1
|
Eitel E, Utter D, Connon S, Orphan V, Murali R. CABO-16S-a Combined Archaea, Bacteria, Organelle 16S rRNA database framework for amplicon analysis of prokaryotes and eukaryotes in environmental samples. NAR Genom Bioinform 2025; 7:lqaf061. [PMID: 40391087 PMCID: PMC12086536 DOI: 10.1093/nargab/lqaf061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/22/2025] [Accepted: 05/11/2025] [Indexed: 05/21/2025] Open
Abstract
Identification of both prokaryotic and eukaryotic microorganisms in environmental samples is currently challenged by the need for additional sequencing to obtain separate 16S and 18S ribosomal RNA (rRNA) amplicons or the constraints imposed by "universal" primers. Organellar 16S rRNA sequences are amplified and sequenced along with prokaryote 16S rRNA and provide an alternative method to identify eukaryotic microorganisms. CABO-16S combines bacterial and archaeal sequences from the SILVA database with 16S rRNA sequences of plastids and other organelles from the PR2 database to enable identification of all 16S rRNA sequences. Comparison of CABO-16S with SILVA 138.2 results in equivalent taxonomic classification of mock communities and increased classification of diverse environmental samples. In particular, identification of phototrophic eukaryotes in shallow seagrass environments, marine waters, and lake waters was increased. The CABO-16S framework allows users to add custom sequences for further classification of underrepresented clades and can be easily updated with future releases of reference databases. Addition of sequences obtained from Sanger sequencing of methane seep sediments and curated sequences of the polyphyletic SEEP-SRB1 clade resulted in differentiation of syntrophic and non-syntrophic SEEP-SRB1 in hydrothermal vent sediments. CABO-16S highlights the benefit of combining and amending existing training sets when studying microorganisms in diverse environments.
Collapse
Affiliation(s)
- Eryn M Eitel
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125, United States
| | - Daniel R Utter
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Stephanie A Connon
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125, United States
| | - Victoria J Orphan
- Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Ranjani Murali
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- School of Life Sciences, University of Nevada, Las Vegas, NV 89154, United States
| |
Collapse
|
2
|
Zhang Y, Ji Y, Liu F, Wang Y, Feng C, Zhou Z, Zhang Z, Han L, Li J, Wang M, Li L. Pseudomonas sp. Strain ADAl3-4 Enhances Aluminum Tolerance in Alfalfa ( Medicago sativa). Int J Mol Sci 2025; 26:4919. [PMID: 40430057 PMCID: PMC12111897 DOI: 10.3390/ijms26104919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/18/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Aluminum toxicity severely inhibits root elongation and nutrient uptake, causing global agricultural yield losses. Dissolved Al3+ are accumulating in plants and subsequently entering food chains via crops and forage plants. Chronic dietary exposure to Al3+ poses a risk to human health. In this study, Pseudomonas sp. strain ADAl3-4, isolated from plant rhizosphere soil, significantly enhanced plant development and biomass. Phenotypic validation using Arabidopsis mutants showed that strain ADAl3-4 regulates plant growth and development under aluminum stress by reprogramming the cell cycle, regulating auxin and ion homeostasis, and enhancing the root absorption of Al3+ from the soil. Transcriptomic and biochemical analyses showed that strain ADAl3-4 promotes plant growth via regulating signal transduction, phytohormone biosynthesis, flavonoid biosynthesis, and antioxidant capacity, etc., under aluminum stress. Our findings indicate that Pseudomonas sp. strain ADAl3-4 enhances plant development and stress resilience under Al3+ toxicity through a coordinated multi-dimensional regulatory network. Furthermore, strain ADAl3-4 promoted the root absorption of aluminum rather than the transportation of Al to the aerial part, endowing it with application prospects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lixin Li
- Key Laboratory of Saline–Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Sciences, Northeast Forestry University, Harbin 150040, China; (Y.Z.); (Y.J.); (F.L.); (Y.W.); (C.F.); (Z.Z.); (Z.Z.); (L.H.); (J.L.); (M.W.)
| |
Collapse
|
3
|
von Ameln Lovison O, Zempulski Volpato FC, Weber LG, Barth AL, Simon Coitinho A, Martins AF. Unveiling the role of the upper respiratory tract microbiome in susceptibility and severity to COVID-19. Front Cell Infect Microbiol 2025; 15:1531084. [PMID: 40433668 PMCID: PMC12106449 DOI: 10.3389/fcimb.2025.1531084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/21/2025] [Indexed: 05/29/2025] Open
Abstract
It is argued that commensal bacteria in the upper respiratory tract (URT) protect against pathogen colonization and infection, including respiratory viruses. Given that the microbiome can mediate immune modulation, a link between the URT microbiome (URTM) and COVID-19 susceptibility and severity is expected. This 16S metagenomics cross-sectional study assessed URTM composition, metabolic prediction, and association with laboratory biomarkers in non-COVID-19 pneumonia (NO-CoV), moderate (M-CoV), severe (S-CoV) COVID-19 patients, as well as COVID-19-negative, asymptomatic (NC) patients. The S-CoV group exhibited reduced URTM diversity, primarily due to a decreased abundance of eubiotic taxa. Some of these taxa (e.g., Haemophilus sp., Neisseria sp.) were also associated with inflammatory biomarkers. Multiple metabolic pathways (e.g., short-chain fatty acids, vitamin B12) linked to immune response, antiviral activity, and host susceptibility showed decreased abundance in S-CoV. These pathways could suggest potential alternatives for the therapeutic arsenal against COVID-19, providing reassurance about the progress in understanding and treating this disease.
Collapse
Affiliation(s)
- Otávio von Ameln Lovison
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Fabiana Caroline Zempulski Volpato
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
| | - Lorenzo Gómez Weber
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - Afonso Luis Barth
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
| | - Adriana Simon Coitinho
- Laboratório de Neuroimunologia do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andreza Francisco Martins
- Laboratório de Pesquisa em Resistência Bacteriana (LABRESIS), Hospital de Clínicas de Porto Alegre (HCPA), Centro de Pesquisa Experimental, Porto Alegre, Brazil
- Laboratório de Microbiologia e Saúde Única do Instituto de Ciências Básicas da Saúde (ICBS) da Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Núcleo de Bioinformática (Bioinformatics Core), Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| |
Collapse
|
4
|
Vázquez-González L, Regueira-Iglesias A, Balsa-Castro C, Tomás I, Carreira MJ. A curated bacterial and archaeal 16S rRNA Gene Oral Sequences dataset. Sci Data 2025; 12:729. [PMID: 40316599 PMCID: PMC12048654 DOI: 10.1038/s41597-025-05050-4] [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: 06/19/2024] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
In a given species, genomes and 16S rRNA gene sequences, along with their intragenomic copy numbers, can vary greatly across environments. The gene copy numbers are crucial for technologies which estimate microbial abundances based on gene counts, such as polymerase chain reaction and high-throughput sequencing. In these, taxa with fewer genes may be underestimated, while those with more genes might be overestimated. Therefore, it is essential to have accurate gene copy number databases specific to the niche under study. The 16S rRNA Gene Oral Sequences dataset (16SGOSeq) contains the number of 16S rRNA genes and their variants in the complete genomes of the bacterial and archaeal species present in the human oral cavity. It includes 3,192 complete genomes of oral bacteria and 191 complete genomes of oral archaea, from which the 16S rRNA gene sequences were extracted, and the sequence variants were identified. This oral-specific dataset of prokaryotic organisms and the pipeline followed for its construction can be applied by clinical microbiologists, bioinformaticians, or microbial ecologists in future microbiome research.
Collapse
Affiliation(s)
- Lara Vázquez-González
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
| | - Alba Regueira-Iglesias
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain
| | - Carlos Balsa-Castro
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain.
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain.
| | - María J Carreira
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain.
- Departamento de Electrónica e Computación, Escola Técnica Superior de Enxeñaría, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain.
| |
Collapse
|
5
|
Yuan J, Sun B, Li M, Yang C, Zhang L, Chen N, Chen F, Li L. OSaMPle workflow for salivary metaproteomics analysis reveals dysbiosis in inflammatory bowel disease patients. NPJ Biofilms Microbiomes 2025; 11:63. [PMID: 40268913 PMCID: PMC12018957 DOI: 10.1038/s41522-025-00692-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: 09/09/2024] [Accepted: 04/03/2025] [Indexed: 04/25/2025] Open
Abstract
The human oral microbiome has been associated with multiple inflammatory conditions including inflammatory bowel disease (IBD). Identifying functional changes in oral microbiome by metaproteomics helps understanding the factors driving dysbiosis related to intestinal diseases. However, enriching bacterial cells from oral samples (such as saliva and mouth rinse) rich in host proteins is challenging. Here, we present an Optimized Salivary MetaProteomic sample analysis workflow (OSaMPle) to enrich salivary bacteria and reduce host-derived interferences for in-depth analysis of the oral metaproteome. Compared to a conventional approach, OSaMPle improved the identification of bacterial peptides and proteins by 3.2 folds and 1.7 folds, respectively. Furthermore, applying OSaMPle to analyze mouth rinse samples from IBD patients revealed significant alterations in bacterial protein expressions under disease conditions. Specifically, proteins involved in the fatty acid elongation pathway in Peptostreptococcus were significantly less abundant in IBD patients, whereas proteins associated with the TCA cycle in Neisseria were significantly more abundant. The OSaMPle workflow is capable of processing small-volume oral samples and adaptable to high-throughput automation. It holds promise as a strategy for investigating the functional responses of oral microbiomes under disease conditions and identifying disease-associated microbes with their proteins, providing critical insights for detecting disease-related biomarkers within the oral microbiome.
Collapse
Affiliation(s)
- Jinhui Yuan
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China
| | - Boyan Sun
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China
| | - Murong Li
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 100081, Beijing, China
| | - Congyi Yang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China
| | - Lingqiang Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing, China.
| | - Feng Chen
- Central Laboratory, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 100081, Beijing, China.
| | - Leyuan Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China.
| |
Collapse
|
6
|
Wang M, Yuan T, Chen J, Yang J, Pu J, Lin W, Dong K, Zhang L, Yuan J, Zheng H, Sun Y, Xu J. A species-level identification pipeline for human gut microbiota based on the V3-V4 regions of 16S rRNA. Front Microbiol 2025; 16:1553124. [PMID: 40226098 PMCID: PMC11985812 DOI: 10.3389/fmicb.2025.1553124] [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: 12/30/2024] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
16S rRNA gene sequencing is pivotal for identifying bacterial species in microbiome studies, especially using the V3-V4 hypervariable regions. A fixed 98.5% similarity threshold is often applied for species-level identification, but this approach can cause misclassification due to varying thresholds among species. To address this, our study integrated data from SILVA, NCBI, and LPSN databases, extracting V3-V4 region sequences and supplementing them with 16S rRNA sequences from 1,082 human gut samples. This resulted in a non-redundant amplicon sequence variants (ASVs) database specific to the V3-V4 regions (positions 341-806). Utilizing this database, we identified flexible classification thresholds for 674 families, 3,661 genera, and 15,735 species, finding clear thresholds for 87.09% of families and 98.38% of genera. For the 896 most common human gut species, we established precise taxonomic thresholds. To leverage these findings, we developed the asvtax pipeline, which applies flexible thresholds for more accurate taxonomic classification, notably improving the identification of new ASVs. The asvtax pipeline not only enhances the precision of species-level classification but also provides a robust framework for analyzing complex microbial communities, facilitating more reliable ecological and functional interpretations in microbiome research.
Collapse
Affiliation(s)
- Min Wang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Tingting Yuan
- School of Medicine, Research Institute of Public Health, Nankai University, Tianjin, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jiali Chen
- School of Medicine, Research Institute of Public Health, Nankai University, Tianjin, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jing Yang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Ji Pu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenchao Lin
- Uniteomics Tianjin Biotechnology Co., Ltd., Tianjin, China
- Beijing Institute of Infectious Diseases, Beijing, China
| | - Kui Dong
- Department of Epidemiology, School of Public Health, Shanxi Medical University, Taiyuan, China
| | - Luqing Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Jiale Yuan
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Han Zheng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yamin Sun
- Beijing Institute of Infectious Diseases, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Jianguo Xu
- School of Medicine, Research Institute of Public Health, Nankai University, Tianjin, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
7
|
Dash M, Thiyageshwari S, Selvi D, Johnson HKV, Ariyan M, Rajan K, Anandham R. Unveiling microbial diversity in slightly and moderately magnesium deficient acidic soils. Sci Rep 2025; 15:3696. [PMID: 39881163 PMCID: PMC11779885 DOI: 10.1038/s41598-025-87943-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Magnesium (Mg) an essential plant nutrient is widespread deficient in the acidic soils of Nilgiris of Tamil nadu, India. The vegetable yield and quality is especially affected due to deficiency of nutrients like Mg. This study investigates soil characteristics and bacterial diversity in the Nilgiris district of Tamil Nadu, India, with respect to Mg deficiency. The soil samples were collected from different vegetable growing regions of the Nilgiris to assess soil physiocochemical parameters, soil enzymes and soil Mg status. 16S rRNA gene-based metagenomic analysis used to investigate the functional potential and structural diversity of the bacterial communities in high Mg and low Mg deficiency soil. Results indicated mildly acidic soils with a sandy loam texture and high organic carbon content. While nitrogen (N), phosphorus (P), and potassium (K) levels were adequate, Mg deficiency was consistent. Soil enzymes such as dehydrogenase, acid phosphatase, urease and aryl sulfatase, varied across the soil samples. Additionally, 16S rRNA gene-based metagenomics analysis revealed the bacterial diversity and functional pathways in soils with high and low Mg deficiency. Low Mg levels were associated with increased bacterial richness, dominated by Proteobacteria, Gemmatimonadetes, Actinobacteria, Bacteroidetes, and Acidobacteria. Functional pathways related to carbon metabolism, amino acid biosynthesis, and various metabolic processes were more abundant in low Mg deficient soils. This research highlights the significant influence of Mg levels on bacterial diversity and functional potentials in acidic soils, providing insights into soil management strategies in Mg-deficient regions.
Collapse
Affiliation(s)
- Munmun Dash
- Department of Soil Science and Agricultural Chemistry, Tamil Nadu Agricultural University, Tamil Nadu, Coimbatore, 641003, India
| | - Subramaniam Thiyageshwari
- Department of Soil Science and Agricultural Chemistry, Tamil Nadu Agricultural University, Tamil Nadu, Coimbatore, 641003, India.
| | - Duraisamy Selvi
- Department of Soil Science and Agricultural Chemistry, Tamil Nadu Agricultural University, Tamil Nadu, Coimbatore, 641003, India
| | - Haina K V Johnson
- Department of Soil Science and Agricultural Chemistry, Tamil Nadu Agricultural University, Tamil Nadu, Coimbatore, 641003, India
| | - Manikandan Ariyan
- Institute of Ecology and Earth Science, University of Tartu, Tartu, Estonia
| | - Karuppusamy Rajan
- Division of Soil Science, ICAR-Indian Institute of Soil and Water Conservation Research Centre, Datia, 475661, Madhya Pradesh, India
| | - Rangasamy Anandham
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, Tamil Nadu, India.
| |
Collapse
|
8
|
Escobar Marcillo DI, Privitera GF, Rollo F, Latini A, Giuliani E, Benevolo M, Giuliani M, Pichi B, Pellini R, Donà MG. Microbiome analysis in individuals with human papillomavirus oral infection. Sci Rep 2025; 15:2953. [PMID: 39848958 PMCID: PMC11757712 DOI: 10.1038/s41598-024-81607-4] [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: 02/23/2024] [Accepted: 11/27/2024] [Indexed: 01/25/2025] Open
Abstract
Microbiome gained attention as a cofactor in cancers originating from epithelial tissues. High-risk (hr)HPV infection causes oropharyngeal squamous cell carcinoma but only in a fraction of hrHPV+ individuals, suggesting that other factors play a role in cancer development. We investigated oral microbiome in cancer-free subjects harboring hrHPV oral infection (n = 33) and matched HPV- controls (n = 30). DNA purified from oral rinse-and-gargles of HIV-infected (HIV+) and HIV-uninfected (HIV-) individuals were used for 16S rRNA gene V3-V4 region amplification and sequencing. Analysis of differential microbial abundance and differential pathway abundance was performed, separately for HIV+ and HIV- individuals. Significant differences in alpha (Chao-1 and Shannon indices) and beta diversity (unweighted UniFrac distance) were observed between hrHPV+ and HPV-negative subjects, but only for the HIV- individuals. Infection by hrHPVs was associated with significant changes in the abundance of Saccharibacteria in HIV+ and Gracilibacteria in HIV- subjects. At the genus level, the greatest change in HIV+ individuals was observed for Bulleidia, which was significantly enriched in hrHPV+ subjects. In HIV- individuals, those hrHPV+ showed a significant enrichment of Parvimonas and depletion of Alloscardovia. Our data suggest a possible interplay between hrHPV infection and oral microbiome, which may vary with the HIV status.
Collapse
Affiliation(s)
- David Israel Escobar Marcillo
- Section of Mechanisms, Biomarkers and Models, Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Grete Francesca Privitera
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Francesca Rollo
- Pathology Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Alessandra Latini
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Eugenia Giuliani
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Maria Benevolo
- Pathology Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Massimo Giuliani
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Barbara Pichi
- Otolaryngology Head Neck Surgery Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Raul Pellini
- Otolaryngology Head Neck Surgery Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Casarin RCV, Silva RVCD, Paz HEDS, Stolf CS, Carvalho LM, Noronha MF, Sallum AW, Monteiro MDF. Metatranscriptomic analysis shows functional alterations in subgingival biofilm in young smokers with periodontitis: a pilot study. J Appl Oral Sci 2024; 32:e20240031. [PMID: 39166556 PMCID: PMC11364450 DOI: 10.1590/1678-7757-2024-0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 06/24/2024] [Indexed: 08/23/2024] Open
Abstract
OBJECTIVE This study aimed to assess the influence of smoking on the subgingival metatranscriptomic profile of young patients affected by stage III/IV and generalized periodontal disease. METHODOLOGY In total, six young patients, both smokers and non-smokers (n=3/group), who were affected by periodontitis were chosen. The STROBE (Strengthening the Reporting of Observational Studies in Epidemiology) guidelines for case-control reporting were followed. Periodontal clinical measurements and subgingival biofilm samples were collected. RNA was extracted from the biofilm and sequenced via Illumina HiSeq. Differential expression analysis used Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and differentially expressed genes were identified using the Sleuth package in R, with a statistical cutoff of ≤0.05. RESULTS This study found 3351 KEGGs in the subgingival biofilm of both groups. Smoking habits altered the functional behavior of subgingival biofilm, resulting in 304 differentially expressed KEGGs between groups. Moreover, seven pathways were modulated: glycan degradation, galactose metabolism, glycosaminoglycan degradation, oxidative phosphorylation, peptidoglycan biosynthesis, butanoate metabolism, and glycosphingolipid biosynthesis. Smoking also altered antibiotic resistance gene levels in subgingival biofilm by significantly overexpressing genes related to beta-lactamase, permeability, antibiotic efflux pumps, and antibiotic-resistant synthetases. CONCLUSION Due to the limitations of a small sample size, our data suggest that smoking may influence the functional behavior of subgingival biofilm, modifying pathways that negatively impact the behavior of subgingival biofilm, which may lead to a more virulent community.
Collapse
Affiliation(s)
- Renato Corrêa Viana Casarin
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| | - Rafaela Videira Clima da Silva
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| | - Hélvis Enri de Sousa Paz
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| | - Camila Schmidt Stolf
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| | - Lucas Miguel Carvalho
- Universidade Estadual de Campinas, Centro de Pesquisas em Engenharias e Ciências Computacionais, Campinas, Brasil
| | - Melline Fontes Noronha
- University of Illinois at Chicago, Research Resource Center, Research Informatics Core, Illinois, USA
| | - Antonio Wilson Sallum
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| | - Mabelle de Freitas Monteiro
- Universidade Estadual de Campinas, Faculdade de Odontologia de Piracicaba, Departamento de Prótese e Periodontia, Piracicaba, Brasil
| |
Collapse
|
12
|
Rocha CM, Kawamoto D, Martins FH, Bueno MR, Ishikawa KH, Ando-Suguimoto ES, Carlucci AR, Arroteia LS, Casarin RV, Saraiva L, Simionato MRL, Mayer MPA. Experimental Inoculation of Aggregatibacter actinomycetemcomitans and Streptococcus gordonii and Its Impact on Alveolar Bone Loss and Oral and Gut Microbiomes. Int J Mol Sci 2024; 25:8090. [PMID: 39125663 PMCID: PMC11312116 DOI: 10.3390/ijms25158090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 08/12/2024] Open
Abstract
Oral bacteria are implicated not only in oral diseases but also in gut dysbiosis and inflammatory conditions throughout the body. The periodontal pathogen Aggregatibacter actinomycetemcomitans (Aa) often occurs in complex oral biofilms with Streptococcus gordonii (Sg), and this interaction might influence the pathogenic potential of this pathogen. This study aims to assess the impact of oral inoculation with Aa, Sg, and their association (Aa+Sg) on alveolar bone loss, oral microbiome, and their potential effects on intestinal health in a murine model. Sg and/or Aa were orally administered to C57Bl/6 mice, three times per week, for 4 weeks. Aa was also injected into the gingiva three times during the initial experimental week. After 30 days, alveolar bone loss, expression of genes related to inflammation and mucosal permeability in the intestine, serum LPS levels, and the composition of oral and intestinal microbiomes were determined. Alveolar bone resorption was detected in Aa, Sg, and Aa+Sg groups, although Aa bone levels did not differ from that of the SHAM-inoculated group. Il-1β expression was upregulated in the Aa group relative to the other infected groups, while Il-6 expression was downregulated in infected groups. Aa or Sg downregulated the expression of tight junction genes Cldn 1, Cldn 2, Ocdn, and Zo-1 whereas infection with Aa+Sg led to their upregulation, except for Cldn 1. Aa was detected in the oral biofilm of the Aa+Sg group but not in the gut. Infections altered oral and gut microbiomes. The oral biofilm of the Aa group showed increased abundance of Gammaproteobacteria, Enterobacterales, and Alloprevotella, while Sg administration enhanced the abundance of Alloprevotella and Rothia. The gut microbiome of infected groups showed reduced abundance of Erysipelotrichaceae. Infection with Aa or Sg disrupts both oral and gut microbiomes, impacting oral and gut homeostasis. While the combination of Aa with Sg promotes Aa survival in the oral cavity, it mitigates the adverse effects of Aa in the gut, suggesting a beneficial role of Sg associations in gut health.
Collapse
Affiliation(s)
- Catarina Medeiros Rocha
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Dione Kawamoto
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Fernando Henrique Martins
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Manuela Rocha Bueno
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Division of Periodontics, Faculdade São Leopoldo Mandic, São Leopoldo Mandic Research Institute, Campinas 13045-755, SP, Brazil
| | - Karin H. Ishikawa
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Ellen Sayuri Ando-Suguimoto
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Aline Ramos Carlucci
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Leticia Sandoli Arroteia
- Department of Prosthesis and Periodontology, School of Dentistry, University of Campinas, Campinas 13083-875, SP, Brazil; (L.S.A.); (R.V.C.)
| | - Renato V. Casarin
- Department of Prosthesis and Periodontology, School of Dentistry, University of Campinas, Campinas 13083-875, SP, Brazil; (L.S.A.); (R.V.C.)
| | - Luciana Saraiva
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Maria Regina Lorenzetti Simionato
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
| | - Marcia Pinto Alves Mayer
- Department of Microbiology, Institute of Biomedical Science, University of São Paulo, São Paulo 05508-000, SP, Brazil; (C.M.R.); (D.K.); (F.H.M.); (M.R.B.); (K.H.I.); (E.S.A.-S.); (A.R.C.); (M.R.L.S.)
- Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
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
|
Manoil D, Parga A, Bostanci N, Belibasakis GN. Microbial diagnostics in periodontal diseases. Periodontol 2000 2024; 95:176-193. [PMID: 38797888 DOI: 10.1111/prd.12571] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
Microbial analytical methods have been instrumental in elucidating the complex microbial etiology of periodontal diseases, by shaping our understanding of subgingival community dynamics. Certain pathobionts can orchestrate the establishment of dysbiotic communities that can subvert the host immune system, triggering inflammation and tissue destruction. Yet, diagnosis and management of periodontal conditions still rely on clinical and radiographic examinations, overlooking the well-established microbial etiology. This review summarizes the chronological emergence of periodontal etiological models and the co-evolution with technological advances in microbial detection. We additionally review the microbial analytical approaches currently accessible to clinicians, highlighting their value in broadening the periodontal assessment. The epidemiological importance of obtaining culture-based antimicrobial susceptibility profiles of periodontal taxa for antibiotic resistance surveillance is also underscored, together with clinically relevant analytical approaches to guide antibiotherapy choices, when necessary. Furthermore, the importance of 16S-based community and shotgun metagenomic profiling is discussed in outlining dysbiotic microbial signatures. Because dysbiosis precedes periodontal damage, biomarker identification offers early diagnostic possibilities to forestall disease relapses during maintenance. Altogether, this review highlights the underutilized potential of clinical microbiology in periodontology, spotlighting the clinical areas most conductive to its diagnostic implementation for enhancing prevention, treatment predictability, and addressing global antibiotic resistance.
Collapse
Affiliation(s)
- Daniel Manoil
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Ana Parga
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Microbiology and Parasitology, CIBUS-Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Georgios N Belibasakis
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| |
Collapse
|
15
|
Vázquez-González L, Regueira-Iglesias A, Balsa-Castro C, Vila-Blanco N, Tomás I, Carreira MJ. PrimerEvalPy: a tool for in-silico evaluation of primers for targeting the microbiome. BMC Bioinformatics 2024; 25:189. [PMID: 38745271 PMCID: PMC11092261 DOI: 10.1186/s12859-024-05805-7] [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: 11/29/2023] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND The selection of primer pairs in sequencing-based research can greatly influence the results, highlighting the need for a tool capable of analysing their performance in-silico prior to the sequencing process. We therefore propose PrimerEvalPy, a Python-based package designed to test the performance of any primer or primer pair against any sequencing database. The package calculates a coverage metric and returns the amplicon sequences found, along with information such as their average start and end positions. It also allows the analysis of coverage for different taxonomic levels. RESULTS As a case study, PrimerEvalPy was used to test the most commonly used primers in the literature against two oral 16S rRNA gene databases containing bacteria and archaea. The results showed that the most commonly used primer pairs in the oral cavity did not match those with the highest coverage. The best performing primer pairs were found for the detection of oral bacteria and archaea. CONCLUSIONS This demonstrates the importance of a coverage analysis tool such as PrimerEvalPy to find the best primer pairs for specific niches. The software is available under the MIT licence at https://gitlab.citius.usc.es/lara.vazquez/PrimerEvalPy .
Collapse
Affiliation(s)
- Lara Vázquez-González
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain.
| | - Alba Regueira-Iglesias
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
| | - Carlos Balsa-Castro
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
| | - Nicolás Vila-Blanco
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain
- Departamento de Electrónica e Computación, Escola Técnica Superior de Enxeñaría, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain.
- Oral Sciences Research Group, Special Needs Unit, Department of Surgery and Medical Surgical Specialities, School of Medicine and Dentistry, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain.
| | - María J Carreira
- Centro Singular de Investigación en Tecnoloxías Intelixentes (CiTIUS), Universidade de Santiago de Compostela, Rúa de Jenaro de la Fuente Domínguez, E15782, Santiago de Compostela, Spain.
- Departamento de Electrónica e Computación, Escola Técnica Superior de Enxeñaría, Universidade de Santiago de Compostela, E15782, Santiago de Compostela, Spain.
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), E15706, Santiago de Compostela, Spain.
| |
Collapse
|
16
|
Solfisburg QS, Baldini F, Baldwin-Hunter B, Austin GI, Lee HH, Park H, Freedberg DE, Lightdale CJ, Korem T, Abrams JA. The Salivary Microbiome and Predicted Metabolite Production Are Associated with Barrett's Esophagus and High-Grade Dysplasia or Adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2024; 33:371-380. [PMID: 38117184 PMCID: PMC10955687 DOI: 10.1158/1055-9965.epi-23-0652] [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: 06/05/2023] [Revised: 09/05/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is rising in incidence, and established risk factors do not explain this trend. Esophageal microbiome alterations have been associated with Barrett's esophagus (BE) and dysplasia and EAC. The oral microbiome is tightly linked to the esophageal microbiome; this study aimed to identify salivary microbiome-related factors associated with BE, dysplasia, and EAC. METHODS Clinical data and oral health history were collected from patients with and without BE. The salivary microbiome was characterized, assessing differential relative abundance of taxa by 16S rRNA gene sequencing and associations between microbiome composition and clinical features. Microbiome metabolic modeling was used to predict metabolite production. RESULTS A total of 244 patients (125 non-BE and 119 BE) were analyzed. Patients with high-grade dysplasia (HGD)/EAC had a significantly higher prevalence of tooth loss (P = 0.001). There were significant shifts with increased dysbiosis associated with HGD/EAC, independent of tooth loss, with the largest shifts within the genus Streptococcus. Modeling predicted significant shifts in the microbiome metabolic capacities, including increases in L-lactic acid and decreases in butyric acid and L-tryptophan production in HGD/EAC. CONCLUSIONS Marked dysbiosis in the salivary microbiome is associated with HGD and EAC, with notable increases within the genus Streptococcus and accompanying changes in predicted metabolite production. Further work is warranted to identify the biological significance of these alterations and to validate metabolic shifts. IMPACT There is an association between oral dysbiosis and HGD/EAC. Further work is needed to establish the diagnostic, predictive, and causal potential of this relationship.
Collapse
Affiliation(s)
- Quinn S Solfisburg
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Federico Baldini
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - George I Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Harry H Lee
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel E Freedberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles J Lightdale
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
- CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
17
|
Dutra TP, Freitas Monteiro M, França-Grohmann IL, Casarin RCV, Casati MZ, Silvério Ruiz KG, Kumar PS, Sallum EA. Clinical, immunological and microbiological evaluation of experimental peri-implant mucositis and gingivitis in subjects with Grade C, stage III/IV periodontitis background. J Clin Periodontol 2024; 51:209-221. [PMID: 37941050 DOI: 10.1111/jcpe.13896] [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: 03/22/2022] [Revised: 10/04/2023] [Accepted: 10/15/2023] [Indexed: 11/10/2023]
Abstract
AIM To compare individuals with a periodontitis background (Grade C, stage III/IV-formerly generalized aggressive periodontitis) (H-GAP) with periodontally healthy subjects (H-Health) in terms of molecular changes (immunological/microbiological) accompanying experimental peri-implant mucositis and gingivitis. MATERIALS AND METHODS H-GAP and control (H-Health) subjects were recruited, and experimental mucositis/gingivitis was induced around a single screw-retained implant and one contralateral tooth. Participants refrained from oral hygiene for 21 days in the selected areas, followed by professional prophylaxis and hygiene instructions for 21 days. Clinical parameters, immunological markers (multiplex analysis) and microbial data (16S rRNA gene sequencing) were collected at baseline, during induction (7, 14 and 21 days) and following remission (42 days). RESULTS Clinically, no significant differences were observed between the groups (n = 10/each group) (H-GAP vs. H-Health) (p > .05, Mann-Whitney test) and the type of site (tooth vs. implant) (p > .05, Wilcoxon test) at the time of onset and resolution, or severity of gingival/mucosal inflammation. H-GAP displayed lower concentrations of the cytokines interleukin (IL)-1B, IL-4, IL-17, tumor necrosis factor-α and interferon-γ around implants than H-Health at baseline and during induction of mucositis (p < .05, Mann-Whitney test). In both groups, implants showed significantly higher inflammatory background at baseline and all subsequent visits when compared with teeth (p < .05, Wilcoxon test). Alpha and β-diversity metrics showed a significant shift in the microbiome composition and abundances of core species during induction and resolution of peri-implant mucositis and gingivitis (p < .05, restricted maximum likelihood method of Shannon and Bray-Curtis indices, respectively). Differences were not significant for these parameters between the H-Health and H-GAP groups when the periodontal and peri-implant microbiomes were compared separately; however, at each time point, the peri-implant microbiome differed significantly from the periodontal microbiome. CONCLUSIONS Within the limitations of this pilot study (e.g. low power), it can be concluded that different microbial shifts contribute to the onset and progression of inflammatory responses around teeth and implants and that history of periodontal disease experience plays an additional role in modulating the immune response of peri-implant and periodontal tissues to biofilm accumulation.
Collapse
Affiliation(s)
- Tamires Pereira Dutra
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
- Department of Periodontics and Oral Medicine, University of Michigan - School of Dentistry, Ann Arbor, Michigan, USA
| | - Mabelle Freitas Monteiro
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Isabela Lima França-Grohmann
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Renato Corrêa Viana Casarin
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Márcio Zaffalon Casati
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Karina Gonzalez Silvério Ruiz
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| | - Purnima S Kumar
- Department of Periodontics and Oral Medicine, University of Michigan - School of Dentistry, Ann Arbor, Michigan, USA
| | - Enílson Antônio Sallum
- Department of Prosthodontics and Periodontics, Division of Periodontics, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo, Brazil
| |
Collapse
|
18
|
Bhaumik D, Salzman E, Davis E, Blostein F, Li G, Neiswanger K, Weyant R, Crout R, McNeil D, Marazita M, Foxman B. Plaque Microbiome in Caries-Active and Caries-Free Teeth by Dentition. JDR Clin Trans Res 2024; 9:61-71. [PMID: 36154330 PMCID: PMC10725180 DOI: 10.1177/23800844221121260] [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] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE Describe associations between dental caries and dental plaque microbiome, by dentition and family membership. METHODS This cross-sectional analysis included 584 participants in the Center for Oral Health Research in Appalachia Cohort 1 (COHRA1). We sequenced the 16S ribosomal RNA gene (V4 region) of frozen supragingival plaque, collected 10 y prior, from 185 caries-active (enamel and dentinal) and 565 caries-free (no lesions) teeth using the Illumina MiSeq platform. Sequences were filtered using the R DADA2 package and assigned taxonomy using the Human Oral Microbiome Database. RESULTS Microbiomes of caries-active and caries-free teeth were most similar in primary dentition and least similar in permanent dentition, but caries-active teeth were significantly less diverse than caries-free teeth in all dentition types. Streptococcus mutans had greater relative abundance in caries-active than caries-free teeth in all dentition types (P < 0.01), as did Veillonella dispar in primary and mixed dentition (P < 0.01). Fusobacterium sp. HMT 203 had significantly higher relative abundance in caries-free than caries-active teeth in all dentition types (P < 0.01). In a linear mixed model adjusted for confounders, the relative abundance of S. mutans was significantly greater in plaque from caries-active than caries-free teeth (P < 0.001), and the relative abundance of Fusobacterium sp. HMT 203 was significantly lower in plaque from caries-active than caries-free teeth (P < 0.001). Adding an effect for family improved model fit for Fusobacterium sp. HMT 203 but notS. mutans. CONCLUSIONS The diversity of supragingival plaque composition from caries-active and caries-free teeth changed with dentition, but S. mutans was positively and Fusobacterium sp. HMT 203 was negatively associated with caries regardless of dentition. There was a strong effect of family on the associations of Fusobacterium sp. HMT 203 with the caries-free state, but this was not true for S. mutans and the caries-active state. KNOWLEDGE TRANSFER STATEMENT Patients' and dentists' concerns about transmission of bacteria within families causing caries should be tempered by the evidence that some shared bacteria may contribute to good oral health.
Collapse
Affiliation(s)
- D. Bhaumik
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - E. Salzman
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - E. Davis
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - F. Blostein
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - G. Li
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - K. Neiswanger
- Center for Craniofacial and Dental Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - R.J. Weyant
- Dental Public Health, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - R. Crout
- Department of Periodontics, West Virginia University, Morgantown, WV, USA
| | - D.W. McNeil
- Departments of Psychology and Dental Practice & Rural Health, and Center for Oral Health Research in Appalachia, West Virginia University, Morgantown, WV, USA
| | - M.L. Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences; Department of Human Genetics, Graduate School of Public Health; Clinical and Translational Science, School of Medicine University of Pittsburgh, Pittsburgh, PA, USA
| | - B. Foxman
- Center of Molecular and Clinical Epidemiology of Infectious Diseases, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Zhang T, Li H, Ma S, Cao J, Liao H, Huang Q, Chen W. The newest Oxford Nanopore R10.4.1 full-length 16S rRNA sequencing enables the accurate resolution of species-level microbial community profiling. Appl Environ Microbiol 2023; 89:e0060523. [PMID: 37800969 PMCID: PMC10617388 DOI: 10.1128/aem.00605-23] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/04/2023] [Indexed: 10/07/2023] Open
Abstract
The long-read amplicon provides a species-level solution for the community. With the improvement of nanopore flowcells, the accuracy of Oxford Nanopore Technologies (ONT) R10.4.1 has been substantially enhanced, with an average of approximately 99%. To evaluate its effectiveness on amplicons, three types of microbiomes were analyzed by 16S ribosomal RNA (hereinafter referred to as "16S") amplicon sequencing using Novaseq, Pacbio sequel II, and Nanopore PromethION platforms (R9.4.1 and R10.4.1) in the current study. We showed the error rate, recall, precision, and bias index in the mock sample. The error rate of ONT R10.4.1 was greatly reduced, with a better recall in the case of the synthetic community. Meanwhile, in different types of environmental samples, ONT R10.4.1 analysis resulted in a composition similar to Pacbio data. We found that classification tools and databases influence ONT data. Based on these results, we conclude that the ONT R10.4.1 16S amplicon can also be used for application in environmental samples. IMPORTANCE The long-read amplicon supplies the community with a species-level solution. Due to the high error rate of nanopore sequencing early on, it has not been frequently used in 16S studies. Oxford Nanopore Technologies (ONT) introduced the R10.4.1 flowcell with Q20+ reagent to achieve more than 99% accuracy as sequencing technology advanced. However, there has been no published study on the performance of commercial PromethION sequencers with R10.4.1 flowcells on 16S sequencing or on the impact of accuracy improvement on taxonomy (R9.4.1 to R10.4.1) using 16S ONT data. In this study, three types of microbiomes were investigated by 16S ribosomal RNA (rRNA) amplicon sequencing using Novaseq, Pacbio sequel II, and Nanopore PromethION platforms (R9.4.1 and R10.4.1). In the mock sample, we displayed the error rate, recall, precision, and bias index. We observed that the error rate in ONT R10.4.1 is significantly lower, especially when deletions are involved. First and foremost, R10.4.1 and Pacific Bioscience platforms reveal a similar microbiome in environmental samples. This study shows that the R10.4.1 full-length 16S rRNA sequences allow for species identification of environmental microbiota.
Collapse
Affiliation(s)
- Tianyuan Zhang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- Wuhan Benagen Technology Co., Ltd., Wuhan, China
| | - Hanzhou Li
- Wuhan Benagen Technology Co., Ltd., Wuhan, China
| | - Silin Ma
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Jian Cao
- Wuhan Benagen Technology Co., Ltd., Wuhan, China
| | - Hao Liao
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Qiaoyun Huang
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Soil Environment and Pollution Remediation, Huazhong Agricultural University, Wuhan, China
| | - Wenli Chen
- National Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
van Dalen R, Elsherbini AMA, Harms M, Alber S, Stemmler R, Peschel A. Secretory IgA impacts the microbiota density in the human nose. MICROBIOME 2023; 11:233. [PMID: 37865781 PMCID: PMC10589987 DOI: 10.1186/s40168-023-01675-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/24/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Respiratory mucosal host defense relies on the production of secretory IgA (sIgA) antibodies, but we currently lack a fundamental understanding of how sIgA is induced by contact with microbes and how such immune responses may vary between humans. Defense of the nasal mucosal barrier through sIgA is critical to protect from infection and to maintain homeostasis of the microbiome, which influences respiratory disorders and hosts opportunistic pathogens. METHODS We applied IgA-seq analysis to nasal microbiota samples from male and female healthy volunteers, to identify which bacterial genera and species are targeted by sIgA on the level of the individual host. Furthermore, we used nasal sIgA from the same individuals in sIgA deposition experiments to validate the IgA-seq outcomes. CONCLUSIONS We observed that the amount of sIgA secreted into the nasal mucosa by the host varied substantially and was negatively correlated with the bacterial density, suggesting that nasal sIgA limits the overall bacterial capacity to colonize. The interaction between mucosal sIgA antibodies and the nasal microbiota was highly individual with no obvious differences between potentially invasive and non-invasive bacterial species. Importantly, we could show that for the clinically relevant opportunistic pathogen and frequent nasal resident Staphylococcus aureus, sIgA reactivity was in part the result of epitope-independent interaction of sIgA with the antibody-binding protein SpA through binding of sIgA Fab regions. This study thereby offers a first comprehensive insight into the targeting of the nasal microbiota by sIgA antibodies. It thereby helps to better understand the shaping and homeostasis of the nasal microbiome by the host and may guide the development of effective mucosal vaccines against bacterial pathogens. Video Abstract.
Collapse
Affiliation(s)
- Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- Present Address: Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ahmed M A Elsherbini
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Mareike Harms
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Svenja Alber
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Regine Stemmler
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
21
|
Galvin S, Anishchuk S, Healy CM, Moran GP. Smoking, tooth loss and oral hygiene practices have significant and site-specific impacts on the microbiome of oral mucosal surfaces: a cross-sectional study. J Oral Microbiol 2023; 15:2263971. [PMID: 37795170 PMCID: PMC10547447 DOI: 10.1080/20002297.2023.2263971] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/13/2023] [Indexed: 10/06/2023] Open
Abstract
We investigated bacterial colonisation patterns of healthy mucosa (buccal, tongue, palate and floor of mouth) in a cohort of adults in order to determine how smoking, tooth loss, plaque levels and oral hygiene practices impacted on mucosal colonisation. A total of 322 swabs were recovered from 256 participants, of whom 46% were current smokers. We analysed colonization by sequencing the V1-V3 regions of the 16S rRNA gene. Palate and tongue microbiomes generally exhibited greater biodiversity than buccal and floor of mouth. Although Neisseria, Lautropia and Haemophilus spp. showed reduced abundance in smokers, buccal mucosa specifically showed a significant increase in Prevotella spp., whereas tongue and floor of mouth tended towards increased abundance of Streptococcus spp. Unexpectedly, tooth brushing frequency had a greater impact on mucosal community structure than plaque levels. Tooth loss was associated with significant reductions in mucosal biodiversity and had site-specific impacts, with buccal communities showing increased abundance of periodontitis-associated species and Rothia mucilaginosa, whereas tongue communities exhibited increased abundance of several streptococcal OTUs and reduced abundance of Haemophilus spp. This study highlights the complex relationship between mucosal colonisation and host factors, highlighting the need for careful consideration of these factors in mucosal microbiome studies.
Collapse
Affiliation(s)
- Sheila Galvin
- Division of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, School of Dental Science, Trinity College Dublin, Dublin Dental University Hospital, Dublin, Ireland
| | - Sviatlana Anishchuk
- Division of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, School of Dental Science, Trinity College Dublin, Dublin Dental University Hospital, Dublin, Ireland
| | - Claire M. Healy
- Division of Oral and Maxillofacial Surgery, Oral Medicine and Oral Pathology, School of Dental Science, Trinity College Dublin, Dublin Dental University Hospital, Dublin, Ireland
| | - Gary P. Moran
- Division of Oral Biosciences, School of Dental Science, Trinity College Dublin, Dublin Dental University Hospital, Dublin, Ireland
| |
Collapse
|
22
|
Zhang M, Tang H, Yuan Y, Ou Z, Chen Z, Xu Y, Fu X, Zhao Z, Sun Y. The Role of Indoor Microbiome and Metabolites in Shaping Children's Nasal and Oral Microbiota: A Pilot Multi-Omic Analysis. Metabolites 2023; 13:1040. [PMID: 37887365 PMCID: PMC10608577 DOI: 10.3390/metabo13101040] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/21/2023] [Accepted: 09/27/2023] [Indexed: 10/28/2023] Open
Abstract
Maintaining a diverse and well-balanced nasal and oral microbiota is vital for human health. However, the impact of indoor microbiome and metabolites on nasal and oral microbiota remains largely unknown. Fifty-six children in Shanghai were surveyed to complete a questionnaire about their personal and environmental characteristics. The indoor microbiome and metabolites from vacuumed indoor dust were profiled via shotgun metagenomics and untargeted liquid chromatography-mass spectrometry (LC-MS). The nasal and oral microbiota in children was characterized using full-length 16S rRNA sequencing from PacBio. Associations between personal/environmental characteristics and the nasal/oral microbiota were calculated using PERMANOVA and regression analyses. We identified 6247, 431, and 342 microbial species in the indoor dust, nasal, and oral cavities, respectively. The overall nasal and oral microbial composition showed significant associations with environmental tobacco smoke (ETS) exposure during pregnancy and early childhood (p = 0.005 and 0.03, respectively), and the abundance of total indoor flavonoids and two mycotoxins (deoxynivalenol and nivalenol) (p = 0.01, 0.02, and 0.03, respectively). Notably, the abundance of several flavonoids, such as baicalein, eupatilin, isoliquiritigenin, tangeritin, and hesperidin, showed positive correlations with alpha diversity and the abundance of protective microbial taxa in nasal and oral cavities (p < 0.02), suggesting their potential beneficial roles in promoting nasal/oral health. Conversely, high carbohydrate/fat food intake and ETS exposure diminished protective microorganisms while augmenting risky microorganisms in the nasal/oral cavities. Further, potential microbial transfer was observed from the indoor environment to the childhood oral cavity (Moraxella catarrhalis, Streptococcus mitis, and Streptococcus salivarius), which could potentially increase virulence factors related to adherence and immune modulation and vancomycin resistance genes in children. This is the first study to reveal the association between the indoor microbiome/metabolites and nasal/oral microbiota using multi-omic approaches. These findings reveal potential protective and risk factors related to the indoor microbial environment.
Collapse
Affiliation(s)
- Mei Zhang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.Y.); (Z.O.)
| | - Hao Tang
- School of Public Health, Fudan University, Shanghai 200032, China; (H.T.); (Y.X.)
| | - Yiwen Yuan
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.Y.); (Z.O.)
| | - Zheyuan Ou
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.Y.); (Z.O.)
| | - Zhuoru Chen
- Children’s Hospital of Fudan University, Shanghai 201102, China;
| | - Yanyi Xu
- School of Public Health, Fudan University, Shanghai 200032, China; (H.T.); (Y.X.)
| | - Xi Fu
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment, School of Public Health, Guangdong Pharmaceutical University, Guangzhou 510006, China;
| | - Zhuohui Zhao
- School of Public Health, Fudan University, Shanghai 200032, China; (H.T.); (Y.X.)
- Key Laboratory of Public Health Safety of the Ministry of Education, NHC Key Laboratory of Health Technology Assessment (Fudan University), Shanghai Typhoon Institute/CMA, Shanghai Key Laboratory of Meteorology and Health, Shanghai 200030, China
| | - Yu Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, China; (M.Z.); (Y.Y.); (Z.O.)
| |
Collapse
|
23
|
Iniesta M, Chamorro C, Ambrosio N, Marín MJ, Sanz M, Herrera D. Subgingival microbiome in periodontal health, gingivitis and different stages of periodontitis. J Clin Periodontol 2023; 50:905-920. [PMID: 36792073 DOI: 10.1111/jcpe.13793] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 02/04/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
AIM To characterize the subgingival microbiome in subjects with different periodontal health statuses. MATERIALS AND METHODS In this cross-sectional observational study, subgingival samples were harvested from Spanish subjects with different periodontal health statuses, based on the 2018 Classification of Periodontal and Peri-Implant Diseases and Conditions. Samples were processed using high-throughput sequencing technologies (Illumina MiSeq). Taxa differentially abundant were identified using Analysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC). α- and β-diversity metrics were calculated using q2-diversity in QIIME2. The analyses were adjusted for age, gender and smoking status. RESULTS The identified subgingival microbiome showed statistically significant differences among subjects, categorized into periodontal health, gingivitis and stages I-II and III-IV periodontitis (p < .05). In patients with severe (stages III-IV) periodontitis, the genera Filifactor and Fretibacterium were detected 24 times more frequently than in periodontally healthy subjects. Similarly, the genera Porphyromonas, Prevotella and Tannerella were detected four times more frequently (p < .05). The genera Granulicatella, Streptococcus, Paracoccus, Pseudomonas, Haemophilus, Actinobacteria, Bergeyella and Capnocytophaga were significantly associated with healthier periodontal status (p < .05). CONCLUSIONS Significant differences were detected in the subgingival microbiome among periodontal health, gingivitis and stages I-II or III-IV periodontitis, suggesting overlapping, yet distinguishable microbial profiles.
Collapse
Affiliation(s)
- Margarita Iniesta
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Cristina Chamorro
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Nagore Ambrosio
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - María José Marín
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
24
|
Solfisburg QS, Baldini F, Baldwin-Hunter BL, Lee HH, Park H, Freedberg DE, Lightdale CJ, Korem T, Abrams JA. The Salivary Microbiome and Predicted Metabolite Production are Associated with Progression from Barrett's Esophagus to Esophageal Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.27.546733. [PMID: 37425673 PMCID: PMC10327009 DOI: 10.1101/2023.06.27.546733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Esophageal adenocarcinoma (EAC) is rising in incidence and associated with poor survival, and established risk factors do not explain this trend. Microbiome alterations have been associated with progression from the precursor Barrett's esophagus (BE) to EAC, yet the oral microbiome, tightly linked to the esophageal microbiome and easier to sample, has not been extensively studied in this context. We aimed to assess the relationship between the salivary microbiome and neoplastic progression in BE to identify microbiome-related factors that may drive EAC development. We collected clinical data and oral health and hygiene history and characterized the salivary microbiome from 250 patients with and without BE, including 78 with advanced neoplasia (high grade dysplasia or early adenocarcinoma). We assessed differential relative abundance of taxa by 16S rRNA gene sequencing and associations between microbiome composition and clinical features and used microbiome metabolic modeling to predict metabolite production. We found significant shifts and increased dysbiosis associated with progression to advanced neoplasia, with these associations occurring independent of tooth loss, and the largest shifts were with the genus Streptococcus. Microbiome metabolic models predicted significant shifts in the metabolic capacities of the salivary microbiome in patients with advanced neoplasia, including increases in L-lactic acid and decreases in butyric acid and L-tryptophan production. Our results suggest both a mechanistic and predictive role for the oral microbiome in esophageal adenocarcinoma. Further work is warranted to identify the biological significance of these alterations, to validate metabolic shifts, and to determine whether they represent viable therapeutic targets for prevention of progression in BE.
Collapse
Affiliation(s)
- Quinn S Solfisburg
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Federico Baldini
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Harry H Lee
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Microbiome and Pathogen Genomics Collaborative Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel E Freedberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Charles J Lightdale
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
- CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
- Digestive and Liver Disease Research Center, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY USA
| |
Collapse
|
25
|
Bedree JK, Kerns K, Chen T, Lima BP, Liu G, Ha P, Shi J, Pan HC, Kim JK, Tran L, Minot SS, Hendrickson EL, Lamont EI, Schulte F, Hardt M, Stephens D, Patel M, Kokaras A, Stodieck L, Shirazi-Fard Y, Wu B, Kwak JH, Ting K, Soo C, McLean JS, He X, Shi W. Specific host metabolite and gut microbiome alterations are associated with bone loss during spaceflight. Cell Rep 2023; 42:112299. [PMID: 37080202 PMCID: PMC10344367 DOI: 10.1016/j.celrep.2023.112299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/30/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023] Open
Abstract
Understanding the axis of the human microbiome and physiological homeostasis is an essential task in managing deep-space-travel-associated health risks. The NASA-led Rodent Research 5 mission enabled an ancillary investigation of the gut microbiome, varying exposure to microgravity (flight) relative to ground controls in the context of previously shown bone mineral density (BMD) loss that was observed in these flight groups. We demonstrate elevated abundance of Lactobacillus murinus and Dorea sp. during microgravity exposure relative to ground control through whole-genome sequencing and 16S rRNA analyses. Specific functionally assigned gene clusters of L. murinus and Dorea sp. capable of producing metabolites, lactic acid, leucine/isoleucine, and glutathione are enriched. These metabolites are elevated in the microgravity-exposed host serum as shown by liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. Along with BMD loss, ELISA reveals increases in osteocalcin and reductions in tartrate-resistant acid phosphatase 5b signifying additional loss of bone homeostasis in flight.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| | - Kristopher Kerns
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Tsute Chen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Bruno P Lima
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Guo Liu
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Ha
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiayu Shi
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hsin Chuan Pan
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jong Kil Kim
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luan Tran
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samuel S Minot
- Microbiome Research Initiative, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Erik L Hendrickson
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Eleanor I Lamont
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Fabian Schulte
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Markus Hardt
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Danielle Stephens
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Michele Patel
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Alexis Kokaras
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Louis Stodieck
- BioServe Space Technologies, Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO 80303, USA
| | - Yasaman Shirazi-Fard
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA
| | - Benjamin Wu
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Hee Kwak
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopedic Surgery, School of Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Wenyuan Shi
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
26
|
Hosokawa M, Iwai N, Arikawa K, Saeki T, Endoh T, Kamata K, Yoda T, Tsuda S, Takeyama H. Target enrichment of uncultured human oral bacteria with phage-derived molecules found by single-cell genomics. J Biosci Bioeng 2023:S1389-1723(23)00116-0. [PMID: 37188549 DOI: 10.1016/j.jbiosc.2023.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023]
Abstract
Advances in culture-independent microbial analysis, such as metagenomics and single-cell genomics, have significantly increased our understanding of microbial lineages. While these methods have uncovered a large number of novel microbial taxa, many remain uncultured, and their function and mode of existence in the environment are still unknown. This study aims to explore the use of bacteriophage-derived molecules as probes for detecting and isolating uncultured bacteria. Here, we proposed multiplex single-cell sequencing to obtain massive uncultured oral bacterial genomes and searched prophage sequences from over 450 obtained human oral bacterial single-amplified genomes (SAGs). The focus was on the cell wall binding domain (CBD) in phage endolysin, and fluorescent protein-fused CBDs were generated based on several CBD gene sequences predicted from Streptococcus SAGs. The ability of the Streptococcus prophage-derived CBDs to detect and enrich specific Streptococcus species from human saliva while maintaining cell viability was confirmed by magnetic separation and flow cytometry. The approach to phage-derived molecule generation based on uncultured bacterial SAG is expected to improve the process of designing molecules that selectively capture or detect specific bacteria, notably from uncultured gram-positive bacteria, and will have applications in isolation and in situ detection of beneficial or pathogenic bacteria.
Collapse
Affiliation(s)
- Masahito Hosokawa
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan; bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan; Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan.
| | - Naoya Iwai
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Koji Arikawa
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan; bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Tatsuya Saeki
- bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Taruho Endoh
- bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Kazuma Kamata
- bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Takuya Yoda
- bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Soichiro Tsuda
- bitBiome, Inc., 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan; Research Organization for Nano and Life Innovation, Waseda University, 513 Wasedatsurumaki-cho, Shinjuku-ku, Tokyo 162-0041, Japan; Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan; Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| |
Collapse
|
27
|
Regueira-Iglesias A, Vázquez-González L, Balsa-Castro C, Vila-Blanco N, Blanco-Pintos T, Tamames J, Carreira MJ, Tomás I. In silico evaluation and selection of the best 16S rRNA gene primers for use in next-generation sequencing to detect oral bacteria and archaea. MICROBIOME 2023; 11:58. [PMID: 36949474 PMCID: PMC10035280 DOI: 10.1186/s40168-023-01481-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Sequencing has been widely used to study the composition of the oral microbiome present in various health conditions. The extent of the coverage of the 16S rRNA gene primers employed for this purpose has not, however, been evaluated in silico using oral-specific databases. This paper analyses these primers using two databases containing 16S rRNA sequences from bacteria and archaea found in the human mouth and describes some of the best primers for each domain. RESULTS A total of 369 distinct individual primers were identified from sequencing studies of the oral microbiome and other ecosystems. These were evaluated against a database reported in the literature of 16S rRNA sequences obtained from oral bacteria, which was modified by our group, and a self-created oral archaea database. Both databases contained the genomic variants detected for each included species. Primers were evaluated at the variant and species levels, and those with a species coverage (SC) ≥75.00% were selected for the pair analyses. All possible combinations of the forward and reverse primers were identified, with the resulting 4638 primer pairs also evaluated using the two databases. The best bacteria-specific pairs targeted the 3-4, 4-7, and 3-7 16S rRNA gene regions, with SC levels of 98.83-97.14%; meanwhile, the optimum archaea-specific primer pairs amplified regions 5-6, 3-6, and 3-6, with SC estimates of 95.88%. Finally, the best pairs for detecting both domains targeted regions 4-5, 3-5, and 5-9, and produced SC values of 95.71-94.54% and 99.48-96.91% for bacteria and archaea, respectively. CONCLUSIONS Given the three amplicon length categories (100-300, 301-600, and >600 base pairs), the primer pairs with the best coverage values for detecting oral bacteria were as follows: KP_F048-OP_R043 (region 3-4; primer pair position for Escherichia coli J01859.1: 342-529), KP_F051-OP_R030 (4-7; 514-1079), and KP_F048-OP_R030 (3-7; 342-1079). For detecting oral archaea, these were as follows: OP_F066-KP_R013 (5-6; 784-undefined), KP_F020-KP_R013 (3-6; 518-undefined), and OP_F114-KP_R013 (3-6; 340-undefined). Lastly, for detecting both domains jointly they were KP_F020-KP_R032 (4-5; 518-801), OP_F114-KP_R031 (3-5; 340-801), and OP_F066-OP_R121 (5-9; 784-1405). The primer pairs with the best coverage identified herein are not among those described most widely in the oral microbiome literature. Video Abstract.
Collapse
Affiliation(s)
- Alba Regueira-Iglesias
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), C/ Entrerrios s/n, 15872 Santiago de Compostela, Spain
| | - Lara Vázquez-González
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Rúa de Jenaro de la Fuente, s/n, 15705 Santiago de Compostela, Spain
| | - Carlos Balsa-Castro
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), C/ Entrerrios s/n, 15872 Santiago de Compostela, Spain
| | - Nicolás Vila-Blanco
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Rúa de Jenaro de la Fuente, s/n, 15705 Santiago de Compostela, Spain
| | - Triana Blanco-Pintos
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), C/ Entrerrios s/n, 15872 Santiago de Compostela, Spain
| | - Javier Tamames
- Microbiome Analysis Laboratory, Systems Biology Department, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Maria José Carreira
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Rúa de Jenaro de la Fuente, s/n, 15705 Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), C/ Entrerrios s/n, 15872 Santiago de Compostela, Spain
| |
Collapse
|
28
|
Guo XJ, Dai SX, Lou JD, Ma XX, Hu XJ, Tu LP, Cui J, Lu H, Jiang T, Xu JT. Distribution characteristics of oral microbiota and its relationship with intestinal microbiota in patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1119201. [PMID: 37025407 PMCID: PMC10072265 DOI: 10.3389/fendo.2023.1119201] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/23/2023] [Indexed: 03/18/2023] Open
Abstract
Introduction Type 2 diabetes mellitus (T2DM) has a high incidence rate globally, increasing the burden of death, disability, and the economy worldwide. Previous studies have found that the compositions of oral and intestinal microbiota changed respectively in T2DM; whether the changes were associated or interacted between the two sites and whether there were some associations between T2DM and the ectopic colonization of oral microbiota in the gut still need to be identified. Research design and methods We performed a cross-sectional observational study; 183 diabetes and 74 controls were enrolled. We used high-throughput sequencing technology to detect the V3-V4 region of 16S rRNA in oral and stool samples. The Source Tracker method was used to identify the proportion of the intestinal microbiota that ectopic colonized from the oral cavity. Results The oral marker bacteria of T2DM were found, such as Actinobacteria, Streptococcus, Rothia, and the intestinal marker bacteria were Bifidobacterium, Streptococcus, and Blautia at the genus level. Among them, Actinobacteria and Blautia played a vital role in different symbiotic relationships of oral and intestinal microbiota. The commonly distributed bacteria, such as Firmicutes, Bacteroidetes, and Actinobacteria, were found in both oral and intestine. Moreover, the relative abundance and composition of bacteria were different between the two sites. The glycine betaine degradation I pathway was the significantly up-regulated pathway in the oral and intestinal flora of T2DM. The main serum indexes related to oral and intestinal flora were inflammatory. The relative abundance of Proteobacteria in the intestine and the Spirochete in oral was positively correlated, and the correlation coefficient was the highest, was 0.240 (P<0.01). The proportion of ectopic colonization of oral flora in the gut of T2DM was 2.36%. Conclusion The dysbacteriosis exited in the oral and intestine simultaneously, and there were differences and connections in the flora composition at the two sites in T2DM. Ectopic colonization of oral flora in the intestine might relate to T2DM. Further, clarifying the oral-gut-transmitting bacteria can provide an essential reference for diagnosing and treating T2DM in the future.
Collapse
Affiliation(s)
- Xiao-jing Guo
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Anesthesiology, Naval Medical University, Shanghai, China
| | - Shi-xuan Dai
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jin-di Lou
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu-xiang Ma
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-juan Hu
- Shanghai Collaborative Innovation Center of Health Service in Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-ping Tu
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji Cui
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hao Lu
- Department of Endocrinology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Jiang
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jia-tuo Xu
- School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
29
|
Zheng T, Wu Y, Guo KX, Tan ZJ, Yang T. The process of hypertension induced by high-salt diet: Association with interactions between intestinal mucosal microbiota, and chronic low-grade inflammation, end-organ damage. Front Microbiol 2023; 14:1123843. [PMID: 36925479 PMCID: PMC10011071 DOI: 10.3389/fmicb.2023.1123843] [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: 12/14/2022] [Accepted: 01/27/2023] [Indexed: 03/06/2023] Open
Abstract
Inflammation and immunity play a major role in the development of hypertension, and a potential correlation between host mucosal immunity and inflammatory response regulation. We explored the changes of intestinal mucosal microbiota in hypertensive rats induced by high-salt diet and the potential link between the intestinal mucosal microbiota and inflammation in rats. Therefore, we used PacBio (Pacific Bioscience) SMRT sequencing technology to determine the structure of intestinal mucosal microbiota, used enzyme-linked immunosorbent assay (ELISA) to determined the proinflammatory cytokines and hormones associated with hypertension in serum, and used histopathology methods to observe the kidney and vascular structure. We performed a potential association analysis between intestinal mucosal characteristic bacteria and significantly different blood cytokines in hypertensive rats induced by high-salt. The results showed that the kidney and vascular structures of hypertensive rats induced by high salt were damaged, the serum concentration of necrosis factor-α (TNF-α), angiotensin II (AngII), interleukin-6 (IL-6), and interleukin-8 (IL-8) were significantly increased (p < 0.05), and the coefficient of immune organ spleen was significantly changed (p < 0.05), but there was no significant change in serum lipids (p > 0.05). From the perspective of gut microbiota, high-salt diet leads to significant changes in intestinal mucosal microbiota. Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were the dominant differential bacteria in intestinal mucosal, with the AUC (area under curve) value of Bifidobacterium animalis subsp. and Brachybacterium paraconglomeratum were 1 and 0.875 according to ROC (receiver operating characteristic) analysis. Correlation analysis showed that Bifidobacterium animalis subsp. was correlated with IL-6, IL-8, TNF-α, and Ang II. Based on our results, we can speculated that high salt diet mediated chronic low-grade inflammation through inhibited the growth of Bifidobacterium animalis subsp. in intestinal mucosa and caused end-organ damage, which leads to hypertension.
Collapse
Affiliation(s)
- Tao Zheng
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Wu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Kang-xiao Guo
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| | - Zhou-jin Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Tao Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
30
|
Hiergeist A, Ruelle J, Emler S, Gessner A. Reliability of species detection in 16S microbiome analysis: Comparison of five widely used pipelines and recommendations for a more standardized approach. PLoS One 2023; 18:e0280870. [PMID: 36795699 PMCID: PMC9934417 DOI: 10.1371/journal.pone.0280870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 01/10/2023] [Indexed: 02/17/2023] Open
Abstract
The use of NGS-based testing of the bacterial microbiota is often impeded by inconsistent or non-reproducible results, especially when applying different analysis pipelines and reference databases. We investigated five frequently used software packages by submitting the same monobacterial datasets to them, representing the V1-2 and the V3-4 regions of the 16S-rRNA gene of 26 well characterized strains, which were sequenced by the Ion Torrent™ GeneStudio S5 system. The results obtained were divergent and calculations of relative abundance did not yield the expected 100%. We investigated these inconsistencies and were able to attribute them to failures either of the pipelines themselves or of the reference databases they rely on. On the basis of these findings, we recommend certain standards which should help to render microbiome testing more consistent and reproducible, and thus useful in clinical practice.
Collapse
Affiliation(s)
- Andreas Hiergeist
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
- * E-mail:
| | - Jean Ruelle
- SmartGene Services SARL, Lausanne, Switzerland
| | | | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
31
|
Regueira-Iglesias A, Vázquez-González L, Balsa-Castro C, Blanco-Pintos T, Vila-Blanco N, Carreira MJ, Tomás I. Impact of 16S rRNA Gene Redundancy and Primer Pair Selection on the Quantification and Classification of Oral Microbiota in Next-Generation Sequencing. Microbiol Spectr 2023; 11:e0439822. [PMID: 36779795 PMCID: PMC10101033 DOI: 10.1128/spectrum.04398-22] [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: 10/27/2022] [Accepted: 01/16/2023] [Indexed: 02/14/2023] Open
Abstract
This study aimed to evaluate the number of 16S rRNA genes in the complete genomes of the bacterial and archaeal species inhabiting the human mouth and to assess how the use of different primer pairs would affect the detection and classification of redundant amplicons and matching amplicons (MAs) from different taxa. A total of 518 oral-bacterial and 191 oral-archaeal complete genomes were downloaded from the NCBI database, and their complete 16S rRNA genes were extracted. The numbers of genes and variants per genome were calculated. Next, 39 primer pairs were used to search for matches in the genomes and obtain amplicons. For each primer, we calculated the number of gene amplicons, variants, genomes, and species detected and the percentage of coverage at the species level with no MAs (SC-NMA). The results showed that 94.09% of oral bacteria and 52.59% of oral archaea had more than one intragenomic 16S rRNA gene. From 1.29% to 46.70% of bacterial species and from 4.65% to 38.89% of archaea detected by the primers had MAs. The best primers were the following (SC-NMA; region; position for Escherichia coli [GenBank version no. J01859.1]): KP_F048-OP_R030 for bacteria (93.55%; V3 to V7; 342 to 1079), KP_F018-KP_R063 for archaea (89.63%; V3 to V9; undefined to 1506), and OP_F114-OP_R121 for both domains (92.52%; V3 to V9; 340 to 1405). In addition to 16S rRNA gene redundancy, the presence of MAs must be controlled to ensure an accurate interpretation of microbial diversity data. The SC-NMA is a more useful parameter than the conventional coverage percentage for selecting the best primer pairs. The pairs used the most in the oral microbiome literature were not among the best performers. IMPORTANCE Hundreds of publications have studied the oral microbiome through 16S rRNA gene sequencing. However, none have assessed the number of 16S rRNA genes in the genomes of oral microbes, or how the use of primer pairs targeting different regions affects the detection of MAs from different taxa. Here, we found that almost all oral bacteria and more than half of oral archaea have more than one intragenomic 16S rRNA gene. The performance of the primer pairs in not detecting MAs increases as the length of the amplicon augments. As none of those most employed in the oral literature were among the best performers, we selected a series of primers to detect bacteria and/or archaea based on their percentage of species detected without MAs. The intragenomic 16S rRNA gene redundancy and the presence of MAs between distinct taxa need to be considered to ensure an accurate interpretation of microbial diversity data.
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, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Lara Vázquez-González
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), 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, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), 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, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Nicolás Vila-Blanco
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Maria José Carreira
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), 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, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| |
Collapse
|
32
|
Blostein F, Bhaumik D, Davis E, Salzman E, Shedden K, Duhaime M, Bakulski KM, McNeil DW, Marazita ML, Foxman B. Evaluating the ecological hypothesis: early life salivary microbiome assembly predicts dental caries in a longitudinal case-control study. MICROBIOME 2022; 10:240. [PMID: 36567334 PMCID: PMC9791751 DOI: 10.1186/s40168-022-01442-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 12/01/2022] [Indexed: 05/09/2023]
Abstract
BACKGROUND Early childhood caries (ECC)-dental caries (cavities) occurring in primary teeth up to age 6 years-is a prevalent childhood oral disease with a microbial etiology. Streptococcus mutans was previously considered a primary cause, but recent research promotes the ecologic hypothesis, in which a dysbiosis in the oral microbial community leads to caries. In this incident, density sampled case-control study of 189 children followed from 2 months to 5 years, we use the salivary bacteriome to (1) prospectively test the ecological hypothesis of ECC in salivary bacteriome communities and (2) identify co-occurring salivary bacterial communities predicting future ECC. RESULTS Supervised classification of future ECC case status using salivary samples from age 12 months using bacteriome-wide data (AUC-ROC 0.78 95% CI (0.71-0.85)) predicts future ECC status before S. mutans can be detected. Dirichlet multinomial community state typing and co-occurrence network analysis identified similar robust and replicable groups of co-occurring taxa. Mean relative abundance of a Haemophilus parainfluenzae/Neisseria/Fusobacterium periodonticum group was lower in future ECC cases (0.14) than controls (0.23, P value < 0.001) in pre-incident visits, positively correlated with saliva pH (Pearson rho = 0.33, P value < 0.001) and reduced in individuals who had acquired S. mutans by the next study visit (0.13) versus those who did not (0.20, P value < 0.01). In a subset of whole genome shotgun sequenced samples (n = 30), case plaque had higher abundances of antibiotic production and resistance gene orthologs, including a major facilitator superfamily multidrug resistance transporter (MFS DHA2 family PBH value = 1.9 × 10-28), lantibiotic transport system permease protein (PBH value = 6.0 × 10-6) and bacitracin synthase I (PBH value = 5.6 × 10-6). The oxidative phosphorylation KEGG pathway was enriched in case plaque (PBH value = 1.2 × 10-8), while the ABC transporter pathway was depleted (PBH value = 3.6 × 10-3). CONCLUSIONS Early-life bacterial interactions predisposed children to ECC, supporting a time-dependent interpretation of the ecological hypothesis. Bacterial communities which assemble before 12 months of age can promote or inhibit an ecological succession to S. mutans dominance and cariogenesis. Intragenera competitions and intergenera cooperation between oral taxa may shape the emergence of these communities, providing points for preventive interventions. Video Abstract.
Collapse
Affiliation(s)
- Freida Blostein
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Deesha Bhaumik
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Elyse Davis
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Elizabeth Salzman
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Kerby Shedden
- Department of Statistics, University of Michigan, Ann Arbor, MI USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Melissa Duhaime
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, MI USA
| | - Kelly M. Bakulski
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| | - Daniel W. McNeil
- Department of Psychology, West Virginia University, WVA, Morgantown, USA
- Department of Dental Practice & Rural Health, West Virginia University, Morgantown, WV USA
| | - Mary L. Marazita
- Department of Oral and Craniofacial Sciences, Center for Craniofacial and Dental Genetics, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA USA
- Clinical and Translational Sciences Institute, and Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA USA
| | - Betsy Foxman
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI USA
| |
Collapse
|
33
|
Gómez-García AP, López-Vidal Y, Pinto-Cardoso S, Aguirre-García MM. Overexpression of proinflammatory cytokines in dental pulp tissue and distinct bacterial microbiota in carious teeth of Mexican Individuals. Front Cell Infect Microbiol 2022; 12:958722. [PMID: 36569197 PMCID: PMC9772992 DOI: 10.3389/fcimb.2022.958722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
The prevalence of dental caries in the Mexican adult population aged 20 to 85 years is around 93.3%, and 50% in Mexican children and adolescents. Worldwide, it is the most common non-communicable disease. One of the main etiological factors for dental caries is the oral microbiome and changes in its structure and function, with an expansion of pathogenic bacteria like Streptococcus mutans. The exposed dental pulp tissue triggers an innate immune response to counteract this bacterial invasion. The relation between oral dysbiosis and innate immune responses remains unclear. We aimed to understand the relationship between innate immune response and the oral microbiota by quantifying the expression of Toll-like receptors (TLRs) and proinflammatory markers (cytokines and a chemokine) in dental pulp tissue, either exposed or not to carious dentin, and to correlate this information with the oral microbiome found in healthy teeth and those with moderate caries. RNA was purified from pulp tissue, subjected to RT-qPCR and analysed with the ΔΔCt method. Supragingival dental plaque of non-carious teeth and dentin of carious teeth were subjected to 16S targeted sequencing. Principal coordinate analysis, permutational multivariate ANOVA, and linear discriminant analysis were used to assess differences between non-carious and carious teeth. Correlations were assessed with Spearman´s test and corrected for multiple comparisons using the FDR method. The relative abundance (RA) of Lactobacillus, Actinomyces, Prevotella, and Mitsuokella was increased in carious teeth; while the RA of Haemophilus and Porphyromonas decreased. Olsenella and Parascardovia were only detected in carious teeth. Significant overexpression of interleukin 1 beta (IL1 β), IL6, and CXCL8 was detected in pulp tissue exposed to carious dentin. IL1β correlated positively with TLR2 and Actinomyces; yet negatively with Porphyromonas. These findings suggest that immune response of pulp tissue chronically exposed to cariogenic microbiome is triggered by proinflammatory cytokines IL1β and IL6 and the chemokine CXCL8.
Collapse
Affiliation(s)
- Ana Pamela Gómez-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
| | - Yolanda López-Vidal
- Programa de Inmunología Molecular Microbiana, Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de Mexico, Ciudad de México, México
| | - Sandra Pinto-Cardoso
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - María Magdalena Aguirre-García
- Unidad de Investigación UNAM-INC, División de Investigación, Facultad de Medicina, UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México,*Correspondence: María Magdalena Aguirre-García,
| |
Collapse
|
34
|
Tricarico PM, Gratton R, dos Santos-Silva CA, de Moura RR, Ura B, Sommella E, Campiglia P, Del Vecchio C, Moltrasio C, Berti I, D’Adamo AP, Elsherbini AMA, Staudenmaier L, Chersi K, Boniotto M, Krismer B, Schittek B, Crovella S. A rare loss-of-function genetic mutation suggest a role of dermcidin deficiency in hidradenitis suppurativa pathogenesis. Front Immunol 2022; 13:1060547. [PMID: 36544771 PMCID: PMC9760663 DOI: 10.3389/fimmu.2022.1060547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/16/2022] [Indexed: 12/11/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic inflammatory skin disease with a multifactorial aetiology that involves a strict interplay between genetic factors, immune dysregulation and lifestyle. Familial forms represent around 40% of total HS cases and show an autosomal dominant mode of inheritance of the disease. In this study, we conducted a whole-exome sequence analysis on an Italian family of 4 members encompassing a vertical transmission of HS. Focusing on rare damaging variants, we identified a rare insertion of one nucleotide (c.225dupA:p.A76Sfs*21) in the DCD gene encoding for the antimicrobial peptide dermcidin (DCD) that was shared by the proband, his affected father and his 11-years old daughter. Since several transcriptome studies have shown a significantly decreased expression of DCD in HS skin, we hypothesised that the identified frameshift insertion was a loss-of-function mutation that might be associated with HS susceptibility in this family. We thus confirmed by mass spectrometry that DCD levels were diminished in the affected members and showed that the antimicrobial activity of a synthetic DCD peptide resulting from the frameshift mutation was impaired. In order to define the consequences related to a decrease in DCD activity, skin microbiome analyses of different body sites were performed by comparing DCD mutant and wild type samples, and results highlighted significant differences between the groins of mutated and wild type groups. Starting from genetic analysis conducted on an HS family, our findings showed, confirming previous transcriptome results, the potential role of the antimicrobial DCD peptide as an actor playing a crucial part in the etio-pathogenesis of HS and in the maintenance of the skin's physiological microbiome composition; so, we can hypothesise that DCD could be used as a novel target for personalised therapeutic approach.
Collapse
Affiliation(s)
- Paola Maura Tricarico
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Rossella Gratton
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | | | - Ronald Rodrigues de Moura
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Blendi Ura
- Maternal-Neonatal Department, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | | | | | - Cecilia Del Vecchio
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Chiara Moltrasio
- Dermatology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Irene Berti
- Pediatric Department, Institute of Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Adamo Pio D’Adamo
- Department of Advanced Diagnostics, Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Ahmed M. A. Elsherbini
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Lena Staudenmaier
- Department of Dermatology, Division of Dermato-oncology, University of Tübingen, Tübingen, Germany
| | - Karin Chersi
- Dermatological Clinic, ASUGI - Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Michele Boniotto
- INSERM, IMRB, Translational Neuropsychiatry, University Paris Est Créteil, Créteil, France
| | - Bernhard Krismer
- Department of Infection Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Birgit Schittek
- Department of Dermatology, Division of Dermato-oncology, University of Tübingen, Tübingen, Germany
| | - Sergio Crovella
- Department of Biological and Environmental Sciences, Biological Sciences Program, College of Arts and Sciences, University of Qatar, Doha, Qatar
| |
Collapse
|
35
|
Huang X, Chen X, Gong X, Xu Y, Xu Z, Gao X. Characteristics of salivary microbiota in children with obstructive sleep apnea: A prospective study with polysomnography. Front Cell Infect Microbiol 2022; 12:945284. [PMID: 36105146 PMCID: PMC9465092 DOI: 10.3389/fcimb.2022.945284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectivesThe present study aimed to investigate the characteristics of salivary microbiota of children with obstructive sleep apnea (OSA) and to assess longitudinal alterations in salivary microbiota before and after adenotonsillectomy.MethodsA set of cross-sectional samples consisted of 36 OSA children (17 boys and 19 girls, 7.47 ± 2.24 years old) and 22 controls (9 boys and 13 girls, 7.55 ± 2.48 years old) were included in the study, among which eight OSA children (five boys and three girls, 8.8 ± 2.0 years old) who underwent treatment of adenotonsillectomy were followed up after 1 year. Saliva samples were collected, and microbial profiles were analyzed by bioinformatics analysis based on 16S rRNA sequencing.ResultsIn cross-sectional samples, the OSA group had higher α-diversity as estimated by Chao1, Shannon, Simpson, Pielou_e, and observed species as compared with the control group (p < 0.05). β-Diversity based on the Bray–Curtis dissimilarities (p = 0.004) and Jaccard distances (p = 0.001) revealed a significant separation between the OSA group and control group. Nested cross-validated random forest classifier identified the 10 most important genera (Lactobacillus, Escherichia, Bifidobacterium, Capnocytophaga, Bacteroidetes_[G-7], Parvimonas, Bacteroides, Klebsiella, Lautropia, and Prevotella) that could differentiate OSA children from controls with an area under the curve (AUC) of 0.94. Linear discriminant analysis effect size (LEfSe) analysis revealed a significantly higher abundance of genera such as Prevotella (p = 0.027), Actinomyces (p = 0.015), Bifidobacterium (p < 0.001), Escherichia (p < 0.001), and Lactobacillus (p < 0.001) in the OSA group, among which Prevotella was further corroborated in longitudinal samples. Prevotella sp_HMT_396 was found to be significantly enriched in the OSA group (p = 0.02) with significantly higher levels as OSA severity increased (p = 0.014), and it had a lower abundance in the post-treatment group (p = 0.003) with a decline in each OSA child 1 year after adenotonsillectomy.ConclusionsA significantly higher microbial diversity and a significant difference in microbial composition and abundance were identified in salivary microbiota of OSA children compared with controls. Meanwhile, some characteristic genera (Prevotella, Actinomyces, Lactobacillus, Escherichia, and Bifidobacterium) were found in OSA children, among which the relationship between Prevotella spp. and OSA is worth further studies.
Collapse
Affiliation(s)
- Xin Huang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xuehui Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xu Gong
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Ying Xu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
| | - Zhifei Xu
- Department of Respiratory Medicine, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- *Correspondence: Xuemei Gao,
| | - Xuemei Gao
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- *Correspondence: Xuemei Gao,
| |
Collapse
|
36
|
Effect of a Stannous Fluoride Dentifrice on Biofilm Composition, Gene Expression and Biomechanical Properties. Microorganisms 2022; 10:microorganisms10091691. [PMID: 36144293 PMCID: PMC9506307 DOI: 10.3390/microorganisms10091691] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
An in situ study was conducted to examine the mode of action of a 0.454% stannous fluoride (SnF2)-containing dentifrice in controlling the composition and properties of oral biofilm. Thirteen generally healthy individuals participated in the study. Each participant wore an intra-oral appliance over a 48-h period to measure differences in the resulting biofilm’s architecture, mechanical properties, and bacterial composition after using two different toothpaste products. In addition, metatranscriptomics analysis of supragingival plaque was conducted to identify the gene pathways influenced. The thickness and volume of the microcolonies formed when brushing with the SnF2 dentifrice were dramatically reduced compared to the control 0.76% sodium monofluorophosphate (MFP)-containing toothpaste. Similarly, the biophysical and nanomechanical properties measured by atomic force microscopy (AFM) demonstrated a significant reduction in biofilm adhesive properties. Metatranscriptomic analysis identified pathways associated with biofilm formation, cell adhesion, quorum sensing, and N-glycosylation that are significantly downregulated with SnF2. This study provides a clinically relevant snapshot of how the use of a stabilized, SnF2 toothpaste formulation can change the spatial organization, nanomechanical, and gene expression properties of bacterial communities.
Collapse
|
37
|
Zheng T, Wu Y, Peng MJ, Xiao NQ, Tan ZJ, Yang T. Hypertension of liver-yang hyperactivity syndrome induced by a high salt diet by altering components of the gut microbiota associated with the glutamate/GABA-glutamine cycle. Front Nutr 2022; 9:964273. [PMID: 36017217 PMCID: PMC9395663 DOI: 10.3389/fnut.2022.964273] [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: 06/08/2022] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
The gut microbiota and metabolites are closely related to hypertension; however, the changes in the composition of the gut microbiome and metabolites linking a high salt diet to elevated blood pressure are not established. In this study, traditional Chinese medicine (TCM) syndrome of hypertension caused by high salt had been diagnosed and the pathogenesis of hypertension was explored from the perspective of intestinal microecology. Rats in a high salt diet-induced hypertension group (CG) and normal group (CZ) were compared by 16S rRNA gene full-length sequencing and liquid chromatography and mass spectrometry to identify differences in the bacterial community structure, metabolites, and metabolic pathways. Hypertension induced by a high salt diet belongs to liver-Yang hyperactivity syndrome. Alpha and beta diversity as well as the composition of microbiota from the phylum to species levels differed substantially between the CG and CZ groups. In an analysis of differential metabolites in the intestines, a high salt diet mainly affected the metabolism of amino acids and their derivatives; in particular, γ-aminobutyric acid (GABA) was down-regulated and glutamic acid and its derivatives were up-regulated under a high salt diet. Based on a KEGG analysis, high salt intake mainly altered pathways related to GABA and the glutamate/glutamine metabolism, such as the GABAergic synapse pathway and glutamatergic synapse pathway. The correlation analysis of differential gut microbes and differential metabolites suggested that a high salt diet promoted hypertension via the inhibition of Clostridiaceae_1 growth and alterations in the GABA metabolic pathway, leading to increased blood pressure. These findings suggest that a high salt diet induces hypertension of liver-Yang hyperactivity syndrome by mediating the microbiota associated with the glutamate/GABA-glutamine metabolic cycle via the gut-brain axis.
Collapse
Affiliation(s)
- Tao Zheng
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Yi Wu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Mai-jiao Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Nen-qun Xiao
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Zhou-jin Tan
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Tao Yang
- College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, China
| |
Collapse
|
38
|
Gao C, Li X, Zhao X, Yang P, Wang X, Chen X, Chen N, Chen F. Standardized studies of the oral microbiome: From technology-driven to hypothesis-driven. IMETA 2022; 1:e19. [PMID: 38868569 PMCID: PMC10989927 DOI: 10.1002/imt2.19] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2024]
Abstract
The microbiome is in a symbiotic relationship with the host. Among the microbial consortia in the human body, that in the oral cavity is complex. Instead of repeatedly confirming biomarkers of oral and systemic diseases, recent studies have focused on a unified clinical diagnostic standard in microbiology that reduces the heterogeneity caused by individual discrepancies. Research has also been conducted on other topics of greater clinical importance, including bacterial pathogenesis, and the effects of drugs and treatments. In this review, we divide existing research into technology-driven and hypothesis-driven, according to whether there is a clear research hypothesis. This classification allows the demonstration of shifts in the direction of oral microbiology research. Based on the shifts, we suggested that establishing clear hypotheses may be the solution to major research challenges.
Collapse
Affiliation(s)
- Chuqi Gao
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xuantao Li
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiaole Zhao
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Peiyue Yang
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiao Wang
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiaoli Chen
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Ning Chen
- Department of GastroenterologyPeking University People's HospitalBeijingChina
| | - Feng Chen
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| |
Collapse
|
39
|
Liao CC, Fu PY, Huang CW, Chuang CH, Yen Y, Lin CY, Chen SH. MetaSquare: an integrated metadatabase of 16S rRNA gene amplicon for microbiome taxonomic classification. Bioinformatics 2022; 38:2930-2931. [PMID: 35561196 PMCID: PMC9113242 DOI: 10.1093/bioinformatics/btac184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 02/18/2022] [Accepted: 03/22/2022] [Indexed: 11/21/2022] Open
Abstract
MOTIVATION Taxonomic classification of 16S ribosomal RNA gene amplicon is an efficient and economic approach in microbiome analysis. 16S rRNA sequence databases like SILVA, RDP, EzBioCloud and HOMD used in downstream bioinformatic pipelines have limitations on either the sequence redundancy or the delay on new sequence recruitment. To improve the 16S rRNA gene-based taxonomic classification, we merged these widely used databases and a collection of novel sequences systemically into an integrated resource. RESULTS MetaSquare version 1.0 is an integrated 16S rRNA sequence database. It is composed of more than 6 million sequences and improves taxonomic classification resolution on both long-read and short-read methods. AVAILABILITY AND IMPLEMENTATION Accessible at https://hub.docker.com/r/lsbnb/metasquare_db and https://github.com/lsbnb/MetaSquare. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Chun-Chieh Liao
- Institute of Information Science, Academia Sinica, 115 Taipei, Taiwan
| | - Po-Ying Fu
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chih-Wei Huang
- Institute of Information Science, Academia Sinica, 115 Taipei, Taiwan
| | - Chia-Hsien Chuang
- Institute of Information Science, Academia Sinica, 115 Taipei, Taiwan
| | - Yun Yen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 110 Taipei, Taiwan
| | | | | |
Collapse
|
40
|
Andersen C, Bergholt B, Ridderberg W, Nørskov-Lauritsen N. Culture on Selective Media and Amplicon-Based Sequencing of 16S rRNA from Spontaneous Brain Abscess-the View from the Diagnostic Laboratory. Microbiol Spectr 2022; 10:e0240721. [PMID: 35404098 PMCID: PMC9045185 DOI: 10.1128/spectrum.02407-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/10/2022] [Indexed: 11/20/2022] Open
Abstract
Forty-one stored samples from cases of spontaneous brain abscess were investigated to gain insight into the natural history, causative agents, and relevant laboratory diagnostics of a rare infection. Samples from a larger collection were selected based on retrospective analysis of patient records. All samples were subjected to amplicon sequencing of 16S rRNA gene fragments. Supplementary culture on selected media was performed as suggested by bioinformatics analysis. For three cases, no microorganism was disclosed, while Toxoplasma gondii, Aspergillus fumigatus, and various bacteria were the cause of 1, 2, and 35 cases, respectively. Bacterial infections were monomicrobial in 20 cases and polymicrobial in 15; the microorganisms of the latter cases were restricted to residents of cavum oris. Amplicon sequencing did not further enhance the importance of the Streptococcus anginosus group, which was involved in 17 cases, and the single primer set used may be suboptimal for amplification of Actinomyces and Nocardia. But, amplicon-based sequencing unquestionably expanded the number of polybacterial infections, with focus on the Fusobacterium nucleatum group, Parvimonas, and Porphyromonas. Culture on selective media confirmed the presence of F. nucleatum group bacteria, which attained a prominence in spontaneous brain abscess similar to the S. anginosus group. Metagenomics is a powerful tool to disclose the spectrum of agents in polymicrobial infections, but a reliable cutoff value for substantial detection is complex. Commercial media for isolation of F. nucleatum group bacteria from mixed infections are available, and these pathogens should be carefully characterized. Isolation of Parvimonas and Porphyromonas in polymicrobial infections has not been resolved. IMPORTANCE Polymicrobial brain abscess is a challenge to the clinical microbiology laboratory due to the aggregative nature of the dental and oral microbiota. Because polymicrobial infections may escape detection by conventional culture methods, directed therapy toward a single detected bacterium is problematic. Amplicon-based sequencing provides important clues to these infections, but only cultured microorganisms can be fully characterized, subjected to antimicrobial susceptibility testing, and formally named. By use of specific selective culture plates, we successfully isolated bacteria of the Fusobacterium nucleatum group, and these bacteria rose to the same prominence as the widely recognized pathogen, the Streptococcus anginosus group. Named and unnamed members of the Fusobacterium nucleatum group must be further investigated to gain insight into a rare but grave disease.
Collapse
Affiliation(s)
- Camilla Andersen
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus, Denmark
| | - Bo Bergholt
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Winnie Ridderberg
- Department of Clinical Microbiology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
41
|
Silva DNDA, Casarin M, Monajemzadeh S, Bezerra BDB, Lux R, Pirih FQ. The Microbiome in Periodontitis and Diabetes. FRONTIERS IN ORAL HEALTH 2022; 3:859209. [PMID: 35464780 PMCID: PMC9024052 DOI: 10.3389/froh.2022.859209] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
Objectives To perform a comprehensive and integrative review of the available literature on the potential changes in the microbiome of healthy and individuals with diabetes under periodontal health and disease. Materials and Methods The review was conducted by two independent reviewers. Indexed electronic databases (PubMed/Medline, Cochrane Library, Web of Science and Scopus) were searched, including articles published in English and dated from 5 years ago until December 2021. A manual search also was performed to identify co-related articles. Following the removal of duplicates and eligibility criteria, the articles were included in tables for analysis and described in the manuscript. Results According to this review, diabetes mellitus was associated with significant changes in the subgingival and salivary microbiome, either in its association with periodontitis or in cases of periodontal health. In addition to affecting microbial diversity in terms of taxonomy, metagenomic studies have shown that this endocrine disorder may also be directly related to increased pathogenicity in the oral microbiome. Conclusion Although the reviewed studies demonstrate important differences in the subgingival and salivary microbiome composition because of diabetes mellitus, further studies are needed to clarify the real effects of hyperglycemia on oral microbial profiles and support new diagnostic approaches and innovative treatments.
Collapse
Affiliation(s)
- Davi Neto de Araújo Silva
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maísa Casarin
- School of Dentistry, Federal University of Pelotas, Pelotas, Brazil
| | - Sepehr Monajemzadeh
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Beatriz de Brito Bezerra
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Renate Lux
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Flavia Q Pirih
- Section of Periodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
42
|
Guo XJ, Jiang T, Ma XX, Hu XJ, Huang JB, Cui LT, Cui J, Yao XH, Shi YL, Li J, Guo ZL, Lou JD, Liang MC, Fu HY, Yuan P, Liu JY, Tu LP, Xu JT. Relationships Between Diurnal Changes of Tongue Coating Microbiota and Intestinal Microbiota. Front Cell Infect Microbiol 2022; 12:813790. [PMID: 35433494 PMCID: PMC9008461 DOI: 10.3389/fcimb.2022.813790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
The oral cavity and the intestine are the main distribution locations of human digestive bacteria. Exploring the relationships between the tongue coating and gut microbiota, the influence of the diurnal variations of the tongue coating microbiota on the intestinal microbiota can provide a reference for the development of the disease diagnosis and monitoring, as well as the medication time. In this work, a total of 39 healthy college students were recruited. We collected their tongue coating microbiota which was collected before and after sleep and fecal microbiota. The diurnal variations of tongue coating microbiota are mainly manifested on the changes in diversity and relative abundance. There are commensal bacteria in the tongue coating and intestines, especially Prevotella which has the higher proportion in both sites. The relative abundance of Prevotella in the tongue coating before sleep has a positive correlation with intestinal Prevotella; the r is 0.322 (p < 0.05). Bacteroides in the intestine had the most bacteria associated with the tongue coating and had the highest correlation coefficient with Veillonella in the oral cavity, which was 0.468 (p < 0.01). These results suggest that the abundance of the same flora in the two sites may have a common change trend. The SourceTracker results show that the proportion of intestinal bacteria sourced from tongue coating is less than 1%. It indicates that oral flora is difficult to colonize in the intestine in healthy people. This will provide a reference for the study on the oral and intestinal microbiota in diseases.
Collapse
Affiliation(s)
- Xiao-jing Guo
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Tao Jiang
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Xu-xiang Ma
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Xiao-juan Hu
- Shanghai Collaborative Innovation Center of Health Service in Traditional Chinese Medicine (TCM), Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jing-bin Huang
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Long-tao Cui
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Ji Cui
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Xing-hua Yao
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Yu-lin Shi
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jun Li
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Zhi-ling Guo
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jin-di Lou
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Meng-chen Liang
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Hong-yuan Fu
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Pei Yuan
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Jia-yi Liu
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
| | - Li-ping Tu
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
- *Correspondence: Li-ping Tu, ; Jia-tuo Xu,
| | - Jia-tuo Xu
- Basic Medical College, Shanghai University of Traditional Chinese Medicine (TCM), Shanghai, China
- *Correspondence: Li-ping Tu, ; Jia-tuo Xu,
| |
Collapse
|
43
|
Hurst JH, McCumber AW, Aquino JN, Rodriguez J, Heston SM, Lugo DJ, Rotta AT, Turner NA, Pfeiffer TS, Gurley TC, Moody MA, Denny TN, Rawls JF, Clark JS, Woods CW, Kelly MS. Age-Related Changes in the Nasopharyngeal Microbiome Are Associated With Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection and Symptoms Among Children, Adolescents, and Young Adults. Clin Infect Dis 2022; 75:e928-e937. [PMID: 35247047 PMCID: PMC8903463 DOI: 10.1093/cid/ciac184] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Children are less susceptible to SARS-CoV-2 infection and typically have milder illness courses than adults, but the factors underlying these age-associated differences are not well understood. The upper respiratory microbiome undergoes substantial shifts during childhood and is increasingly recognized to influence host defense against respiratory pathogens. Thus, we sought to identify upper respiratory microbiome features associated with SARS-CoV-2 infection susceptibility and illness severity. METHODS We collected clinical data and nasopharyngeal swabs from 285 children, adolescents, and young adults (<21 years) with documented SARS-CoV-2 exposure. We used 16S ribosomal RNA gene sequencing to characterize the nasopharyngeal microbiome and evaluated for age-adjusted associations between microbiome characteristics and SARS-CoV-2 infection status and respiratory symptoms. RESULTS Nasopharyngeal microbiome composition varied with age (PERMANOVA, P < .001; R2 = 0.06) and between SARS-CoV-2-infected individuals with and without respiratory symptoms (PERMANOVA, P = .002; R2 = 0.009). SARS-CoV-2-infected participants with Corynebacterium/Dolosigranulum-dominant microbiome profiles were less likely to have respiratory symptoms than infected participants with other nasopharyngeal microbiome profiles (OR: .38; 95% CI: .18-.81). Using generalized joint attributed modeling, we identified 9 bacterial taxa associated with SARS-CoV-2 infection and 6 taxa differentially abundant among SARS-CoV-2-infected participants with respiratory symptoms; the magnitude of these associations was strongly influenced by age. CONCLUSIONS We identified interactive relationships between age and specific nasopharyngeal microbiome features that are associated with SARS-CoV-2 infection susceptibility and symptoms in children, adolescents, and young adults. Our data suggest that the upper respiratory microbiome may be a mechanism by which age influences SARS-CoV-2 susceptibility and illness severity.
Collapse
Affiliation(s)
| | | | - Jhoanna N Aquino
- Division of Infectious Diseases, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Javier Rodriguez
- Children’s Clinical Research Unit, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sarah M Heston
- Division of Infectious Diseases, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Debra J Lugo
- Division of Infectious Diseases, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alexandre T Rotta
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Nicholas A Turner
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Trevor S Pfeiffer
- Division of Infectious Diseases, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Thaddeus C Gurley
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - M Anthony Moody
- Division of Infectious Diseases, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina, USA,Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, USAand
| | - James S Clark
- Nicholas School of the Environment, Duke University, Durham, North Carolina, USA
| | - Christopher W Woods
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA,Duke Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Matthew S Kelly
- Correspondence: M. S. Kelly, 2301 Erwin Road, Durham, NC 27710 USA ()
| |
Collapse
|
44
|
Kelly MS, Plunkett C, Yu Y, Aquino JN, Patel SM, Hurst JH, Young RR, Smieja M, Steenhoff AP, Arscott-Mills T, Feemster KA, Boiditswe S, Leburu T, Mazhani T, Patel MZ, Rawls JF, Jawahar J, Shah SS, Polage CR, Cunningham CK, Seed PC. Non-diphtheriae Corynebacterium species are associated with decreased risk of pneumococcal colonization during infancy. THE ISME JOURNAL 2022; 16:655-665. [PMID: 34511605 PMCID: PMC8857224 DOI: 10.1038/s41396-021-01108-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/22/2021] [Accepted: 09/03/2021] [Indexed: 02/08/2023]
Abstract
Streptococcus pneumoniae (pneumococcus) is a leading cause of severe infections among children and adults. Interactions between commensal microbes in the upper respiratory tract and S. pneumoniae are poorly described. In this study, we sought to identify interspecies interactions that modify the risk of S. pneumoniae colonization during infancy and to describe development of the upper respiratory microbiome during infancy in a sub-Saharan African setting. We collected nasopharyngeal swabs monthly (0-6 months of age) or bimonthly (6-12 months of age) from 179 mother-infant dyads in Botswana. We used 16S ribosomal RNA gene sequencing to characterize the nasopharyngeal microbiome and identified S. pneumoniae colonization using a species-specific PCR assay. We detect S. pneumoniae colonization in 144 (80%) infants at a median age of 71 days and identify a strong negative association between the relative abundance of the bacterial genera Corynebacterium within the infant nasopharyngeal microbiome and the risk of S. pneumoniae colonization. Using in vitro cultivation experiments, we demonstrate growth inhibition of S. pneumoniae by secreted factors from strains of several Corynebacterium species isolated from these infants. Finally, we demonstrate that antibiotic exposures and the winter season are associated with a decline in the relative abundance of Corynebacterium within the nasopharyngeal microbiome, while breastfeeding is associated with an increase in the Corynebacterium relative abundance. Our findings provide novel insights into the interspecies interactions that contribute to colonization resistance to S. pneumoniae and suggest that the nasopharyngeal microbiome may be a previously unrecognized mechanism by which environmental factors influence the risk of pneumococcal infections during childhood. Moreover, this work lays the foundation for future studies seeking to use targeted manipulation of the nasopharyngeal microbiome to prevent infections caused by S. pneumoniae.
Collapse
Affiliation(s)
- Matthew S. Kelly
- grid.7621.20000 0004 0635 5486Botswana-University of Pennsylvania Partnership, Gaborone, Botswana ,grid.26009.3d0000 0004 1936 7961Division of Pediatric Infectious Diseases, Duke University, Durham, NC USA
| | - Catherine Plunkett
- grid.16753.360000 0001 2299 3507Division of Pediatric Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| | - Yahe Yu
- grid.40803.3f0000 0001 2173 6074Department of Mathematics, North Carolina State University, Raleigh, NC USA
| | - Jhoanna N. Aquino
- grid.26009.3d0000 0004 1936 7961Division of Pediatric Infectious Diseases, Duke University, Durham, NC USA
| | - Sweta M. Patel
- grid.26009.3d0000 0004 1936 7961Division of Pulmonary Allergy, and Critical Care Medicine, Duke University, Durham, NC USA
| | - Jillian H. Hurst
- grid.26009.3d0000 0004 1936 7961Division of Pediatric Infectious Diseases, Duke University, Durham, NC USA
| | - Rebecca R. Young
- grid.26009.3d0000 0004 1936 7961Division of Pediatric Infectious Diseases, Duke University, Durham, NC USA
| | - Marek Smieja
- grid.25073.330000 0004 1936 8227Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON Canada
| | - Andrew P. Steenhoff
- grid.7621.20000 0004 0635 5486Botswana-University of Pennsylvania Partnership, Gaborone, Botswana ,grid.239552.a0000 0001 0680 8770Global Health Center, Children’s Hospital of Philadelphia, Philadelphia, PA USA ,grid.239552.a0000 0001 0680 8770Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Tonya Arscott-Mills
- grid.7621.20000 0004 0635 5486Botswana-University of Pennsylvania Partnership, Gaborone, Botswana ,grid.239552.a0000 0001 0680 8770Global Health Center, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Kristen A. Feemster
- grid.239552.a0000 0001 0680 8770Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA USA
| | - Sefelani Boiditswe
- grid.7621.20000 0004 0635 5486Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Tirayaone Leburu
- grid.7621.20000 0004 0635 5486Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
| | - Tiny Mazhani
- grid.7621.20000 0004 0635 5486University of Botswana School of Medicine, Gaborone, Botswana
| | - Mohamed Z. Patel
- grid.7621.20000 0004 0635 5486University of Botswana School of Medicine, Gaborone, Botswana
| | - John F. Rawls
- grid.26009.3d0000 0004 1936 7961Department of Molecular Genetics and Microbiology, Duke University, Durham, NC USA
| | - Jayanth Jawahar
- grid.26009.3d0000 0004 1936 7961Department of Molecular Genetics and Microbiology, Duke University, Durham, NC USA
| | - Samir S. Shah
- grid.239573.90000 0000 9025 8099Divisions of Hospital Medicine and Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH USA
| | - Christopher R. Polage
- grid.26009.3d0000 0004 1936 7961Department of Pathology, Duke University, Durham, NC USA
| | - Coleen K. Cunningham
- grid.26009.3d0000 0004 1936 7961Division of Pediatric Infectious Diseases, Duke University, Durham, NC USA
| | - Patrick C. Seed
- grid.16753.360000 0001 2299 3507Division of Pediatric Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL USA
| |
Collapse
|
45
|
Regueira-Iglesias A, Vázquez-González L, Balsa-Castro C, Blanco-Pintos T, Martín-Biedma B, Arce VM, Carreira MJ, Tomás I. In-Silico Detection of Oral Prokaryotic Species With Highly Similar 16S rRNA Sequence Segments Using Different Primer Pairs. Front Cell Infect Microbiol 2022; 11:770668. [PMID: 35223533 PMCID: PMC8863748 DOI: 10.3389/fcimb.2021.770668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/31/2021] [Indexed: 11/13/2022] Open
Abstract
Although clustering by operational taxonomic units (OTUs) is widely used in the oral microbial literature, no research has specifically evaluated the extent of the limitations of this sequence clustering-based method in the oral microbiome. Consequently, our objectives were to: 1) evaluate in-silico the coverage of a set of previously selected primer pairs to detect oral species having 16S rRNA sequence segments with ≥97% similarity; 2) describe oral species with highly similar sequence segments and determine whether they belong to distinct genera or other higher taxonomic ranks. Thirty-nine primer pairs were employed to obtain the in-silico amplicons from the complete genomes of 186 bacterial and 135 archaeal species. Each fasta file for the same primer pair was inserted as subject and query in BLASTN for obtaining the similarity percentage between amplicons belonging to different oral species. Amplicons with 100% alignment coverage of the query sequences and with an amplicon similarity value ≥97% (ASI97) were selected. For each primer, the species coverage with no ASI97 (SC-NASI97) was calculated. Based on the SC-NASI97 parameter, the best primer pairs were OP_F053-KP_R020 for bacteria (region V1-V3; primer pair position for Escherichia coli J01859.1: 9-356); KP_F018-KP_R002 for archaea (V4; undefined-532); and OP_F114-KP_R031 for both (V3-V5; 340-801). Around 80% of the oral-bacteria and oral-archaea species analyzed had an ASI97 with at least one other species. These very similar species play different roles in the oral microbiota and belong to bacterial genera such as Campylobacter, Rothia, Streptococcus and Tannerella, and archaeal genera such as Halovivax, Methanosarcina and Methanosalsum. Moreover, ~20% and ~30% of these two-by-two similarity relationships were established between species from different bacterial and archaeal genera, respectively. Even taxa from distinct families, orders, and classes could be grouped in the same possible OTU. Consequently, regardless of the primer pair used, sequence clustering with a 97% similarity provides an inaccurate description of oral-bacterial and oral-archaeal species, which can greatly affect microbial diversity parameters. As a result, OTU clustering conditions the credibility of associations between some oral species and certain health and disease conditions. This significantly limits the comparability of the microbial diversity findings reported in oral microbiome literature.
Collapse
Affiliation(s)
- Alba Regueira-Iglesias
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Lara Vázquez-González
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Carlos Balsa-Castro
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Triana Blanco-Pintos
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Benjamín Martín-Biedma
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Víctor M. Arce
- Department of Physiology and Center for Disease in Molecular Medicine and Chronic Diseases, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria J. Carreira
- Centro Singular de Investigación en Tecnoloxías Intelixentes and Departamento de Electrónica e Computación, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| | - Inmaculada Tomás
- Oral Sciences Research Group, Department of Surgery and Medical-Surgical Specialties, School of Medicine and Dentistry, Universidade de Santiago de Compostela, Health Research Institute Foundation of Santiago (FIDIS), Santiago de Compostela, Spain
| |
Collapse
|
46
|
Uranga C, Nelson KE, Edlund A, Baker JL. Tetramic Acids Mutanocyclin and Reutericyclin A, Produced by Streptococcus mutans Strain B04Sm5 Modulate the Ecology of an in vitro Oral Biofilm. FRONTIERS IN ORAL HEALTH 2022; 2:796140. [PMID: 35048077 PMCID: PMC8757879 DOI: 10.3389/froh.2021.796140] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/29/2021] [Indexed: 01/04/2023] Open
Abstract
The human oral microbiome consists of diverse microbes actively communicating and interacting through a variety of biochemical mechanisms. Dental caries is a major public health issue caused by fermentable carbohydrate consumption that leads to dysbiosis of the oral microbiome. Streptococcus mutans is a known major contributor to caries pathogenesis, due to its exceptional ability to form biofilms in the presence of sucrose, as well as to its acidophilic lifestyle. S. mutans can also kill competing bacteria, which are typically health associated, through the production of bacteriocins and other small molecules. A subset of S. mutans strains encode the muc biosynthetic gene cluster (BGC), which was recently shown to produce the tetramic acids, mutanocyclin and reutericyclins A, B, and C. Reutericyclin A displayed strong antimicrobial activity and mutanocyclin appeared to be anti-inflammatory; however the effect of these compounds, and the carriage of muc by S. mutans, on the ecology of the oral microbiota is not known, and was examined here using a previously developed in vitro biofilm model derived from human saliva. While reutericyclin significantly inhibited in vitro biofilm formation and acid production at sub-nanomolar concentrations, mutanocyclin did not present any activity until the high micromolar range. 16S rRNA gene sequencing revealed that reutericyclin drastically altered the biofilm community composition, while mutanocyclin showed a more specific effect, reducing the relative abundance of cariogenic Limosilactobacillus fermentum. Mutanocyclin or reutericyclin produced by the S. mutans strains amended to the community did not appear to affect the community in the same way as the purified compounds, although the results were somewhat confounded by the differing growth rates of the S. mutans strains. Regardless of the strain added, the addition of S. mutans to the in vitro community significantly increased the abundance of S. mutans and Veillonella infantium, only. Overall, this study illustrates that reutericyclin A and mutanocyclin do impact the ecology of a complex in vitro oral biofilm; however, further research is needed to determine the extent to which the production of these compounds affects the virulence of S. mutans.
Collapse
Affiliation(s)
- Carla Uranga
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA, United States
| | - Karen E Nelson
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA, United States
| | - Anna Edlund
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA, United States.,Department of Pediatrics, UC San Diego School of Medicine, San Diego, CA, United States
| | - Jonathon L Baker
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA, United States.,Department of Pediatrics, UC San Diego School of Medicine, San Diego, CA, United States
| |
Collapse
|
47
|
Melo-Dias S, Valente C, Andrade L, Marques A, Sousa A. Saliva as a non-invasive specimen for COPD assessment. Respir Res 2022; 23:16. [PMID: 35093093 PMCID: PMC8800366 DOI: 10.1186/s12931-022-01935-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND People with COPD have been reported to bear a distinct airway microbiota from healthy individuals based on bronchoalveolar lavage (BAL) and sputum samples. Unfortunately, the collection of these samples involves relatively invasive procedures and is resource-demanding, limiting its regular use. Non-invasive samples from the upper airways could constitute an interesting alternative, but its relationship with COPD is still underexplored. We examined the merits of saliva to identify the typical profile of COPD oral bacteria and test its association with the disease. METHODS Outpatients with COPD and age-sex matched healthy controls were recruited and characterised based on clinical parameters and 16S rRNA profiling of oral bacteria. A clustering analysis based on patients' oral bacteria beta-diversity and logistic regressions were performed to evaluate the association between oral bacteria composition and COPD. RESULTS 128 individuals participated (70 patients and 58 controls). Differential abundance analyses showed differences in patients comparable to the ones previously observed in samples from the lower respiratory tract, i.e., an increase in Proteobacteria (particularly Haemophilus) and loss of microbiota diversity. An unsupervised clustering analysis separated patients in two groups based on microbiota composition differing significantly in the frequency of patients hospitalized due to severe acute exacerbation of COPD (AECOPD) and in the frequency of GOLD D patients. Furthermore, a low frequency of Prevotella was associated with a significantly higher risk of recent severe AECOPD and of being GOLD D. CONCLUSION Salivary bacteria showed an association with COPD, particularly with severe exacerbations, supporting the use of this non-invasive specimen for future studies of heterogeneous respiratory diseases like COPD.
Collapse
Affiliation(s)
- Sara Melo-Dias
- grid.7311.40000000123236065Department of Medical Sciences, University of Aveiro, Aveiro, Portugal ,grid.7311.40000000123236065Lab3R-Respiratory Research and Rehabilitation Laboratory, School of Health Sciences (ESSUA), University of Aveiro, Aveiro, Portugal ,grid.7311.40000000123236065Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Carla Valente
- Department of Pulmonology, Hospital Center of Baixo Vouga, Aveiro, Portugal
| | - Lília Andrade
- Department of Pulmonology, Hospital Center of Baixo Vouga, Aveiro, Portugal
| | - Alda Marques
- grid.7311.40000000123236065Lab3R-Respiratory Research and Rehabilitation Laboratory, School of Health Sciences (ESSUA), University of Aveiro, Aveiro, Portugal ,grid.7311.40000000123236065Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Sousa
- grid.7311.40000000123236065Department of Medical Sciences, University of Aveiro, Aveiro, Portugal ,grid.7311.40000000123236065Institute of Biomedicine (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
48
|
Sundh J, Tanash H, Arian R, Neves-Guimaraes A, Broberg K, Lindved G, Kern T, Zych K, Nielsen HB, Halling A, Ohlsson B, Jönsson D. Advanced Dental Cleaning is Associated with Reduced Risk of COPD Exacerbations - A Randomized Controlled Trial. Int J Chron Obstruct Pulmon Dis 2021; 16:3203-3215. [PMID: 34858021 PMCID: PMC8629912 DOI: 10.2147/copd.s327036] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/13/2021] [Indexed: 01/02/2023] Open
Abstract
Purpose Infections from the oral microbiome may lead to exacerbations of chronic obstructive pulmonary disease (COPD). We investigated whether advanced dental cleaning could reduce exacerbation frequency. Secondary outcomes were disease-specific health status, lung function, and whether the bacterial load and composition of plaque microbiome at baseline were associated with a difference in outcomes. Patients and Methods One-hundred-one primary and secondary care patients with COPD were randomized to intervention with advanced dental cleaning or to dental examination only, repeated after six months. At baseline and at 12 months, data of exacerbations, lung function, COPD Assessment Test (CAT) score, and periodontal status were collected from questionnaires, record review, and periodontal examination. Student’s t-test and Mann–Whitney-U (MWU) test compared changes in outcomes. The primary outcome variable was also assessed using multivariable linear regression with adjustment for potential confounders. Microbiome analyses of plaque samples taken at baseline were performed using Wilcoxon signed ranks tests for calculation of alpha diversity, per mutational multivariate analysis of variance for beta diversity, and receiver operating characteristic curves for prediction of outcomes based on machine learning models. Results In the MWU test, the annual exacerbation frequency was significantly reduced in patients previously experiencing frequent exacerbations (p = 0.020) and in those with repeated advanced dental cleaning (p = 0.039) compared with the non-treated control group, but not in the total population including both patients with a single and repeated visits (p = 0.207). The result was confirmed in multivariable linear regression, where the risk of new exacerbations was significantly lower in patients both in the intention to treat analysis (regression coefficient 0.36 (95% CI 0.25–0.52), p < 0.0001) and in the population with repeated dental cleaning (0.16 (0.10–0.27), p < 0.0001). The composition of microbiome at baseline was moderately predictive of an increased risk of worsened health status at 12 months (AUC = 0.723). Conclusion Advanced dental cleaning is associated with a reduced frequency of COPD exacerbations. Regular periodontal examination and dental cleaning may be of clinical importance to prevent COPD exacerbations.
Collapse
Affiliation(s)
- Josefin Sundh
- Department of Respiratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Hanan Tanash
- Department of Respiratory Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Rahi Arian
- Department of Periodontology and Implantology, Public Dental Service, Örebro, Sweden
| | | | | | | | - Timo Kern
- Clinical Microbiomics, Copenhagen, Denmark
| | | | | | - Anders Halling
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - Bodil Ohlsson
- Department of Internal Medicine, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Daniel Jönsson
- Public Dental Service of Skåne, Lund, Sweden.,Hypertension and Cardiovascular Disease, Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden.,Faculty of Odontology, Malmö University, Malmö, Sweden
| |
Collapse
|
49
|
Cameron ES, Schmidt PJ, Tremblay BJM, Emelko MB, Müller KM. Enhancing diversity analysis by repeatedly rarefying next generation sequencing data describing microbial communities. Sci Rep 2021; 11:22302. [PMID: 34785722 PMCID: PMC8595385 DOI: 10.1038/s41598-021-01636-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022] Open
Abstract
Amplicon sequencing has revolutionized our ability to study DNA collected from environmental samples by providing a rapid and sensitive technique for microbial community analysis that eliminates the challenges associated with lab cultivation and taxonomic identification through microscopy. In water resources management, it can be especially useful to evaluate ecosystem shifts in response to natural and anthropogenic landscape disturbances to signal potential water quality concerns, such as the detection of toxic cyanobacteria or pathogenic bacteria. Amplicon sequencing data consist of discrete counts of sequence reads, the sum of which is the library size. Groups of samples typically have different library sizes that are not representative of biological variation; library size normalization is required to meaningfully compare diversity between them. Rarefaction is a widely used normalization technique that involves the random subsampling of sequences from the initial sample library to a selected normalized library size. This process is often dismissed as statistically invalid because subsampling effectively discards a portion of the observed sequences, yet it remains prevalent in practice and the suitability of rarefying, relative to many other normalization approaches, for diversity analysis has been argued. Here, repeated rarefying is proposed as a tool to normalize library sizes for diversity analyses. This enables (i) proportionate representation of all observed sequences and (ii) characterization of the random variation introduced to diversity analyses by rarefying to a smaller library size shared by all samples. While many deterministic data transformations are not tailored to produce equal library sizes, repeatedly rarefying reflects the probabilistic process by which amplicon sequencing data are obtained as a representation of the amplified source microbial community. Specifically, it evaluates which data might have been obtained if a particular sample's library size had been smaller and allows graphical representation of the effects of this library size normalization process upon diversity analysis results.
Collapse
Affiliation(s)
- Ellen S Cameron
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
| | - Philip J Schmidt
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
| | - Benjamin J-M Tremblay
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
| | - Monica B Emelko
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada
| | - Kirsten M Müller
- Department of Biology, University of Waterloo, 200 University Ave. W, Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|
50
|
RESCRIPt: Reproducible sequence taxonomy reference database management. PLoS Comput Biol 2021; 17:e1009581. [PMID: 34748542 PMCID: PMC8601625 DOI: 10.1371/journal.pcbi.1009581] [Citation(s) in RCA: 349] [Impact Index Per Article: 87.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/18/2021] [Accepted: 10/21/2021] [Indexed: 12/22/2022] Open
Abstract
Nucleotide sequence and taxonomy reference databases are critical resources for widespread applications including marker-gene and metagenome sequencing for microbiome analysis, diet metabarcoding, and environmental DNA (eDNA) surveys. Reproducibly generating, managing, using, and evaluating nucleotide sequence and taxonomy reference databases creates a significant bottleneck for researchers aiming to generate custom sequence databases. Furthermore, database composition drastically influences results, and lack of standardization limits cross-study comparisons. To address these challenges, we developed RESCRIPt, a Python 3 software package and QIIME 2 plugin for reproducible generation and management of reference sequence taxonomy databases, including dedicated functions that streamline creating databases from popular sources, and functions for evaluating, comparing, and interactively exploring qualitative and quantitative characteristics across reference databases. To highlight the breadth and capabilities of RESCRIPt, we provide several examples for working with popular databases for microbiome profiling (SILVA, Greengenes, NCBI-RefSeq, GTDB), eDNA and diet metabarcoding surveys (BOLD, GenBank), as well as for genome comparison. We show that bigger is not always better, and reference databases with standardized taxonomies and those that focus on type strains have quantitative advantages, though may not be appropriate for all use cases. Most databases appear to benefit from some curation (quality filtering), though sequence clustering appears detrimental to database quality. Finally, we demonstrate the breadth and extensibility of RESCRIPt for reproducible workflows with a comparison of global hepatitis genomes. RESCRIPt provides tools to democratize the process of reference database acquisition and management, enabling researchers to reproducibly and transparently create reference materials for diverse research applications. RESCRIPt is released under a permissive BSD-3 license at https://github.com/bokulich-lab/RESCRIPt. Generating and managing sequence and taxonomy reference data presents a bottleneck to many researchers, whether they are generating custom databases or attempting to format existing, curated reference databases for use with standard sequence analysis tools. Evaluating database quality and choosing the “best” database can be an equally formidable challenge. We developed RESCRIPt to alleviate this bottleneck, supporting reproducible, streamlined generation, curation, and evaluation of reference sequence databases. RESCRIPt uses QIIME 2 artifact file formats, which store all processing steps as data provenance within each file, allowing researchers to retrace the computational steps used to generate any given file. We used RESCRIPt to benchmark several commonly used marker-gene sequence databases for 16S rRNA genes, ITS, and COI sequences, demonstrating both the utility of RESCRIPt to streamline use of these databases, but also to evaluate several qualitative and quantitative characteristics of each database. We show that larger databases are not always best, and curation steps to reduce redundancy and filter out noisy sequences may be beneficial for some applications. We anticipate that RESCRIPt will streamline the use, management, and evaluation/selection of reference database materials for microbiomics, diet metabarcoding, eDNA, and other diverse applications.
Collapse
|