1
|
Hickman HD, Moutsopoulos NM. Viral infection and antiviral immunity in the oral cavity. Nat Rev Immunol 2024:10.1038/s41577-024-01100-x. [PMID: 39533045 DOI: 10.1038/s41577-024-01100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
Individual tissues have distinct antiviral properties garnered through various mechanisms, including physical characteristics, tissue-resident immune cells and commensal organisms. Although the oral mucosa has long been appreciated as a critical barrier tissue that is exposed to a continuous barrage of pathogens, many fundamental aspects of the antiviral immune response in this tissue remain unknown. Several viral pathogens, such as herpesviruses and human papillomaviruses, have been acknowledged both historically and at present for infections in the oral cavity that result in substantial clinical burden. However, recent viral outbreaks, including those with SARS-CoV-2 and mpox, featured oral symptoms even though these viruses are not generally considered oral pathogens. Ensuing studies have shown that the oral cavity is an important locale for viral infection and potential transmission of newly emergent or re-emergent pathogens, highlighting the need for an increased understanding of the mechanisms of antiviral immunity at this site. In this Review, we provide a broad overview of antiviral immune responses in the oral cavity and discuss common viral infections and their manifestations in the oral mucosa. In addition, we present current mouse models for the study of oral viral infections.
Collapse
Affiliation(s)
- Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Niki M Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
2
|
Gallo M, Ferrari E, Giovati L, Pertinhez TA, Artesani L, Conti S, Ciociola T. The Variability of the Salivary Antimicrobial Peptide Profile: Impact of Lifestyle. Int J Mol Sci 2024; 25:11501. [PMID: 39519054 PMCID: PMC11547034 DOI: 10.3390/ijms252111501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Saliva is crucial in maintaining oral health; its composition reflects the body's physiological and diseased state. Among salivary components, antimicrobial peptides (AMPs) stand out for their broad antimicrobial activities and role in modulating the oral microbiota and innate immune response. Local and systemic diseases can affect the levels of AMPs in saliva, making them attractive biomarkers. However, the large variability in their concentrations hampers their use in diagnostics. Knowledge of the various factors influencing the profile of salivary AMPs is essential for their use as biomarkers. Here, we examine how lifestyle factors such as physical activity, dietary supplementation, tobacco smoking, and psychological stress impact salivary AMP levels. By understanding these sources of variability, we can take a step forward in using AMPs for diagnostics and prognostics and develop new tailored and preventative approaches.
Collapse
Affiliation(s)
- Mariana Gallo
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (M.G.); (E.F.)
| | - Elena Ferrari
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (M.G.); (E.F.)
| | - Laura Giovati
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (L.G.); (L.A.); (S.C.); (T.C.)
- Microbiome Research Hub, University of Parma, 43124 Parma, Italy
| | - Thelma A. Pertinhez
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (M.G.); (E.F.)
| | - Lorenza Artesani
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (L.G.); (L.A.); (S.C.); (T.C.)
| | - Stefania Conti
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (L.G.); (L.A.); (S.C.); (T.C.)
- Microbiome Research Hub, University of Parma, 43124 Parma, Italy
| | - Tecla Ciociola
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (L.G.); (L.A.); (S.C.); (T.C.)
- Microbiome Research Hub, University of Parma, 43124 Parma, Italy
| |
Collapse
|
3
|
Winter J, Jepsen S. Role of innate host defense proteins in oral cancerogenesis. Periodontol 2000 2024; 96:203-220. [PMID: 38265172 PMCID: PMC11579821 DOI: 10.1111/prd.12552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024]
Abstract
It is nowadays well accepted that chronic inflammation plays a pivotal role in tumor initiation and progression. Under this aspect, the oral cavity is predestined to examine this connection because periodontitis is a highly prevalent chronic inflammatory disease and oral squamous cell carcinomas are the most common oral malignant lesions. In this review, we describe how particular molecules of the human innate host defense system may participate as molecular links between these two important chronic noncommunicable diseases (NCDs). Specific focus is directed toward antimicrobial polypeptides, such as the cathelicidin LL-37 and human defensins, as well as S100 proteins and alarmins. We report in which way these peptides and proteins are able to initiate and support oral tumorigenesis, showing direct mechanisms by binding to growth-stimulating cell surface receptors and/or indirect effects, for example, inducing tumor-promoting genes. Finally, bacterial challenges with impact on oral cancerogenesis are briefly addressed.
Collapse
Affiliation(s)
- Jochen Winter
- Faculty of Medicine, Department of Periodontology, Operative and Preventive Dentistry, University HospitalUniversity of BonnBonnGermany
| | - Søren Jepsen
- Faculty of Medicine, Department of Periodontology, Operative and Preventive Dentistry, University HospitalUniversity of BonnBonnGermany
| |
Collapse
|
4
|
Bosch TCG, Blaser MJ, Ruby E, McFall-Ngai M. A new lexicon in the age of microbiome research. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230060. [PMID: 38497258 PMCID: PMC10945402 DOI: 10.1098/rstb.2023.0060] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/04/2023] [Indexed: 03/19/2024] Open
Abstract
At a rapid pace, biologists are learning the many ways in which resident microbes influence, and sometimes even control, their hosts to shape both health and disease. Understanding the biochemistry behind these interactions promises to reveal completely novel and targeted ways of counteracting disease processes. However, in our protocols and publications, we continue to describe these new results using a language that originated in a completely different context. This language developed when microbial interactions with hosts were perceived to be primarily pathogenic, as threats that had to be vanquished. Biomedicine had one dominating thought: winning this war against microorganisms. Today, we know that beyond their defensive roles, host tissues, especially epithelia, are vital to ensuring association with the normal microbiota, the communities of microbes that persistently live with the host. Thus, we need to adopt a language that better encompasses the newly appreciated importance of host-microbiota associations. We also need a language that frames the onset and progression of pathogenic conditions within the context of the normal microbiota. Such a reimagined lexicon should make it clear, from the very nature of its words, that microorganisms are primarily vital to our health, and only more rarely the cause of disease. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
| | - Martin J. Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ 08854, USA
| | - Edward Ruby
- California Institute of Technology, Pasadena, CA 91125, USA
| | | |
Collapse
|
5
|
Radu CM, Radu CC, Arbănaşi EM, Hogea T, Murvai VR, Chiș IA, Zaha DC. Exploring the Efficacy of Novel Therapeutic Strategies for Periodontitis: A Literature Review. Life (Basel) 2024; 14:468. [PMID: 38672739 PMCID: PMC11050937 DOI: 10.3390/life14040468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/28/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Periodontitis, a prevalent oral condition, is facing difficulties in therapeutic approaches, sometimes leading to failure. This literature review was conducted to investigate the diversity of other therapeutic approaches and their potential contributions to the successful management of the disease. This research scrutinized the alterations in microbial diversity and imbalances in crucial microbial species, which contribute significantly to the pathogenesis of periodontitis. Within the limitations of this study, we highlight the importance of understanding the treatment plan's role in periodontitis disease, opening the way for further research and innovative treatment plans to mitigate the impact of periodontitis on oral health. This will aid both healthcare professionals and patients in preventing and effectively treating periodontitis, ultimately improving oral health outcomes and overall systemic health and well-being.
Collapse
Affiliation(s)
- Casandra-Maria Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
| | - Carmen Corina Radu
- Department of Forensic Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 38 Gheorghe Marinescu Street, 540139 Targu Mures, Romania
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Emil-Marian Arbănaşi
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
- Clinic of Vascular Surgery, Mureș County Emergency Hospital, 540136 Targu Mures, Romania
- Department of Vascular Surgery, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 38 Gheorghe Marinescu Street, 540139 Targu Mures, Romania
| | - Timur Hogea
- Department of Forensic Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 38 Gheorghe Marinescu Street, 540139 Targu Mures, Romania
- Institute of Forensic Medicine, 540141 Targu Mures, Romania
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Viorela Romina Murvai
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq, 410028 Oradea, Romania
| | - Ioana-Andreea Chiș
- Department of Oral Rehabilitation, Faculty of Dentistry, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania
| | - Dana Carmen Zaha
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 1 University Street, 410087 Oradea, Romania
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 1 December Sq, 410028 Oradea, Romania
| |
Collapse
|
6
|
R PA, Anbarasu A. Antimicrobial Peptides as Immunomodulators and Antimycobacterial Agents to Combat Mycobacterium tuberculosis: a Critical Review. Probiotics Antimicrob Proteins 2023; 15:1539-1566. [PMID: 36576687 DOI: 10.1007/s12602-022-10018-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/29/2022]
Abstract
Tuberculosis (TB) is a devastating disease foisting a significantly high morbidity, prepotent in low- and middle-income developing countries. Evolution of drug resistance among Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, has made the TB treatment more complicated. The protracted nature of present TB treatment, persistent and tolerant Mtb populations, interaction with antiretroviral therapy and existing toxicity concerned with conventional anti-TB drugs are the four major challenges inflicted with emergence of drug-resistant mycobacterial strains, and the standard medications are unable to combat these strains. These factors emphasize an exigency to develop new drugs to overcome these barriers in current TB therapy. With this regard, antimycobacterial peptides derived from various sources such as human cells, bacterial sources, mycobacteriophages, fungal, plant and animal sources could be considered as antituberculosis leads as most of these peptides are associated with dual advantages of having both bactericidal activity towards Mtb as well as immuno-regulatory property. Some of the peptides possess the additional advantage of interacting synergistically with antituberculosis medications too, thereby increasing their efficiency, underscoring the vigour of antimicrobial peptides (AMPs) as best possible alternative therapeutic candidates or adjuvants in TB treatment. Albeit the beneficiary features of these peptides, few obstacles allied with them like cytotoxicity and proteolytic degradation are matter of concerns too. In this review, we have focused on structural hallmarks, targeting mechanisms and specific structural aspects contributing to antimycobacterial activity and discovered natural and synthetic antimycobacterial peptides along with their sources, anti-TB, immuno-regulatory properties, merits and demerits and possible delivery methods of AMPs.
Collapse
Affiliation(s)
- Preethi A R
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India
| | - Anand Anbarasu
- Medical & Biological Computing Laboratory, School of Bio-Sciences & Technology, Vellore Institute of Technology, Vellore-632014, India.
- Department of Biotechnology, SBST, VIT, Vellore-632014, Tamil Nadu, India.
| |
Collapse
|
7
|
Bassani B, Cucchiara M, Butera A, Kayali O, Chiesa A, Palano MT, Olmeo F, Gallazzi M, Dellavia CPB, Mortara L, Parisi L, Bruno A. Neutrophils' Contribution to Periodontitis and Periodontitis-Associated Cardiovascular Diseases. Int J Mol Sci 2023; 24:15370. [PMID: 37895050 PMCID: PMC10607037 DOI: 10.3390/ijms242015370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Neutrophils represent the primary defense against microbial threats playing a pivotal role in maintaining tissue homeostasis. This review examines the multifaceted involvement of neutrophils in periodontitis, a chronic inflammatory condition affecting the supporting structures of teeth summarizing the contribution of neutrophil dysfunction in periodontitis and periodontal-related comorbidities. Periodontitis, a pathological condition promoted by dysbiosis of the oral microbiota, is characterized by the chronic inflammation of the gingiva and subsequent tissue destruction. Neutrophils are among the first immune cells recruited to the site of infection, releasing antimicrobial peptides, enzymes, and reactive oxygen species to eliminate pathogens. The persistent inflammatory state in periodontitis can lead to aberrant neutrophil activation and a sustained release of proinflammatory mediators, finally resulting in tissue damage, bone resorption, and disease progression. Growing evidence now points to the correlation between periodontitis and systemic comorbidities. Indeed, the release of inflammatory mediators, immune complexes, and oxidative stress by neutrophils, bridge the gap between local and systemic immunity, thus highlighting neutrophils as key players in linking periodontal inflammation to chronic conditions, including cardiovascular diseases, diabetes mellitus, and rheumatoid arthritis. This review underscores the crucial role of neutrophils in the pathogenesis of periodontitis and the complex link between neutrophil dysfunction, local inflammation, and systemic comorbidities. A comprehensive understanding of neutrophil contribution to periodontitis development and their impact on periodontal comorbidities holds significant implications for the management of oral health. Furthermore, it highlights the need for the development of novel approaches aimed at limiting the persistent recruitment and activation of neutrophils, also reducing the impact of periodontal inflammation on broader health contexts, offering promising avenues for improved disease management and patient care.
Collapse
Affiliation(s)
- Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Martina Cucchiara
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Andrea Butera
- Unit of Dental Hygiene, Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (A.B.); (A.C.)
| | - Omar Kayali
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Alessandro Chiesa
- Unit of Dental Hygiene, Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (A.B.); (A.C.)
| | - Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Francesca Olmeo
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | | | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Luca Parisi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy;
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| |
Collapse
|
8
|
Hiroshima Y, Kido JI, Kido R, Yoshida K, Bando M, Kajimoto K, Yumoto H, Shinohara Y. β-defensin 2 synthesized by a cell-free protein synthesis system and encapsulated in liposomes inhibits adhesion of Porphyromonas gingivalis to oral epithelial cells. Odontology 2023; 111:830-838. [PMID: 36745267 DOI: 10.1007/s10266-023-00789-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/29/2023] [Indexed: 02/07/2023]
Abstract
β-defensin 2 (BD-2), an antimicrobial peptide (AMP), is expressed by oral epithelial cells and plays an important role in innate immunity of the oral cavity. Cell-free protein synthesis (CFPS) systems have been studied for the synthesis of various proteins, however, the synthesis of BD-2 by a CFPS system has not been extensively explored. Liposomes have been developed as tools for drug delivery. A delivery of liposome-encapsulated AMP to oral epithelium may be useful to prevent oral infectious diseases. In the present study, we investigated the antimicrobial activity of the BD-2 protein, artificially synthesized using a CFPS system and encapsulated in liposomes. BD-2 protein was artificially synthesized using template DNA and a reconstituted CFPS system and was identified by western blotting. Bilayer liposomes were prepared using 1,2-dioleoyl-sn-glycero-3-phospho-choline and 3-sn-phosphatidylcholine from egg yolk. The artificially synthesized BD-2 was encapsulated in liposomes, collected by ultrafiltration, and detected by western blotting. Human oral epithelial cells were cultured with the liposome-encapsulated BD-2 and the concentration of BD-2 in the cell lysate of the culture with the synthesized BD-2 was higher than that of the control cultures. The antimicrobial activity of the synthesized BD-2 was investigated by an adhesion assay of Porphyromonas gingivalis to oral epithelial cells. The artificially synthesized BD-2 and its liposome significantly inhibited adhesion of P. gingivalis to oral epithelial cells. These results suggest that artificially synthesized BD-2 and liposome-encapsulated BD-2 show antimicrobial activity and can potentially play a role in oral healthcare for periodontal diseases.
Collapse
Affiliation(s)
- Yuka Hiroshima
- Department of Oral Microbiology, Tokushima University, 3-18-15, Kuramoto, Tokushima, 770-8504, Japan.
| | - Jun-Ichi Kido
- Department of Periodontology and Endodontology, Tokushima University, Tokushima, Japan
| | - Rie Kido
- Department of Periodontology and Endodontology, Tokushima University, Tokushima, Japan
| | - Kaya Yoshida
- Department of Oral Healthcare Education, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Mika Bando
- Department of Periodontology and Endodontology, Tokushima University, Tokushima, Japan
| | - Kazuaki Kajimoto
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Hiromichi Yumoto
- Department of Periodontology and Endodontology, Tokushima University, Tokushima, Japan
| | - Yasuo Shinohara
- Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
9
|
Kzar WA, Abbas RF, Hussein HM. The Antimicrobial Peptide LL-37 as a Predictor Biomarker for Periodontitis with the Presence and Absence of Smoking: A Case-Control Study. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5581267. [PMID: 37711877 PMCID: PMC10499532 DOI: 10.1155/2023/5581267] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/16/2023]
Abstract
Background A major issue is finding a valid biomarker able to diagnose periodontal disease with the presence and absence of risk factors. Indeed, the association between smoking and periodontal diseases and its impact on the manifestation of antimicrobial peptides has been delineated in clinical and epidemiological investigations. The antimicrobial peptide cathelicidin (LL-37) is pivotal in preserving periodontal health. Objectives This investigation examines and contrasts the levels of cathelicidin in the saliva of smokers and nonsmokers of periodontitis. The study also seeks to establish this biomarker's diagnostic ability to differentiate between periodontal health and disease. Materials and Methods The study involved the collection of unstimulated saliva samples from 160 participants, comprising 80 patients diagnosed with periodontitis (40 of whom were smokers and 40 were nonsmokers) and 80 periodontitis-free individuals (40 smokers and 40 nonsmokers). The clinical periodontal parameters were assessed, including recording the probing pocket depth, the level of clinical attachment, and the percentage of bleeding on probing. Subsequently, enzyme-linked immunosorbent assays were conducted to quantify the protein levels of LL-37 in the saliva samples obtained from the subjects mentioned above. Results The highest level of salivary LL-37 was found in the nonsmoker periodontitis (NSP) patients, followed by the group of smoker periodontitis (SP) and then nonsmoker healthy (NSH) group, while the lowest level was found in the healthy smoker (HS) group. At the same time, the LL-37 seems to be a very good biomarker in differentiating periodontal health from disease with the presence and absence of smoking. Conclusion Periodontitis results in a significant elevation of salivary LL-37 levels in smoker and nonsmoker patients compared to healthy individuals. These levels are positively correlated with the periodontal parameter and can serve as a valuable diagnostic tool to predict periodontitis, whereas smoking significantly reduces these levels.
Collapse
Affiliation(s)
- Wael Abdulazeez Kzar
- Department of Periodontology, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Raghad Fadhil Abbas
- Department of Periodontology, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Hashim Mueen Hussein
- Department of Conservative Dentistry, College of Dentistry, Mustansiriyah University, Baghdad, Iraq
| |
Collapse
|
10
|
Ren J, Guo X. The germicidal effect, biosafety and mechanical properties of antibacterial resin composite in cavity filling. Heliyon 2023; 9:e19078. [PMID: 37662807 PMCID: PMC10474440 DOI: 10.1016/j.heliyon.2023.e19078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/22/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
In recent years, dental resin materials have become increasingly popular for cavity filling. However, these materials can shrink during polymerization, leading to microleakages that enable bacteria to erode tooth tissue and cause secondary caries. As a result, there is great clinical demand for the development of antibacterial resins. The principle of antibacterial resin includes contact killing and filler-release killing of bacteria. For contact killing, quaternary ammonium salts (QACs) and antibacterial peptides (AMPs) can be added. For filler-release killing, chlorhexidine (CHX) and nanoparticles are used. These antibacterial agents are effective against gram-positive bacteria, gram-negative bacteria, fungi, and more. Among them, QACs has a lasting antibacterial effect, and silver nanoparticles even have a certain ability to kill viruses. Biocompatibility-wise, QACs, AMPs, and CHX have low cytotoxicity to cells when added into the resin. However, nanoparticles with smaller particle sizes have higher cytotoxicity. In terms of mechanical properties, QACs, AMPs, and CHX do not negatively affect the resin. However, the addition of magnesium oxide can have a negative impact. This paper reviews the types and antibacterial principles of commonly used antibacterial resins in recent years, evaluates their antibacterial effect, biological safety, and mechanical properties, and provides references for selecting clinical filling materials.
Collapse
Affiliation(s)
- Jiamu Ren
- Yanbian University, Jilin, 133002, China
| | - Xinwei Guo
- Peking University, Haidian District, Beijing, 100871, China
| |
Collapse
|
11
|
Vasconcelos MA, da Silva BR, Andrade AL, de Azevedo Pinheiro A, Evaristo FFV, Arruda FVS, Lorenzón EN, Cilli EM, Teixeira EH. Antimicrobial and Antibiofilm Activity of Synthetic Peptide [W7]KR12-KAEK Against Enterococcus faecalis Strains. Curr Microbiol 2023; 80:325. [PMID: 37606794 DOI: 10.1007/s00284-023-03445-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
The emergence of infections caused by microorganisms in the oral cavity and increasing concerns regarding the use of antibiotics have resulted in the development of novel antimicrobial molecules, such as antimicrobial synthetic peptides. The purpose of this study was to evaluate the antimicrobial and antibiofilm activities of the native peptide KR-12 and its derivative, the synthetic peptide [W7]KR12-KAEK, against planktonic and biofilms Enterococcus faecalis strains. The methods used to evaluate the antimicrobial activity in planktonic cultures include minimum inhibitory concentration and minimum bactericidal concentration assays. The effects of [W7]KR12-KAEK on biofilm formation and mature biofilms were evaluated by quantifying biomass (crystal violet staining) and counting colony-forming units. Structural assessments of the biofilms and cellular morphological changes were performed using scanning electron microscopy. Peptide [W7]KR12-KAEK showed potential antimicrobial activity against planktonic cells. Interestingly, the native peptide KR-12 showed no antimicrobial activity. Moreover, it inhibited biofilm formation and disrupted the mature biofilms of E. faecalis strains. These results suggest that [W7]KR12-KAEK may be a potential molecule for the development of auxiliary antimicrobial therapies against oral infections.
Collapse
Affiliation(s)
- Mayron Alves Vasconcelos
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil
- Universidade do Estado de Minas Gerais, Unidade de Divinópolis, Divinópolis, MG, Brazil
- Faculdade de Educação de Itapipoca (FACEDI), Universidade Estadual do Ceará, Itapipoca, CE, Brazil
| | - Bruno Rocha da Silva
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil
- Universidade de Fortaleza (UNIFOR), Fortaleza, CE, Brazil
| | - Alexandre Lopes Andrade
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - Aryane de Azevedo Pinheiro
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | | | - Francisco Vassiliepe Sousa Arruda
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil
- UNINTA, Sobral, CE, Brazil
| | | | - Eduardo Maffud Cilli
- Instituto de Química, Universidade Estadual Paulista (UNESP), Araraquara, SP, Brazil
| | - Edson Holanda Teixeira
- Laboratório Integrado de Biomoléculas, Departamento de Patologia e Medicina Legal, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| |
Collapse
|
12
|
Irie K, Azuma T, Tomofuji T, Yamamoto T. Exploring the Role of IL-17A in Oral Dysbiosis-Associated Periodontitis and Its Correlation with Systemic Inflammatory Disease. Dent J (Basel) 2023; 11:194. [PMID: 37623290 PMCID: PMC10453731 DOI: 10.3390/dj11080194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Oral microbiota play a pivotal role in maintaining homeostasis, safeguarding the oral cavity, and preventing the onset of disease. Oral dysbiosis has the potential to trigger pro-inflammatory effects and immune dysregulation, which can have a negative impact on systemic health. It is regarded as a key etiological factor for periodontitis. The emergence and persistence of oral dysbiosis have been demonstrated to mediate inflammatory pathology locally and at distant sites. The heightened inflammation observed in oral dysbiosis is dependent upon the secretion of interleukin-17A (IL-17A) by various innate and adaptive immune cells. IL-17A has been found to play a significant role in host defense mechanisms by inducing antibacterial peptides, recruiting neutrophils, and promoting local inflammation via cytokines and chemokines. This review seeks to present the current knowledge on oral dysbiosis and its prevention, as well as the underlying role of IL-17A in periodontitis induced by oral dysbiosis and its impact on systemic inflammatory disease.
Collapse
Affiliation(s)
- Koichiro Irie
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| | - Tetsuji Azuma
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Takaaki Tomofuji
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Tatsuo Yamamoto
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| |
Collapse
|
13
|
Laberge S, Akoum D, Wlodarczyk P, Massé JD, Fournier D, Semlali A. The Potential Role of Epigenetic Modifications on Different Facets in the Periodontal Pathogenesis. Genes (Basel) 2023; 14:1202. [PMID: 37372382 DOI: 10.3390/genes14061202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Periodontitis is a chronic inflammatory disease that affects the supporting structures of teeth. In the literature, the association between the pathogenicity of bacteria and environmental factors in this regard have been extensively examined. In the present study, we will shed light on the potential role that epigenetic change can play on different facets of its process, more particularly the modifications concerning the genes involved in inflammation, defense, and immune systems. Since the 1960s, the role of genetic variants in the onset and severity of periodontal disease has been widely demonstrated. These make some people more susceptible to developing it than others. It has been documented that the wide variation in its frequency for various racial and ethnic populations is due primarily to the complex interplay among genetic factors with those affecting the environment and the demography. In molecular biology, epigenetic modifications are defined as any change in the promoter for the CpG islands, in the structure of the histone protein, as well as post-translational regulation by microRNAs (miRNAs), being known to contribute to the alteration in gene expression for complex multifactorial diseases such as periodontitis. The key role of epigenetic modification is to understand the mechanism involved in the gene-environment interaction, and the development of periodontitis is now the subject of more and more studies that attempt to identify which factors are stimulating it, but also affect the reduced response to therapy.
Collapse
Affiliation(s)
- Samuel Laberge
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Daniel Akoum
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Piotr Wlodarczyk
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | - Jean-Daniel Massé
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| | | | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
14
|
Qi S, Zhao S, Zhang H, Liu S, Liu J, Yang J, Qi Y, Zhao Q, Jin Y, Wang F. Novel casein antimicrobial peptides for the inhibition of oral pathogenic bacteria. Food Chem 2023; 425:136454. [PMID: 37276666 DOI: 10.1016/j.foodchem.2023.136454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 04/22/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023]
Abstract
Milk casein is a rich source of antimicrobial peptides (AMPs) and the most common way to produce AMPs is enzymatic hydrolysis in vitro. In this study, active casein antimicrobial peptide (CAMPs) mixtures were generated by optimized proteolytic cleavage of milk casein. These natural-safe CAMPs mixtures exhibited high activity in the inhibition of Streptococcus mutans and Porphyromonas gingivalis. Morphological characterization suggested the pathogenic bacteria presented incomplete or irregular collapsed membrane surface after the treatment with active CAMPs mixtures. The CAMPs inhibition activity was also effective in the attachment and development of microbial biofilm. Potential CAMPs sequences were unambiguously determined by unbiased proteomic analysis and 301 potential CAMPs were obtained. The activity of 4 novel CAMPs was successfully confirmed by using synthetic standards. This study provides a promising milk CAMPs resource for the development of safe agents in oral bacteria inhibition and functional foods.
Collapse
Affiliation(s)
- Shizhe Qi
- College of Food Science and Engineering, Ocean University of Dalian, No. 52 Heishijiao Street, Dalian, Liaoning Province 116023, China; CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Shan Zhao
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Huiyan Zhang
- College of Food Science and Engineering, Ocean University of Dalian, No. 52 Heishijiao Street, Dalian, Liaoning Province 116023, China; CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Shiwen Liu
- College of Food Science and Engineering, Ocean University of Dalian, No. 52 Heishijiao Street, Dalian, Liaoning Province 116023, China; CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Jiaxin Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Jian Yang
- Shubeideng (Hangzhou) Technology Co. Ltd, No.1818-2 Wenyi Road, Hangzhou, Zhejiang Province 311121, China
| | - Yanxia Qi
- College of Food Science and Engineering, Ocean University of Dalian, No. 52 Heishijiao Street, Dalian, Liaoning Province 116023, China.
| | - Qiancheng Zhao
- College of Food Science and Engineering, Ocean University of Dalian, No. 52 Heishijiao Street, Dalian, Liaoning Province 116023, China.
| | - Yan Jin
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China
| | - Fangjun Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, No. 457 Zhongshan Road, Dalian, Liaoning Province 116023, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
15
|
Li Y, Ma Y, Yu J, Li C, Yu D, Dai R, Li Q, Cao CY. A dual functional polypeptide with antibacterial and anti-inflammatory properties for the treatment of periodontitis. Int J Biol Macromol 2023; 242:124920. [PMID: 37196724 DOI: 10.1016/j.ijbiomac.2023.124920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/29/2023] [Accepted: 05/14/2023] [Indexed: 05/19/2023]
Abstract
Periodontitis has been reported as the sixth most prevalent disease in human beings. This destructive disease is closely related to systemic diseases. Existing local drug delivery systems for periodontitis suffer from poor antibacterial effect and drug resistance. Inspired by the pathogenesis of periodontitis, we implemented a strategy to construct a dual functional polypeptide LL37-C15, which exhibited remarkable antibacterial effect against P. gingivalis and A. actinomycetemcomitans. In addition, LL37-C15 inhibits the release of pro-inflammatory cytokines by controlling the inflammatory pathway and reversing macrophage M1. Furthermore, the anti-inflammatory effect of LL37-C15 was also verified in vivo in a periodontitis rat model through the morphometry and histological observations of alveolar bone, hematoxylin-eosin, and Trap staining in gingival tissue. The results of molecular dynamics simulations showed that LL37-C15 could selectively destroy the bacterial cell membrane and protect the animal cell membrane in a self-destructive manner. The results showed that the polypeptide LL37-C15, as a novel promising therapeutic agent, exhibited a great potential for the periodontitis management. What's more, this dual functional polypeptide provides a promising strategy for building a multifunctional therapeutic platform against the inflammation and other diseases.
Collapse
Affiliation(s)
- Yuexiang Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Yunfeng Ma
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Jianan Yu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Cancan Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Da Yu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Ruoxi Dai
- Tufts University School of Dental Medicine, Department of Comprehensive Care, Boston, MA 02111, USA
| | - Quanli Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Chris Ying Cao
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| |
Collapse
|
16
|
Abstract
Mucosal tissues are constantly exposed to the outside environment. They receive signals from the commensal microbiome and tissue-specific triggers including alimentary and airborne elements and are tasked to maintain balance in the absence of inflammation and infection. Here, we present neutrophils as sentinel cells in mucosal immunity. We discuss the roles of neutrophils in mucosal homeostasis and overview clinical susceptibilities in patients with neutrophil defects. Finally, we present concepts related to specification of neutrophil responses within specific mucosal tissue microenvironments.
Collapse
Affiliation(s)
- Lakmali M. Silva
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Tae Sung Kim
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| | - Niki M. Moutsopoulos
- Oral Immunity and Infection Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
17
|
Salivary bactericidal/permeability-increasing protein: A novel inflammatory marker associated with periodontitis. Clin Oral Investig 2023:10.1007/s00784-023-04922-6. [PMID: 36805805 DOI: 10.1007/s00784-023-04922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/13/2023] [Indexed: 02/23/2023]
Abstract
OBJECTIVES The present case-control study aims to investigate the salivary levels of bactericidal/permeability-increasing protein (BPI) and interleukin-1beta (IL-1ß) in systemically healthy individuals with periodontitis and periodontally healthy for the evaluation of BPI's relation with periodontal inflammation and clinical diagnosis of periodontitis. MATERIALS AND METHODS A total of 100 participants were enrolled in this study and divided into periodontitis (P group) (n = 50) and periodontally healthy (H group) (n = 50) groups based on their full-mouth periodontal examination results including plaque index, probing pocket depth, gingival index, bleeding on probing, and clinical attachment level. Unstimulated whole saliva was collected. Salivary BPI and IL-1β levels were determined using an enzyme-linked immunosorbent assay. Receiver operating characteristic (ROC) curves were created to determine the diagnostic value of BPI. RESULTS The levels of BPI and IL-1ß in saliva were significantly higher in the P group than in the H group (p<0.001). Moreover, salivary BPI and IL-1ß levels correlated significantly with all clinical periodontal parameters (all p<0.001). Interestingly, there was a strong positive correlation between salivary levels of BPI and IL-1ß (r=0.544, p<0.001). In addition, the results of the ROC curve analysis showed that BPI had a high diagnostic potential to distinguish periodontitis from healthy controls with an area under the curve value of 0.94% (p<0.000). CONCLUSION The significantly higher salivary levels of BPI in periodontitis patients together with strong positive correlations between all periodontal parameters and salivary IL-1ß levels suggest that BPI may be involved in the inflammatory process of periodontal disease. CLINICAL RELEVANCE The present study for the first time report that salivary BPI levels may serve as a potential biomarker of inflammation in periodontal disease. TRIAL REGISTRATION NUMBER Thai Clinical Trials.gov (TCTR20211222008) (22 December 2021).
Collapse
|
18
|
Neculae E, Gosav EM, Valasciuc E, Dima N, Floria M, Tanase DM. The Oral Microbiota in Valvular Heart Disease: Current Knowledge and Future Directions. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010182. [PMID: 36676130 PMCID: PMC9862471 DOI: 10.3390/life13010182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Oral microbiota formation begins from birth, and everything from genetic components to the environment, alongside the host's behavior (such as diet, smoking, oral hygiene, and even physical activity), contributes to oral microbiota structure. Even though recent studies have focused on the gut microbiota's role in systemic diseases, the oral microbiome represents the second largest community of microorganisms, making it a new promising therapeutic target. Periodontitis and dental caries are considered the two main consequences of oral bacterial imbalance. Studies have shown that oral dysbiosis effects are not limited locally. Due to technological advancement, research identified oral bacterial species in heart valves. This evidence links oral dysbiosis with the development of valvular heart disease (VHD). This review focuses on describing the mechanism behind prolonged local inflammation and dysbiosis, that can induce bacteriemia by direct or immune-mediated mechanisms and finally VHD. Additionally, we highlight emerging therapies based on controlling oral dysbiosis, periodontal disease, and inflammation with immunological and systemic effects, that exert beneficial effects in VHD management.
Collapse
Affiliation(s)
- Ecaterina Neculae
- Department of Gastroenterology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Institute of Gastroenterology and Hepatology, “Sf. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Nicoleta Dima
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Correspondence:
| | - Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| |
Collapse
|
19
|
Li X, Chaouhan HS, Wang YM, Wang IK, Lin CL, Shen TC, Li CY, Sun KT. Risk of Periodontitis in Patients with Gastroesophageal Reflux Disease: A Nationwide Retrospective Cohort Study. Biomedicines 2022; 10:biomedicines10112980. [PMID: 36428548 PMCID: PMC9687120 DOI: 10.3390/biomedicines10112980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Background: Gastroesophageal reflux disease (GERD) is the most common digestive clinical problem worldwide that affects approximately 20% of the adult populations in Western countries. Poor oral hygiene has been reported to be associated with GERD as an atypical clinical complication. However, evidence showing the relationship between GERD and the risk of periodontitis is less clear. The present study aimed to use a retrospective cohort study design to further clarify the association between GERD and the subsequent risk of periodontitis. Methods: The risk of periodontitis in patients with GERD was investigated by analyzing epidemiological data from the Taiwan National Health Insurance Research Database from 2008 to 2018. We selected 20,125 participants with a minimum age of 40 years in the GERD group and 1:1 propensity-matched these with non-GERD individuals by sex, age, and comorbidities. The incidence of periodontitis was determined at the end of 2018. A Cox proportional hazards regression model was used to evaluate the risk of periodontitis in patients with GERD. Results: The overall incidence rate of the periodontitis risk was 1.38-fold higher (30.0 vs. 21.7/1000 person years, adjusted hazard ratio (aHR) = 1.36, 95% confidence interval (CI) = 1.28−1.45) in patients with GERD than in those without GERD. After stratified analyses for sex, age, and comorbidity, patients with GERD had a higher risk of periodontitis for age (aHR = 1.31, 95% CI = 1.20−1.42 for 40−54 years and aHR = 1.42, 95% CI =1.28−1.57 for 55−69 years), sex (aHR = 1.40, 95% CI = 1.28−1.54 for men and aHR = 1.33, 95% CI = 1.23−1.45 for women), and presence (aHR = 1.36, 95% CI = 1.27−1.45) and absence (aHR = 1.40, 95% CI = 1.21−1.62) of comorbidity than those without GERD. Among the GERD cohort, the risk for periodontitis was increased with an increasing number of emergency room visits (≥ 1 vs. <1, aHR = 5.19, 95% CI = 2.16−12.5). Conclusions: Our results revealed that patients with GERD have a higher risk of periodontitis development than those without GERD. Clinicians should pay more attention to identifying and managing periodontitis in patients with GERD.
Collapse
Affiliation(s)
- Xin Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Hitesh Singh Chaouhan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Yao-Ming Wang
- Department of Radiology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 40402, Taiwan
| | - I-Kuan Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Cheng-Li Lin
- Management Office for Health Data, China Medical University Hospital, Taichung 40402, Taiwan
| | - Te-Chun Shen
- Department of Internal Medicine, China Medical University Hospital, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Correspondence: (T.-C.S.); (C.-Y.L.); (K.-T.S.)
| | - Chi-Yuan Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- School of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Anesthesiology, China Medical University Hospital, Taichung 40402, Taiwan
- Correspondence: (T.-C.S.); (C.-Y.L.); (K.-T.S.)
| | - Kuo-Ting Sun
- Department of Pediatric Dentistry, China Medical University Hospital, Taichung 40402, Taiwan
- School of Dentistry, China Medical University, Taichung 40402, Taiwan
- Correspondence: (T.-C.S.); (C.-Y.L.); (K.-T.S.)
| |
Collapse
|
20
|
Ramírez Thomé S, Ávila Curiel B, Hernández Huerta MT, Solórzano Mata C. β-defensinas como posibles indicadores de la actividad inflamatoria en la enfermedad periodontal. INVESTIGACIÓN CLÍNICA 2022. [DOI: 10.54817/ic.v63n4a08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Periodontal disease (gingivitis and periodontitis) is an inflam-matory process caused by the activity of pathogenic bacteria and their products on the gingival sulcus, with the consequent activation of the immune response. Saliva and crevicular fluid contain a wide variety of enzymes and antimicrobial factors that are in contact with the supragingival and subgingival region, in-cluding β-defensins (hBDs). hHBDs are non-glycosylated, cysteine-rich cationic peptides produced by epithelial cells with antimicrobial and immunoregulatory effects, thus contributing to maintaining homeostasis in periodontal tissues. The changes in the microbiota and the immune response from a healthy peri-odontium to gingivitis and, finally, to periodontitis are complex. Their sever-ity depends on a dynamic balance between bacteria associated with plaque, genetic and environmental factors. Recent advances have made it possible to understand the implication of hBDs in the detection, diagnosis, and therapy of periodontal disease and the relationship between periodontitis and other inflammatory conditions. This review aims to describe the effect of hBDs on the immune response and its use as a possible marker of the inflammatory activity of the periodontal disease.
Collapse
Affiliation(s)
- Saira Ramírez Thomé
- Facultad de Odontología. Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| | | | | | - Carlos Solórzano Mata
- Facultad de Odontología. Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca, México
| |
Collapse
|
21
|
El Mobadder M, Nammour S, Grzech-Leśniak Z, Grzech-Leśniak K. Efficacy of the Adjunct Use of Povidone-Iodine or Sodium Hypochlorite with Non-Surgical Management of Periodontitis: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:jcm11216593. [PMID: 36362818 PMCID: PMC9658783 DOI: 10.3390/jcm11216593] [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: 10/23/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
This systematic review sought to assess the efficacy of combining either sodium hypochlorite or povidone-iodine as disinfection solutions with non-surgical treatment of periodontitis. An electronic search was conducted through PubMed, Scopus, Web of Science, CENTRAL, and Google Scholar from inception until 10 September 2022. Outcomes included clinical outcomes (probing pocket depth, plaque index, clinical attachment level, relative-horizontal attachment level, bleeding on probing, gingival recession, the position of gingival margin) and biochemical (BAPNA level) properties. A subgroup analysis was conducted according to the assessment timepoint. Ten studies reporting the use of povidone-iodine and five studies reporting the use of sodium hypochlorite were included in this review. Overall, in the meta-analysis of povidone-iodine, no significant changes were noted in any of the assessed outcomes; however, minor changes were noted in probing pocket depth and clinical attachment level at a specific timepoint. Regarding sodium hypochlorite, a significant reduction in all clinical outcomes, except for bleeding on probing, was noted. In conclusion, the use of povidone-iodine does not result in an improvement in clinical outcomes, whereas sodium hypochlorite has promising properties that result in significant improvement in probing pocket depth and clinical attachment level. However, more studies are needed to confirm these observations.
Collapse
Affiliation(s)
- Marwan El Mobadder
- Dental Surgery Department, Wroclaw Medical University, 50-425 Wroclaw, Poland
- Correspondence: or ; Tel.: +961-7134-3767
| | - Samir Nammour
- Department of Dental Sciences, Faculty of Medicine, University of Liege, 4000 Liege, Belgium
| | | | | |
Collapse
|
22
|
Functional biomaterials for comprehensive periodontitis therapy. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
23
|
Acyldepsipeptide Analogues: A Future Generation Antibiotics for Tuberculosis Treatment. Pharmaceutics 2022; 14:pharmaceutics14091956. [PMID: 36145704 PMCID: PMC9502522 DOI: 10.3390/pharmaceutics14091956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Acyldepsipeptides (ADEPs) are a new class of emerging antimicrobial peptides (AMPs), which are currently explored for treatment of pathogenic infections, including tuberculosis (TB). These cyclic hydrophobic peptides have a unique bacterial target to the conventional anti-TB drugs, and present a therapeutic window to overcome Mycobacterium Tuberculosis (M. tb) drug resistance. ADEPs exerts their antibacterial activity on M. tb strains through activation of the protein homeostatic regulatory protease, the caseinolytic protease (ClpP1P2). ClpP1P2 is normally regulated and activated by the ClpP-ATPases to degrade misfolded and toxic peptides and/or short proteins. ADEPs bind and dysregulate all the homeostatic capabilities of ClpP1P2 while inducing non-selective proteolysis. The uncontrolled proteolysis leads to M. tb cell death within the host. ADEPs analogues that have been tested possess cytotoxicity and poor pharmacokinetic and pharmacodynamic properties. However, these can be improved by drug design techniques. Moreover, the use of nanomaterial in conjunction with ADEPs would yield effective synergistic effect. This new mode of action has potential to combat and eradicate the extensive multi-drug resistance (MDR) problem that is currently faced by the public health pertaining bacterial infections, especially TB.
Collapse
|
24
|
Safe-by-Design Antibacterial Peroxide-Substituted Biomimetic Apatites: Proof of Concept in Tropical Dentistry. J Funct Biomater 2022; 13:jfb13030144. [PMID: 36135579 PMCID: PMC9503752 DOI: 10.3390/jfb13030144] [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: 07/21/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Bone infections are a key health challenge with dramatic consequences for affected patients. In dentistry, periodontitis is a medically compromised condition for efficient dental care and bone grafting, the success of which depends on whether the surgical site is infected or not. Present treatments involve antibiotics associated with massive bacterial resistance effects, urging for the development of alternative antibacterial strategies. In this work, we established a safe-by-design bone substitute approach by combining bone-like apatite to peroxide ions close to natural in vivo oxygenated species aimed at fighting pathogens. In parallel, bone-like apatites doped with Ag+ or co-doped Ag+/peroxide were also prepared for comparative purposes. The compounds were thoroughly characterized by chemical titrations, FTIR, XRD, SEM, and EDX analyses. All doped apatites demonstrated significant antibacterial properties toward four major pathogenic bacteria involved in periodontitis and bone infection, namely Porphyromonas gingivalis (P. gingivalis), Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans), Fusobacterium nucleatum (F. nucleatum), and S. aureus. By way of complementary tests to assess protein adsorption, osteoblast cell adhesion, viability and IC50 values, the samples were also shown to be highly biocompatible. In particular, peroxidated apatite was the safest material tested, with the lowest IC50 value toward osteoblast cells. We then demonstrated the possibility to associate such doped apatites with two biocompatible polymers, namely gelatin and poly(lactic-co-glycolic) acid PLGA, to prepare, respectively, composite 2D membranes and 3D scaffolds. The spatial distribution of the apatite particles and polymers was scrutinized by SEM and µCT analyses, and their relevance to the field of bone regeneration was underlined. Such bio-inspired antibacterial apatite compounds, whether pure or associated with (bio)polymers are thus promising candidates in dentistry and orthopedics while providing an alternative to antibiotherapy.
Collapse
|
25
|
Trivedi R, Upadhyay TK, Kausar MA, Saeed A, Sharangi AB, Almatroudi A, Alabdallah NM, Saeed M, Aqil F. Nanotechnological interventions of the microbiome as a next-generation antimicrobial therapy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 833:155085. [PMID: 35398124 DOI: 10.1016/j.scitotenv.2022.155085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 03/22/2022] [Accepted: 04/03/2022] [Indexed: 06/14/2023]
Abstract
The rise of antimicrobial resistance (AMR) impacts public health due to the diminished potency of existing antibiotics. The microbiome plays an important role in the host's immune system activity and shows the history of exposure to antimicrobials and its manipulation in combating antimicrobial resistance. Advancements in gene technologies, DNA sequencing, and computational biology have emerged as powerful platforms to better understand the relationship between animals and microorganisms (MOs). The past few years have witnessed an increase in the use of nanotechnology, both in industry and in academia, as tools to tackle antimicrobial resistance. New strategies of microbiome manipulation have been developed, such as the use of prebiotics, probiotics, peptides, antibodies, an appropriate diet, phage therapy, and the use of various nanotechnological techniques. Owing to the research outcomes, targeted delivery of antimicrobials with some modifications with nanoparticles can lead to the destruction of resistant microbial cells. In addition, nanoparticles have been studied for their potential antimicrobial effects both in vitro and in vivo. In this review, we highlight key opportunistic areas for applying nanotechnologies with the aim of manipulating the microbiome for the treatment of antimicrobial resistance. Besides providing a detailed review on various nanomaterials, technologies, opportunities, technical needs, and potential approaches for the manipulation of the microbiome to address these challenges, we discuss future challenges and our perspective.
Collapse
Affiliation(s)
- Rashmi Trivedi
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India
| | - Tarun Kumar Upadhyay
- Department of Biotechnology, Parul Institute of Applied Sciences and Animal Cell Culture and Immunobiochemistry Lab, Centre of Research for Development, Parul University, Vadodara 391760, India.
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, PO Box 2240, Hail, Saudi Arabia
| | - Amir Saeed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, PO Box 2240, Hail, Saudi Arabia
| | - Amit Baran Sharangi
- Department of Plantation Spices Medicinal and Aromatic Crops, Bidhan Chandra Krishi Viswavidyalaya, Mohanpur 741252, India
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Qassim 51431, Saudi Arabia
| | - Nadiyah M Alabdallah
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Hail, PO Box 2240, Hail, Saudi Arabia.
| | - Farrukh Aqil
- UofL Health - Brown Cancer Center and Department of Medicine, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
26
|
Johnstone KF, Herzberg MC. Antimicrobial peptides: Defending the mucosal epithelial barrier. FRONTIERS IN ORAL HEALTH 2022; 3:958480. [PMID: 35979535 PMCID: PMC9376388 DOI: 10.3389/froh.2022.958480] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The recent epidemic caused by aerosolized SARS-CoV-2 virus illustrates the importance and vulnerability of the mucosal epithelial barrier against infection. Antimicrobial proteins and peptides (AMPs) are key to the epithelial barrier, providing immunity against microbes. In primitive life forms, AMPs protect the integument and the gut against pathogenic microbes. AMPs have also evolved in humans and other mammals to enhance newer, complex innate and adaptive immunity to favor the persistence of commensals over pathogenic microbes. The canonical AMPs are helictical peptides that form lethal pores in microbial membranes. In higher life forms, this type of AMP is exemplified by the defensin family of AMPs. In epithelial tissues, defensins, and calprotectin (complex of S100A8 and S100A9) have evolved to work cooperatively. The mechanisms of action differ. Unlike defensins, calprotectin sequesters essential trace metals from microbes, which inhibits growth. This review focuses on defensins and calprotectin as AMPs that appear to work cooperatively to fortify the epithelial barrier against infection. The antimicrobial spectrum is broad with overlap between the two AMPs. In mice, experimental models highlight the contribution of both AMPs to candidiasis as a fungal infection and periodontitis resulting from bacterial dysbiosis. These AMPs appear to contribute to innate immunity in humans, protecting the commensal microflora and restricting the emergence of pathobionts and pathogens. A striking example in human innate immunity is that elevated serum calprotectin protects against neonatal sepsis. Calprotectin is also remarkable because of functional differences when localized in epithelial and neutrophil cytoplasm or released into the extracellular environment. In the cytoplasm, calprotectin appears to protect against invasive pathogens. Extracellularly, calprotectin can engage pathogen-recognition receptors to activate innate immune and proinflammatory mechanisms. In inflamed epithelial and other tissue spaces, calprotectin, DNA, and histones are released from degranulated neutrophils to form insoluble antimicrobial barriers termed neutrophil extracellular traps. Hence, calprotectin and other AMPs use several strategies to provide microbial control and stimulate innate immunity.
Collapse
Affiliation(s)
| | - Mark C. Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
27
|
Soybean peptide inhibits the biofilm of periodontopathic bacteria via bactericidal activity. Arch Oral Biol 2022; 142:105497. [PMID: 35849907 DOI: 10.1016/j.archoralbio.2022.105497] [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: 01/21/2022] [Revised: 06/09/2022] [Accepted: 06/29/2022] [Indexed: 11/20/2022]
Abstract
OBJECTIVE This study aimed to clarify the antibacterial mechanism and antibiofilm effect of soybean-derived peptide BCBS-11 against periodontopathic bacteria. DESIGN The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of BCBS-11 against Porphyromonas gingivalis (P. gingivalis), Fusobacterium nucleatum (F. nucleatum), and Streptococcus mitis (S. mitis) were determined for the antibacterial mechanism. The effect of BCBS-11 on membrane permeability and depolarization activity were investigated using propidium iodide (PI) staining and 3, 3'-dipropylthiadicarbocyanine iodide (DiSC3-(5)) analysis. Monospecies and multispecies biofilms were cultured on 96-well plates. The amount of biofilm was determined using crystal violet staining to determine the inhibition of biofilm formation and the eradication of established biofilm using BCBS-11. The cytotoxicity of BCBS-11 was evaluated using 3-(4, 5-Dimethylthiazol-2-yl)- 2, 5-diphenyltetrazolium bromide (MTT) assay. RESULTS The MIC and MBC indicated the bactericidal activity of BCBS-11 against P. gingivalis and F. nucleatum. The PI staining revealed that BCBS-11 disrupted the bacterial membrane integrity. The DiSC3-(5) analysis indicated that BCBS-11 depolarized the bacterial cytoplasmic membrane. These results indicate the antimicrobial action of BCBS-11 through membrane disruption and the collapse of membrane electrochemical gradient. BCBS-11 significantly inhibited the monospecies biofilm formation of P. gingivalis and F. nucleatum and also inhibited dual-species biofilm. BCBS-11 was not cytotoxic toward human oral epithelial cells. CONCLUSIONS BCBS-11 inhibits the monospecies and multispecies biofilm formation of P. gingivalis and F. nucleatum, and their bactericidal activity results from membrane disruption.
Collapse
|
28
|
Sarvari R, Naghili B, Agbolaghi S, Abbaspoor S, Bannazadeh Baghi H, Poortahmasebi V, Sadrmohammadi M, Hosseini M. Organic/polymeric antibiofilm coatings for surface modification of medical devices. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2066668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Raana Sarvari
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Naghili
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Agbolaghi
- Chemical Engineering Department, Faculty of Engineering, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahdat Poortahmasebi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Sadrmohammadi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hosseini
- Chemical Engineering Department, Faculty of Engineering, Azarbaijan Shahid Madani University, Tabriz, Iran
| |
Collapse
|
29
|
Inhibitory Activity of Antibacterial Mouthwashes and Antiseptic Substances against Neisseria gonorrhoeae. Antimicrob Agents Chemother 2022; 66:e0004222. [PMID: 35579459 PMCID: PMC9211398 DOI: 10.1128/aac.00042-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Improved treatment and prevention strategies, such as antimicrobial mouthwashes, may be important for addressing the public health threat of antimicrobial-resistant Neisseria gonorrhoeae. Here, we describe the activity of seven common antibacterial mouthwashes and antiseptics against N. gonorrhoeae isolates, incorporating the use of a human saliva test matrix. Our data demonstrate that antibacterial mouthwashes and antiseptics vary in their ability to inhibit the in vitro growth of N. gonorrhoeae and saliva may impact this inhibitory activity.
Collapse
|
30
|
Tokajuk J, Deptuła P, Piktel E, Daniluk T, Chmielewska S, Wollny T, Wolak P, Fiedoruk K, Bucki R. Cathelicidin LL-37 in Health and Diseases of the Oral Cavity. Biomedicines 2022; 10:1086. [PMID: 35625823 PMCID: PMC9138798 DOI: 10.3390/biomedicines10051086] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 02/07/2023] Open
Abstract
The mechanisms for maintaining oral cavity homeostasis are subject to the constant influence of many environmental factors, including various chemicals and microorganisms. Most of them act directly on the oral mucosa, which is the mechanical and immune barrier of the oral cavity, and such interaction might lead to the development of various oral pathologies and systemic diseases. Two important players in maintaining oral health or developing oral pathology are the oral microbiota and various immune molecules that are involved in controlling its quantitative and qualitative composition. The LL-37 peptide is an important molecule that upon release from human cathelicidin (hCAP-18) can directly perform antimicrobial action after insertion into surface structures of microorganisms and immunomodulatory function as an agonist of different cell membrane receptors. Oral LL-37 expression is an important factor in oral homeostasis that maintains the physiological microbiota but is also involved in the development of oral dysbiosis, infectious diseases (including viral, bacterial, and fungal infections), autoimmune diseases, and oral carcinomas. This peptide has also been proposed as a marker of inflammation severity and treatment outcome.
Collapse
Affiliation(s)
- Joanna Tokajuk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
- Dentistry and Medicine Tokajuk, Zelazna 9/7, 15-297 Bialystok, Poland
| | - Piotr Deptuła
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Mickiewicza 2B, 15-222 Białystok, Poland;
| | - Tamara Daniluk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
| | - Sylwia Chmielewska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
| | - Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734 Kielce, Poland;
| | - Przemysław Wolak
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland;
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Mickiewicza 2C, 15-222 Białystok, Poland; (J.T.); (P.D.); (T.D.); (S.C.); (K.F.)
| |
Collapse
|
31
|
Ebersole JL, Kirakodu S, Nguyen L, Gonzalez OA. Gingival Transcriptome of Innate Antimicrobial Factors and the Oral Microbiome With Aging and Periodontitis. FRONTIERS IN ORAL HEALTH 2022; 3:817249. [PMID: 35330821 PMCID: PMC8940521 DOI: 10.3389/froh.2022.817249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/04/2022] [Indexed: 12/30/2022] Open
Abstract
The epithelial barrier at mucosal sites comprises an important mechanical protective feature of innate immunity, and is intimately involved in communicating signals of infection/tissue damage to inflammatory and immune cells in these local environments. A wide array of antimicrobial factors (AMF) exist at mucosal sites and in secretions that contribute to this innate immunity. A non-human primate model of ligature-induced periodontitis was used to explore characteristics of the antimicrobial factor transcriptome (n = 114 genes) of gingival biopsies in health, initiation and progression of periodontal lesions, and in samples with clinical resolution. Age effects and relationship of AMF to the dominant members of the oral microbiome were also evaluated. AMF could be stratified into 4 groups with high (n = 22), intermediate (n = 29), low (n = 18) and very low (n = 45) expression in healthy adult tissues. A subset of AMF were altered in healthy young, adolescent and aged samples compared with adults (e.g., APP, CCL28, DEFB113, DEFB126, FLG2, PRH1) and were affected across multiple age groups. With disease, a greater number of the AMF genes were affected in the adult and aged samples with skewing toward decreased expression, for example WDC12, PGLYRP3, FLG2, DEFB128, and DEF4A/B, with multiple age groups. Few of the AMF genes showed a >2-fold increase with disease in any age group. Selected AMF exhibited significant positive correlations across the array of AMF that varied in health and disease. In contrast, a rather limited number of the AMF significantly correlated with members of the microbiome; most prominent in healthy samples. These correlated microbes were different in younger and older samples and differed in health, disease and resolution samples. The findings supported effects of age on the expression of AMF genes in healthy gingival tissues showing a relationship to members of the oral microbiome. Furthermore, a dynamic expression of AMF genes was related to the disease process and showed similarities across the age groups, except for low/very low expressed genes that were unaffected in young samples. Targeted assessment of AMF members from this large array may provide insight into differences in disease risk and biomolecules that provide some discernment of early transition to disease.
Collapse
Affiliation(s)
- Jeffrey L. Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
| | - Linh Nguyen
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, NV, United States
| | - Octavio A. Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, United States
- Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
32
|
Andrukhov O, Blufstein A, Behm C. A Review of Antimicrobial Activity of Dental Mesenchymal Stromal Cells: Is There Any Potential? FRONTIERS IN ORAL HEALTH 2022; 2:832976. [PMID: 35098213 PMCID: PMC8795861 DOI: 10.3389/froh.2021.832976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/19/2022] Open
Abstract
Antimicrobial defense is an essential component of host-microbial homeostasis and contributes substantially to oral health maintenance. Dental mesenchymal stromal cells (MSCs) possess multilineage differentiation potential, immunomodulatory properties and play an important role in various processes like regeneration and disease progression. Recent studies show that dental MSCs might also be involved in antibacterial defense. This occurs by producing antimicrobial peptides or attracting professional phagocytic immune cells and modulating their activity. The production of antimicrobial peptides and immunomodulatory abilities of dental MSCs are enhanced by an inflammatory environment and influenced by vitamin D3. Antimicrobial peptides also have anti-inflammatory effects in dental MSCs and improve their differentiation potential. Augmentation of antibacterial efficiency of dental MSCs could broaden their clinical application in dentistry.
Collapse
Affiliation(s)
- Oleh Andrukhov
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Alice Blufstein
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
- Division of Conservative Dentistry and Periodontology, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| | - Christian Behm
- Competence Center for Periodontal Research, University Clinic of Dentistry, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
33
|
Vinel A, Al Halabi A, Roumi S, Le Neindre H, Millavet P, Simon M, Cuny C, Barthet JS, Barthet P, Laurencin-Dalicieux S. Non-surgical Periodontal Treatment: SRP and Innovative Therapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:303-327. [DOI: 10.1007/978-3-030-96881-6_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
34
|
Host Defence Peptides in Diabetes Mellitus Type 2 Patients with Periodontal Disease. A Systematic Review. Diagnostics (Basel) 2021; 11:diagnostics11122210. [PMID: 34943445 PMCID: PMC8700015 DOI: 10.3390/diagnostics11122210] [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: 11/02/2021] [Revised: 11/20/2021] [Accepted: 11/24/2021] [Indexed: 12/09/2022] Open
Abstract
The aim of the study was to critically assess and review the latest evidence relating the associations between host defence peptides (HDPs), periodontal diseases (PD) and diabetes mellitus type 2 (DM2). To explore studies on HDPs, periodontal disease, and DM2, researchers utilised specific key phrases to search the electronic databases PubMed (National Library of Medicine), Embase (Ovid), Medline (EBSCO), and Dentistry and Oral Sciences (EBSCO). Quality assessment was conducted by means of the Newcastle Ottawa scale and the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) tool. Following a thorough screening process, a total of 12 papers (4 case-control, 6 cross-sectional, 1 animal, and 1 in vitro) fulfilled the selection criteria and were included. The majority of research found that HDPs were upregulated in DM2 patients with PD. Three investigations, however, found that HDPs were downregulated in DM2 patients with PD. HDPs play a part in the pathophysiology of PD and DM2. Nonetheless, more human, animal and laboratory investigations are needed to fully understand validation of the link, as the evidence is limited. Understanding HDPs as common moderators is critical, aimed at unlocking their potential as therapeutic and diagnostic agents.
Collapse
|
35
|
Lin B, Li R, Handley TN, Wade JD, Li W, O’Brien-Simpson NM. Cationic Antimicrobial Peptides Are Leading the Way to Combat Oropathogenic Infections. ACS Infect Dis 2021; 7:2959-2970. [PMID: 34587737 DOI: 10.1021/acsinfecdis.1c00424] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oral dental infections are one of the most common diseases affecting humans, with caries and periodontal disease having the highest incidence. Caries and periodontal disease arise from infections caused by oral bacterial pathogens. Current misuse and overuse of antibiotic treatments have led to the development of antimicrobial resistance. However, recent studies have shown that cationic antimicrobial peptides are a promising family of antibacterial agents that are active against oral pathogenic bacteria and also possess less propensity for development of antimicrobial resistance. This timely Review has a focus on two primary subjects: (i) the oral bacterial pathogens associated with dental infections and (ii) the current development of antimicrobial peptides targeting oral pathogens.
Collapse
Affiliation(s)
- Bruce Lin
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Rong Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
- Department of Biochemistry & Pharmacology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Thomas N.G. Handley
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - John D. Wade
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Victoria 3010, Australia
- School of Chemistry, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Wenyi Li
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Neil M. O’Brien-Simpson
- The Bio21 Institute of Molecular Science and Biotechnology, Melbourne Dental School, Centre for Oral Health Research, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
36
|
Coker MO, Cairo C, Garzino-Demo A. HIV-Associated Interactions Between Oral Microbiota and Mucosal Immune Cells: Knowledge Gaps and Future Directions. Front Immunol 2021; 12:676669. [PMID: 34616391 PMCID: PMC8488204 DOI: 10.3389/fimmu.2021.676669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/06/2021] [Indexed: 02/02/2023] Open
Abstract
Even with sustained use of antiretroviral therapy (ART), HIV-infected individuals have an increased risk of systemic comorbid conditions and oral pathologies, including opportunistic infections, oral mucosal inflammation, and gingival and periodontal diseases. The immune-mediated mechanisms that drive this increased risk, in the context of sustained viral suppression, are unclear. HIV infection, even when controlled, alters microbial communities contributing to a chronic low-grade inflammatory state that underlies these non-HIV co-morbidities. The higher prevalence of dental caries, and mucosal and periodontal inflammation reported in HIV-infected individuals on ART is often associated with differentially abundant oral microbial communities, possibly leading to a heightened susceptibility to inflammation. This mini-review highlights current gaps in knowledge regarding the microbe-mediated oral mucosal immunity with HIV infection while discussing opportunities for future research investigations and implementation of novel approaches to elucidate these gaps. Interventions targeting both inflammation and microbial diversity are needed to mitigate oral inflammation-related comorbidities, particularly in HIV-infected individuals. More broadly, additional research is needed to bolster general models of microbiome-mediated chronic immune activation and aid the development of precise microbiota-targeted interventions to reverse or mitigate adverse outcomes.
Collapse
Affiliation(s)
- Modupe O Coker
- Department of Oral Biology, School of Dental Medicine at Rutgers, Newark, NJ, United States.,Department of Epidemiology, School of Public Health at Rutgers, Newark, NJ, United States
| | - Cristiana Cairo
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, United States.,Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - Alfredo Garzino-Demo
- Institute of Human Virology, School of Medicine, University of Maryland, Baltimore, MD, United States.,Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, United States.,Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
37
|
Caiaffa KS, Dos Santos VR, Abuna GF, Santos-Filho NA, Cilli EM, Sakai VT, Cintra LTA, Duque C. Cytocompatibility and Synergy of EGCG and Cationic Peptides Against Bacteria Related to Endodontic Infections, in Planktonic and Biofilm Conditions. Probiotics Antimicrob Proteins 2021; 13:1808-1819. [PMID: 34402021 DOI: 10.1007/s12602-021-09830-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 12/01/2022]
Abstract
This study evaluated the cytocompatibility and antimicrobial/antibiofilm effects of epigallocatechin-3-gallate (EGCG) associated with peptide LL-37 and its analogue KR-12-a5 against oral pathogens. The effect of the compounds on metabolism of fibroblasts was evaluated by methyltetrazolium assays. Antimicrobial activity of the compounds was evaluated on Streptococcus mutans, Enterococcus faecalis, Actinomyces israelii, and Fusobacterium nucleatum under planktonic conditions, on single- and dual-species biofilms and E. faecalis biofilms in dentinal tubules and analyzed by bacterial counts and confocal microscopy. Data were statistically analyzed considering p < 0.05. EGCG and peptide combinations were not toxic to fibroblasts. KR-12-a5 showed synergistic or addictive effects with EGCG and LL-37 against all bacteria tested. However, EGCG associated with KR-12-a5 demonstrated the highest bactericidal activity on all bacteria tested, at lower concentrations. In single-species biofilms, EGCG + KR-12-a5 eliminated S. mutans and A. israelii and reduced E. faecalis and F. nucleatum counts around 5 log CFU/mL. EGCG + KR-12-a5 reduced E. faecalis (-3.93 log CFU/mL) and eliminated S. mutans in dual-species biofilms. No growth of E. faecalis and significant reduction in A. israelii (-6.24 log CFU/mL) and F. nucleatum (-4.62 log CFU/mL) counts were detected in dual-species biofilms. The combination of EGCG and KR-12-a5 led to 88% of E. faecalis dead cells inside dentin tubules. The association of EGCG and KR-12-a5 was cytocompatible and promoted synergistic effect against biofilms of bacteria associated with endodontic infections.
Collapse
Affiliation(s)
- Karina Sampaio Caiaffa
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, São Paulo State University (UNESP), Aracatuba, São Paulo, Brazil
| | - Vanessa Rodrigues Dos Santos
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, São Paulo State University (UNESP), Aracatuba, São Paulo, Brazil
| | - Gabriel Flores Abuna
- Department of Restorative Dentistry, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Norival Alves Santos-Filho
- Department of Biochemistry and Organic Chemistry, São Paulo State University - Institute of Chemistry (UNESP), Araraquara, São Paulo, Brazil
| | - Eduardo Maffud Cilli
- Department of Biochemistry and Organic Chemistry, São Paulo State University - Institute of Chemistry (UNESP), Araraquara, São Paulo, Brazil
| | - Vivien Thiemy Sakai
- Department of Clinics and Surgery, School of Dentistry, Federal University of Alfenas (UNIFAL-MG), Alfenas, Minas Gerais, Brazil
| | - Luciano Tavares Angelo Cintra
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, São Paulo State University (UNESP), Aracatuba, São Paulo, Brazil
| | - Cristiane Duque
- Department of Preventive and Restorative Dentistry, Araçatuba Dental School, São Paulo State University (UNESP), Aracatuba, São Paulo, Brazil.
| |
Collapse
|
38
|
Thomas C, Minty M, Vinel A, Canceill T, Loubières P, Burcelin R, Kaddech M, Blasco-Baque V, Laurencin-Dalicieux S. Oral Microbiota: A Major Player in the Diagnosis of Systemic Diseases. Diagnostics (Basel) 2021; 11:1376. [PMID: 34441309 PMCID: PMC8391932 DOI: 10.3390/diagnostics11081376] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
The oral cavity is host to a complex and diverse microbiota community which plays an important role in health and disease. Major oral infections, i.e., caries and periodontal diseases, are both responsible for and induced by oral microbiota dysbiosis. This dysbiosis is known to have an impact on other chronic systemic diseases, whether triggering or aggravating them, making the oral microbiota a novel target in diagnosing, following, and treating systemic diseases. In this review, we summarize the major roles that oral microbiota can play in systemic disease development and aggravation and also how novel tools can help investigate this complex ecosystem. Finally, we describe new therapeutic approaches based on oral bacterial recolonization or host modulation therapies. Collaboration in diagnosis and treatment between oral specialists and general health specialists is of key importance in bridging oral and systemic health and disease and improving patients' wellbeing.
Collapse
Affiliation(s)
- Charlotte Thomas
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
| | - Matthieu Minty
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
| | - Alexia Vinel
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
| | - Thibault Canceill
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- UMR CNRS 5085, Centre Interuniversitaire de Recherche et d’Ingénierie des Matériaux (CIRIMAT), Université Paul Sabatier, 35 Chemin des Maraichers, CEDEX 9, 31062 Toulouse, France
| | - Pascale Loubières
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
| | - Remy Burcelin
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
| | - Myriam Kaddech
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
| | - Vincent Blasco-Baque
- INSERM UMR 1297 Inserm, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Avenue Jean Poulhès 1, CEDEX 4, 31432 Toulouse, France; (A.V.); (P.L.); (R.B.); (V.B.-B.)
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
| | - Sara Laurencin-Dalicieux
- Faculté de Chirurgie Dentaire, Université Paul Sabatier III (UPS), 118 Route de Narbonne, CEDEX 9, 31062 Toulouse, France; (T.C.); (M.K.); (S.L.-D.)
- Service d’Odontologie Rangueil, CHU de Toulouse, 3 Chemin des Maraîchers, CEDEX 9, 31062 Toulouse, France
- INSERM UMR 1295, Centre d’Epidémiologie et de Recherche en Santé des Populations de Toulouse (CERPOP), Epidémiologie et Analyse en Santé Publique, Risques, Maladies Chroniques et Handicaps, 37 Allées Jules Guesdes, 31000 Toulouse, France
| |
Collapse
|
39
|
An Overview of Physical, Microbiological and Immune Barriers of Oral Mucosa. Int J Mol Sci 2021; 22:ijms22157821. [PMID: 34360589 PMCID: PMC8346143 DOI: 10.3390/ijms22157821] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023] Open
Abstract
The oral mucosa, which is the lining tissue of the oral cavity, is a gateway to the body and it offers first-line protection against potential pathogens, exogenous chemicals, airborne allergens, etc. by means of its physical and microbiological-immune barrier functions. For this reason, oral mucosa is considered as a mirror to the health of the individual as well as a guard or early warning system. It is organized in two main components: a physical barrier, which consists of stratified epithelial cells and cell-cell junctions, and a microbiological-immune barrier that keeps the internal environment in a condition of homeostasis. Different factors, including microorganism, saliva, proteins and immune components, have been considered to play a critical role in disruption of oral epithelial barrier. Altered mucosal structure and barrier functions results in oral pathologies as well as systemic diseases. About 700 kinds of microorganisms exist in the human mouth, constituting the oral microbiota, which plays a significant role on the induction, training and function of the host immune system. The immune system maintains the symbiotic relationship of the host with this microbiota. Crosstalk between the oral microbiota and immune system includes various interactions in homeostasis and disease. In this review, after reviewing briefly the physical barriers of oral mucosa, the fundamentals of oral microbiome and oral mucosal immunity in regard to their barrier properties will be addressed. Furthermore, their importance in development of new diagnostic, prophylactic and therapeutic strategies for certain diseases as well as in the application for personalized medicine will be discussed.
Collapse
|
40
|
Inomata M, Horie T, Into T. Effect of the Antimicrobial Peptide LL-37 on Gene Expression of Chemokines and 29 Toll-like Receptor-Associated Proteins in Human Gingival Fibroblasts Under Stimulation with Porphyromonas gingivalis Lipopolysaccharide. Probiotics Antimicrob Proteins 2021; 12:64-72. [PMID: 31686299 DOI: 10.1007/s12602-019-09600-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The antimicrobial peptide LL-37 neutralizes the biological activity of lipopolysaccharide (LPS), while it upregulates the expression of several immune-related genes. We investigated the effect of LL-37 on gene regulation of human gingival fibroblasts (HGFs), stimulated with or without Porphyromonas gingivalis-derived LPS, a ligand for Toll-like receptor (TLR). LL-37 was non-toxic to HGFs up to a concentration of 10 μg/ml. P. gingivalis LPS upregulated the expression of IL8, CXCL10, and CCL2, whereas LL-37 reduced this upregulation. In absence of LPS, LL-37 itself upregulated the expression of IL8 and CCL2. LL-37 increased the expression of P2X7, which was constitutively expressed in HGFs. The P2X7 antagonist A-438079 suppressed the cytotoxicity and upregulatory effect of LL-37 on chemokine response, but not its downregulatory effect on P. gingivalis LPS-induced chemokine response. Whether LL-37 alters the expression of 29 genes that encode TLR-associated proteins, including TLRs, co-receptors, signaling molecules, and negative regulators, in HGFs, under stimulation with LPS, was examined. Among TLRs, P. gingivalis LPS upregulated the level of TLR4, whereas LL-37 reduced it. In co-receptors, LL-37 downregulated the level of CD14. Among signaling molecules, LL-37 augmented the LPS-upregulated expression of IRAK1. Similar effects were observed in the specific negative regulators TNFAIP3, RNF216, TOLLIP, and SIGIRR. Our results suggest that LL-37 exerts cytotoxicity and upregulation of chemokine response via the P2X7 receptor, while it induces downregulation of P. gingivalis LPS-induced chemokine response through alteration in the expression of 7 specific TLR-associated genes: downregulation of TLR4 and CD14 and upregulation of IRAK1, TNFAIP3, RNF216, TOLLIP, and SIGIRR.
Collapse
Affiliation(s)
- Megumi Inomata
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, 501-0296, Japan.
| | - Toshi Horie
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, 501-0296, Japan
| | - Takeshi Into
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, 501-0296, Japan.
| |
Collapse
|
41
|
Yamamoto M, Aizawa R. Maintaining a protective state for human periodontal tissue. Periodontol 2000 2021; 86:142-156. [PMID: 33690927 DOI: 10.1111/prd.12367] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Periodontitis, caused by infection with periodontal pathogens, is primarily characterized by inflammatory bone resorption and destruction of connective tissue. Simply describing periodontitis as a specific bacterial infection cannot completely explain the various periodontal tissue destruction patterns observed. Periodontal tissue damage is thought to be caused by various factors. In recent years, research goals for periodontal pathogens have shifted from searching for specific pathogens to investigating mechanisms that damage periodontal tissues. Bacteria interact directly with the host in several ways, influencing expression and activity of molecules that evade host defenses, and destroying local tissues and inhibiting their repair. The host's innate and acquired immune systems are important defense mechanisms that protect periodontal tissues from attack and invasion of periodontal pathogens, thus preventing infection. Innate and acquired immunity have evolved to confront the microbial challenge, forming a seamless defense network in periodontal tissues. In the innate immune response, host cells quickly detect, via specialized receptors, macromolecules and nucleic acids present on bacterial cell walls, and this triggers a protective, inflammatory response. The work of this subsystem of host immunity is performed mainly by phagocytes, beta-defensin, and the complement system. In addition, the first line of defense in oral innate immunity is the junctional epithelium, which acts as a physical barrier to the entry of oral bacteria and other nonself substances. In the presence of a normal flora, junctional epithelial cells differentiate actively and proliferate apically, with concomitant increase in chemotactic factor expression recruiting neutrophils. These immune cells play an important role in maintaining homeostasis and the protective state in periodontal tissue because they eliminate unwanted bacteria over time. Previous studies indicate a mechanism for attracting immune cells to periodontal tissue with the purpose of maintaining a protective state; although this mechanism can function without bacteria, it is enhanced by the normal flora. A better understanding of the relationship between the protective state and its disruption in periodontal disease could lead to the development of new treatment strategies for periodontal disease.
Collapse
Affiliation(s)
- Matsuo Yamamoto
- Department of Periodontology, School of Dentistry, Showa University, Tokyo, Japan
| | - Ryo Aizawa
- Department of Periodontology, School of Dentistry, Showa University, Tokyo, Japan
| |
Collapse
|
42
|
Pan L, Zhang X, Gao Q. Effects and mechanisms of histatins as novel skin wound-healing agents. J Tissue Viability 2021; 30:190-195. [PMID: 33551241 DOI: 10.1016/j.jtv.2021.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 12/27/2020] [Accepted: 01/11/2021] [Indexed: 12/31/2022]
Abstract
Wound healing is a complex and important physiological process that maintains the integrity of skin after various injuries. Abnormal wound healing, especially of chronic wounds, impairs normal physical function. Therefore, the search for effective and safe healing agents is one of the main concerns. Histatins are histidine-rich low molecular weight peptides that are expressed in the saliva of both humans and higher primates. Histatins have two main biological effects, cell stimulation and bacteria killing, with the former playing an important role in wound healing by promoting epithelial cell and fibroblast migration and angiogenesis and enhancing the re-epithelialization of the wounded area. Because of these biological effects, histatins have been shown to be promising agents of improved wound healing. Histatins are categorized into many subtypes, of which histatin 1 and its hydrolysates are the most effective in promoting wound healing. This review addresses the bioactivity of histatins in wound healing, such as their stimulatory effects on epithelial cells and fibroblasts, and elucidates the possible mechanisms by which histatin subtypes induce their biological effects.
Collapse
Affiliation(s)
- Li Pan
- Department of Cardiopulmonary Bypass, Lanzhou University Second Hospital, Lanzhou, China
| | - Xuanfen Zhang
- Department of Orthopaedic Surgery, Lanzhou University Second Hospital, Lanzhou, China.
| | - Qiong Gao
- Department of Orthopaedic Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
43
|
Weinberg A, Tugizov S, Pandiyan P, Jin G, Rakshit S, Vyakarnam A, Naglik JR. Innate immune mechanisms to oral pathogens in oral mucosa of HIV-infected individuals. Oral Dis 2020; 26 Suppl 1:69-79. [PMID: 32862519 PMCID: PMC7570967 DOI: 10.1111/odi.13470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A crucial aspect of mucosal HIV transmission is the interaction between HIV, the local environmental milieu and immune cells. The oral mucosa comprises many host cell types including epithelial cells, CD4 + T cells, dendritic cells and monocytes/macrophages, as well as a diverse microbiome predominantly comprising bacterial species. While the oral epithelium is one of the first sites exposed to HIV through oral-genital contact and nursing infants, it is largely thought to be resistant to HIV transmission via mechanisms that are still unclear. HIV-1 infection is also associated with predisposition to secondary infections, such as tuberculosis, and other diseases including cancer. This review addresses the following questions that were discussed at the 8th World Workshop on Oral Health and Disease in AIDS held in Bali, Indonesia, 13 September –15 September 2019: (a) How does HIV infection affect epithelial cell signalling? (b) How does HIV infection affect the production of cytokines and other innate antimicrobial factors, (c) How is the mucosal distribution and function of immune cells altered in HIV infection? (d) How do T cells affect HIV (oral) pathogenesis and cancer? (e) How does HIV infection lead to susceptibility to TB infections?
Collapse
Affiliation(s)
- Aaron Weinberg
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Sharof Tugizov
- Department of Medicine, University of California-San Francisco, San Francisco, CA, USA
| | - Pushpa Pandiyan
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ge Jin
- Department of Biological Sciences, School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Srabanti Rakshit
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Annapurna Vyakarnam
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Laboratory of Immunology of HIV-TB co-infection, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, India
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
44
|
Boyapati R, Vudathaneni VKP, Nadella SB, Bollepalli AC, Marella Y, Adurty C. Reflex Gastroesophageal Disorders and Functional Dyspepsia: Potential Confounding Variables for the Progression of Chronic Periodontitis: A Clinical Study. Int J Prev Med 2020; 11:138. [PMID: 33088466 PMCID: PMC7554558 DOI: 10.4103/ijpvm.ijpvm_141_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/08/2019] [Indexed: 01/10/2023] Open
Abstract
Aim: To probe into the possible connection between gastroesophageal reflux disorders (GERDs) and functionally occurring dyspepsia as a factor raising the risk of chronic periodontitis. Materials and Methods: A cross-sectional study was carried out on 40 patients with chronic periodontitis with age group between 40–60 years. The test group included 20 people diagnosed with gastroesophageal reflux disease (GERD), according to the Montreal Definition and Classification agreement, and chronic periodontitis. Symptomatic diagnoses were done to confirm functional dyspepsia. The control group comprised 20 systematically healthy people suffering from chronic periodontitis. Indices measured included flow-rate of saliva, repetitive saliva swallowing test for swallowing function, papillary marginal attachment index of gingiva, oral hygiene index-simplified and decayed, missing, filled teeth index. Data was analyzed using SPSS version 22 (IBM Inc. Chicago, USA). Descriptive statistics, such as mean and standard deviation (SD) for continuous variables and frequency and percentage for categorical variables were determined. T test was performed for intergroup comparison and Pearson correlation test was done for evaluating correlation between various parameters. P ≤ 0.05 considered as significant. Results: Statistically significant differences were observed between the test and control groups with regard to all the clinical parameters of interest. Pearson's correlation test revealed a strong negative correlation between salivary flow rate and OHI-S and DMFT scores. The RSST swallow function values demonstrated a moderate negative correlation with OHI-S scores, while OHI-I scores and DMFT scores were observed to be strongly correlated in a positive direction. A statistically significant difference was present in the probing depth and CAL levels between both the groups with higher levels in test group. Conclusion: GERD was linked to incremental incidences of chronic periodontitis and was established as an independent risk-raising factor.
Collapse
Affiliation(s)
- Ramanarayana Boyapati
- Department of Periodontics, Sibar Institute of Dental Sciences, Takkellapadu, Guntur, Andhra Pradesh, India
| | - Vijaya K P Vudathaneni
- Department of Internal Medicine, Physician, North Central Bronx Hospital, 3424 Kossuth Ave, Bronx, New York, USA
| | - Swetha B Nadella
- Department of Internal Medicine, Resident Physician, James J Peter VA Medical Center, 130W Kingsbridge Road, Bronx, New York, USA
| | - Appaiah C Bollepalli
- Department of Periodontics, Sibar Institute of Dental Sciences, Takkellapadu, Guntur, Andhra Pradesh, India
| | - Yamuna Marella
- Department of Periodontics, Sibar Institute of Dental Sciences, Takkellapadu, Guntur, Andhra Pradesh, India
| | - Chaitanya Adurty
- Department of Periodontics, Sibar Institute of Dental Sciences, Takkellapadu, Guntur, Andhra Pradesh, India
| |
Collapse
|
45
|
Abstract
Porphyromonas gingivalis is a key pathogen of periodontitis, a polymicrobial disease characterized by a chronic inflammation that destroys the tissues supporting the teeth. Thus, understanding the virulence potential of P. gingivalis is essential to maintaining a healthy oral microbiome. In nonoral organisms, CRISPR-Cas systems have been shown to modulate a variety of microbial processes, including protection from exogenous nucleic acids, and, more recently, have been implicated in bacterial virulence. Previously, our clinical findings identified activation of the CRISPR-Cas system in patient samples at the transition to disease; however, the mechanism of contribution to disease remained unknown. The importance of the present study resides in that it is becoming increasingly clear that CRISPR-associated proteins have broader functions than initially thought and that those functions now include their role in the virulence of periodontal pathogens. Studying a P. gingivaliscas3 mutant, we demonstrate that at least one of the CRISPR-Cas systems is involved in the regulation of virulence during infection. The CRISPR (clustered regularly interspaced short palindromic repeat)-Cas system is a unique genomic entity that provides prokaryotic cells with adaptive and heritable immunity. Initial studies identified CRISPRs as central elements used by bacteria to protect against foreign nucleic acids; however, emerging evidence points to CRISPR involvement in bacterial virulence. The present study aimed to identify the participation of one CRISPR-Cas protein, Cas3, in the virulence of the oral pathogen Porphyromonas gingivalis, an organism highly associated with periodontitis. Our results show that compared to the wild type, a mutant with a deletion of the Cas3 gene, an essential nuclease part of the class 1 type I CRISPR-Cas system, increased the virulence of P. gingivalis. In vitro infection modeling revealed only mildly enhanced production of proinflammatory cytokines by THP-1 cells when infected with the mutant strain. Dual transcriptome sequencing (RNA-seq) analysis of infected THP-1 cells showed an increase in expression of genes associated with pathogenesis in response to Δcas3 mutant infection, with the target of Cas3 activities in neutrophil chemotaxis and gene silencing. The importance of cas3 in controlling virulence was corroborated in a Galleria mellonella infection model, where the presence of the Δcas3 mutant resulted in a statistically significant increase in mortality of G. mellonella. A time-series analysis of transcription patterning during infection showed that G. mellonella elicited very different immune responses to the wild-type and the Δcas3 mutant strains and revealed a rearrangement of association in coexpression networks. Together, these observations show for the first time that Cas3 plays a significant role in regulating the virulence of P. gingivalis. IMPORTANCEPorphyromonas gingivalis is a key pathogen of periodontitis, a polymicrobial disease characterized by a chronic inflammation that destroys the tissues supporting the teeth. Thus, understanding the virulence potential of P. gingivalis is essential to maintaining a healthy oral microbiome. In nonoral organisms, CRISPR-Cas systems have been shown to modulate a variety of microbial processes, including protection from exogenous nucleic acids, and, more recently, have been implicated in bacterial virulence. Previously, our clinical findings identified activation of the CRISPR-Cas system in patient samples at the transition to disease; however, the mechanism of contribution to disease remained unknown. The importance of the present study resides in that it is becoming increasingly clear that CRISPR-associated proteins have broader functions than initially thought and that those functions now include their role in the virulence of periodontal pathogens. Studying a P. gingivaliscas3 mutant, we demonstrate that at least one of the CRISPR-Cas systems is involved in the regulation of virulence during infection.
Collapse
|
46
|
Bayirli BA, Öztürk A, Avci B. Serum vitamin D concentration is associated with antimicrobial peptide level in periodontal diseases. Arch Oral Biol 2020; 117:104827. [DOI: 10.1016/j.archoralbio.2020.104827] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/01/2022]
|
47
|
Tonomura S, Ihara M, Friedland RP. Microbiota in cerebrovascular disease: A key player and future therapeutic target. J Cereb Blood Flow Metab 2020; 40:1368-1380. [PMID: 32312168 PMCID: PMC7308516 DOI: 10.1177/0271678x20918031] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Stroke is the second leading cause of death and a significant cause of disability worldwide. Recent advances in DNA sequencing, proteomics, metabolomics, and computational tools are dramatically increasing access to the identification of host-microbiota interactions in systemic diseases. In this review, we describe the accumulating evidence showing how human microbiota plays an essential role in cerebrovascular diseases. We introduce the symbiotic relationships between microbiota and the mucosal immune system, focusing on differences by anatomical sites. Microbiota directly or indirectly contributes to the pathogenesis of traditional vascular risk factors including age, obesity, diabetes mellitus, dyslipidemia, and hypertension. Moreover, recent studies proposed independent effects of the microbiome on the progression of various subtypes of stroke through direct microbial invasion, exotoxins, functional amyloids, inflammation, and microbe-derived metabolites. We propose the critical concept of gene-microbial interaction to elucidate the heterogeneity of stroke and provide possible therapeutic avenues. We suggest ways to resolve the vast inter-individual diversity of cerebrovascular disease and mechanisms for personalized prevention and treatment.
Collapse
Affiliation(s)
- Shuichi Tonomura
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan.,Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Robert P Friedland
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
48
|
Stojković B, Igić M, Jevtović Stoimenov T, Tričković Janjić O, Ignjatović A, Kostić M, Petrović M, Stojanović S. Can Salivary Biomarkers Be Used as Predictors of Dental Caries in Young Adolescents? Med Sci Monit 2020; 26:e923471. [PMID: 32518218 PMCID: PMC7301671 DOI: 10.12659/msm.923471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background Identifying caries predictors in the subpopulation at risk is one of the preconditions for developing effective caries prevention measures. The present exploratory study aimed to examine the significance of socio-demographic characteristics, dietary-hygiene habits, salivary pH, and salivary antimicrobial HNP-1, hBD-2, and LL-37 peptides as potential caries risk predictors in children ages 11–13 years. Material/Methods This prospective 1-year study enrolled 213 children ages 11–13 years. The subjects underwent a dental examination and their mothers were interviewed. Unstimulated saliva was collected from the subjects to determine its pH value, as well as the salivary levels of HNP-1, hBD-2, and LL-37 peptides in 85 of the subjects. After 12 months, the 1-year caries incidence rate was recorded. Logistic regression analysis was used to estimate the ability of selected variables to predict caries risk. Results The univariable logistic regression analysis determined that the most significant independent caries risk predictors were: sex (female) (OR=2.132, p=0.007), mothers’ education (OR=1.986, p=0.020), salivary pH (OR=0.270, p=0.043), oral hygiene index (OR=1.886, p=0.015), and daily tooth brushing frequency (OR=0.565, p=0.042). The multivariable model showed that sex and oral hygiene-related variables were the most important caries predictors. Conclusions Salivary HNP-1, hBD-2, and LL-37 peptides were not found to have a significant predictive value. Therefore, socio-demographic and oral hygiene variables remain important caries predictors in early adolescents, suggesting the importance of the mechanical control of biofilm as the key measure for preventing caries. However, there is still a need for effective caries risk biomarkers, and additional research is needed in this area of caries risk prediction.
Collapse
Affiliation(s)
| | - Marija Igić
- Department of Dentistry, Medical Faculty Niš, University of Niš, Niš, Serbia
| | | | | | - Aleksandra Ignjatović
- Department of Medical Statistics and Informatics, Medical Faculty Niš, University of Niš, Niš, Serbia
| | - Miloš Kostić
- Department of Immunology, Medical Faculty Niš, University of Niš, Niš, Serbia
| | - Milica Petrović
- Department of Dentistry, Medical Faculty Niš, University of Niš, Niš, Serbia
| | - Simona Stojanović
- Department of Dentistry, Medical Faculty Niš, University of Niš, Niš, Serbia
| |
Collapse
|
49
|
Wu L, Li F, Ran L, Gao Y, Xie P, Yang J, Ke F, Liu L, Wang Q, Gao X. Insight Into the Effects of Nisin and Cecropin on the Oral Microbial Community of Rats by High-Throughput Sequencing. Front Microbiol 2020; 11:1082. [PMID: 32582069 PMCID: PMC7292207 DOI: 10.3389/fmicb.2020.01082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/30/2020] [Indexed: 12/22/2022] Open
Abstract
The oral microbiome has major impacts on oral health and disease. Antimicrobial peptides (AMPs), such as nisin and cecropin, have been widely used as food preservatives or feed additives, and are thus inevitably ingested by consumers through their oral cavity. However, as broad-spectrum antimicrobial reagents, the effect of AMPs on the oral microbiome of consumer's remains poorly characterized. In this study, we performed 16S rDNA high-throughput sequencing to investigate the effect of nisin and cecropin on the oral microbiomes of rats. Our results suggest that although nisin and cecropin have different effects on the oral microbiome of rats, both AMPs impact the composition of oral microbial communities at the phylum and genus levels. Cecropin significantly reduced the diversity and richness of rat oral microbial communities. Notably, the relative abundance of the pathogen Acinetobacter baumannii increased in the oral microbial community of rats fed cecropin-containing feed. In addition, nisin significantly reduced the amount of secretory immunoglobulin A in the saliva of rats.
Collapse
Affiliation(s)
- Lijuan Wu
- Department of Endocrinology, The Affiliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
| | - Fei Li
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Lisha Ran
- College of Integrated Traditional Chinese and Western Medicine, Southwest Medical University, Luzhou, China
| | - Yanping Gao
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Peijuan Xie
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Li Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qin Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaowei Gao
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
50
|
Nylund KM, Ruokonen H, Sorsa T, Heikkinen AM, Meurman JH, Ortiz F, Tervahartiala T, Furuholm J, Bostanci N. Association of the salivary triggering receptor expressed on myeloid cells/its ligand peptidoglycan recognition protein 1 axis with oral inflammation in kidney disease. J Periodontol 2019; 89:117-129. [PMID: 28846062 DOI: 10.1902/jop.2017.170218] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 08/16/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Triggering receptor expressed on myeloid cells (TREM-1) is a cell-surface receptor involved in amplification of inflammatory response to bacterial infections, along with its ligand peptidoglycan recognition protein 1 (PGLYRP1). TREM-1 is shed by matrix metalloproteinases (MMPs) to its soluble (s) form. The aim of the study is to investigate association of sTREM-1 and PGLYRP1 with oral inflammatory burden among patients with chronic kidney disease (CKD) at predialysis and posttransplantation stages. METHODS One hundred forty-four patients with CKD were examined at predialysis, and oral infection foci were treated prior to kidney transplantation. Fifty-three patients were available for follow-up after transplantation. Oral inflammatory burden was assessed by the Periodontal Inflammatory Burden Index (PIBI) and Total Dental Index. sTREM-1, PGLYRP1, and interleukin (IL)-1β were measured in saliva by enzyme-linked immunosorbent assay, and MMP-8 was measured by immunofluorometric assay. RESULTS In the predialysis stage, sTREM-1 and PGLYRP1 were positively associated with IL-1β, MMP-8, and PIBI. More specifically, patients with deeper probing depth (PD) (at least two sites with ≥6 mm) had higher concentrations of salivary sTREM-1 and PGLYRP1 compared with those with shallower PD. Higher concentrations of PGLYRP1 and IL-1β were associated with a higher number of teeth (> 25). On follow-up, higher PGLYRP1 and sTREM-1 were associated with one or more sites with ≥4 mm PD. CONCLUSIONS sTREM-1 and PGLYRP1 are elevated in patients with CKD with poor oral health and positively correlate with number of active periodontal pockets after oral infection therapy. Moreover, they positively correlate with MMP-8 and IL-1β. Hence, the salivary sTREM-1/PGLYRP1 axis could be useful as a diagnostic marker for oral infection within patients with CKD.
Collapse
Affiliation(s)
- Karita M Nylund
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hellevi Ruokonen
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Timo Sorsa
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland.,Department of Dental Medicine, Karolinska Insitutet, Huddinge, Sweden
| | - Anna Maria Heikkinen
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jukka H Meurman
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Fernanda Ortiz
- Department of Nephrology, Abdominal Center, Helsinki University Hospital.,Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland
| | - Taina Tervahartiala
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Furuholm
- Department of Oral and Maxillofacial Diseases, Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Nagihan Bostanci
- Department of Dental Medicine, Karolinska Insitutet, Huddinge, Sweden
| |
Collapse
|