogenic microorganism invasion and maintaining periodontal tissue health. Pathogenic microorganism invasion of the gingival mucosa produces a large number of cytokines. Among them, pyroptosis is an important player in exacerbating immune-inflammatory responses, leading to tissue destruction. However, the mechanism of pyroptosis and the immune response it triggers have not been fully elucidated. We provide an overview of recent advances in understanding gingival physical barrier pyroptosis and inflammation-induced hyperimmunity. METHODS PubMed, Web of Science databases were searched for articles, reviews, and clinical studies published until March 2024. RESULTS We summarised the importance of the gingival barrier in terms of the functions of different cells, described the progress in research on gingival epithelial cell and gingival fibroblast pyroptosis and the immune-inflammatory response it induces, and discussed the relationship between pyroptosis and systemic diseases, association of multiple cell death systems. Finally, we propose future directions for pyroptosis research. CONCLUSIONS Pyroptosis often triggers a range of inflammatory immune responses that lead to associated diseases. Therefore, further study of the molecular mechanisms of pyroptosis and the immune responses is warranted.
Collapse
Affiliation(s)
- Xueyu Xiang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zhang
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Yuan Yue
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Jiang W, Yu W, Hu S, Shi Y, Lin L, Yang R, Tang J, Gu Y, Gong Y, Jin M, Lu E. Differential expression of FSTL1 and its correlation with the pathological process of periodontitis. J Periodontal Res 2024; 59:1005-1016. [PMID: 38807492 DOI: 10.1111/jre.13275] [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: 01/17/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/30/2024]
Abstract
AIMS This study aimed to elucidate the alterations in Follistatin-like protein 1 (FSTL1) and its association with the pathological process of periodontitis. METHODS This study included 48 patients with periodontitis and 42 healthy controls. The expression level of FSTL1 in the gingiva was determined by RT-qPCR, validated using the dataset GSE16134, and subsequently examined by western blotting. Bioinformatics analysis revealed a single-cell distribution of FSTL1, characteristic of angiogenesis and immune cell infiltration. The expression and distribution of FSTL1, vascular endothelial marker protein CD31 and myeloperoxidase (MPO), the indicator of neutrophil activity, were determined by immunohistochemistry (IHC). A series of correlation analyses was performed to determine the associations between FSTL1 and clinical parameters, including probing depth (PD) and clinical attachment loss (CAL), and their potential role in angiogenesis (CD31) and neutrophil infiltration (MPO). RESULTS FSTL1 was significantly upregulated in the gingiva of patients with periodontitis compared to their healthy counterparts. In addition, FSTL1 was positively correlated with the clinical parameters PD (r = .5971, p = .0005) and CAL (r = .6078, p = .0004). Bioinformatic analysis and IHC indicated that high FSTL1 expression was significantly correlated with angiogenesis and neutrophil infiltration in periodontitis. Moreover, receiver operating characteristic (ROC) analysis demonstrated that FSTL1 could serve as an independent indicator for evaluating the severity of periodontitis (area under the curve [AUC] = 0.9011, p < .0001). CONCLUSION This study demonstrated FSTL1 upregulation in periodontitis and its potential contribution to the disease via angiogenesis and neutrophil infiltration.
Collapse
Affiliation(s)
- Wenxin Jiang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijun Yu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shucheng Hu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanjie Shi
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lu Lin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruhan Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiaqi Tang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhua Gong
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
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
|
40
|
Mandel VD, Marconi GD, Pizzicannella J, Paganelli A. Editorial: Inflammatory disorders of the oral mucosa: current challenges and future perspectives. FRONTIERS IN ORAL HEALTH 2024; 5:1497187. [PMID: 39398105 PMCID: PMC11466937 DOI: 10.3389/froh.2024.1497187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024] Open
Affiliation(s)
- Victor Desmond Mandel
- Porphyria and Rare Diseases Unit, San Gallicano Dermatological Institute-IRCCS, Rome, Italy
| | - Guya Diletta Marconi
- Department of Innovative Technologies in Medicine and Dentistry, University “G. D'Annunzio” Chieti-Pescara, Chieti, Italy
| | - Jacopo Pizzicannella
- Dipartimento di Ingegneria e Geologia, University “G. D'Annunzio” Chieti-Pescara, Chieti, Italy
| | - Alessia Paganelli
- Clinical Dermatology Unit, IDI- IRCCS Istituto Dermopatico Dell'Immacolata, Rome, Italy
| |
Collapse
|
41
|
Xuanyuan X, Zhang L, Zheng Y, Jiang R, Ma Y, Liu R, Hou P, Lei M, Xu H, Zeng H. SPRR1B+ keratinocytes prime oral mucosa for rapid wound healing via STAT3 activation. Commun Biol 2024; 7:1155. [PMID: 39300285 PMCID: PMC11413210 DOI: 10.1038/s42003-024-06864-5] [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: 12/09/2022] [Accepted: 09/06/2024] [Indexed: 09/22/2024] Open
Abstract
Oral mucosal wounds exhibit accelerated healing with reduced scarring compared to cutaneous wounds, representing an optimal wound healing paradigm. However, the specific cellular subtypes orchestrating the efficient healing of mucosal tissues remain elusive. Through a comprehensive analysis integrating bulk-mRNA and single-cell sequencing data during the wound healing process in oral mucosa and skin, we have delineated a distinct set of genes markedly upregulated during tissue repair. This collection of wound healing-associated genesets was highly enriched in a specific keratinocyte subpopulation identified as STAT3-activated SPRR1B+ keratinocytes. Notably, despite the inherent rapidity of oral mucosal healing, the induction of SPRR1B+ keratinocytes is evident in both skin and mucosal wound healing processes in murine model. Intriguingly, these wound healing-promoting SPRR1B+ keratinocytes, which are induced via STAT3 activation, inherently abundant in unwounded normal mucosa but absent in normal skin. SPRR1B knockdown significantly inhibits mucosal keratinocyte migration, a critical attribute for effective wound healing. In summary, through analysis of human oral and skin wound healing processes at single-cell resolution, coupled with validation in murine model, suggests STAT3-activated SPRR1B+ keratinocytes are associated with the rapid mucosal repair process. This discovery underscores the potential application of SPRR1B+ keratinocytes in the therapeutic management of chronic or non-healing wounds.
Collapse
Affiliation(s)
- Xinyang Xuanyuan
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Li Zhang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yang Zheng
- Department of Oral & Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ruixin Jiang
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yanni Ma
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ruixin Liu
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Pengcong Hou
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Ming Lei
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hui Xu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Hanlin Zeng
- Shanghai Institute of Precision Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
- Department of Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
42
|
Borowiec BM, Dyszkiewicz-Konwińska M, Bukowska D, Nowicki M, Budna-Tukan J. Small Extracellular Vesicles and Oral Mucosa: The Power Couple in Regenerative Therapies? Cells 2024; 13:1514. [PMID: 39329698 PMCID: PMC11429515 DOI: 10.3390/cells13181514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Although ongoing debates persist over the scope of phenomena classified as regenerative processes, the most up-to-date definition of regeneration is the replacement or restoration of damaged or missing cells, tissues, organs, or body parts to full functionality. Despite extensive research on this topic, new methods in regenerative medicine are continually sought, and existing ones are being improved. Small extracellular vesicles (sEVs) have gained attention for their regenerative potential, as evidenced by existing studies conducted by independent research groups. Of particular interest are sEVs derived from the oral mucosa, a tissue renowned for its rapid regeneration and minimal scarring. While the individual regenerative potential of both sEVs and the oral mucosa is somewhat understood, the combined potential of sEVs derived from the oral mucosa has not been sufficiently explored and highlighted in the existing literature. Serving as a broad compendium, it aims to provide scientists with essential and detailed information on this subject, including the nature of the materials employed, isolation and analysis methodologies, and clinical applications. The content of this survey aims to facilitate the comparison of diverse methods for working with sEVs derived from the oral mucosa, aiding in the planning of research endeavors and identifying potential research gaps.
Collapse
Affiliation(s)
- Blanka Maria Borowiec
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland (M.N.)
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | | | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 87-100 Torun, Poland;
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland (M.N.)
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland (M.N.)
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, 65-046 Zielona Gora, Poland
| |
Collapse
|
43
|
Wang L, Dos Santos Sanches N, Panahipour L, Imani A, Yao Y, Zhang Y, Li L, Gruber R. Dimethyl Fumarate-Loaded Gellan Gum Hydrogels Can Reduce In Vitro Chemokine Expression in Oral Cells. Int J Mol Sci 2024; 25:9485. [PMID: 39273432 PMCID: PMC11395421 DOI: 10.3390/ijms25179485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Dimethyl fumarate (DMF), originally proposed to treat multiple sclerosis, is considered to have a spectrum of anti-inflammatory effects that effectively control periodontitis, mainly when applied with a hydrogel delivery system. Chemokine expression by gingival fibroblasts is a significant driver of periodontitis; thus, hydrogel-based strategies to deliver DMF, which in turn dampen chemokine expression, are of potential clinical relevance. To test this approach, we have established a bioassay where chemokine expression is induced by exposing gingival fibroblast to IL1β and TNFα, or with saliva. We show herein that DMF effectively reduced the expression of CXCL8, CXCL1, CXCL2, and CCL2-and lowered the phosphorylation of ERK and JNK-without affecting cell viability. This observation was confirmed by immunoassays with CXCL8. Consistently, the forced chemokine expression in HSC2 oral squamous epithelial cells was greatly diminished by DMF. To implement our hydrogel-based delivery system, gingival fibroblasts were cocultured with gellan gum hydrogels enriched for DMF. In support of our strategy, DMF-enriched gellan gum hydrogels significantly reduced the forced chemokine expression in gingival fibroblasts. Our data suggest that DMF exerts its anti-inflammatory activity in periodontal cells when released from gellan gum hydrogels, suggesting a potential clinical relevance to control overshooting chemokine expression under chronic inflammatory conditions.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China
| | - Natalia Dos Santos Sanches
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Department of Diagnosis and Surgery, Araçatuba Dental School of Sao Paulo, Sao Paulo 16015-050, Brazil
| | - Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Atefe Imani
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Yili Yao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou 325027, China
| | - Yan Zhang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China
| | - Lingli Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou 325027, China
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, 1090 Vienna, Austria
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
44
|
Napiórkowska-Baran K, Darwish S, Kaczor J, Treichel P, Szymczak B, Szota M, Koperska K, Bartuzi Z. Oral Diseases as a Manifestation of Inborn Errors of Immunity. J Clin Med 2024; 13:5079. [PMID: 39274292 PMCID: PMC11396297 DOI: 10.3390/jcm13175079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Oral findings such as inflammation, ulcerations, or lesions can indicate serious systemic diseases and should prompt suspicion of acquired chronic conditions or inborn errors of immunity (IEIs). Currently, there are approximately 500 disease entities classified as IEIs, with the list expanding annually. The awareness of the existence of such conditions is of paramount importance, as patients with these disorders frequently necessitate the utilization of enhanced diagnostic techniques. This is exemplified by patients with impaired antibody production, in whom conventional serological methods may prove to be undiagnostic. Patients with IEI may require distinct therapeutic approaches or antimicrobial prophylaxis throughout their lives. An accurate diagnosis and, more importantly, early identification of patients with immune deficiencies is crucial to ensure the quality and longevity of their lives. It is important to note that the failure to establish a proper diagnosis or to provide adequate treatment could also have legal implications for medical professionals. The article presents IEIs, which may manifest in the oral cavity, and their diagnosis alongside therapeutic procedures.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Samira Darwish
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Justyna Kaczor
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Paweł Treichel
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Maciej Szota
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Kinga Koperska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| |
Collapse
|
45
|
Cieri N, Hookeri N, Stromhaug K, Li L, Keating J, Díaz-Fernández P, Gómez-García de Soria V, Stevens J, Kfuri-Rubens R, Shao Y, Kooshesh KA, Powell K, Ji H, Hernandez GM, Abelin J, Klaeger S, Forman C, Clauser KR, Sarkizova S, Braun DA, Penter L, Kim HT, Lane WJ, Oliveira G, Kean LS, Li S, Livak KJ, Carr SA, Keskin DB, Muñoz-Calleja C, Ho VT, Ritz J, Soiffer RJ, Neuberg D, Stewart C, Getz G, Wu CJ. Systematic identification of minor histocompatibility antigens predicts outcomes of allogeneic hematopoietic cell transplantation. Nat Biotechnol 2024:10.1038/s41587-024-02348-3. [PMID: 39169264 DOI: 10.1038/s41587-024-02348-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/02/2024] [Indexed: 08/23/2024]
Abstract
T cell alloreactivity against minor histocompatibility antigens (mHAgs)-polymorphic peptides resulting from donor-recipient (D-R) disparity at sites of genetic polymorphisms-is at the core of the therapeutic effect of allogeneic hematopoietic cell transplantation (allo-HCT). Despite the crucial role of mHAgs in graft-versus-leukemia (GvL) and graft-versus-host disease (GvHD) reactions, it remains challenging to consistently link patient-specific mHAg repertoires to clinical outcomes. Here we devise an analytic framework to systematically identify mHAgs, including their detection on HLA class I ligandomes and functional verification of their immunogenicity. The method relies on the integration of polymorphism detection by whole-exome sequencing of germline DNA from D-R pairs with organ-specific transcriptional- and proteome-level expression. Application of this pipeline to 220 HLA-matched allo-HCT D-R pairs demonstrated that total and organ-specific mHAg load could independently predict the occurrence of acute GvHD and chronic pulmonary GvHD, respectively, and defined promising GvL targets, confirmed in a validation cohort of 58 D-R pairs, for the prevention or treatment of post-transplant disease recurrence.
Collapse
Affiliation(s)
- Nicoletta Cieri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nidhi Hookeri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kari Stromhaug
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Liang Li
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Julia Keating
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Paula Díaz-Fernández
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Valle Gómez-García de Soria
- Department of Hematology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Jonathan Stevens
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Raphael Kfuri-Rubens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Yiren Shao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Kaila Powell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Helen Ji
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gabrielle M Hernandez
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jennifer Abelin
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Susan Klaeger
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Department of Proteomic and Genomic Technologies, Genentech Inc, South San Francisco, CA, USA
| | - Cleo Forman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karl R Clauser
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Siranush Sarkizova
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - David A Braun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Livius Penter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Hematology, Oncology, and Tumorimmunology, Campus Virchow Klinikum, Berlin, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Haesook T Kim
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William J Lane
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Leslie S Kean
- Harvard Medical School, Boston, MA, USA
- Division Hematology/Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Shuqiang Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kenneth J Livak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Steven A Carr
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Derin B Keskin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Computer Science, Metropolitan College, Boston University, Boston, MA, USA
- Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Cecilia Muñoz-Calleja
- Department of Immunology, Instituto de Investigación Sanitaria Princesa (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Vincent T Ho
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jerome Ritz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Donna Neuberg
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Chip Stewart
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Gad Getz
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
46
|
Zhang J, Yang L, Zeng H, Zhao Z, Han Y, Zhao Y, Qu S, Gong Z, Wang Z, Bai Y, Zhao Q. Targeted Reprogramming of Pathogenic Fibroblast Genes at the 3'-Untranslated Regions by DNA Nanorobots for Periodontitis. ACS NANO 2024; 18:22139-22152. [PMID: 39110572 DOI: 10.1021/acsnano.4c05475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Periodontitis, with its persistent nature, causes significant distress for most sufferers. Current treatments, such as mechanical cleaning and surgery, often fail to fully address the underlying overactivation of fibroblasts that drives this degradation. Targeting the post-transcriptional regulation of fibroblasts, particularly at the 3'-untranslated regions (3'UTR) of pathogenic genes, offers a therapeutic strategy for periodontitis. Herein, we developed a DNA nanorobot for this purpose. This system uses a dynamic DNA nanoframework to incorporate therapeutic microRNAs through molecular recognition and covalent bonds, facilitated by DNA monomers modified with disulfide bonds. The assembled-DNA nanoframework is encapsulated in a cell membrane embedded with a fibroblast-targeting peptide. By analyzing the 3'UTR regions of pathogenic fibroblast genes FOSB and JUND, we identified the therapeutic microRNA as miR-1-3p and integrated it into this system. As expected, the DNA nanorobot delivered the internal components to fibroblasts by the targeting peptide and outer membrane that responsively releases miR-1-3p under intracellular glutathione. It resulted in a precise reduction of mRNA and suppression of protein function in pathogenic genes, effectively reprogramming fibroblast behavior. Our results confirm that this approach not only mitigates the inflammation but also promotes tissue regeneration in periodontal models, offering a promising therapeutic avenue for periodontitis.
Collapse
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Liu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hao Zeng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zifan Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yue Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yilong Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Shuyuan Qu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zijian Gong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ziming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yi Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
47
|
Wang T, Liu X, Li J, Yue Y, Li J, Wang M, Wei N, Hao L. Mechanisms of mechanical force in periodontal homeostasis: a review. Front Immunol 2024; 15:1438726. [PMID: 39221238 PMCID: PMC11361942 DOI: 10.3389/fimmu.2024.1438726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Mechanical forces affect periodontal health through multiple mechanisms. Normally, mechanical forces can boost soft and hard tissue metabolism. However, excessive forces may damage the periodontium or result in irreversible inflammation, whereas absence of occlusion forces also leads to tissue atrophy and bone resorption. We systemically searched the PubMed and Web of Science databases and found certain mechanisms of mechanical forces on immune defence, extracellular matrix (ECM) metabolism, specific proteins, bone metabolism, characteristic periodontal ligament stem cells (PDLSCs) and non-coding RNAs (ncRNAs) as these factors contribute to periodontal homeostasis. The immune defence functions change under forces; genes, signalling pathways and proteinases are altered under forces to regulate ECM metabolism; several specific proteins are separately discussed due to their important functions in mechanotransduction and tissue metabolism. Functions of osteocytes, osteoblasts, and osteoclasts are activated to maintain bone homeostasis. Additionally, ncRNAs have the potential to influence gene expression and thereby, modify tissue metabolism. This review summarizes all these mechanisms of mechanical forces on periodontal homeostasis. Identifying the underlying causes, this review provides a new perspective of the mechanisms of force on periodontal health and guides for some new research directions of periodontal homeostasis.
Collapse
Affiliation(s)
- Tianqi Wang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xinran Liu
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiaxin Li
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Yue
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jinle Li
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of General Clinic, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Min Wang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Wei
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Hao
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Deep skin fibroblast-mediated macrophage recruitment supports acute wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607357. [PMID: 39149286 PMCID: PMC11326280 DOI: 10.1101/2024.08.09.607357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single nuclei RNA-sequencing, we defined changes in gene expression associated with inflammation at 1-day post-wounding (dpw) in mouse skin. Compared to keratinocytes and myeloid cells, we detected enriched expression of pro-inflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL33 compared to SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound bed macrophages and monocytes during injury-induced inflammation with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of deep skin fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M. Amuso
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - MaryEllen R. Haas
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Paula O. Cooper
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Sana Hafiz
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Shatha Salameh
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Miguel F. Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Violet Josephson
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Khatereh Khorsandi
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Dermatology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
49
|
Feng MH, Lai YR, Deng YW, Li XY, Pan L, Tian Z, Tang GY, Wang YF. B Cells Infiltration Potentially Responded Better to Systemic Corticoids in Oral Lichen Planus and Oral Lichenoid Lesions. Inflammation 2024:10.1007/s10753-024-02112-4. [PMID: 39117788 DOI: 10.1007/s10753-024-02112-4] [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: 04/19/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024]
Abstract
Oral lichen planus (OLP) and oral lichenoid lesion (OLL) are chronic inflammatory diseases involving the oral mucosa. B cells infiltration in OLP and OLL, however, little is known about these cells in OLP and OLL. To analyze the function and infiltrating features of B lymphocytes in OLP and OLL, and to preliminarily evaluate their correlation with clinical outcomes. Tissue samples were collected from OLP, OLL, and healthy mucosa. The phenotypes and amounts of B cells in tissues were analyzed by single-cell sequencing. Their proportion and infiltrating features in tissues were examined by immunohistochemistry and immunofluorescence. With the systemic medication of corticoids, the correlation between B cells infiltrating characteristics and the clinical outcomes were evaluated. A quantified proportion increase of B cells was shown in both OLP and OLL. B cells in OLP demonstrated heightened activation and enhanced regulation in immune response. A cohort of 100 patients with OLP/OLL and 13 healthy controls were examined to investigate the B cells infiltration pattern. B cells were distributed in the superficial layer of lamina propria in 92.9% and 41.9% of OLP and OLL, respectively(P < 0.01); focally distributed in 25.0% and 62.9% of OLP and OLL, respectively(P < 0.01). With the systemic medication of corticoids, the cases with B cell infiltration (B+) in OLP and OLL groups showed a statistically significant reduction in REU scores before and after treatment (P < 0.01). B cells are widely present in OLP and OLL, and B cell infiltration in OLP and OLL are related to the better therapeutic effect of oral corticoids.
Collapse
Grants
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
- 82270976, 82205200, 82020108010 The National Natural Science Foundation of China
Collapse
Affiliation(s)
- Ming-Hua Feng
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Department of Stomatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Yi-Rao Lai
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Yi-Wen Deng
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xi-Ye Li
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Lei Pan
- Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhen Tian
- Department of Oral Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Guo-Yao Tang
- Shanghai Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yu-Feng Wang
- Department of Oral Medicine, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai, 200011, China.
- College of Stomatology, Shanghai Jiao Tong University; Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China.
| |
Collapse
|
50
|
McClure FA, Wemyss K, Cox JR, Bridgeman HM, Prise IE, King JI, Jaigirdar S, Whelan A, Jones GW, Grainger JR, Hepworth MR, Konkel JE. Th17-to-Tfh plasticity during periodontitis limits disease pathology. J Exp Med 2024; 221:e20232015. [PMID: 38819409 PMCID: PMC11143381 DOI: 10.1084/jem.20232015] [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: 11/02/2023] [Revised: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
Th17 cell plasticity is crucial for development of autoinflammatory disease pathology. Periodontitis is a prevalent inflammatory disease where Th17 cells mediate key pathological roles, yet whether they exhibit any functional plasticity remains unexplored. We found that during periodontitis, gingival IL-17 fate-mapped T cells still predominantly produce IL-17A, with little diversification of cytokine production. However, plasticity of IL-17 fate-mapped cells did occur during periodontitis, but in the gingiva draining lymph node. Here, some Th17 cells acquired features of Tfh cells, a functional plasticity that was dependent on IL-6. Notably, Th17-to-Tfh diversification was important to limit periodontitis pathology. Preventing Th17-to-Tfh plasticity resulted in elevated periodontal bone loss that was not simply due to increased proportions of conventional Th17 cells. Instead, loss of Th17-to-Tfh cells resulted in reduced IgG levels within the oral cavity and a failure to restrict the biomass of the oral commensal community. Thus, our data identify a novel protective function for a subset of otherwise pathogenic Th17 cells during periodontitis.
Collapse
Affiliation(s)
- Flora A. McClure
- 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
| | - 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
| | - Hayley M. Bridgeman
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Ian E. Prise
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - James I. King
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Shafqat Jaigirdar
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Annie Whelan
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Gareth W. Jones
- Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - John R. Grainger
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Matthew R. Hepworth
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - 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
| |
Collapse
|