51
|
Homayouni Rad A, Pourjafar H, Mirzakhani E. A comprehensive review of the application of probiotics and postbiotics in oral health. Front Cell Infect Microbiol 2023; 13:1120995. [PMID: 36968114 PMCID: PMC10031100 DOI: 10.3389/fcimb.2023.1120995] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Oral diseases are among the most common diseases around the world that people usually suffer from during their lifetime. Tooth decay is a multifactorial disease, and the composition of oral microbiota is a critical factor in its development. Also, Streptococcus mutans is considered the most important caries-causing species. It is expected that probiotics, as they adjust the intestinal microbiota and reduce the number of pathogenic bacteria in the human intestine, can exert their health-giving effects, especially the anti-pathogenic effect, in the oral cavity, which is part of the human gastrointestinal tract. Therefore, numerous in vitro and in vivo studies have been conducted on the role of probiotics in the prevention of tooth decay. In this review, while investigating the effect of different strains of probiotics Lactobacillus and Bifidobacteria on oral diseases, including dental caries, candida yeast infections, periodontal diseases, and halitosis, we have also discussed postbiotics as novel non-living biological compounds derived from probiotics.
Collapse
Affiliation(s)
- Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Pourjafar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
- *Correspondence: Esmaeel Mirzakhani, ; Hadi Pourjafar,
| | - Esmaeel Mirzakhani
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Esmaeel Mirzakhani, ; Hadi Pourjafar,
| |
Collapse
|
52
|
Zhang J, Duan Z. Identification of a new probiotic strain, Lactiplantibacillus plantarum VHProbi ® V38, and its use as an oral health agent. Front Microbiol 2022; 13:1000309. [PMID: 36583042 PMCID: PMC9793799 DOI: 10.3389/fmicb.2022.1000309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Introduction Probiotics can be used to treat oral diseases such as dental caries, gingivitis, periodontitis, and halitosis. Methods This study screened for strains capable of inhibiting Streptococcus mutans,one of the primary pathogenic bacteria responsible for dental caries by agar diffusion in different samples. Strain identification was performed by 16S rDNA sequencing and the API 50CH system. The potential functions of the strains in terms of oral health properties were also tested by agglutination assays, growth inhibition assays, adhesion assays, biofilm removal assays and inhibition of adhesion in human primary gingival epithelial (HPGE) cells assays. Results This study identified a probiotic strain from fermented cabbages that has a strong inhibitory effect on Streptococcus mutans. The API 50CH system and 16S rDNA sequencing verified that this was a new strain and it was given the name, Lactiplantibacillus plantarum VHProbi®V38. Agglutination, growth inhibition and adhesion, and biofilm removal tests indicated that L. plantarum VHProbi® V38 inhibited and reduced S. mutans. This probiotic was shown to have a broad antibacterial spectrum, simultaneously inhibiting the growth of periodontal pathogenic bacteria such as Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans, and Fusobacterium nucleatum. After 2 hours of co-cultivation with these pathogens, L. plantarum VHProbi® V38 was able to significantly reduce pathogens adhesion on human primary gingival epithelial (HPGE) cells. Discussion These findings suggest that L. plantarum VHProbi® V38 could potentially prevent and treat periodontal diseases caused by these pathogenic bacteria. L. plantarum VHProbi® V38 also adheres strongly to HPGE cells and thus has potential as an oral probiotic. This study describes new methods that can be used to aid the screening and identification of oral probiotics.
Collapse
|
53
|
Tonon CC, Panariello B, Chorilli M, Spolidorio DMP, Duarte S. Effect of curcumin-loaded photoactivatable polymeric nanoparticle on peri-implantitis-related biofilm. Photodiagnosis Photodyn Ther 2022; 40:103150. [PMID: 36244678 DOI: 10.1016/j.pdpdt.2022.103150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Curcumin has been used as a photosensitizer (PS) for antimicrobial photodynamic chemotherapy (PACT). However, its low solubility, instability, and poor bioavailability challenge its in vivo application. This study aimed to synthesize curcumin-loaded polymeric nanoparticles (curcumin-NP) and determine their antimicrobial and cytotoxic effects. Nanoparticles (NP) were synthesized using polycaprolactone (PCL) as a polymer by the nanoprecipitation method. Curcumin-NP was characterized by particle size, polydispersity index and zeta potential, scanning electron microscopy, and curcumin encapsulation efficiency (EE). Curcumin-NP was compared to free curcumin solubilized in 10% DMSO as photosensitizers for PACT in single and multispecies Porphyromonas gingivalis, Fusobacterium nucleatum, and Streptococcus oralis biofilms. Chlorhexidine 0.12% (CHX) and ultrapure water were used as positive and negative controls. The cytotoxic effect of curcumin-NP was evaluated on human periodontal ligament fibroblast cells (HPLF). Data were analyzed by ANOVA (α=0.05). Curcumin-NP exhibited homogeneity and stability in solution, small particle size, and 67.5% EE of curcumin. Curcumin-NP presented reduced antibiofilm activity at 500 µg/ml, although in planktonic cultures it showed inhibitory and bactericidal effect. Curcumin-NP and curcumin with and without photoactivation were not cytotoxic to HPLF cells. Curcumin-NP has antimicrobial and antibiofilm properties, with better effects when associated with blue light, being a promising therapy for preventing and treating peri-implant diseases.
Collapse
Affiliation(s)
- Caroline Coradi Tonon
- Department of Oral Diagnosis and Surgery, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Beatriz Panariello
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Marlus Chorilli
- Department of Drugs and Pharmaceuticals, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | | | - Simone Duarte
- American Dental Association Science and Research Institute, 211 E. Chicago Ave, Chicago, IL 60611, USA.
| |
Collapse
|
54
|
Anju VT, Busi S, Imchen M, Kumavath R, Mohan MS, Salim SA, Subhaswaraj P, Dyavaiah M. Polymicrobial Infections and Biofilms: Clinical Significance and Eradication Strategies. Antibiotics (Basel) 2022; 11:antibiotics11121731. [PMID: 36551388 PMCID: PMC9774821 DOI: 10.3390/antibiotics11121731] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 12/03/2022] Open
Abstract
Biofilms are population of cells growing in a coordinated manner and exhibiting resistance towards hostile environments. The infections associated with biofilms are difficult to control owing to the chronicity of infections and the emergence of antibiotic resistance. Most microbial infections are contributed by polymicrobial or mixed species interactions, such as those observed in chronic wound infections, otitis media, dental caries, and cystic fibrosis. This review focuses on the polymicrobial interactions among bacterial-bacterial, bacterial-fungal, and fungal-fungal aggregations based on in vitro and in vivo models and different therapeutic interventions available for polymicrobial biofilms. Deciphering the mechanisms of polymicrobial interactions and microbial diversity in chronic infections is very helpful in anti-microbial research. Together, we have discussed the role of metagenomic approaches in studying polymicrobial biofilms. The outstanding progress made in polymicrobial research, especially the model systems and application of metagenomics for detecting, preventing, and controlling infections, are reviewed.
Collapse
Affiliation(s)
- V T Anju
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
- Correspondence:
| | - Madangchanok Imchen
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Ranjith Kumavath
- Department of Genomic Science, School of Biological Sciences, Central University of Kerala, Kerala 671316, India
- Department of Biotechnology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Mahima S. Mohan
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Simi Asma Salim
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| | - Pattnaik Subhaswaraj
- Department of Biotechnology and Bioinformatics, Sambalpur University, Burla, Sambalpur 768019, India
| | - Madhu Dyavaiah
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry 605014, India
| |
Collapse
|
55
|
Rabe A, Gesell Salazar M, Michalik S, Kocher T, Below H, Völker U, Welk A. Impact of different oral treatments on the composition of the supragingival plaque microbiome. J Oral Microbiol 2022; 14:2138251. [PMID: 36338832 PMCID: PMC9629129 DOI: 10.1080/20002297.2022.2138251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Dental plaque consists of a diverse microbial community embedded in a complex structure of exopolysaccharides. Dental biofilms form a natural barrier against pathogens but lead to oral diseases in a dysbiotic state. Objective Using a metaproteome approach combined with a standard plaque-regrowth study, this pilot study examined the impact of different concentrations of lactoperoxidase (LPO) on early plaque formation, and active biological processes. Design Sixteen orally healthy subjects received four local treatments as a randomized single-blind study based on a cross-over design. Two lozenges containing components of the LPO-system in different concentrations were compared to a placebo and Listerine®. The newly formed dental plaque was analyzed by mass spectrometry (nLC-MS/MS). Results On average 1,916 metaproteins per sample were identified, which could be assigned to 116 genera and 1,316 protein functions. Listerine® reduced the number of metaproteins and their relative abundance, confirming the plaque inhibiting effect. The LPO-lozenges triggered mainly higher metaprotein abundances of early and secondary colonizers as well as bacteria associated with dental health but also periodontitis. Functional information indicated plaque biofilm growth. Conclusion In conclusion, the mechanisms on plaque biofilm formation of Listerine® and the LPO-system containing lozenges are different. In contrast to Listerine®, the lozenges led to a higher bacterial diversity.
Collapse
Affiliation(s)
- Alexander Rabe
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany,CONTACT Alexander Rabe University Medicine Greifswald, Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, Felix-Hausdorff-Str. 8, 17489Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Thomas Kocher
- Center for Dentistry, Oral and Maxillofacial Medicine, Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, Dental School of University Medicine Greifswald, Fleischmannstraße 42-44, 17489
| | - Harald Below
- Institute for Hygiene and Environmental Medicine, University Medicine Greifswald, Walter-Rathenau-Straße 49 A17475Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Felix-Hausdorff-Str. 8, 17475Greifswald, Germany
| | - Alexander Welk
- Center for Dentistry, Oral and Maxillofacial Medicine, Department of Restorative Dentistry, Periodontology, Endodontology, and Preventive and Pediatric Dentistry, Dental School of University Medicine Greifswald, Fleischmannstraße 42-44, 17489
| |
Collapse
|
56
|
Abstract
The morphogenesis of two-dimensional bacterial colonies has been well studied. However, little is known about the colony morphologies of bacteria growing in three dimensions, despite the prevalence of three-dimensional environments (e.g., soil, inside hosts) as natural bacterial habitats. Using experiments on bacteria in granular hydrogel matrices, we find that dense multicellular colonies growing in three dimensions undergo a common morphological instability and roughen, adopting a characteristic broccoli-like morphology when they exceed a critical size. Analysis of a continuum “active fluid” model of the expanding colony reveals that this behavior originates from an interplay of competition for nutrients with growth-driven colony expansion, both of which vary spatially. These results shed light on the fundamental biophysical principles underlying growth in three dimensions. How do growing bacterial colonies get their shapes? While colony morphogenesis is well studied in two dimensions, many bacteria grow as large colonies in three-dimensional (3D) environments, such as gels and tissues in the body or subsurface soils and sediments. Here, we describe the morphodynamics of large colonies of bacteria growing in three dimensions. Using experiments in transparent 3D granular hydrogel matrices, we show that dense colonies of four different species of bacteria generically become morphologically unstable and roughen as they consume nutrients and grow beyond a critical size—eventually adopting a characteristic branched, broccoli-like morphology independent of variations in the cell type and environmental conditions. This behavior reflects a key difference between two-dimensional (2D) and 3D colonies; while a 2D colony may access the nutrients needed for growth from the third dimension, a 3D colony inevitably becomes nutrient limited in its interior, driving a transition to unstable growth at its surface. We elucidate the onset of the instability using linear stability analysis and numerical simulations of a continuum model that treats the colony as an “active fluid” whose dynamics are driven by nutrient-dependent cellular growth. We find that when all dimensions of the colony substantially exceed the nutrient penetration length, nutrient-limited growth drives a 3D morphological instability that recapitulates essential features of the experimental observations. Our work thus provides a framework to predict and control the organization of growing colonies—as well as other forms of growing active matter, such as tumors and engineered living materials—in 3D environments.
Collapse
|
57
|
Bu F, Liu M, Xie Z, Chen X, Li G, Wang X. Targeted Anti-Biofilm Therapy: Dissecting Targets in the Biofilm Life Cycle. Pharmaceuticals (Basel) 2022; 15:1253. [PMID: 36297365 PMCID: PMC9611117 DOI: 10.3390/ph15101253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 06/13/2024] Open
Abstract
Biofilm is a crucial virulence factor for microorganisms that causes chronic infection. After biofilm formation, the bacteria present improve drug tolerance and multifactorial defense mechanisms, which impose significant challenges for the use of antimicrobials. This indicates the urgent need for new targeted technologies and emerging therapeutic strategies. In this review, we focus on the current biofilm-targeting strategies and those under development, including targeting persistent cells, quorum quenching, and phage therapy. We emphasize biofilm-targeting technologies that are supported by blocking the biofilm life cycle, providing a theoretical basis for design of targeting technology that disrupts the biofilm and promotes practical application of antibacterial materials.
Collapse
Affiliation(s)
| | | | | | | | | | - Xing Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
58
|
Ponath F, Zhu Y, Cosi V, Vogel J. Expanding the genetic toolkit helps dissect a global stress response in the early-branching species Fusobacterium nucleatum. Proc Natl Acad Sci U S A 2022; 119:e2201460119. [PMID: 36161895 PMCID: PMC9546586 DOI: 10.1073/pnas.2201460119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/13/2022] [Indexed: 11/18/2022] Open
Abstract
Fusobacterium nucleatum, long known as a common oral microbe, has recently garnered attention for its ability to colonize tissues and tumors elsewhere in the human body. Clinical and epidemiological research has now firmly established F. nucleatum as an oncomicrobe associated with several major cancer types. However, with the current research focus on host associations, little is known about gene regulation in F. nucleatum itself, including global stress-response pathways that typically ensure the survival of bacteria outside their primary niche. This is due to the phylogenetic distance of Fusobacteriota to most model bacteria, their limited genetic tractability, and paucity of known gene functions. Here, we characterize a global transcriptional stress-response network governed by the extracytoplasmic function sigma factor, σE. To this aim, we developed several genetic tools for this anaerobic bacterium, including four different fluorescent marker proteins, inducible gene expression, scarless gene deletion, and transcriptional and translational reporter systems. Using these tools, we identified a σE response partly reminiscent of phylogenetically distant Proteobacteria but induced by exposure to oxygen. Although F. nucleatum lacks canonical RNA chaperones, such as Hfq, we uncovered conservation of the noncoding arm of the σE response in form of the noncoding RNA FoxI. This regulatory small RNA acts as an mRNA repressor of several membrane proteins, thereby supporting the function of σE. In addition to the characterization of a global stress response in F. nucleatum, the genetic tools developed here will enable further discoveries and dissection of regulatory networks in this early-branching bacterium.
Collapse
Affiliation(s)
- Falk Ponath
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, D-97080 Germany
| | - Yan Zhu
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, D-97080 Germany
| | - Valentina Cosi
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, D-97080 Germany
| | - Jörg Vogel
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, D-97080 Germany
- Institute for Molecular Infection Biology, University of Würzburg, Würzburg, D-97080 Germany
| |
Collapse
|
59
|
Alves CH, Russi KL, Rocha NC, Bastos F, Darrieux M, Parisotto TM, Girardello R. Host-microbiome interactions regarding peri-implantitis and dental implant loss. Lab Invest 2022; 20:425. [PMID: 36138430 PMCID: PMC9502891 DOI: 10.1186/s12967-022-03636-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022]
Abstract
In the last decades, the ortho-aesthetic-functional rehabilitation had significant advances with the advent of implantology. Despite the success in implantology surgeries, there is a percentage of failures mainly due to in loco infections, through bacterial proliferation, presence of fungi and biofilm formation, originating peri-implantitis. In this sense, several studies have been conducted since then, seeking answers to numerous questions that remain unknown. Thus, the present work aims to discuss the interaction between host-oral microbiome and the development of peri-implantitis. Peri-implantitis was associated with a diversity of bacterial species, being Porphiromonas gingivalis, Treponema denticola and Tannerella forsythia described in higher proportion of peri-implantitis samples. In a parallel role, the injury of peri-implant tissue causes an inflammatory response mediated by activation of innate immune cells such as macrophages, dendritic cells, mast cells, and neutrophils. In summary, the host immune system activation may lead to imbalance of oral microbiota, and, in turn, the oral microbiota dysbiosis is reported leading to cytokines, chemokines, prostaglandins, and proteolytic enzymes production. These biological processes may be responsible for implant loss.
Collapse
Affiliation(s)
- Carlos Henrique Alves
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil
| | - Karolayne Larissa Russi
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil
| | - Natália Conceição Rocha
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil
| | | | - Michelle Darrieux
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil
| | - Thais Manzano Parisotto
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil
| | - Raquel Girardello
- Laboratório de Microbiologia Molecular E Clínica, Programa de Pós-Graduação Em Ciências da Saúde, Universidade São Francisco, 218, São Francisco Ave., Bragança Paulista, São Paulo, Zip code: # 12916900, Brazil.
| |
Collapse
|
60
|
Zhang Q, Wang M, Ma X, Li Z, Jiang C, Pan Y, Zeng Q. In vitro investigation on lactic acid bacteria isolatedfrom Yak faeces for potential probiotics. Front Cell Infect Microbiol 2022; 12:984537. [PMID: 36189367 PMCID: PMC9523120 DOI: 10.3389/fcimb.2022.984537] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
In order to evaluate the potential and safety of lactic acid bacteria (LAB) isolated from faeces samples of Ganan yak as probiotic for prevention and/or treatment of yak diarrhea, four strains of LAB including Latilactobacillus curvatus (FY1), Weissella cibaria (FY2), Limosilactobacillus mucosae (FY3), and Lactiplantibacillus pentosus (FY4) were isolated and identified in this study. Cell surface characteristics (hydrophobicity and cell aggregation), acid resistance and bile tolerance, compatibility, antibacterial activity and in vitro cell adhesion tests were also carried out to evaluate the probiotic potential of LAB. The results showed that the four isolates had certain acid tolerance, bile salt tolerance, hydrophobicity and cell aggregation, all of which contribute to the survival and colonization of LAB in the gastrointestinal tract. There is no compatibility between the four strains, so they can be combined into a mixed probiotic formula. Antimicrobial tests showed that the four strains were antagonistic to Escherichia coli, Staphylococcus aureus, and Salmonella typhimurium. Moreover, the in vitro safety of the four isolates were determined through hemolytic analysis, gelatinase activity, and antibacterial susceptibility experiments. The results suggest that all the four strains were considered as safe because they had no hemolytic activity, no gelatinase activity and were sensitive to most antibacterial agents. Moreover, the acute oral toxicity test of LAB had no adverse effect on body weight gain, food utilization and organ indices in Kunming mice. In conclusion, the four LAB isolated from yak feces have considerable potential to prevent and/or treat yak bacterial disease-related diarrhea.
Collapse
Affiliation(s)
- Qingli Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xin Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Zhijie Li
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Chenghui Jiang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| |
Collapse
|
61
|
Krutyhołowa A, Strzelec K, Dziedzic A, Bereta GP, Łazarz-Bartyzel K, Potempa J, Gawron K. Host and bacterial factors linking periodontitis and rheumatoid arthritis. Front Immunol 2022; 13:980805. [PMID: 36091038 PMCID: PMC9453162 DOI: 10.3389/fimmu.2022.980805] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/27/2022] [Indexed: 02/05/2023] Open
Abstract
Observations from numerous clinical, epidemiological and serological studies link periodontitis with severity and progression of rheumatoid arthritis. The strong association is observed despite totally different aetiology of these two diseases, periodontitis being driven by dysbiotic microbial flora on the tooth surface below the gum line, while rheumatoid arthritis being the autoimmune disease powered by anti-citrullinated protein antibodies (ACPAs). Here we discuss genetic and environmental risk factors underlying development of both diseases with special emphasis on bacteria implicated in pathogenicity of periodontitis. Individual periodontal pathogens and their virulence factors are argued as potentially contributing to putative causative link between periodontal infection and initiation of a chain of events leading to breakdown of immunotolerance and development of ACPAs. In this respect peptidylarginine deiminase, an enzyme unique among prokaryotes for Porphyromonas gingivalis, is elaborated as a potential mechanistic link between this major periodontal pathogen and initiation of rheumatoid arthritis development.
Collapse
Affiliation(s)
- Anna Krutyhołowa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Karolina Strzelec
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Agata Dziedzic
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Grzegorz P. Bereta
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Katarzyna Łazarz-Bartyzel
- Department of Periodontology and Oral Medicine, Faculty of Medicine, Medical College, Jagiellonian University, Krakow, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland,Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States,*Correspondence: Katarzyna Gawron, ; Jan Potempa,
| | - Katarzyna Gawron
- Department of Molecular Biology and Genetics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland,*Correspondence: Katarzyna Gawron, ; Jan Potempa,
| |
Collapse
|
62
|
Zelikman H, Slutzkey G, Rosner O, Levartovsky S, Matalon S, Beitlitum I. Bacterial Growth on Three Non-Resorbable Polytetrafluoroethylene (PTFE) Membranes-An In Vitro Study. MATERIALS (BASEL, SWITZERLAND) 2022; 15:5705. [PMID: 36013840 PMCID: PMC9414989 DOI: 10.3390/ma15165705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 06/15/2023]
Abstract
GBR (Guided Bone Regeneration) procedure is challenged by the risk of membrane exposure to the oral cavity and contamination. The barrier quality of these membranes serve as a mechanical block from bacterial penetration into the GBR site. The purpose of this in vitro study was to evaluate the antibacterial effect of three commercial non-resorbable polytetrafluoroethylene membranes. (Two d-PTFE membranes and one double layer e-PTFE +d-PTFE membrane). A validated in vitro model with two bacterial species (Streptococcus sanguinis and Fusobacterium nucleatum) was used. Eight samples from membrane each were placed in a 96-well microtiter plate. The experimental and positive control groups were exposed to a bacterial suspension which involved one bacterial species in each plate. Bacterial growth was monitored spectrophotometrically at 650 nm for 24 h in temperature controlled microplate spectrophotometer under anaerobic conditions. One- Sample Kolmogorov−Smirnov Normal test and the Kruskal−Wallis test was used for the statistical analysis. As shown by the bacterial growth curves obtained from the spectrophotometer readings, all three membranes resulted in bacterial growth. We have not found a statistical difference in F. nucleatum growth between different membrane samples and the positive control group. However, S. sanguinis growth was reduced significantly in the presence of two membranes (CYTOPLAST TXT-200 and NeoGenTM) when compared to the control (p < 0.01). The presence of Permamem® had no significant influence on S. sanguinis growth. Some types of commercial non-resorbable PTFE membranes may have an impact on the growth dynamics of specific bacterial species.
Collapse
Affiliation(s)
- Helena Zelikman
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Gil Slutzkey
- Department of Periodontology and Dental Implantology, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofir Rosner
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shifra Levartovsky
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shlomo Matalon
- Department of Oral Rehabilitation, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ilan Beitlitum
- Department of Periodontology and Dental Implantology, Goldschleger School of Dental Medicine, Sackler Medical Faculty, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
63
|
Fusobacterium nucleatum and Malignant Tumors of the Digestive Tract: A Mechanistic Overview. Bioengineering (Basel) 2022; 9:bioengineering9070285. [PMID: 35877336 PMCID: PMC9312082 DOI: 10.3390/bioengineering9070285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 11/16/2022] Open
Abstract
Fusobacterium nucleatum (F. nucleatum) is an oral anaerobe that plays a role in several oral diseases. However, F. nucleatum is also found in other tissues of the digestive tract, and several studies have recently reported that the level of F. nucleatum is significantly elevated in malignant tumors of the digestive tract. F. nucleatum is proposed as one of the risk factors in the initiation and progression of digestive tract malignant tumors. In this review, we summarize recent reports on F. nucleatum and its role in digestive tract cancers and evaluate the mechanisms underlying the action of F. nucleatum in digestive tract cancers.
Collapse
|
64
|
Costa RC, Bertolini M, Costa Oliveira BE, Nagay BE, Dini C, Benso B, Klein MI, Barāo VAR, Souza JGS. Polymicrobial biofilms related to dental implant diseases: unravelling the critical role of extracellular biofilm matrix. Crit Rev Microbiol 2022; 49:370-390. [PMID: 35584310 DOI: 10.1080/1040841x.2022.2062219] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Biofilms are complex tri-dimensional structures that encase microbial cells in an extracellular matrix comprising self-produced polymeric substances. The matrix rich in extracellular polymeric substance (EPS) contributes to the unique features of biofilm lifestyle and structure, enhancing microbial accretion, biofilm virulence, and antimicrobial resistance. The role of the EPS matrix of biofilms growing on biotic surfaces, especially dental surfaces, is largely unravelled. To date, there is a lack of a broad overview of existing literature concerning the relationship between the EPS matrix and the dental implant environment and its role in implant-related infections. Here, we discuss recent advances in the critical role of the EPS matrix on biofilm growth and virulence on the dental implant surface and its effect on the etiopathogenesis and progression of implant-related infections. Similar to other biofilms associated with human diseases/conditions, EPS-enriched biofilms on implant surfaces promote microbial accumulation, microbiological shift, cross-kingdom interaction, antimicrobial resistance, biofilm virulence, and, consequently, peri-implant tissue damage. But intriguingly, the protagonism of EPS role on implant-related infections and the development of matrix-target therapeutic strategies has been neglected. Finally, we highlight the need for more in-depth analyses of polymicrobial interactions within EPS matrix and EPS-targeting technologies' rationale for disrupting the complex biofilm microenvironment with more outstanding translation to implant applications in the near future.
Collapse
Affiliation(s)
- Raphael C Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Martinna Bertolini
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | | | - Bruna E Nagay
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Bruna Benso
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, CA, Chile
| | - Marlise I Klein
- Department of Dental Materials and Prosthodontics, São Paulo State University, São Paulo, Brazil
| | - Valentim A R Barāo
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil
| | - Joāo Gabriel S Souza
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Brazil.,Dental Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Brazil.,Dental Research Division, Guarulhos University, Sāo Paulo, Brazil
| |
Collapse
|
65
|
Bensing BA, Stubbs HE, Agarwal R, Yamakawa I, Luong K, Solakyildirim K, Yu H, Hadadianpour A, Castro MA, Fialkowski KP, Morrison KM, Wawrzak Z, Chen X, Lebrilla CB, Baudry J, Smith JC, Sullam PM, Iverson TM. Origins of glycan selectivity in streptococcal Siglec-like adhesins suggest mechanisms of receptor adaptation. Nat Commun 2022; 13:2753. [PMID: 35585145 PMCID: PMC9117288 DOI: 10.1038/s41467-022-30509-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 04/26/2022] [Indexed: 11/22/2022] Open
Abstract
Bacterial binding to host receptors underlies both commensalism and pathogenesis. Many streptococci adhere to protein-attached carbohydrates expressed on cell surfaces using Siglec-like binding regions (SLBRs). The precise glycan repertoire recognized may dictate whether the organism is a strict commensal versus a pathogen. However, it is currently not clear what drives receptor selectivity. Here, we use five representative SLBRs and identify regions of the receptor binding site that are hypervariable in sequence and structure. We show that these regions control the identity of the preferred carbohydrate ligand using chimeragenesis and single amino acid substitutions. We further evaluate how the identity of the preferred ligand affects the interaction with glycoprotein receptors in human saliva and plasma samples. As point mutations can change the preferred human receptor, these studies suggest how streptococci may adapt to changes in the environmental glycan repertoire.
Collapse
Affiliation(s)
- Barbara A Bensing
- Division of Infectious Diseases, Veterans Affairs Medical Center, Department of Medicine, University of California, San Francisco, CA, USA
- the Northern California Institute for Research and Education, San Francisco, CA, 94121, USA
| | - Haley E Stubbs
- Graduate Program in Chemical and Physical Biology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Rupesh Agarwal
- University of Tennessee/Oak Ridge National Laboratory, Center for Molecular Biophysics, Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6309, USA
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Izumi Yamakawa
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- School of Nursing, Belmont University, Nashville, TN, 37212, USA
| | - Kelvin Luong
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kemal Solakyildirim
- Department of Chemistry, Erzincan Binali Yildirim University, Erzincan, 24100, Turkey
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Hai Yu
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Azadeh Hadadianpour
- Department of Microbiology, Pathology, and Immunology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Manuel A Castro
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kevin P Fialkowski
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
- College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - KeAndreya M Morrison
- Department of Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, 37208, USA
| | - Zdzislaw Wawrzak
- LS-CAT Synchrotron Research Center, Northwestern University, Argonne, IL, 60439, USA
| | - Xi Chen
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - Jerome Baudry
- Department of Biological Sciences, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Jeremy C Smith
- University of Tennessee/Oak Ridge National Laboratory, Center for Molecular Biophysics, Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, 37831-6309, USA
- Department of Biochemistry and Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Paul M Sullam
- Division of Infectious Diseases, Veterans Affairs Medical Center, Department of Medicine, University of California, San Francisco, CA, USA
- the Northern California Institute for Research and Education, San Francisco, CA, 94121, USA
| | - T M Iverson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
66
|
Combined Transcriptomic and Protein Array Cytokine Profiling of Human Stem Cells from Dental Apical Papilla Modulated by Oral Bacteria. Int J Mol Sci 2022; 23:ijms23095098. [PMID: 35563488 PMCID: PMC9103834 DOI: 10.3390/ijms23095098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/22/2022] [Accepted: 04/30/2022] [Indexed: 11/18/2022] Open
Abstract
Stem cells from the apical papilla (SCAP) are a promising resource for use in regenerative endodontic treatment (RET) that may be adversely affected by oral bacteria, which in turn can exert an effect on the success of RET. Our work aims to study the cytokine profile of SCAP upon exposure to oral bacteria and their supernatants—Fusobacterium nucleatum and Enterococcus faecalis—as well as to establish their effect on the osteogenic and immunogenic potentials of SCAP. Further, we target the presence of key proteins of the Wnt/β-Catenin, TGF-β, and NF-κB signaling pathways, which play a crucial role in adult osteogenic differentiation of mesenchymal stem cells, using the Western blot (WB) technique. The membrane-based sandwich immunoassay and transcriptomic analysis showed that, under the influence of F. nucleatum (both bacteria and supernatant), the production of pro-inflammatory cytokines IL-6, IL-8, and MCP-1 occurred, which was also confirmed at the mRNA level. Conversely, E. faecalis reduced the secretion of the aforementioned cytokines at both mRNA and protein levels. WB analysis showed that SCAP co-cultivation with E. faecalis led to a decrease in the level of the key proteins of the Wnt/β-Catenin and NF-κB signaling pathways: β-Catenin (p = 0.0068 *), LRP-5 (p = 0.0059 **), and LRP-6 (p = 0.0329 *), as well as NF-kB (p = 0.0034 **) and TRAF6 (p = 0.0285 *). These results suggest that oral bacteria can up- and downregulate the immune and inflammatory responses of SCAP, as well as influence the osteogenic potential of SCAP, which may negatively regulate the success of RET.
Collapse
|
67
|
Hwang G. In it together: Candida-bacterial oral biofilms and therapeutic strategies. ENVIRONMENTAL MICROBIOLOGY REPORTS 2022; 14:183-196. [PMID: 35218311 PMCID: PMC8957517 DOI: 10.1111/1758-2229.13053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 05/16/2023]
Abstract
Under natural environmental settings or in the human body, the majority of microorganisms exist in complex polymicrobial biofilms adhered to abiotic and biotic surfaces. These microorganisms exhibit symbiotic, mutualistic, synergistic, or antagonistic relationships with other species during biofilm colonization and development. These polymicrobial interactions are heterogeneous, complex and hard to control, thereby often yielding worse outcomes than monospecies infections. Concerning fungi, Candida spp., in particular, Candida albicans is often detected with various bacterial species in oral biofilms. These Candida-bacterial interactions may induce the transition of C. albicans from commensal to pathobiont or dysbiotic organism. Consequently, Candida-bacterial interactions are largely associated with various oral diseases, including dental caries, denture stomatitis, periodontitis, peri-implantitis, and oral cancer. Given the severity of oral diseases caused by cross-kingdom consortia that develop hard-to-remove and highly drug-resistant biofilms, fundamental research is warranted to strategically develop cost-effective and safe therapies to prevent and treat cross-kingdom interactions and subsequent biofilm development. While studies have shed some light, targeting fungal-involved polymicrobial biofilms has been limited. This mini-review outlines the key features of Candida-bacterial interactions and their impact on various oral diseases. In addition, current knowledge on therapeutic strategies to target Candida-bacterial polymicrobial biofilms is discussed.
Collapse
Affiliation(s)
- Geelsu Hwang
- Department of Preventive and Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Corresponding Author: Geelsu Hwang,
| |
Collapse
|
68
|
Geremias TC, Batistella MA, Magini RRS, Guelli U. de Souza SMA, Franco CV, Barbosa LCA, Pereira UA, Hinestroza JP, Pimenta AL, Ulson de Souza AA. Functionalization of poly(lactic‐co‐glycolic acid) nanofibrous membranes with antibiofilm compounds. CAN J CHEM ENG 2022. [DOI: 10.1002/cjce.24115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Thaise C. Geremias
- Centre for Research on Dental Implants (CEPID), School of Dentistry (ODT) Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| | - Marcos A. Batistella
- Laboratory of Mass Transfer (LABMASSA), Department of Chemical and Food Engineering (EQA) Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| | - Ricardo R. S. Magini
- Centre for Research on Dental Implants (CEPID), School of Dentistry (ODT) Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| | - Selene M. A. Guelli U. de Souza
- Laboratory of Mass Transfer (LABMASSA), Department of Chemical and Food Engineering (EQA) Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| | - Cesar V. Franco
- Laboratory of Inorganic Synthesis and Nanoparticles (LabSiN), Department of Chemistry Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| | - Luiz C. A. Barbosa
- Department of Chemistry Universidade Federal de Minas Gerais, Campus Pampulha Belo Horizonte Brazil
| | - Ulisses A. Pereira
- Institute of Agricultural Sciences Universidade Federal de Minas Gerais, Campus Regional de Montes Claros Montes Claros Brazil
| | | | - Andréa L. Pimenta
- Department of Biology, ERRMECe, Université de Cergy Pontoise Maison Internationale de la Recherche Neuville sur Oise Cedex France
- Integrated Laboratories Technologies (InteLab), Department of Chemical and Food Engineering (EQA) Federal University of Santa Catarina (UFSC) Florianópolis SC 88040‐970 Brazil
| | - Antônio A. Ulson de Souza
- Laboratory of Mass Transfer (LABMASSA), Department of Chemical and Food Engineering (EQA) Federal University of Santa Catarina (UFSC) Florianópolis Brazil
| |
Collapse
|
69
|
Vazquez-Munoz R, Thompson A, Russell JT, Sobue T, Zhou Y, Dongari-Bagtzoglou A. Insights From the Lactobacillus johnsonii Genome Suggest the Production of Metabolites With Antibiofilm Activity Against the Pathobiont Candida albicans. Front Microbiol 2022; 13:853762. [PMID: 35330775 PMCID: PMC8940163 DOI: 10.3389/fmicb.2022.853762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Lactobacillus johnsonii is a probiotic bacterial species with broad antimicrobial properties; however, its antimicrobial activities against the pathobiont Candida albicans are underexplored. The aim of this study was to study the interactions of L. johnsonii with C. albicans and explore mechanisms of bacterial anti-fungal activities based on bacterial genomic characterization coupled with experimental data. We isolated an L. johnsonii strain (MT4) from the oral cavity of mice and characterized its effect on C. albicans growth in the planktonic and biofilm states. We also identified key genetic and phenotypic traits that may be associated with a growth inhibitory activity exhibited against C. albicans. We found that L. johnsonii MT4 displays pH-dependent and pH-independent antagonistic interactions against C. albicans, resulting in inhibition of C. albicans planktonic growth and biofilm formation. This antagonism is influenced by nutrient availability and the production of soluble metabolites with anticandidal activity.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Angela Thompson
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Jordan T Russell
- Department of Psychiatry/Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Takanori Sobue
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Yanjiao Zhou
- Department of Psychiatry/Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
70
|
Li B, Cai Q, Wang Z, Qiao S, Ou Y, Ma R, Luo C, Meng W. D-arginine Enhances the Effect of Alpha-Amylase on Disassembling Actinomyces viscosus Biofilm. Front Bioeng Biotechnol 2022; 10:864012. [PMID: 35309977 PMCID: PMC8927782 DOI: 10.3389/fbioe.2022.864012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/17/2022] [Indexed: 11/25/2022] Open
Abstract
Peri-implantitis is the leading cause of dental implant failure, initially raised by biofilm accumulation on the implant surface. During the development of biofilm, Actinomyces viscosus (A. viscosus) plays a pivotal role in initial attachment as well as the bacterial coaggregation of multispecies pathogens. Hence, eliminating the A. viscosus-associated biofilm is fundamental for the regeneration of the lost bone around implants. Whereas clinical evidence indicated that antimicrobials and debridement did not show significant effects on the decontamination of biofilm on the implant surface. In this study, alpha-amylase was investigated for its effects on disassembling A. viscosus biofilm. Then, in order to substantially disperse biofilm under biosafety concentration, D-arginine was employed to appraise its enhancing effects on alpha-amylase. In addition, molecular dynamics simulations and molecular docking were conducted to elucidate the mechanism of D-arginine enhancing alpha-amylase. 0.1–0.5% alpha-amylase showed significant effects on disassembling A. viscosus biofilm, with definite cytotoxicity toward MC3T3-E1 cells meanwhile. Intriguingly, 8 mM D-arginine drastically enhanced the eradication of A. viscosus biofilm biomass by 0.01% alpha-amylase with biosafety in 30 min. The exopolysaccharides of biofilm were also thoroughly hydrolyzed by 0.01% alpha-amylase with 8 mM D-arginine. The biofilm thickness and integrity were disrupted, and the exopolysaccharides among the extracellular matrix were elusive. Molecular dynamics simulations showed that with the hydrogen bonding of D-arginine to the catalytic triad and calcium-binding regions of alpha-amylase, the atom fluctuation of the structure was attenuated. The distances between catalytic triad were shortened, and the calcium-binding regions became more stable. Molecular docking scores revealed that D-arginine facilitated the maltotetraose binding process of alpha-amylase. In conclusion, these results demonstrate that D-arginine enhances the disassembly effects of alpha-amylase on A. viscosus biofilm through potentiating the catalytic triad and stabilizing the calcium-binding regions, thus providing a novel strategy for the decontamination of biofilm contaminated implant surface.
Collapse
Affiliation(s)
- Baosheng Li
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Changchun, China
| | - Qing Cai
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zixuan Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Changchun, China
| | - Shuwei Qiao
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Changchun, China
| | - Yanzhen Ou
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Changchun, China
| | - Rui Ma
- Xinjiang Laboratory of Phase Transitions and Microstructures in Condensed Matters, College of Physical Science and Technology, Yili Normal University, Yining, China
| | - Chuanfu Luo
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, China
| | - Weiyan Meng
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
- *Correspondence: Weiyan Meng,
| |
Collapse
|
71
|
Chen Y, Huang Z, Tang Z, Huang Y, Huang M, Liu H, Ziebolz D, Schmalz G, Jia B, Zhao J. More Than Just a Periodontal Pathogen –the Research Progress on Fusobacterium nucleatum. Front Cell Infect Microbiol 2022; 12:815318. [PMID: 35186795 PMCID: PMC8851061 DOI: 10.3389/fcimb.2022.815318] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Fusobacterium nucleatum is a common oral opportunistic bacterium that can cause different infections. In recent years, studies have shown that F. nucleatum is enriched in lesions in periodontal diseases, halitosis, dental pulp infection, oral cancer, and systemic diseases. Hence, it can promote the development and/or progression of these conditions. The current study aimed to assess research progress in the epidemiological evidence, possible pathogenic mechanisms, and treatment methods of F. nucleatum in oral and systemic diseases. Novel viewpoints obtained in recent studies can provide knowledge about the role of F. nucleatum in hosts and a basis for identifying new methods for the diagnosis and treatment of F. nucleatum-related diseases.
Collapse
Affiliation(s)
- Yuanxin Chen
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhijie Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zhengming Tang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yisheng Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Mingshu Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hongyu Liu
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Dirk Ziebolz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - Gerhard Schmalz
- Department of Cariology, Endodontology and Periodontology, University of Leipzig, Leipzig, Germany
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Bo Jia, ; Jianjiang Zhao,
| | - Jianjiang Zhao
- Shenzhen Stomatological Hospital, Southern Medical University, Shenzhen, China
- *Correspondence: Bo Jia, ; Jianjiang Zhao,
| |
Collapse
|
72
|
Liu T, Yang R, Zhou J, Lu X, Yuan Z, Wei X, Guo L. Interactions Between Streptococcus gordonii and Fusobacterium nucleatum Altered Bacterial Transcriptional Profiling and Attenuated the Immune Responses of Macrophages. Front Cell Infect Microbiol 2022; 11:783323. [PMID: 35071038 PMCID: PMC8776643 DOI: 10.3389/fcimb.2021.783323] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 12/15/2021] [Indexed: 12/24/2022] Open
Abstract
Interspecies coaggregation promotes transcriptional changes in oral bacteria, affecting bacterial pathogenicity. Streptococcus gordonii (S. gordonii) and Fusobacterium nucleatum (F. nucleatum) are common oral inhabitants. The present study investigated the transcriptional profiling of S. gordonii and F. nucleatum subsp. polymorphum in response to the dual-species coaggregation using RNA-seq. Macrophages were infected with both species to explore the influence of bacterial coaggregation on both species' abilities to survive within macrophages and induce inflammatory responses. Results indicated that, after the 30-min dual-species coaggregation, 116 genes were significantly up-regulated, and 151 genes were significantly down-regulated in S. gordonii; 97 genes were significantly down-regulated, and 114 genes were significantly up-regulated in F. nucleatum subsp. polymorphum. Multiple S. gordonii genes were involved in the biosynthesis and export of cell-wall proteins and carbohydrate metabolism. F. nucleatum subsp. polymorphum genes were mostly associated with translation and protein export. The coaggregation led to decreased expression levels of genes associated with lipopolysaccharide and peptidoglycan biosynthesis. Coaggregation between S. gordonii and F. nucleatum subsp. polymorphum significantly promoted both species' intracellular survival within macrophages and attenuated the production of pro-inflammatory cytokines IL-6 and IL-1β. Physical interactions between these two species promoted a symbiotic lifestyle and repressed macrophage's killing and pro-inflammatory responses.
Collapse
Affiliation(s)
- Tingjun Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Ruiqi Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Jiani Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xianjun Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Zijian Yuan
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| | - Lihong Guo
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, China
| |
Collapse
|
73
|
Jacob KM, Reguera G. Competitive advantage of oral streptococci for colonization of the middle ear mucosa. Biofilm 2022; 4:100067. [PMID: 35146417 PMCID: PMC8818537 DOI: 10.1016/j.bioflm.2022.100067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 12/17/2021] [Accepted: 01/10/2022] [Indexed: 10/29/2022] Open
|
74
|
Fungi—A Component of the Oral Microbiome Involved in Periodontal Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:113-138. [DOI: 10.1007/978-3-030-96881-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
75
|
Redox Sensing Modulates the Activity of the ComE Response Regulator of Streptococcus mutans. J Bacteriol 2021; 203:e0033021. [PMID: 34516285 DOI: 10.1128/jb.00330-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus mutans, a dental pathogen, encodes the ComDE two-component system comprised of a histidine kinase (ComD) and a response regulator (ComE). This system is necessary for production of bacteriocins and development of genetic competence. ComE interacts with its cognate promoters to activate the transcription of bacteriocin and competence-related genes. Previous transcriptomic studies indicated that expressions of bacteriocin genes were upregulated in the presence of oxygen. To understand the relationship between the aerobic condition and bacteriocin expression, we analyzed the S. mutans ComE sequence and its close homologs. Surprisingly, we noticed the presence of cysteine (Cys) residues located at positions 200 and 229, which are highly conserved among the ComE homologs. Here, we investigated the role of Cys residues of S. mutans ComE in the activation of bacteriocin transcription using the PnlmA promoter that expresses bacteriocin NlmA. We constructed both single mutants and double mutants by replacing the Cys residues with serine and performed complementation assays. We observed that the presence of Cys residues is essential for PnlmA activation. With purified ComE mutant proteins, we found that ComE double mutants displayed a nearly 2-fold lower association rate than wild-type ComE. Furthermore, 1-anilinonaphthalene-8-sulfonic acid (ANS) fluorescence studies indicated that the double mutants displayed wider conformation changes than wild-type ComE. Finally, we demonstrated that close streptococcal ComE homologs successfully activate the PnlmA expression in vivo. This is the first report suggesting that S. mutans ComE and its homologs can sense the oxidation status of the cell, a phenomenon similar to the AgrA system of Staphylococcus aureus but with different outcomes. IMPORTANCE Streptococci are an important species that prefer to grow under anaerobic or microaerophilic environments. Studies have shown that streptococci growth in an aerobic environment generates oxidative stress responses by activating various defense systems, including production of antimicrobial peptides called bacteriocins. This study highlights the importance of a two-component response regulator (ComE) that senses the aerobic environment and induces bacteriocin production in Streptococcus mutans, a dental pathogen. We believe increased bacteriocin secretion under aerobic conditions is necessary for survival and colonization of S. mutans in the oral cavity by inhibiting other competing organisms. Redox sensing by response regulator might be a widespread phenomenon since two other ComE homologs from pathogenic streptococci that inhabit diverse environmental niches also perform a similar function.
Collapse
|
76
|
Hong P, Yang K, Shu Y, Xiao B, Wu H, Xie Y, Gu Y, Qian F, Wu X. Efficacy of auto-aggregating aerobic denitrifiers with coaggregation traits for bioaugmentation performance in biofilm-formation and nitrogen-removal. BIORESOURCE TECHNOLOGY 2021; 337:125391. [PMID: 34139566 DOI: 10.1016/j.biortech.2021.125391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/02/2021] [Accepted: 06/05/2021] [Indexed: 06/12/2023]
Abstract
To promote efficiency nitrogen-rich wastewater treatment from a sequencing batch biofilm reactor (SBBR), three aerobic denitrifiers (Pseudomonas mendocinaIHB602, Methylobacterium gregansDC-1 and Pseudomonas stutzeriIHB618) with dual-capacities of strong auto-aggregation and high nitrogen removal efficiency were studied. The aggregation index analysis indicated that coaggregation of the three strains co-existed was better when compared with one or two strains grown alone. Optimal coaggregation strains were used to bioaugmente a test reactor (SBBRT), which exhibited a shorter time for biofilm-formation than uninoculated control reactor (SBBRC). With different influent ammonia-N loads (150, 200 and 300 mg·L-1), the average ammonia-N and nitrate-N removal efficiency were all higher than that in SBBRC, as well as a lower nitrite-N accumulation. Microbial community structure analysis revealed coaggregation strains may successfully colonize in the bioreactor and be very tolerant of high nitrogen concentrations, and contribute to the high efficiency of inorganic nitrogen-removal and biofilm-formation.
Collapse
Affiliation(s)
- Pei Hong
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China; Key Laboratory of Algal Biology of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Keyin Yang
- Key Laboratory for the Conservation and Utilization of Important Biological Resources of Anhui Province, Anhui Normal University, Wuhu 241000, China
| | - Yilin Shu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Bangding Xiao
- Key Laboratory of Algal Biology of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Hailong Wu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Yunyun Xie
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Yali Gu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Fangping Qian
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Anhui Normal University, Wuhu 241000, China
| | - Xingqiang Wu
- Key Laboratory of Algal Biology of the Chinese Academy of Sciences, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.
| |
Collapse
|
77
|
Ali Mohammed MM, Pettersen VK, Nerland AH, Wiker HG, Bakken V. Label-free quantitative proteomic analysis of the oral bacteria Fusobacterium nucleatum and Porphyromonas gingivalis to identify protein features relevant in biofilm formation. Anaerobe 2021; 72:102449. [PMID: 34543761 DOI: 10.1016/j.anaerobe.2021.102449] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/24/2021] [Accepted: 09/14/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The opportunistic pathogens Fusobacterium nucleatum and Porphyromonas gingivalis are Gram-negative bacteria associated with oral biofilm and periodontal disease. This study investigated interactions between F. nucleatum and P. gingivalis proteomes with the objective to identify proteins relevant in biofilm formation. METHODS We applied liquid chromatography-tandem mass spectrometry to determine the expressed proteome of F. nucleatum and P. gingivalis cells grown in biofilm or planktonic culture, and as mono- and dual-species models. The detected proteins were classified into functional categories and their label-free quantitative (LFQ) intensities statistically compared. RESULTS The proteomic analyses detected 1,322 F. nucleatum and 966 P. gingivalis proteins, including abundant virulence factors. Using univariate statistics, we identified significant changes between biofilm and planktonic culture (p-value ≤0.05) in 0,4% F. nucleatum, 7% P. gingivalis, and 14% of all proteins in the dual-species model. For both species, proteins involved in vitamin B2 (riboflavin) metabolism had significantly increased levels in biofilm. In both mono- and dual-species biofilms, P. gingivalis increased the production of proteins for translation, oxidation-reduction, and amino acid metabolism compared to planktonic cultures. However, when we compared LFQ intensities between mono- and dual-species, over 90% of the significantly changed P. gingivalis proteins had their levels reduced in biofilm and planktonic settings of the dual-species model. CONCLUSIONS The findings suggest that P. gingivalis reduces the production of multiple proteins because of the F. nucleatum presence. The results highlight the complex interactions of bacteria contributing to oral biofilms, which need to be considered in the design of prevention strategies.
Collapse
Affiliation(s)
| | | | - Audun H Nerland
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Harald G Wiker
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Vidar Bakken
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
78
|
Villar CC, Dongari-Bagtzoglou A. Fungal diseases: Oral dysbiosis in susceptible hosts. Periodontol 2000 2021; 87:166-180. [PMID: 34463992 DOI: 10.1111/prd.12378] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The oral cavity is colonized by a large number of microorganisms that are referred to collectively as the oral microbiota. These indigenous microorganisms have evolved in symbiotic relationships with the oral mucosal immune system and are involved in maintaining homeostasis in the oral cavity. Although Candida species are commonly found in the healthy oral cavity without causing infection, these fungi can become pathogenic. Recents advances indicate that the development of oral candidiasis is driven both by Candida albicans overgrowth in a dysbiotic microbiome and by disturbances in the host's immune system. Perturbation of the oral microbiota triggered by host-extrinsic (ie, medications), host-intrinsic (ie, host genetics), and microbiome-intrinsic (ie, microbial interactions) factors may increase the risk of oral candidiasis. In this review, we provide an overview of the oral mycobiome, with a particular focus on the interactions of Candida albicans with some of the most common oral bacteria and the oral mucosal immune system. Also, we present a summary of our current knowledge of the host-intrinsic and host-extrinsic factors that can predispose to oral candidiasis.
Collapse
Affiliation(s)
- Cristina Cunha Villar
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Anna Dongari-Bagtzoglou
- Department of Oral Health and Diagnostic Sciences, University of Connecticut School of Dental Medicine, Farmington, CT, USA
| |
Collapse
|
79
|
Tett A, Pasolli E, Masetti G, Ercolini D, Segata N. Prevotella diversity, niches and interactions with the human host. Nat Rev Microbiol 2021; 19:585-599. [PMID: 34050328 PMCID: PMC11290707 DOI: 10.1038/s41579-021-00559-y] [Citation(s) in RCA: 370] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
The genus Prevotella includes more than 50 characterized species that occur in varied natural habitats, although most Prevotella spp. are associated with humans. In the human microbiome, Prevotella spp. are highly abundant in various body sites, where they are key players in the balance between health and disease. Host factors related to diet, lifestyle and geography are fundamental in affecting the diversity and prevalence of Prevotella species and strains in the human microbiome. These factors, along with the ecological relationship of Prevotella with other members of the microbiome, likely determine the extent of the contribution of Prevotella to human metabolism and health. Here we review the diversity, prevalence and potential connection of Prevotella spp. in the human host, highlighting how genomic methods and analysis have improved and should further help in framing their ecological role. We also provide suggestions for future research to improve understanding of the possible functions of Prevotella spp. and the effects of the Western lifestyle and diet on the host-Prevotella symbiotic relationship in the context of maintaining human health.
Collapse
Affiliation(s)
- Adrian Tett
- Centre for Microbiology and Environmental Systems Science, University of Vienna, Vienna, Austria
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | | | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy.
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy.
- European Institute of Oncology IRCCS, Milan, Italy.
| |
Collapse
|
80
|
Dewake N, Ma X, Sato K, Nakatsu S, Yoshimura K, Eshita Y, Fujinaka H, Yano Y, Yoshinari N, Yoshida A. β-Glycyrrhetinic acid inhibits the bacterial growth and biofilm formation by supragingival plaque commensals. Microbiol Immunol 2021; 65:343-351. [PMID: 33860563 DOI: 10.1111/1348-0421.12884] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/30/2022]
Abstract
β-Glycyrrhetinic acid (BGA) is a natural antibacterial agent. Previous studies reported that BGA has antibacterial effects against several bacteria. This study evaluated the effects of BGA on the regulation of supragingival plaque bacteria. First, the minimum inhibitory concentrations (MICs) of BGA against oral bacteria were measured. Next, the minimum concentrations for inhibition of biofilm formation were evaluated against Streptococcus mutans and Streptococcus sobrinus, possessing insoluble glucan synthesis abilities. The MICs of biofilm formation by these bacteria ranged from 1/8 to 2× MIC. Furthermore, the inhibition effects of BGA against the coaggregation of Porphyromonas gingivalis and Streptococcus gordonii were evaluated. BGA at 32 or 64 μg/mL inhibited the coaggregation of these bacteria after a 30 min incubation. Lastly, the inhibition effects of BGA against human supragingival plaque bacteria were evaluated. Human supragingival plaque samples were obtained from 12 healthy donors. The inhibition effects of BGA against biofilm formation by these plaque bacteria were evaluated. Of 12 samples, the biofilm formation by 11 was significantly attenuated by 128-256 μg/mL of BGA. The number of colony forming units in these biofilms was also significantly attenuated. In conclusion, it was revealed that BGA inhibits the growth and biofilm formation of bacteria, furthermore, the same effect was confirmed with supragingival plaque bacteria. BGA is a good candidate for a natural agent that prevents the outbreak and progression of periodontal disease because it suppresses not only the growth and biofilm formation of bacteria, but also the coaggregation of P. gingivalis with plaque bacteria.
Collapse
Affiliation(s)
- Nanae Dewake
- Department of Periodontology, Faculty of Dentistry, Matsumoto Dental University, Shiojiri, Nagano, Japan
| | - Xiangtao Ma
- Department of Periodontology, Faculty of Dentistry, Matsumoto Dental University, Shiojiri, Nagano, Japan
| | - Kayo Sato
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Susumu Nakatsu
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Kenji Yoshimura
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Yoshiyuki Eshita
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Hidetake Fujinaka
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Yoshitaka Yano
- Personal Health Care Product Research, Kao Corporation, Tokyo, Japan
| | - Nobuo Yoshinari
- Department of Periodontology, Faculty of Dentistry, Matsumoto Dental University, Shiojiri, Nagano, Japan
| | - Akihiro Yoshida
- Department of Oral Microbiology, Faculty of Dentistry, Matsumoto Dental University, Shiojiri, Nagano, Japan
| |
Collapse
|
81
|
Kawajiri J, Nagata K, Nakamura A, Fujieda A, Ino K, Nomura J, Arai N, Ohishi K, Katayama N, Nakase K. Clinical utility of oral management in allogeneic hematopoietic stem cell transplantation recipients: microbiological evidence based on molecular analysis of oral bacteria. Support Care Cancer 2021; 30:757-764. [PMID: 34374846 DOI: 10.1007/s00520-021-06462-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/22/2021] [Indexed: 12/01/2022]
Abstract
PURPOSE This study aimed to clarify the clinical utility of oral management to prevent bloodstream infections by oral bacteria microbiologically in patients undergoing allogeneic hematopoietic stem cell transplantation (ASCT). METHODS Ten consecutive patients with hematological malignancies undergoing ASCT were enrolled in this study. We implemented dental treatments before transplantation, if required, and carried out oral hygiene instructions and oral management every other day after transplantation. Molecular analysis of bacterial DNA for seven oral species using a polymerase chain reaction (PCR) assay was performed for oral samples and peripheral blood once a week for 3 weeks after transplantation. RESULTS Periodontitis was found in all 10 patients (mild grade in 3 and middle grade in 7) for whom basic periodontal therapy was conducted. Necessary dental procedures, including tooth extraction were performed in 5 patients. After transplantation, oral mucositis occurred in 10 patients (grade 1 in 3, grade 2 in 2, and grade 3 in 5) for whom oral hygiene instruction and oral care were continued every other day. PCR-identified three to six bacterial species in oral samples from nine patients, but none in peripheral blood from any patient during the observation period. CONCLUSIONS Our study suggests that oral management could prevent bloodstream infections by oral bacteria in ASCT recipients despite the existence of periodontitis or oral mucositis. Its utility was confirmed by microbiological evidence based on molecular data.
Collapse
Affiliation(s)
- Jumpei Kawajiri
- Faculty of Nursing, Suzuka University of Medical Science, Mie, Japan.,Department of Translational Medical Science, Mie University Graduate School of Medicine, Mie, Japan
| | - Kokoro Nagata
- Department of Oral and Maxillofacial Surgery, Mie University Hospital, Mie, Japan
| | - Akiko Nakamura
- Central Clinical Laboratories, Mie University Hospital, Mie, Japan
| | - Atsushi Fujieda
- Department of Hematology and Oncology, Mie University Hospital, Mie, Japan
| | - Kazuko Ino
- Department of Hematology and Oncology, Mie University Hospital, Mie, Japan
| | - Jouji Nomura
- Department of Oral and Maxillofacial Surgery, Mie University Hospital, Mie, Japan
| | - Naoya Arai
- Department of Oral and Maxillofacial Surgery, Mie University Hospital, Mie, Japan
| | - Kohshi Ohishi
- Transfusion Service, Mie University Hospital, Mie, Japan
| | - Naoyuki Katayama
- Faculty of Nursing, Suzuka University of Medical Science, Mie, Japan.,Department of Hematology and Oncology, Mie University Hospital, Mie, Japan
| | - Kazunori Nakase
- Department of Hematology and Oncology, Mie University Hospital, Mie, Japan. .,Cancer Center, Mie University Hospital, Mie, Japan.
| |
Collapse
|
82
|
Marya A, Steier L, Karobari MI, Venugopal A. Benefits of Using Fluorescence Induced Theragnosis in Fixed Orthodontic Therapy: Status, Technology and Future Trends. Dent J (Basel) 2021; 9:90. [PMID: 34436002 PMCID: PMC8393472 DOI: 10.3390/dj9080090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 01/05/2023] Open
Abstract
Dental biofilm is often found to be the source of bacteria that releases toxins, peptides, lipopolysaccharides as well as organic acids, which lead to gingival inflammation and tooth caries. Further, the persistent plaque may result in the continued destruction of the surrounding soft and hard tissues. During fixed orthodontic therapy, arch-wires, brackets, and elastic modules have been shown to be sites of significant plaque accumulation, making it difficult for a patient to maintain proper oral hygiene. The problem most dentists face is that they cannot visualize this biofilm completely to be able to carry out efficient plaque removal. Visual assessment is, to date, the most common method for plaque visualization, and various indexes have been demonstrated to be sufficient for quantification of the amount of plaque present. However, the problem is that visual assessments are inconsistent, operator dependent and often subjective, which can lead to inconsistency in results. Fluorescence is one such method that can be explored for its use in effective plaque identification and removal. Literature has it that dentists and patients find it particularly useful for monitoring oral hygiene status during treatment. Fluorescence has the capability of offering clinical orthodontists and researchers a new method of detection of demineralization during orthodontic treatment, furthermore, for efficient removal of orthodontic adhesive cements, fluorescent light may be used in conjunction with high-speed burs to deliver fast, less time consuming, and safer results. The benefit of direct visual treatment using fluorescence enhanced theragnosis is that the patient receives controlled and guided therapy. It has multiple benefits, such as early diagnosis of caries, biofilm identification, and even helps to achieve improved treatment outcomes by better resin selection for esthetic procedures.
Collapse
Affiliation(s)
- Anand Marya
- Department of Orthodontics, Faculty of Dentistry, University of Puthisastra, Phnom Penh 12211, Cambodia
- Department of Orthodontics, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College, Saveetha University, Chennai 600077, India;
| | - Liviu Steier
- Department of Restorative Dentistry, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Mohmed Isaqali Karobari
- Conservative Dentistry Unit, Health Campus, School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Kelantan, Malaysia;
| | - Adith Venugopal
- Department of Orthodontics, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College, Saveetha University, Chennai 600077, India;
| |
Collapse
|
83
|
Structure-function characterization of Streptococcus intermedius surface antigen Pas. J Bacteriol 2021; 203:e0017521. [PMID: 34339301 DOI: 10.1128/jb.00175-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus intermedius, an oral commensal bacterium, is found at various sites including subgingival dental plaque, purulent infections, and in cystic fibrosis lungs. Oral streptococci utilize proteins on their surface to adhere to tissues and/or surfaces localizing the bacteria, which subsequently leads to the development of biofilms, colonization and infection. Among the 19 genomically annotated cell-wall attached surface proteins on S. intermedius, Pas is an adhesin that belongs to the Antigen I/II (AgI/II) family. Here we have structurally and functionally characterized Pas, particularly focusing on its microbial-host as well as microbial-microbial interactions. The crystal structures of VPas and C123Pas show high similarity with AgI/II of S. mutans. VPas hosts a conserved metal binding site, and likewise the C123Pas structure retains its conserved metal binding sites and isopeptide bonds within its three DEv-IgG domains. Pas interacts with nanomolar affinity to lung alveolar glycoprotein 340 (Gp340), its scavenger receptor cysteine rich domains (SRCRs) and with fibrinogen. Both Candida albicans and Pseudomonas aeruginosa, the opportunistic pathogens that cohabitate with S. intermedius in the lungs of CFTR patients were studied in dual-species biofilm studies. The Pas deficient mutant (Δpas) displayed significant reduction in dual biofilm formation with C. albicans. In similar studies with P. aeruginosa, Pas did not mediate the biofilm formation with either the acute isolate (PAO1), or the chronic isolate (FRD1). However, the Sortase A deficient mutant (ΔsrtA) displayed reduced biofilm formation with both C. albicans and P. aeruginosa FRD1. Taken together, our findings highlight the role of Pas in both microbial-host and interkingdom interactions and expose its potential role in disease outcomes. Importance Streptococcus intermedius, an oral commensal bacterium, has been clinically observed in subgingival dental plaque, purulent infections, and in cystic fibrosis lungs. In this study, we have (a) determined the crystal structure of the V- and C-regions of Pas; (b) shown that its surface protein Pas adheres to fibrinogen, which could potentially ferry the microbe through the blood stream from the oral cavity; (c) characterized Pas's high affinity adherence to lung alveolar protein Gp340 that could fixate the microbe on lung epithelial cells; and (d) most importantly shown that these surface proteins on the oral commensal S. intermedius enhances biofilms of known pathogens Candida albicans and Pseudomonas aeruginosa.
Collapse
|
84
|
Boesch M, Baty F, Rothschild SI, Tamm M, Joerger M, Früh M, Brutsche MH. Tumour neoantigen mimicry by microbial species in cancer immunotherapy. Br J Cancer 2021; 125:313-323. [PMID: 33824481 PMCID: PMC8329167 DOI: 10.1038/s41416-021-01365-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/02/2021] [Accepted: 03/10/2021] [Indexed: 02/08/2023] Open
Abstract
Tumour neoantigens arising from cancer-specific mutations generate a molecular fingerprint that has a definite specificity for cancer. Although this fingerprint perfectly discriminates cancer from healthy somatic and germline cells, and is therefore therapeutically exploitable using immune checkpoint blockade, gut and extra-gut microbial species can independently produce epitopes that resemble tumour neoantigens as part of their natural gene expression programmes. Such tumour molecular mimicry is likely not only to influence the quality and strength of the body's anti-cancer immune response, but could also explain why certain patients show favourable long-term responses to immune checkpoint blockade while others do not benefit at all from this treatment. This article outlines the requirement for tumour neoantigens in successful cancer immunotherapy and draws attention to the emerging role of microbiome-mediated tumour neoantigen mimicry in determining checkpoint immunotherapy outcome, with far-reaching implications for the future of cancer immunotherapy.
Collapse
Affiliation(s)
| | - Florent Baty
- Lung Center, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology and Comprehensive Cancer Center, University Hospital of Basel, Basel, Switzerland
| | - Michael Tamm
- Department of Pulmonology, University Hospital of Basel, Basel, Switzerland
| | - Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Martin Früh
- Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
- Department of Medical Oncology, University Hospital Bern, Bern, Switzerland
| | | |
Collapse
|
85
|
In Vivo Biological Evaluation of Biodegradable Nanofibrous Membranes Incorporated with Antibiofilm Compounds. Polymers (Basel) 2021; 13:polym13152457. [PMID: 34372057 PMCID: PMC8347157 DOI: 10.3390/polym13152457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/20/2022] Open
Abstract
Guided bone regeneration involves excluding non-osteogenic cells from the surrounding soft tissues and allowing osteogenic cells originating from native bone to inhabit the defect. The aim of this work was to fabricate, analyze antibiofilm activity and evaluate in vivo biological response of poly (lactic-co-glycolic acid) (PLGA) electrospun membranes incorporated with tea tree oil and furan-2(5H)-one. Samples were exposed to Streptococcus mutans culture and after 48 h incubation, biofilm was evaluated by colony forming units (CFU/mL) followed by scanning electron microscopy. Additionally, seventy-five Balb-C mice were divided into five experimental groups for subcutaneous implantation: tea tree oil loaded PLGA electrospun fiber membrane, furanone loaded PLGA electrospun fiber membrane, neat PLGA electrospun fiber membrane, a commercially available PLGA membrane –Pratix® and Sham (no-membrane implantation). Post implantation period of each experimental group (1, 3 and 9 weeks), samples were collected and processed for by histological descriptive and semiquantitative evaluation. Results showed a significant reduction of bacterial attachment on tea tree oil and furan-2(5H)-one incorporated membranes. Macrophage counts were significant found in all the materials implanted, although giant cells were predominantly associated with electrospun fiber membranes. The incorporation of antibiofilm compounds in nanofibers membranes did not incite inflammatory response significantly different in comparison with pure PLGA electrospun membranes, indicating its potential for development of novel functionalized membranes targeting the inhibition of bacterial biofilms on membrane-grafting materials.
Collapse
|
86
|
Hottmann I, Borisova M, Schäffer C, Mayer C. Peptidoglycan Salvage Enables the Periodontal Pathogen Tannerella forsythia to Survive within the Oral Microbial Community. Microb Physiol 2021; 31:123-134. [PMID: 34107471 DOI: 10.1159/000516751] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/22/2021] [Indexed: 11/19/2022]
Abstract
Tannerella forsythia is an anaerobic, fusiform Gram-negative oral pathogen strongly associated with periodontitis, a multibacterial inflammatory disease that leads to the destruction of the teeth-supporting tissue, ultimately causing tooth loss. To survive in the oral habitat, T. forsythia depends on cohabiting bacteria for the provision of nutrients. For axenic growth under laboratory conditions, it specifically relies on the external supply of N-acetylmuramic acid (MurNAc), which is an essential constituent of the peptidoglycan (PGN) of bacterial cell walls. T. forsythia comprises a typical Gram-negative PGN; however, as evidenced by genome sequence analysis, the organism lacks common enzymes required for the de novo synthesis of precursors of PGN, which rationalizes its MurNAc auxotrophy. Only recently insights were obtained into how T. forsythia gains access to MurNAc in its oral habitat, enabling synthesis of the own PGN cell wall. This report summarizes T. forsythia's strategies to survive in the oral habitat by means of PGN salvage pathways, including recovery of exogenous MurNAc and PGN-derived fragments but also polymeric PGN, which are all derived from cohabiting bacteria either via cell wall turnover or decay of cells. Salvage of polymeric PGN presumably requires the removal of peptides from PGN by an unknown amidase, concomitantly with the translocation of the polymer across the outer membrane. Two recently identified exo-lytic N-acetylmuramidases (Tf_NamZ1 and Tf_NamZ2) specifically cleave the peptide-free, exogenous (nutrition source) PGN in the periplasm and release the MurNAc and disaccharide substrates for the transporters Tf_MurT and Tf_AmpG, respectively, whereas the peptide-containing, endogenous (the self-cell wall) PGN stays unattached. This review also outlines how T. forsythia synthesises the PGN precursors UDP-MurNAc and UDP-N-acetylglucosamine (UDP-GlcNAc), involving homologs of the Pseudomonas sp. recycling enzymes AmgK/MurU and a monofunctional uridylyl transferase (named Tf_GlmU*), respectively.
Collapse
Affiliation(s)
- Isabel Hottmann
- Interfaculty Institute of Microbiology and Infection Medicine, Organismic Interactions/Glycobiology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Marina Borisova
- Interfaculty Institute of Microbiology and Infection Medicine, Organismic Interactions/Glycobiology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Christina Schäffer
- Department of NanoBiotechnology, NanoGlycobiology Unit, Universität für Bodenkultur Wien, Vienna, Austria
| | - Christoph Mayer
- Interfaculty Institute of Microbiology and Infection Medicine, Organismic Interactions/Glycobiology, Eberhard Karls Universität Tübingen, Tübingen, Germany
| |
Collapse
|
87
|
Frazão Câmara JV, Araujo TT, Mendez DAC, da Silva NDG, de Medeiros FF, Santos LA, de Souza Carvalho T, Reis FN, Martini T, Moraes SM, Shibao PYT, Groisman S, Magalhães AC, Henrique-Silva F, Buzalaf MAR. Effect of a sugarcane cystatin on the profile and viability of microcosm biofilm and on dentin demineralization. Arch Microbiol 2021; 203:4133-4139. [PMID: 34059945 DOI: 10.1007/s00203-021-02403-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/27/2022]
Abstract
To analyze the effect of a sugarcane cystatin (CaneCPI-5) on the microbial profile and viability, as well as on the prevention of dentin demineralization using a microcosm biofilm model. Ninety bovine dentine specimens were divided into five experimental groups according with the solution they were treated for 60 s: (1) PBS (negative control), (2) 0.12% chlorhexidine (positive control), (3) Fluoride (500 ppm F, as NaF), (4) 0.025 mg/ml CaneCPI-5, and (5) 0.05 mg/ml CaneCPI-5. Specimens were incubated with inoculum (McBain's saliva plus human saliva) in the first 8 h, and from then on, they were exposed to McBain saliva containing sucrose and daily treated (60 s) with the solutions for 5 days. Resazurin and colony-forming unit counting assays were performed. Dentin demineralization was measured by transverse micro-radiography (TMR). 0.12% chlorhexidine significantly reduced the metabolic activity of the microcosm biofilm in relation to the negative control and treated groups (p < 0.01). CHX and F significantly reduced the counts of total microorganisms, mutans group streptococci, and lactobacilli when compared with the negative control. None of the treatments was able to significantly reduce dentin demineralization in comparison with the negative control. In the model evaluated, CaneCPI-5 neither altered the microcosm biofilm profile and viability nor protected dentin against demineralization.
Collapse
Affiliation(s)
- João Victor Frazão Câmara
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil.
| | - Tamara Teodoro Araujo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Daniela Alejandra Cusicanqui Mendez
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Natara Dias Gomes da Silva
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Felipe Fávaro de Medeiros
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Lethycia Almeida Santos
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Thamyris de Souza Carvalho
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Fabiana Navas Reis
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Tatiana Martini
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Samanta Mascarenhas Moraes
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | | | - Sonia Groisman
- School of Dentistry, Federal University of Rio de Janeiro, Cidade Universitária da Universidade Federal Do Rio de Janeiro, R. Prof. Rodolpho Paulo Rocco, 325, Rio de Janeiro, RJ, 21941-617, Brazil
| | - Ana Carolina Magalhães
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| | - Flavio Henrique-Silva
- Federal University of São Carlos, Rod. Washington Luiz, s/n, São Carlos, SP, 13565-905, Brazil
| | - Marília Afonso Rabelo Buzalaf
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75-Vila Regina, Bauru, SP, 17011-220, Brazil
| |
Collapse
|
88
|
Ben Lagha A, Maquera Huacho P, Grenier D. A cocoa (Theobroma cacao L.) extract impairs the growth, virulence properties, and inflammatory potential of Fusobacterium nucleatum and improves oral epithelial barrier function. PLoS One 2021; 16:e0252029. [PMID: 34029354 PMCID: PMC8143394 DOI: 10.1371/journal.pone.0252029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022] Open
Abstract
Fusobacterium nucleatum is associated with many conditions and diseases, including periodontal diseases that affect tooth-supporting tissues. The aim of the present study was to investigate the effects of a cocoa extract (Theobroma cacao L.) on F. nucleatum with respect to growth, biofilm formation, adherence, and hydrogen sulfide (H2S) production. The anti-inflammatory properties and the effect on epithelial barrier function of the cocoa extract were also assessed. The cocoa extract, whose major phenolic compound is epicatechin, dose-dependently inhibited the growth, biofilm formation, adherence properties (basement membrane matrix, oral epithelial cells), and H2S production of F. nucleatum. It also decreased IL-6 and IL-8 production by F. nucleatum-stimulated oral epithelial cells and inhibited F. nucleatum-induced NF-κB activation in monocytes. Lastly, the cocoa extract enhanced the barrier function of an oral epithelial model by increasing the transepithelial electrical resistance. We provide evidence that the beneficial properties of an epicatechin-rich cocoa extract may be useful for preventing and/or treating periodontal diseases.
Collapse
Affiliation(s)
- Amel Ben Lagha
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
| | - Patricia Maquera Huacho
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
| | - Daniel Grenier
- Oral Ecology Research Group, Faculty of Dentistry, Université Laval, Quebec City, QC, Canada
- * E-mail:
| |
Collapse
|
89
|
Afonso AC, Gomes IB, Saavedra MJ, Giaouris E, Simões LC, Simões M. Bacterial coaggregation in aquatic systems. WATER RESEARCH 2021; 196:117037. [PMID: 33751976 DOI: 10.1016/j.watres.2021.117037] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 06/12/2023]
Abstract
The establishment of a sessile community is believed to occur in a sequence of steps where genetically distinct bacteria can become attached to partner cells via specific molecules, in a process known as coaggregation. The presence of bacteria with the ability to autoaggregate and coaggregate has been described for diverse aquatic systems, particularly freshwater, drinking water, wastewater, and marine water. In these aquatic systems, coaggregation already demonstrated a role in the development of complex multispecies sessile communities, including biofilms. While specific molecular aspects on coaggregation in aquatic systems remain to be understood, clear evidence exist on the impact of this mechanism in multispecies biofilm resilience and homeostasis. The identification of bridging bacteria among coaggregating consortia has potential to improve the performance of wastewater treatment plants and/or to contribute for the development of strategies to control undesirable biofilms. This study provides a comprehensive analysis on the occurrence and role of bacterial coaggregation in diverse aquatic systems. The potential of this mechanism in water-related biotechnology is further described, with particular emphasis on the role of bridging bacteria.
Collapse
Affiliation(s)
- Ana C Afonso
- LEPABE, Faculty of Engineering, Department of Chemical Engineering, University of Porto, Porto, Portugal
| | - Inês B Gomes
- LEPABE, Faculty of Engineering, Department of Chemical Engineering, University of Porto, Porto, Portugal
| | - Maria José Saavedra
- CITAB, Centre for the Research and Technology for Agro-Environment and Biological Sciences, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Efstathios Giaouris
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, Ierou Lochou 10 & Makrygianni, Myrina 81400, Lemnos, Greece
| | - Lúcia C Simões
- CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Manuel Simões
- LEPABE, Faculty of Engineering, Department of Chemical Engineering, University of Porto, Porto, Portugal.
| |
Collapse
|
90
|
Li ZR, Sun J, Du Y, Pan A, Zeng L, Maboudian R, Burne RA, Qian PY, Zhang W. Mutanofactin promotes adhesion and biofilm formation of cariogenic Streptococcus mutans. Nat Chem Biol 2021; 17:576-584. [PMID: 33664521 DOI: 10.1038/s41589-021-00745-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
Cariogenic Streptococcus mutans is known as a predominant etiological agent of dental caries due to its exceptional capacity to form biofilms. From strains of S. mutans isolated from dental plaque, we discovered, in the present study, a polyketide/nonribosomal peptide biosynthetic gene cluster, muf, which directly correlates with a strong biofilm-forming capability. We then identified the muf-associated bioactive product, mutanofactin-697, which contains a new molecular scaffold, along with its biosynthetic logic. Further mode-of-action studies revealed that mutanofactin-697 binds to S. mutans cells and also extracellular DNA, increases bacterial hydrophobicity, and promotes bacterial adhesion and subsequent biofilm formation. Our findings provided an example of a microbial secondary metabolite promoting biofilm formation via a physicochemical approach, highlighting the importance of secondary metabolism in mediating critical processes related to the development of dental caries.
Collapse
Affiliation(s)
- Zhong-Rui Li
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Jin Sun
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yongle Du
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Aifei Pan
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Lin Zeng
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Roya Maboudian
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Robert A Burne
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Pei-Yuan Qian
- Department of Ocean Science and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
91
|
Presence of non-oral bacteria in the oral cavity. Arch Microbiol 2021; 203:2747-2760. [PMID: 33791834 PMCID: PMC8012020 DOI: 10.1007/s00203-021-02300-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/20/2020] [Accepted: 03/22/2021] [Indexed: 12/30/2022]
Abstract
A homeostatic balance exists between the resident microbiota in the oral cavity and the host. Perturbations of the oral microbiota under particular conditions can contribute to the growth of non-oral pathogens that are hard to kill because of their higher resistance to antimicrobials, raising the probability of treatment failure and reinfection. The presence of these bacteria in the oral cavity has been proven to be associated with several oral diseases such as periodontitis, caries, and gingivitis, and systemic diseases of importance in clinical medicine such as cystic fibrosis, HIV, and rheumatoid arthritis. However, it is still controversial whether these species are merely transient members or unique to the oral cavity. Mutualistic and antagonistic interactions between the oral microbiota and non-oral pathogens can also occur, though the mechanisms used by these bacteria are not clear. Therefore, this review presents an overview of the current knowledge about the presence of non-oral bacteria in the oral cavity, their relationship with systemic and oral diseases, and their interactions with oral bacteria.
Collapse
|
92
|
Cugini C, Ramasubbu N, Tsiagbe VK, Fine DH. Dysbiosis From a Microbial and Host Perspective Relative to Oral Health and Disease. Front Microbiol 2021; 12:617485. [PMID: 33763040 PMCID: PMC7982844 DOI: 10.3389/fmicb.2021.617485] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The significance of microbiology and immunology with regard to caries and periodontal disease gained substantial clinical or research consideration in the mid 1960's. This enhanced emphasis related to several simple but elegant experiments illustrating the relevance of bacteria to oral infections. Since that point, the understanding of oral diseases has become increasingly sophisticated and many of the original hypotheses related to disease causality have either been abandoned or amplified. The COVID pandemic has reminded us of the importance of history relative to infectious diseases and in the words of Churchill "those who fail to learn from history are condemned to repeat it." This review is designed to present an overview of broad general directions of research over the last 60 years in oral microbiology and immunology, reviewing significant contributions, indicating emerging foci of interest, and proposing future directions based on technical advances and new understandings. Our goal is to review this rich history (standard microbiology and immunology) and point to potential directions in the future (omics) that can lead to a better understanding of disease. Over the years, research scientists have moved from a position of downplaying the role of bacteria in oral disease to one implicating bacteria as true pathogens that cause disease. More recently it has been proposed that bacteria form the ecological first line of defense against "foreign" invaders and also serve to train the immune system as an acquired host defensive stimulus. While early immunological research was focused on immunological exposure as a modulator of disease, the "hygiene hypothesis," and now the "old friends hypothesis" suggest that the immune response could be trained by bacteria for long-term health. Advanced "omics" technologies are currently being used to address changes that occur in the host and the microbiome in oral disease. The "omics" methodologies have shaped the detection of quantifiable biomarkers to define human physiology and pathologies. In summary, this review will emphasize the role that commensals and pathobionts play in their interaction with the immune status of the host, with a prediction that current "omic" technologies will allow researchers to better understand disease in the future.
Collapse
Affiliation(s)
- Carla Cugini
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
| | | | | | | |
Collapse
|
93
|
Sadiq FA, Burmølle M, Heyndrickx M, Flint S, Lu W, Chen W, Zhao J, Zhang H. Community-wide changes reflecting bacterial interspecific interactions in multispecies biofilms. Crit Rev Microbiol 2021; 47:338-358. [PMID: 33651958 DOI: 10.1080/1040841x.2021.1887079] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Existence of most bacterial species, in natural, industrial, and clinical settings in the form of surface-adhered communities or biofilms has been well acknowledged for decades. Research predominantly focusses on single-species biofilms as these are relatively easy to study. However, microbiologists are now interested in studying multispecies biofilms and revealing interspecific interactions in these communities because of the existence of a plethora of different bacterial species together in almost all natural settings. Multispecies biofilms-led emergent properties are triggered by bacterial social interactions which have huge implication for research and practical knowledge useful for the control and manipulation of these microbial communities. Here, we discuss some important bacterial interactions that take place in multispecies biofilm communities and provide insights into community-wide changes that indicate bacterial interactions and elucidate underlying mechanisms.
Collapse
Affiliation(s)
- Faizan Ahmed Sadiq
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Mette Burmølle
- Section of Microbiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Marc Heyndrickx
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Technology & Food Sciences Unit, Melle, Belgium.,Department of Pathology, Bacteriology and Poultry Diseases, Ghent University, Merelbeke, Belgium
| | - Steve Flint
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,School of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| |
Collapse
|
94
|
Pignatelli P, Iezzi L, Pennese M, Raimondi P, Cichella A, Bondi D, Grande R, Cotellese R, Di Bartolomeo N, Innocenti P, Piattelli A, Curia MC. The Potential of Colonic Tumor Tissue Fusobacterium nucleatum to Predict Staging and Its Interplay with Oral Abundance in Colon Cancer Patients. Cancers (Basel) 2021; 13:1032. [PMID: 33804585 PMCID: PMC7957509 DOI: 10.3390/cancers13051032] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/16/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Intestinal microbiota dysbiosis may enhance the carcinogenicity of colon cancer (CC) by the proliferation and differentiation of epithelial cells. Oral Fusobacterium nucleatum (Fn) and Porphyromonas gingivalis (Pg) have the ability to invade the gut epithelium, promoting tumor progression. The aim of the study was to assess whether the abundance of these odontopathogenic bacteria was associated with colon cancer. We also investigated how lifestyle factors could influence the oral Fn and Pg abundance and CC. METHODS Thirty-six CC patients were included in the study to assess the Pg and Fn oral and colon tissue abundance by qPCR. Oral health data, food habits and lifestyles were also recorded. RESULTS Patients had a greater quantity of Fn in the oral cavity than matched CC and adjacent non-neoplastic mucosa (adj t) tissues (p = 0.004 and p < 0.001). Instead, Pg was not significantly detected in colonic tissues. There was an association between the Fn quantity in the oral and CC tissue and a statistically significant relation between the Fn abundance in adenocarcinoma (ADK) and staging (p = 0.016). The statistical analysis revealed a tendency towards a greater Fn quantity in CC (p = 0.073, η2p = 0.12) for high-meat consumers. CONCLUSION In our study, Pg was absent in colon tissues but was correlated with the oral inflammation gingival and plaque indices. For the first time, there was evidence that the Fn oral concentration can influence colon tissue concentrations and predict CC prognosis.
Collapse
Affiliation(s)
- Pamela Pignatelli
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
| | - Lorena Iezzi
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
| | - Martina Pennese
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
| | - Paolo Raimondi
- Department of General Surgery, Private Hospital “Villa Serena”, Città Sant’Angelo, 65013 Pescara, Italy; (P.R.); (A.C.); (N.D.B.); (P.I.)
| | - Anna Cichella
- Department of General Surgery, Private Hospital “Villa Serena”, Città Sant’Angelo, 65013 Pescara, Italy; (P.R.); (A.C.); (N.D.B.); (P.I.)
| | - Danilo Bondi
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
| | - Rossella Grande
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy;
| | - Roberto Cotellese
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
- Villa Serena Foundation for Research, Città Sant’Angelo, 65013 Pescara, Italy
| | - Nicola Di Bartolomeo
- Department of General Surgery, Private Hospital “Villa Serena”, Città Sant’Angelo, 65013 Pescara, Italy; (P.R.); (A.C.); (N.D.B.); (P.I.)
| | - Paolo Innocenti
- Department of General Surgery, Private Hospital “Villa Serena”, Città Sant’Angelo, 65013 Pescara, Italy; (P.R.); (A.C.); (N.D.B.); (P.I.)
- Villa Serena Foundation for Research, Città Sant’Angelo, 65013 Pescara, Italy
| | - Adriano Piattelli
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
- Villa Serena Foundation for Research, Città Sant’Angelo, 65013 Pescara, Italy
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini, 66100 Chieti, Italy; (P.P.); (L.I.); (M.P.); (R.C.); (A.P.)
| |
Collapse
|
95
|
Motey GA, Owusu-Kwarteng J, Obiri-Danso K, Ofori LA, Ellis WO, Jespersen L. In vitro properties of potential probiotic lactic acid bacteria originating from Ghanaian indigenous fermented milk products. World J Microbiol Biotechnol 2021; 37:52. [PMID: 33594545 DOI: 10.1007/s11274-021-03013-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 01/28/2021] [Indexed: 01/17/2023]
Abstract
Fermented milk products are a major source of health-promoting microorganisms known as probiotics. To characterize the probiotic properties of lactic acid bacteria isolated from Ghanaian traditionally fermented milk, thirty (30) isolates comprising Enterococcus faecium (1), Lactobacillus fermentum (14), Lb. plantarum (2) and Pediococcus acidilactici (13) identified by 16S rRNA gene sequencing, were tested for survival at low pH (2.5) and bile salts (0.3% (w/v)), hydrophobicity, co-aggregation, auto-aggregation and antimicrobial activities against selected pathogens. Safety of potential probiotic bacteria was assessed by hemolytic activity on blood agar and susceptibility to nine different antibiotics. Majority (90%) of the strains showed survival rates above 80% at pH (2.5) and in bile salts (0.3% (w/v)). Hydrophobicity ranged from 5 to 61% while cell auto-aggregation ranged from 41 to 80% after 24 h. Co-aggregation with E. coli (3.7-43.9%) and S. Typhimurium (1.3-49.5%) were similar for the LAB strains at 24 h. Cell- free supernatants of all LAB strains inhibited E. coli while S. Typhimurium was not sensitive to cell-free supernatants of five Pd. acidilactici strains: OS24h20, OS18h3, OY9h19, OS9h8 and 24NL38. None of the LAB strains showed β-hemolysis but 38% of strains showed α-hemolysis. Susceptibilities to antibiotics were strain-specific; only four strains, two Lb. fermentum and two Pd. acidilactici were susceptible to all nine antibiotics tested. Based on high survival rates in bile salts, low pH and generally good hydrophobicity, auto-aggregation, co-aggregation and inhibitory activities, 15 out of 30 strains tested were considered qualified candidates for development of probiotic cultures for fermented milk products in sub-Saharan Africa.
Collapse
Affiliation(s)
- Grace Adzo Motey
- Department of Applied Biology, C. K. Tedam University of Technology and Applied Sciences, Navrongo, Ghana.
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana.
| | - James Owusu-Kwarteng
- Department of Food Science and Technology, University of Energy and Natural Resources, Sunyani, Ghana
| | - Kwasi Obiri-Danso
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Linda Aurelia Ofori
- Department of Theoretical and Applied Biology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - William Otoo Ellis
- Department of Food Science and Technology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Lene Jespersen
- Department of Food Science, University of Copenhagen, Rolighedsvej 26, 1958, Frederiksberg C, Denmark
| |
Collapse
|
96
|
Jalali F, Ellett F, Balani P, Duncan MJ, Dewhirst FE, Borisy GG, Irimia D. No man's land: Species-specific formation of exclusion zones bordering Actinomyces graevenitzii microcolonies in nanoliter cultures. Microbiologyopen 2021; 10:e1137. [PMID: 33544453 PMCID: PMC7882712 DOI: 10.1002/mbo3.1137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/07/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022] Open
Abstract
To survive within complex environmental niches, including the human host, bacteria have evolved intricate interspecies communities driven by competition for limited nutrients, cooperation via complementary metabolic proficiencies, and establishment of homeostatic relationships with the host immune system. The study of such complex, interdependent relationships is often hampered by the challenges of culturing many bacterial strains in research settings and the limited set of tools available for studying the dynamic behavior of multiple bacterial species at the microscale. Here, we utilize a microfluidic‐based co‐culture system and time‐lapse imaging to characterize dynamic interactions between Streptococcus species, Staphylococcus aureus, and Actinomyces species. Co‐culture of Streptococcus cristatus or S. salivarius in nanoliter compartments with Actinomyces graevenitzii revealed localized exclusion of Streptococcus and Staphylococcus from media immediately surrounding A. graevenitzii microcolonies. This community structure did not occur with S. mitis or S. oralis strains or in co‐cultures containing other Actinomycetaceae species such as S. odontolyticus or A. naeslundii. Moreover, fewer neutrophils were attracted to compartments containing both A. graevenitzii and Staphylococcus aureus than to an equal number of either species alone, suggesting a possible survival benefit together during immune responses.
Collapse
Affiliation(s)
- Fatemeh Jalali
- Division of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Felix Ellett
- Division of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Pooja Balani
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Margaret J Duncan
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Floyd E Dewhirst
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA.,Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Gary G Borisy
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Daniel Irimia
- Division of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
97
|
Cheng Z, Do T, Mankia K, Meade J, Hunt L, Clerehugh V, Speirs A, Tugnait A, Emery P, Devine D. Dysbiosis in the oral microbiomes of anti-CCP positive individuals at risk of developing rheumatoid arthritis. Ann Rheum Dis 2021; 80:162-168. [PMID: 33004333 DOI: 10.1136/annrheumdis-2020-216972] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES An increased prevalence of periodontitis and perturbation of the oral microbiome has been identified in patients with rheumatoid arthritis (RA). The periodontal pathogen Porphyromonas gingivalis may cause local citrullination of proteins, potentially triggering anti-citrullinated protein antibody production. However, it is not known if oral dysbiosis precedes the onset of clinical arthritis. This study comprehensively characterised the oral microbiome in anti-cyclic citrullinated peptide (anti-CCP) positive at-risk individuals without clinical synovitis (CCP+at risk). METHODS Subgingival plaque was collected from periodontally healthy and diseased sites in 48 CCP+at risk, 26 early RA and 32 asymptomatic healthy control (HC) individuals. DNA libraries were sequenced on the Illumina HiSeq 3000 platform. Taxonomic profile and functional capability of the subgingival microbiome were compared between groups. RESULTS At periodontally healthy sites, CCP+at risk individuals had significantly lower microbial richness compared with HC and early RA groups (p=0.004 and 0.021). Microbial community alterations were found at phylum, genus and species levels. A large proportion of the community differed significantly in membership (523 species; 35.6%) and structure (575 species; 39.1%) comparing CCP+at risk and HC groups. Certain core species, including P. gingivalis, had higher relative abundance in the CCP+at risk group. Seventeen clusters of orthologous gene functional units were significantly over-represented in the CCP+at risk group compared with HC (adjusted p value <0.05). CONCLUSION Anti-CCP positive at-risk individuals have dysbiotic subgingival microbiomes and increased abundance of P. gingivalis compared with controls. This supports the hypothesis that the oral microbiome and specifically P. gingivalis are important in RA initiation.
Collapse
Affiliation(s)
- Zijian Cheng
- Division of Oral Biology, University of Leeds, School of Dentistry, Leeds, UK
- The Affiliated Stomatology Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Zhejiang University School of Stomatology, Hangzhou, China
| | - Thuy Do
- Division of Oral Biology, University of Leeds, School of Dentistry, Leeds, UK
| | - Kulveer Mankia
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, School of Medicine, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Josephine Meade
- Division of Oral Biology, University of Leeds, School of Dentistry, Leeds, UK
| | - Laura Hunt
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, School of Medicine, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Val Clerehugh
- Division of Restorative Dentistry, University of Leeds, School of Dentistry, Leeds, UK
| | - Alastair Speirs
- Leeds Dental Institute, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Aradhna Tugnait
- Division of Restorative Dentistry, University of Leeds, School of Dentistry, Leeds, UK
| | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, School of Medicine, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Deirdre Devine
- Division of Oral Biology, University of Leeds, School of Dentistry, Leeds, UK
| |
Collapse
|
98
|
Relucenti M, Familiari G, Donfrancesco O, Taurino M, Li X, Chen R, Artini M, Papa R, Selan L. Microscopy Methods for Biofilm Imaging: Focus on SEM and VP-SEM Pros and Cons. BIOLOGY 2021; 10:biology10010051. [PMID: 33445707 PMCID: PMC7828176 DOI: 10.3390/biology10010051] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/11/2022]
Abstract
Simple Summary Bacterial biofilms cause infections that are often resistant to antibiotic treatments. Research about the formation and elimination of biofilms cannot be undertaken without detailed imaging techniques. In this review, traditional and cutting-edge microscopy methods to study biofilm structure, ultrastructure, and 3-D architecture, with particular emphasis on conventional scanning electron microscopy and variable pressure scanning electron microscopy, are addressed, with the respective advantages and disadvantages. When ultrastructural characterization of biofilm matrix and its embedded bacterial cells is needed, as in studies on the effects of drug treatments on biofilm, scanning electron microscopy with customized protocols such as the osmium tetroxide (OsO4), ruthenium red (RR), tannic acid (TA), and ionic liquid (IL) must be preferred over other methods for the following: unparalleled image quality, magnification and resolution, minimal sample loss, and actual sample structure preservation. The first step to make a morphological assessment of the effect of the various pharmacological treatments on clinical biofilms is the production of images that faithfully reflect the structure of the sample. The extraction of quantitative parameters from images, possible using specific software, will allow for the scanning electron microscopy morphological evaluation to no longer be considered as an accessory technique, but a quantitative method to all effects. Abstract Several imaging methodologies have been used in biofilm studies, contributing to deepening the knowledge on their structure. This review illustrates the most widely used microscopy techniques in biofilm investigations, focusing on traditional and innovative scanning electron microscopy techniques such as scanning electron microscopy (SEM), variable pressure SEM (VP-SEM), environmental SEM (ESEM), and the more recent ambiental SEM (ASEM), ending with the cutting edge Cryo-SEM and focused ion beam SEM (FIB SEM), highlighting the pros and cons of several methods with particular emphasis on conventional SEM and VP-SEM. As each technique has its own advantages and disadvantages, the choice of the most appropriate method must be done carefully, based on the specific aim of the study. The evaluation of the drug effects on biofilm requires imaging methods that show the most detailed ultrastructural features of the biofilm. In this kind of research, the use of scanning electron microscopy with customized protocols such as osmium tetroxide (OsO4), ruthenium red (RR), tannic acid (TA) staining, and ionic liquid (IL) treatment is unrivalled for its image quality, magnification, resolution, minimal sample loss, and actual sample structure preservation. The combined use of innovative SEM protocols and 3-D image analysis software will allow for quantitative data from SEM images to be extracted; in this way, data from images of samples that have undergone different antibiofilm treatments can be compared.
Collapse
Affiliation(s)
- Michela Relucenti
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy; (G.F.); (O.D.)
- Correspondence: ; Tel.: +39-0649918061
| | - Giuseppe Familiari
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy; (G.F.); (O.D.)
| | - Orlando Donfrancesco
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via Alfonso Borelli 50, 00161 Rome, Italy; (G.F.); (O.D.)
| | - Maurizio Taurino
- Department of Clinical and Molecular Medicine, Unit of Vascular Surgery, Sant’Andrea Hospital, Sapienza University of Rome, Via di Grottarossa 1039, 00189 Rome, Italy;
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210096, China; (X.L.); (R.C.)
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210096, China; (X.L.); (R.C.)
| | - Marco Artini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (R.P.); (L.S.)
| | - Rosanna Papa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (R.P.); (L.S.)
| | - Laura Selan
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (M.A.); (R.P.); (L.S.)
| |
Collapse
|
99
|
Araujo TT, Camiloti GD, Valle AD, Silva NDG, Souza BM, Carvalho TDS, Câmara JVF, Shibao PYT, Henrique-Silva F, Cruvinel T, Magalhães AC, Buzalaf MAR. A sugarcane cystatin (CaneCPI-5) alters microcosm biofilm formation and reduces dental caries. BIOFOULING 2021; 37:109-116. [PMID: 33588651 DOI: 10.1080/08927014.2021.1881065] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 06/12/2023]
Abstract
The antimicrobial and anticaries effects of CaneCPI-5 were evaluated. Ninety bovine enamel samples were treated for 60 s with either phosphate-buffered-saline (PBS), 0.12% chlorhexidine (CHX), 0.05 mg ml-1 CaneCPI-5, 0.1 mg ml-1 CaneCPI-5 or 0.5 mg ml-1 CaneCPI-5. They were incubated with inoculum (human saliva + McBain's saliva) for the first 8 h. From then until the end of the experiment, the enamel was exposed to McBain saliva with sucrose and, once a day, for 5 days, they were treated with the solutions. At the end of the experimental period, resazurin and viable plate count assays were performed. Enamel demineralization was also measured. All concentrations of CaneCPI-5 and CHX significantly reduced the activity of biofilms compared with PBS. For viable plate counts, all treatments similarly reduced the lactobacilli and total streptococci; for the mutans streptococci, 0.05 mg ml-1 CaneCPI-5 performed better than CHX. All CaneCPI-5 concentrations significantly reduced the integrated mineral loss. This study represents the first step regarding the use of CaneCPI-5 within the concept of acquired enamel pellicle and biofilm engineering to prevent dental caries.
Collapse
Affiliation(s)
- Tamara Teodoro Araujo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Gabriel Domingues Camiloti
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Aline Dionizio Valle
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Natara Dias Gomes Silva
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Beatriz Martines Souza
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Thamyris de Souza Carvalho
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - João Victor Frazão Câmara
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Priscila Yumi Tanaka Shibao
- Laboratory of Molecular Biology, Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, Brazil
| | - Flavio Henrique-Silva
- Laboratory of Molecular Biology, Department of Genetics and Evolution, Federal University of São Carlos, São Carlos, Brazil
| | - Thiago Cruvinel
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Ana Carolina Magalhães
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | | |
Collapse
|
100
|
Jia YJ, Liao Y, He YQ, Zheng MQ, Tong XT, Xue WQ, Zhang JB, Yuan LL, Zhang WL, Jia WH. Association Between Oral Microbiota and Cigarette Smoking in the Chinese Population. Front Cell Infect Microbiol 2021; 11:658203. [PMID: 34123872 PMCID: PMC8195269 DOI: 10.3389/fcimb.2021.658203] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/19/2021] [Indexed: 01/04/2023] Open
Abstract
The oral microbiota has been observed to be influenced by cigarette smoking and linked to several human diseases. However, research on the effect of cigarette smoking on the oral microbiota has not been systematically conducted in the Chinese population. We profiled the oral microbiota of 316 healthy subjects in the Chinese population by 16S rRNA gene sequencing. The alpha diversity of oral microbiota was different between never smokers and smokers (P = 0.002). Several bacterial taxa were first reported to be associated with cigarette smoking by LEfSe analysis, including Moryella (q = 1.56E-04), Bulleidia (q = 1.65E-06), and Moraxella (q = 3.52E-02) at the genus level and Rothia dentocariosa (q = 1.55E-02), Prevotella melaninogenica (q = 8.48E-08), Prevotella pallens (q = 4.13E-03), Bulleidia moorei (q = 1.79E-06), Rothia aeria (q = 3.83E-06), Actinobacillus parahaemolyticus (q = 2.28E-04), and Haemophilus parainfluenzae (q = 4.82E-02) at the species level. Two nitrite-producing bacteria that can increase the acidity of the oral cavity, Actinomyces and Veillonella, were also enriched in smokers with FDR-adjusted q-values of 3.62E-06 and 1.10E-06, respectively. Notably, we observed that two acid production-related pathways, amino acid-related enzymes (q = 6.19E-05) and amino sugar and nucleotide sugar metabolism (q = 2.63E-06), were increased in smokers by PICRUSt analysis. Finally, the co-occurrence analysis demonstrated that smoker-enriched bacteria were significantly positively associated with each other and were negatively correlated with the bacteria decreased in smokers. Our results suggested that cigarette smoking may affect oral health by creating a different environment by altering bacterial abundance, connections among oral microbiota, and the microbiota and their metabolic function.
Collapse
Affiliation(s)
- Yi-Jing Jia
- School of Public Health, Sun Yat‐sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Ying Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Yong-Qiao He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Mei-Qi Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Xia-Ting Tong
- School of Public Health, Sun Yat‐sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Wen-Qiong Xue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Jiang-Bo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Lei-Lei Yuan
- School of Public Health, Sun Yat‐sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Wen-Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
| | - Wei-Hua Jia
- School of Public Health, Sun Yat‐sen University, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat‐sen University Cancer Center, Guangzhou, China
- *Correspondence: Wei-Hua Jia,
| |
Collapse
|