1
|
Chen X, Zeng C, Chen X, Sun J, Jin F, Chen Z, Song J. Relationship between three body obesity indicators, WWI, BMI, WtHR, and periodontitis. Sci Rep 2025; 15:697. [PMID: 39753692 DOI: 10.1038/s41598-024-83963-7] [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: 02/03/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025] Open
Abstract
Many scholars have studied the relationship between obesity and periodontitis. The relationship between different obesity indicators and periodontitis seems to vary. In this study, we would like to explore the relationship between three commonly used obesity indicators, WWI (weight-adjusted-waist index), BMI (body mass index), WtHR (waist-to-height ratio), and periodontitis, and try to find the most clinically valuable indicators. The NHANES 2009-2014 provided the data for this study. 10,289, 10,600 and 10,290 participants were included to explore the relationship between WWI, BMI, WtHR and periodontitis respectively. Multiple logistic regression, smooth curve fitting, threshold effects analysis, and subgroup analysis were used to explore their relationships. WWI and periodontitis positively correlated in model III (OR = 1.35; 95% CI, 1.26-1.45, p < 0.001). This result persisted after the quartile for WWI was created. Furthermore, in smooth curve fitting, the relationship between WWI and periodontitis with an inflection point of 12.68 was observed. In the fully adjusted model, WtHR also showed a positive correlation with periodontitis (OR = 2.94; 95% CI, 1.57-5.50, p < 0.001). However, BMI did not express a significant association with periodontitis either as a continuous or categorical variable. This cross-sectional study exposed a positive correlation between WWI, WtHR and periodontitis and this positive correlation showed an age difference.
Collapse
Affiliation(s)
- XingJin Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - ChongWen Zeng
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - XianRun Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - JiangLing Sun
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - FuQian Jin
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Zhu Chen
- Guiyang Stomatological Hospital, 253 Jiefang Road, Nanming District, Guiyang, Guizhou, China.
- School of Stomatology, Zunyi Medical University, Zunyi, China.
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, No. 9, Beijing Road, Yunyan District, Guiyang, 550005, Guizhou, China.
| |
Collapse
|
2
|
Duan ZM, Wu LF. Role of oral-gut-brain axis in psychiatric and neurological disorders. Shijie Huaren Xiaohua Zazhi 2024; 32:878-886. [DOI: 10.11569/wcjd.v32.i12.878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/06/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
The oral cavity and gut are two important microbial habitats in the human body, harboring the most ecologically rich and taxonomically diverse microbial communities in humans, which play a key role in microbiome related diseases. In recent years, the emerging concept of the oral-gut-brain axis has attracted widespread attention in the fields of neuroscience, digestive science, and microbiology. It is not only an anatomical description, but also a comprehensive concept that covers multiple physiological functions and pathological mechanisms. Simply put, the oral-gut-brain axis refers to the complex network that connects the mouth, gut, and brain tightly together through neural connections and hormonal and immune pathways. With the deepening of research on the oral-gut-brain axis theory, more and more evidence shows that it plays an important role in depression, Parkinson's disease, and other neurodegenerative diseases. This article reviews the recent progress in research of the oral-gut-brain axis in psychiatric and neurological diseases.
Collapse
Affiliation(s)
- Zhi-Min Duan
- Department of Gastroenterology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Ling-Fei Wu
- Department of Gastroenterology, The Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
3
|
Cao YH, Zhou YM, Wang SY, Guo J, Cen LS. Promise of the gut microbiota in prevention and traditional Chinese medicine treatment of diabetic peripheral neuropathy. World J Diabetes 2024; 15:2387-2393. [PMID: 39676801 PMCID: PMC11580588 DOI: 10.4239/wjd.v15.i12.2387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/22/2024] [Accepted: 10/12/2024] [Indexed: 11/18/2024] Open
Abstract
The pathogenesis of diabetic peripheral neuropathy (DPN) has not been fully elucidated, and treatment options are limited. Currently, the main strategies for treating DPN are strict glycemic control and symptomatic pain relief. In this editorial, we comment on an article by Li et al, which suggested that modulating the gut microbiota using traditional Chinese medicine (TCM) may be a promising strategy for alleviating DPN symptoms. The regulation of the gut microbiota has received widespread attention in the study of various diseases. TCM can participate in the regulation of gut microbiota through multiple mechanisms, and this regulatory effect can alleviate the clinical symptoms of DPN. We briefly analyzed the promise of the gut microbiota in the early diagnosis, treatment, and clinical efficacy of TCM for DPN. The gut microbiota has potential value at multiple nodes in the occurrence and progression of DPN.
Collapse
Affiliation(s)
- Yuan-Hao Cao
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Yi-Mai Zhou
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Si-Yu Wang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang Province, China
| | - Jing Guo
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| | - Lu-Sha Cen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
4
|
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
|
5
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024; 22:791-805. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
6
|
Gupta U, Dey P. The oral microbial odyssey influencing chronic metabolic disease. Arch Physiol Biochem 2024; 130:831-847. [PMID: 38145405 DOI: 10.1080/13813455.2023.2296346] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 11/30/2023] [Accepted: 12/03/2023] [Indexed: 12/26/2023]
Abstract
INTRODUCTION Since the oral cavity is the gateway to the gut, oral microbes likely hold the potential to influence metabolic disease by affecting the gut microbiota. METHOD A thorough review of literature has been performed to link the alterations in oral microbiota with chronic metabolic disease by influencing the gut microbiota. RESULT A strong correlation exists between abnormalities in oral microbiota and several systemic disorders, such as cardiovascular disease, diabetes, and obesity, which likely initially manifest as oral diseases. Ensuring adequate oral hygiene practices and cultivating diverse oral microflora are crucial for the preservation of general well-being. Oral bacteria have the ability to establish and endure in the gastrointestinal tract, leading to the development of prolonged inflammation and activation of the immune system. Oral microbe-associated prophylactic strategies could be beneficial in mitigating metabolic diseases. CONCLUSION Oral microbiota can have a profound impact on the gut microbiota and influence the pathogenesis of metabolic diseases.
Collapse
Affiliation(s)
- Upasana Gupta
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, India
| |
Collapse
|
7
|
Yu S, Huang F, Huang Y, Yan F, Li Y, Xu S, Zhao Y, Zhang X, Chen R, Chen X, Zhang P. Deciphering the influence of gut and oral microbiomes on menopause for healthy aging. J Genet Genomics 2024:S1673-8527(24)00311-4. [PMID: 39577767 DOI: 10.1016/j.jgg.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Menopause is characterized by the cessation of menstruation and a decline in reproductive function, which is an intrinsic component of the aging process; however, it has been a frequently overlooked field of women's health. The oral and gut microbiota, constituting the largest ecosystem within the human body, are important for maintaining human health and notably contribute to the healthy aging of menopausal women. Thus, a comprehensive review elucidating the impact of the gut and oral microbiota on menopause for healthy aging is of paramount importance. This paper presents the current understanding of the microbiome during menopause, with a particular focus on alterations in the oral and gut microbiota. Our study elucidates the complex interplay between the microbiome and sex hormone levels, explores microbial crosstalk dynamics, and investigates the associations between the microbiome and diseases linked to menopause. Additionally, this review explores the potential of microbiome-targeting therapies for managing menopause-related diseases. Given that menopause can last for approximately 30 years, gaining insights into how the microbiome and menopause interact could pave the way for innovative interventions, which may result in symptomatic relief from menopause and an increase in quality of life in women.
Collapse
Affiliation(s)
- Shuting Yu
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China
| | - Yixuan Huang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Fangxu Yan
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yi Li
- Hunan Agriculture University, Changsha, Hunan 410128, CHINA
| | - Shenglong Xu
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, CHINA
| | - Yao Zhao
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, CHINA
| | - Xinlei Zhang
- Beijing ClouDNA Technology Co., Ltd., Beijing 101407, China
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing 100730, China.
| | - Xingming Chen
- Department of Otolaryngology-Head and Neck Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
8
|
Xi M, Ruan Q, Zhong S, Li J, Qi W, Xie C, Wang X, Abuduxiku N, Ni J. Periodontal bacteria influence systemic diseases through the gut microbiota. Front Cell Infect Microbiol 2024; 14:1478362. [PMID: 39619660 PMCID: PMC11604649 DOI: 10.3389/fcimb.2024.1478362] [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: 08/09/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Many systemic diseases, including Alzheimer disease (AD), diabetes mellitus (DM) and cardiovascular disease, are associated with microbiota dysbiosis. The oral and intestinal microbiota are directly connected anatomically, and communicate with each other through the oral-gut microbiome axis to establish and maintain host microbial homeostasis. In addition to directly, periodontal bacteria may also be indirectly involved in the regulation of systemic health and disease through the disturbed gut. This paper provides evidence for the role of periodontal bacteria in systemic diseases via the oral-gut axis and the far-reaching implications of maintaining periodontal health in reducing the risk of many intestinal and parenteral diseases. This may provide insight into the underlying pathogenesis of many systemic diseases and the search for new preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Mengying Xi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Qijun Ruan
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Sulan Zhong
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Jiatong Li
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Weijuan Qi
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Congman Xie
- Department of Orthodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Xiaoyan Wang
- Department of Periodontics, Shenzhen Longgang Otolaryngology hospital, Shenzhen, China
| | - Nuerbiya Abuduxiku
- Department of Stomatology, The First People’s Hospital of Kashi, Kashi, China
| | - Jia Ni
- Department of Periodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Liang J, Ren Y, Zheng Y, Lin X, Song W, Zhu J, Zhang X, Zhou H, Wu Q, He Y, Yin J. Functional Outcome Prediction of Acute Ischemic Stroke Based on the Oral and Gut Microbiota. Mol Neurobiol 2024:10.1007/s12035-024-04618-2. [PMID: 39546118 DOI: 10.1007/s12035-024-04618-2] [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: 04/02/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Although several studies have identified a distinct gut microbiota in individuals with acute ischemic stroke (AIS), there is a limited amount of research that has simultaneously investigated alterations in the oral and intestinal microbiota in AIS patients and their correlation with clinical prognosis. This was a prospective and observational single-center cohort study in which we included 160 AIS patients who were admitted within 24 h after a stroke event. We collected oral and rectal swab samples for analysis using 16S rRNA high-throughput sequencing. Our study revealed that patients with unfavorable outcomes after AIS showed early disruptions in their oral and intestinal microbiota. Rectal swabs showed increased levels of facultatively anaerobic bacteria in patients with a poor prognosis, while the oral cavity exhibited higher levels of anaerobic and opportunistic pathogenic bacteria. By employing machine learning analysis, we found that the microbiota composition at both rectal and oral sites could predict early and long-term outcomes. Moreover, patients with a poor prognosis displayed increased oral bacterial colonization in the rectal microbiota and altered interactions between the oral and gut microbiota. This study reveals distinct rectal and oral bacteria that could predict unfavorable outcomes for AIS patients. Monitoring the microbiota of various body sites during the early stages after admission may hold prognostic value and inform personalized treatment strategies. The presence of oral bacteria colonizing the intestines during the acute phase of stroke could serve as an early indication of poor outcomes for AIS patients.
Collapse
Affiliation(s)
- Jingru Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yueran Ren
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaofei Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Song
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiajia Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaomei Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong, China
| | - Qiheng Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China.
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong, China.
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
10
|
Sun J, Wang X, Xiao J, Yang Q, Huang X, Yang Z, Liu H, Liu Y, Wang H, Huang Z, Ma L, Cao Z. Autophagy mediates the impact of Porphyromonas gingivalis on short-chain fatty acids metabolism in periodontitis-induced gut dysbiosis. Sci Rep 2024; 14:26291. [PMID: 39487211 PMCID: PMC11530519 DOI: 10.1038/s41598-024-77909-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis), the main pathogen responsible for periodontitis, is linked to systemic disorders via the oral-gut axis. Short-chain fatty acids (SCFAs) are vital for gut health, but their role in P. gingivalis-induced gut disorders remains unclear. This study utilized metabolomics and 16 S rRNA sequencing to explore gut microbiota and SCFAs levels in P. gingivalis-induced periodontitis mouse models. Significant changes were observed in gut, including a reduction in SCFAs-producing bacteria, such as Lactobacillus, Ligilactobacillus, Allobucalum, and a notable decrease in Firmicutes and Actinobacteriota. The intestinal permeability tests and histological analyses revealed that periodontitis led to epithelial inflammation, reduced mucin secretion, and compromised gut barrier integrity. In vitro experiments with Caco-2 cells co-cultured with P. gingivalis showed that the bacterium disrupted cellular junctions by impairing autophagy, specifically through the ATG5-LC3 pathway, leading to decreased expression of tight junction proteins and reduced SCFA absorption. Remarkably, rapamycin treatment both in vitro and in vivo restored gut barrier function by enhancing autophagy, increasing tight junction protein expression, and promoting SCFAs absorption via MCT1 and SMCT1, alongside GPR43/GPR109a pathway activation. These findings reveal autophagy's novel role in regulating SCFAs metabolism in P. gingivalis-induced gut dysbiosis, offering insights for preventing and treating periodontitis-related systemic diseases.
Collapse
Affiliation(s)
- Jiahui 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, China
| | - Xiaoxuan 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, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junhong Xiao
- 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
| | - Qiudong 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, China
| | - Xin Huang
- 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 Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengkun 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, China
| | - Heyu Liu
- 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
| | - Yuqi Liu
- 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
| | - Huiyi 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, China
| | - Zhendong Huang
- 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
| | - Li Ma
- 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 Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhengguo Cao
- 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 Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Gan G, Luo Y, Zeng Y, Lin S, Lu B, Zhang R, Chen S, Lei H, Cai Z, Huang X. Gut microbiota dysbiosis links chronic apical periodontitis to liver fibrosis in nonalcoholic fatty liver disease: Insights from a mouse model. Int Endod J 2024; 57:1608-1622. [PMID: 38958220 DOI: 10.1111/iej.14119] [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: 01/11/2024] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024]
Abstract
AIM In this study, we investigated the systemic implications of chronic apical periodontitis (CAP). CAP may contribute to the nonalcoholic fatty liver disease (NAFLD) progression through the gut microbiota and its metabolites, which are related to the degree of fibrosis. METHODOLOGY Sixteen 7-week-old male apolipoprotein E knockout (apoE-/-) mice were randomly divided into two groups: the CAP and Con groups. A CAP model was established by sealing the first- and second-maxillary molars with bacterium-containing cotton balls. Apical lesions were evaluated by micro-CT. Histological evaluations of NAFLD were performed using second harmonic generation/two-photon excitation fluorescence (SHG/TPEF) assays. Additionally, we comprehensively analyzed the gut microbiota using 16S rRNA gene sequencing and explored metabolic profiles by liquid chromatography-mass spectrometry (LC-MS). Immunofluorescence analysis was used to examine the impact of CAP on tight junction proteins and mucin expression. Transcriptome assays have elucidated gene expression alterations in liver tissues. RESULTS Micro-CT scans revealed an evident periapical bone loss in the CAP group, and the total collagen percentage was increased (Con, 0.0361 ± 0.00510%, CAP, 0.0589 ± 0.00731%, p < .05). 16S rRNA sequencing revealed reduced diversity and distinct taxonomic enrichment in the CAP group. Metabolomic assessments revealed that differentially enriched metabolites, including D-galactosamine, were enriched and that 16-hydroxyhexadecanoic acid and 3-methylindole were depleted in the CAP group. Immunofluorescence analyses revealed disruptions in tight junction proteins and mucin production, indicating intestinal barrier integrity disruption. Liver transcriptome analysis revealed upregulation of Lpin-1 expression in the CAP group. CONCLUSION This study provides comprehensive evidence of the systemic effects of CAP on liver fibrosis in NAFLD patients by elucidating alterations in the gut microbiota composition and metabolism.
Collapse
Affiliation(s)
- Guowu Gan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yufang Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yu Zeng
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shihan Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Beibei Lu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ren Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Huaxiang Lei
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatology Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
12
|
Zhang H, Lin X. Research progress on the potential correlation between polycystic ovary syndrome and periodontal disease. J Int Med Res 2024; 52:3000605241300096. [PMID: 39600040 PMCID: PMC11603522 DOI: 10.1177/03000605241300096] [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: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024] Open
Abstract
Over the past few years, the correlation between periodontal disease (PD) and polycystic ovary syndrome (PCOS) has attracted widespread attention owing to the increased incidence of these diseases. Several studies have suggested a possible link between the two. In this narrative review, we examined the epidemiology, common risk factors, and pathological mechanisms of PCOS and PD to investigate the potential association between these diseases. Evidence from the literature indicates that PCOS and PD can interact with each other. Common risk factors, such as microbial homeostasis imbalance owing to dysbiosis, along with multiple hormone and inflammatory mediators, as well as inflammatory responses owing to oxidative stress and oxidative responses owing to ferroptosis, are all associated with the pathogenesis of both diseases. Further studies are needed to clarify the specific mechanisms of interaction between PCOS and PD, which could clarify future directions in disease management and combined multidisciplinary treatment.
Collapse
Affiliation(s)
- Huishan Zhang
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoping Lin
- Department of Stomatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
13
|
Qian X, Lin X, Hu W, Zhang L, Chen W, Zhang S, Ge S, Xu X, Luo K. Intestinal homeostasis disrupted by Periodontitis exacerbates Alzheimer's Disease in APP/PS1 mice. J Neuroinflammation 2024; 21:263. [PMID: 39425119 PMCID: PMC11489998 DOI: 10.1186/s12974-024-03256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
Periodontitis exacerbates Alzheimer's disease (AD) through multiple pathways. Both periodontitis and AD are intricately correlated to intestinal homeostasis, yet there is still a lack of direct evidence regarding whether periodontitis can regulate the progression of AD by modulating intestinal homeostasis. The current study induced experimental periodontitis in AD mice by bilaterally ligating the maxillary second molars with silk and administering Pg-LPS injections in APPswe/PS1ΔE9 (APP/PS1) mice. Behavioral tests and histological analyses of brain tissue were conducted after 8 weeks. Gut microbiota was analyzed and colon tissue were also evaluated. Then, fecal microbiota from mice with periodontitis was transplanted into antibiotic-treated mice to confirm the effects of periodontitis on AD and the potential mechanism was explored. The results indicated periodontitis exacerbated cognitive impairment and anxious behaviour in APP/PS1 mice, with increased Aβ deposition, microglial overactivation and neuroinflammation in brain. Moreover, the intestinal homeostasis of AD mice was altered by periodontitis, including affecting gut microbiota composition, causing colon inflammation and destroyed intestinal epithelial barrier. Furthermore, AD mice that underwent fecal transplantation from mice with periodontitis exhibited worsened AD progression and disrupted intestinal homeostasis. It also impaired intestinal barrier function, elevated peripheral inflammation, damaged blood-brain barrier (BBB) and caused neuroinflammation and synapses impairment. Taken together, the current study demonstrated that periodontitis could disrupt intestinal homeostasis to exacerbate AD progression potential via causing gut microbial dysbiosis, intestinal inflammation and intestinal barrier impairment to induce peripheral inflammation and damage BBB, ultimately leading to neuroinflammation and synapse impairment. It underscores the importance of maintaining both periodontal health and intestinal homeostasis to reduce the risk of AD.
Collapse
Affiliation(s)
- Xueshen Qian
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Xuxin Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Weiqiang Hu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Wenqian Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Shuang Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, Jiangsu, 210008, P.R. China
| | - Song Ge
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, 563003, P.R. China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| |
Collapse
|
14
|
Catalan EA, Seguel-Fuentes E, Fuentes B, Aranguiz-Varela F, Castillo-Godoy DP, Rivera-Asin E, Bocaz E, Fuentes JA, Bravo D, Schinnerling K, Melo-Gonzalez F. Oral Pathobiont-Derived Outer Membrane Vesicles in the Oral-Gut Axis. Int J Mol Sci 2024; 25:11141. [PMID: 39456922 PMCID: PMC11508520 DOI: 10.3390/ijms252011141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Oral pathobionts are essential in instigating local inflammation within the oral cavity and contribute to the pathogenesis of diseases in the gastrointestinal tract and other distant organs. Among the Gram-negative pathobionts, Porphyromonas gingivalis and Fusobacterium nucleatum emerge as critical drivers of periodontitis, exerting their influence not only locally but also as inducers of gut dysbiosis, intestinal disturbances, and systemic ailments. This dual impact is facilitated by their ectopic colonization of the intestinal mucosa and the subsequent mediation of distal systemic effects by releasing outer membrane vesicles (OMVs) into circulation. This review elucidates the principal components of oral pathobiont-derived OMVs implicated in disease pathogenesis within the oral-gut axis, detailing virulence factors that OMVs carry and their interactions with host epithelial and immune cells, both in vitro and in vivo. Additionally, we shed light on the less acknowledged interplay between oral pathobionts and the gut commensal Akkermansia muciniphila, which can directly impede oral pathobionts' growth and modulate bacterial gene expression. Notably, OMVs derived from A. muciniphila emerge as promoters of anti-inflammatory effects within the gastrointestinal and distant tissues. Consequently, we explore the potential of A. muciniphila-derived OMVs to interact with oral pathobionts and prevent disease in the oral-gut axis.
Collapse
Affiliation(s)
- Eduardo A. Catalan
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Emilio Seguel-Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Brandon Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Aranguiz-Varela
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Daniela P. Castillo-Godoy
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elizabeth Rivera-Asin
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elisa Bocaz
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Denisse Bravo
- Cellular Interactions Laboratory, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370133, Chile;
| | - Katina Schinnerling
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Melo-Gonzalez
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| |
Collapse
|
15
|
Ge J, Zhu X, Weng C, Yuan D, Zhao J, Zhao L, Wang T, Wu Y. Periodontitis impacts on thrombotic diseases: from clinical aspect to future therapeutic approaches. Int J Oral Sci 2024; 16:58. [PMID: 39402049 PMCID: PMC11473739 DOI: 10.1038/s41368-024-00325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/08/2024] [Accepted: 09/12/2024] [Indexed: 10/17/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease initiated by biofilm microorganisms and mediated by host immune imbalance. Uncontrolled periodontal infections are the leading cause of tooth loss in adults. Thrombotic diseases can lead to partial or complete obstruction of blood flow in the circulatory system, manifesting as organ or tissue ischemia and necrosis in patients with arterial thrombosis, and local edema, pain and circulatory instability in patients with venous thrombosis, which may lead to mortality or fatality in severe case. Recent studies found that periodontitis might enhance thrombosis through bacterial transmission or systemic inflammation by affecting platelet-immune cell interactions, as well as the coagulation, and periodontal therapy could have a prophylactic effect on patients with thrombotic diseases. In this review, we summarized clinical findings on the association between periodontitis and thrombotic diseases and discussed several novel prothrombotic periodontitis-related agents, and presented a perspective to emphasize the necessity of oral health management for people at high risk of thrombosis.
Collapse
Affiliation(s)
- Jinting Ge
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuanzhi Zhu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengxin Weng
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ding Yuan
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jichun Zhao
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tiehao Wang
- Division of Vascular Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
| | - Yafei Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, China.
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
16
|
Da J, Ren Y, Liu S, Wang N, Wang L, Fu Z, Liang Y, Pan Y, Li J, Chen J. Effect of the gut microbiome and inflammation-related proteins on oral leukoplakia: a Mendelian randomization study and mediation analysis. Front Oncol 2024; 14:1443123. [PMID: 39386195 PMCID: PMC11461448 DOI: 10.3389/fonc.2024.1443123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Background Oral leukoplakia (OL) is the most common potentially malignant disease of the oral cavity. In recent years, studies have identified a correlation between the gut microbiota (GM) and oral cancer, in addition, inflammation-related proteins have been reported to play an important role in the development of OL. However, the causal relationship between gut microbiota and OL, as well as whether cytokines play a mediating role, remain unclear. Methods In this Mendelian randomization (MR) study, the genome-wide association studies (GWAS) (n=18340) of the MiBioGen consortium microbiome was used as exposure data. Genetic variation data related to OL were extracted from the Finngen R9 project (513 cases of OL and 411668 controls). The 91 inflammation-related proteins obtained in the literature serve as potential mediators. Two-sample Mendelian randomization analysis was applied to infer causality using Inverse Variance Weighted (IVW), MR Egger, weighted media, simple mode and weighted mode method. Subsequently, sensitivity analysis was conducted to ensure the robustness of the MR results. In addition, we conducted reverse MR analysis to alleviate reverse causality. Finally, we used mediation analysis to determine the pathway mediated by inflammation-related proteins from the gut microbiota to OL. Results The five bacterial taxa in the gut microbiota indicate a potential causal relationship with the development of OL. Notably, family Clostridiaceae1 was negatively correlated with the risk of OL development, while genus Dorea, genus Ruminococcus1, genus Senegalimasilia and genus Veillonella were positively associated with the risk of OL development. In addition, this study identified a potential causal relationship between interleukin-10 receptor subunit alpha (IL-10RA), interleukin-18 receptor 1(IL18-R1) and the occurrence of OL. In addition, intermediary analysis indicates that IL18-R1 mediated the pathway between the gut microbiota genus Senegalimasilia and OL. Conclusions In summary, our research emphasize the complex relationship between gut microbiota, inflammation-related proteins and OL. The identified associations and mediating effects provide new insights into potential therapeutic approaches for targeting the gut microbiota in the management of OL, and contribute to its prevention, diagnosis and treatment.
Collapse
Affiliation(s)
- Junlong Da
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Yinting Ren
- Department of Stomatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, China
| | - Shiwei Liu
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Nanyan Wang
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Lei Wang
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Zhifeng Fu
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Yongtang Liang
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Yu Pan
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Jin Li
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| | - Jufeng Chen
- Department of Stomatology Center, The First People’s Hospital of Foshan, Foshan, China
| |
Collapse
|
17
|
Yuan Y, Miao X, Hou Y, Huang Y, Qiu B, Shi W. Association between sleep and periodontitis: NHANES 2009-2014 and Mendelian randomization study. Cranio 2024:1-10. [PMID: 39321071 DOI: 10.1080/08869634.2024.2406737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
OBJECTIVE We assessed association between periodontitis and sleep patterns. METHODS Our study included 7289 participants based on a large-scale study in NHANES. Periodontitis was defined through an oral examination. We used logistic regression to explore association between sleep status and risk of periodontitis. Further, stratified analysis was conducted. We performed a Mendelian randomization (MR) analysis using publicly available genetic data from corresponding studies. RESULTS Odds Ratio (OR) of periodontitis was 1.25 (1.07-1.46) in individuals with < 7 hours/per night. Stratification analysis showed individuals under 45 years old (OR: 1.28, 95% CI: 1.07-1.54), women (1.28(1.07-1.54)), education levels below high school (1.45(1.03-2.05)) and higher family income (-1.28(1.06-1.56)) were more likely to deteriorate to periodontitis if sleep time less than 7 hours. IVW-MR showed periodontitis risk was not associated with genetically increased levels of sleep time (0.22 (0.02-3.05)) or insomnia (0.83(0.55-1.24)). CONCLUSION There was no indication that sleep status was associated with periodontitis.
Collapse
Affiliation(s)
- Yong Yuan
- Clinical Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xujie Miao
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Yingqi Hou
- Department of Laboratory Medicine, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yanqiu Huang
- School of public health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beibei Qiu
- Department of Chronic Communicable Disease, Nanjing Municipal Center for Disease Control and Prevention, Nanjing, China
| | - Wentao Shi
- Clinical Research Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Kerstens R, Ng YZ, Pettersson S, Jayaraman A. Balancing the Oral-Gut-Brain Axis with Diet. Nutrients 2024; 16:3206. [PMID: 39339804 PMCID: PMC11435118 DOI: 10.3390/nu16183206] [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: 08/04/2024] [Revised: 09/14/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Background: The oral microbiota is the second largest microbial community in humans. It contributes considerably to microbial diversity and health effects, much like the gut microbiota. Despite physical and chemical barriers separating the oral cavity from the gastrointestinal tract, bidirectional microbial transmission occurs between the two regions, influencing overall host health. Method: This review explores the intricate interplay of the oral-gut-brain axis, highlighting the pivotal role of the oral microbiota in systemic health and ageing, and how it can be influenced by diet. Results: Recent research suggests a relationship between oral diseases, such as periodontitis, and gastrointestinal problems, highlighting the broader significance of the oral-gut axis in systemic diseases, as well as the oral-gut-brain axis in neurological disorders and mental health. Diet influences microbial diversity in the oral cavity and the gut. While certain diets/dietary components improve both gut and oral health, others, such as fermentable carbohydrates, can promote oral pathogens while boosting gut health. Conclusions: Understanding these dynamics is key for promoting a healthy oral-gut-brain axis through dietary interventions that support microbial diversity and mitigate age-related health risks.
Collapse
Affiliation(s)
- Rebecca Kerstens
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Yong Zhi Ng
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
- Duke-NUS Medical School, 8 College Rd., Singapore 169857, Singapore
| | - Sven Pettersson
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
- Faculty of Medical Sciences, Sunway University, Subang Jaya 47500, Selangor, Malaysia
- Department of Microbiology and Immunology, National University Singapore, Singapore 117545, Singapore
| | - Anusha Jayaraman
- ASEAN Microbiome Nutrition Centre, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| |
Collapse
|
19
|
Kozman R, Mussie KM, Elger B, Wienand I, Jotterand F. Ethical Challenges in Oral Healthcare Services Provided by Non-Governmental Organizations for Refugees in Germany. JOURNAL OF BIOETHICAL INQUIRY 2024; 21:491-500. [PMID: 38353921 DOI: 10.1007/s11673-023-10327-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2024]
Abstract
Oral healthcare is attracting much attention after decades of neglect from policymakers. Recent studies have shown a strong association between oral and overall health, which can lead to serious health problems. Availability of oral healthcare services is an essential part of ensuring universal healthcare coverage. More importantly, current gaps in its accessibility by minority or marginalized population groups are crucial public health as well as ethical concerns. One notable effort to address this issue comes from Non-Governmental Organizations (NGOs), which offer oral healthcare services for non-insured refugees. However, the challenge remains that these care services are not comprehensive, which has implications for the refugees' oral and general health. In this article, we discuss this complex issue in the German healthcare context by including ethical reflections. Therefore, the purpose of this article is to discuss the ethical challenges related to oral healthcare services provided by NGOs for refugees in Germany. First, we will introduce the general oral healthcare context worldwide and in Germany. Second, we will provide a general description of the oral healthcare services provided by NGOs for refugees in Germany, as well as an overview of existing gaps. This will provide us with the context for our third and most important task-discussing the ethical implications of the gaps. In doing so, and since the ethical implications can be several, we demarcate the scope of our analysis by focusing on the specific ethical issues of justice, harm, and autonomy. Finally, we offer some recommendations for how to move forward.
Collapse
Affiliation(s)
- R Kozman
- Institute for Biomedical Ethics, University of Basel, 4056, Basel, Switzerland.
| | - K M Mussie
- Institute for Biomedical Ethics, University of Basel, 4056, Basel, Switzerland
| | - B Elger
- Institute for Biomedical Ethics, University of Basel, 4056, Basel, Switzerland
| | - I Wienand
- Institute for Biomedical Ethics, University of Basel, 4056, Basel, Switzerland
| | - F Jotterand
- Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
Li J, O’Toole PW. Disease-associated microbiome signature species in the gut. PNAS NEXUS 2024; 3:pgae352. [PMID: 39228810 PMCID: PMC11370893 DOI: 10.1093/pnasnexus/pgae352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/12/2024] [Indexed: 09/05/2024]
Abstract
There is an accumulation of evidence that the human gut microbiota plays a role in maintaining health, and that an altered gut microbiota (sometimes called dysbiosis) associates with risk for many noncommunicable diseases. However, the dynamics of disease-linked bacteria in the gut and other body sites remain unclear. If microbiome alterations prove causative in particular diseases, therapeutic intervention may be possible. Furthermore, microbial signature taxa have been established for the diagnosis of some diseases like colon cancer. We identified 163 disease-enriched and 98 disease-depleted gut microbiome signature taxa at species-level resolution (signature species) from 10 meta-analyses of multiple diseases such as colorectal cancer, ulcerative colitis, Crohn's disease, irritable bowel syndrome, pancreatic cancer, and COVID-19 infection. Eight signature species were enriched and nine were depleted across at least half of the diseases studied. Compared with signature species depleted in diseases, a significantly higher proportion of disease-enriched signature species were identified as extra-intestinal (primarily oral) inhabitants, had been reported in bacteremia cases from the literature, and were aerotolerant anaerobes. These findings highlight the potential involvement of oral microbes, bacteremia isolates, and aerotolerant anaerobes in disease-associated gut microbiome alterations, and they have implications for patient care and disease management.
Collapse
Affiliation(s)
- Junhui Li
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- School of Microbiology, University College Cork, Cork T12 K8AF, Ireland
| | - Paul W O’Toole
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland
- School of Microbiology, University College Cork, Cork T12 K8AF, Ireland
| |
Collapse
|
21
|
Leonov G, Salikhova D, Starodubova A, Vasilyev A, Makhnach O, Fatkhudinov T, Goldshtein D. Oral Microbiome Dysbiosis as a Risk Factor for Stroke: A Comprehensive Review. Microorganisms 2024; 12:1732. [PMID: 39203574 PMCID: PMC11357103 DOI: 10.3390/microorganisms12081732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Stroke represents a significant global health burden, with a substantial impact on mortality, morbidity, and long-term disability. The examination of stroke biomarkers, particularly the oral microbiome, offers a promising avenue for advancing our understanding of the factors that contribute to stroke risk and for developing strategies to mitigate that risk. This review highlights the significant correlations between oral diseases, such as periodontitis and caries, and the onset of stroke. Periodontal pathogens within the oral microbiome have been identified as a contributing factor in the exacerbation of risk factors for stroke, including obesity, dyslipidemia, atherosclerosis, hypertension, and endothelial dysfunction. The alteration of the oral microbiome may contribute to these conditions, emphasizing the vital role of oral health in the prevention of cardiovascular disease. The integration of dental and medical health practices represents a promising avenue for enhancing stroke prevention efforts and improving patient outcomes.
Collapse
Affiliation(s)
- Georgy Leonov
- Federal Research Center of Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia;
| | - Diana Salikhova
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| | - Antonina Starodubova
- Federal Research Center of Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia;
- Therapy Faculty, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Andrey Vasilyev
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
- E.V. Borovsky Institute of Dentistry, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
- Central Research Institute of Dental and Maxillofacial Surgery, 119021 Moscow, Russia
| | - Oleg Makhnach
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| | - Timur Fatkhudinov
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| |
Collapse
|
22
|
Sun X, Wu T, Yang Z, Chen S, Zhao Z, Hu C, Wu S, Wu J, Mao Y, Liu J, Guo C, Cao G, Xu X, Huang S, Liang G. Regulatory role of PDK1 via integrated gene analysis of mitochondria-immune response in periodontitis. Gene 2024; 918:148476. [PMID: 38657876 DOI: 10.1016/j.gene.2024.148476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
AIMS To investigate the association between mitochondrial events and immune response in periodontitis and related regulatory genes. MAIN METHODS Gene expression profiles in gingival tissues were retrieved from the Gene Expression Omnibus. Mitochondria-immune response-related differentially expressed genes (MIR-DEGs) between the healthy and periodontitis samples were determined. WGCNA, GO, and KEGG were used to investigate the function and the enriched pathways of MIR-DEGs. The correlation between MIR-DEGs expression and clinical probing pocket depth was analyzed. The MIR-DEGs were further identified and verified in animal samples. A periodontitis model was established in C57BL/6 mice with silk ligation. Micro-computed tomography was used to assess alveolar bone loss. Western blot, quantitative real-time polymerase chain reaction, and immunohistochemical analyses further validated the differential expression of the MIR-DEGs. KEY FINDINGS A total of ten MIR-DEGs (CYP24A1, PRDX4, GLDC, PDK1, BCL2A1, CBR3, ARMCX3, BNIP3, IFI27, and UNG) were identified, the expression of which could effectively distinguish patients with periodontitis from the healthy controls. Enhanced immune response was detected in the periodontitis group with that in the healthy controls, especially in B cells. PDK1 was a critical MIR-DEG correlated with B cell immune response and clinical periodontal probing pocket depth. Both animal and clinical periodontal samples presented higher gene and protein expression of PDK1 than the control samples. Additionally, PDK1 colocalized with B cells in both animal and clinical periodontal tissues. SIGNIFICANCE Mitochondria participate in the regulation of the immune response in periodontitis. PDK1 may be the key mitochondria-related gene regulating B-cell immune response in periodontitis.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China; 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
| | - Tong Wu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhan Yang
- 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
| | - Zheyu Zhao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chaoming Hu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengzhuang Wu
- School and Hospital of Stomatology, Hangzhou Medical University, Wenzhou, China
| | - Jiayu Wu
- School of Medicine, Jiujiang University, 320 Xunyang East Road, Jiujiang City, Jiangxi Province 332000, China
| | - Yixin Mao
- 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
| | - Jiefan Liu
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China; Department of Oral and Maxillofacial Surgery/Pathology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chen Guo
- School/Hospital of Stomatology, Lanzhou University, Lanzhou 730000, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Xiangwei Xu
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.
| | - Guang Liang
- Affiliated Yongkang First People's Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310012, China.
| |
Collapse
|
23
|
Qing X, Zhang C, Zhong Z, Zhang T, Wang L, Fang S, Jiang T, Luo X, Yang Y, Song G, Wei W. Causal Association Analysis of Periodontitis and Inflammatory Bowel Disease: A Bidirectional Mendelian Randomization Study. Inflamm Bowel Dis 2024; 30:1251-1257. [PMID: 38408068 PMCID: PMC11291616 DOI: 10.1093/ibd/izad188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Indexed: 02/28/2024]
Abstract
BACKGROUND Periodontitis has been reported to be associated with inflammatory bowel disease (IBD), including ulcerative colitis (UC), and Crohn's disease (CD). However, the causality of these 2 diseases remains unclear. We conducted bidirectional Mendelian randomization (MR) to investigate the causal relationship between periodontitis and IBD. METHODS We obtained the genome-wide association study (GWAS) summary data of European populations from FinnGen database (for IBD) and a published article (for periodontitis), from which independent single nucleotide polymorphisms were selected as instrumental variables. Inverse variance-weighted (IVW), MR-Egger, and weighted median (WM) methods were utilized for MR analysis. Heterogeneity or pleiotropy was detected through Cochran's Q test and MR-Egger intercept, respectively. Outlier was identified with MR-PRESSO (Mendelian Randomization Pleiotropy RESidual Sum and Outlier) and leave-one-out analysis. All statistical analyses were performed with R 4.2.1 and the packages of TwoSampleMR version 0.5.6. RESULTS Genetic prediction showed that periodontitis was the risk factor of UC (odds ratio [OR], 1.13; 95% confidence interval [CI], 1.01-1.26; P = .027), rather than of CD (OR, 0.92; 95% CI, 0.74-1.15; P = .456) and IBD (OR, 0.96; 95% CI, 0.81-1.13; P = .619). To the contrary, CD, not UC or IBD, resulted in exacerbating periodontitis in terms of the results of the IVW (OR, 1.09; 95% CI, 1.01-1.17; P = .021) and WM (OR, 1.10; 95% CI, 1.01-1.20; P = .030) methods. Heterogeneity or pleiotropy was acceptable. CONCLUSIONS Our results indicated that CD was the risk factor for periodontitis; conversely, periodontitis was responsible for the exacerbation of UC, enhancing the existence of mouth-gut axis. Patients with UC should pay more attention to periodontal health, while patients with periodontitis should actively pay close heed to intestinal health.
Collapse
Affiliation(s)
- Xiangli Qing
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenhui Zhang
- Department of General Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Zhuotai Zhong
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tao Zhang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lin Wang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuangshuang Fang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tianyuan Jiang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoying Luo
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Yang
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Gengqing Song
- Department of Gastroenterology and Hepatology, MetroHealth Medical Center/Case Western Reserve University, Cleveland, OH, USA
| | - Wei Wei
- Department of Gastroenterology, Beijing Key Laboratory of Functional Gastrointestinal Disorders Diagnosis and Treatment of Traditional Chinese Medicine, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
24
|
Mo C, Huang M, Yan F, Song M, Fan J, Zhang J. Correlation Between Gut Microbiota Composition and Serum Interleukin 17 (IL-17) in Mice With Type 2 Diabetes and Experimental Periodontitis. Cureus 2024; 16:e68005. [PMID: 39211822 PMCID: PMC11360949 DOI: 10.7759/cureus.68005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Objective To preliminarily explore the composition characteristics of gut microbiota in mice with type 2 diabetes mellitus (T2DM) and experimental periodontitis, and their correlation with serum IL-17 levels, aiming to provide new insights and evidence for related experimental studies. Methods A total of 42 SPF-grade C57BL/6J mice were randomly selected, with 24 used for T2DM modeling. Successfully modeled T2DM mice were divided into the T2DM group (ND group, n=8) and T2DM with experimental periodontitis group (PD group, n=8). Non-T2DM mice were divided into the blank control group (NC group, n=8) and the experimental periodontitis group (NP group, n=8). After modeling, body weight and fasting plasma glucose (FPG) were measured weekly. Each group of mice underwent an oral glucose tolerance test (OGTT) and an insulin tolerance test (ITT). Six weeks after modeling experimental periodontitis, serum IL-17 levels were measured using ELISA, intestinal inflammation was assessed using HE staining, and gut microbiota composition in cecal contents was analyzed by 16S rRNA sequencing to determine its correlation with serum IL-17 levels. Results FPG in the PD group was higher than in the ND group, with a statistically significant difference in the 12th week (p<0.05). The glucose tolerance level in the PD group was lower than in the ND group (p<0.01). Compared with the NC group, other groups showed varying degrees of inflammatory cell infiltration in the intestinal mucosa, and serum IL-17 levels were lower in both the ND and PD groups compared to the NC group (p<0.01), with the PD group also lower than the NP group (p<0.01). The Shannon and Pielou-e indices of gut microbiota in the PD group were significantly lower than those in the NP group (p<0.05). In terms of microbiota composition, Firmicutes were increased in both the ND and PD groups compared to the NC and NP groups (p<0.05), while Bacteroidetes were decreased (p<0.05). Proteobacteria were increased in the PD group compared to the ND group (p<0.05). The abundance of Bacteroidetes and the Bacteroidetes/Firmicutes ratio was moderately positively correlated with serum IL-17 levels (p<0.01) and moderately negatively correlated with blood glucose levels (p<0.01); serum IL-17 levels were strongly negatively correlated with blood glucose levels (p<0.01). Conclusion Comorbidity of experimental periodontitis and T2DM may exacerbate glucose metabolism impairment in T2DM mice by increasing the abundance of Proteobacteria and intestinal mucosal damage. Serum IL-17 levels may serve as an indicator of gut microbiota dysbiosis in T2DM mice with experimental periodontitis.
Collapse
Affiliation(s)
- Chaolun Mo
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, CHN
| | - Mingkun Huang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, CHN
| | - Fuhua Yan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing University Medical School & Nanjing University Institute of Stomatology, Nanjing, CHN
| | - Minghui Song
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, CHN
| | - Jiabing Fan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, CHN
| | - Junmei Zhang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, CHN
| |
Collapse
|
25
|
Rocha CM, Kawamoto D, Martins FH, Bueno MR, Ishikawa KH, Ando-Suguimoto ES, Carlucci AR, Arroteia LS, Casarin RV, Saraiva L, Simionato MRL, Mayer MPA. Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes. Int J Mol Sci 2024; 25:8090. [PMID: 39125663 PMCID: PMC11312116 DOI: 10.3390/ijms25158090] [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: 04/15/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 08/12/2024] Open
Abstract
Oral bacteria are implicated not only in oral diseases but also in gut dysbiosis and inflammatory conditions throughout the body. The periodontal pathogen Aggregatibacter actinomycetemcomitans (Aa) often occurs in complex oral biofilms with Streptococcus gordonii (Sg), and this interaction might influence the pathogenic potential of this pathogen. This study aims to assess the impact of oral inoculation with Aa, Sg, and their association (Aa+Sg) on alveolar bone loss, oral microbiome, and their potential effects on intestinal health in a murine model. Sg and/or Aa were orally administered to C57Bl/6 mice, three times per week, for 4 weeks. Aa was also injected into the gingiva three times during the initial experimental week. After 30 days, alveolar bone loss, expression of genes related to inflammation and mucosal permeability in the intestine, serum LPS levels, and the composition of oral and intestinal microbiomes were determined. Alveolar bone resorption was detected in Aa, Sg, and Aa+Sg groups, although Aa bone levels did not differ from that of the SHAM-inoculated group. Il-1β expression was upregulated in the Aa group relative to the other infected groups, while Il-6 expression was downregulated in infected groups. Aa or Sg downregulated the expression of tight junction genes Cldn 1, Cldn 2, Ocdn, and Zo-1 whereas infection with Aa+Sg led to their upregulation, except for Cldn 1. Aa was detected in the oral biofilm of the Aa+Sg group but not in the gut. Infections altered oral and gut microbiomes. The oral biofilm of the Aa group showed increased abundance of Gammaproteobacteria, Enterobacterales, and Alloprevotella, while Sg administration enhanced the abundance of Alloprevotella and Rothia. The gut microbiome of infected groups showed reduced abundance of Erysipelotrichaceae. Infection with Aa or Sg disrupts both oral and gut microbiomes, impacting oral and gut homeostasis. While the combination of Aa with Sg promotes Aa survival in the oral cavity, it mitigates the adverse effects of Aa in the gut, suggesting a beneficial role of Sg associations in gut health.
Collapse
Affiliation(s)
- Catarina Medeiros Rocha
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Fernando Henrique Martins
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Manuela Rocha Bueno
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Division of Periodontics, Faculdade São Leopoldo Mandic, São Leopoldo Mandic Research Institute, Campinas 13045-755, SP, Brazil
| | - Karin H. Ishikawa
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Ellen Sayuri Ando-Suguimoto
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Aline Ramos Carlucci
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Leticia Sandoli Arroteia
- Department of Prosthesis and Periodontology, School of Dentistry, University of Campinas, Campinas 13083-875, SP, Brazil; (L.S.A.); (R.V.C.)
| | - Renato V. Casarin
- Department of Prosthesis and Periodontology, School of Dentistry, University of Campinas, Campinas 13083-875, SP, Brazil; (L.S.A.); (R.V.C.)
| | - Luciana Saraiva
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Maria Regina Lorenzetti Simionato
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
26
|
Ge J, Li M, Yao J, Guo J, Li X, Li G, Han X, Li Z, Liu M, Zhao J. The potential of EGCG in modulating the oral-gut axis microbiota for treating inflammatory bowel disease. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155643. [PMID: 38820660 DOI: 10.1016/j.phymed.2024.155643] [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: 02/24/2024] [Revised: 04/07/2024] [Accepted: 04/13/2024] [Indexed: 06/02/2024]
Abstract
Inflammatory bowel disease (IBD) is a recurrent chronic intestinal disorder that includes ulcerative colitis (UC) and Crohn's disease (CD). Its pathogenesis involves intricate interactions between pathogenic microorganisms, native intestinal microorganisms, and the intestinal immune system via the oral-gut axis. The strong correlation observed between oral diseases and IBD indicates the potential involvement of oral pathogenic microorganisms in IBD development. Consequently, therapeutic strategies targeting the proliferation, translocation, intestinal colonization and exacerbated intestinal inflammation of oral microorganisms within the oral-gut axis may partially alleviate IBD. Tea consumption has been identified as a contributing factor in reducing IBD, with epigallocatechin gallate (EGCG) being the primary bioactive compound used for IBD treatment. However, the precise mechanism by which EGCG mediates microbial crosstalk within the oral-gut axis remains unclear. In this review, we provide a comprehensive overview of the diverse oral microorganisms implicated in the pathogenesis of IBD and elucidate their colonization pathways and mechanisms. Subsequently, we investigated the antibacterial properties of EGCG and its potential to attenuate microbial translocation and colonization in the gut, emphasizing its role in attenuating exacerbations of IBD. We also elucidated the toxic and side effects of EGCG. Finally, we discuss current strategies for enhancing EGCG bioavailability and propose novel multi-targeted nano-delivery systems for the more efficacious management of IBD. This review elucidates the role and feasibility of EGCG-mediated modulation of the oral-gut axis microbiota in the management of IBD, contributing to a better understanding of the mechanism of action of EGCG in the treatment of IBD and the development of prospective treatment strategies.
Collapse
Affiliation(s)
- Jiaming Ge
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengyuan Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingwen Yao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinling Guo
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiankuan Li
- Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Gang Li
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Xiangli Han
- Department of Geriatric, Fourth Teaching Hospital of Tianjin University of TCM, Tianjin 300450, China
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ming Liu
- Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, 236 Baidi Road, Nankai District, Tianjin 300192, China.
| | - Jing Zhao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent TCM Diagnosis and Treatment Technology and Equipment, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
27
|
Chew RJJ, Tan KS, Chen T, Al-Hebshi NN, Goh CE. Quantifying periodontitis-associated oral dysbiosis in tongue and saliva microbiomes-An integrated data analysis. J Periodontol 2024. [PMID: 39007741 DOI: 10.1002/jper.24-0120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Periodontitis is primarily driven by subgingival biofilm dysbiosis. However, the quantification and impact of this periodontal dysbiosis on other oral microbial niches remain unclear. This study seeks to quantify the dysbiotic changes in tongue and salivary microbiomes resulting from periodontitis by applying a clinically relevant dysbiosis index to an integrated data analysis. METHODS The National Center for Biotechnology Information (NCBI) database was searched to identify BioProjects with published studies on salivary and tongue microbiomes of healthy and periodontitis subjects. Raw sequence datasets were processed using a standardized bioinformatic pipeline and categorized by their ecological niche and periodontal status. The subgingival microbial dysbiosis index (SMDI), a dysbiosis index originally developed using the subgingival microbiome, was computed at species and genus levels and customized for each niche. Its diagnostic accuracy for periodontitis was evaluated using receiver operating characteristic curves. RESULTS Four studies, contributing 328 microbiome samples, were included. At both species and genus levels, periodontitis samples had a higher SMDI, but the differences were only significant for subgingival biofilm and saliva (p < 0.001). However, SMDI showed good diagnostic accuracy for periodontitis status for all three niches (area under curve ranging from 0.76 to 0.90, p < 0.05). The dysbiosis index of subgingival biofilm was positively correlated with saliva consistently (p < 0.001) and with the tongue at the genus level (p = 0.036). CONCLUSIONS While the impact on the tongue microbiome requires further investigation, periodontitis-associated dysbiosis affects the salivary microbiome and is quantifiable using the dysbiosis index. The diagnostic potential of salivary microbial dysbiosis as a convenient periodontal biomarker for assessing periodontal status has potential public health and clinical applications. PLAIN LANGUAGE SUMMARY Periodontitis, a severe inflammation of the gums which causes bone loss, is a disease caused by an imbalance of good and bad bacteria under the gums. However, it is unclear how this bacterial imbalance in the gums affects the bacterial balance of other distinct parts of the mouth, such as the saliva and tongue. This study uses bacteria datasets of four previously published studies, contributing a total of 328 bacterial samples. The data were processed using a uniform data analysis workflow, and a bacterial score, the subgingival microbial dysbiosis index (SMDI), previously shown to capture periodontitis-associated bacteria imbalance, was calculated separately for samples from under the gums, the saliva, and the tongue. The SMDI was able to distinguish between health and periodontitis within each oral location, and in general, the scores were higher for periodontitis samples, though this difference was significant only for bacteria under the gums and in saliva. Saliva scores were also consistently correlated with bacteria under the gums. This study shows that periodontitis-associated bacterial imbalances are observed in oral locations beyond just under the gums, particularly the saliva. Thus, saliva bacteria may be used as a convenient biomarker for assessing gum disease, allowing for potential public health and clinical applications.
Collapse
Affiliation(s)
- Ren Jie Jacob Chew
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Kai Soo Tan
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| | - Tsute Chen
- The Forsyth Institute, Cambridge, Massachusetts, USA
| | - Nezar Noor Al-Hebshi
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania, USA
| | - Charlene Enhui Goh
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
| |
Collapse
|
28
|
Zhao Y, Liu Y, Jia L. Gut microbial dysbiosis and inflammation: Impact on periodontal health. J Periodontal Res 2024. [PMID: 38991951 DOI: 10.1111/jre.13324] [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: 02/07/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024]
Abstract
Periodontitis is widely acknowledged as the most prevalent type of oral inflammation, arising from the dynamic interplay between oral pathogens and the host's immune responses. It is also recognized as a contributing factor to various systemic diseases. Dysbiosis of the oral microbiota can significantly alter the composition and diversity of the gut microbiota. Researchers have delved into the links between periodontitis and systemic diseases through the "oral-gut" axis. However, whether the associations between periodontitis and the gut microbiota are simply correlative or driven by causative mechanistic interactions remains uncertain. This review investigates how dysbiosis of the gut microbiota impacts periodontitis, drawing on existing preclinical and clinical data. This study highlights potential mechanisms of this interaction, including alterations in subgingival microbiota, oral mucosal barrier function, neutrophil activity, and abnormal T-cell recycling, and offers new perspectives for managing periodontitis, especially in cases linked to systemic diseases.
Collapse
Affiliation(s)
- Yifan Zhao
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| | - Lu Jia
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
Kamer AR, Pushalkar S, Hamidi B, Janal MN, Tang V, Annam KRC, Palomo L, Gulivindala D, Glodzik L, Saxena D. Periodontal Inflammation and Dysbiosis Relate to Microbial Changes in the Gut. Microorganisms 2024; 12:1225. [PMID: 38930608 PMCID: PMC11205299 DOI: 10.3390/microorganisms12061225] [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: 05/06/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Periodontal disease (PerioD) is a chronic inflammatory disease of dysbiotic etiology. Animal models and few human data showed a relationship between oral bacteria and gut dysbiosis. However, the effect of periodontal inflammation and subgingival dysbiosis on the gut is unknown. We hypothesized that periodontal inflammation and its associated subgingival dysbiosis contribute to gut dysbiosis even in subjects free of known gut disorders. We evaluated and compared elderly subjects with Low and High periodontal inflammation (assessed by Periodontal Inflamed Surface Area (PISA)) for stool and subgingival derived bacteria (assayed by 16S rRNA sequencing). The associations between PISA/subgingival dysbiosis and gut dysbiosis and bacteria known to produce short-chain fatty acid (SCFA) were assessed. LEfSe analysis showed that, in Low PISA, species belonging to Lactobacillus, Roseburia, and Ruminococcus taxa and Lactobacillus zeae were enriched, while species belonging to Coprococcus, Clostridiales, and Atopobium were enriched in High PISA. Regression analyses showed that PISA associated with indicators of dysbiosis in the gut mainly reduced abundance of SCFA producing bacteria (Radj = -0.38, p = 0.03). Subgingival bacterial dysbiosis also associated with reduced levels of gut SCFA producing bacteria (Radj = -0.58, p = 0.002). These results suggest that periodontal inflammation and subgingival microbiota contribute to gut bacterial changes.
Collapse
Affiliation(s)
- Angela R. Kamer
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Smruti Pushalkar
- Center for Genomics and Systems Biology, New York University, 12 Waverly Place, New York, NY 10003, USA;
| | - Babak Hamidi
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Malvin N. Janal
- Department of Epidemiology and Health Promotion, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA;
| | - Vera Tang
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Kumar Raghava Chowdary Annam
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Leena Palomo
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Deepthi Gulivindala
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA; (B.H.); (V.T.); (K.R.C.A.); (L.P.); (D.G.)
| | - Lidia Glodzik
- Department of Radiology, Weill Cornell Medicine, Brain Health Imaging Institute Cornell University, New York, NY 10021, USA
| | - Deepak Saxena
- Department of Basic Sciences and Craniofacial Biology, College of Dentistry, New York University, 345 East 24th Street, New York, NY 10010, USA;
| |
Collapse
|
30
|
Sun B, Wang Y, Wu M, Feng G, Guo T. Key periodontal pathogens may mediate potential pathogenic relationships between periodontitis and crohn's disease. BMC Oral Health 2024; 24:668. [PMID: 38849764 PMCID: PMC11161938 DOI: 10.1186/s12903-024-04425-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/30/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Crohn's disease (CD)-associated periodontitis is common. However, the role of periodontal pathogens in the Coexistence of CD and periodontal disease remains unclear. METHODS To investigate the potential relationship mediated by periodontal pathogens between periodontitis and CD, we collected salivary samples from healthy participants (H group, n = 12), patients with CD (Ch group, n = 10), patients with periodontitis (Ps group, n = 12), and patients with Coexistence of CD and periodontal disease (Cp group, n = 12) and analyzed them by 16 S rRNA sequencing. RESULTS Patients with Coexistence of CD and periodontal disease had increased levels of Fusobacterium, Actinomyces, Leptotrichia, and Prevotella, which correlated with the severity of periodontitis. Conversely, the levels of Streptococcus, Neisseria, Haemophilus, and Gemella, which decreased in Coexistence of CD and periodontal disease, were negatively correlated with the severity of periodontitis. To further investigate the role of periodontal pathogens in CD development, representative periodontal pathogens causing periodontitis, Porphyromonas gingivalis and Fusobacterium nucleatum, were administered to mice. These pathogens migrate to, and colonize, the gut, accelerating CD progression and aggravating colitis, and even systemic inflammation. In vitro experiments using a Caco-2/periodontal pathogen coculture revealed that P. gingivalis and F. nucleatum increased intestinal permeability by directly disrupting the tight junctions of intestinal epithelial cells. CONCLUSION Our findings strongly suggest that periodontal pathogens play a role in the relationship between periodontitis and CD. These results provide a basis for understanding the pathogenesis of Coexistence of CD and periodontal disease and may lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Boyang Sun
- Department of General Dentistry, Research institute of Stomatology, Nanjing stomatological Hospital, Affiliated hospital of medical school, Nanjing University, Nanjing, 210008, China
| | - Ying Wang
- Department of General Dentistry, Research institute of Stomatology, Nanjing stomatological Hospital, Affiliated hospital of medical school, Nanjing University, Nanjing, 210008, China
| | - Mengmeng Wu
- Department of Pharmacy, Research institute of Stomatology, Nanjing stomatological Hospital, Affiliated hospital of medical school, Nanjing University, Nanjing, 210008, China
| | - Geng Feng
- Nanjing Fengzi Bio-pharm Technology Co. Ltd, Nanjing, 210018, China
| | - Ting Guo
- Department of General Dentistry, Research institute of Stomatology, Nanjing stomatological Hospital, Affiliated hospital of medical school, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
31
|
Sampath C, Chukkapalli SS, Raju AV, Alluri LSC, Srisai D, Gangula PR. Cinnamaldehyde Protects against P. gingivalis Induced Intestinal Epithelial Barrier Dysfunction in IEC-6 Cells via the PI3K/Akt-Mediated NO/Nrf2 Signaling Pathway. Int J Mol Sci 2024; 25:4734. [PMID: 38731952 PMCID: PMC11083591 DOI: 10.3390/ijms25094734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Porphyromonas gingivalis (Pg), a Gram-negative oral pathogen, promotes and accelerates periodontitis-associated gut disorders. Intestinal epithelial barrier dysfunction is crucial in the pathogenesis of intestinal and systemic diseases. In this study, we sought to elucidate the protective role of cinnamaldehyde (CNM, an activator of Nrf2) against P. gingivalis (W83) and Pg-derived lipopolysaccharide (Pg-LPS) induced intestinal epithelial barrier dysfunction via antioxidative mechanisms in IEC-6 cells. IEC-6 (ATCC, CRL-1592) cells were pretreated with or without CNM (100 µM), in the presence or absence of P. gingivalis (strain W83, 109 MOI) or Pg-LPS (1, 10, and 100 µg/mL), respectively, between 0-72 h time points by adopting a co-culture method. Intestinal barrier function, cytokine secretion, and intestinal oxidative stress protein markers were analyzed. P. gingivalis or Pg-LPS significantly (p < 0.05) increased reactive oxygen species (ROS) and malondialdehyde (MDA) levels expressing oxidative stress damage. Pg-LPS, as well as Pg alone, induces inflammatory cytokines via TLR-4 signaling. Furthermore, infection reduced Nrf2 and NAD(P)H quinone dehydrogenase 1 (NQO1). Interestingly, inducible nitric oxide synthase (iNOS) protein expression significantly (p < 0.05) increased with Pg-LPS or Pg infection, with elevated levels of nitric oxide (NO). CNM treatment suppressed both Pg- and Pg-LPS-induced intestinal oxidative stress damage by reducing ROS, MDA, and NO production. Furthermore, CNM treatment significantly upregulated the expression of tight junction proteins via increasing the phosphorylation levels of PI3K/Akt/Nrf2 suppressing inflammatory cytokines. CNM protected against Pg infection-induced intestinal epithelial barrier dysfunction by activating the PI3K/Akt-mediated Nrf2 signaling pathway in IEC-6 cells.
Collapse
Affiliation(s)
- Chethan Sampath
- Department of Diabetes, Metabolism and Endocrinology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| | - Sasanka S. Chukkapalli
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA;
| | - Abhinav V. Raju
- College of Osteopathic Medicine, Kansas City University, Kansas City, MO 64106, USA;
| | | | - Dollada Srisai
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| | - Pandu R. Gangula
- Department of ODS & Research, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
32
|
Zhao D, Li MH, Pan T, Guo J, Li J, Shi C, Wang N, Huang H, Wang C, Yang G. Preventive and Therapeutic Potential of Streptococcus cristatus CA119 in Experimental Periodontitis in Rats. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10254-y. [PMID: 38607584 DOI: 10.1007/s12602-024-10254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Periodontitis is an inflammatory condition of the oral cavity caused by a mixed infection of various bacteria, which not only severely affects the alveolar bone and connective tissues but also displays potential correlations with distal intestinal inflammation. In this study, we aimed to elucidate the therapeutic effects of Streptococcus cristatus CA119 on experimental periodontitis in rats and its impact on intestinal morphology. The results demonstrate that CA119 is capable of colonizing the oral cavity and exerting antagonistic effects on Porphyromonas gingivalis and Fusobacterium nucleatum, thus leading to a significant reduction in the oral pathogen load. Following CA119 intervention, there was a significant alleviation of weight loss in rats induced by periodontitis (P < 0.001). CA119 also regulated the expression of IL-6 (P < 0.05), IL-1β (P < 0.001), IL-18 (P < 0.001), COX-2 (P < 0.001), iNOS (P < 0.001), and MCP-1 (P < 0.01) in the gingival tissue. Additionally, CA119 reduced oxidative stress levels in rats and enhanced their antioxidant capacity. Microcomputed tomography (micro-CT) and histological analysis revealed that CA119 significantly reduced alveolar bone loss and reversed the downregulation of OPG/RANKL (P < 0.001). Furthermore, CA119 exhibited a significant protective effect against intestinal inflammation induced by periodontal disease and improved the colonic morphology in rats. In conclusion, this study demonstrates the role of CA119 as a potential oral probiotic in the prevention and treatment of experimental periodontitis, underscoring the potential of probiotics as a complementary approach to traditional periodontal care.
Collapse
Affiliation(s)
- Dongyu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Ming-Han Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Tianxu Pan
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Jialin Guo
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Junyi Li
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunwei Shi
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Haibin Huang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Chunfeng Wang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China.
| | - Guilian Yang
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, China.
- Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Jilin Agricultural University, Changchun, China.
- Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
33
|
Liang C, Ma L, Chen Y, Li J, Wang B, Ma C, Yuan Z, Nong X. Artesunate Alleviates Kidney Fibrosis in Type 1 Diabetes with Periodontitis Rats via Promoting Autophagy and Suppression of Inflammation. ACS OMEGA 2024; 9:16358-16373. [PMID: 38617690 PMCID: PMC11007779 DOI: 10.1021/acsomega.4c00020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 04/16/2024]
Abstract
To explore the effect of periodontal disease on the progression of diabetic kidney disease (DKD), to observe the effects of artesunate (ART) intervention on periodontal and kidney tissues in type 1 diabetic rats with periodontitis, and to explore the possibility of ART for the treatment of DKD. Rat models of diabetes mellitus, periodontitis, and diabetes mellitus with periodontitis were established through streptozotocin (STZ) intraperitoneal injection, maxillary first molar ligation, and P. gingivalis ligation applied sequentially. Ten weeks after modeling, ART gavage treatment was given for 4 weeks. Immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR), and Western blot were used to investigate the inflammatory factors, fibrogenisis, autophagy-related factors, and proteins in periodontal and kidney tissues, and 16S rDNA sequencing was used to detect the changes in dental plaque fluid and kidney tissue flora. Compared to the control group, the protein expression levels of transforming growth factor β1 (TGF-β1) and COL-IV in the periodontal disease (PD) group were increased. The protein expression of TGF-β1, Smad3, and COL-IV increased in the DM group and the DM + PD group, and the expression of TGF-β1, Smad3, and COL-IV was upregulated in the DM + PD group. These results suggest that periodontal disease enhances renal fibrosis and that this process is related to the TGF-β1/Smad/COL-IV signaling pathway. Among the top five dominant bacteria in the kidney of the DM + PD group, the abundance of Proteobacteria increased most significantly, followed by Actinobacteria and Firmicutes with mild increases. The relative abundance of Proteobacteria, Actinobacteria, and Firmicutes in the kidney tissues of DM and PD groups also showed an increasing trend compared with the CON group. Proteobacteria and Firmicutes in the kidney of the PD group and DM + PD group showed an increasing trend, which may mediate the increase of oxidative stress in the kidney and promote the occurrence and development of DN. Periodontal disease may lead to an imbalance of renal flora, aggravate renal damage in T1DM, cause glomerular inflammation and renal tubulointerstitial fibrosis, and reduce the level of autophagy. ART delays the process of renal fibrosis by inhibiting the TGF-β-Smad signaling pathway.
Collapse
Affiliation(s)
- Chen Liang
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Licheng Ma
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Yi Chen
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Jiaquan Li
- Medical
Science Research Center, Guangxi Medical
University, Nanning 530021, Guangxi, China
| | - Binge Wang
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Chubin Ma
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Zhong Yuan
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
| | - Xiaolin Nong
- College
of Stomatology, Hospital of Stomatology, Guangxi Medical University, No. 10 Shuangyong Road, Nanning 530021, Guangxi, China
- Guangxi
Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning 530021, Guangxi, China
| |
Collapse
|
34
|
Chen H, Peng L, Wang Z, He Y, Zhang X. Exploring the causal relationship between periodontitis and gut microbiome: Unveiling the oral-gut and gut-oral axes through bidirectional Mendelian randomization. J Clin Periodontol 2024; 51:417-430. [PMID: 38016486 DOI: 10.1111/jcpe.13906] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 11/05/2023] [Accepted: 11/07/2023] [Indexed: 11/30/2023]
Abstract
AIM This Mendelian randomization (MR) study was performed to explore the potential bidirectional causal relationship between the gut microbiome (GM) and periodontitis. MATERIALS AND METHODS We used genetic instruments from the genome-wide association study of European descent for periodontitis from the GeneLifestyle Interactions in Dental Endpoints (GLIDE) consortium (17,353 cases and 28,210 controls) and the FinnGen consortium (4434 cases and 259,234 controls) to investigate the causal relationship with GM (the MiBioGen consortium, 18,340 samples), and vice versa. Several MR techniques, which include inverse variance weighting (IVW), MR-Egger, weighted median, simple mode and weighted mode approaches, were employed to investigate the causal relationship between the exposures and the outcomes. Cochran's Q-test was performed to detect heterogeneity. The MR-Egger regression intercept and MR pleiotropy residual sum and outlier test (MR-PRESSO) were conducted to test potential horizontal pleiotropy. Leave-one-out sensitivity analyses were used to assess the stabilities of single nucleotide polymorphisms (SNPs). Finally, the IVW results from the two databases were analysed using meta-analysis. RESULTS We confirmed three potential causal relationships between GM taxa and periodontitis at the genus level. Among them, the genera Alistipes and Holdemanella were genetically associated with an increased risk of periodontitis. In reverse, periodontitis may lead to a decreased abundance of the genus Ruminococcaceae UCG014. CONCLUSIONS The demonstration of a causal link between GM and periodontitis provides compelling evidence, highlighting the interconnectivity and interdependence of the gut-oral and oral-gut axes.
Collapse
Affiliation(s)
- Hang Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
| | - Limin Peng
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
| | - Zhenxiang Wang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
| | - Yujuan He
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xiaonan Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
35
|
Baima G, Ferrocino I, Del Lupo V, Colonna E, Thumbigere-Math V, Caviglia GP, Franciosa I, Mariani GM, Romandini M, Ribaldone DG, Romano F, Aimetti M. Effect of Periodontitis and Periodontal Therapy on Oral and Gut Microbiota. J Dent Res 2024; 103:359-368. [PMID: 38362600 DOI: 10.1177/00220345231222800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
Mounting evidence indicates that periodontitis-related oral bacteria may contribute to gut microbial dysbiosis. This clinical study aimed to explore the oral-gut microbial signatures associated with periodontitis and to longitudinally evaluate the effect of periodontal treatment on the oral and gut microbial composition. Stool and saliva samples from generalized stage III/IV periodontitis patients (n = 47) were collected and analyzed by 16S ribosomal RNA gene amplicon sequencing, before and 3 mo after steps I to II of periodontal therapy. Periodontally healthy matched subjects (n = 47) were used as controls. Principal component analysis was carried out to identify oral-gut microbial profiles between periodontitis patients at baseline and healthy subjects; periodontitis samples were longitudinally compared before and after treatment. β-Diversity of gut microbial profiles of periodontitis patients before treatment significantly differed from healthy controls (P < 0.001). Periodontal therapy was associated with a significant change in gut microbiota (P < 0.001), with post-treatment microbial profiles similar to healthy volunteers. A higher abundance of Bacteroides, Faecalibacterium, Fusobacterium, and Lachnospiraceae was noted in fecal samples of periodontitis patients at baseline compared to healthy controls. In contrast, Lactobacillus was the only genus more abundant in the latter. Additionally, periodontal therapy led to a parallel reduction in the salivary carriage of periodontal pathobionts, as well as gut Bacteroides, Lachnoclostridium, Lachnospiraceae, Oscillospiraceae, and Ruminococcaceae, to levels similar to healthy controls. Collectively, discriminating oral-gut microbial signatures of periodontitis were found. Periodontal treatment both mitigated oral dysbiosis and altered gut microbial composition, signifying potential broader implications for gastrointestinal health and disease.
Collapse
Affiliation(s)
- G Baima
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - I Ferrocino
- Department of Agricultural, Forestry and Food Science, University of Turin, Turin, Italy
| | - V Del Lupo
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - E Colonna
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - V Thumbigere-Math
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - G P Caviglia
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - I Franciosa
- Department of Agricultural, Forestry and Food Science, University of Turin, Turin, Italy
| | - G M Mariani
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - M Romandini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - D G Ribaldone
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - F Romano
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - M Aimetti
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| |
Collapse
|
36
|
Yay E, Yilmaz M, Toygar H, Balci N, Alvarez Rivas C, Bolluk Kılıç B, Zirh A, Paster BJ, Kantarci A. Oral and gut microbial profiling in periodontitis and Parkinson's disease. J Oral Microbiol 2024; 16:2331264. [PMID: 38528960 PMCID: PMC10962298 DOI: 10.1080/20002297.2024.2331264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/11/2024] [Indexed: 03/27/2024] Open
Abstract
Objectives We tested the hypothesis that Parkinson's disease (PA) alters the periodontitis-associated oral microbiome. Method Patients with periodontitis with Parkinson's disease (PA+P) and without PA (P) and systemically and periodontally healthy individuals (HC) were enrolled. Clinical, periodontal and neurological parameters were recorded. The severity of PA motor functions was measured. Unstimulated saliva samples and stool samples were collected. Next-generation sequencing of 16S ribosomal RNA (V1-V3 regions) was performed. Results PA patients had mild-to-moderate motor dysfunction and comparable plaque scores as those without, indicating that oral hygiene was efficient in the PA+P group. In saliva, there were statistically significant differences in beta diversity between HC and PA+P (p = 0.001), HC and P (p = 0.001), and P and PA+P (p = 0.028). The microbial profiles of saliva and fecal samples were distinct. Mycoplasma faucium, Tannerella forsythia, Parvimonas micra, and Saccharibacteria (TM7) were increased in P; Prevotella pallens, Prevotella melaninogenica, Neisseria multispecies were more abundant in PA+P group, Ruthenibacterium lactatiformans, Dialister succinatiphilus, Butyrivibrio crossotus and Alloprevotella tannerae were detected in fecal samples in P groups compared to healthy controls. Conclusions No significant differences were detected between Parkinson's and non-Parkinson's gut microbiomes, suggesting that Parkinson's disease modifies the oral microbiome in periodontitis subjects independent of the gut microbiome.
Collapse
Affiliation(s)
- Ekin Yay
- Department of Applied Oral Sciences, The ADA Forsyth Institute, Cambridge, MA, USA
- Periodontist, Private Practice, Istanbul, Turkey
| | - Melis Yilmaz
- Department of Applied Oral Sciences, The ADA Forsyth Institute, Cambridge, MA, USA
- Department of Periodontology, Istanbul Medipol University, Istanbul, Turkey
| | - Hilal Toygar
- Department of Periodontology, Istanbul Medipol University, Istanbul, Turkey
| | - Nur Balci
- Department of Periodontology, Istanbul Medipol University, Istanbul, Turkey
| | - Carla Alvarez Rivas
- Department of Applied Oral Sciences, The ADA Forsyth Institute, Cambridge, MA, USA
- Department of Oral Microbiology and Infection, Harvard School of Dental Medicine, Boston, MA, USA
| | | | - Ali Zirh
- Department of Neurology, Istanbul Medipol University, Istanbul, Turkey
| | - Bruce J. Paster
- Department of Applied Oral Sciences, The ADA Forsyth Institute, Cambridge, MA, USA
- Department of Oral Microbiology and Infection, Harvard School of Dental Medicine, Boston, MA, USA
| | - Alpdogan Kantarci
- Department of Applied Oral Sciences, The ADA Forsyth Institute, Cambridge, MA, USA
- Department of Oral Microbiology and Infection, Harvard School of Dental Medicine, Boston, MA, USA
| |
Collapse
|
37
|
Fang L, Zhai Q, Zhang H, Ji P, Chen C, Zhang H. Comparisons of different extraction methods and solvents for saliva samples. Metabolomics 2024; 20:38. [PMID: 38460055 DOI: 10.1007/s11306-024-02105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 02/19/2024] [Indexed: 03/11/2024]
Abstract
INTRODUCTION Changes in the categories and concentrations of salivary metabolites may be closely related to oral, intestinal or systemic diseases. To study salivary metabolites, the first analytical step is to extract them from saliva samples as much as possible, while reducing interferences to a minimum. Frequently used extraction methods are protein precipitation (PPT), liquid-liquid extraction (LLE) and solid-phase extraction (SPE), with various organic solvents. The types and quantities of metabolites extracted with different methods may vary greatly, but few studies have systematically evaluated them. OBJECTIVES This study aimed to select the most suitable methods and solvents for the extraction of saliva according to different analytical targets. METHODS An untargeted metabolomics approach based on liquid chromatography-mass spectrometry was applied to obtain the raw data. The numbers of metabolites, repeatability of the data and intensities of mass spectrometry signals were used as evaluation criteria. RESULTS PPT resulted in the highest coverage. Among the PPT solvents, acetonitrile displayed the best repeatability and the highest coverage, while acetone resulted in the best signal intensities for the extracted compounds. LLE with the mixture of chloroform and methanol was the most suitable for the extraction of small hydrophobic compounds. CONCLUSION PPT with acetonitrile or acetone was recommended for untargeted analysis, while LLE with the mixture of chloroform and methanol was recommended for small hydrophobic compounds.
Collapse
Affiliation(s)
- Lingli Fang
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Qiming Zhai
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Zhang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Chang Chen
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| | - Hongmei Zhang
- Department of Pediatric Dentistry, Stomatological Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
38
|
Shen S, Sun T, Ding X, Gu X, Wang Y, Ma X, Li Z, Gao H, Ge S, Feng Q. The exoprotein Gbp of Fusobacterium nucleatum promotes THP-1 cell lipid deposition by binding to CypA and activating PI3K-AKT/MAPK/NF-κB pathways. J Adv Res 2024; 57:93-105. [PMID: 37100345 PMCID: PMC10918358 DOI: 10.1016/j.jare.2023.04.007] [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: 01/09/2023] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/28/2023] Open
Abstract
INTRODUCTION Growing evidence has shown the correlation between periodontitis and atherosclerosis, while our knowledge on the pathogenesis of periodontitis-promoting atherosclerosis is far from sufficient. OBJECTIVES Illuminate the pathogenic effects of Fusobacterium nucleatum (F. nucleatum) on intracellular lipid deposition in THP-1-derived macrophages and elucidate the underlying pathogenic mechanism of how F. nucleatum promoting atherosclerosis. METHODS AND RESULTS F. nucleatum was frequently detected in different kinds of atherosclerotic plaques and its abundance was positively correlated with the proportion of macrophages. In vitro assays showed F. nucleatum could adhere to and invade THP-1 cells, and survive continuously in macrophages for 24 h. F. nucleatum stimulation alone could significantly promote cellular inflammation, lipid uptake and inhibit lipid outflow. The dynamic gene expression of THP-1 cells demonstrated that F. nucleatum could time-serially induce the over-expression of multiple inflammatory related genes and activate NF-κB, MAPK and PI3K-AKT signaling pathways. The exoprotein of F. nucleatum, D-galactose-binding protein (Gbp), acted as one of the main pathogenic proteins to interact with the Cyclophilin A (CypA) of THP-1 cells and induced the activation of the NF- κB, MAPK and PI3K-AKT signaling pathways. Furthermore, use of six candidate drugs targeting to the key proteins in NF- κB, MAPK and PI3K-AKT pathways could dramatically decrease F. nucleatum induced inflammation and lipid deposition in THP-1 cells. CONCLUSIONS This study suggests that the periodontal pathogen F. nucleatum can activate macrophage PI3K-AKT/MAPK/NF-κB signal pathways, promotes inflammation, enhances cholesterol uptake, reduces lipid excretion, and promotes lipid deposition, which may be one of its main strategies promoting the development of atherosclerosis.
Collapse
Affiliation(s)
- Song Shen
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Tianyong Sun
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Xiangjiu Ding
- Department of Vascular Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Xiufeng Gu
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Yushang Wang
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Xiaomei Ma
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Zixuan Li
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Haiting Gao
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China
| | - Shaohua Ge
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China.
| | - Qiang Feng
- Department of Human Microbiome & Periodontology & Implantology & Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan 250012, China; State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| |
Collapse
|
39
|
Buetas E, Jordán-López M, López-Roldán A, Mira A, Carda-Diéguez M. Impact of Periodontitis on the Leakage of Oral Bacteria to the Gut. J Dent Res 2024; 103:289-297. [PMID: 38193290 DOI: 10.1177/00220345231221709] [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: 01/10/2024] Open
Abstract
Colorectal cancer (CRC) and periodontitis have recently been related due to the higher incidence of CRC in periodontal patients and the involvement of periodontal pathogens in carcinogenesis, suggesting that leakage from the oral cavity to the gut occurs. However, the magnitude of this pass-through in healthy individuals is controversial, and the effect that periodontitis could play in it is understudied. To evaluate the rate of bacterial leakage from the oral cavity to the gut, we analyzed the microbial composition of saliva, subgingival plaque, and fecal samples in healthy individuals without gastrointestinal disorders, including 20 periodontitis patients and 20 oral healthy controls, using PacBio full-length 16S rRNA gene sequencing. As expected, we observed a higher abundance of periodontal pathogens in the subgingival plaque and saliva of periodontal patients. In contrast, no significant differences were found between the fecal samples of both groups, implying that gut samples from periodontal patients were not enriched in periodontal pathogens. Fusobacterium nucleatum, a biomarker of CRC, was not found in the fecal samples of any participant. Our study does show a small leakage of some oral bacteria (mainly streptococci) to the gut, regardless of periodontal health status. Future studies should test whether other host factors and/or the preexistence of a gut disorder must be present in addition to periodontitis to promote the colonization of the gut by oral pathogens. The absence of periodontal pathogens in feces supports the idea that these bacteria could be used as biomarkers of intestinal disorders, including CRC.
Collapse
Affiliation(s)
- E Buetas
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
| | - M Jordán-López
- Department of Periodontics, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - A López-Roldán
- Department of Periodontics, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - A Mira
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
- CIBER Center for Epidemiology and Public Health, Madrid, Spain
| | - M Carda-Diéguez
- Genomics & Health Department, FISABIO Foundation, Valencia, Spain
| |
Collapse
|
40
|
Hernández-Ruiz P, Escalona Montaño AR, Amezcua-Guerra LM, González-Pacheco H, Niccolai E, Amedei A, Aguirre-García MM. Potential Association of the Oral Microbiome with Trimethylamine N-Oxide Quantification in Mexican Patients with Myocardial Infarction. Mediators Inflamm 2024; 2024:3985731. [PMID: 38415052 PMCID: PMC10898950 DOI: 10.1155/2024/3985731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/29/2024] Open
Abstract
Many attempts have been proposed to evaluate the linkage between the oral-gut-liver axis and the mechanisms related to the diseases' establishment. One of them is the oral microbiota translocation into the bloodstream, liver, and gut, promoting a host dysbiosis and triggering the presence of some metabolites such as trimethylamine N-oxide (TMAO), known as a risk marker for cardiovascular disease, and especially the myocardial infarction (MI). In the present pilot study, the involvement of oral dysbiosis related to the presence of TMAO has been considered an independent component of the standard risk factors (SRs) in the development of MI, which has not been previously described in human cohorts. A positive and significant correlation of TMAO levels with Porphyromonas was identified; likewise, the increase of the genus Peptidiphaga in patients without SRs was observed. We determined that the presence of SRs does not influence the TMAO concentration in these patients. This report is the first study where the relationship between oral dysbiosis and TMAO is specified in the Mexican population. Our findings provide information on the possible contribution of the oral pathogens associated with gut dysbiosis in the development of MI, although further analysis should be performed.
Collapse
Affiliation(s)
- Paulina Hernández-Ruiz
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - Alma R Escalona Montaño
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - Luis M Amezcua-Guerra
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - Héctor González-Pacheco
- Unidad de Cuidados Coronarios, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence 50134, Italy
| | - María M Aguirre-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico
| |
Collapse
|
41
|
Hu H, Yao Y, Liu F, Luo L, Liu J, Wang X, Wang Q. Integrated microbiome and metabolomics revealed the protective effect of baicalin on alveolar bone inflammatory resorption in aging. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155233. [PMID: 38181526 DOI: 10.1016/j.phymed.2023.155233] [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: 08/28/2023] [Revised: 11/01/2023] [Accepted: 11/19/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND With the growing aging population and longer life expectancy, periodontitis and tooth loss have become major health concerns. The gut microbiota, as a key regulator in bone homeostasis, has gathered immense interest. Baicalin, a flavonoid compound extracted from Scutellaria baicalensis Georgi, has shown antioxidant and anti-inflammatory activities. PURPOSE This study investigated, for the first time, the protective mechanism of baicalin against alveolar bone inflammatory resorption in aging mice by regulating intestinal flora and metabolites, as well as intestinal barrier function. METHODS A ligature-induced periodontitis model was established in d-galactose (D-gal)-induced aging mice, and baicalin was administered at different dosages for 13 weeks. Body weight was measured weekly. The antioxidant and anti-inflammatory activity of baicalin were evaluated using serum superoxide dismutase (SOD), malonaldehyde (MDA), interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-α) levels. The immune capability was assessed by thymus and spleen indices. Histopathological changes were observed in the heart, liver, ileum, and periodontal tissues. Alveolar bone absorption of maxillary second molars was examined, and osteoclasts were counted by tartrate-resistant acid phosphatase (TRAP) staining. Furthermore, fecal samples were analyzed using 16S rRNA sequencing and non-targeted metabolomics to identify differences in intestinal bacterial composition and metabolites. RESULTS Baicalin exhibited anti-aging properties, as evidenced by increased SOD activity and decreased levels of MDA, IL-6, and TNF-α in serum compared to the control group. Baicalin also ameliorated alveolar bone loss in the d-gal-induced aging-periodontitis group (p < 0.05). Furthermore, baicalin restored ileal permeability by up-regulating the expression of ZO-1 and occludin in aging-periodontitis groups (p < 0.05). Alpha diversity analysis indicated that baicalin-treated mice harbored a higher diversity of gut microbe. PCoA and ANOSIM results revealed significant dissimilarity between groups. The Firmicutes/Bacteroidetes (F/B) ratio, which decreased in periodontitis mice, was restored by baicalin treatment. Additionally, medium-dosage baicalin promoted the production of beneficial flavonoids, and enriched short-chain fatty acids (SCFAs)-producing bacteria. CONCLUSION Intestinal homeostasis is a potential avenue for treating age-related alveolar bone loss. Baicalin exerts anti-inflammatory, antioxidant, and osteo-protective properties by regulating the gut microbiota and metabolites.
Collapse
Affiliation(s)
- Huan Hu
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China; School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Yanzi Yao
- School of Stomatology, Zunyi Medical University, Zunyi, China; Luoyang Maternal and Child Health Hospital & Henan Second Children's Hospital, Luoyang, China
| | - Fangzhou Liu
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Liangliang Luo
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi, China
| | - Jianguo Liu
- School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China.
| | - Qian Wang
- Microbial Resources and Drug Development Key Laboratory of Guizhou Tertiary Institution, Zunyi Medical University, Zunyi, China; School of Stomatology, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
42
|
Liu J, Jiang J, Lan Y, Li C, Han R, Wang J, Wang T, Zhao Z, Fan Z, He L, Fang J. Metagenomic analysis of oral and intestinal microbiome of patients during the initial stage of orthodontic treatment. Am J Orthod Dentofacial Orthop 2024; 165:161-172.e3. [PMID: 37966405 DOI: 10.1016/j.ajodo.2023.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/01/2023] [Accepted: 07/01/2023] [Indexed: 11/16/2023]
Abstract
INTRODUCTION This prospective study analyzed changes in the oral and intestinal microbiomes in patients before and after fixed orthodontic treatment, elucidating the impacts of fixed orthodontic treatment on patient health and metabolism. METHODS Metagenomic analysis was conducted on stool, dental plaque, and saliva samples from 10 fixed orthodontic patients. All the samples were sequenced with Illumina NovaSeq 6000 with a paired-end sequencing length of 150 bp. Identification of taxa in metagenomes and functional annotation of genes of the microbiota were performed using the data after quality control. Clinical periodontal parameters, including the gingiva index, plaque index, and pocket probing depth, were examined at each time point in triplicates. Patients also received a table to record their oral hygiene habits of brushing, flossing, and dessert consumption frequency over 1 month. RESULTS The brushing and flossing times per day of patients were significantly increased after treatment compared with baseline. The number of times a patient ate dessert daily was also fewer after treatment than at baseline. In addition, the plaque index decreased significantly, whereas the pH value of saliva, gingiva index, and pocket probing depth did not change. No significant differences were observed between the participants before and after orthodontic treatment regarding alpha-diversity analysis of the gut, dental plaque, or saliva microbiota. However, on closer analysis, periodontal disease-associated bacteria levels in the oral cavity remain elevated. Alterations in gut microbiota were also observed after orthodontic treatment. CONCLUSIONS The richness and diversity of the microbiome did not change significantly during the initial stage of fixed orthodontic treatment. However, the levels of periodontal disease-associated bacteria increased.
Collapse
Affiliation(s)
- Jialing Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiyang Jiang
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Yue Lan
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Chengyan Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ruiying Han
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiao Wang
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Tianyi Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zhenxin Fan
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Libang He
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| | - Jie Fang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
43
|
Ye HL, Zhi MF, Chen BY, Lin WZ, Li YL, Huang SJ, Zhou LJ, Xu S, Zhang J, Zhang WC, Feng Q, Duan SZ. Alterations of oral and gut viromes in hypertension and/or periodontitis. mSystems 2024; 9:e0116923. [PMID: 38108668 PMCID: PMC10804974 DOI: 10.1128/msystems.01169-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
The microbiota plays an important role in both hypertension (HTN) and periodontitis (PD), and PD exacerbates the development of HTN by oral and gut microbiota. Previous studies have focused on exploring the importance of the bacteriome in HTN and PD but overlooked the impact of the virome, which is also a member of the microbiota. We collected 180 samples of subgingival plaques, saliva, and feces from a cohort of healthy subjects (nHTNnPD), subjects with HTN (HTNnPD) or PD (PDnHTN), and subjects with both HTN and PD (HTNPD). We performed metagenomic sequencing to assess the roles of the oral and gut viromes in HTN and PD. The HTNnPD, PDnHTN, and HTNPD groups all showed significantly distinct beta diversity from the nHTNnPD group in saliva. We analyzed alterations in oral and gut viral composition in HTN and/or PD and identified significantly changed viruses in each group. Many viruses across three sites were significantly associated with blood pressure and other clinical parameters. Combined with these clinical associations, we found that Gillianvirus in subgingival plaques was negatively associated with HTN and that Torbevirus in saliva was positively associated with HTN. We found that Pepyhexavirus from subgingival plaques was indicated to be transferred to the gut. We finally evaluated viral-bacterial transkingdom interactions and found that viruses and bacteria may cooperate to affect HTN and PD. Correspondingly, HTN and PD may synergize to improve communications between viruses and bacteria.IMPORTANCEPeriodontitis (PD) and hypertension (HTN) are both highly prevalent worldwide and cause serious adverse outcomes. Increasing studies have shown that PD exacerbates HTN by oral and gut microbiota. Previous studies have focused on exploring the importance of the bacteriome in HTN and PD but overlooked the impact of the virome, even though viruses are common inhabitants in humans. Alterations in oral and gut viral diversity and composition contribute to diseases. The present study, for the first time, profiled the oral and gut viromes in HTN and/or PD. We identified key indicator viruses and their clinical implications in HTN and/or PD. We also investigated interactions between viruses and bacteria. This work improved the overall understanding of the viromes in HTN and PD, providing vital insights into the role of the virome in the development of HTN and PD.
Collapse
Affiliation(s)
- Hui-Lin Ye
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Meng-Fan Zhi
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Bo-Yan Chen
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wen-Zhen Lin
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Yu-Lin Li
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shi-Jia Huang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Lu-Jun Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Shuo Xu
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jun Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Wu-Chang Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Qiang Feng
- Department of Human Microbiome, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Sheng-Zhong Duan
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People’s Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, Shanghai Key Laboratory of Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
44
|
Xu M, Shao Q, Zhou Y, Yu Y, Wang S, Wang A, Cai Y. Potential effects of specific gut microbiota on periodontal disease: a two-sample bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1322947. [PMID: 38314435 PMCID: PMC10834673 DOI: 10.3389/fmicb.2024.1322947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Introduction Periodontal disease (PD) presents a substantial global health challenge, encompassing conditions from reversible gingivitis to irreversible periodontitis, often culminating in tooth loss. The gut-oral axis has recently emerged as a focal point, with potential gut microbiota dysbiosis exacerbating PD. Methods In this study, we employed a double-sample bidirectional Mendelian randomized (MR) approach to investigate the causal relationship between specific gut microbiota and periodontal disease (PD) and bleeding gum (BG) development, while exploring the interplay between periodontal health and the gut microenvironment. We performed genome-wide association studies (GWAS) with two cohorts, totalling 346,731 (PD and control) and 461,113 (BG and control) participants, along with data from 14,306 participants' intestinal flora GWAS, encompassing 148 traits (31 families and 117 genera). Three MR methods were used to assess causality, with the in-verse-variance-weighted (IVW) measure as the primary outcome. Cochrane's Q test, MR-Egger, and MR-PRESSO global tests were used to detect heterogeneity and pleiotropy. The leave-one-out method was used to test the stability of the MR results. An F-statistic greater than 10 was accepted for instrument exposure association. Results and conclusion Specifically, Eubacterium xylanophilum and Lachnoclostridium were associated with reduced gum bleeding risk, whereas Anaerotruncus, Eisenbergiella, and Phascolarctobacterium were linked to reduced PD risk. Conversely, Fusicatenibacter was associated with an elevated risk of PD. No significant heterogeneity or pleiotropy was detected. In conclusion, our MR analysis pinpointed specific gut flora with causal connections to PD, offering potential avenues for oral health interventions.
Collapse
Affiliation(s)
- Meng Xu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Shao
- IT Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Yinglu Zhou
- Nursing Department, Huashan Hospital, Fudan University, Shanghai, China
| | - Yili Yu
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuwei Wang
- Dental Diseases Prevention and Treatment Center of Jiading District, Shanghai, China
| | - An Wang
- Shanghai Jingan Dental Clinic, Shanghai, China
| | - Yida Cai
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Kuraji R, Ye C, Zhao C, Gao L, Martinez A, Miyashita Y, Radaic A, Kamarajan P, Le C, Zhan L, Range H, Sunohara M, Numabe Y, Kapila YL. Nisin lantibiotic prevents NAFLD liver steatosis and mitochondrial oxidative stress following periodontal disease by abrogating oral, gut and liver dysbiosis. NPJ Biofilms Microbiomes 2024; 10:3. [PMID: 38233485 PMCID: PMC10794237 DOI: 10.1038/s41522-024-00476-x] [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: 04/17/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Oral microbiome dysbiosis mediates chronic periodontal disease, gut microbial dysbiosis, and mucosal barrier disfunction that leads to steatohepatitis via the enterohepatic circulation. Improving this dysbiosis towards health may improve liver disease. Treatment with antibiotics and probiotics have been used to modulate the microbial, immunological, and clinical landscape of periodontal disease with some success. The aim of the present investigation was to evaluate the potential for nisin, an antimicrobial peptide produced by Lactococcus lactis, to counteract the periodontitis-associated gut dysbiosis and to modulate the glycolipid-metabolism and inflammation in the liver. Periodontal pathogens, namely Porphyromonas gingivalis, Treponema denticola, Tannerella forsythia and Fusobacterium nucleatum, were administrated topically onto the oral cavity to establish polymicrobial periodontal disease in mice. In the context of disease, nisin treatment significantly shifted the microbiome towards a new composition, commensurate with health while preventing the harmful inflammation in the small intestine concomitant with decreased villi structural integrity, and heightened hepatic exposure to bacteria and lipid and malondialdehyde accumulation in the liver. Validation with RNA Seq analyses, confirmed the significant infection-related alteration of several genes involved in mitochondrial dysregulation, oxidative phosphorylation, and metal/iron binding and their restitution following nisin treatment. In support of these in vivo findings indicating that periodontopathogens induce gastrointestinal and liver distant organ lesions, human autopsy specimens demonstrated a correlation between tooth loss and severity of liver disease. Nisin's ability to shift the gut and liver microbiome towards a new state commensurate with health while mitigating enteritis, represents a novel approach to treating NAFLD-steatohepatitis-associated periodontal disease.
Collapse
Affiliation(s)
- Ryutaro Kuraji
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Changchang Ye
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontology, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Chuanjiang Zhao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Li Gao
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - April Martinez
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Yukihiro Miyashita
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Allan Radaic
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Pachiyappan Kamarajan
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Charles Le
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Ling Zhan
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
| | - Helene Range
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA
- Department of Periodontology, University of Rennes, UFR of Odontology; Service d'Odontologie, CHU de Rennes, Rennes, France
- INSERM CHU Rennes, Institut NUMECAN (Nutrition Metabolisms and Cancer); CIC 1414, Rennes, France
| | - Masataka Sunohara
- Department of Anatomy, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Yukihiro Numabe
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Tokyo, Tokyo, Japan
| | - Yvonne L Kapila
- Orofacial Sciences Department, School of Dentistry, University of California, San Francisco, San Francisco, CA, USA.
- Sections of Biosystems and Function and Periodontics, School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
46
|
Qian J, Lu J, Cheng S, Zou X, Tao Q, Wang M, Wang N, Zheng L, Liao W, Li Y, Yan F. Periodontitis salivary microbiota exacerbates colitis-induced anxiety-like behavior via gut microbiota. NPJ Biofilms Microbiomes 2023; 9:93. [PMID: 38062089 PMCID: PMC10703887 DOI: 10.1038/s41522-023-00462-9] [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: 06/27/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
The gut-brain axis is a bidirectional communication system between the gut and central nervous system. Many host-related factors can affect gut microbiota, including oral bacteria, making the brain a vulnerable target via the gut-brain axis. Saliva contains a large number of oral bacteria, and periodontitis, a common oral disease, can change the composition of salivary microbiota. However, the role and mechanism of periodontitis salivary microbiota (PSM) on the gut-brain axis remain unclear. Herein, we investigated the nature and mechanisms of this relationship using the mice with dextran sulfate sodium salt (DSS)-induced anxiety-like behavior. Compared with healthy salivary microbiota, PSM worsened anxiety-like behavior; it significantly reduced the number of normal neurons and activated microglia in DSS mice. Antibiotic treatment eliminated the effect of PSM on anxiety-like behavior, and transplantation of fecal microbiota from PSM-gavaged mice exacerbated anxiety-like behavior. These observations indicated that the anxiety-exacerbating effect of PSM was dependent on the gut microbiota. Moreover, the PSM effect on anxiety-like behavior was not present in non-DSS mice, indicating that DSS treatment was a prerequisite for PSM to exacerbate anxiety. Mechanistically, PSM altered the histidine metabolism in both gut and brain metabolomics. Supplementation of histidine-related metabolites had a similar anxiety-exacerbating effect as that of PSM, suggesting that histidine metabolism may be a critical pathway in this process. Our results demonstrate that PSM can exacerbate colitis-induced anxiety-like behavior by directly affecting the host gut microbiota, emphasizing the importance of oral diseases in the gut-brain axis.
Collapse
Affiliation(s)
- Jun Qian
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiangyue Lu
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shuyu Cheng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xihong Zou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Min Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Nannan Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lichun Zheng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenzheng Liao
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yanfen Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Fuhua Yan
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
47
|
Song J, Wu Y, Yin X, Ma H, Zhang J. Mendelian Randomisation Study on Association of Gut Microbiota and Periodontitis. Int Dent J 2023; 73:847-853. [PMID: 37316410 PMCID: PMC10658424 DOI: 10.1016/j.identj.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 06/16/2023] Open
Abstract
OBJECTIVE Several studies have demonstrated the possible association between gut microbiota and periodontitis. The mechanism by which gut microbiota contribute to periodontitis remains unknown. METHODS A 2-sample Mendelian randomisation (MR) study was conducted using publicly available Genome-Wide Association Studies (GWAS) data of European ancestry. The relationships between gut microbiota and tooth loss and periodontitis were assessed using summary-level data. Moreover, inverse variance weighted (IVW), MR-Egger, weighted median, and simple Mendelian were used. The results were further validated using sensitivity analyses. RESULTS A total of 211 gut microbiota were studied, including 9 phyla, 16 classes, 20 orders, 35 families, and 131 genera. The IVW method identified 16 bacterial genera related to the risk of periodontitis and tooth loss. Lactobacillaceae was associated with an increased risk of periodontitis (odds ratio [OR], 1.40, 95% confidence interval [CI], 1.03-1.91, P<.001) and tooth loss (OR, 1.12; 95% CIs, 1.02-1.24, P = .002), whereas Lachnospiraceae UCG008 was linked to a lower risk of tooth loss (P = .041). There was no heterogeneity and horizontal pleiotropy in the sensitivity analysis. CONCLUSIONS Several microorganisms were identified to be linked to the risk of periodontitis. Furthermore, the findings improved our understanding of gut microbiota and periodontitis pathology.
Collapse
Affiliation(s)
- Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Yadong Wu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Xinhai Yin
- Department of Oral and Maxillofacial Surgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Hong Ma
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| | - Junmei Zhang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
48
|
Nibali L, Stephen AS, Allaker RP, Di Pino A, Terranova V, Pisano M, Di Marca S, Ferrara V, Scicali R, Purrello F, Donos N, Regolo M, Malatino L. Associations between Host Genetic Variants and Subgingival Microbiota in Patients with the Metabolic Syndrome. Int J Mol Sci 2023; 24:16649. [PMID: 38068972 PMCID: PMC10706808 DOI: 10.3390/ijms242316649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Host genetic variants may affect oral biofilms, playing a role in the periodontitis-systemic disease axis. This is the first study to assess the associations between host genetic variants and subgingival microbiota in patients with metabolic syndrome (MetS); 103 patients with MetS underwent medical and periodontal examinations and had blood and subgingival plaque samples taken. DNA was extracted and processed, assessing a panel of selected single nucleotide polymorphisms (SNPs) first (hypothesis testing) and then expanding to a discovery phase. The subgingival plaque microbiome from these patients was profiled. Analysis of associations between host genetic and microbial factors was performed and stratified for periodontal diagnosis. Specific SNPs within RUNX2, CAMTA1 and VDR genes were associated with diversity metrics with no genome-wide associations detected for periodontitis severity or Mets components at p < 10-7. Severe periodontitis was associated with pathogenic genera and species. Some SNPs correlated with specific bacterial genera as well as with microbial taxa, notably VDR (rs12717991) with Streptococcus mutans and RUNX2 (rs3749863) with Porphyromonas gingivalis. In conclusion, variation in host genotypes may play a role in the dysregulated immune responses characterizing periodontitis and thus the oral microbiome, suggesting that systemic health-associated host traits further interact with oral health and the microbiome.
Collapse
Affiliation(s)
- Luigi Nibali
- Periodontology Unit, Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London WC2R 2LS, UK;
| | - Abish S. Stephen
- Centre for Immunobiology & Regenerative Medicine and Centre for Oral Clinical Research, Institute of Dentistry, Faculty of Medicine and Dentistry, Queen Mary University of London (QMUL), London E1 4NS, UK; (A.S.S.); (R.P.A.); (N.D.)
| | - Robert P. Allaker
- Centre for Immunobiology & Regenerative Medicine and Centre for Oral Clinical Research, Institute of Dentistry, Faculty of Medicine and Dentistry, Queen Mary University of London (QMUL), London E1 4NS, UK; (A.S.S.); (R.P.A.); (N.D.)
| | - Antonino Di Pino
- Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95123 Catania, Italy (R.S.); (F.P.)
| | - Valentina Terranova
- Department of Clinical and Experimental Medicine, Cannizzaro Hospital, University of Catania, 95123 Catania, Italy (M.R.)
| | - Marcella Pisano
- Department of Clinical and Experimental Medicine, Cannizzaro Hospital, University of Catania, 95123 Catania, Italy (M.R.)
| | - Salvatore Di Marca
- Department of Clinical and Experimental Medicine, Cannizzaro Hospital, University of Catania, 95123 Catania, Italy (M.R.)
| | - Viviana Ferrara
- Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95123 Catania, Italy (R.S.); (F.P.)
| | - Roberto Scicali
- Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95123 Catania, Italy (R.S.); (F.P.)
| | - Francesco Purrello
- Department of Clinical and Experimental Medicine, Garibaldi-Nesima Hospital, University of Catania, 95123 Catania, Italy (R.S.); (F.P.)
| | - Nikolaos Donos
- Centre for Immunobiology & Regenerative Medicine and Centre for Oral Clinical Research, Institute of Dentistry, Faculty of Medicine and Dentistry, Queen Mary University of London (QMUL), London E1 4NS, UK; (A.S.S.); (R.P.A.); (N.D.)
| | - Matteo Regolo
- Department of Clinical and Experimental Medicine, Cannizzaro Hospital, University of Catania, 95123 Catania, Italy (M.R.)
- Academic Unit of Internal Medicine, Cannizzaro Hospital, Via Messina 829, 95126 Catania, Italy
| | - Lorenzo Malatino
- Department of Clinical and Experimental Medicine, Cannizzaro Hospital, University of Catania, 95123 Catania, Italy (M.R.)
- Academic Unit of Internal Medicine, Cannizzaro Hospital, Via Messina 829, 95126 Catania, Italy
| |
Collapse
|
49
|
Lee HJ, Lee Y, Hong SH, Park JW. Decoding the Link between Periodontitis and Neuroinflammation: The Journey of Bacterial Extracellular Vesicles. Curr Genomics 2023; 24:132-135. [PMID: 38178987 PMCID: PMC10761334 DOI: 10.2174/0113892029258657231010065320] [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: 04/19/2023] [Revised: 07/26/2023] [Accepted: 09/08/2023] [Indexed: 01/06/2024] Open
Affiliation(s)
- Heon-Jin Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea, South
| | - Youngkyun Lee
- Department of Biochemistry, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea, South
| | - Su-Hyung Hong
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea, South
| | - Jin-Woo Park
- Department of Periodontology, School of Dentistry, Kyungpook National University, Daegu, 41940, Korea, South
| |
Collapse
|
50
|
Hammad MI, Conrads G, Abdelbary MMH. Isolation, identification, and significance of salivary Veillonella spp., Prevotella spp., and Prevotella salivae in patients with inflammatory bowel disease. Front Cell Infect Microbiol 2023; 13:1278582. [PMID: 38053528 PMCID: PMC10694262 DOI: 10.3389/fcimb.2023.1278582] [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: 08/16/2023] [Accepted: 10/30/2023] [Indexed: 12/07/2023] Open
Abstract
The global prevalence of inflammatory bowel disease (IBD) is on the rise, prompting significant attention from researchers worldwide. IBD entails chronic inflammatory disorders of the intestinal tract, characterized by alternating flares and remissions. Through high-throughput sequencing, numerous studies have unveiled a potential microbial signature for IBD patients showing intestinal enrichment of oral-associated bacteria. Simultaneously, the oral microbiome can be perturbed by intestinal inflammation. Our prior investigation, based on 16S rRNA amplicon sequencing, underscored elevated abundance of Veillonella spp. and Prevotella spp. in the salivary microbiomes of IBD patients. Noteworthy, Prevotella salivae emerged as a distinct species significantly associated with IBD. P. salivae is an under-recognized pathogen that was found to play a role in both oral and systemic diseases. In this study, we delve deeper into the salivary microbiomes of both IBD patients and healthy controls. Employing diverse cultivation techniques and real-time quantitative polymerase chain reactions (RT-qPCR), we gauged the prevalence and abundance of Veillonella spp., Prevotella spp., and P. salivae. Our isolation efforts yielded 407 and 168 strains of Veillonella spp., as well as 173 and 90 strains of Prevotella spp., from the saliva samples of IBD patients and healthy controls, respectively. Veillonella-vancomycin agar emerged as the discerning choice for optimal Veillonella spp. cultivation, while Schaedler kanamycin-vancomycin agar proved to be the most suitable medium for cultivating Prevotella spp. strains. Comparing our RT-qPCR findings to the previous 16S rRNA amplicon sequencing data, the results corroborated the higher abundance of Veillonella spp., Prevotella spp., and P. salivae in the saliva of IBD patients compared to healthy controls. However, it's worth noting that in contrast to RT-qPCR, the 16S rRNA amplicon sequencing data revealed greater absolute abundance of all three bacterial groups in both IBD patients and controls.
Collapse
Affiliation(s)
- Moshira I. Hammad
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule University Hospital, Aachen, Germany
| | - Georg Conrads
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule University Hospital, Aachen, Germany
| | - Mohamed M. H. Abdelbary
- Division of Oral Microbiology and Immunology, Department of Operative Dentistry, Periodontology and Preventive Dentistry, Rheinisch-Westfälische Technische Hochschule University Hospital, Aachen, Germany
- Division of Nosocomial Pathogens and Antibiotic Resistances, Department of Infectious Diseases, Robert Koch Institute, Wernigerode, Germany
| |
Collapse
|