1
|
Saravanan C, S M N Mydin RB, Mohamed Sheriff NR, Kaur G, Singh Dhaliwal S, Musa MY. Salivaomics in head and neck cancer. Clin Chim Acta 2025; 565:119952. [PMID: 39216814 DOI: 10.1016/j.cca.2024.119952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Salivaomics is a promising method for the early detection and monitoring of head and neck cancer (HNC). By analyzing salivary proteomics, RNA, and DNA, it identifies biomarkers that distinguish HNC patients from healthy individuals. Saliva's non-invasive, easily collectible nature and affordability make it an advantageous screening tool. Multiomics approaches, which explore genetic mutations, gene expression patterns, protein profiles, and metabolite levels, provide a comprehensive molecular perspective that enhances clinical applicability. The approaches enhance the precision of diagnoses, enable the development and application of targeted therapies, and contribute to the overall advancement of personalized medicine. Despite its potential, larger-scale studies are essential for validating biomarkers, and assessing sensitivity, accuracy, and specificity in detecting HNC. This review highlights salivaomics' potential as a non-invasive, accessible biological sample for early disease detection in HNC and underscores the value of multiomics in advancing this research. Salivaomics offers significant insights into the underlying mechanisms of HNC, enabling the discovery of robust, non-invasive biomarkers for improved disease management.
Collapse
Affiliation(s)
- Chandrarohini Saravanan
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM), 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Rabiatul Basria S M N Mydin
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM), 13200 Kepala Batas, Pulau Pinang, Malaysia.
| | - Nur Rizikin Mohamed Sheriff
- School of Distance Education, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia; Division of Research & Innovation, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Satvinder Singh Dhaliwal
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia; Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia; Duke-NUS Medical School, National University of Singapore, Queenstown, Singapore; Singapore University of Social Sciences, 463 Clementi Road, Clementi 599494, Singapore
| | - Muhamad Yusri Musa
- Department of Clinical Medicine, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Malaysia; Pusat Perubatan, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Pulau Pinang 84001, Malaysia
| |
Collapse
|
2
|
Esberg A, Fries N, Haworth S, Johansson I. Saliva microbiome profiling by full-gene 16S rRNA Oxford Nanopore Technology versus Illumina MiSeq sequencing. NPJ Biofilms Microbiomes 2024; 10:149. [PMID: 39695121 DOI: 10.1038/s41522-024-00634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Affiliation(s)
- Anders Esberg
- Department of Odontology, Umeå University, Umeå, Sweden.
| | - Niklas Fries
- Department of Odontology, Umeå University, Umeå, Sweden
| | | | | |
Collapse
|
3
|
Zhong Y, Kang X, Bai X, Pu B, Smerin D, Zhao L, Xiong X. The Oral-Gut-Brain Axis: The Influence of Microbes as a Link of Periodontitis With Ischemic Stroke. CNS Neurosci Ther 2024; 30:e70152. [PMID: 39675010 DOI: 10.1111/cns.70152] [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: 08/14/2024] [Revised: 11/03/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024] Open
Abstract
Periodontitis, a non-communicable chronic inflammation disease resulting from dysbiosis of the oral microbiota, has been demonstrated to have a positive association with the risk of ischemic stroke (IS). The major periodontal pathogens contribute to the progression of stroke-related risk factors such as obesity, diabetes, atherosclerosis, and hypertension. Transcriptional changes in periodontitis pathogens have been detected in oral samples from stroke patients, suggesting a new conceptual framework involving microorganisms. The bidirectional regulation between the gut and the central nervous system (CNS) is mediated by interactions between intestinal microflora and brain cells. The connection between the oral cavity and gut through microbiota indicates that the oral microbial community may play a role in mediating complex communication between the oral cavity and the CNS; however, underlying mechanisms have yet to be fully understood. In this review, we present an overview of key concepts and potential mechanisms of interaction between the oral-gut-brain axis based on previous research, focusing on how the oral microbiome (especially the periodontal pathogens) impacts IS and its risk factors, as well as the mediating role of immune system homeostasis, and providing potential preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xianhui Kang
- Department of Anesthesiology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaofeng Bai
- Department of Oral and Maxillofacial Surgery, Stomatology Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Bei Pu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Daniel Smerin
- Department of Neurosurgery, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Liang Zhao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Xu Y, Yu M, Huang X, Wang G, Wang H, Zhang F, Zhang J, Gao X. Differences in salivary microbiome among children with tonsillar hypertrophy and/or adenoid hypertrophy. mSystems 2024; 9:e0096824. [PMID: 39287377 PMCID: PMC11494981 DOI: 10.1128/msystems.00968-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Children diagnosed with severe tonsillar hypertrophy display discernible craniofacial features distinct from those with adenoid hypertrophy, prompting illuminating considerations regarding microbiota regulation in this non-inflammatory condition. The present study aimed to characterize the salivary microbial profile in children with tonsillar hypertrophy and explore the potential functionality therein. A total of 112 children, with a mean age of 7.79 ± 2.41 years, were enrolled and divided into the tonsillar hypertrophy (TH) group (n = 46, 8.4 ± 2.5 years old), adenoid hypertrophy (AH) group (n = 21, 7.6 ± 2.8 years old), adenotonsillar hypertrophy (ATH) group (n = 23, 7.2 ± 2.1 years old), and control group (n = 22, 8.6 ± 2.1 years old). Unstimulated saliva samples were collected, and microbial profiles were analyzed by 16S rRNA sequencing of V3-V4 regions. Diversity and composition of salivary microbiome and the correlation with parameters of overnight polysomnography and complete blood count were investigated. As a result, children with tonsillar hypertrophy had significantly higher α-diversity indices (P<0.05). β-diversity based on Bray-Curtis distance revealed that the salivary microbiome of the tonsillar hypertrophy group had a slight separation from the other three groups (P<0.05). The linear discriminant analysis effect size (LEfSe) analysis indicated that Gemella was most closely related to tonsillar hypertrophy, and higher abundance of Gemella, Parvimonas, Dialister, and Lactobacillus may reflect an active state of immune regulation. Meanwhile, children with different degrees of tonsillar hypertrophy shared similar salivary microbiome diversity. This study demonstrated that the salivary microbiome in pediatric tonsillar hypertrophy patients had different signatures, highlighting that the site of upper airway obstruction primarily influences the salivary microbiome rather than hypertrophy severity.IMPORTANCETonsillar hypertrophy is the most frequent cause of upper airway obstruction and one of the primary risk factors for pediatric obstructive sleep apnea (OSA). Studies have discovered that children with isolated tonsillar hypertrophy exhibit different craniofacial morphology features compared with those with isolated adenoid hypertrophy or adenotonsillar hypertrophy. Furthermore, characteristic salivary microbiota from children with OSA compared with healthy children has been identified in our previous research. However, few studies provided insight into the relationship between the different sites of upper airway obstruction resulting from the enlargement of pharyngeal lymphoid tissue at different sites and the alterations in the microbiome. Here, to investigate the differences in the salivary microbiome of children with tonsillar hypertrophy and/or adenoid hypertrophy, we conducted a cross-sectional study and depicted the unique microbiome profile of pediatric tonsillar hypertrophy, which was mainly characterized by a significantly higher abundance of genera belonging to phyla Firmicutes and certain bacteria involving in the immune response in tonsillar hypertrophy, offering novel perspectives for future related research.
Collapse
Affiliation(s)
- Ying Xu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Min Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xin Huang
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Guixiang Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Hua Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Fengzhen Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jie Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xuemei Gao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| |
Collapse
|
5
|
Yuan K, Xu S, Liu G, Han Y, Hu J, Zhang W, Zhang Z, Liu L, Huang Z, Zhu Y, Liu S. Porphyromonas gingivalis Promotes Oral Squamous Cell Carcinoma Progression by Modulating Autophagy. Oral Dis 2024. [PMID: 39435608 DOI: 10.1111/odi.15157] [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: 04/09/2024] [Revised: 08/16/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVES Porphyromonas gingivalis (P. gingivalis) is a keystone periodontal pathogen associated with various gastro-intestinal tract cancers. However, whether P. gingivalis can promote oral squamous cell carcinoma (OSCC) and the underlying mechanism associated with such promotion remain unclear. MATERIALS AND METHODS In this study, OSCC xenograft models were used to evaluate the effects of P. gingivalis on tumor progression. The functional studies were done on several OSCC cell lines in vitro. P. gingivalis-specific 16S rRNA fluorescent in situ hybridization (FISH) was used to test its prevalence in clinical samples. RESULTS We found that P. gingivalis increased tumor volume and tumor growth in OSCC nude models. Functional studies demonstrated that P. gingivalis inhibited the apoptosis of OSCC cells by promoting cellular autophagy. P. gingivalis was more prevalent in FISH samples from patients with OSCC than from patients with leukoplakia or healthy subjects (70% vs. 47.2% vs. 33.3%, p = 0.045 and p < 0.001, respectively). CONCLUSION These data suggest that P. gingivalis plays an accelerating role in OSCC progression and contributes to OSCC by enhancing the autophagy pathway to reduce carcinoma apoptosis.
Collapse
Affiliation(s)
- Keyong Yuan
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengming Xu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglong Liu
- Department of Stomatology, Tengzhou Central People's Hospital, Jining Medical College, Tengzhou, China
| | - Yong Han
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingzhou Hu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wuchang Zhang
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyuan Zhang
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liu Liu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengwei Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Implant Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuli Liu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Nurkolis F, Utami TW, Alatas AI, Wicaksono D, Kurniawan R, Ratmandhika SR, Sukarno KT, Pahu YGP, Kim B, Tallei TE, Tjandrawinata RR, Alhasyimi AA, Surya R, Helen H, Halim P, Muhar AM, Syahputra RA. Can salivary and skin microbiome become a biodetector for aging-associated diseases? Current insights and future perspectives. FRONTIERS IN AGING 2024; 5:1462569. [PMID: 39484071 PMCID: PMC11524912 DOI: 10.3389/fragi.2024.1462569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Growth and aging are fundamental elements of human development. Aging is defined by a decrease in physiological activities and higher illness vulnerability. Affected by lifestyle, environmental, and hereditary elements, aging results in disorders including cardiovascular, musculoskeletal, and neurological diseases, which accounted for 16.1 million worldwide deaths in 2019. Stress-induced cellular senescence, caused by DNA damage, can reduce tissue regeneration and repair, promoting aging. The root cause of many age-related disorders is inflammation, encouraged by the senescence-associated secretory phenotype (SASP). Aging's metabolic changes and declining immune systems raise illness risk via promoting microbiome diversity. Stable, individual-specific skin and oral microbiomes are essential for both health and disease since dysbiosis is linked with periodontitis and eczema. Present from birth to death, the human microbiome, under the influence of diet and lifestyle, interacts symbiotically with the body. Poor dental health has been linked to Alzheimer's and Parkinson's diseases since oral microorganisms and systemic diseases have important interactions. Emphasizing the importance of microbiome health across the lifetime, this study reviews the understanding of the microbiome's role in aging-related diseases that can direct novel diagnosis and treatment approaches.
Collapse
Affiliation(s)
- Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | - Trianna Wahyu Utami
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Aiman Idrus Alatas
- Program of Clinical Microbiology Residency, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Danar Wicaksono
- Alumnus Department of Dermatology and Venereology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rudy Kurniawan
- Graduate School of Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | | | | | | | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, Indonesia
| | | | - Ananto Ali Alhasyimi
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
7
|
Lin B, Pathak JL, Gao H, Zhou Z, Ser HL, Wu L, Lee LH, Wang L, Chen J, Zhong M. A pilot study examining periodontally healthy middle-aged humans and monkeys display different levels of alveolar bone resorption, gingival inflammatory infiltrate, and salivary microbiota profile. PLoS One 2024; 19:e0311282. [PMID: 39413077 PMCID: PMC11482700 DOI: 10.1371/journal.pone.0311282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/13/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Monkeys are an appropriate model for periodontal research owing to their similar dental anatomy and physiology unlike humans. Extensive literature exists on pathological periodontitis in monkeys and humans, although concerns regarding whether healthy middle-aged monkeys and humans display the same periodontal and oral microbial status remains unclear. AIMS AND OBJECTIVES The current study aimed to compare alveolar bone resorption, gingival inflammatory infiltrate, and salivary microbiota profile in periodontally healthy middle-aged humans and monkeys. METHODS CBCT examination and histological analysis were performed to compare the periodontal status in middle-aged healthy humans and monkeys. Oral saliva16S rRNA sequencing was performed to analyze the oral microbial profile. RESULTS The alveolar resorption was compared between humans and monkeys, to determine the periodontal health. The percentage attachment of attachment loss was more around the posteriors teeth in humans when compared to monkeys (p<0.05). The degree of gingival inflammation was analyzed in both the groups, the expression of CD 34,45was higher in humans. 16S rRNA analysis demonstrated less diversity of salivary microorganisms in humans than in monkeys. The relative abundance of Aggregatibacter, Haemophilus, Gemella, and Porphyromonas at the genus level was significantly less in humans than in monkeys (p(<0.05). CONCLUSION The periodontally healthy middle-aged humans and monkeys display different alveolar bone resorption and gingival inflammatory infiltrate levels. Furthermore, the salivary microbiota profile showed distinctly different oral microbiomes in these two primates. Our results suggest that the difference in alveolar bone status and gingival inflammatory infiltrate in healthy humans and monkeys might be associated with the diversity of the oral microbiome.
Collapse
Affiliation(s)
- Bingpeng Lin
- Department of Orthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Janak L. Pathak
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongbin Gao
- Guangdong Laboratory Animals Monitoring Institute, Key Laboratory of Guangdong Laboratory Animals, Guangzhou, China
| | - Zijun Zhou
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hooi-Leng Ser
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Lihong Wu
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery (NBDD) Research Group, Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute(CBI), University of Nottingham Ningbo China, Ningbo, China
| | - Lijing Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jianming Chen
- Department of Orthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mei Zhong
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
8
|
Cui X, Yin L, Zhang Y, Jiang X, Li L, Bi X. Salivary microbiota composition before and after use of proton pump inhibitors in patients with laryngopharyngeal reflux: a self-control study. BMC Oral Health 2024; 24:1194. [PMID: 39379876 PMCID: PMC11460238 DOI: 10.1186/s12903-024-05000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Issues associated with proton pump inhibitor (PPI) usage have been documented. PPIs affect the gastrointestinal microbiome, as well as the saliva microbiota of healthy individuals. However, the alterations in the saliva microbiota of laryngopharyngeal reflux (LPR) patients remain unclear. This study aims to examine the composition of saliva microbiota in LPR patients before and after PPI usage through a self-controlled study. METHODS Thirty-two adult LPR patients participated in the study. Saliva samples were collected before and after an 8-week regimen of twice-daily administration of 20-mg esomeprazole. The impact of PPI administration on bacterial communities was assessed using 16 S rRNA gene sequencing. The functional and metabolic changes in saliva microbial communities after PPI usage were analyzed using PICRUSt2 based on our 16 S rRNA gene sequencing results. RESULTS The alpha diversity within the salivary microbiota, as measured by the PD-whole-tree index, exhibited a significant difference between samples collected before and after PPI application (P = 0.038). Additionally, PCoA analysis of unweighted UniFrac distances (beta diversity) revealed distinct separation of saliva sample microbiota structures before and after PPI application in LPR patients, with statistical significance (Adonis test, R2 = 0.063, P< 0.010). Taxon-based analysis indicated that PPI administration increased the abundance of Epsilonproteobacteria, Campylobacterales, Campylobacteraceae, Campylobacter, and Campylobacter_gracilis, while reducing the abundance of Lactobacillaceae and Lactobacillus in salivary samples ( P< 0.050). Using LEfSe to compare bacterial abundances, Bacillaceae and Anoxybacillus were found to be enriched before PPI usage in LPR patients. Furthermore, the proportion of genes responsible for indole alkaloid biosynthesis in the salivary microbiota of LPR patients significantly increased after PPI therapy (P< 0.050). CONCLUSIONS These findings indicate that PPIs induce alterations in the salivary microbiota of LPR patients. CHINESE CLINICAL TRIAL REGISTRY No. ChiCTR2300067507. Registered on January 10,2023 retrospectively. LEVEL OF EVIDENCE: 4
Collapse
Affiliation(s)
- Xiaohuan Cui
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
- Department of Otorhinolaryngology Head and Neck Surgery, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100091, China
| | - Longlong Yin
- Hebei North University, Zhangjiakou, 075051, China
| | - Yanping Zhang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China.
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China.
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China.
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China.
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China.
- Department of Otorhinolaryngology Head and Neck Surgery, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100091, China.
| | - Xingwang Jiang
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
- Department of Otorhinolaryngology Head and Neck Surgery, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100091, China
| | - Lina Li
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
- Department of Otorhinolaryngology Head and Neck Surgery, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100091, China
| | - Xinxin Bi
- Senior Department of Otolaryngology Head and Neck Surgery, the Sixth Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, 100048, China
- State Key Laboratory of Hearing and Balance Science, Beijing, 100853, China
- National Clinical Research Center for Otolaryngologic Diseases, Beijing, 100853, China
- Key Laboratory of Hearing Science, Ministry of Education, Beijing, 100853, China
- Beijing Key Laboratory of Hearing Impairment Prevention and Treatment, Beijing, 100853, China
- Department of Otorhinolaryngology Head and Neck Surgery, the Eighth Medical Center, Chinese PLA General Hospital, Beijing, 100091, China
| |
Collapse
|
9
|
Sulaiman Y, Pacauskienė IM, Šadzevičienė R, Anuzyte R. Oral and Gut Microbiota Dysbiosis Due to Periodontitis: Systemic Implications and Links to Gastrointestinal Cancer: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1416. [PMID: 39336457 PMCID: PMC11433653 DOI: 10.3390/medicina60091416] [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: 07/02/2024] [Revised: 08/19/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024]
Abstract
Periodontitis can disrupt oral and gut microbiota, leading to dysbiosis that affects overall systemic health. Besides the spread of periodontal pathogens by the hematogenous route, they can also be translocated into the gastrointestinal tract, possibly intervening in the neoplastic process in the gastrointestinal tract. This manuscript reviews the relationship between oral and gut microbiota due to periodontitis, discussing systemic health implications and potential links to gastrointestinal cancer. This article highlights the significance and effect of dysbiosis in the gut, emphasizing the importance of maintaining oral health to prevent systemic diseases. Lastly, it will go through therapeutic innovations such as probiotics and oral microbiota analysis tools for systemic disease detection. These findings will mark the integration of oral health management in clinical practice to lower systemic disease risk and improve overall patient outcomes. Aim of work: This manuscript aims to unravel the pathological interaction between oral and gut microbiota and their bidirectional effect on systemic diseases. Materials and methods: The review was performed using the MEDLINE and ScienceDirect databases. Reviewed articles were published in English between the year 2015 and 2024. The search used keywords such as ("oral microbiota" AND "periodontal disease") OR ("oral microbiota" AND "gastrointestinal cancer") OR ("Porphyromonas gingivalis" AND "periodontal disease") OR ("Helicobacter pylori" AND "gastric cancer") OR ("gut microbiome" AND "inflammatory bowel disease") OR ("oral microbiome" AND "systemic diseases"). Conclusions: The dysbiotic change in the oral cavity due to periodontitis is linked directly and indirectly to systemic diseases such as IBS, neurodegenerative diseases, muscle joint diseases, respiratory infections, and gastrointestinal cancer; this underscores the importance of maintaining oral hygiene for prophylaxis of oral diseases and the prevention of systemic diseases. A better understanding of the interconnections between oral health and systemic diseases will integrate oral health management to offer new prevention, diagnostic, and treatment opportunities to improve overall patient outcomes.
Collapse
Affiliation(s)
- Yaman Sulaiman
- Clinic of Dental and Oral Pathology, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50161 Kaunas, Lithuania
| | - Ingrida Marija Pacauskienė
- Clinic of Dental and Oral Pathology, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50161 Kaunas, Lithuania
| | - Renata Šadzevičienė
- Clinic of Dental and Oral Pathology, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50161 Kaunas, Lithuania
| | - Rugile Anuzyte
- Clinic of Dental and Oral Pathology, Faculty of Odontology, Medical Academy, Lithuanian University of Health Sciences, Eivenių Str. 2, LT-50161 Kaunas, Lithuania
| |
Collapse
|
10
|
Shang L, Deng D, Krom BP, Gibbs S. Oral host-microbe interactions investigated in 3D organotypic models. Crit Rev Microbiol 2024; 50:397-416. [PMID: 37166371 DOI: 10.1080/1040841x.2023.2211665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
The oral cavity is inhabited by abundant microbes which continuously interact with the host and influence the host's health. Such host-microbe interactions (HMI) are dynamic and complex processes involving e.g. oral tissues, microbial communities and saliva. Due to difficulties in mimicking the in vivo complexity, it is still unclear how exactly HMI influence the transition between healthy status and disease conditions in the oral cavity. As an advanced approach, three-dimensional (3D) organotypic oral tissues (epithelium and mucosa/gingiva) are being increasingly used to study underlying mechanisms. These in vitro models were designed with different complexity depending on the research questions to be answered. In this review, we summarised the existing 3D oral HMI models, comparing designs and readouts, discussing applications as well as future perspectives.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Betser-Cohen G, Mayuoni-Kirsehenbaum L, Zubeidat K, Horev Y, Berchoer LE, Cohen A, Aframian D, Amiel M, Hovav AH. Identification of oral bacteria as a new forensic tool for saliva detection. Forensic Sci Int 2024; 361:112112. [PMID: 38917506 DOI: 10.1016/j.forsciint.2024.112112] [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: 01/18/2024] [Revised: 05/12/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024]
Abstract
Body fluid detection is an important component in the toolbox of forensic scientists, with saliva playing a particularly critical role in forensic evidence. Given that each body fluid possesses a distinct microbiome, the identification of body fluid based on specific representatives of the microbiota presents an appealing approach for forensic applications. In this study, we have developed a real-time polymerase chain reaction (RT-PCR)-based method for the precise identification of saliva, focusing on three bacteria highly associated with saliva but not with other tested body fluids -Porphyromonas gingivalis, Fusobacterium nucleatum, and Streptococcus salivarius. The inclusion of these three bacterial species enhances the accuracy of detection and reinforces validation. Notably, specific identification of saliva was achievable even at low concentrations where Phadebas, a commonly used method for saliva detection, proved ineffective. Importantly, bacteria-based saliva detection utilizes DNA generated for small tandem repeats (STR) profiling, facilitating seamless integration into forensic laboratories and optimizing DNA sample utilization. This study collectively proposes an effective bacterial DNA-based approach for saliva identification, demonstrating promising potential for forensic applications.
Collapse
Affiliation(s)
- Gili Betser-Cohen
- Division of Identification and Forensic Science, Police National HQ, Haim Bar-Lev 1 Road, Jerusalem, Israel.
| | - Lina Mayuoni-Kirsehenbaum
- Division of Identification and Forensic Science, Police National HQ, Haim Bar-Lev 1 Road, Jerusalem, Israel
| | - Khaled Zubeidat
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yael Horev
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Luba Eli Berchoer
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Adiel Cohen
- Department of Obstetrics and Gynecology, Hadassah Medical Organization and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Doron Aframian
- Department of Oral Medicine, Sedation and Imaging, Faculty of Dental Medicine, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Merav Amiel
- Division of Identification and Forensic Science, Police National HQ, Haim Bar-Lev 1 Road, Jerusalem, Israel
| | - Avi-Hai Hovav
- Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
12
|
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
|
13
|
Regueira-Iglesias A, Suárez-Rodríguez B, Blanco-Pintos T, Relvas M, Alonso-Sampedro M, Balsa-Castro C, Tomás I. The salivary microbiome as a diagnostic biomarker of periodontitis: a 16S multi-batch study before and after the removal of batch effects. Front Cell Infect Microbiol 2024; 14:1405699. [PMID: 39071165 PMCID: PMC11272481 DOI: 10.3389/fcimb.2024.1405699] [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: 03/23/2024] [Accepted: 06/17/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Microbiome-based clinical applications that improve diagnosis related to oral health are of great interest to precision dentistry. Predictive studies on the salivary microbiome are scarce and of low methodological quality (low sample sizes, lack of biological heterogeneity, and absence of a validation process). None of them evaluates the impact of confounding factors as batch effects (BEs). This is the first 16S multi-batch study to analyze the salivary microbiome at the amplicon sequence variant (ASV) level in terms of differential abundance and machine learning models. This is done in periodontally healthy and periodontitis patients before and after removing BEs. Methods Saliva was collected from 124 patients (50 healthy, 74 periodontitis) in our setting. Sequencing of the V3-V4 16S rRNA gene region was performed in Illumina MiSeq. In parallel, searches were conducted on four databases to identify previous Illumina V3-V4 sequencing studies on the salivary microbiome. Investigations that met predefined criteria were included in the analysis, and the own and external sequences were processed using the same bioinformatics protocol. The statistical analysis was performed in the R-Bioconductor environment. Results The elimination of BEs reduced the number of ASVs with differential abundance between the groups by approximately one-third (Before=265; After=190). Before removing BEs, the model constructed using all study samples (796) comprised 16 ASVs (0.16%) and had an area under the curve (AUC) of 0.944, sensitivity of 90.73%, and specificity of 87.16%. The model built using two-thirds of the specimens (training=531) comprised 35 ASVs (0.36%) and had an AUC of 0.955, sensitivity of 86.54%, and specificity of 90.06% after being validated in the remaining one-third (test=265). After removing BEs, the models required more ASVs (all samples=200-2.03%; training=100-1.01%) to obtain slightly lower AUC (all=0.935; test=0.947), lower sensitivity (all=81.79%; test=78.85%), and similar specificity (all=91.51%; test=90.68%). Conclusions The removal of BEs controls false positive ASVs in the differential abundance analysis. However, their elimination implies a significantly larger number of predictor taxa to achieve optimal performance, creating less robust classifiers. As all the provided models can accurately discriminate health from periodontitis, implying good/excellent sensitivities/specificities, the salivary microbiome demonstrates potential clinical applicability as a precision diagnostic tool for periodontitis.
Collapse
Affiliation(s)
- Alba Regueira-Iglesias
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Berta Suárez-Rodríguez
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Triana Blanco-Pintos
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marta Relvas
- Instituto Universitário de Ciências da Saúde, Cooperativa de Ensino Superior Politécnico e Universitário (IUCS-CESPU), Unidade de Investigação em Patologia e Reabilitação Oral (UNIPRO), Gandra, Portugal
| | - Manuela Alonso-Sampedro
- Department of Internal Medicine and Clinical Epidemiology, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario, Santiago de Compostela, Spain
| | - Carlos Balsa-Castro
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Instituto de Investigación Sanitaria de Santiago (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
14
|
Liu J, Yue Q, Zhang S, Xu J, Jiang X, Su Q, Sun L, Li B, Li K, Su L, Zhao L. A pilot study on oral microbiome in electronic cigarettes consumers versus traditional cigarettes smokers. Folia Microbiol (Praha) 2024:10.1007/s12223-024-01185-w. [PMID: 38954243 DOI: 10.1007/s12223-024-01185-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
Oral microorganisms are closely related to oral health, the occurrence of some oral diseases is associated with changes in the oral microbiota, and many studies have demonstrated that traditional smoking can affect the oral microbial community. However, due to the short time since the emergence of e-cigarettes, fewer studies are comparing oral microorganisms for users of e-cigarettes versus cigarettes. We collected saliva from 40 non-smokers (NS), 46 traditional cigarette smokers (TS), and 27 e-cigarette consumers (EC), aged between 18 and 35 years. We performed 16S rRNA gene sequencing on the saliva samples collected to study the effects of e-cigarettes versus traditional cigarettes on the oral microbiome. The results showed that compared with the NS group, the alpha diversity of oral flora in saliva was altered in the TS group, with no significant change in the e-cigarette group. Compared with the NS and EC groups, the relative abundance of Actinomyces and Prevotella was increased in the TS group. However, compared with the NS and TS groups, the relative abundance of Veillonella was increased, and the relative abundance of Porphyromonas and Peptostreptococcus was decreased in the EC group. These results showed that both e-cigarettes and traditional cigarettes could alter the structure and composition of oral microbiota. The use of traditional cigarettes promotes the growth of some anaerobic bacteria, which may contribute to dental decay and bad breath over time. E-cigarettes have a different effect on the structure and composition of the oral microbial community compared to conventional cigarettes. In order to better understand the effects of e-cigarettes and traditional cigarettes on users' mouths, future studies will investigate the relationship between diseases such as dental caries and periodontitis and changes in oral microbial species levels.
Collapse
Affiliation(s)
- Jilong Liu
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, People's Republic of China
| | - Qiulin Yue
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, People's Republic of China
| | - Song Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, People's Republic of China
| | - Jing Xu
- RELX Tech. Co., Ltd, Shenzhen, People's Republic of China
| | - Xingtao Jiang
- RELX Tech. Co., Ltd, Shenzhen, People's Republic of China
| | - Qun Su
- Shandong Baoyuan Biotechnology Co., Ltd, Yantai, People's Republic of China
| | - Lei Sun
- Shandong Baoyuan Biotechnology Co., Ltd, Yantai, People's Republic of China
| | - Baojun Li
- Shandong Danhe Biotechnology Co., Ltd, Jinan, People's Republic of China
| | - Kunlun Li
- Shengshengxiangrong (Shandong) Biotechnology Co., Ltd, Jinan, People's Republic of China
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, People's Republic of China.
| | - Lin Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, People's Republic of China.
| |
Collapse
|
15
|
He B, Cao Y, Zhuang Z, Deng Q, Qiu Y, Pan L, Zheng X, Shi B, Lin L, Chen F. The potential value of oral microbial signatures for prediction of oral squamous cell carcinoma based on machine learning algorithms. Head Neck 2024; 46:1660-1670. [PMID: 38695435 DOI: 10.1002/hed.27795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 03/30/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE This study aimed to explore the potential predictive value of oral microbial signatures for oral squamous cell carcinoma (OSCC) risk based on machine learning algorithms. METHODS The oral microbiome signatures were assessed in the unstimulated saliva samples of 80 OSCC patients and 179 healthy individuals using 16S rRNA gene sequencing. Four different machine learning classifiers were used to develop prediction models. RESULTS Compared with control participants, OSCC patients had a higher microbial dysbiosis index (MDI, p < 0.001). Among four machine learning classifiers, random forest (RF) provided the best predictive performance, followed by the support vector machines, artificial neural networks and naive Bayes. After controlling the potential confounders using propensity score matching, the optimal RF model was further developed incorporating a minimal set of 20 bacteria genera, exhibiting better predictive performance than the MDI (AUC: 0.992 vs. 0.775, p < 0.001). CONCLUSIONS The novel MDI and RF model developed in this study based on oral microbiome signatures may serve as noninvasive tools for predicting OSCC risk.
Collapse
Affiliation(s)
- Baochang He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yujie Cao
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhaocheng Zhuang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingrong Deng
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yu Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lizhen Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoyan Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lisong Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Clinical Research Unit, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
16
|
Xiang X, Peng B, Liu K, Wang T, Ding P, Zhu Y, Cheng K, Ming Y. Prediction of delayed graft function by early salivary microbiota following kidney transplantation. Appl Microbiol Biotechnol 2024; 108:402. [PMID: 38951204 PMCID: PMC11217047 DOI: 10.1007/s00253-024-13236-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 07/03/2024]
Abstract
Delayed graft function (DGF) is a frequently observed complication following kidney transplantation (KT). Our prior research revealed dynamic shifts in salivary microbiota post-KT with immediate graft function (IGF), yet its behavior during DGF remains unexplored. Five recipients with DGF and 35 recipients with IGF were enrolled. Saliva samples were collected during the perioperative period, and 16S rRNA gene sequencing was performed. The salivary microbiota of IGFs changed significantly and gradually stabilized with the recovery of renal function. The salivary microbiota composition of DGFs was significantly different from that of IGFs, although the trend of variation appeared to be similar to that of IGFs. Salivary microbiota that differed significantly between patients with DGF and IGF at 1 day after transplantation were able to accurately distinguish the two groups in the randomForest algorithm (accuracy = 0.8333, sensitivity = 0.7778, specificity = 1, and area under curve = 0.85), with Selenomonas playing an important role. Bacteroidales (Spearman's r = - 0.4872 and p = 0.0293) and Veillonella (Spearmen's r = - 0.5474 and p = 0.0125) were significantly associated with the serum creatinine in DGF patients. Moreover, the significant differences in overall salivary microbiota structure between DGF and IGF patients disappeared upon long-term follow-up. This is the first study to investigate the dynamic changes in salivary microbiota in DGFs. Our findings suggested that salivary microbiota was able to predict DGF in the early stages after kidney transplantation, which might help the perioperative clinical management and early-stage intervention of kidney transplant recipients. KEY POINTS: • Salivary microbiota on the first day after KT could predict DGF. • Alterations in salivary taxa after KT are related to recovery of renal function.
Collapse
Affiliation(s)
- Xuyu Xiang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Bo Peng
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Kai Liu
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Tianyin Wang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Peng Ding
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yi Zhu
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Ke Cheng
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, 410013, China.
- Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China.
| |
Collapse
|
17
|
Ahmad P, Moussa DG, Siqueira WL. Metabolomics for dental caries diagnosis: Past, present, and future. MASS SPECTROMETRY REVIEWS 2024. [PMID: 38940512 DOI: 10.1002/mas.21896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/22/2024] [Accepted: 06/15/2024] [Indexed: 06/29/2024]
Abstract
Dental caries, a prevalent global infectious condition affecting over 95% of adults, remains elusive in its precise etiology. Addressing the complex dynamics of caries demands a thorough exploration of taxonomic, potential, active, and encoded functions within the oral ecosystem. Metabolomic profiling emerges as a crucial tool, offering immediate insights into microecosystem physiology and linking directly to the phenotype. Identified metabolites, indicative of caries status, play a pivotal role in unraveling the metabolic processes underlying the disease. Despite challenges in metabolite variability, the use of metabolomics, particularly via mass spectrometry and nuclear magnetic resonance spectroscopy, holds promise in caries research. This review comprehensively examines metabolomics in caries prevention, diagnosis, and treatment, highlighting distinct metabolite expression patterns and their associations with disease-related bacterial communities. Pioneering in approach, it integrates singular and combinatory metabolomics methodologies, diverse biofluids, and study designs, critically evaluating prior limitations while offering expert insights for future investigations. By synthesizing existing knowledge, this review significantly advances our comprehension of caries, providing a foundation for improved prevention and treatment strategies.
Collapse
Affiliation(s)
- Paras Ahmad
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Dina G Moussa
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Walter L Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
18
|
Marruganti C, Romandini M, Gaeta C, Trovato E, Cinotti E, Rubegni P, D'Aiuto F, Grandini S. Treatment of periodontitis ameliorates the severity and extent of psoriasis-A randomized clinical trial. J Periodontal Res 2024. [PMID: 38899599 DOI: 10.1111/jre.13314] [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: 04/06/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
AIM To assess the impact of non-surgical periodontitis treatment over conventional dermatological treatment on the severity and extent of psoriasis in patients affected by comorbid psoriasis and periodontitis. METHODS Seventy-four patients affected by both psoriasis and Stages I-IV periodontitis were randomized to receive either Steps 1-2 (non-surgical) of periodontal therapy (test group; n = 37) or no treatment (control group; n = 37). The two groups were balanced in terms of psoriasis medications, with the majority of the included patients undergoing biologics (74.0%) as monotherapy, while minor proportions were under systemic medications (13.7%) or none/topical/phototherapy (12.3%). The psoriasis area severity index (PASI) was regarded as the primary outcome. The Body Surface Area (BSA) and the Dermatology Life Quality Index (DLQI) were additionally considered as dermatological outcomes. Probing pocket depth, recession depth, clinical attachment level, periodontal inflamed surface area, and full-mouth plaque and bleeding scores were also measured. [Correction added on July 5, 2024, after first online publication: The preceding sentence has been revised]. RESULTS Periodontal therapy in the test group led to statistically significant lower PASI scores at 10 weeks (mean = 3.15; standard deviation [SD] = 3.78) compared to the control group (mean = 7.11; SD = 6.09) (mean difference [MD] = -4.0; 95% confidence interval [CI]: -6.3, -1.6; p = .001). The test group also showed improvements in BSA (MD = -4.3) and periodontal parameters compared to the control group. DLQI only showed a non-statistically significant tendency (MD = -2.0). CONCLUSION Steps 1-2 of periodontal therapy showed an additional effect over conventional dermatological treatment in reducing the severity and extent of psoriasis (Clinicaltrials.gov: NCT05311501).
Collapse
Affiliation(s)
- Crystal Marruganti
- Unit of Periodontology, Endodontology and Restorative Dentistry, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Unit of Periodontology, UCL Eastman Dental Institute, London, UK
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Carlo Gaeta
- Unit of Periodontology, Endodontology and Restorative Dentistry, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Emanuele Trovato
- Unit of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Siena, Italy
| | - Elisa Cinotti
- Unit of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Siena, Italy
| | - Pietro Rubegni
- Unit of Dermatology, Department of Medical, Surgical and Neurological Science, University of Siena, Siena, Italy
| | | | - Simone Grandini
- Unit of Periodontology, Endodontology and Restorative Dentistry, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
19
|
Agurto MG, Bozorgi SS, Carpenter G, Ramirez V, Burke M, Felipe Gutierrez M, Cordova C, Banerjee A. Longitudinal study of the role of salivary proteins on radiation-related caries onset in head and neck cancer patients using 5000 ppm fluoride dentifrice up to one-year post-intensity modulated radiotherapy. Clin Oral Investig 2024; 28:379. [PMID: 38886263 DOI: 10.1007/s00784-024-05788-y] [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: 04/01/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
OBJECTIVES Longitudinal assessment of the role of specific proteins on radiotherapy caries (RC) onset in head and neck cancer patients(HNC) up to one-year post-IMRT using a 5000ppm fluoride paste daily. MATERIALS AND METHODS Dental status/salivary protein data were obtained from 40 HNC patients pre-IMRT, six months (T1) and 12 months (T2) post-IMRT (ethical approval/consent). DMFT/salivary parameters were quantified, including flow rate, mucin 5B/7, Immunoglobulin A (IgA), cystatin S and α-amylase. RESULTS 45% patients had at least one carious lesion at T2, a significant reduction in the number of remaining teeth (65% <21), salivary flow rate (< 50%) and, protein secretion (< 0.05) post-IMRT. T1 IgA concentration/secretion rate was associated with RC (p < 0.05). Finally, IgA and total protein concentration obtained at T1 could provide a predictive pattern (AUC 82.3%) for the patients more predisposed to developing RC at T2. CONCLUSIONS This study demonstrated the significant association of RC with salivary proteins in HNC patients treated with IMRT, revealing the potential role of salivary proteins in the early diagnosis of RC. CLINICAL RELEVANCE This research contributes to revealing salivary proteins association with RC, and its role in early diagnosis. Therefore, this could be the first step towards personalized medicine approaches to improve this group quality-of-life.
Collapse
Affiliation(s)
- Maria Gonzalez Agurto
- Universidad de los Andes, Santiago, Chile.
- Faculty of Dentistry, Oral & Craniofacial Sciences, King?s College London, London, UK.
| | - Sophie S Bozorgi
- Faculty of Dentistry, Oral & Craniofacial Sciences, King?s College London, London, UK
| | - Guy Carpenter
- Faculty of Dentistry, Oral & Craniofacial Sciences, King?s College London, London, UK
| | | | - Mary Burke
- Faculty of Dentistry, Oral & Craniofacial Sciences, King?s College London, London, UK
| | | | | | - Avijit Banerjee
- Faculty of Dentistry, Oral & Craniofacial Sciences, King?s College London, London, UK
| |
Collapse
|
20
|
Easter QT, Fernandes Matuck B, Beldorati Stark G, Worth CL, Predeus AV, Fremin B, Huynh K, Ranganathan V, Ren Z, Pereira D, Rupp BT, Weaver T, Miller K, Perez P, Hasuike A, Chen Z, Bush M, Qu X, Lee J, Randell SH, Wallet SM, Sequeira I, Koo H, Tyc KM, Liu J, Ko KI, Teichmann SA, Byrd KM. Single-cell and spatially resolved interactomics of tooth-associated keratinocytes in periodontitis. Nat Commun 2024; 15:5016. [PMID: 38876998 PMCID: PMC11178863 DOI: 10.1038/s41467-024-49037-y] [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: 08/26/2023] [Accepted: 05/20/2024] [Indexed: 06/16/2024] Open
Abstract
Periodontitis affects billions of people worldwide. To address relationships of periodontal niche cell types and microbes in periodontitis, we generated an integrated single-cell RNA sequencing (scRNAseq) atlas of human periodontium (34-sample, 105918-cell), including sulcular and junctional keratinocytes (SK/JKs). SK/JKs displayed altered differentiation states and were enriched for effector cytokines in periodontitis. Single-cell metagenomics revealed 37 bacterial species with cell-specific tropism. Fluorescence in situ hybridization detected intracellular 16 S and mRNA signals of multiple species and correlated with SK/JK proinflammatory phenotypes in situ. Cell-cell communication analysis predicted keratinocyte-specific innate and adaptive immune interactions. Highly multiplexed immunofluorescence (33-antibody) revealed peri-epithelial immune foci, with innate cells often spatially constrained around JKs. Spatial phenotyping revealed immunosuppressed JK-microniches and SK-localized tertiary lymphoid structures in periodontitis. Here, we demonstrate impacts on and predicted interactomics of SK and JK cells in health and periodontitis, which requires further investigation to support precision periodontal interventions in states of chronic inflammation.
Collapse
Affiliation(s)
- Quinn T Easter
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Bruno Fernandes Matuck
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
| | | | | | | | | | - Khoa Huynh
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Zhi Ren
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Diana Pereira
- Center for Oral Immunobiology and Regenerative Medicine, Barts Centre for Squamous Cancer, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Brittany T Rupp
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Theresa Weaver
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
| | | | - Paola Perez
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Akira Hasuike
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, Japan
| | - Zhaoxu Chen
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mandy Bush
- Respiratory TRACTS Core, Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xufeng Qu
- VCU Massey Comprehensive Cancer Center, Bioinformatics Shared Resource Core, Virginia Commonwealth University, Richmond, VA, USA
| | - Janice Lee
- Craniofacial Anomalies & Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shannon M Wallet
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Inês Sequeira
- Center for Oral Immunobiology and Regenerative Medicine, Barts Centre for Squamous Cancer, Institute of Dentistry, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Hyun Koo
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katarzyna M Tyc
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
- VCU Massey Comprehensive Cancer Center, Bioinformatics Shared Resource Core, Virginia Commonwealth University, Richmond, VA, USA
| | - Jinze Liu
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
- VCU Massey Comprehensive Cancer Center, Bioinformatics Shared Resource Core, Virginia Commonwealth University, Richmond, VA, USA
| | - Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Physics, Cavendish Laboratory, Cambridge, UK
| | - Kevin M Byrd
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation & Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA.
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
21
|
Ding J, Li J, Zhang C, Tan L, Zhao C, Gao L. High-Throughput Combined Analysis of Saliva Microbiota and Metabolomic Profile in Chinese Periodontitis Patients: A Pilot Study. Inflammation 2024; 47:874-890. [PMID: 38148454 DOI: 10.1007/s10753-023-01948-6] [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: 09/03/2023] [Revised: 11/28/2023] [Accepted: 12/11/2023] [Indexed: 12/28/2023]
Abstract
The onset and progression of periodontitis involves complicated interactions between the dysbiotic oral microbiota and disrupted host immune-inflammatory response, which can be mirrored by the changes in salivary metabolites profile. This pilot study sought to examine the saliva microbiome and metabolome in the Chinese population by the combined approach of 16s rRNA sequencing and high-throughput targeted metabolomics to discover potential cues for host-microbe metabolic interactions. Unstimulated whole saliva samples were collected from eighteen Stage III and IV periodontitis patients and thirteen healthy subjects. Full-mouth periodontal parameters were recorded. The taxonomic composition of microbiota was obtained by 16s rRNA sequencing, and the metabolites were identified and measured by ultra-high performance liquid chromatography and mass spectrometry-based metabolomic analysis. The oral microbiota composition displayed marked changes where the abundance of 93 microbial taxa differed significantly between the periodontitis and healthy group. Targeted metabolomics identified 103 differential metabolites between the patients and healthy individuals. Functional enrichment analysis demonstrated the upregulation of protein digestion and absorption, histidine metabolism, and nicotinate and nicotinamide metabolism pathways in the dysbiotic microbiota, while the ferroptosis, tryptophan metabolism, glutathione metabolism, and carbon metabolism pathways were upregulated in the patients. Correlation analysis confirmed positive relationships between the clinical parameters, pathogen abundances, and disease-related metabolite levels. The integral analysis of the saliva microbiome and metabolome yielded an accurate presentation of the dysbiotic oral microbiome and functional alterations in host-microbe metabolism. The microbial and metabolic profiling of the saliva could be a potential tool in the diagnosis, prognosis evaluation, and pathogenesis study of periodontitis.
Collapse
Affiliation(s)
- Jing Ding
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Jinyu Li
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Chi Zhang
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Lingping Tan
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Chuanjiang Zhao
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Li Gao
- Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China.
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
22
|
Shinde DB, Mahore JG, Giram PS, Singh SL, Sharda A, Choyan D, Musale S. Microbiota of Saliva: A Non-invasive Diagnostic Tool. Indian J Microbiol 2024; 64:328-342. [PMID: 39010986 PMCID: PMC11246313 DOI: 10.1007/s12088-024-01219-4] [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: 06/23/2023] [Accepted: 01/30/2024] [Indexed: 07/17/2024] Open
Abstract
Potential of salivary microbiota as a non-invasive diagnostic tool for various diseases are explained in the present review. Traditional diagnostic methods rely on blood, which has limitations in terms of collection and biomarker specificity. We discuss the concept of normal flora and how disruptions in oral microbiota can be indicative of diseases. Saliva, harboring a diverse microbial community, offers promise as a diagnostic biomarker source for oral and non-oral conditions. We delve into the role of microbial dysbiosis in disease pathogenesis and the prospects of using biological indicators like dysbiosis for diagnosis, prediction, and monitoring. This review also emphasizes the significance of saliva microbiota in advancing early disease detection and timely intervention. We addressed the following research question and objectives: Can the microbiota of saliva serve as a non-invasive diagnostic tool for the early detection and monitoring of both oral and non-oral diseases? To achieve this, we will explore the normal flora of microorganisms in the oral cavity, the impact of microbial dysbiosis, and the potential of using specific pathogenic microorganisms as biomarkers. Additionally, we will investigate the correlation between oral and non-oral diseases by analyzing total saliva or site-specific dental biofilms for signs of symbiosis or dysbiosis. This research seeks to contribute valuable insights into the development of a non-invasive diagnostic approach with broad applications in healthcare.
Collapse
Affiliation(s)
- Dasharath B Shinde
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University), Pune, 412115 India
| | - Jayashri G Mahore
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
- Sinhgad College of Pharmacy, Vadgaon (Bk.), Pune, 411041 India
| | - Prabhanjan S Giram
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
- Department of Pharmaceutical Sciences, The State University of New York, Buffalo, NY 14214 USA
| | - Shaktikumar L Singh
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Aditi Sharda
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Divya Choyan
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| | - Shubham Musale
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, 411018 India
| |
Collapse
|
23
|
He Y, Li XY, Hu AQ, Qian D. Salivary microbiome is associated with the response to chemoradiotherapy in initially inoperable patients with esophageal squamous cell carcinoma. J Oral Microbiol 2024; 16:2359887. [PMID: 38813524 PMCID: PMC11134033 DOI: 10.1080/20002297.2024.2359887] [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: 02/02/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Background The salivary microbiome may interact with chemoradiotherapy through dynamic changes in microbial composition and systemic immunity. We aimed to explore the association between the salivary microbiome and response to chemoradiotherapy in initially inoperable patients with local advanced esophageal squamous cell carcinoma (LAESCC). Methods Salivary and peripheral blood samples were collected before and after chemoradiotherapy. The microbiome and metabolic pathways were analyzed by 16S ribosomal RNA sequencing and liquid chromatography tandem mass spectrometry/Mass spectrometry analyses. Results The salivary microbiome exhibited characteristic variations between patients and healthy controls. A significant correlation was found between Prevotella_salivae, Saccharibacteria_TM7_G3_bacterium_HMT_351, and Veillonellaceae_G1_bacterium_HMT_129 and pathological complete response (pCR) in initially inoperable patients who underwent surgery. The PICRUSt suggested that immune diseases and cell motility were different in tumor compared to normal groups. KEGG enrichment analysis showed enriched lipid metabolism, signal transduction, and membrane transport in the tumor group. CD3+CD8 T cells, IL6, IL10, and IFNγ exhibited an increasing trend during the treatment process of chemoradiotherapy. Conclusions Our study demonstrated that variations in specific saliva taxa associated with host immunomodulatory cells and cytokines could be promising for early efficacy prediction of chemoradiotherapy in initially inoperable patients with LAESCC.
Collapse
Affiliation(s)
- Yuan He
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiao-Yang Li
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - An-Qi Hu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
24
|
Min K, Glowacki AJ, Bosma ML, McGuire JA, Tian S, McAdoo K, DelSasso A, Fourre T, Gambogi RJ, Milleman J, Milleman KR. Quantitative analysis of the effects of essential oil mouthrinses on clinical plaque microbiome: a parallel-group, randomized trial. BMC Oral Health 2024; 24:578. [PMID: 38762482 PMCID: PMC11102605 DOI: 10.1186/s12903-024-04365-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
BACKGROUND The rich diversity of microorganisms in the oral cavity plays an important role in the maintenance of oral health and development of detrimental oral health conditions. Beyond commonly used qualitative microbiome metrics, such as relative proportions or diversity, both the species-level identification and quantification of bacteria are key to understanding clinical disease associations. This study reports the first-time application of an absolute quantitative microbiome analysis using spiked DNA standards and shotgun metagenome sequencing to assess the efficacy and safety of product intervention on dental plaque microbiome. METHODS In this parallel-group, randomized clinical trial, essential oil mouthrinses, including LISTERINE® Cool Mint Antiseptic (LCM), an alcohol-containing prototype mouthrinse (ACPM), and an alcohol-free prototype mouthrinse (AFPM), were compared against a hydroalcohol control rinse on clinical parameters and the oral microbiome of subjects with moderate gingivitis. To enable a sensitive and clinically meaningful measure of bacterial abundances, species were categorized according to their associations with oral conditions based on published literature and quantified using known amounts of spiked DNA standards. RESULTS Multivariate analysis showed that both LCM and ACPM shifted the dysbiotic microbiome composition of subjects with gingivitis to a healthier state after 4 weeks of twice-daily use, resembling the composition of subjects with clinically healthy oral conditions recruited for observational reference comparison at baseline. The essential oil-containing mouthrinses evaluated in this study showed statistically significant reductions in clinical gingivitis and plaque measurements when compared to the hydroalcohol control rinse after 6 weeks of use. CONCLUSIONS By establishing a novel quantitative method for microbiome analysis, this study sheds light on the mechanisms of LCM mouthrinse efficacy on oral microbial ecology, demonstrating that repeated usage non-selectively resets a gingivitis-like oral microbiome toward that of a healthy oral cavity. TRIAL REGISTRATION The trial was registered on ClinicalTrials.gov on 10/06/2021. The registration number is NCT04921371.
Collapse
Affiliation(s)
- Kyungrok Min
- Johnson & Johnson Consumer Inc, Skillman, NJ, USA.
| | | | | | | | - Sandy Tian
- Johnson & Johnson Consumer Inc, Skillman, NJ, USA
| | | | | | - Tara Fourre
- Johnson & Johnson Consumer Inc, Skillman, NJ, USA
| | | | | | | |
Collapse
|
25
|
Onyia NE, Osagie E, Akhigbe P, Idemudia NL, Obuekwe O, Omoigberale A, Richards V, Coker MO. Impact of perinatal HIV exposure and infection on salivary properties among Nigerian children. BMC Oral Health 2024; 24:460. [PMID: 38627731 PMCID: PMC11020910 DOI: 10.1186/s12903-024-04159-z] [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: 01/31/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND There is growing evidence that perinatal HIV infection and exposure affect salivary pH and flow rate in children in most parts of the world, but not against the background of caries and the African demographic. This study aimed to evaluate the impact of HIV infection as well as exposure on salivary properties and their influence upon the dental caries experience among school-aged children in Nigeria. METHOD This cross-sectional study assessed the salivary flow rates and salivary pH of HIV infected and exposed school-aged (4-11) children receiving care at a Nigerian tertiary hospital. A total of 266 consenting participants which comprised of three groups as follows: (1) HIV Infected (HI) (n = 87), (2) HIV Exposed and Uninfected (HEU) (n = 82) and (3) HIV Unexposed and Uninfected (HUU) (n = 97) were recruited for the study. Questionnaires completed by parents/guardians were used for data collection. Three calibrated dentists performed oral examinations for dental caries. International Caries Detection and Assessment Scores (ICDAS) was used and presented as dmft/DMFT. Salivary pH was measured using MColourpHast™ pH indicator strips, while salivary flow rate was determined by collecting unstimulated whole saliva using the suction method. Data analysis relied on comparative statistics to determine the correlation between HIV exposure and infection on salivary pH and flow rates. RESULT Across the groups, (HI, HEU, and HUU) mean pH of the HI was significantly less than that of HEU and HUU. Similarly, there was a statistically significant difference in the SFR across the three groups (p = 0.004). Other variables such as gender, age and oral hygiene status expressed by the gingival inflammatory scores had no significant influence on the pH and SFR of study participants. There was a rather unexpected positive correlation of DMFT of HI and HEU groups with increasing salivary flow rate; though, the relationship was weak and not significant. CONCLUSION Perinatal HIV exposure and infection significantly impact salivary pH and flow rate among school-aged children in Nigeria. The findings of this study imply that HIV infection influenced the salivary pH, while HIV maternal exposure (without infection) impacted salivary flow rates when compared to the controls.
Collapse
Affiliation(s)
- Nonso E Onyia
- Department of Oral Pathology Oral Medicine Oral Diagnosis, University of Calabar, Calabar, Cross River State, Nigeria
| | - Esosa Osagie
- Research Department, Institute of Human Virology, Abuja, Nigeria
| | - Paul Akhigbe
- Research Department, Institute of Human Virology, Abuja, Nigeria
| | - Nosakhare L Idemudia
- Medical Microbiology Division, Medical Laboratory Services, University of Benin Teaching Hospital, Benin City, Nigeria
| | - Ozo Obuekwe
- Department of Oral and Maxillofacial Surgery, University of Benin, Benin City, Nigeria
| | | | - Vincent Richards
- Department of Biological Sciences, Clemson University, Clemson, SC, USA
| | - Modupe O Coker
- Department of Oral Biology, Rutgers University, Newark, NJ, USA.
| |
Collapse
|
26
|
Manzoor M, Leskelä J, Pietiäinen M, Martinez-Majander N, Könönen E, Niiranen T, Lahti L, Sinisalo J, Putaala J, Pussinen PJ, Paju S. Shotgun metagenomic analysis of the oral microbiome in gingivitis: a nested case-control study. J Oral Microbiol 2024; 16:2330867. [PMID: 38528961 PMCID: PMC10962305 DOI: 10.1080/20002297.2024.2330867] [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: 12/19/2023] [Accepted: 03/08/2024] [Indexed: 03/27/2024] Open
Abstract
Background Gingivitis, i.e. inflammation of the gums, is often induced by dentalplaque. However, its exact link to the oral microbiota remains unclear. Methods In a case-control study involving 120 participants, comprising 60 cases and 60 controls (mean age (SD) 36.6 (7.6) years; 50% males), nested within a prospective multicentre cohort study, we examined theoral microbiome composition of gingivitis patients and their controlsusing shotgun metagenomic sequencing of saliva samples. Participants underwent clinical and radiographic oral health examinations, including bleeding on probing (BOP), at six tooth sites. BOP ≥33%was considered 'generalized gingivitis/initial periodontitis'(GG/IP), and BOP <33% as 'healthy and localized gingivitis'(H/LG). Functional potential was inferred using HUMANn3. Results GG/IP exhibited an increase in the abundance of Actinomyces, Porphyromonas, Aggregatibacter, Corynebacterium, Olsenella, and Treponema, whereas H/LG exhibited an increased abundance of Candidatus Nanosynbacter. Nineteen bacterial species and fourmicrobial functional profiles, including L-methionine, glycogen, andinosine-5'-phosphate biosynthesis, were associated with GG/IP. Constructing models with multiple markers resulted in a strong predictive value for GG/IP, with an area under the curve (ROC) of 0.907 (95% CI: 0.848-0.966). Conclusion We observed distinct differences in the oral microbiome between the GG/IP and H/LG groups, indicating similar yet unique microbial profiles and emphasizing their potential role in progression of periodontal diseases.
Collapse
Affiliation(s)
- Muhammed Manzoor
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Jaakko Leskelä
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Milla Pietiäinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- Industrial Biotechnology and Food Protein Production, VTT Technical Research Centre of Finland, Espoo, Finland
| | | | - Eija Könönen
- Institute of Dentistry, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Internal Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Central Hospital, and Helsinki University, Helsinki, Finland
| | - Jukka Putaala
- Department of Neurology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Pirkko J. Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
- School of Medicine, Institute of Dentistry, University of Eastern Finland, Kuopio, Finland
| | - Susanna Paju
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Wang M, Chen J, Wang Z, Wang Y, Zhang Y, Feng Q, Wei F. Salivary microbiomes vary among orthodontic appliances and associate with clinical periodontal parameters. Orthod Craniofac Res 2024; 27:174-184. [PMID: 37985447 DOI: 10.1111/ocr.12733] [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] [Accepted: 11/06/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE To investigate the salivary bacterial communities during the first 6-month orthodontic treatment with Clear Aligners (CA) and Fixed Appliances (FA), and its correlation with clinical periodontal parameters. MATERIALS AND METHODS Saliva and periodontal parameters were sampled from individuals wearing CA or FA before treatment (T0), and after 3- (T3) and 6-month (T6) treatments. Salivary bacterial communities characterized based on the 16S rRNA V3-V4 region were compared between FA and CA and correlated with clinical periodontal parameters. RESULTS Probing Depth (PD) significantly increased at T6 in the FA group versus T0, whereas it remained stable in the CA group. The Shannon and Pielou indices were significantly higher in the FA group and significantly positively correlated with periodontal inflammation parameters. β-diversity analysis revealed distinct communities between the FA group and CA group at T6. The relative abundances of 3 genera and 15 species were significantly higher in the FA group. Among the above appliance-type related taxa, bacterial genera Selenomonas, Stomatobaculum, Olsenella and Faecalicoccus and bacterial species Selenomonas_sputigena, Dialister_invisus, Olsenella_profus, Prevotella_buccae, Cryptobacterium_curtum and Clostridium_spiroforme were significantly positively associated with periodontal parameters. CONCLUSIONS Orthodontic treatments trigger appliance-related salivary bacterial communities, highlighting the importance of developing appliance-orientated periodontal strategies during orthodontic treatments. Salivary bacterial communities harboured by patients wearing FA possess higher bacterial parameters which were associated with increasing PD, PI and Gingival Index.
Collapse
Affiliation(s)
- Mengqiao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Jitian Chen
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Ziyao Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yihua Wang
- Department of 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
| | - Yue Zhang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Qiang Feng
- Department of 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
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| |
Collapse
|
28
|
Dinis M, Tran NC. Oral immune system and microbes. MICROBES, MICROBIAL METABOLISM, AND MUCOSAL IMMUNITY 2024:147-228. [DOI: 10.1016/b978-0-323-90144-4.00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Zhang Z, Zhao H, Chen X, Tian G, Liu G, Cai J, Jia G. Implications of Jatropha curcas L. cake feed on swine health: A microbiota-gut-brain axis perspective. Anim Sci J 2024; 95:e13953. [PMID: 38783533 DOI: 10.1111/asj.13953] [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: 08/05/2023] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
The safety of Jatropha curcas L. cake (JCC) in animal feed remains under scrutiny, despite the advent of low phorbol ester (PE) variants. This study investigates the impact of low PE JCC on swine health when used as a protein feed. Pigs were fed a 5% JCC diet with a PE concentration of 0.98 mg/kg, which surprisingly still induced toxicity. Symptoms included depression, decreased food intake, increased diarrhea, along with hypothalamus and colon lesions. The toxicity was associated with a decrease in antioxidant enzymes, an increase in inflammatory cytokines in the hypothalamus, plasma, and colon, and a rise in pro-inflammatory colon microbes and metabolites. Disturbances in neurotransmitters further suggest that this toxicity is related to disruption of the microbiota-gut-brain axis, indicating that JCC's toxic elements are not solely due to PE. The sensitivity of pigs to JCC underscores the need for thorough detoxification prior to its use as feed. These findings significantly contribute to the discourse on the safety of low PE JCC in animal feed, highlighting implications for both the feed industry and public health.
Collapse
Affiliation(s)
- Zhenyu Zhang
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Animal Husbandry and Veterinary Medicine, Meishan Vocational Technical College, Meishan, Sichuan, China
- Agricultural and Rural Bureau of Dongpo District, Meishan, China
| | - Hua Zhao
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xiaoling Chen
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Tian
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Guangmang Liu
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jingyi Cai
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Jia
- Animal Nutrition Institute, Key Laboratory for Animal Disease-Resistance Nutrition of China, Sichuan Agricultural University, Chengdu, Sichuan, China
| |
Collapse
|
30
|
Wang W, Ou Z, Huang X, Wang J, Li Q, Wen M, Zheng L. Microbiota and glioma: a new perspective from association to clinical translation. Gut Microbes 2024; 16:2394166. [PMID: 39185670 DOI: 10.1080/19490976.2024.2394166] [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: 05/13/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Gliomas pose a significant challenge in oncology due to their malignant nature, aggressive growth, frequent recurrence, and complications posed by the blood-brain barrier. Emerging research has revealed the critical role of gut microbiota in influencing health and disease, indicating its possible impact on glioma pathogenesis and treatment responsiveness. This review focused on existing evidence and hypotheses on the relationship between microbiota and glioma from progression to invasion. By discussing possible mechanisms through which microbiota may affect glioma biology, this paper offers new avenues for targeted therapies and precision medicine in oncology.
Collapse
Affiliation(s)
- Wenhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Minghui Wen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
31
|
Manzoor M, Leskelä J, Pietiäinen M, Martinez-Majander N, Ylikotila P, Könönen E, Niiranen T, Lahti L, Sinisalo J, Putaala J, Pussinen PJ, Paju S. Multikingdom oral microbiome interactions in early-onset cryptogenic ischemic stroke. ISME COMMUNICATIONS 2024; 4:ycae088. [PMID: 38988699 PMCID: PMC11235082 DOI: 10.1093/ismeco/ycae088] [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: 12/27/2023] [Revised: 04/15/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024]
Abstract
Although knowledge of the role of the oral microbiome in ischemic stroke is steadily increasing, little is known about the multikingdom microbiota interactions and their consequences. We enrolled participants from a prospective multicentre case-control study and investigated multikingdom microbiome differences using saliva metagenomic datasets (n = 308) from young patients diagnosed with cryptogenic ischemic stroke (CIS) and age- and sex-matched stroke-free controls. Differentially abundant taxa were identified using Analysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC2). Functional potential was inferred using HUMANn3. Our findings revealed significant differences in the composition and functional capacity of the oral microbiota associated with CIS. We identified 51 microbial species, including 47 bacterial, 3 viral, and one fungal species associated with CIS in the adjusted model. Co-abundance network analysis highlighted a more intricate microbial network in CIS patients, indicating potential interactions and co-occurrence patterns among microbial species across kingdoms. The results of our metagenomic analysis reflect the complexity of the oral microbiome, with high diversity and multikingdom interactions, which may play a role in health and disease.
Collapse
Affiliation(s)
- Muhammed Manzoor
- Department of Oral and Maxillofacial Diseases, University of Helsinki, 00014 Helsinki, Finland
| | - Jaakko Leskelä
- Department of Oral and Maxillofacial Diseases, University of Helsinki, 00014 Helsinki, Finland
| | - Milla Pietiäinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, 00014 Helsinki, Finland
- Industrial Biotechnology and Food Protein Production, VTT Technical Research Centre of Finland, 02044 Espoo, Finland
| | - Nicolas Martinez-Majander
- Department of Neurology, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland
| | - Pauli Ylikotila
- Neurocenter, Turku University Hospital, University of Turku, 20521 Turku, Finland
| | - Eija Könönen
- Institute of Dentistry, University of Turku, 20500 Turku, Finland
| | - Teemu Niiranen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, 00271 Helsinki, Finland
- Department of Internal Medicine, Turku University Hospital and University of Turku, 20521 Turku, Finland
| | - Leo Lahti
- Department of Computing, University of Turku, 20500 Turku, Finland
| | - Juha Sinisalo
- Heart and Lung Center, Helsinki University Central Hospital, and Helsinki University, 00260 Helsinki, Finland
| | - Jukka Putaala
- Department of Neurology, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland
| | - Pirkko J Pussinen
- Department of Oral and Maxillofacial Diseases, University of Helsinki, 00014 Helsinki, Finland
- School of Medicine, Institute of Dentistry, University of Eastern Finland, 70211 Kuopio, Finland
| | - Susanna Paju
- Department of Oral and Maxillofacial Diseases, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
32
|
Liu Z, Liu J, Geng J, Wu E, Zhu J, Cong B, Wu R, Sun H. Metatranscriptomic characterization of six types of forensic samples and its potential application to body fluid/tissue identification: A pilot study. Forensic Sci Int Genet 2024; 68:102978. [PMID: 37995518 DOI: 10.1016/j.fsigen.2023.102978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
Microorganisms are potential markers for identifying body fluids (venous and menstrual blood, semen, saliva, and vaginal secretion) and skin tissue in forensic genetics. Existing published studies have mainly focused on investigating microbial DNA by 16 S rRNA gene sequencing or metagenome shotgun sequencing. We rarely find microbial RNA level investigations on common forensic body fluid/tissue. Therefore, the use of metatranscriptomics to characterize common forensic body fluids/tissue has not been explored in detail, and the potential application of metatranscriptomics in forensic science remains unknown. Here, we performed 30 metatranscriptome analyses on six types of common forensic sample from healthy volunteers by massively parallel sequencing. After quality control and host RNA filtering, a total of 345,300 unigenes were assembled from clean reads. Four kingdoms, 137 phyla, 267 classes, 488 orders, 985 families, 2052 genera, and 4690 species were annotated across all samples. Alpha- and beta-diversity and differential analysis were also performed. As a result, the saliva and skin groups demonstrated high alpha diversity (Simpson index), while the venous blood group exhibited the lowest diversity despite a high Chao1 index. Specifically, we discussed potential microorganism contamination and the "core microbiome," which may be of special interest to forensic researchers. In addition, we implemented and evaluated artificial neural network (ANN), random forest (RF), and support vector machine (SVM) models for forensic body fluid/tissue identification (BFID) using genus- and species-level metatranscriptome profiles. The ANN and RF prediction models discriminated six forensic body fluids/tissue, demonstrating that the microbial RNA-based method could be applied to BFID. Unlike metagenomic research, metatranscriptomic analysis can provide information about active microbial communities; thus, it may have greater potential to become a powerful tool in forensic science for microbial-based individual identification. This study represents the first attempt to explore the application potential of metatranscriptome profiles in forensic science. Our findings help deepen our understanding of the microorganism community structure at the RNA level and are beneficial for other forensic applications of metatranscriptomics.
Collapse
Affiliation(s)
- Zhiyong Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiajun Liu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiaojiao Geng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Enlin Wu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China
| | - Jianzhang Zhu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou 510080, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Shijiazhuang 050017, China.
| | - Riga Wu
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China.
| | - Hongyu Sun
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
33
|
Ramanathan K, Padmanabhan G, Gulilat H, Malik T. Salivary microbiome in kidney diseases: A narrative review. Cell Biochem Funct 2023; 41:988-995. [PMID: 37795946 DOI: 10.1002/cbf.3864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/01/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023]
Abstract
Many research has been conducted since the microbiota's discovery that have focused on the role it plays in health and disease. Microbiota can be divided into categories like intestinal, oral, respiratory, and skin microbiota based on the specific localized areas. To maintain homeostasis and control immunological response, the microbial populations live in symbiosis with the host. On the other hand, dysbiosis of the microbiota can cause diseases including kidney diseases and the deregulation of body functioning. We discuss the current understanding of how various kidney diseases are caused by the salivary microbiome (SM) in this overview. First, we review the studies on the salivary microbiota in diverse clinical situations. The importance of the SM in diabetic kidney disease, chronic kidney disease, membranous nephropathy, and IgA nephropathy is next highlighted. We conclude that the characteristics of the SM of patients with various kidney diseases have revealed the potential of salivary microbial markers as noninvasive tool for the detection of various kidney diseases.
Collapse
Affiliation(s)
- Kumaresan Ramanathan
- Department of Biomedical Sciences, Institute of Health, Faculty of Medical Sciences, Jimma University, Jimma, Ethiopia
| | | | - Henok Gulilat
- Department of Biomedical Sciences, Institute of Health, Faculty of Medical Sciences, Jimma University, Jimma, Ethiopia
| | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Faculty of Medical Sciences, Jimma University, Jimma, Ethiopia
| |
Collapse
|
34
|
Qian J, Yang M, Xu D, Zhang G, Cai Y, Yang B, Wang X, Yu Y. Alterations of the salivary microbiota in gastroesophageal reflux disease. J Oral Biosci 2023; 65:280-286. [PMID: 37595742 DOI: 10.1016/j.job.2023.08.005] [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: 03/13/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 08/20/2023]
Abstract
OBJECTIVES Gastroesophageal reflux disease (GERD) is among the most prevalent gastrointestinal disorders. The oral microbiota plays an important role in human health and may be altered by the presence of GERD. Here, we aimed to investigate the alterations of salivary microbiota in GERD patients. METHODS We collected clinical information and salivary samples from 60 individuals. All participants underwent combined pH/impedance monitoring measurement and submitted samples for salivary microbiota sequencing. According to acid exposure time and DeMeester score, participants were divided into two groups: GERD + (Group G) and GERD - (Group C). RESULTS There was no significant difference in alpha diversity between study groups. Regarding beta diversity, principal coordinate analysis plots indicated that the microbiota composition data of the participants were grouped within partial overlapping clusters. The statistical analysis of the distance matrices was performed using the Adonis test (p = 0.017). Based on linear discriminant analysis effect size, the relative abundances of the phylum Bacteroidetes, class Bacteroidia, order Bacteroidales, family Prevotellaceae, and genus unidentified_Prevotellaceae were enriched in Group G. Compared with Group C, the phylum Actinobacteria, classes unidentified_Actinobacteria and Bacilli, orders Micrococcales and Lactobacillales, families Micrococcaceae and Streptococcaceae, and genuses Rothia and Streptococcus were decreased in Group G. At the genus level, the abundances of Streptococcus and Rothia were negatively correlated with DeMeester score and acid exposure time. CONCLUSIONS This study revealed alterations of the salivary microbiota in GERD patients, suggesting that acid reflux changes the oral ecosystem.
Collapse
Affiliation(s)
- Jun Qian
- Department of Colorectal Surgery, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Meilin Yang
- Department of Gastroenterology, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Duiyue Xu
- Department of Gastroenterology, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Gaosong Zhang
- Department of Gastroenterology, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Youhong Cai
- Department of Geriatrics, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Bin Yang
- Department of Gastroenterology, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China
| | - Xiying Wang
- Department of Geriatrics, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China.
| | - Yanbo Yu
- Department of Geriatrics, Affiliated Xinchang Hospital, Wenzhou Medical University, 312500, Shaoxing, Zhejiang, China.
| |
Collapse
|
35
|
Lê S, Cecchin-Albertoni C, Thomas C, Kemoun P, Minty M, Blasco-Baque V. The Role of Dysbiotic Oral Microbiota in Cardiometabolic Diseases: A Narrative Review. Diagnostics (Basel) 2023; 13:3184. [PMID: 37892006 PMCID: PMC10605832 DOI: 10.3390/diagnostics13203184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/25/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Over the past decade, there have been significant advancements in the high-flow analysis of "omics," shedding light on the relationship between the microbiota and the host. However, the full recognition of this relationship and its implications in cardiometabolic diseases are still underway, despite advancements in understanding the pathophysiology of these conditions. Cardiometabolic diseases, which include a range of conditions from insulin resistance to cardiovascular disease and type 2 diabetes, continue to be the leading cause of mortality worldwide, with a persistently high morbidity rate. While the link between the intestinal microbiota and cardiometabolic risks has been extensively explored, the role of the oral microbiota, the second-largest microbiota in the human body, and specifically the dysbiosis of this microbiota in causing these complications, remains incompletely defined. This review aims to examine the association between the oral microbiota and cardiometabolic diseases, focusing on the dysbiosis of the oral microbiota, particularly in periodontal disease. Additionally, we will dive into the mechanistic aspects of this dysbiosis that contribute to the development of these complications. Finally, we will discuss potential prevention and treatment strategies, including the use of prebiotics, probiotics, and other interventions.
Collapse
Affiliation(s)
- Sylvie Lê
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Chiara Cecchin-Albertoni
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- RESTORE Research Center, CNRS, EFS, ENVT, Batiment INCERE, INSERM, Université de Toulouse, 4 bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Charlotte Thomas
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Philippe Kemoun
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- RESTORE Research Center, CNRS, EFS, ENVT, Batiment INCERE, INSERM, Université de Toulouse, 4 bis Avenue Hubert Curien, 31100 Toulouse, France
| | - Matthieu Minty
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| | - Vincent Blasco-Baque
- Département Dentaire, Université Paul Sabatier III (UPS), 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France; (S.L.); (C.C.-A.); (C.T.); (P.K.); (M.M.)
- Service d’Odontologie Toulouse, CHU Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- Team InCOMM/Intestine ClinicOmics Metabolism & Microbiota, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC)—UMR1297 Inserm, Université Paul Sabatier, 1 Avenue Jean Poulhes, 31432 Toulouse, France
| |
Collapse
|
36
|
Koerner R, Prescott S, McSkimming D, Alman A, Duffy A, Groer M. The Salivary Microbiome During Pregnancy: Associations With Clinical and Sociodemographic Characteristics. J Perinat Neonatal Nurs 2023; 37:287-294. [PMID: 37878513 DOI: 10.1097/jpn.0000000000000715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
PURPOSE Poor oral health has been associated with adverse pregnancy outcomes, and the oral microbiome may play a role in these mechanisms. We aimed to examine the salivary microbiome for alterations in diversity or relative abundance throughout pregnancy and its associations with adverse pregnancy outcomes and sociodemographic characteristics. STUDY DESIGN AND METHODS We conducted an ancillary study from a previous cohort study of 37 women during their second and third trimesters of pregnancy using preexisting, participant-collected salivary samples to examine the oral microbiome using 16S rRNA sequencing. RESULTS The salivary microbiome demonstrated stability throughout pregnancy, as there were no significant differences in alpha or beta diversity. Individuals who were diagnosed with preeclampsia had differences in beta diversity at the genus level (F = 2.65, df = 1, P = .015). There were also differences in beta diversity at the species level in Hispanic individuals compared with non-Hispanic individuals (F = 1.7183, df = 1, P = .04). CONCLUSION The salivary microbiome demonstrated stability throughout the second and third trimesters but may be different in Hispanics or those diagnosed with preeclampsia. As such, clinical providers need to demonstrate culturally competent care during pregnancy and continue to educate women about the importance of oral healthcare during the perinatal period. Future research is needed to examine the mechanisms associated with oral microbiome dysbiosis in Hispanic women during pregnancy and in women with preeclampsia.
Collapse
Affiliation(s)
- Rebecca Koerner
- University of South Florida College of Nursing, Tampa, Florida (Drs Koerner, Prescott, Duffy, and Groer); State University of New York at Buffalo State, Buffalo (Dr McSkimming); and University of South Florida College of Public Health, Tampa (Dr Alman)
| | | | | | | | | | | |
Collapse
|
37
|
Siddiqui R, Badran Z, Boghossian A, Alharbi AM, Alfahemi H, Khan NA. The increasing importance of the oral microbiome in periodontal health and disease. Future Sci OA 2023; 9:FSO856. [PMID: 37621848 PMCID: PMC10445586 DOI: 10.2144/fsoa-2023-0062] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/30/2023] [Indexed: 08/26/2023] Open
Abstract
Herein, the aim is to discuss the current knowledge of microbiome and periodontal diseases. Current treatment strategies include mechanical therapy such as root planing, scaling, deep pocket debridement and antimicrobial chemotherapy as an adjuvant therapy. Among promising therapeutic strategies, dental probiotics and oral microbiome transplantation have gained attention, and may be used to treat bacterial imbalances by competing with pathogenic bacteria for nutrients and adhesion surfaces, as well as probiotics targeting the gut microbiome. Development of strategies to prevent and treat periodontal diseases are warranted as both are highly prevalent and can affect human health. Further studies are necessary to better comprehend the microbiome in order to develop innovative preventative measures as well as efficacious therapies against periodontal diseases.
Collapse
Affiliation(s)
- Ruqaiyyah Siddiqui
- College of Arts & Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
- Department of Medical Biology, Faculty of Medicine, Istinye University, Istanbul, 34010, Turkey
| | - Zahi Badran
- Periodontology Unit, Department of Preventive & Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Anania Boghossian
- College of Arts & Sciences, American University of Sharjah, Sharjah, 26666, United Arab Emirates
| | - Ahmad M Alharbi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, 21944, Saudi Arabia
| | - Hasan Alfahemi
- Department of Medical Microbiology, Faculty of Medicine, Al-Baha University, Al-Baha, 65799, Saudi Arabia
| | - Naveed Ahmed Khan
- Department of Medical Biology, Faculty of Medicine, Istinye University, Istanbul, 34010, Turkey
- Department of Clinical Sciences, College of Medicine, University of Sharjah, University City, Sharjah, United Arab Emirates
| |
Collapse
|
38
|
Araújo V, Fehn AM, Phiri A, Wills J, Rocha J, Gayà-Vidal M. Oral microbiome homogeneity across diverse human groups from southern Africa: first results from southwestern Angola and Zimbabwe. BMC Microbiol 2023; 23:226. [PMID: 37596536 PMCID: PMC10436416 DOI: 10.1186/s12866-023-02970-2] [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: 03/31/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND While the human oral microbiome is known to play an important role in systemic health, its average composition and diversity patterns are still poorly understood. To gain better insights into the general composition of the microbiome on a global scale, the characterization of microbiomes from a broad range of populations, including non-industrialized societies, is needed. Here, we used the portion of non-human reads obtained through an expanded exome capture sequencing approach to characterize the saliva microbiomes of 52 individuals from eight ethnolinguistically diverse southern African populations from Angola (Kuvale, Kwepe, Himba, Tjimba, Kwisi, Twa, !Xun) and Zimbabwe (Tshwa), including foragers, food-producers, and peripatetic groups (low-status communities who provide services to their dominant neighbors). RESULTS Our results indicate that neither host genetics nor livelihood seem to influence the oral microbiome profile, with Neisseria, Streptococcus, Prevotella, Rothia, and Porphyromonas being the five most frequent genera in southern African groups, in line with what has been shown for other human populations. However, we found that some Tshwa and Twa individuals display an enrichment of pathogenic genera from the Enterobacteriaceae family (i.e. Enterobacter, Citrobacter, Salmonella) of the Proteobacteria phylum, probably reflecting deficient sanitation and poor health conditions associated with social marginalization. CONCLUSIONS Taken together, our results suggest that socio-economic status, rather than ethnolinguistic affiliation or subsistence mode, is a key factor in shaping the salivary microbial profiles of human populations in southern Africa.
Collapse
Affiliation(s)
- Vítor Araújo
- Centro de Investigação em Biodiversidade e Recursos Genéticos, CIBIO, InBIO Laboratório Associado, Universidade do Porto, Campus de Vairão, Vairão, 4485-661, Portugal
- Program in Genomics, Biodiversity and Land Planning, CIBIO, BIOPOLIS, Campus de Vairão, Vairão, 4485-661, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, 4169-007, Portugal
| | - Anne-Maria Fehn
- Centro de Investigação em Biodiversidade e Recursos Genéticos, CIBIO, InBIO Laboratório Associado, Universidade do Porto, Campus de Vairão, Vairão, 4485-661, Portugal
- Program in Genomics, Biodiversity and Land Planning, CIBIO, BIOPOLIS, Campus de Vairão, Vairão, 4485-661, Portugal
| | - Admire Phiri
- Department of Linguistics and Language Practice, University of Free State, Bloemfontein, South Africa
| | | | - Jorge Rocha
- Centro de Investigação em Biodiversidade e Recursos Genéticos, CIBIO, InBIO Laboratório Associado, Universidade do Porto, Campus de Vairão, Vairão, 4485-661, Portugal
- Program in Genomics, Biodiversity and Land Planning, CIBIO, BIOPOLIS, Campus de Vairão, Vairão, 4485-661, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, 4169-007, Portugal
| | - Magdalena Gayà-Vidal
- Centro de Investigação em Biodiversidade e Recursos Genéticos, CIBIO, InBIO Laboratório Associado, Universidade do Porto, Campus de Vairão, Vairão, 4485-661, Portugal.
- Program in Genomics, Biodiversity and Land Planning, CIBIO, BIOPOLIS, Campus de Vairão, Vairão, 4485-661, Portugal.
| |
Collapse
|
39
|
Makkena VK, Jaramillo AP, Awosusi BL, Ayyub J, Dabhi KN, Gohil NV, Tanveer N, Hussein S, Pingili S, Khan S. Probing the Relationship Between the Human Gut Microbiome and Prospects of Prostate Cancer: A Systematic Review. Cureus 2023; 15:e43892. [PMID: 37746426 PMCID: PMC10511825 DOI: 10.7759/cureus.43892] [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: 07/24/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Prostate neoplasia is one of the most commonly occurring neoplasias in males and has a high mortality rate. Prostate cancer (PCA) risk factors include tall stature, male sex, known family history, obesity, high blood pressure, lack of fitness, higher levels of testosterone for a long time, increasing age, and ethnicity are well known. The association and role of the gut microbiota in different diseases in our body have been highlighted recently. Therefore, finding the influence of gut microbiota on the prostatic cells can be useful for preventing prostatic neoplasia and/or reducing its severity. We aimed to assess its impact on PCA risk. We thoroughly searched databases for the relevant literature for our systematic review. The final research papers analyzed how bacteria played a role in the risk of PCA, either through inflammation or the production of metabolites that increase/decrease the risk of PCA. Based on the studies reviewed, we found that some gut bacteria play a role in the formation of PCA. In contrast, some bacteria can help prevent PCA, but the metabolism of the dietary components is the major factor for PCA.
Collapse
Affiliation(s)
- Vijaya Krishna Makkena
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Medicine, Osmania Medical College, Hyderabad, IND
| | - Arturo P Jaramillo
- Department of Internal Medicine, Universidad Estatal de Guayaquil, Machala, ECU
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Babatope L Awosusi
- Department of Pathology and Laboratory Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Javaria Ayyub
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Karan Nareshbha Dabhi
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Namra V Gohil
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Internal Medicine, Medical College Baroda, Vadodara, IND
| | - Nida Tanveer
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Sally Hussein
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Shravya Pingili
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Department of Medicine, Kakatiya Medical College, Hyderabad, IND
| | - Safeera Khan
- Department of Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
40
|
Ribeiro ASP, Marquezin MCS, Pacheco ERP, Rasera I, Klein MI, de Vasconcellos SP, Landgraf RG, Okamoto D, Calixto LA, Castelo PM. Bypass gastroplasty impacts oral health, salivary inflammatory biomarkers, and microbiota: a controlled study. Clin Oral Investig 2023; 27:4735-4746. [PMID: 37294353 DOI: 10.1007/s00784-023-05101-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/29/2023] [Indexed: 06/10/2023]
Abstract
OBJECTIVES Knowledge about the impact of gastroplasty on oral health and salivary biomarkers is limited. The aim was to prospectively evaluate oral health status, salivary inflammatory markers, and microbiota in patients undergoing gastroplasty compared with a control group undergoing a dietary program. MATERIALS AND METHODS Forty participants with obesity class II/III were included (20 individuals in each sex-matched group; 23-44 years). Dental status, salivary flow, buffering capacity, inflammatory cytokines, and uric acid were assessed. Salivary microbiological analysis (16S-rRNA sequencing) assessed the abundance of genus, species, and alpha diversity. Cluster analysis and mixed-model ANOVA were applied. RESULTS Oral health status, waist-to-hip ratio, and salivary alpha diversity were associated at baseline. A subtle improvement in food consumption markers was observed, although caries activity increased in both groups, and the gastroplasty group showed worse periodontal status after three months. IFNγ and IL10 levels decreased in the gastroplasty group at 3 months, while a decrease was observed in the control group at 6 months; IL6 decreased in both groups (p < 0.001). Salivary flow and buffering capacity did not change. Significant changes in Prevotella nigrescens and Porphyromonas endodontalis abundance were observed in both groups, while alpha diversity (Sobs, Chao1, Ace, Shannon, and Simpson) increased in the gastroplasty group. CONCLUSIONS Both interventions changed in different degrees the salivary inflammatory biomarkers and microbiota, but did not improve the periodontal status after 6 months. CLINICAL RELEVANCE Although the observed discrete improvement in dietary habits, caries activity increased with no clinical improvement in the periodontal status, emphasizing the need of oral health monitoring during obesity treatment.
Collapse
Affiliation(s)
- Aianne Souto Pizzolato Ribeiro
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | - Maria Carolina Salomé Marquezin
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | | | - Irineu Rasera
- Faculdade de Ensino Superior da Amazônia Reunida, Av. Brasil, 1435, Redenção, Brazil
| | - Marlise Inês Klein
- Faculdade de Odontologia de Piracicaba, Universidade de Campinas, Av. Limeira, 901, Piracicaba, Brazil
| | - Suzan Pantaroto de Vasconcellos
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | - Richardt Gama Landgraf
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | - Debora Okamoto
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | - Leandro Augusto Calixto
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil
| | - Paula Midori Castelo
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), R. São Nicolau, 210 - 1. Andar, Diadema, SP, Brazil.
| |
Collapse
|
41
|
Basilicata M, Pieri M, Marrone G, Nicolai E, Di Lauro M, Paolino V, Tomassetti F, Vivarini I, Bollero P, Bernardini S, Noce A. Saliva as Biomarker for Oral and Chronic Degenerative Non-Communicable Diseases. Metabolites 2023; 13:889. [PMID: 37623833 PMCID: PMC10456419 DOI: 10.3390/metabo13080889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
Saliva is a very complex fluid and it is essential to maintain several physiological processes and functions, including oral health, taste, digestion and immunological defenses. Saliva composition and the oral microbiome can be influenced by several factors, like diet and smoking habits, and their alteration can represent an important access point for pathogens and, thus, for systemic illness onset. In this review, we explore the potentiality of saliva as a new tool for the early detection of some pathological conditions, such as oral diseases, chronic degenerative non-communicable diseases, among these chronic kidney disease (CKD). We also examined the possible correlation between oral and systemic diseases and oral and gut microbiota dysbiosis. In particular, we deeply analyzed the relationship between oral diseases and CKD. In this context, some salivary parameters can represent a new device to detect either oral or systemic pathologies. Moreover, the positive modulation of oral and gut microbiota induced by prebiotics, postbiotics, or symbiotics could represent a new possible adjuvant therapy in the clinical management of oral diseases and CKD.
Collapse
Affiliation(s)
- Michele Basilicata
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Giulia Marrone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Nicolai
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Manuela Di Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Vincenza Paolino
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Flaminia Tomassetti
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Vivarini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Patrizio Bollero
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Noce
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- UOSD Nephrology and Dialysis, Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
42
|
Dass M, Singh Y, Ghai M. A Review on Microbial Species for Forensic Body Fluid Identification in Healthy and Diseased Humans. Curr Microbiol 2023; 80:299. [PMID: 37491404 PMCID: PMC10368579 DOI: 10.1007/s00284-023-03413-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/08/2023] [Indexed: 07/27/2023]
Abstract
Microbial communities present in body fluids can assist in distinguishing between types of body fluids. Metagenomic studies have reported bacterial genera which are core to specific body fluids and are greatly influenced by geographical location and ethnicity. Bacteria in body fluids could also be due to bacterial infection; hence, it would be worthwhile taking into consideration bacterial species associated with diseases. The present review reports bacterial species characteristic of diseased and healthy body fluids across geographical locations, and bacteria described in forensic studies, with the aim of collating a set of bacteria to serve as the core species-specific markers for forensic body fluid identification. The most widely reported saliva-specific bacterial species are Streptococcus salivarius, Prevotella melaninogenica, Neisseria flavescens, with Fusobacterium nucleatum associated with increased diseased state. Lactobacillus crispatus and Lactobacillus iners are frequently dominant in the vaginal microbiome of healthy women. Atopobium vaginae, Prevotella bivia, and Gardnerella vaginalis are more prevalent in women with bacterial vaginosis. Semen and urine-specific bacteria at species level have not been reported, and menstrual blood bacteria are indistinguishable from vaginal fluid. Targeting more than one bacterial species is recommended for accurate body fluid identification. Although metagenomic sequencing provides information of a broad microbial profile, the specific bacterial species could be used to design biosensors for rapid body fluid identification. Validation of microbial typing methods and its application in identifying body fluids in a mixed sample would allow regular use of microbial profiling in a forensic workflow.
Collapse
Affiliation(s)
- Mishka Dass
- Department of Genetics, School of Life Sciences, University of KwaZulu Natal, Westville Campus, Private Bag X 54001, Durban, KwaZulu-Natal South Africa
| | - Yashna Singh
- Department of Genetics, School of Life Sciences, University of KwaZulu Natal, Westville Campus, Private Bag X 54001, Durban, KwaZulu-Natal South Africa
| | - Meenu Ghai
- Department of Genetics, School of Life Sciences, University of KwaZulu Natal, Westville Campus, Private Bag X 54001, Durban, KwaZulu-Natal South Africa
| |
Collapse
|
43
|
Shen MT, Shahin B, Chen Z, Adami GR. Unexpected lower level of oral periodontal pathogens in patients with high numbers of systemic diseases. PeerJ 2023; 11:e15502. [PMID: 37465146 PMCID: PMC10351517 DOI: 10.7717/peerj.15502] [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: 01/18/2023] [Accepted: 05/14/2023] [Indexed: 07/20/2023] Open
Abstract
Background Periodontal disease is associated with systemic conditions such as diabetes, arthritis, and cardiovascular disease, all diseases with large inflammatory components. Some, but not all, reports show periopathogens Porphyromonas gingivialis and Tannerella forsythia at higher levels orally in people with one of these chronic diseases and in people with more severe cases. These oral pathogens are thought to be positively associated with systemic inflammatory diseases through induction of oral inflammation that works to distort systemic inflammation or by directly inducing inflammation at distal sites in the body. This study aimed to determine if, among patients with severe periodontal disease, those with multi-morbidity (or many chronic diseases) showed higher levels of periodontal pathogens. Methods A total of 201 adult subjects, including 84 with severe periodontal disease were recruited between 1/2017 and 6/2019 at a city dental clinic. Electronic charts supplied self-reported diseases and conditions which informed a morbidity index based on the number of chronic diseases and conditions present. Salivary composition was determined by 16S rRNA gene sequencing. Results As expected, patients with severe periodontal disease showed higher levels of periodontal pathogens in their saliva. Also, those with severe periodontal disease showed higher levels of multiple chronic diseases (multimorbidity). An examination of the 84 patients with severe periodontal disease revealed some subjects despite being of advanced age were free or nearly free of systemic disease. Surprisingly, the salivary microbiota of the least healthy of these 84 subjects, defined here as those with maximal multimorbidity, showed significantly lower relative numbers of periodontal pathogens, including Porphyromonas gingivalis and Tannerella Forsythia, after controlling for active caries, tobacco usage, age, and gender. Analysis of a control group with none to moderate periodontal disease revealed no association of multimorbidity or numbers of medications used and specific oral bacteria, indicating the importance of severe periodontal disease as a variable of interest. Conclusion The hypothesis that periodontal disease patients with higher levels of multimorbidity would have higher levels of oral periodontal pathogens is false. Multimorbidity is associated with a reduced relative number of periodontal pathogens Porphyromonas gingivalis and Tannerella forsythia.
Collapse
Affiliation(s)
- Michael T Shen
- Oral Medicine and Diagnostic Sciences, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States of America
| | - Betti Shahin
- Restorative Dentistry, University of Illinois Chicago, Chicago, IL, United States of America
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois at Chicago, Chicago, IL, United States of America
- Biostatistics Shared Resource Core, University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL, United States of America
| | - Guy R Adami
- Oral Medicine and Diagnostic Sciences, College of Dentistry, University of Illinois Chicago, Chicago, IL, United States of America
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL, United States of America
| |
Collapse
|
44
|
Murugesan S, Al Khodor S. Salivary microbiome and hypertension in the Qatari population. J Transl Med 2023; 21:454. [PMID: 37422685 PMCID: PMC10329805 DOI: 10.1186/s12967-023-04247-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND The prevalence of hypertension in Qatar is 33 percent of the adult population. It is postulated that the salivary microbiome can regulate blood pressure (BP). However, limited investigations exist to prove this hypothesis. Therefore, we examined the difference in the salivary microbiome composition between hypertensive and normotensive Qatari subjects. METHODS A total of 1190 Qatar Genome Project (QGP) participants (Mean age = 43 years) were included in this study. BP for all participants was classified into Normal (n = 357), Stage1 (n = 336), and Stage2: (n = 161) according to the American Heart Association guidelines. 16S-rRNA libraries were sequenced and analyzed using QIIME-pipeline, and PICRUST was used to predict functional metabolic routes. Machine Learning (ML) strategies were applied to identify salivary microbiome-based predictors of hypertension. RESULTS Differential abundant analysis (DAA) revealed that Bacteroides and Atopobium were the significant members of the hypertensive groups. Alpha and beta diversity indices indicated dysbiosis between the normotensive and hypertensive groups. ML-based prediction models revealed that these markers could predict hypertension with an AUC (Area under the curve) of 0.89. Functional predictive analysis disclosed that Cysteine and Methionine metabolism and the sulphur metabolic pathways involving the renin-angiotensin system were significantly higher in the normotensive group. Therefore, members of Bacteroides and Atopobium can serve as predictors of hypertension. Likewise, Prevotella, Neisseria, and Haemophilus can be the protectors that regulate BP via nitric acid synthesis and regulation of the renin-angiotensin system. CONCLUSION It is one of the first studies to assess salivary microbiome and hypertension as disease models in a large cohort of the Qatari population. Further research is needed to confirm these findings and validate the mechanisms involved.
Collapse
Affiliation(s)
- Selvasankar Murugesan
- Maternal and Child Health Division, Research Department, Sidra Medicine, 26999, Doha, Qatar
| | - Souhaila Al Khodor
- Maternal and Child Health Division, Research Department, Sidra Medicine, 26999, Doha, Qatar.
| |
Collapse
|
45
|
Ma T, Wu Z, Lin J, Shan C, Abasijiang A, Zhao J. Characterization of the oral and gut microbiome in children with obesity aged 3 to 5 years. Front Cell Infect Microbiol 2023; 13:1102650. [PMID: 37065198 PMCID: PMC10090557 DOI: 10.3389/fcimb.2023.1102650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
The ever-increasing global prevalence of obesity has trended towards a younger age. The ecological characteristics and changes of the oral and gut microbial community during childhood are poorly understood.In this study, we analyzed the salivary and fecal microbiota of 30 children with obesity and 30 normal weight children aged 3-5 years via third-generation long-range DNA sequencing,with the aim of understanding the structure of childhood microbiota and identifying specific oral and gut microbial lineages and genera in children that may be associated with obesity.The results revealed significant variation in alpha diversity indices among the four groups (Chao1: P < 0.001; observed species: P < 0.001; Shannon < 0.001). Principal coordinate analysis (PCoA) and nonmetric multidimensional scaling (NMDS) revealed significant differences in oral and gut microbial community structure between obesity and controls. The Firmicutes/Bacteroidetes (F/B) abundance ratios of oral and intestinal flora among children with obesity were higher than those of controls. The most abundant phyla and genera found in oral and intestinal flora were Firmicutes, Proteobacteria, Bacteroidetes, Neisseria, Bacteroides, Faecalibacterium, Streptococcus, Prevotella and so on. Linear discriminant analysis effect size (LEfSe) revealed higher proportions of Filifactor (LDA= 3.98; P < 0.05) and Butyrivibrio (LDA = 2.54; P < 0.001) in the oral microbiota of children with obesity, while the fecal microbiota of children with obesity were more enriched with Faecalibacterium (LDA = 5.02; P < 0.001), Tyzzerella (LDA=3.25; P < 0.01), Klebsiella (LDA = 4.31; P < 0.05),which could be considered as dominant bacterial biomarkers for obesity groups.A total of 148 functional bacterial pathways were found to significantly differ in the oral and gut microbiota among controls and obesity using PICRUSt 2. Most predicted functional pathways were clustered in biosynthesis. In conclusion, This work suggests there were significant differences in oral and gut microbiota in controls and obesity groups, microbiota dysbiosis in childhood might have significant effect on the development of obesity.
Collapse
Affiliation(s)
- Ting Ma
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
| | - Zeyu Wu
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
| | - Jing Lin
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
| | - Chao Shan
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
| | - Aisaiti Abasijiang
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
| | - Jin Zhao
- Department of Cariology and Endodontics, The First Affiliated Hospital of Xinjiang Medical University, The Affiliated Stomatology Hospital of Xinjiang Medical University, Urumqi, China
- Stomatology Disease Institute of Xinjiang Uyghur Autonomous Region, Xinjiang Medical University, Urumqi, China
- *Correspondence: Jin Zhao,
| |
Collapse
|
46
|
Saliva Metabolomic Profile in Dental Medicine Research: A Narrative Review. Metabolites 2023; 13:metabo13030379. [PMID: 36984819 PMCID: PMC10052075 DOI: 10.3390/metabo13030379] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Metabolomic research tends to increase in popularity over the years, leading to the identification of new biomarkers related to specific health disorders. Saliva is one of the most newly introduced and systematically developed biofluids in the human body that can serve as an informative substance in the metabolomic profiling armamentarium. This review aims to analyze the current knowledge regarding the human salivary metabolome, its alterations due to physiological, environmental and external factors, as well as the limitations and drawbacks presented in the most recent research conducted, focusing on pre—analytical and analytical workflows. Furthermore, the use of the saliva metabolomic profile as a promising biomarker for several oral pathologies, such as oral cancer and periodontitis will be investigated.
Collapse
|
47
|
Younginger BS, Mayba O, Reeder J, Nagarkar DR, Modrusan Z, Albert ML, Byrd AL. Enrichment of oral-derived bacteria in inflamed colorectal tumors and distinct associations of Fusobacterium in the mesenchymal subtype. Cell Rep Med 2023; 4:100920. [PMID: 36706753 PMCID: PMC9975273 DOI: 10.1016/j.xcrm.2023.100920] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/22/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023]
Abstract
While the association between colorectal cancer (CRC) features and Fusobacterium has been extensively studied, less is known of other intratumoral bacteria. Here, we leverage whole transcriptomes from 807 CRC samples to dually characterize tumor gene expression and 74 intratumoral bacteria. Seventeen of these species, including 4 Fusobacterium spp., are classified as orally derived and are enriched among right-sided, microsatellite instability-high (MSI-H), and BRAF-mutant tumors. Across consensus molecular subtypes (CMSs), integration of Fusobacterium animalis (Fa) presence and tumor expression reveals that Fa has the most significant associations in mesenchymal CMS4 tumors despite a lower prevalence than in immune CMS1. Within CMS4, the prevalence of Fa is uniquely associated with collagen- and immune-related pathways. Additional Fa pangenome analysis reveals that stress response genes and the adhesion FadA are commonly expressed intratumorally. Overall, this study identifies oral-derived bacteria as enriched in inflamed tumors, and the associations of bacteria and tumor expression are context and species specific.
Collapse
Affiliation(s)
- Brett S Younginger
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Oleg Mayba
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA, USA
| | - Jens Reeder
- Department of Oncology Bioinformatics, Genentech, Inc., South San Francisco, CA, USA
| | - Deepti R Nagarkar
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA
| | - Zora Modrusan
- Microchemistry, Proteomics, Lipidomics and Next Generation Sequencing, Genentech, Inc., South San Francisco, CA, USA
| | | | - Allyson L Byrd
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
48
|
Fungal composition in saliva and plaque in children with caries: Differences and influencing factors. MEDICINE IN MICROECOLOGY 2023. [DOI: 10.1016/j.medmic.2023.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
|
49
|
Wang M, Yan LY, Qiao CY, Zheng CC, Niu CG, Huang ZW, Pan YH. Ecological shifts of salivary microbiota associated with metabolic-associated fatty liver disease. Front Cell Infect Microbiol 2023; 13:1131255. [PMID: 36864882 PMCID: PMC9971218 DOI: 10.3389/fcimb.2023.1131255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease related to metabolic syndrome. However, ecological shifts in the saliva microbiome in patients with MAFLD remain unknown. This study aimed to investigate the changes to the salivary microbial community in patients with MAFLD and explore the potential function of microbiota. Methods Salivary microbiomes from ten MAFLD patients and ten healthy participants were analyzed by 16S rRNA amplicon sequencing and bioinformatics analysis. Body composition, plasma enzymes, hormones, and blood lipid profiles were assessed with physical examinations and laboratory tests. Results The salivary microbiome of MAFLD patients was characterized by increased α-diversity and distinct β-diversity clustering compared with control subjects. Linear discriminant analysis effect size analysis showed a total of 44 taxa significantly differed between the two groups. Genera Neisseria, Filifactor, and Capnocytophaga were identified as differentially enriched genera for comparison of the two groups. Co-occurrence networks suggested that the salivary microbiota from MAFLD patients exhibited more intricate and robust interrelationships. The diagnostic model based on the salivary microbiome achieved a good diagnostic power with an area under the curve of 0.82(95% CI: 0.61-1). Redundancy analysis and spearman correlation analysis revealed that clinical variables related to insulin resistance and obesity were strongly associated with the microbial community. Metagenomic predictions based on Phylogenetic Investigation of Communities by Reconstruction of Unobserved States revealed that pathways related to metabolism were more prevalent in the two groups. Conclusions Patients with MAFLD manifested ecological shifts in the salivary microbiome, and the saliva microbiome-based diagnostic model provides a promising approach for auxiliary MAFLD diagnosis.
Collapse
Affiliation(s)
- Min Wang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Li-Ya Yan
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Cai-Yun Qiao
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chu-Chu Zheng
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Chen-Guang Niu
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zheng-Wei Huang
- Department of Endodontics, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Oral Diseases, National Center for Stomatology, Shanghai, China
- Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
- *Correspondence: Zheng-Wei Huang, ; Yi-Huai Pan,
| | - Yi-Huai Pan
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zheng-Wei Huang, ; Yi-Huai Pan,
| |
Collapse
|
50
|
Sex differences in the oral microbiome, host traits, and their causal relationships. iScience 2022; 26:105839. [PMID: 36660475 PMCID: PMC9843272 DOI: 10.1016/j.isci.2022.105839] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/09/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
The oral microbiome has been implicated in a growing number of diseases; however, determinants of the oral microbiome and their roles remain elusive. Here, we investigated the oral (saliva and tongue dorsum) metagenome, the whole genome, and other omics data in a total of 4,478 individuals and demonstrated that the oral microbiome composition and its major contributing host factors significantly differed between sexes. We thus conducted a sex-stratified metagenome-genome-wide-association study (M-GWAS) and identified 11 differential genetic associations with the oral microbiome (p sex-difference < 5 × 10-8). Furthermore, we performed sex-stratified Mendelian randomization (MR) analyses and identified abundant causalities between the oral microbiome and serum metabolites. Notably, sex-specific microbes-hormonal interactions explained the mostly observed sex hormones differences such as the significant causalities enrichments for aldosterone in females and androstenedione in males. These findings illustrate the necessity of sex stratification and deepen our understanding of the interplay between the oral microbiome and serum metabolites.
Collapse
|