1
|
Zhu B, Anandan V, Bao L, Xu P. High-throughput characterization of the influence of Streptococcus sanguinis genes on the interaction between Streptococcus sanguinis and Porphyromonas gingivalis. Mol Oral Microbiol 2024; 39:461-467. [PMID: 39054378 DOI: 10.1111/omi.12478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 05/30/2024] [Accepted: 07/14/2024] [Indexed: 07/27/2024]
Abstract
Porphyromonas gingivalis is a keystone pathogen in periodontitis, and Streptococcus sanguinis is an abundant oral commensal bacterium associated with periodontal health. However, the interaction between P. gingivalis and S. sanguinis remains obscure. Here, we established a strategy for high-throughput measurement of the cell number of P. gingivalis in the coculture with S. sanguinis by detecting the concentration of hydrogen sulfate. The interaction between P. gingivalis and over 2000 S. sanguinis single-gene mutants was characterized using this strategy, and several interaction-associated genes in S. sanguinis were determined by detecting more P. gingivalis cells in the coculture with matched S. sanguinis mutants. Three S. sanguinis interaction-associated genes were predicted to be responsible for cysteine metabolism, and the supplementation of exogenous L-cysteine promoted the cell number of P. gingivalis in the coculture with S. sanguinis. Thus, exogenous L-cysteine and the compromised cysteine metabolism in S. sanguinis enhanced the growth of P. gingivalis in the existence of S. sanguinis. Additionally, the interaction between P. gingivalis and other Streptococcus spp. was examined, and S. pneumoniae was the only streptococci that had no inhibition on the cell number of P. gingivalis. In total, this study established a new strategy for high-throughput screening of the interaction between Streptococcus and P. gingivalis and discovered a set of genes in S. sanguinis that impacted the interaction. The influence of exogenous L-cysteine on the interaction between P. gingivalis and S. sanguinis in the oral cavity needs further investigation.
Collapse
Affiliation(s)
- Bin Zhu
- Department of Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Vysakh Anandan
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Liang Bao
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ping Xu
- Department of Microbiology & Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, Virginia, USA
- The Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
2
|
Liao L, Wang Q, Feng Y, Li G, Lai R, Jameela F, Zhan X, Liu B. Advances and challenges in the development of periodontitis vaccines: A comprehensive review. Int Immunopharmacol 2024; 140:112650. [PMID: 39079346 DOI: 10.1016/j.intimp.2024.112650] [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: 02/14/2024] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 09/01/2024]
Abstract
Periodontitis is a prevalent polymicrobial disease. It damages soft tissues and alveolar bone, and causes a significant public-health burden. Development of an advanced therapeutic approach and exploration of vaccines against periodontitis hold promise as potential treatment avenues. Clinical trials for a periodontitis vaccine are lacking. Therefore, it is crucial to address the urgent need for developing strategies to implement vaccines at the primary level of prevention in public health. A deep understanding of the principles and mechanisms of action of vaccines plays a crucial role in the successful development of vaccines and their clinical translation. This review aims to provide a comprehensive summary of potential directions for the development of highly efficacious periodontitis vaccines. In addition, we address the limitations of these endeavors and explore future possibilities for the development of an efficacious vaccine against periodontitis.
Collapse
Affiliation(s)
- Lingzi Liao
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China
| | - Qi Wang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, China
| | - Yujia Feng
- School of Stomatology, Jinan University, Guangzhou, China
| | - Guojiang Li
- School of Stomatology, Jinan University, Guangzhou, China
| | - Renfa Lai
- Hospital of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, China; School of Stomatology, Jinan University, Guangzhou, China
| | - Fatima Jameela
- Modern American Dental Clinic, West Warren Avenue, MI, USA
| | - Xiaozhen Zhan
- Hospital of Stomatology, the First Affiliated Hospital of Jinan University, Guangzhou, China; School of Stomatology, Jinan University, Guangzhou, China.
| | - Bin Liu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School and Hospital of Stomatology, Lanzhou University, Lanzhou, China.
| |
Collapse
|
3
|
Catalan EA, Seguel-Fuentes E, Fuentes B, Aranguiz-Varela F, Castillo-Godoy DP, Rivera-Asin E, Bocaz E, Fuentes JA, Bravo D, Schinnerling K, Melo-Gonzalez F. Oral Pathobiont-Derived Outer Membrane Vesicles in the Oral-Gut Axis. Int J Mol Sci 2024; 25:11141. [PMID: 39456922 PMCID: PMC11508520 DOI: 10.3390/ijms252011141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Oral pathobionts are essential in instigating local inflammation within the oral cavity and contribute to the pathogenesis of diseases in the gastrointestinal tract and other distant organs. Among the Gram-negative pathobionts, Porphyromonas gingivalis and Fusobacterium nucleatum emerge as critical drivers of periodontitis, exerting their influence not only locally but also as inducers of gut dysbiosis, intestinal disturbances, and systemic ailments. This dual impact is facilitated by their ectopic colonization of the intestinal mucosa and the subsequent mediation of distal systemic effects by releasing outer membrane vesicles (OMVs) into circulation. This review elucidates the principal components of oral pathobiont-derived OMVs implicated in disease pathogenesis within the oral-gut axis, detailing virulence factors that OMVs carry and their interactions with host epithelial and immune cells, both in vitro and in vivo. Additionally, we shed light on the less acknowledged interplay between oral pathobionts and the gut commensal Akkermansia muciniphila, which can directly impede oral pathobionts' growth and modulate bacterial gene expression. Notably, OMVs derived from A. muciniphila emerge as promoters of anti-inflammatory effects within the gastrointestinal and distant tissues. Consequently, we explore the potential of A. muciniphila-derived OMVs to interact with oral pathobionts and prevent disease in the oral-gut axis.
Collapse
Affiliation(s)
- Eduardo A. Catalan
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Emilio Seguel-Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Brandon Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Aranguiz-Varela
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Daniela P. Castillo-Godoy
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elizabeth Rivera-Asin
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elisa Bocaz
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Denisse Bravo
- Cellular Interactions Laboratory, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370133, Chile;
| | - Katina Schinnerling
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Melo-Gonzalez
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| |
Collapse
|
4
|
Talapko J, Erić S, Meštrović T, Stipetić MM, Juzbašić M, Katalinić D, Bekić S, Muršić D, Flam J, Belić D, Lešić D, Fureš R, Markanović M, Škrlec I. The Impact of Oral Microbiome Dysbiosis on the Aetiology, Pathogenesis, and Development of Oral Cancer. Cancers (Basel) 2024; 16:2997. [PMID: 39272855 PMCID: PMC11394246 DOI: 10.3390/cancers16172997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common head and neck cancer. Although the oral cavity is an easily accessible area for visual examination, the OSCC is more often detected at an advanced stage. The global prevalence of OSCC is around 6%, with increasing trends posing a significant health problem due to the increase in morbidity and mortality. The oral cavity microbiome has been the target of numerous studies, with findings highlighting the significant role of dysbiosis in developing OSCC. Dysbiosis can significantly increase pathobionts (bacteria, viruses, fungi, and parasites) that trigger inflammation through their virulence and pathogenicity factors. In contrast, chronic bacterial inflammation contributes to the development of OSCC. Pathobionts also have other effects, such as the impact on the immune system, which can alter immune responses and contribute to a pro-inflammatory environment. Poor oral hygiene and carbohydrate-rich foods can also increase the risk of developing oral cancer. The risk factors and mechanisms of OSCC development are not yet fully understood and remain a frequent research topic. For this reason, this narrative review concentrates on the issue of dysbiosis as the potential cause of OSCC, as well as the underlying mechanisms involved.
Collapse
Affiliation(s)
- Jasminka Talapko
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Suzana Erić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Radiotherapy and Oncology, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tomislav Meštrović
- University Centre Varaždin, University North, 42000 Varaždin, Croatia
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA 98195, USA
- Department for Health Metrics Sciences, School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Marinka Mravak Stipetić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Martina Juzbašić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Darko Katalinić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Sanja Bekić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Family Medicine Practice, 31000 Osijek, Croatia
| | - Dora Muršić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Radiotherapy and Oncology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Josipa Flam
- Department of Radiotherapy and Oncology, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Dino Belić
- Department of Radiotherapy and Oncology, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | | | - Rajko Fureš
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Gynecology and Obstetrics, Zabok General Hospital and Croatian Veterans Hospital, 49210 Zabok, Croatia
| | - Manda Markanović
- Department of Clinical and Molecular Microbiology, University Hospital Centre Zagreb, 10000 Zagreb, Croatia
| | - Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
5
|
Leonov G, Salikhova D, Starodubova A, Vasilyev A, Makhnach O, Fatkhudinov T, Goldshtein D. Oral Microbiome Dysbiosis as a Risk Factor for Stroke: A Comprehensive Review. Microorganisms 2024; 12:1732. [PMID: 39203574 PMCID: PMC11357103 DOI: 10.3390/microorganisms12081732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Stroke represents a significant global health burden, with a substantial impact on mortality, morbidity, and long-term disability. The examination of stroke biomarkers, particularly the oral microbiome, offers a promising avenue for advancing our understanding of the factors that contribute to stroke risk and for developing strategies to mitigate that risk. This review highlights the significant correlations between oral diseases, such as periodontitis and caries, and the onset of stroke. Periodontal pathogens within the oral microbiome have been identified as a contributing factor in the exacerbation of risk factors for stroke, including obesity, dyslipidemia, atherosclerosis, hypertension, and endothelial dysfunction. The alteration of the oral microbiome may contribute to these conditions, emphasizing the vital role of oral health in the prevention of cardiovascular disease. The integration of dental and medical health practices represents a promising avenue for enhancing stroke prevention efforts and improving patient outcomes.
Collapse
Affiliation(s)
- Georgy Leonov
- Federal Research Center of Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia;
| | - Diana Salikhova
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| | - Antonina Starodubova
- Federal Research Center of Nutrition, Biotechnology and Food Safety, 109240 Moscow, Russia;
- Therapy Faculty, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Andrey Vasilyev
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
- E.V. Borovsky Institute of Dentistry, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 119991 Moscow, Russia
- Central Research Institute of Dental and Maxillofacial Surgery, 119021 Moscow, Russia
| | - Oleg Makhnach
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| | - Timur Fatkhudinov
- Institute of Molecular and Cellular Medicine, RUDN University, 117198 Moscow, Russia; (D.S.); (A.V.); (T.F.)
| | - Dmitry Goldshtein
- Research Centre for Medical Genetics, 115522 Moscow, Russia; (O.M.); (D.G.)
| |
Collapse
|
6
|
Zhang H, Lin Y, Li S, Bi J, Zeng J, Mo C, Xu S, Jia B, Lu Y, Liu C, Liu Z. Effects of bacterial extracellular vesicles derived from oral and gastrointestinal pathogens on systemic diseases. Microbiol Res 2024; 285:127788. [PMID: 38833831 DOI: 10.1016/j.micres.2024.127788] [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: 03/04/2024] [Revised: 04/18/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024]
Abstract
Oral microbiota and gastrointestinal microbiota, the two largest microbiomes in the human body, are closely correlated and frequently interact through the oral-gut axis. Recent research has focused on the roles of these microbiomes in human health and diseases. Under normal conditions, probiotics and commensal bacteria can positively impact health. However, altered physiological states may induce dysbiosis, increasing the risk of pathogen colonization. Studies suggest that oral and gastrointestinal pathogens contribute not only to localized diseases at their respective colonized sites but also to the progression of systemic diseases. However, the mechanisms by which bacteria at these local sites are involved in systemic diseases remain elusive. In response to this gap, the focus has shifted to bacterial extracellular vesicles (BEVs), which act as mediators of communication between the microbiota and the host. Numerous studies have reported the targeted delivery of bacterial pathogenic substances from the oral cavity and the gastrointestinal tract to distant organs via BEVs. These pathogenic components subsequently elicit specific cellular responses in target organs, thereby mediating the progression of systemic diseases. This review aims to elucidate the extensive microbial communication via the oral-gut axis, summarize the types and biogenesis mechanisms of BEVs, and highlight the translocation pathways of oral and gastrointestinal BEVs in vivo, as well as the impacts of pathogens-derived BEVs on systemic diseases.
Collapse
Affiliation(s)
- Han Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chengxia Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
7
|
Tang Y, Qi Y, Chen Y, Wang YQ, Zhang C, Sun Y, Huang C, Zhang XZ. Erythrocyte-Mimicking Nanovesicle Targeting Porphyromonas gingivalis for Periodontitis. ACS NANO 2024. [PMID: 39088785 DOI: 10.1021/acsnano.4c02316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
Abstract
Porphyromonas gingivalis has been demonstrated to have the strongest association with periodontitis. Within the host, P. gingivalis relies on acquiring iron and heme through the aggregation and lysis of erythrocytes, which are important factors in the growth and virulence of P. gingivalis. Additionally, the excess obtained heme is deposited on the surface of P. gingivalis, protecting the cells from oxidative damage. Based on these biological properties of the interaction between P. gingivalis and erythrocytes, this study developed an erythrocyte membrane nanovesicle loaded with gallium porphyrins to mimic erythrocytes. The nanovesicle can target and adhere with P. gingivalis precisely, being lysed and utilized by P. gingivalis as erythrocytes. Ingested gallium porphyrin replaces iron porphyrin in P. gingivalis, causing intracellular metabolic disruption. Deposited porphyrin generates a large amount of reactive oxygen species (ROS) under blue light, causing oxidative damage, and its lethality is enhanced by bacterial metabolic disruption, synergistically killing P. gingivalis. Our results demonstrate that this strategy can target and inhibit P. gingivalis, reduce its invasion of epithelial cells, and alleviate the progression of periodontitis.
Collapse
Affiliation(s)
- Ying Tang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yongdan Qi
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yu-Qiang Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Yunxia Sun
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, China
| |
Collapse
|
8
|
Nadaf R, Kumbar VM, Ghagane S. Unravelling the intricacies of Porphyromonas gingivalis: virulence factors, lifecycle dynamics and phytochemical interventions for periodontal disease management. APMIS 2024. [PMID: 39030947 DOI: 10.1111/apm.13440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024]
Abstract
Porphyromonas gingivalis is a gram-negative anaerobic bacterium recognized for its pivotal role in the pathogenesis of periodontal diseases. This review covers an overview of the virulence factors and lifecycle stages of P. gingivalis, with a specific focus on attachment and colonization, biofilm formation, growth and multiplication, dormancy survival and dissemination. Additionally, we explore the significance of inter-bacterial cross-feeding within biofilms. Furthermore, we discuss potential phytochemical-based strategies to target P. gingivalis, including the use of curcumin, apigenin, quercetin and resveratrol. Understanding the virulence factors and lifecycle stages of P. gingivalis, along with the promising phytochemical-based interventions, holds promise for advancing strategies in periodontal disease management and oral health promotion.
Collapse
Affiliation(s)
- Rubeen Nadaf
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Vijay M Kumbar
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Shridhar Ghagane
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| |
Collapse
|
9
|
Baz AA, Hao H, Lan S, Li Z, Liu S, Jin X, Chen S, Chu Y. Emerging insights into macrophage extracellular traps in bacterial infections. FASEB J 2024; 38:e23767. [PMID: 38924166 DOI: 10.1096/fj.202400739r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Macrophages possess a diverse range of well-defined capabilities and roles as phagocytes, encompassing the regulation of inflammation, facilitation of wound healing, maintenance of tissue homeostasis, and serving as a crucial element in the innate immune response against microbial pathogens. The emergence of extracellular traps is a novel strategy of defense that has been observed in several types of innate immune cells. In response to infection, macrophages are stimulated and produce macrophage extracellular traps (METs), which take the form of net-like structures, filled with strands of DNA and adorned with histones and other cellular proteins. METs not only capture and eliminate microorganisms but also play a role in the development of certain diseases such as inflammation and autoimmune disorders. The primary objective of this study is to examine the latest advancements in METs for tackling bacterial infections. We also delve into the current knowledge and tactics utilized by bacteria to elude or endure the effects of METs. Through this investigation, we hope to shed light on the intricate interactions between bacteria and the host's immune system, particularly in the context of microbicidal effector mechanisms of METs. The continued exploration of METs and their impact on host defense against various pathogens opens up new avenues for understanding and potentially manipulating the immune system's response to infections.
Collapse
Affiliation(s)
- Ahmed Adel Baz
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut, Egypt
| | - Huafang Hao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shimei Lan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Zhangcheng Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shuang Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Xiangrui Jin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Shengli Chen
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| | - Yuefeng Chu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, China
- Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Ruminant Disease Prevention and Control (West), Ministry of Agricultural and Rural Affairs, Lanzhou, China
| |
Collapse
|
10
|
Chuang WC, Yang CN, Wang HW, Lin SK, Yu CC, Syu JH, Chiang CP, Shiao YJ, Chen YW. The mechanisms of Porphyromonas gingivalis-derived outer membrane vesicles-induced neurotoxicity and microglia activation. J Dent Sci 2024; 19:1434-1442. [PMID: 39035337 PMCID: PMC11259672 DOI: 10.1016/j.jds.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 07/23/2024] Open
Abstract
Background/purpose Periodontitis is associated with various systemic diseases, potentially facilitated by the passage of Porphyromonas gingivalis outer membrane vesicles (Pg-OMVs). Several recent studies have suggested a connection between Pg-OMVs and neuroinflammation and neurodegeneration, but the precise causal relationship remains unclear. This study aimed to investigate the mechanisms underlying these associations using in vitro models. Materials and methods Isolated Pg-OMVs were characterized by morphology, size, and gingipain activity. We exposed SH-SY5Y neuroblastoma cells and BV-2 microglial cells to various concentrations of Pg-OMVs. Cell morphology, a 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, an enzyme-linked immunosorbent assay, and Western blot analysis were used to evaluate the cellular mechanism underlying Pg-OMV-induced neurotoxicity in neuronal cells and inflammatory responses in microglial cells. Results Exposure to Pg-OMVs induced neurotoxicity in SH-SY5Y cells, as evidenced by cellular shrinkage, reduced viability, activation of apoptotic pathways, and diminished neuronal differentiation markers. Gingipain inhibition mitigated these effects, suggesting that gingipain mediates Pg-OMVs-induced neurotoxicity in SH-SY5Y cells. Our research on neuroinflammation suggests that upon endocytosis of Pg-OMVs by BV-2 cells, lipopolysaccharide (LPS) can modulate the production of inducible nitric oxide synthase and tumor necrosis factor-alpha by activating pathways that involve phosphorylated AKT and the phosphorylated JNK pathway. Conclusion Our study demonstrated that following the endocytosis of Pg-OMVs, gingipain can induce neurotoxicity in SH-SY5Y cells. Furthermore, the Pg-OMVs-associated LPS can trigger neuroinflammation via AKT and JNK signaling pathways in BV-2 cells.
Collapse
Affiliation(s)
- Wei-Chun Chuang
- Department of Dentistry, Fu Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Han-Wei Wang
- Department of Dentistry, National Taiwan University Hospital, Taipei, Taiwan
| | - Sze-Kwan Lin
- Department of Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ching-Chu Yu
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Jhe-Hao Syu
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Chun-Pin Chiang
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Young-Ji Shiao
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Yi-Wen Chen
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
11
|
Lusta KA, Summerhill VI, Khotina VA, Sukhorukov VN, Glanz VY, Orekhov AN. The Role of Bacterial Extracellular Membrane Nanovesicles in Atherosclerosis: Unraveling a Potential Trigger. Curr Atheroscler Rep 2024; 26:289-304. [PMID: 38805145 DOI: 10.1007/s11883-024-01206-6] [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] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
PURPOSE OF REVIEW In this review, we explore the intriguing and evolving connections between bacterial extracellular membrane nanovesicles (BEMNs) and atherosclerosis development, highlighting the evidence on molecular mechanisms by which BEMNs can promote the athero-inflammatory process that is central to the progression of atherosclerosis. RECENT FINDINGS Atherosclerosis is a chronic inflammatory disease primarily driven by metabolic and lifestyle factors; however, some studies have suggested that bacterial infections may contribute to the development of both atherogenesis and inflammation in atherosclerotic lesions. In particular, the participation of BEMNs in atherosclerosis pathogenesis has attracted special attention. We provide some general insights into how the immune system responds to potential threats such as BEMNs during the development of atherosclerosis. A comprehensive understanding of contribution of BEMNs to atherosclerosis pathogenesis may lead to the development of targeted interventions for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Konstantin A Lusta
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Volha I Summerhill
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| | - Victoria A Khotina
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Vasily N Sukhorukov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Victor Y Glanz
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia
| | - Alexander N Orekhov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Russian National Center of Surgery, Moscow, 119991, Russia.
- Department of Research and Development, Institute for Atherosclerosis Research, Moscow, 121609, Russia.
| |
Collapse
|
12
|
Wang S, Yan T, Zhang B, Chen Y, Li Z. Porphyromonas gingivalis Vaccine: Antigens and Mucosal Adjuvants. Vaccines (Basel) 2024; 12:619. [PMID: 38932348 PMCID: PMC11209493 DOI: 10.3390/vaccines12060619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Porphyromonas gingivalis (Pg), a Gram-negative anaerobic bacterium found in dental plaque biofilm within periodontal pockets, is the primary pathogenic microorganism responsible for chronic periodontitis. Infection by Pg significantly impacts the development and progression of various diseases, underscoring the importance of eliminating this bacterium for effective clinical treatment. While antibiotics are commonly used to combat Pg, the rise of antibiotic resistance poses a challenge to complete eradication. Thus, the prevention of Pg infection is paramount. Research suggests that surface antigens of Pg, such as fimbriae, outer membrane proteins, and gingipains, can potentially be utilized as vaccine antigens to trigger protective immune responses. This article overviews these antigens, discusses advancements in mucosal adjuvants (including immunostimulant adjuvants and vaccine-delivery adjuvants), and their application in Pg vaccine development. Furthermore, the review examines the advantages and disadvantages of different immune pathways and common routes of Pg vaccine immunization. By summarizing the current landscape of Pg vaccines, addressing existing challenges, and highlighting the potential of mucosal vaccines, this review offers new insights for the advancement and clinical implementation of Pg vaccines.
Collapse
Affiliation(s)
- Shuo Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China; (S.W.); (T.Y.); (B.Z.); (Y.C.)
| | - Tong Yan
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China; (S.W.); (T.Y.); (B.Z.); (Y.C.)
| | - Bingtao Zhang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China; (S.W.); (T.Y.); (B.Z.); (Y.C.)
| | - Yixiang Chen
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China; (S.W.); (T.Y.); (B.Z.); (Y.C.)
- Henan Engineering Research Center for Key Immunological Biomaterials, Luoyang Polytechnic, Luoyang 471000, China
| | - Zhitao Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang 471003, China; (S.W.); (T.Y.); (B.Z.); (Y.C.)
| |
Collapse
|
13
|
Puca V, Marinacci B, Pellegrini B, Campanile F, Santagati M, Grande R. Biofilm and bacterial membrane vesicles: recent advances. Expert Opin Ther Pat 2024; 34:475-491. [PMID: 38578180 DOI: 10.1080/13543776.2024.2338101] [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: 11/14/2023] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
INTRODUCTION Bacterial Membrane Vesicles (MVs) play important roles in cell-to-cell communication and transport of several molecules. Such structures are essential components of Extracellular Polymeric Substances (EPS) biofilm matrix of many bacterial species displaying a structural function and a role in virulence and pathogenesis. AREAS COVERED In this review were included original articles from the last ten years by searching the keywords 'biofilm' and 'vesicles' on PUBMED and Scopus databases. The articles available in literature mainly describe a positive correlation between bacterial MVs and biofilms formation. The research on Espacenet and Google Patent databases underlines the available patents related to the application of both biofilm MVs and planktonic MVs in inhibiting biofilm formation. EXPERT OPINION This review covers and analyzes recent advances in the study of the relationship between bacterial vesicles and biofilm. The huge number of papers discussing the role of MVs confirms the interest aimed at developing new applications in the medical field. The study of the MVs composition and biogenesis may contribute to the identification of components which could be (i) the target for the development of new drugs inhibiting the biofilm establishment; (ii) candidates for the development of vaccines; (iii) biomarkers for the diagnosis of bacterial infections.
Collapse
Affiliation(s)
- Valentina Puca
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Beatrice Marinacci
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Benedetta Pellegrini
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Floriana Campanile
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences (BIOMETEC) - Microbiology Section, University of Catania, Catania, Italy
| | - Rossella Grande
- Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
14
|
Shimazu K, Ookoshi K, Fukumitsu S, Kagami H, Mitsuhata C, Nomura R, Aida K. Effects of Oleanolic Acid Derived from Wine Pomace on Periodontopathic Bacterial Growth in Healthy Individuals: A Randomized Placebo-Controlled Study. Dent J (Basel) 2024; 12:133. [PMID: 38786531 PMCID: PMC11119493 DOI: 10.3390/dj12050133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/07/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
Periodontal disease is caused by oral pathogenic bacteria and is associated with systemic disease and frailty. Therefore, its prevention is crucial in extending healthy life expectancy. This study aimed to evaluate the effect of orally administered oleanolic acid, extracted from wine pomace, on periodontopathic bacterial growth in healthy individuals. In this randomized, placebo-controlled, double-blind, parallel-group comparison study, 84 healthy adults were assigned to a placebo (n = 29), low-dose (n = 29, 9 mg oleanolic acid), or high-dose (n = 26, 27 mg oleanolic acid) groups. The number of oral bacteria in their saliva, collected before and 5 h after administration, was determined using the polymerase chain reaction-invader technique. The proportion of periodontopathic bacteria among the total oral bacteria in the saliva was calculated. Oleanolic acid significantly decreased the proportion of Porphyromonas gingivalis among the total oral bacteria in a dose-dependent manner (p = 0.005 (low-dose) and p = 0.003 (high-dose) vs. placebo, Williams' test). Moreover, high-dose oleanolic acid decreased the proportion of Tannerella forsythia (p = 0.064 vs. placebo, Williams' test). Periodontopathic bacteria are closely associated with the development and progression of periodontal disease; thus, the continuous daily intake of oleanolic acid derived from pomace may be helpful in maintaining a healthy oral microbiome by controlling the proportion of periodontopathic bacteria.
Collapse
Affiliation(s)
- Kyoko Shimazu
- Innovation Center, Central Research Laboratory, Nippn Corporation, Yokohama 243-0041, Japan; (K.O.); (S.F.); (K.A.)
| | - Kouta Ookoshi
- Innovation Center, Central Research Laboratory, Nippn Corporation, Yokohama 243-0041, Japan; (K.O.); (S.F.); (K.A.)
| | - Satoshi Fukumitsu
- Innovation Center, Central Research Laboratory, Nippn Corporation, Yokohama 243-0041, Japan; (K.O.); (S.F.); (K.A.)
| | | | - Chieko Mitsuhata
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (C.M.); (R.N.)
| | - Ryota Nomura
- Department of Pediatric Dentistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8553, Japan; (C.M.); (R.N.)
| | - Kazuhiko Aida
- Innovation Center, Central Research Laboratory, Nippn Corporation, Yokohama 243-0041, Japan; (K.O.); (S.F.); (K.A.)
| |
Collapse
|
15
|
Miranda-López DC, Pérez-Rueda E, Rojas-Vargas J, Cortez CH, Saldaña-Padilla A, Castelán-Sánchez HG, Castro-Escarpulli G. Comprehensive comparative analysis of the periodontal pathogen Porphyromonas gingivalis: exploring the pan-genome, the reconstruction of the gene regulatory network and genome-scale metabolic network. Lett Appl Microbiol 2024; 77:ovae048. [PMID: 38769598 DOI: 10.1093/lambio/ovae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/23/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Porphyromonas gingivalis is a nonmotile, obligate anaerobic, Gram-negative bacterium known for its association with periodontal disease and its involvement in systemic diseases such as atherosclerosis, cardiovascular disease, colon cancer, and Alzheimer's disease. This bacterium produces several virulence factors, including capsules, fimbriae, lipopolysaccharides, proteolytic enzymes, and hemagglutinins. A comparative genomic analysis revealed the open pangenome of P. gingivalis and identified complete type IV secretion systems in strain KCOM2805 and almost complete type VI secretion systems in strains KCOM2798 and ATCC49417, which is a new discovery as previous studies did not find the proteins involved in secretion systems IV and VI. Conservation of some virulence factors between different strains was observed, regardless of their genetic diversity and origin. In addition, we performed for the first time a reconstruction analysis of the gene regulatory network, identifying transcription factors and proteins involved in the regulatory mechanisms of bacterial pathogenesis. In particular, QseB regulates the expression of hemagglutinin and arginine deaminase, while Rex may suppress the release of gingipain through interactions with PorV and the formatum/nitrate transporter. Our study highlights the central role of conserved virulence factors and regulatory pathways, particularly QseB and Rex, in P. gingivalis and provides insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Diana C Miranda-López
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, México
| | - Ernesto Pérez-Rueda
- Unidad Académica del Estado de Yucatán, Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de México, Mérida 97357, México
| | - Jorge Rojas-Vargas
- Department of Biology, University of Western, London, Ontario N6A 3K7, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western, London, Ontario N6A 3K7, Canada
| | - Cecilia Hernández Cortez
- Laboratorio de Bioquímica Microbiana, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, México
| | - Andres Saldaña-Padilla
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, México
| | - Hugo G Castelán-Sánchez
- Programa de Investigadoras e Investigadores por México. Grupo de Genómica y Dinámica Evolutiva de Microorganismos Emergentes, Consejo Nacional de Humanidades, Ciencias y Tecnologías, Ciudad de México 03940, México
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario N6A 3K7, Canada
| | - Graciela Castro-Escarpulli
- Laboratorio de Investigación Clínica y Ambiental, Departamento de Microbiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 11340, México
| |
Collapse
|
16
|
Ye W, Wu J, Jiang Q, Su Z, Liao H, Liu Z, Tao R, Yong X. Antibacterial activity of corydalis saxicola bunting total alkaloids against Porphyromonas gingivalis in vitro. Future Microbiol 2024; 19:595-606. [PMID: 38629885 PMCID: PMC11229583 DOI: 10.2217/fmb-2023-0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/16/2024] [Indexed: 07/04/2024] Open
Abstract
Aim: To investigate the antibacterial effects of Corydalis Saxicola bunting total alkaloid (CSBTA) on Porphyromonas gingivalis. Methods: SEM, chemical staining, RT-qPCR and ELISA were used to detect effects of CSBTA on P. gingivalis. Results: CSBTA treatment caused shrinkage and rupture of P. gingivalis morphology, decreased biofilm density and live bacteria in biofilm, as well as reduced mRNA expression of virulence genes hagA, hagB, kgp, rgpA and rgpB of P. gingivalis. Furthermore, NOK cells induced by CSBTA-treated P. gingivalis exhibited lower IL-6 and TNF-α expression levels. Conclusion: CSBTA is able to kill free P. gingivalis, disrupt the biofilm and weaken the pathogenicity of P. gingivalis. It has the potential to be developed as a drug against P. gingivalis infection.
Collapse
Affiliation(s)
- Wenli Ye
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
| | - Jiaxuan Wu
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
| | - Qiaozhi Jiang
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
| | - Zhiheng Su
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Haiqing Liao
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
- Guangxi Key Laboratory of Oral & Maxillofacial Rehabilitation & Reconstruction, Nanning, China
| | - Zhenmin Liu
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
- Guangxi Key Laboratory of Oral & Maxillofacial Rehabilitation & Reconstruction, Nanning, China
| | - Renchuan Tao
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
- Guangxi Key Laboratory of Oral & Maxillofacial Rehabilitation & Reconstruction, Nanning, China
| | - Xiangzhi Yong
- Department of Periodontics & Oral Medicine, College of Stomatology, Guangxi Medical University, Nanning, China
- Guangxi Health Commission Key Laboratory of Prevention & Treatment for Oral Infectious Diseases, Nanning, China
- Guangxi Key Laboratory of Oral & Maxillofacial Rehabilitation & Reconstruction, Nanning, China
| |
Collapse
|
17
|
Irfan M, Solbiati J, Duran-Pinedo A, Rocha FG, Gibson FC, Frias-Lopez J. A Porphyromonas gingivalis hypothetical protein controlled by the type I-C CRISPR-Cas system is a novel adhesin important in virulence. mSystems 2024; 9:e0123123. [PMID: 38323815 PMCID: PMC10949514 DOI: 10.1128/msystems.01231-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 12/26/2023] [Indexed: 02/08/2024] Open
Abstract
The ability of many human pathogens to infect requires their ability to adhere to the host surfaces as a first step in the process. Porphyromonas gingivalis, a keystone oral pathogen, uses adhesins to adhere to the surface of the gingival epithelium and other members of the oral microbiome. In a previous study, we identified several proteins potentially linked to virulence whose mRNA levels are regulated by CRISPR-Cas type I-C. Among those, PGN_1547 was highly upregulated in the CRISPR-Cas 3 mutant. PGN_1547 is annotated as a hypothetical protein. Employing homology searching, our data support that PGN_1547 resembles an auto-transporter adhesin of P. gingivalis based on containing the DUF2807 domain. To begin to characterize the function of PGN_1547, we found that a deletion mutant displayed a significant decrease in virulence using a Galleria mellonela model. Furthermore, this mutant was significantly impaired in forming biofilms and attaching to the macrophage-like cell THP-1. Luminex revealed that the PGN_1547 mutant elicited a less robust cytokine and chemokine response from THP-1 cells, and TLR2 predominantly sensed that recombinant PGN_1547. Taken together, these findings broaden our understanding of the toolbox of virulence factors possessed by P. gingivalis. Importantly, PGN_1547, a hypothetical protein, has homologs in another member of the order Bacteroidales whose function is unknown, and our results could shed light on the role of this family of proteins as auto-transport adhesins in this phylogenetic group.IMPORTANCEPeriodontal diseases are among humans' most common infections, and besides their effect on the oral cavity, they have been associated with systemic inflammatory conditions. Among members of the oral microbiome implicated in the development of periodontitis, Porphyromonas gingivalis is considered a keystone pathogen. We have identified a new adhesin that acts as a virulence factor, PGN_1547, which contains the DUF2807 domain, which belongs to the putative auto-transporter adhesin, head GIN domain family. Deletion of this gene lowers the virulence of P. gingivalis and impacts the ability of P. gingivalis to form biofilm and attach to host cells. Furthermore, the broad distribution of these receptors in the order Bacteroidales suggests their importance in colonization by this important group of organisms.
Collapse
Affiliation(s)
- Muhammad Irfan
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Jose Solbiati
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Ana Duran-Pinedo
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Fernanda Godoy Rocha
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Frank C. Gibson
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Jorge Frias-Lopez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
18
|
Wenck C, Leopoldt D, Habib M, Hegermann J, Stiesch M, Doll-Nikutta K, Heisterkamp A, Torres-Mapa ML. Colorimetric detection of oral bacteria using functionalized gold nanoparticles as a plasmonic biosensor array. NANOSCALE ADVANCES 2024; 6:1447-1459. [PMID: 38419865 PMCID: PMC10898432 DOI: 10.1039/d3na00477e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024]
Abstract
Early detection of specific oral bacterial species would enable timely treatment and prevention of certain oral diseases. In this work, we investigated the sensitivity and specificity of functionalized gold nanoparticles for plasmonic sensing of oral bacteria. This approach is based on the aggregation of positively charged gold nanoparticles on the negatively charged bacteria surface and the corresponding localized surface plasmon resonance (LSPR) shift. Gold nanoparticles were synthesized in different sizes, shapes and functionalization. A biosensor array was developed consisting of spherical- and anisotropic-shaped (1-hexadecyl) trimethylammonium bromide (CTAB) and spherical mercaptoethylamine (MEA) gold nanoparticles. It was used to detect four oral bacterial species (Aggregatibacter actinomycetemcomitans, Actinomyces naeslundii, Porphyromonas gingivalis and Streptococcus oralis). The plasmonic response was measured and analysed using RGB and UV-vis absorbance values. Both methods successfully detected the individual bacterial species based on their unique responses to the biosensor array. We present an in-depth study relating the bacteria zeta potential and AuNP aggregation to plasmonic response. The sensitivity depends on multiple parameters, such as bacterial species and concentration as well as gold nanoparticle shape, concentration and functionalization.
Collapse
Affiliation(s)
- Christina Wenck
- Institute of Quantum Optics, Leibniz University Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Dorthe Leopoldt
- Institute of Quantum Optics, Leibniz University Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Mosaieb Habib
- Institute of Inorganic Chemistry, Leibniz University Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Jan Hegermann
- Research Core Unit Electron Microscopy, Institute of Functional and Applied Anatomy, Hannover Medical School Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Katharina Doll-Nikutta
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| | - Maria Leilani Torres-Mapa
- Institute of Quantum Optics, Leibniz University Hannover Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE) Germany
| |
Collapse
|
19
|
Luo S, Xu T, Zheng Q, Jiang A, Zhao J, Ying Y, Liu N, Pan Y, Zhang D. Mitochondria: An Emerging Unavoidable Link in the Pathogenesis of Periodontitis Caused by Porphyromonas gingivalis. Int J Mol Sci 2024; 25:737. [PMID: 38255811 PMCID: PMC10815845 DOI: 10.3390/ijms25020737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a key pathogen of periodontitis. Increasing evidence shows that P. gingivalis signals to mitochondria in periodontal cells, including gingival epithelial cells, gingival fibroblast cells, immune cells, etc. Mitochondrial dysfunction affects the cellular state and participates in periodontal inflammatory response through the aberrant release of mitochondrial contents. In the current review, it was summarized that P. gingivalis induced mitochondrial dysfunction by altering the mitochondrial metabolic state, unbalancing mitochondrial quality control, prompting mitochondrial reactive oxygen species (ROS) production, and regulating mitochondria-mediated apoptosis. This review outlines the impacts of P. gingivalis and its virulence factors on the mitochondrial function of periodontal cells and their role in periodontitis.
Collapse
Affiliation(s)
- Shiyin Luo
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Tong Xu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Qifan Zheng
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Aijia Jiang
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Jiahui Zhao
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yue Ying
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Nan Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang 110002, China; (S.L.); (T.X.); (Q.Z.); (A.J.); (J.Z.); (Y.Y.); (N.L.)
| | - Yaping Pan
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| | - Dongmei Zhang
- Department of Periodontics and Oral Biology, School of Stomatology, China Medical University, Shenyang 110002, China;
| |
Collapse
|
20
|
Chen Q, Shan T, Liang Y, Xu Y, Shi E, Wang Y, Li C, Wang Y, Cao M. A biomimetic phototherapeutic nanoagent based on bacterial double-layered membrane vesicles for comprehensive treatment of oral squamous cell carcinoma. J Mater Chem B 2023; 11:11265-11279. [PMID: 37974456 DOI: 10.1039/d3tb02046k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
As one of the most common malignancies, oral squamous cell carcinoma (OSCC) with high rates of invasiveness and metastasis threatens people's health worldwide, while traditional therapeutic approaches have not met the requirement of its cure. Phototherapies including photothermal therapy (PTT) and photodynamic therapy (PDT) have shown great potential for OSCC treatment due to their noninvasiveness or minimal invasiveness, high selectivity and little tolerance. However, PTT or PDT alone makes it difficult to eradicate OSCC and prevent its metastasis and recurrence. Here, double-layered membrane vesicles (DMVs) were extracted from attenuated Porphyromonas gingivalis, one of the most common pathogens inside the oral region, and served as an immune adjuvant to develop a biomimetic phototherapeutic nanoagent named PBAE/IR780@DMV for OSCC treatment via combining dual PTT/PDT and robust antitumor immunity. To obtain PBAE/IR780@DMV, poly(β-amino) ester (PBAE) was used as a carrier material to prepare the nanoparticles for loading IR780, a widely known photosensitizer possessing both PTT and PDT capabilities, followed by surface wrapping with DMVs. Upon 808 nm laser irradiation, PBAE/IR780@DMV exerted strong antitumor effects against OSCC both in vitro and in vivo, via combining PTT/PDT and specific immune responses triggered by tumor-associated antigens and DMVs. Altogether, this study provides a promising biomimetic phototherapeutic nanoagent for comprehensive treatment of OSCC.
Collapse
Affiliation(s)
- Qian Chen
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Tianhe Shan
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Yanjie Liang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Yujing Xu
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Enyu Shi
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Yue Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Changyi Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Yinsong Wang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| | - Mingxin Cao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
21
|
Wong SP, Er YX, Tan SM, Lee SC, Rajasuriar R, Lim YAL. Oral and Gut Microbiota Dysbiosis is Associated with Mucositis Severity in Autologous Hematopoietic Stem Cell Transplantation: Evidence from an Asian Population. Transplant Cell Ther 2023; 29:633.e1-633.e13. [PMID: 37422196 DOI: 10.1016/j.jtct.2023.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/17/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
Mucositis is a debilitating complication of hematopoietic stem cell transplantation (HSCT). It is unclear how changes in the composition of microbiota, which are modulated by geographical location and ethnicity, may influence immune regulation leading to the development of mucositis, and the study of both oral and gut microbiota in a single population of autologous HSCT in the Asian region is lacking. The present study aimed to characterize the oral and gut microbiota changes, and the impact on both oral and lower gastrointestinal (GI) mucositis, with associated temporal changes in a population of adult recipients of autologous HSCT. Autologous HSCT recipients age ≥18 years were recruited from Hospital Ampang, Malaysia, between April 2019 and December 2020. Mucositis assessments were conducted daily, and blood, saliva, and fecal samples were collected prior to conditioning, on day 0, and at 7 days and 6 months post-transplantation. Longitudinal differences in alpha diversity and beta diversity were determined using the Wilcoxon signed-rank test and permutational multivariate analysis of variance, respectively. Changes in relative abundances of bacteria across time points were assessed using the microbiome multivariate analysis by linear models function. The combined longitudinal effects of clinical, inflammatory, and microbiota variables on mucositis severity were measured using the generalized estimating equation. Among the 96 patients analyzed, oral mucositis and diarrhea (representing lower GI mucositis) occurred in 58.3% and 95.8%, respectively. Alpha and beta diversities were significantly different between sample types (P < .001) and across time points, with alpha diversity reaching statistical significance at day 0 in fecal samples (P < .001) and at day +7 in saliva samples (P < .001). Diversities normalized to baseline by 6 months post-transplantation. Significant microbiota, clinical, and immunologic factors were associated with increasing mucositis grades. Increasing relative abundances of saliva Paludibacter, Leuconostoc, and Proteus were associated with higher oral mucositis grades, whereas increasing relative abundances of fecal Rothia and Parabacteroides were associated with higher GI mucositis grades. Meanwhile, increasing relative abundances of saliva Lactococcus and Acidaminococcus and fecal Bifidobacterium were associated with protective effects against worsening oral and GI mucositis grades, respectively. This study provides real-world evidence and insights into the dysbiosis of the microbiota in patients exposed to conditioning regimen during HSCT. Independent of clinical and immunologic factors, we demonstrated significant associations between relative bacteria abundances with the increasing severity of oral and lower GI mucositis. Our findings offer a potential rationale to consider the inclusion of preventive and restorative measures targeting oral and lower GI dysbiosis as interventional strategies to ameliorate mucositis outcome in HSCT recipients.
Collapse
Affiliation(s)
- Shu Ping Wong
- Department of Pharmacy, Ampang Hospital, Ministry of Health, Ampang, Selangor Darul Ehsan, Malaysia
| | - Yi Xian Er
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Sen Mui Tan
- Department of Haematology, Ampang Hospital, Ministry of Health, Ampang, Selangor Darul Ehsan, Malaysia
| | - Soo Ching Lee
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Reena Rajasuriar
- Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yvonne Ai Lian Lim
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia.
| |
Collapse
|
22
|
Tang K, Wu Y, Zheng Q, Chen X. Bibliometric research on analysis of links between periodontitis and cardiovascular diseases. Front Cardiovasc Med 2023; 10:1255722. [PMID: 37745126 PMCID: PMC10512184 DOI: 10.3389/fcvm.2023.1255722] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/10/2023] [Indexed: 09/26/2023] Open
Abstract
Background Periodontitis (PD) and cardiovascular diseases (CVD) rank among the most prevalent pathologies worldwide, and their correlation has been a subject of prolonged investigation. Numerous studies suggest shared etiological factors; however, a definitive causal connection remains unestablished. The objective of this study was to employ bibliometric and visual analyses in order to comprehensively examine the overarching characteristics, focal areas of research, and prospective trends pertaining to the PD-CVD relationship. Methods We sourced articles, reviews, and online publications on PD- and CVD- research from the Web of Science Core Collection (WoSCC) spanning from January 1, 1993, to May 15, 2023. A triad of analytical tools (R-Bibliometrix, VOSviewer 1.6.19, and CiteSpace 6.2.R3) were utilized to facilitate collaboration network analysis, co-citation analysis, co-occurrence analysis, and citation burst detection. Results Out of the 1,116 publications that fulfilled the eligibility criteria in the WoSCC database, the comprehensive characteristics analysis divulged a sustained growth trend in publication frequency. In the cluster analysis of reference co-citation and keyword co-occurrence, prominent themes such as "periodontitis", "cardiovascular diseases", "inflammation", "Porphyromonas gingivalis", and "atherosclerosis" consistently emerged. Contemporary topics such as "peri-implantitis," "COVID-19", "cardiovascular risk factors," and "endocarditis" were pinpointed as burgeoning research hotspots. Conclusion Based on this bibliometric study, in the field of association studies between PD and CVD, the etiologic mechanisms of both diseases have been intensively studied in the last three decades. Periodontal pathogens might serve as potential initiating factors linking PD and CVD. Inflammation may constitute a significant etiological factor shared by both diseases. Several emerging topics, such as COVID-19 and peri-implantitis, exhibit promising potential. This exhaustive overview casts light on pivotal research arenas, augmenting the field's understanding and stimulating further scholarly investigations.
Collapse
Affiliation(s)
| | | | | | - Xuepeng Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
Bechir ES. The Clinical and Microbiological Effects of LANAP Compared to Scaling and Root Planing Alone in the Management of Periodontal Conditions. Diagnostics (Basel) 2023; 13:2450. [PMID: 37510194 PMCID: PMC10378219 DOI: 10.3390/diagnostics13142450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/30/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The purpose of this study was to evaluate the efficiency of two therapeutic procedures clinically and microbiologically in the management of periodontally affected teeth: scaling and root planing alone and the laser-assisted new attachment procedure (LANAP). Molecular biological determinations of bacterial markers through the polymerase chain reaction (real-time PCR) method with standard PET tests (species-specific DNA probes at a time) were used for the quantification of three of the most important periodontal pathogens (Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, and Treponema denticola). Both nonsurgical periodontal therapies were proven effective in patients with chronic periodontal disease; however, LANAP was associated with a greater reduction in pocket depth and improved clinical outcomes, associated with a significant decrease in the amount of Porphyromonas gingivalis. The clinical results included a decrease in periodontal pocket depth, bleeding on probing, and dental plaque, with LANAP having better overall outcomes than SRP alone. The use of Nd:YAG lasers in LANAP therapy is a safe and effective procedure that is well accepted by patients.
Collapse
Affiliation(s)
- Edwin Sever Bechir
- Faculty of Dental Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 38 Gh. Marinescu Str., 540142 Targu Mures, Romania
| |
Collapse
|
24
|
Morales-Olavarría M, Nuñez-Belmar J, González D, Vicencio E, Rivas-Pardo JA, Cortez C, Cárdenas JP. Phylogenomic analysis of the Porphyromonas gingivalis - Porphyromonas gulae duo: approaches to the origin of periodontitis. Front Microbiol 2023; 14:1226166. [PMID: 37538845 PMCID: PMC10394638 DOI: 10.3389/fmicb.2023.1226166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/04/2023] [Indexed: 08/05/2023] Open
Abstract
Porphyromonas gingivalis is an oral human pathogen associated with the onset and progression of periodontitis, a chronic immune-inflammatory disease characterized by the destruction of the teeth-supporting tissue. P. gingivalis belongs to the genus Porphyromonas, which is characterized by being composed of Gram-negative, asaccharolytic, non-spore-forming, non-motile, obligatory anaerobic species, inhabiting niches such as the oral cavity, urogenital tract, gastrointestinal tract and infected wound from different mammals including humans. Among the Porphyromonas genus, P. gingivalis stands out for its specificity in colonizing the human oral cavity and its keystone pathogen role in periodontitis pathogenesis. To understand the evolutionary process behind P. gingivalis in the context of the Pophyoromonas genus, in this study, we performed a comparative genomics study with publicly available Porphyromonas genomes, focused on four main objectives: (A) to confirm the phylogenetic position of P. gingivalis in the Porphyromonas genus by phylogenomic analysis; (B) the definition and comparison of the pangenomes of P. gingivalis and its relative P. gulae; and (C) the evaluation of the gene family gain/loss events during the divergence of P. gingivalis and P. gulae; (D) the evaluation of the evolutionary pressure (represented by the calculation of Tajima-D values and dN/dS ratios) comparing gene families of P. gingivalis and P. gulae. Our analysis found 84 high-quality assemblies representing P. gingivalis and 14 P. gulae strains (from a total of 233 Porphyromonas genomes). Phylogenomic analysis confirmed that P. gingivalis and P. gulae are highly related lineages, close to P. loveana. Both organisms harbored open pangenomes, with a strong core-to-accessory ratio for housekeeping genes and a negative ratio for unknown function genes. Our analyses also characterized the gene set differentiating P. gulae from P. gingivalis, mainly associated with unknown functions. Relevant virulence factors, such as the FimA, Mfa1, and the hemagglutinins, are conserved in P. gulae, P. gingivalis, and P. loveana, suggesting that the origin of those factors occurred previous to the P. gulae - P. gingivalis divergence. These results suggest an unexpected evolutionary relationship between the P. gulae - P. gingivalis duo and P. loveana, showing more clues about the origin of the role of those organisms in periodontitis.
Collapse
Affiliation(s)
- Mauricio Morales-Olavarría
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Josefa Nuñez-Belmar
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Dámariz González
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Emiliano Vicencio
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jaime Andres Rivas-Pardo
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| | - Cristian Cortez
- Escuela de Tecnología Médica, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Juan P. Cárdenas
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| |
Collapse
|
25
|
Boyanova L, Markovska R, Yordanov D, Gergova R, Hadzhiyski P. Anaerobes in specific infectious and noninfectious diseases: new developments. Anaerobe 2023; 81:102714. [PMID: 37349047 DOI: 10.1016/j.anaerobe.2023.102714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 01/21/2023] [Accepted: 02/03/2023] [Indexed: 06/24/2023]
Abstract
With the buildup of new research data, newer associations between anaerobic bacteria and diseases/conditions were evaluated. The aim of the mini-review was to draw attention and to encourage further multidisciplinary studies of the associations. We considered microbiome-disease correlations such as a decrease of fecal Faecalibacterium prausnitzii abundance in inflammatory bowel disease (IBD) and IBD recurrence, suggesting that F. prausnitzii could be a good biomarker for IBD. A link of subgingival Porphyromonas gingivalis with cardiovascular diseases was reported. Decreased Roseburia abundance was observed in the gut of Alzheimer's and Parkinson's disease patients. Akkermansia muciniphila was found to improve adipose/glucose metabolism, however, its intestinal abundance was observed in neurodegenerative diseases as well. Severe Clostridioides difficile infections have been reported in neonates and young children. Carcinogenic potential of anaerobes has been suggested. Fusobacterium nucleatum was implicated in the development of oral and colorectal cancer, Porphyromonas gingivalis and Tannerella forsythia were linked to esophageal cancer and Cutibacterium acnes subsp. defendens was associated with prostate cancer. However, there are some controversies about the results. In a Swedish longitudinal study, neither P. gingivalis nor T. forsythia exhibited oncogenic potential. The present data can enrich knowledge of anaerobic bacteria and their multifaceted significance for health and disease and can draw future research directions. However, more studies on large numbers of patients over prolonged periods are needed, taking into account the possible changes in the microbiota over time.
Collapse
Affiliation(s)
- Lyudmila Boyanova
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria.
| | - Rumyana Markovska
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| | - Daniel Yordanov
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| | - Raina Gergova
- Department of Medical Microbiology, Medical University of Sofia, 2 Zdrave Str., 1431, Sofia, Bulgaria
| | - Petyo Hadzhiyski
- Specialized Hospital for Active Pediatric Treatment, Medical University of Sofia, "Acad. Ivan Evstatiev Geshov" Blvd, 1606, Sofia, Bulgaria
| |
Collapse
|
26
|
Butrungrod W, Chaiyasut C, Makhamrueang N, Peerajan S, Chaiyana W, Sirilun S. Postbiotic Metabolite of Lactiplantibacillus plantarum PD18 against Periodontal Pathogens and Their Virulence Markers in Biofilm Formation. Pharmaceutics 2023; 15:pharmaceutics15051419. [PMID: 37242661 DOI: 10.3390/pharmaceutics15051419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/27/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Alternative methods to reduce infectious diseases caused by bacterial pathogens and their virulence factors, biofilm formations, have arisen to reduce the pressure on existing or currently developed disinfectants and antimicrobial agents. The current strategies for reducing the severity of periodontal pathogen-caused disease by using beneficial bacteria and their metabolites are highly desirable. Probiotic strains of lactobacilli related to foods from Thai-fermented foods were selected and their postbiotic metabolites (PM) were isolated with inhibitory activity on periodontal pathogens and their biofilm formation. The PM from Lactiplantibacillus plantarum PD18 (PD18 PM) with the highest antagonistic effect against Streptococcus mutans, Porphyromonas gingivalis, Tannerella forsythia and Prevotella loescheii was selected from 139 Lactobacillus isolates. The minimal inhibitory concentration (MIC) and minimum biofilm inhibitory concentration (MBIC) values of PD18 PM against the pathogens ranged from 1:2 to 1:4. The PD18 PM demonstrated the ability to prevent the biofilm formation of S. mutans and P. gingivalis by showing a significant reduction in viable cells, high percentages of biofilm inhibition at 92.95 and 89.68%, and the highest effective contact times at 5 and 0.5 min, respectively. L. plantarum PD18 PM showed potential as a promising natural adjunctive agent to inhibit periodontal pathogens and their biofilms.
Collapse
Affiliation(s)
- Widawal Butrungrod
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyavat Chaiyasut
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Netnapa Makhamrueang
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | | | - Wantida Chaiyana
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasithorn Sirilun
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
27
|
Lee HJ, Lee SJ, Lee SK, Choi BK, Lee DR. Magnolia kobus Extract Inhibits Periodontitis-Inducing Mediators in Porphyromonas gingivalis Lipopolysaccharide-Activated RAW 264.7 Cells. Curr Issues Mol Biol 2023; 45:538-554. [PMID: 36661522 PMCID: PMC9858207 DOI: 10.3390/cimb45010036] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Periodontitis, a disease caused by inflammation of oral bacteria, contributes to the loss of alveolar bone and destruction of connective tissues. Porphyromonas gingivalis, a Gram-negative bacterium, is known to possess important pathogenic factors for periodontal disease. In this study, we investigated the anti-periodontitis effects of Magnolia kobus extract (MKE) and magnolin as a component of Magnolia kobus (MK) in murine macrophage RAW 264.7 cells stimulated with Porphyromonas gingivalis lipopolysaccharide (LPS). Effects of MKE and magnolin on the mechanism of RAW 264.7 cellular inflammation were determined by analyzing nitric oxide (NO) production and Western blot protein expression (n = 3). MKE/magnolin inhibited NO production without affecting cell survival. MKE/magnolin treatment inhibited LPS-induced pro-inflammatory cytokines, expression levels of matrix metalloproteinases (MMPs such as MMP-1, 3, 8, 9, and 13), and protein levels of inflammatory mediators (such as TNF-α, IL-1β, and mPGES-1). MKE/magnolin also suppressed NF-κB activation by inhibiting the TLR4 signaling pathway. These findings suggest that MKE has a therapeutic effect on inflammatory periodontal disease caused by oral bacterium P. gingivalis and that magnolin is a major functional component in the anti-inflammatory effect of MKE.
Collapse
|
28
|
Huang W, Tang X, Bao J. Commentary: The oral microbiome in young women at different stages of periodontitis: Prevotella dominant in stage III periodontitis. Front Cell Infect Microbiol 2023; 13:1141285. [PMID: 36816593 PMCID: PMC9936083 DOI: 10.3389/fcimb.2023.1141285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Affiliation(s)
- Weiqiang Huang
- The Fourth Affiliated College Of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinyu Tang
- The Fourth Affiliated College Of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianfeng Bao
- Department of Hepatology, Affiliated Hangzhou Xixi Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- *Correspondence: Jianfeng Bao,
| |
Collapse
|
29
|
Liu L, Geng Y, Xiong C. Impact of Porphyromonas gingivalis-odontogenic infection on the pathogenesis of non-alcoholic fatty liver disease. Ann Med 2023; 55:2255825. [PMID: 37708866 PMCID: PMC10503456 DOI: 10.1080/07853890.2023.2255825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/15/2023] [Accepted: 09/01/2023] [Indexed: 09/16/2023] Open
Abstract
Aim: Non-alcoholic fatty liver disease is characterized by diffuse hepatic steatosis and has quickly risen to become the most prevalent chronic liver disease. Its incidence is increasing yearly, but the pathogenesis is still not fully understood. Porphyromonas gingivalis (P. gingivalis) is a major pathogen widely prevalent in periodontitis patients. Its infection has been reported to be a risk factor for developing insulin resistance, non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and metabolic syndrome. The aim of this review is to evaluate the association between P. gingivalis infection and NAFLD, identify the possible etiopathogenetic mechanisms, and raise public awareness of oral health to prevent and improve NAFLD.Methods: After searching in PubMed and Web of Science databases using 'Porphyromonas gingivalis', 'non-alcoholic fatty liver disease', and 'hepatic steatosis' as keywords, studies related were compiled and examined.Results: P. gingivalis infection is a direct risk factor for NAFLD based on clinical and basic research. Moreover, it induces systematic changes and systemic abnormalities by disrupting metabolic, inflammatory, and immunologic homeostasis.Conclusion: P. gingivalis-odontogenic infection promotes the occurrence and development of NAFLD. Further concerns are needed to emphasize oral health and maintain good oral hygiene for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Linbo Liu
- Department of Clinical Laboratory, Yulin No.2 Hospital, Yulin, Shaanxi, China
| | - Yan Geng
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chaoliang Xiong
- Department of Clinical Laboratory, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|