51
|
Huang S, Cao G, Dai D, Xu Q, Ruiz S, Shindo S, Nakamura S, Kawai T, Lin J, Han X. Porphyromonas gingivalis outer membrane vesicles exacerbate retinal microvascular endothelial cell dysfunction in diabetic retinopathy. Front Microbiol 2023; 14:1167160. [PMID: 37250057 PMCID: PMC10213754 DOI: 10.3389/fmicb.2023.1167160] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/14/2023] [Indexed: 05/31/2023] Open
Abstract
Diabetic retinopathy (DR) is one of the leading causes of blindness. Periodontitis is one of the highest oral incidences and has been closely related to various systemic conditions through Porphyromonas gingivalis (P. gingivalis). P. gingivalis OMVs, derived from P. gingivalis, can cause endothelial dysfunction and potentially affect microvascular diseases. Current epidemiological studies provide limited evidence suggesting that periodontitis is associated with DR. However, there is a lack of basic research elucidating how periodontitis affects the severity of DR. This study aimed to explore the potential of P. gingivalis OMVs to contribute to the pathogenesis of DR and explore how it affect the retinal microvascular endothelium. The results demonstrated that P. gingivalis OMVs accelerated the blood-retinal barrier damage in DR mice. In vitro studies showed that the expression of inflammatory factors in human retinal microvascular endothelial cells (HRMECs) was increased after P. gingivalis OMVs stimulation, and the increased reactive oxygen species production, mitochondrial dysfunction, apoptosis, and altered endothelial permeability were observed in HRMECs under P. gingivalis OMVs stimulation. In addition, we found that protease-activated receptor-2 (PAR-2) regulated OMVs-induced TNF-α, MMP-9 mRNA expression, cell death, and endothelial permeability. Overall, we suggested that P. gingivalis OMVs induced mitochondria-related cell death of HRMECs and accelerated endothelial dysfunction, thus aggravating DR, in which PAR-2 plays a potential role. This study is the first research report to delineate the potential molecular mechanism of P. gingivalis OMVs on DR pathogenesis, which uniquely focused on elucidating the possible impact of periodontal pathogen derivatives on DR progression.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Guoqin Cao
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Dong Dai
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qiuping Xu
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Sunniva Ruiz
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Satoru Shindo
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Shin Nakamura
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Toshihisa Kawai
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Jiang Lin
- Department of Stomatology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaozhe Han
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| |
Collapse
|
52
|
Bruno JS, Al-Qadami GH, Laheij AMGA, Bossi P, Fregnani ER, Wardill HR. From Pathogenesis to Intervention: The Importance of the Microbiome in Oral Mucositis. Int J Mol Sci 2023; 24:ijms24098274. [PMID: 37175980 PMCID: PMC10179181 DOI: 10.3390/ijms24098274] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Oral mucositis (OM) is a common and impactful toxicity of standard cancer therapy, affecting up to 80% of patients. Its aetiology centres on the initial destruction of epithelial cells and the increase in inflammatory signals. These changes in the oral mucosa create a hostile environment for resident microbes, with oral infections co-occurring with OM, especially at sites of ulceration. Increasing evidence suggests that oral microbiome changes occur beyond opportunistic infection, with a growing appreciation for the potential role of the microbiome in OM development and severity. This review collects the latest articles indexed in the PubMed electronic database which analyse the bacterial shift through 16S rRNA gene sequencing methodology in cancer patients under treatment with oral mucositis. The aims are to assess whether changes in the oral and gut microbiome causally contribute to oral mucositis or if they are simply a consequence of the mucosal injury. Further, we explore the emerging role of a patient's microbial fingerprint in OM development and prediction. The maintenance of resident bacteria via microbial target therapy is under constant improvement and should be considered in the OM treatment.
Collapse
Affiliation(s)
- Julia S Bruno
- Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo 01308-060, Brazil
| | - Ghanyah H Al-Qadami
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Alexa M G A Laheij
- Department of Oral Medicine, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Department of Preventive Dentistry, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Department of Oral Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 LA Amsterdam, The Netherlands
| | - Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Eduardo R Fregnani
- Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo 01308-060, Brazil
| | - Hannah R Wardill
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5005, Australia
- The Supportive Oncology Research Group, Precision Cancer Medicine Theme, The South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| |
Collapse
|
53
|
Luo R, Chang Y, Liang H, Zhang W, Song Y, Li G, Yang C. Interactions between extracellular vesicles and microbiome in human diseases: New therapeutic opportunities. IMETA 2023; 2:e86. [PMID: 38868436 PMCID: PMC10989913 DOI: 10.1002/imt2.86] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/21/2022] [Accepted: 01/14/2023] [Indexed: 06/14/2024]
Abstract
In recent decades, accumulating research on the interactions between microbiome homeostasis and host health has broadened new frontiers in delineating the molecular mechanisms of disease pathogenesis and developing novel therapeutic strategies. By transporting proteins, nucleic acids, lipids, and metabolites in their versatile bioactive molecules, extracellular vesicles (EVs), natural bioactive cell-secreted nanoparticles, may be key mediators of microbiota-host communications. In addition to their positive and negative roles in diverse physiological and pathological processes, there is considerable evidence to implicate EVs secreted by bacteria (bacterial EVs [BEVs]) in the onset and progression of various diseases, including gastrointestinal, respiratory, dermatological, neurological, and musculoskeletal diseases, as well as in cancer. Moreover, an increasing number of studies have explored BEV-based platforms to design novel biomedical diagnostic and therapeutic strategies. Hence, in this review, we highlight the recent advances in BEV biogenesis, composition, biofunctions, and their potential involvement in disease pathologies. Furthermore, we introduce the current and emerging clinical applications of BEVs in diagnostic analytics, vaccine design, and novel therapeutic development.
Collapse
Affiliation(s)
- Rongjin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Spine Surgery, Honghui HospitalXi'an Jiaotong UniversityXi'anChina
| | - Yanmin Chang
- Department of Neurology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yu Song
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
54
|
Liu M, Shao J, Zhao Y, Ma B, Ge S. Porphyromonas gingivalis Evades Immune Clearance by Regulating Lysosome Efflux. J Dent Res 2023; 102:555-564. [PMID: 36800907 DOI: 10.1177/00220345221146097] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023] Open
Abstract
Porphyromonas gingivalis, a major periodontal pathogen, invades autophagosomes of cells, including gingival epithelial cells, endothelial cells, gingival fibroblasts, macrophages, and dendritic cells, to escape antimicrobial autophagy and lysosome fusion. However, it is not known how P. gingivalis resists autophagic immunity, survives within cells, and induces inflammation. Thus, we investigated whether P. gingivalis could escape antimicrobial autophagy by promoting lysosome efflux to block autophagic maturation, leading to intracellular survival, and whether the growth of P. gingivalis within cells results in cellular oxidative stress, causing mitochondrial damage and inflammatory responses. P. gingivalis invaded human immortalized oral epithelial cells in vitro and mouse oral epithelial cells of gingival tissues in vivo. The production of reactive oxygen species (ROS) increased upon bacterial invasion, as well as mitochondrial dysfunction-related parameters with downregulated mitochondrial membrane potential and intracellular adenosine triphosphate (ATP), upregulated mitochondrial membrane permeability, intracellular Ca2+ influx, mitochondrial DNA expression, and extracellular ATP. Lysosome excretion was elevated, the number of intracellular lysosomes was diminished, and lysosomal-associated membrane protein 2 was downregulated. Expression of autophagy-related proteins, microtubule-associated protein light chain 3, sequestosome-1, the NLRP3 inflammasome, and interleukin-1β increased with P. gingivalis infection. P. gingivalis may survive in vivo by promoting lysosome efflux, blocking autophagosome-lysosome fusion, and destroying autophagic flux. As a result, ROS and damaged mitochondria accumulated and activated the NLRP3 inflammasome, which recruited the adaptor protein ASC and caspase 1, leading to the production of proinflammatory factor interleukin-1β and inflammation.
Collapse
Affiliation(s)
- M Liu
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - J Shao
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Y Zhao
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - B Ma
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - S Ge
- Department of Periodontology & Tissue Engineering and Regeneration, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| |
Collapse
|
55
|
de Medeiros RA, da Silva YM, Miranda YMS, Gomes DDS, Carvalho TRB, Tanaka EB, de Oliveira PGFP, Nogueira JSE, de Menezes SAF, Menezes TODA, Laurentino RV, Fonseca RRDS, Machado LFA. Digital Form for Assessing Dentistry Undergraduates Regarding Periodontal Disease Associated with Cardiovascular Diseases. Medicina (B Aires) 2023; 59:medicina59030509. [PMID: 36984510 PMCID: PMC10053880 DOI: 10.3390/medicina59030509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023] Open
Abstract
Background: Throughout recent years, periodontal disease (PD) has been linked to innumerable medical systemic conditions, such as cardiovascular disease (CVD). This association could negatively impact oral health, so the knowledge of dentists who have graduated must follow modern dentistry in order to promote oral health, mainly in systemically compromised patients. Therefore, the present study aimed to determine and evaluate the knowledge level of dentistry undergraduate students (DUS) regarding the correct periodontal treatment and management of cardiac patients with PD. Methods: This cross-sectional and populational-based study was conducted between March and June 2022 in northern Brazil. A total of 153 DUS received an anonymous digital form (Google Forms Platform) using a non-probabilistic “snowball” sampling technique. The digital form was composed of four blocks of dichotomous and multiple-choice questions. After signing the informed consent term, DUS were divided into three groups according to their period/semester in dentistry graduation during the study time (G1: 1st period/semester; G2: 5th period/semester and G3: 10th period/semester). A total of 25 questions referring to demographic, educational and knowledge data about the dental and periodontal care of cardiac patients with PD were asked, and all data were presented as descriptive percentages and then analyzed using the Kappa test. Results: From a total of 153 (100%) DUS, the sample was mostly composed of 104 (68%) female participants, with an average age of 21.1 years. Regarding basic knowledge, the majority of answers were no, with G1 being higher than G2 and G3. Regarding clinical questions, 1247 (58.3%) answers were no. Additionally, regarding fundamental clinical questions 1, 2, 3, 7, 9, 11, 13 and 14, the majority of G1, G2 and G3 answered no, demonstrating a major lack of knowledge. Conclusions: In our study, DUS demonstrated a low knowledge level of the dental and periodontal care of cardiac patients with PD and its bi-directional link. Thus, according to our results, an improvement in dentistry educational programs regarding periodontal medicine must be implemented.
Collapse
Affiliation(s)
| | | | | | | | | | - Erich Brito Tanaka
- School of Dentistry, University Center of State of Pará, Belém 66060-575, PA, Brazil
| | | | | | | | | | - Rogério Valois Laurentino
- Biology of Infectious and Parasitic Agents Post-Graduate Program, Federal University of Pará, Belém 66075-110, PA, Brazil
- Virology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Ricardo Roberto de Souza Fonseca
- Biology of Infectious and Parasitic Agents Post-Graduate Program, Federal University of Pará, Belém 66075-110, PA, Brazil
- Virology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Luiz Fernando Almeida Machado
- Biology of Infectious and Parasitic Agents Post-Graduate Program, Federal University of Pará, Belém 66075-110, PA, Brazil
- Virology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Correspondence:
| |
Collapse
|
56
|
Posada-López A, Duque JD, Pineda-Tamayo RA, Bedoya-Giraldo E, Botero JE. Lack of association between periodontitis and rheumatoid arthritis. REUMATOLOGIA CLINICA 2023; 19:123-129. [PMID: 36906387 DOI: 10.1016/j.reumae.2022.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/17/2022] [Indexed: 03/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Periodontitis and rheumatoid arthritis (RA) have been associated in a bidirectional way. The objective of this study was to determine the association between clinical parameters of periodontitis and RA. MATERIALS AND METHODS Seventy-five (75) participants distributed in 3 groups (21 patients with periodontitis without RA, 33 patients with periodontitis with RA and 21 patients with reduced periodontium with RA) were included in this cross-sectional study. A full periodontal and medical examination was performed in each patient. Additionally, subgingival plaque samples for the detection of Porphyromonas gingivalis (P. gingivalis) and blood samples for biochemical markers of RA were also taken. Logistic regression analysis adjusted for confounding variables, Spearman's rank correlation coefficient and a linear multivariate regression were used to analyze the data. RESULTS Patients with RA presented less severity of periodontal parameters. The highest levels of anti-citrullinated protein antibodies were detected in non-periodontitis patients with RA. Covariates such as age, P. gingivalis, diabetes, smoking, osteoporosis and use of medication were not associated with RA. All periodontal variables and P. gingivalis expressed a negative correlation with biochemical markers of RA (P<0.05). CONCLUSIONS Periodontitis was not associated with RA. Furthermore, there was no correlation between periodontal clinical parameters and biochemical markers of RA.
Collapse
Affiliation(s)
| | - Jose Daniel Duque
- Facultad de Odontología, Universidad de Antioquia, Medellín, Colombia
| | | | | | | |
Collapse
|
57
|
Oliveira MOA, Leonço ÁR, Pavani VB, Barbosa IR, Campos MM. Omega-3 Effects on Ligature-Induced Periodontitis in Rats with Fructose-Induced Metabolic Syndrome. Inflammation 2023; 46:388-403. [PMID: 36171491 DOI: 10.1007/s10753-022-01741-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/05/2022] [Accepted: 09/14/2022] [Indexed: 11/05/2022]
Abstract
Both periodontal disease (PD) and metabolic syndrome (MS) represent disorders of concern worldwide. Current evidence indicates that PD and MS might negatively influence each other, increasing the risk for cardiovascular diseases (CVD), via mutual inflammatory pathways. A failure of the inflammation resolution mechanisms is crucial for these comorbidities. Fish oil-derived omega-3 has been linked with resolution-driven responses in different pathological conditions during the last years. This study evaluated the impacts of omega-3 supplementation in a rat model combining ligature-induced PD and 10% fructose intake-elicited MS. Our main findings show that 10% fructose ingestion led to an elevation of Lee index and white adipose tissue (WAT) weight, along with hepatic alterations, accompanied by an increase of leptin, and a decrement of adiponectin serum amounts, regardless of PD induction. Noteworthy, the co-induction of PD and MS resulted in higher levels of glycemia and triglycerides, being this latter effect lessened by omega-3 supplementation. In this case, the beneficial effects of omega-3 might be associated with its ability to recover the decline of serum adiponectin levels in rats with PD plus MS. As expected, PD induction led to alveolar bone loss, independent of MS induction. However, the supplementation with omega-3 restored alveolar bone in PD control animals, but not in the rats with PD combined with MS. Our study extends the knowledge about PD and MS as comorbidities, showing novel effects of omega-3 supplementation in this context.
Collapse
Affiliation(s)
- Maysa O A Oliveira
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Álvaro R Leonço
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Curso de Graduação em Medicina, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vinícius B Pavani
- Curso de Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre, RS, 90619-900, Brazil
| | - Isadora R Barbosa
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria M Campos
- Programa de Pós-Graduação em Biologia Celular e Molecular, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Centro de Pesquisa em Toxicologia e Farmacologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, RS, Brazil.
- Curso de Graduação em Odontologia, Escola de Ciências da Saúde e da Vida, Pontifícia Universidade Católica do Rio Grande do Sul, Avenida Ipiranga, 6681, Partenon, Porto Alegre, RS, 90619-900, Brazil.
| |
Collapse
|
58
|
Ximinies AD, Dou Y, Mishra A, Zhang K, Deivanayagam C, Wang C, Fletcher HM. The Oxidative Stress-Induced Hypothetical Protein PG_0686 in Porphyromonas gingivalis W83 Is a Novel Diguanylate Cyclase. Microbiol Spectr 2023; 11:e0441122. [PMID: 36719196 PMCID: PMC10101095 DOI: 10.1128/spectrum.04411-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/06/2023] [Indexed: 02/01/2023] Open
Abstract
The survival/adaptation of Porphyromonas gingivalis to the inflammatory environment of the periodontal pocket requires an ability to overcome oxidative stress. Several functional classes of genes, depending on the severity and duration of the exposure, were induced in P. gingivalis under H2O2-induced oxidative stress. The PG_0686 gene was highly upregulated under prolonged oxidative stress. PG_0686, annotated as a hypothetical protein of unknown function, is a 60 kDa protein that carries several domains including hemerythrin, PAS10, and domain of unknown function (DUF)-1858. Although PG_0686 showed some relatedness to several diguanylate cyclases (DGCs), it is missing the classical conserved, active site sequence motif (GGD[/E]EF), commonly observed in other bacteria. PG_0686-related proteins are observed in other anaerobic bacterial species. The isogenic mutant P. gingivalis FLL361 (ΔPG_0686::ermF) showed increased sensitivity to H2O2, and decreased gingipain activity compared to the parental strain. Transcriptome analysis of P. gingivalis FLL361 showed the dysregulation of several gene clusters/operons, known oxidative stress resistance genes, and transcriptional regulators, including PG_2212, CdhR and PG_1181 that were upregulated under normal anaerobic conditions. The intracellular level of c-di-GMP in P. gingivalis FLL361 was significantly decreased compared to the parental strain. The purified recombinant PG_0686 (rPG_0686) protein catalyzed the formation of c-di-GMP from GTP. Collectively, our data suggest a global regulatory property for PG_0686 that may be part of an unconventional second messenger signaling system in P. gingivalis. Moreover, it may coordinately regulate a pathway(s) vital for protection against environmental stress, and is significant in the pathogenicity of P. gingivalis and other anaerobes. IMPORTANCE Porphyromonas gingivalis is an important etiological agent in periodontitis and other systemic diseases. There is still a gap in our understanding of the mechanisms that P. gingivalis uses to survive the inflammatory microenvironment of the periodontal pocket. The hypothetical PG_0686 gene was highly upregulated under prolonged oxidative stress. Although the tertiary structure of PG_0686 showed little relatedness to previously characterized diguanylate cyclases (DGCs), and does not contain the conserved GGD(/E)EF catalytic domain motif sequence, an ability to catalyze the formation of c-di-GMP from GTP is demonstrated. The second messenger pathway for c-di-GMP was previously predicted to be absent in P. gingivalis. PG_0686 paralogs are identified in other anaerobic bacteria. Thus, PG_0686 may represent a novel class of DGCs, which is yet to be characterized. In conclusion, we have shown, for the first time, evidence for the presence of c-di-GMP signaling with environmental stress protective function in P. gingivalis.
Collapse
Affiliation(s)
- Alexia D. Ximinies
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yuetan Dou
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Arunima Mishra
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Kangling Zhang
- Department of Pharmacology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Champion Deivanayagam
- Department of Biochemistry and Molecular Genetics, University of Alabama, Birmingham, Alabama, USA
| | - Charles Wang
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M. Fletcher
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
59
|
Kreth J, Merritt J. Illuminating the oral microbiome and its host interactions: tools and approaches for molecular ecological studies. FEMS Microbiol Rev 2023; 47:fuac052. [PMID: 36564013 PMCID: PMC9936263 DOI: 10.1093/femsre/fuac052] [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: 01/31/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022] Open
Abstract
A more comprehensive understanding of oral diseases like caries and periodontitis is dependent on an intimate understanding of the microbial ecological processes that are responsible for disease development. With this review, we provide a comprehensive overview of relevant molecular ecology techniques that have played critical roles in the current understanding of human oral biofilm development, interspecies interactions, and microbiome biogeography. The primary focus is on relevant technologies and examples available in the oral microbiology literature. However, most, if not all, of the described technologies should be readily adaptable for studies of microbiomes from other mucosal sites in the body. Therefore, this review is intended to serve as a reference guide used by microbiome researchers as they inevitably transition into molecular mechanistic studies of the many significant phenotypes observed clinically.
Collapse
Affiliation(s)
- Jens Kreth
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, MRB433, 3181 SW Sam Jackson Park Rd., #L595, Portland, OR 97239, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Justin Merritt
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, MRB433, 3181 SW Sam Jackson Park Rd., #L595, Portland, OR 97239, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
60
|
Cai R, Wang L, Zhang W, Liu B, Wu Y, Pang J, Ma C. The role of extracellular vesicles in periodontitis: pathogenesis, diagnosis, and therapy. Front Immunol 2023; 14:1151322. [PMID: 37114060 PMCID: PMC10126335 DOI: 10.3389/fimmu.2023.1151322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Periodontitis is a prevalent disease and one of the leading causes of tooth loss. Biofilms are initiating factor of periodontitis, which can destroy periodontal tissue by producing virulence factors. The overactivated host immune response is the primary cause of periodontitis. The clinical examination of periodontal tissues and the patient's medical history are the mainstays of periodontitis diagnosis. However, there is a lack of molecular biomarkers that can be used to identify and predict periodontitis activity precisely. Non-surgical and surgical treatments are currently available for periodontitis, although both have drawbacks. In clinical practice, achieving the ideal therapeutic effect remains a challenge. Studies have revealed that bacteria produce extracellular vesicles (EVs) to export virulence proteins to host cells. Meanwhile, periodontal tissue cells and immune cells produce EVs that have pro- or anti-inflammatory effects. Accordingly, EVs play a critical role in the pathogenesis of periodontitis. Recent studies have also presented that the content and composition of EVs in saliva and gingival crevicular fluid (GCF) can serve as possible periodontitis diagnostic indicators. In addition, studies have indicated that stem cell EVs may encourage periodontal regeneration. In this article, we mainly review the role of EVs in the pathogenesis of periodontitis and discuss their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Rong Cai
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Lu Wang
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Bing Liu
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
| | - Yiqi Wu
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianliang Pang
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| | - Chufan Ma
- Department of Stomatology, Air Force Medical Center, The Fourth Military Medical University, Beijing, China
- *Correspondence: Chufan Ma, ; Jianliang Pang,
| |
Collapse
|
61
|
Yao C, Lan D, Li X, Wang Y, Qi S, Liu Y. Porphyromonas gingivalis is a risk factor for the development of nonalcoholic fatty liver disease via ferroptosis. Microbes Infect 2023; 25:105040. [PMID: 35987459 DOI: 10.1016/j.micinf.2022.105040] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/20/2022] [Accepted: 08/08/2022] [Indexed: 02/04/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a metabolic liver disease that can eventually lead to liver cirrhosis and hepatocellular carcinoma. Porphyromonas gingivalis (P.g) is the main pathogen that causes periodontal disease, which participates in the development of NAFLD. The purpose of our study was to further study the direct role of P.g in NAFLD and the underlying molecular mechanism. An animal model of oral P.g administration was established, and liver function and pathology in this model were evaluated. The gut microbiome and metabolic products were analysed. Furthermore, the Th17/Treg balance in the spleen and liver was assessed. In our study, NAFLD was observed in all the mice that were orally administered P.g. The gut microbiome and metabolic products were altered after oral P.g administration. P.g and ferroptosis were observed in the livers of the mice after oral P.g administration. Additionally, ferroptosis was observed in hepatocytes in vitro, but it was reversed by ferroptosis inhibitors. In addition, P.g triggered an imbalance in the Th17/Treg ratio in the liver and spleen in vivo. These findings suggest that oral P.g administration directly induced NAFLD in mice, which may be dependent on the ferroptosis of liver cells that occurs through the Th17/Treg imbalance induced by disordered microbial metabolism. Therefore, improving the periodontal environment is a novel treatment strategy for preventing NAFLD.
Collapse
Affiliation(s)
- Chao Yao
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Medical College, Anhui University of Science and Technology, Huainan, China
| | - Dongmei Lan
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Medical College, Anhui University of Science and Technology, Huainan, China
| | - Xue Li
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Medical College, Anhui University of Science and Technology, Huainan, China
| | - Yan Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Department of Preventive Dentistry, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China
| | - Shengcai Qi
- Department of Prothodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China; Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Medical College, Anhui University of Science and Technology, Huainan, China.
| | - Yuehua Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China; Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, China.
| |
Collapse
|
62
|
Zhang X, Li X, Xu H, Fu Z, Wang F, Huang W, Wu K, Li C, Liu Y, Zou J, Zhu H, Yi H, Kaiming S, Gu M, Guan J, Yin S. Changes in the oral and nasal microbiota in pediatric obstructive sleep apnea. J Oral Microbiol 2023; 15:2182571. [PMID: 36875426 PMCID: PMC9980019 DOI: 10.1080/20002297.2023.2182571] [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] [Indexed: 03/04/2023] Open
Abstract
Background Several clinical studies have demonstrated that pediatric obstructive sleep apnea (OSA) is associated with dysbiosis of airway mucosal microbiota. However, how oral and nasal microbial diversity, composition, and structure are altered in pediatric OSA has not been systemically explored. Methods 30 polysomnography-confirmed OSA patients with adenoid hypertrophy, and 30 controls who did not have adenoid hypertrophy, were enrolled. Swabs from four surface oral tissue sites (tongue base, soft palate, both palatine tonsils, and adenoid) and one nasal swab from both anterior nares were collected. The 16S ribosomal RNA (rRNA) V3-V4 region was sequenced to identify the microbial communities. Results The beta diversity and microbial profiles were significantly different between pediatric OSA patients and controls at the five upper airway sites. The abundances of Haemophilus, Fusobacterium, and Porphyromonas were higher at adenoid and tonsils sites of pediatric patients with OSA. Functional analysis revealed that the differential pathway between the pediatric OSA patients and controls involved glycerophospholipids and amino acid metabolism. Conclusions In this study, the oral and nasal microbiome of pediatric OSA patients exhibited certain differences in composition compared with the controls. However, the microbiota data could be useful as a reference for studies on the upper airway microbiome.
Collapse
Affiliation(s)
- Xiaoman Zhang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Li
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huajun Xu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihui Fu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weijun Huang
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejia Wu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenyang Li
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yupu Liu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianyin Zou
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaming Zhu
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongliang Yi
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Su Kaiming
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meizhen Gu
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Guan
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery & Shanghai Key Laboratory of Sleep Disordered Breathing & Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
63
|
Lim Y, Kim HY, Sun-Jin An, Choi BK. Activation of bone marrow-derived dendritic cells and CD4 + T cell differentiation by outer membrane vesicles of periodontal pathogens. J Oral Microbiol 2022; 14:2123550. [PMID: 36312320 PMCID: PMC9616074 DOI: 10.1080/20002297.2022.2123550] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Outer membrane vesicles (OMVs) released from gram-negative bacteria harbor diverse molecules to communicate with host cells. In this study, we evaluated the OMVs of periodontal pathogens for their effects on the activation of dendritic cells and CD4+ T cell differentiation. OMVs of Porphyromonas gingivalis ATCC 33277, Treponema denticola ATCC 33521, and Tannerella forsythia ATCC 43037 (‘red complex’ pathogens) were isolated by density gradient ultracentrifugation. Mouse bone marrow-derived dendritic cells (BMDCs) were treated with OMVs, and OMV-primed BMDCs were cocultured with naïve CD4+ T cells to analyze the polarization of effector helper T cells. The OMVs upregulated maturation markers, including MHC class II, CD80, CD86, and CD40, on BMDCs. OMVs of P. gingivalis and T. forsythia induced the expression of the proinflammatory cytokines IL-1β, IL-6, IL-23, and IL-12p70 in BMDCs. In T. denticola OMV-primed BMDCs, proinflammatory cytokines were poorly detected, which may be attributed to posttranslational degradation due to the highly proteolytic nature of OMVs. In cocultures of naïve CD4+ T cells with OMV-primed BMDCs, OMVs of P. gingivalis and T. denticola induced the differentiation of Th17 cells, whereas T. forsythia OMVs induced Th1 cell differentiation. These results demonstrate that OMVs derived from the ‘red complex’ periodontal pathogens induce maturation of BMDCs and differentiation of naïve CD4+ T cells to Th1 or Th17 cells.
Collapse
Affiliation(s)
- Younggap Lim
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | | | - Bong-Kyu Choi
- Department of Oral Microbiology and Immunology, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
64
|
Deng DK, Zhang JJ, Gan D, Zou JK, Wu RX, Tian Y, Yin Y, Li X, Chen FM, He XT. Roles of extracellular vesicles in periodontal homeostasis and their therapeutic potential. J Nanobiotechnology 2022; 20:545. [PMID: 36585740 PMCID: PMC9801622 DOI: 10.1186/s12951-022-01757-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/23/2022] [Indexed: 01/01/2023] Open
Abstract
Periodontal tissue is a highly dynamic and frequently stimulated area where homeostasis is easily destroyed, leading to proinflammatory periodontal diseases. Bacteria-bacteria and cell-bacteria interactions play pivotal roles in periodontal homeostasis and disease progression. Several reviews have comprehensively summarized the roles of bacteria and stem cells in periodontal homeostasis. However, they did not describe the roles of extracellular vesicles (EVs) from bacteria and cells. As communication mediators evolutionarily conserved from bacteria to eukaryotic cells, EVs secreted by bacteria or cells can mediate interactions between bacteria and their hosts, thereby offering great promise for the maintenance of periodontal homeostasis. This review offers an overview of EV biogenesis, the effects of EVs on periodontal homeostasis, and recent advances in EV-based periodontal regenerative strategies. Specifically, we document the pathogenic roles of bacteria-derived EVs (BEVs) in periodontal dyshomeostasis, focusing on plaque biofilm formation, immune evasion, inflammatory pathway activation and tissue destruction. Moreover, we summarize recent advancements in cell-derived EVs (CEVs) in periodontal homeostasis, emphasizing the multifunctional biological effects of CEVs on periodontal tissue regeneration. Finally, we discuss future challenges and practical perspectives for the clinical translation of EV-based therapies for periodontitis.
Collapse
Affiliation(s)
- Dao-Kun Deng
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiu-Jiu Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Dian Gan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jie-Kang Zou
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Rui-Xin Wu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yi Tian
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yuan Yin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xuan Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Fa-Ming Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| | - Xiao-Tao He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
65
|
Karaçam S, Tunçer S. Exploiting the Acidic Extracellular pH: Evaluation of Streptococcus salivarius M18 Postbiotics to Target Cancer Cells. Probiotics Antimicrob Proteins 2022; 14:995-1011. [PMID: 34080175 DOI: 10.1007/s12602-021-09806-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2021] [Indexed: 12/24/2022]
Abstract
Previously, we showed that the growth, antibiotic resistance, and biofilm formation properties of the pathogens Pseudomonas aeruginosa and Klebsiella pneumonia were tremendously inhibited by the cell-free supernatant of the oral probiotic Streptococcus salivarius M18. These anti-pathogenic activities of the supernatant were more efficient under acidic conditions. The present approach takes advantage of the acidic nature of the tumor microenvironment to evaluate the effect of the S. salivarius M18 postbiotics on colon cancer cells. In both two-dimensional (2D) and three-dimensional (3D) cell culture models, S. salivarius M18 cell-free supernatant showed anti-cancer actions in the pH conditions mimicking the acidity of the tumor. The inhibitory effect was more prominent when the colon cancer cells have been treated with the cell-free supernatant obtained from the inulin incubated S. salivarius M18. The results of this study point out the potential of the S. salivarius M18 functional probiotic products to be used for targeting low pH environments including the unique acidic microenvironment of tumors.
Collapse
Affiliation(s)
- Sevinç Karaçam
- Department of Biotechnology, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey
- Biotechnology Application and Research Center, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey
| | - Sinem Tunçer
- Biotechnology Application and Research Center, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey.
- Department of Medical Services and Techniques, Vocational School of Health Services, Bilecik Şeyh Edebali University, 11230, Bilecik, Turkey.
| |
Collapse
|
66
|
Involvement of Bacterial Extracellular Membrane Nanovesicles in Infectious Diseases and Their Application in Medicine. Pharmaceutics 2022; 14:pharmaceutics14122597. [PMID: 36559091 PMCID: PMC9784355 DOI: 10.3390/pharmaceutics14122597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/02/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Bacterial extracellular membrane nanovesicles (EMNs) are attracting the attention of scientists more and more every year. These formations are involved in the pathogenesis of numerous diseases, among which, of course, the leading role is occupied by infectious diseases, the causative agents of which are a range of Gram-positive and Gram-negative bacteria. A separate field for the study of the role of EMN is cancer. Extracellular membrane nanovesicles nowadays have a practical application as vaccine carriers for immunization against many infectious diseases. At present, the most essential point is their role in stimulating immune response to bacterial infections and tumor cells. The possibility of nanovesicles' practical use in several disease treatments is being evaluated. In our review, we listed diseases, focusing on their multitude and diversity, for which EMNs are essential, and also considered in detail the possibilities of using EMNs in the therapy and prevention of various pathologies.
Collapse
|
67
|
du Teil Espina M, Fu Y, van der Horst D, Hirschfeld C, López-Álvarez M, Mulder LM, Gscheider C, Haider Rubio A, Huitema M, Becher D, Heeringa P, van Dijl JM. Coating and Corruption of Human Neutrophils by Bacterial Outer Membrane Vesicles. Microbiol Spectr 2022; 10:e0075322. [PMID: 36000865 PMCID: PMC9602476 DOI: 10.1128/spectrum.00753-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 08/07/2022] [Indexed: 12/30/2022] Open
Abstract
Porphyromonas gingivalis is a keystone oral pathogen that successfully manipulates the human innate immune defenses, resulting in a chronic proinflammatory state of periodontal tissues and beyond. Here, we demonstrate that secreted outer membrane vesicles (OMVs) are deployed by P. gingivalis to selectively coat and activate human neutrophils, thereby provoking degranulation without neutrophil killing. Secreted granule components with antibacterial activity, especially LL-37 and myeloperoxidase (MPO), are subsequently degraded by potent OMV-bound proteases known as gingipains, thereby ensuring bacterial survival. In contrast to neutrophils, the P. gingivalis OMVs are efficiently internalized by macrophages and epithelial cells. Importantly, we show that neutrophil coating is a conserved feature displayed by OMVs of at least one other oral pathogen, namely, Aggregatibacter actinomycetemcomitans. We conclude that P. gingivalis deploys its OMVs for a neutrophil-deceptive strategy to create a favorable inflammatory niche and escape killing. IMPORTANCE Severe periodontitis is a dysbiotic inflammatory disease that affects about 15% of the adult population, making it one of the most prevalent diseases worldwide. Importantly, periodontitis has been associated with the development of nonoral diseases, such as rheumatoid arthritis, pancreatic cancer, and Alzheimer's disease. Periodontal pathogens implicated in periodontitis can survive in the oral cavity only by avoiding the insults of neutrophils while at the same time promoting an inflamed environment where they successfully thrive. Our present findings show that outer membrane vesicles secreted by the keystone pathogen Porphyromonas gingivalis provide an effective delivery tool of virulence factors that protect the bacterium from being killed while simultaneously activating human neutrophils.
Collapse
Affiliation(s)
- Marines du Teil Espina
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yanyan Fu
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Demi van der Horst
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Claudia Hirschfeld
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Marina López-Álvarez
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lianne M. Mulder
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Costanza Gscheider
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna Haider Rubio
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Minke Huitema
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Dörte Becher
- Institute for Microbiology, Ernst-Moritz-Arndt-University Greifswald, Greifswald, Germany
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
68
|
Howard KC, Garneau-Tsodikova S. Selective Inhibition of the Periodontal Pathogen Porphyromonas gingivalis by Third-Generation Zafirlukast Derivatives. J Med Chem 2022; 65:14938-14956. [DOI: 10.1021/acs.jmedchem.2c01471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kaitlind C. Howard
- Department of Pharmaceutical Sciences, University of Kentucky, Lee T. Todd, Jr. Building, 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| | - Sylvie Garneau-Tsodikova
- Department of Pharmaceutical Sciences, University of Kentucky, Lee T. Todd, Jr. Building, 789 South Limestone Street, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
69
|
Uemura Y, Hiroshima Y, Tada A, Murakami K, Yoshida K, Inagaki Y, Kuwahara T, Murakami A, Fujii H, Yumoto H. Porphyromonas gingivalis Outer Membrane Vesicles Stimulate Gingival Epithelial Cells to Induce Pro-Inflammatory Cytokines via the MAPK and STING Pathways. Biomedicines 2022; 10:biomedicines10102643. [PMID: 36289904 PMCID: PMC9599832 DOI: 10.3390/biomedicines10102643] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 11/28/2022] Open
Abstract
Porphyromonas gingivalis (Pg) is a keystone pathogen associated with chronic periodontitis and produces outer membrane vesicles (OMVs) that contain lipopolysaccharide (LPS), gingipains, and pathogen-derived DNA and RNA. Pg-OMVs are involved in the pathogenesis of periodontitis. Pg-OMV-activated pathways that induce the production of the pro-inflammatory cytokines, interleukin (IL)-6, and IL-8 in the human gingival epithelial cell line, OBA-9, were investigated. The role of mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB in levels of Pg-OMV-induced pro-inflammatory cytokines was investigated using Western blot analysis and specific pathway inhibitors. Pg-OMVs induced IL-6 and IL-8 production via the extracellular signal-regulated kinase (Erk) 1/2, c-Jun N-terminal kinase (JNK), p38 MAPK, and NF-κB signaling pathways in OBA-9 cells. In addition, the stimulator of interferon genes (STING), an essential innate immune signaling molecule, was triggered by a cytosolic pathogen DNA. Pg-OMV-induced IL-6 and IL-8 mRNA expression and production were significantly suppressed by STING-specific small interfering RNA. Taken together, these results demonstrated that Pg-OMV-activated Erk1/2, JNK, p38 MAPK, STING, and NF-κB signaling pathways resulting in increased IL-6 and IL-8 expression in human gingival epithelial cells. These results suggest that Pg-OMVs may play important roles in periodontitis exacerbation by stimulating various pathways.
Collapse
Affiliation(s)
- Yuta Uemura
- Department of Periodontology and Endodontology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| | - Yuka Hiroshima
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
- Correspondence: ; Tel.: +81-88-633-7330
| | - Ayano Tada
- Department of Microbiology, Faculty of Medicine, Kagawa University, 1750-1 Miki, Takamatsu 761-0793, Japan
| | - Keiji Murakami
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
- Department of Clinical Nutrition, Faculty of Health Science and Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki 701-0193, Japan
| | - Kaya Yoshida
- Department of Oral Healthcare Education, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| | - Yuji Inagaki
- Department of Periodontology and Endodontology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| | - Tomomi Kuwahara
- Department of Microbiology, Faculty of Medicine, Kagawa University, 1750-1 Miki, Takamatsu 761-0793, Japan
| | - Akikazu Murakami
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| | - Hideki Fujii
- Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
- Department of Biology, Keio University School of Medicine, 4-1-1 Hiyoshi, Kohoku-ku, Yokohama 223-8521, Japan
| | - Hiromichi Yumoto
- Department of Periodontology and Endodontology, Graduate School of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima 770-8504, Japan
| |
Collapse
|
70
|
Aleksijević LH, Aleksijević M, Škrlec I, Šram M, Šram M, Talapko J. Porphyromonas gingivalis Virulence Factors and Clinical Significance in Periodontal Disease and Coronary Artery Diseases. Pathogens 2022; 11:pathogens11101173. [PMID: 36297228 PMCID: PMC9609396 DOI: 10.3390/pathogens11101173] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/27/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Porphyromonas gingivalis is a gram-negative, anaerobic bacterium that lives in the oral cavity. It is an integral part of the oral microbiome, which includes more than 500 types of bacteria. Under certain circumstances, as a consequence of virulence factors, it can become very destructive and proliferate to many cells in periodontal lesions. It is one of the causative agents present extremely often in dental plaque and is the main etiological factor in the development of periodontal disease. During various therapeutic procedures, P. gingivalis can enter the blood and disseminate through it to distant organs. This primarily refers to the influence of periodontal agents on the development of subacute endocarditis and can facilitate the development of coronary heart disease, atherosclerosis, and ischemic infarction. The action of P. gingivalis is facilitated by numerous factors of virulence and pathogenicity such as fimbriae, hemolysin, hemagglutinin, capsules, outer membrane vesicles, lipopolysaccharides, and gingipains. A special problem is the possibility of biofilm formation. P. gingivalis in a biofilm is 500 to 1000 times less sensitive to antimicrobial drugs than planktonic cells, which represents a significant problem in the treatment of infections caused by this pathogen.
Collapse
Affiliation(s)
- Lorena Horvat Aleksijević
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Correspondence: (L.H.A.); (J.T.)
| | - Marko Aleksijević
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marko Šram
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Miroslav Šram
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Cardiology, Clinical Hospital Center Osijek, 31000 Osijek, Croatia
| | - Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Correspondence: (L.H.A.); (J.T.)
| |
Collapse
|
71
|
Chow YC, Yam HC, Gunasekaran B, Lai WY, Wo WY, Agarwal T, Ong YY, Cheong SL, Tan SA. Implications of Porphyromonas gingivalis peptidyl arginine deiminase and gingipain R in human health and diseases. Front Cell Infect Microbiol 2022; 12:987683. [PMID: 36250046 PMCID: PMC9559808 DOI: 10.3389/fcimb.2022.987683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Porphyromonas gingivalis is a major pathogenic bacterium involved in the pathogenesis of periodontitis. Citrullination has been reported as the underlying mechanism of the pathogenesis, which relies on the interplay between two virulence factors of the bacterium, namely gingipain R and the bacterial peptidyl arginine deiminase. Gingipain R cleaves host proteins to expose the C-terminal arginines for peptidyl arginine deiminase to citrullinate and generate citrullinated proteins. Apart from carrying out citrullination in the periodontium, the bacterium is found capable of citrullinating proteins present in the host synovial tissues, atherosclerotic plaques and neurons. Studies have suggested that both virulence factors are the key factors that trigger distal effects mediated by citrullination, leading to the development of some non-communicable diseases, such as rheumatoid arthritis, atherosclerosis, and Alzheimer’s disease. Thus, inhibition of these virulence factors not only can mitigate periodontitis, but also can provide new therapeutic solutions for systematic diseases involving bacterial citrullination. Herein, we described both these proteins in terms of their unique structural conformations and biological relevance to different human diseases. Moreover, investigations of inhibitory actions on the enzymes are also enumerated. New approaches for identifying inhibitors for peptidyl arginine deiminase through drug repurposing and virtual screening are also discussed.
Collapse
Affiliation(s)
- Yoke Chan Chow
- Department of Bioscience, Faculty of Applied Sciences, Tunku Abdul Rahman University College, Kuala Lumpur, Malaysia
| | - Hok Chai Yam
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Baskaran Gunasekaran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Weng Yeen Lai
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Weng Yue Wo
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Tarun Agarwal
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Guntur, India
| | - Yien Yien Ong
- Department of Bioscience, Faculty of Applied Sciences, Tunku Abdul Rahman University College, Kuala Lumpur, Malaysia
| | - Siew Lee Cheong
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- *Correspondence: Sheri-Ann Tan, ; Siew Lee Cheong,
| | - Sheri-Ann Tan
- Department of Bioscience, Faculty of Applied Sciences, Tunku Abdul Rahman University College, Kuala Lumpur, Malaysia
- *Correspondence: Sheri-Ann Tan, ; Siew Lee Cheong,
| |
Collapse
|
72
|
The Relationship between Mutations in Gene-Specific Domains of Salivary Fibronectin (cFn) and Dynamin-2 (Dynm-2) and the Development of Porphyromonas gingivalis-Initiated Periodontitis. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3030015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by the destruction of the supporting structures of the teeth. Its high prevalence and negative effects on quality of life make it one of the current problems in dentistry. Porphyromonas gingivalis (P. gingivalis) is the predominant periodontal pathogen that expresses a number of virulence factors involved in the pathogenesis of periodontitis. P. gingivalis fimbriae are a critical factor in the interaction between the organism and the host tissue. They promote both bacterial adhesion and invasion into the target sites. Fimbriae are capable of binding to human saliva components, extracellular matrix proteins, and commensal bacteria, as well as firmly binding to the cellular integrin α5β1. After attachment to α5β1-integrin, P. gingivalis is captured by cellular pseudopodia, which makes invagination through an actin-mediated pathway possible. It has been proven that the invagination event also requires the participation of the host cell dynamin, actin fibers, microtubules and lipid rafts. Work has emerged investigating mutations in the proline-rich terminal domain (PRD) and their impact on disease development. Salivary antimicrobial peptides are early protective factors against microbial attack. Of great interest is fibronectin (FN) as the main competitor of P. gingivalis fimbriae. The FN can interact with cells in three different regions: the central cell-binding domain (CCBD), the COOH terminal heparin-binding domain (Hep2), and the type III connecting segment (IIICS), including the CS1 region (Yamada, 1991). CCBD is the major cell-adhesion domain of FN and contains an Arg–Gly–Asp (RGD) motif that is recognized by members of the cell adhesion receptor integrin family, including a5b1, which is the primary FN receptor in many cell types. The work focuses on identifying the relationship between the development of periodontitis and the presence of mutations in the adhesion domains of salivary proteins such as cellular fibronectin (cFN) and dynamin-2 (DYNM2).
Collapse
|
73
|
Pathobiont-responsive Th17 cells in gut-mouth axis provoke inflammatory oral disease and are modulated by intestinal microbiome. Cell Rep 2022; 40:111314. [PMID: 36070692 DOI: 10.1016/j.celrep.2022.111314] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/20/2022] [Accepted: 08/12/2022] [Indexed: 11/21/2022] Open
Abstract
Host immune response via Th17 cells against oral pathobionts is a key mediator in periodontitis development. However, where and how the Th17-type immune response is induced during the development of periodontitis is not well understood. Here, we demonstrate that gut translocation of the oral pathobiont Porphyromonas gingivalis (Pg) exacerbates oral pathobiont-induced periodontitis with enhanced Th17 cell differentiation. The oral pathobiont-responsive Th17 cells are differentiated in Peyer's patches and translocated systemically in the peripheral immune tissues. They are also capable of migrating to and accumulating in the mouth upon oral infection. Development of periodontitis via the oral pathobiont-responsive Th17 cells is regulated by the intestinal microbiome, and altering the intestinal microbiome composition with antibiotics affects the development of periodontitis. Our study highlights that pathobiont-responsive Th17 cells in the gut-mouth axis and the intestinal microbiome work together to provoke inflammatory oral diseases, including periodontitis.
Collapse
|
74
|
Porphyromonas gingivalis-mediated disruption in spiral artery remodeling is associated with altered uterine NK cell populations and dysregulated IL-18 and Htra1. Sci Rep 2022; 12:14799. [PMID: 36042379 PMCID: PMC9427787 DOI: 10.1038/s41598-022-19239-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/26/2022] [Indexed: 11/25/2022] Open
Abstract
Impaired spiral artery remodeling (IRSA) underpins the great obstetrical syndromes. We previously demonstrated that intrauterine infection with the periodontal pathogen, Porphyromonas gingivalis, induces IRSA in rats. Since our previous studies only examined the end stage of arterial remodeling, the aim of this study was to identify the impact of P. gingivalis infection on the earlier stages of remodeling. Gestation day (GD) 11 specimens, a transition point between trophoblast-independent remodeling and the start of extravillous trophoblast invasion, were compared to late stage GD18 tissues. P. gingivalis was found in decidual stroma of GD11 specimens that already had reduced spiral artery remodeling defined as smaller arterial lumen size, increased retention of vascular smooth muscle, and decreased invasion by extravillous trophoblasts. At GD11, P. gingivalis-induced IRSA coincided with altered uterine natural killer (uNK) cell populations, decreased placental bed expression of interleukin-18 (IL-18) with increased production of temperature requirement A1 (Htra1), a marker of oxidative stress. By GD18, placental bed IL-18 and Htra1 levels, and uNK cell numbers were equivalent in control and infected groups. However, infected GD18 placental bed specimens had decreased TNF + T cells. These results suggest disturbances in placental bed decidual stroma and uNK cells are involved in P. gingivalis-mediated IRSA.
Collapse
|
75
|
In Vitro and In Vivo Anti-Inflammatory Effects of TEES-10®, a Mixture of Ethanol Extracts of Ligularia stenocephala Matsum. & Koidz. and Secale cereale L. Sprout, on Gingivitis and Periodontitis. Dent J (Basel) 2022; 10:dj10080143. [PMID: 36005241 PMCID: PMC9406350 DOI: 10.3390/dj10080143] [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: 06/07/2022] [Revised: 07/13/2022] [Accepted: 07/26/2022] [Indexed: 11/25/2022] Open
Abstract
Gingivitis and periodontitis are inflammatory disorders caused by dental plaque and calculus. These disorders often lead to tooth loss if not treated properly. Although antibiotics can be used, it is hard to treat them due to the difficulty in supplying effective doses of antibiotics to lesion areas and side effects associated with long-term use of antibiotics. In the present study, attempts were made to provide in vitro and in vivo evidence to support anti-inflammatory activities of TEES-10®, a mixture of ethanol extracts of Ligularia stenocephala (LSE) and Secale cereale L. sprout (SCSE) toward gingivitis and periodontitis by performing the following experiments. TEES-10® with a ratio of 6:4 (LSE:SCSE) showed the best effects in both stimulating the viability and inhibiting the cytotoxicity. In in vitro experiments, TEES-10® showed an ability to scavenge 2,2-diphenyl-1-picrylhydrazyl and superoxide radicals and remove ROS generated in periodontal ligament cells treated with lipopolysaccharide. TEES-10® also enhanced the viability of stem cells from human exfoliated deciduous teeth and stimulated the osteogenic differentiation of deciduous teeth cells. In in vivo experiments using rats with induced periodontitis, TEES-10® significantly decreased inflammatory cell infiltration and the numbers of osteoclasts, increased alveolar process volume and the numbers of osteoblasts, decreased serum levels of IL-1β and TNF-α (pro-inflammatory cytokines), and increased serum levels of IL-10 and IL-13 (anti-inflammatory cytokines). These results strongly support the theory that TEES-10® has the potential to be developed as a health functional food that can treat and prevent gingival and periodontal diseases and improve dental health.
Collapse
|
76
|
Porphyromonas gingivalis outer membrane vesicles modulate cytokine and chemokine production by gingipain-dependent mechanisms in human macrophages. Arch Oral Biol 2022; 140:105453. [DOI: 10.1016/j.archoralbio.2022.105453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 11/19/2022]
|
77
|
Porphyromonas gingivalis resistance and virulence: An integrated functional network analysis. Gene 2022; 839:146734. [PMID: 35835406 DOI: 10.1016/j.gene.2022.146734] [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: 01/31/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND The gram-negative bacteria Porphyromonas gingivalis (PG) is the most prevalent cause of periodontal diseases and multidrug-resistant (MDR) infections. Periodontitis and MDR infections are severe due to PG's ability to efflux antimicrobial and virulence factors. This gives rise to colonisation, biofilm development, evasion, and modulation of the host defence system. Despite extensive studies on the MDR efflux pump in other pathogens, little is known about the efflux pump and its association with the virulence factor in PG. Prolonged infection of PG leads to complete loss of teeth and other systemic diseases. This necessitates the development of new therapeutic interventions to prevent and control MDR. OBJECTIVE The study aims to identify the most indispensable proteins that regulate both resistance and virulence in PG, which could therefore be used as a target to fight against the MDR threat to antibiotics. METHODS We have adopted a hierarchical network-based approach to construct a protein interaction network. Firstly, individual networks of four major efflux pump proteins and two virulence regulatory proteins were constructed, followed by integrating them into one. The relationship between proteins was investigated using a combination of centrality scores, k-core network decomposition, and functional annotation, to computationally identify the indispensable proteins. RESULTS Our study identified four topologically significant genes, PG_0538, PG_0539, PG_0285, and PG_1797, as potential pharmacological targets. PG_0539 and PG_1797 were identified to have significant associations between the efflux pump and virulence genes. This type of underpinning research may help in narrowing the drug spectrum used for treating periodontal diseases, and may also be exploited to look into antibiotic resistance and pathogenicity in bacteria other than PG.
Collapse
|
78
|
Moradali MF, Ghods S, Bähre H, Lamont RJ, Scott DA, Seifert R. Atypical cyclic di-AMP signaling is essential for Porphyromonas gingivalis growth and regulation of cell envelope homeostasis and virulence. NPJ Biofilms Microbiomes 2022; 8:53. [PMID: 35794154 PMCID: PMC9259658 DOI: 10.1038/s41522-022-00316-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/13/2022] [Indexed: 02/04/2023] Open
Abstract
Microbial pathogens employ signaling systems through cyclic (di-) nucleotide monophosphates serving as second messengers to increase fitness during pathogenesis. However, signaling schemes via second messengers in Porphyromonas gingivalis, a key Gram-negative anaerobic oral pathogen, remain unknown. Here, we report that among various ubiquitous second messengers, P. gingivalis strains predominantly synthesize bis-(3',5')-cyclic di-adenosine monophosphate (c-di-AMP), which is essential for their growth and survival. Our findings demonstrate an unusual regulation of c-di-AMP synthesis in P. gingivalis. P. gingivalis c-di-AMP phosphodiesterase (PDE) gene (pdepg) positively regulates c-di-AMP synthesis and impedes a decrease in c-di-AMP concentration despite encoding conserved amino acid motifs for phosphodiesterase activity. Instead, the predicted regulator gene cdaR, unrelated to the c-di-AMP PDE genes, serves as a potent negative regulator of c-di-AMP synthesis in this anaerobe. Further, our findings reveal that pdepg and cdaR are required to regulate the incorporation of ATP into c-di-AMP upon pyruvate utilization, leading to enhanced biofilm formation. We show that shifts in c-di-AMP signaling change the integrity and homeostasis of cell envelope, importantly, the structure and immunoreactivity of the lipopolysaccharide layer. Additionally, microbe-microbe interactions and the virulence potential of P. gingivalis were modulated by c-di-AMP. These studies provide the first glimpse into the scheme of second messenger signaling in P. gingivalis and perhaps other Bacteroidetes. Further, our findings indicate that c-di-AMP signaling promotes the fitness of the residents of the oral cavity and the development of a pathogenic community.
Collapse
Affiliation(s)
- M Fata Moradali
- Department of Oral Immunology and Infectious Diseases, University of Louisville, School of Dentistry, Louisville, KY, USA.
| | - Shirin Ghods
- Department of Oral Immunology and Infectious Diseases, University of Louisville, School of Dentistry, Louisville, KY, USA
| | - Heike Bähre
- Research Core Unit Metabolomics, Hannover Medical School, Hanover, Germany
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville, School of Dentistry, Louisville, KY, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville, School of Dentistry, Louisville, KY, USA
| | - Roland Seifert
- Research Core Unit Metabolomics, Hannover Medical School, Hanover, Germany
| |
Collapse
|
79
|
Peridontitis as a Risk Factor for Attention Deficit Hyperactivity Disorder: Possible Neuro-inflammatory Mechanisms. Neurochem Res 2022; 47:2925-2935. [PMID: 35764847 DOI: 10.1007/s11064-022-03650-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 10/17/2022]
Abstract
Periodontitis is a condition caused mostly by the creation of a biofilm by the bacterium P. gingivalis, which releases toxins and damages the tooth structure. Recent research studies have reported association between dental health and neuropsychiatric illnesses. Neuroinflammation triggered by the first systemic inflammation caused by the bacterium present in the oral cavities is a plausible explanation for such a relationship. Substantial amount of evidence supports the role of neuroinflammation and dysfunction of the dopaminergic system in the pathology of ADHD (Attention deficit hyperactivity disorders). Recent epidemiological, microbiological and inflammatory findings strengthen that, periodontal bacteria, which cause systemic inflammation can contribute to neuroinflammation and finally ADHD. Although both diseases are characterized by inflammation, the specific pathways and crosslink's between periodontitis and ADHD remain unknown. Here, the authors describe the inflammatory elements of periodontitis, how this dental illness causes systemic inflammation, and how this systemic inflammation contributes to deteriorating neuroinflammation in the evolution of ADHD. Therefore, the aim of this review is to present possible links and mechanisms that could confirm the evidence of this association.
Collapse
|
80
|
Wen C, Muhetaer HJ, Gao Z, Wu J. Dual response of fibroblasts viability and
Porphyromonas gingivalis
adhesion on nanostructured zirconia abutment surfaces. J Biomed Mater Res A 2022; 110:1645-1654. [PMID: 35676876 DOI: 10.1002/jbm.a.37414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Cheng Wen
- Department of Stomatology The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital Shenzhen Guangdong China
| | - Huo Jia Muhetaer
- Department of Stomatology The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital Shenzhen Guangdong China
| | - Zhengyang Gao
- Department of Stomatology The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital Shenzhen Guangdong China
| | - Jincheng Wu
- Department of Stomatology The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group Luohu People's Hospital Shenzhen Guangdong China
| |
Collapse
|
81
|
Effect of the technique of photodynamic therapy against the main microorganisms responsible for periodontitis: A systematic review of in-vitro studies. Arch Oral Biol 2022; 138:105425. [DOI: 10.1016/j.archoralbio.2022.105425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
|
82
|
Ge D, Wang F, Hu Y, Wang B, Gao X, Chen Z. Fast, Simple, and Highly Specific Molecular Detection of Porphyromonas gingivalis Using Isothermal Amplification and Lateral Flow Strip Methods. Front Cell Infect Microbiol 2022; 12:895261. [PMID: 35694545 PMCID: PMC9174636 DOI: 10.3389/fcimb.2022.895261] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/27/2022] [Indexed: 12/16/2022] Open
Abstract
Porphyromonas gingivalis is an important oral pathogen that causes periodontal disease and is difficult to culture under conventional conditions. Therefore, a reliable technique for detecting this pathogenic bacterium is required. Here, isothermal recombinase polymerase amplification (RPA), a new nucleic acid amplification method, was combined with a visualization method based on nanoparticle-based lateral flow strips (LFS) for the rapid detection of P. gingivalis. The species-specific 16S rRNA sequence of P. gingivalis was used as the target for RPA, and a set of specific primer–probe combinations were designed and screened to amplify the target sequences. As a thermostatic amplification method, the RPA reaction, under optimized conditions, takes only 30 min to complete at a constant temperature (37°C). The amplification reaction products can be detected visually by LFS without any need for special equipment. The RPA-LFS method established for the detection of P. gingivalis was shown to be highly specific in distinguishing P. gingivalis from other pathogenic organisms by using 20 clinical isolates of P. gingivalis and 23 common pathogenic microorganisms. Susceptibility measurements and probit regression analysis were performed with gradient dilutions of P. gingivalis genomic DNA. The method was obtained to be highly sensitive, with a detection limit of 9.27 CFU per reaction at 95% probability. By analyzing the gingival sulcus fluid specimens from 130 patients with chronic periodontitis, the results showed that the RPA-LFS method detected 118 positive cases and 12 negative cases of P. gingivalis, and the results obtained were consistent with those of a conventional PCR assay. The RPA–LFS method is an efficient, rapid, and convenient diagnostic method that simplifies the tedious process of detecting P. gingivalis.
Collapse
Affiliation(s)
| | | | | | | | - Xuzhu Gao
- *Correspondence: Zhenxing Chen, ; Xuzhu Gao,
| | | |
Collapse
|
83
|
Kabwe M, Dashper S, Tucci J. The Microbiome in Pancreatic Cancer-Implications for Diagnosis and Precision Bacteriophage Therapy for This Low Survival Disease. Front Cell Infect Microbiol 2022; 12:871293. [PMID: 35663462 PMCID: PMC9160434 DOI: 10.3389/fcimb.2022.871293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
While the mortality rates for many cancers have decreased due to improved detection and treatments, that of pancreatic cancer remains stubbornly high. The microbiome is an important factor in the progression of many cancers. Greater understanding of the microbiome in pancreatic cancer patients, as well as its manipulation, may assist in diagnosis and treatment of this disease. In this report we reviewed studies that compared microbiome changes in pancreatic cancer patients and non-cancer patients. We then identified which bacterial genera were most increased in relative abundance across the oral, pancreatic, duodenal, and faecal tissue microbiomes. In light of these findings, we discuss the potential for utilising these bacteria as diagnostic biomarkers, as well as their potential control using precision targeting with bacteriophages, in instances where a causal oncogenic link is made.
Collapse
Affiliation(s)
- Mwila Kabwe
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| | - Stuart Dashper
- Melbourne Dental School, University of Melbourne, Melbourne, VIC, Australia
| | - Joseph Tucci
- Department of Rural Clinical Sciences, La Trobe Rural Health School, La Trobe University, Bendigo, VIC, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bendigo, VIC, Australia
| |
Collapse
|
84
|
Hauk V, D'Eramo L, Calo G, Merech F, Doga L, Lara B, Gliosca L, Massone C, Molgatini S, Ramhorst R, Squassi A, Pérez Leirós C. Gingival crevicular fluid from pregnant women impairs trophoblast cell function and trophoblast-neutrophil interaction. Am J Reprod Immunol 2022; 88:e13558. [PMID: 35511077 DOI: 10.1111/aji.13558] [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: 12/15/2021] [Revised: 03/10/2022] [Accepted: 04/19/2022] [Indexed: 12/15/2022] Open
Abstract
PROBLEM A strong association between periodontitis and higher susceptibility to pregnancy complications like preeclampsia has been reported although the mechanisms remain elusive. Trophoblast cells modulate the recruitment and functional shaping of maternal leukocytes at early stages to sustain an antiinflammatory microenvironment and fetal growth. Neutrophil activation with reactive oxygen species (ROS) release is associated with preeclampsia. Our aim was to study the effect of the gingival crevicular fluid (GCF) from pregnant women on trophoblast cell function and trophoblast-neutrophil interaction. METHOD OF STUDY Pregnant women at 16-20 weeks of gestation (n = 27) and non-pregnant women (n = 8) as the control group were studied for gingivoperiodontal clinical score evaluation and GCF collection. Total bacteria and common periodontal pathogens were analyzed in GCF samples. The effect of each GCF sample was tested on first trimester trophoblast-derived cells to assess cell migration, cytokine expression and glucose uptake. Also, the effect of GCF on human peripheral neutrophil chemoattraction by trophoblast cells and ROS formation was assessed. RESULTS Gingival crevicular fluid from pregnant women reduced trophoblast cell migration, increased proinflammatory marker expression and glucose uptake. A significant correlation between gingivoperiodontal score and trophoblast dysfunction was observed. Upon conditioning of trophoblast cells with GCF, only the GCF from pregnant women stimulated neutrophil chemoattraction. Similarly, GCF from pregnant but not from non-pregnant controls stimulated ROS formation in neutrophils. CONCLUSIONS Gingival crevicular fluid from pregnant women is deleterious for first trimester trophoblast cell function. These effects could lead to placental homeostasis disruption underlying a pathogenic mechanism of pregnancy complications associated to periodontal disease.
Collapse
Affiliation(s)
- Vanesa Hauk
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Luciana D'Eramo
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Guillermina Calo
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Fátima Merech
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Luciana Doga
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Brenda Lara
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Laura Gliosca
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Carla Massone
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Susana Molgatini
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Microbiología, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| | - Aldo Squassi
- Universidad de Buenos Aires - Facultad de Odontología, Cátedra de Odontología Preventiva y Comunitaria, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires - CONICET, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Laboratorio de Inmunofarmacología, Buenos Aires, Argentina
| |
Collapse
|
85
|
Seymour T, Zhang J. Porphyromonas Gingivalis in the Pathogenesis of Alzheimer’s Disease and Its Therapeutic Target. JOURNAL OF EXPLORATORY RESEARCH IN PHARMACOLOGY 2022; 7:45-53. [DOI: 10.14218/jerp.2021.00030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
86
|
Park DY, Park JY, Lee D, Hwang I, Kim HS. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells 2022; 11:1079. [PMID: 35406643 PMCID: PMC8997512 DOI: 10.3390/cells11071079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
The oral cavity is the gateway for microorganisms into your body where they disseminate not only to the directly connected respiratory and digestive tracts but also to the many remote organs. Oral microbiota, travelling to the end of the intestine and circulating in our bodies through blood vessels, not only affect a gut microbiome profile but also lead to many systemic diseases. By gathering information accumulated from the era of focal infection theory to the age of revolution in microbiome research, we propose a pivotal role of "leaky gum", as an analogy of "leaky gut", to underscore the importance of the oral cavity in systemic health. The oral cavity has unique structures, the gingival sulcus (GS) and the junctional epithelium (JE) below the GS, which are rarely found anywhere else in our body. The JE is attached to the tooth enamel and cementum by hemidesmosome (HD), which is structurally weaker than desmosome and is, thus, vulnerable to microbial infiltration. In the GS, microbial biofilms can build up for life, unlike the biofilms on the skin and intestinal mucosa that fall off by the natural process. Thus, we emphasize that the GS and the JE are the weakest leaky point for microbes to invade the human body, making the leaky gum just as important as, or even more important than, the leaky gut.
Collapse
Affiliation(s)
- Do-Young Park
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Jin Young Park
- Department of Gastrointestinal Endoscopy, Apple Tree Healthcare Center, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| | - Inseong Hwang
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Hye-Sung Kim
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| |
Collapse
|
87
|
Albuquerque-Souza E, Sahingur SE. Periodontitis, chronic liver diseases, and the emerging oral-gut-liver axis. Periodontol 2000 2022; 89:125-141. [PMID: 35244954 PMCID: PMC9314012 DOI: 10.1111/prd.12427] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The liver carries out a wide range of functions ranging from the control of metabolites, nutrient storage, and detoxification to immunosurveillance. While inflammation is essential for the tissue remodeling and maintenance of homeostasis and normal liver physiology, constant exposure to dietary and microbial products creates a niche for potentially prolonged immune activation and unresolved inflammation in susceptible host. Failure to restrain inflammation can lead to development of chronic liver diseases characterized by fibrosis, cirrhosis and eventually liver failure. The liver maintains close interactions with numerous organs which can influence its metabolism and physiology. It is also known that oral cavity microenvironment can influence the physiological conditions of other organs and emerging evidence implicates that this could be true for the liver as well. Presence of chronic inflammation and dysbiotic microbiota is a common feature leading to clinical pathology both in periodontitis and chronic liver diseases (CLDs). In fact, known CLDs appear to have some relationship with periodontitis, which impacts the onset or progression of these conditions in a bidirectional crosstalk. In this review, we explore the emerging association between oral‐gut‐liver axis focusing on periodontitis and common CLDs including nonalcoholic fatty liver disease, chronic viral hepatitis, liver cirrhosis, and hepatocellular cancer. We highlight the immune pathways and oral microbiome interactions which can link oral cavity and liver health and offer perspectives for future research.
Collapse
Affiliation(s)
- Emmanuel Albuquerque-Souza
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sinem E Sahingur
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
88
|
Ren L, Shen D, Liu C, Ding Y. Protein Tyrosine and Serine/Threonine Phosphorylation in Oral Bacterial Dysbiosis and Bacteria-Host Interaction. Front Cell Infect Microbiol 2022; 11:814659. [PMID: 35087767 PMCID: PMC8787120 DOI: 10.3389/fcimb.2021.814659] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/13/2021] [Indexed: 02/05/2023] Open
Abstract
The human oral cavity harbors approximately 1,000 microbial species, and dysbiosis of the microflora and imbalanced microbiota-host interactions drive many oral diseases, such as dental caries and periodontal disease. Oral microbiota homeostasis is critical for systemic health. Over the last two decades, bacterial protein phosphorylation systems have been extensively studied, providing mounting evidence of the pivotal role of tyrosine and serine/threonine phosphorylation in oral bacterial dysbiosis and bacteria-host interactions. Ongoing investigations aim to discover novel kinases and phosphatases and to understand the mechanism by which these phosphorylation events regulate the pathogenicity of oral bacteria. Here, we summarize the structures of bacterial tyrosine and serine/threonine kinases and phosphatases and discuss the roles of tyrosine and serine/threonine phosphorylation systems in Porphyromonas gingivalis and Streptococcus mutans, emphasizing their involvement in bacterial metabolism and virulence, community development, and bacteria-host interactions.
Collapse
Affiliation(s)
- Liang Ren
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Daonan Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Ding
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
89
|
Nonaka S, Kadowaki T, Nakanishi H. Secreted gingipains from Porphyromonas gingivalis increase permeability in human cerebral microvascular endothelial cells through intracellular degradation of tight junction proteins. Neurochem Int 2022; 154:105282. [PMID: 35032577 DOI: 10.1016/j.neuint.2022.105282] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/21/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Despite a clear correlation between the infiltration of periodontal pathogens in the brain and cognitive decline in Alzheimer's disease (AD), the precise mechanism underlying bacteria crossing the blood-brain barrier (BBB) remains unclear. The periodontal pathogen Porphyromonas gingivalis produces a unique class of cysteine proteases termed gingipains. Gingipains appear to be key virulence factors that exacerbate sporadic AD. We herein report that gingipains are involved in increasing permeability of hCMEC/D3 cell monolayer, human cerebral microvascular endothelial cell lines, through degradation of tight junction proteins including zonula occludens (ZO-1) and occludin. There was a significant decrease in the mean protein levels of ZO-1 and occludin after infection of hCMEC/D3 cells with wild-type (WT) P. gingivalis. However, infection of these cells with a gingipain-deficient P. gingivalis strain showed significantly lower reduction of the mean protein levels of either ZO-1 and occludin, compared to the WT strain. Similar results were obtained after treatment with culture supernatant from WT and gingipain-deficient P. gingivalis strains. In vitro digestion of human recombinant ZO-1 and occludin by WT P. gingivalis culture supernatant in the absence or presence of gingipain inhibitors indicated that gingipains directly degraded these tight junction proteins. A close immunohistochemical examination using anti-gingipain antibody further revealed that gingipains localized in the cytosol and nuclei of hCMEC/D3 cells after infection with WT P. gingivalis and treatment with its culture supernatant. Furthermore, intracellular localization of outer membrane vesicles (OMVs) bound gingipains from WT P. gingivalis and OMV-induced degradation of ZO-1 and occludin were also observed in hCMEC/D3 cells. Thus, the delivery of gingipains into the cerebral microvascular endothelial cells, probably through OMV, may be responsible for the BBB damage through intracellular degradation of ZO-1 and occludin.
Collapse
Affiliation(s)
- Saori Nonaka
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan.
| | - Tomoko Kadowaki
- Department of Frontier Oral Science, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588, Japan
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan.
| |
Collapse
|
90
|
Wang T, Ishikawa T, Sasaki M, Chiba T. Oral and Gut Microbial Dysbiosis and Non-alcoholic Fatty Liver Disease: The Central Role of Porphyromonas gingivalis. Front Med (Lausanne) 2022; 9:822190. [PMID: 35308549 PMCID: PMC8924514 DOI: 10.3389/fmed.2022.822190] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/19/2022] [Indexed: 02/05/2023] Open
Abstract
Gut microbiota play many important roles, such as the regulation of immunity and barrier function in the intestine, and are crucial for maintaining homeostasis in living organisms. The disruption in microbiota is called dysbiosis, which has been associated with various chronic inflammatory conditions, food allergies, colorectal cancer, etc. The gut microbiota is also affected by several other factors such as diet, antibiotics and other medications, or bacterial and viral infections. Moreover, there are some reports on the oral-gut-liver axis indicating that the disruption of oral microbiota affects the intestinal biota. Non-alcoholic fatty liver disease (NAFLD) is one of the systemic diseases caused due to the dysregulation of the oral-gut-liver axis. NAFLD is the most common liver disease reported in the developed countries. It includes liver damage ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), cirrhosis, and cancer. Recently, accumulating evidence supports an association between NAFLD and dysbiosis of oral and gut microbiota. Periodontopathic bacteria, especially Porphyromonas gingivalis, have been correlated with the pathogenesis and development of NAFLD based on the clinical and basic research, and immunology. P. gingivalis was detected in the liver, and lipopolysaccharide from this bacteria has been shown to be involved in the progression of NAFLD, thereby indicating a direct role of P. gingivalis in NAFLD. Moreover, P. gingivalis induces dysbiosis of gut microbiota, which promotes the progression of NAFLD, through disrupting both metabolic and immunologic pathways. Here, we review the roles of microbial dysbiosis in NAFLD. Focusing on P. gingivalis, we evaluate and summarize the most recent advances in our understanding of the relationship between oral-gut microbiome symbiosis and the pathogenesis and progression of non-alcoholic fatty liver disease, as well as discuss novel strategies targeting both P. gingivalis and microbial dysbiosis.
Collapse
Affiliation(s)
- Ting Wang
- Division of Internal Medicine, Department of Oral Medicine, Iwate Medical University, Morioka, Japan
- Ting Wang
| | - Taichi Ishikawa
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, Morioka, Japan
| | - Minoru Sasaki
- Division of Molecular Microbiology, Department of Microbiology, Iwate Medical University, Morioka, Japan
| | - Toshimi Chiba
- Division of Internal Medicine, Department of Oral Medicine, Iwate Medical University, Morioka, Japan
- *Correspondence: Toshimi Chiba
| |
Collapse
|
91
|
Zhao X, Yang Y, Yu J, Ding R, Pei D, Zhang Y, He G, Cheng Y, Li A. Injectable hydrogels with high drug loading through B–N coordination and ROS-triggered drug release for efficient treatment of chronic periodontitis in diabetic rats. Biomaterials 2022; 282:121387. [DOI: 10.1016/j.biomaterials.2022.121387] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/10/2022] [Accepted: 01/22/2022] [Indexed: 12/27/2022]
|
92
|
Reid LV, Spalluto CM, Watson A, Staples KJ, Wilkinson TMA. The Role of Extracellular Vesicles as a Shared Disease Mechanism Contributing to Multimorbidity in Patients With COPD. Front Immunol 2021; 12:754004. [PMID: 34925327 PMCID: PMC8675939 DOI: 10.3389/fimmu.2021.754004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/04/2021] [Indexed: 01/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the leading causes of death worldwide. Individuals with COPD typically experience a progressive, debilitating decline in lung function as well as systemic manifestations of the disease. Multimorbidity, is common in COPD patients and increases the risk of hospitalisation and mortality. Central to the genesis of multimorbidity in COPD patients is a self-perpetuating, abnormal immune and inflammatory response driven by factors including ageing, pollutant inhalation (including smoking) and infection. As many patients with COPD have multiple concurrent chronic conditions, which require an integrative management approach, there is a need to greater understand the shared disease mechanisms contributing to multimorbidity. The intercellular transfer of extracellular vesicles (EVs) has recently been proposed as an important method of local and distal cell-to-cell communication mediating both homeostatic and pathological conditions. EVs have been identified in many biological fluids and provide a stable capsule for the transfer of cargo including proteins, lipids and nucleic acids. Of these cargo, microRNAs (miRNAs), which are short 17-24 nucleotide non-coding RNA molecules, have been amongst the most extensively studied. There is evidence to support that miRNA are selectively packaged into EVs and can regulate recipient cell gene expression including major pathways involved in inflammation, apoptosis and fibrosis. Furthermore changes in EV cargo including miRNA have been reported in many chronic diseases and in response to risk factors including respiratory infections, noxious stimuli and ageing. In this review, we discuss the potential of EVs and EV-associated miRNA to modulate shared pathological processes in chronic diseases. Further delineating these may lead to the identification of novel biomarkers and therapeutic targets for patients with COPD and multimorbidities.
Collapse
Affiliation(s)
- Laura V Reid
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - C Mirella Spalluto
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom.,Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
93
|
Veith PD, Glew MD, Gorasia DG, Cascales E, Reynolds EC. The Type IX Secretion System and Its Role in Bacterial Function and Pathogenesis. J Dent Res 2021; 101:374-383. [PMID: 34889148 DOI: 10.1177/00220345211051599] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Porphyromonas, Tannerella, and Prevotella species found in severe periodontitis use the Type IX Secretion System (T9SS) to load their outer membrane surface with an array of virulence factors. These virulence factors are then released on outer membrane vesicles (OMVs), which penetrate the host to dysregulate the immune response to establish a positive feedback loop of chronic, inflammatory destruction of the tooth's supporting tissues. In this review, we present the latest information on the molecular architecture of the T9SS and provide mechanistic insight into its role in secretion and attachment of cargo proteins to produce a virulence coat on cells and OMVs. The recent molecular structures of the T9SS motor comprising PorL and PorM as well as the secretion pore Sov, together with advances in the overall interactome, have provided insight into the possible mechanisms of secretion. We propose the presence of PorL/M motors arranged in a circle at the inner membrane with bent periplasmic rotors interacting with the PorN protein. At the outer membrane, we envisage a slide carousel model where the PorN protein is driven around a circular track composed of PorK. Cargo proteins are transported by PorN to PorW and the Sov translocon just as slides are rotated to the projection window. Secreted proteins are proposed to then be shuttled along highways consisting of the PorV shuttle protein to an array of attachment complexes distributed around the cell. The cell surface attachment of cargo is a hallmark of the T9SS, and in Porphyromonas gingivalis and Tannerella forsythia, this attachment is achieved via covalent bonding to a linking sugar synthesized by the Wbp/Vim pathway. The cell-surface attached cargo are enriched on OMVs, which are then released from the cell.
Collapse
Affiliation(s)
- P D Veith
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - M D Glew
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - D G Gorasia
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| | - E Cascales
- Laboratoire d'Ingénierie des Syst èmes Macromol éculaires (LISM), Institut de Microbiologie, Bioénergies and Biotechnologie (IM2B), Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), UMR7255, Marseille Cedex, France
| | - E C Reynolds
- Oral Health CRC, Melbourne Dental School, Bio21 Institute, The University of Melbourne, Victoria, Australia
| |
Collapse
|
94
|
Delivery of Toxins and Effectors by Bacterial Membrane Vesicles. Toxins (Basel) 2021; 13:toxins13120845. [PMID: 34941684 PMCID: PMC8703475 DOI: 10.3390/toxins13120845] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/22/2023] Open
Abstract
Pathogenic bacteria interact with cells of their host via many factors. The surface components, i.e., adhesins, lipoproteins, LPS and glycoconjugates, are particularly important in the initial stages of colonization. They enable adhesion and multiplication, as well as the formation of biofilms. In contrast, virulence factors such as invasins and toxins act quickly to damage host cells, causing tissue destruction and, consequently, organ dysfunction. These proteins must be exported from the bacterium and delivered to the host cell in order to function effectively. Bacteria have developed a number of one- and two-step secretion systems to transport their proteins to target cells. Recently, several authors have postulated the existence of another transport system (sometimes called "secretion system type zero"), which utilizes extracellular structures, namely membrane vesicles (MVs). This review examines the role of MVs as transporters of virulence factors and the interaction of toxin-containing vesicles and other protein effectors with different human cell types. We focus on the unique ability of vesicles to cross the blood-brain barrier and deliver protein effectors from intestinal or oral bacteria to the central nervous system.
Collapse
|
95
|
Tetrahydroimidazo[4,5- c]pyridine-Based Inhibitors of Porphyromonas gingivalis Glutaminyl Cyclase. Pharmaceuticals (Basel) 2021; 14:ph14121206. [PMID: 34959608 PMCID: PMC8709289 DOI: 10.3390/ph14121206] [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: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 01/05/2023] Open
Abstract
Periodontitis is a severe yet underestimated oral disease. Since it is linked to several systemic diseases, such as diabetes, artheriosclerosis, and even Alzheimer’s disease, growing interest in treating periodontitis has emerged recently. The major cause of periodontitis is a shift in the oral microbiome. A keystone pathogen that is associated with this shift is Porphyromonas gingivalis. Hence, targeting P. gingivalis came into focus of drug discovery for the development of novel antiinfective compounds. Among others, glutaminyl cyclases (QCs) of oral pathogens might be promising drug targets. Here, we report the discovery and structure–activity relationship of a novel class of P. gingivalis QC inhibitors according to a tetrahydroimidazo[4,5-c]pyridine scaffold. Some compounds exhibited activity in the lower nanomolar range and thus were further characterized with regard to their selectivity and toxicity.
Collapse
|
96
|
Ezhilarasan D. Deciphering the toxicological role of Porphyromonas gingivalis derived endotoxins in liver diseases. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 88:103755. [PMID: 34662732 DOI: 10.1016/j.etap.2021.103755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 06/13/2023]
Abstract
Periodontitis is a most prevalent and infectious multifactorial inflammatory disease and is characterized by the progressive destruction of the tooth-supporting tissues. Porphyromonas gingivalis, a Gram‑negative oral anaerobe, mainly causes periodontitis and it is one of the most important risk factors responsible for aggravation of existing systemic diseases. Several experimental and clinical studies have shown the positive association between periodontitis and different forms of liver disease. Periodontal diseases increase the prevalence of non-alcoholic fatty liver diseases and cirrhosis. Infected periodontium and pathogens in the periodontal microenvironments release pathogen-associated molecular patterns such as peptidoglycan, lipopolysaccharides, gingipain, fimbria, bacterial DNA, etc, and damage-associated molecular patterns such as interleukins-1α, β, - 8, and galectin-3, etc. These virulence factors and cytokines enter the bloodstream, disseminate into the whole body, and induce a variety of systemic pathological effects, including liver diseases (steatosis and fibrosis). Maintaining oral hygiene by scaling and root planning significantly improves liver damage in patients with periodontitis. Dentists and physicians should have more awareness in understanding the bidirectional nature of the relationship between oral and systemic diseases. Importantly, periodontitis condition aggravates simple fatty liver into fibrotic disease and therefore, the aim of this review is to understand the possible link between periodontitis and liver diseases.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, The Blue Lab, Molecular Medicine and Toxicology Division, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 600 077, India.
| |
Collapse
|
97
|
Zhang Z, Liu S, Zhang S, Li Y, Shi X, Liu D, Pan Y. Porphyromonas gingivalis outer membrane vesicles inhibit the invasion of Fusobacterium nucleatum into oral epithelial cells by downregulating FadA and FomA. J Periodontol 2021; 93:515-525. [PMID: 34458990 PMCID: PMC9415117 DOI: 10.1002/jper.21-0144] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/13/2021] [Accepted: 08/09/2021] [Indexed: 02/02/2023]
Abstract
Background Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum) participate in the formation and progression of periodontitis. They can exert virulence by invading into host cells, but the interaction between them and their specific mechanisms remain unclear. The purpose of this study was to study the effect of P. gingivalis outer membrane vesicles (OMVs) on the ability of F. nucleatum to invade oral epithelial cells, and the reasons for the influence. Methods The invasion abilities of the two bacteria were detected separately after mixed infection of P. gingivalis and F. nucleatum. Next, P. gingivalis OMVs were extracted with the kit, and their influence on the invasion ability of F. nucleatum was tested. The effects of P. gingivalis OMVs on F. nucleatum were evaluated by assessment of bacterial morphology, growth curves, auto‐aggregation morphology, and the expression of adhesion‐related proteins FadA and FomA. Results Our results showed that P. gingivalis inhibited the invasion of F. nucleatum into oral epithelial cells but F. nucleatum promoted the invasion of P. gingivalis. In subsequent experiments, we extracted P. gingivalis OMVs successfully and revealed that proteases in P. gingivalis OMVs inhibited the invasion of F. nucleatum into oral epithelial cells. Furthermore, P. gingivalis OMVs did not affect the morphology and proliferation of F. nucleatum, but proteases inside decreased the auto‐aggregation of F. nucleatum. Additionally, proteases in P. gingivalis OMVs reduced the expression levels of F. nucleatum surface adhesion‐related proteins FadA and FomA. Conclusion Our study demonstrated that proteases in P. gingivalis OMVs inhibited the invasion of F. nucleatum into oral epithelial cells by downregulating FadA and FomA.
Collapse
Affiliation(s)
- Zhiying Zhang
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Sai Liu
- Department of Dental Materials, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Shuwei Zhang
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yuchao Li
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Xiaoting Shi
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Dongjuan Liu
- Department of Emergency and Oral Medicine, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yaping Pan
- Department of Periodontics, Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
98
|
Nara PL, Sindelar D, Penn MS, Potempa J, Griffin WST. Porphyromonas gingivalis Outer Membrane Vesicles as the Major Driver of and Explanation for Neuropathogenesis, the Cholinergic Hypothesis, Iron Dyshomeostasis, and Salivary Lactoferrin in Alzheimer's Disease. J Alzheimers Dis 2021; 82:1417-1450. [PMID: 34275903 PMCID: PMC8461682 DOI: 10.3233/jad-210448] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/22/2022]
Abstract
Porphyromonas gingivalis (Pg) is a primary oral pathogen in the widespread biofilm-induced "chronic" multi-systems inflammatory disease(s) including Alzheimer's disease (AD). It is possibly the only second identified unique example of a biological extremophile in the human body. Having a better understanding of the key microbiological and genetic mechanisms of its pathogenesis and disease induction are central to its future diagnosis, treatment, and possible prevention. The published literature around the role of Pg in AD highlights the bacteria's direct role within the brain to cause disease. The available evidence, although somewhat adopted, does not fully support this as the major process. There are alternative pathogenic/virulence features associated with Pg that have been overlooked and may better explain the pathogenic processes found in the "infection hypothesis" of AD. A better explanation is offered here for the discrepancy in the relatively low amounts of "Pg bacteria" residing in the brain compared to the rather florid amounts and broad distribution of one or more of its major bacterial protein toxins. Related to this, the "Gingipains Hypothesis", AD-related iron dyshomeostasis, and the early reduced salivary lactoferrin, along with the resurrection of the Cholinergic Hypothesis may now be integrated into one working model. The current paper suggests the highly evolved and developed Type IX secretory cargo system of Pg producing outer membrane vesicles may better explain the observed diseases. Thus it is hoped this paper can provide a unifying model for the sporadic form of AD and guide the direction of research, treatment, and possible prevention.
Collapse
Affiliation(s)
| | | | - Marc S. Penn
- Summa Heart Health and Vascular Institute, Akron, OH, USA
| | - Jan Potempa
- Department of Oral Immunology and Infectious Diseases in the School of Dentistry, University of Louisville, Louisville, KY, USA
| | - W. Sue T. Griffin
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|