1
|
Lai Y, Liu S, Song C, Long T, Song L, Jiang M. An update on the role and mechanisms of periodontitis in cardiovascular diseases. Cell Signal 2025; 132:111770. [PMID: 40164419 DOI: 10.1016/j.cellsig.2025.111770] [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: 02/03/2025] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Despite extensive studies into the causes and therapies for CVDs, their incidence and prevalence continue to increase. Periodontitis is a multifactorial, chronic inflammatory disease related to systemic health. Current research suggests that periodontitis may be an unconventional risk factor for CVDs and it may increase the risk of CVDs such as atherosclerosis, coronary heart disease, myocardial infarction, hypertension, heart failure as well as cardiomyopathy. For all these reasons, it is quite plausible that prevention of periodontitis has an impact on the onset or progression of CVDs. Therefore, in this review, we investigated the association between periodontitis caused by oral microorganisms and different CVDs. In addition, we discuss the various mechanisms by which periodontitis contributes to the onset and progression of CVDs. Our review aims to raise global awareness of periodontitis, particularly its role in CVDs, provide a basis for the prevention and treatment of CVDs and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Yuping Lai
- The Huankui Academy, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Sibo Liu
- The Queen Mary school, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Chenxin Song
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China
| | - Ting Long
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; The Institute of Periodontal Disease, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China
| | - Li Song
- Center of Stomatology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; The Institute of Periodontal Disease, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China; JXHC Key Laboratory of Periodontology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 1 Minde Road, Nanchang, Jiangxi Province 330000, China.
| | - Meixiu Jiang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, 999 Xuefu Road, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
2
|
Albahri J, Allison H, Whitehead KA, Muhamadali H. The role of salivary metabolomics in chronic periodontitis: bridging oral and systemic diseases. Metabolomics 2025; 21:24. [PMID: 39920480 PMCID: PMC11805826 DOI: 10.1007/s11306-024-02220-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/31/2024] [Indexed: 02/09/2025]
Abstract
BACKGROUND Chronic periodontitis is a condition impacting approximately 50% of the world's population. As chronic periodontitis progresses, the bacteria in the oral cavity change resulting in new microbial interactions which in turn influence metabolite production. Chronic periodontitis manifests with inflammation of the periodontal tissues, which is progressively developed due to bacterial infection and prolonged bacterial interaction with the host immune response. The bi-directional relationship between periodontitis and systemic diseases has been reported in many previous studies. Traditional diagnostic methods for chronic periodontitis and systemic diseases such as chronic kidney diseases (CKD) have limitations due to their invasiveness, requiring practised individuals for sample collection, frequent blood collection, and long waiting times for the results. More rapid methods are required to detect such systemic diseases, however, the metabolic profiles of the oral cavity first need to be determined. AIM OF REVIEW In this review, we explored metabolomics studies that have investigated salivary metabolic profiles associated with chronic periodontitis and systemic illnesses including CKD, oral cancer, Alzheimer's disease, Parkinsons's disease, and diabetes to highlight the most recent methodologies that have been applied in this field. KEY SCIENTIFIC CONCEPTS OF THE REVIEW Of the rapid, high throughput techniques for metabolite profiling, Nuclear magnetic resonance (NMR) spectroscopy was the most applied technique, followed by liquid chromatography-mass spectrometry (LC-MS) and gas chromatography-mass spectrometry (GC-MS). Furthermore, Raman spectroscopy was the most used vibrational spectroscopic technique for comparison of the saliva from periodontitis patients to healthy individuals, whilst Fourier Transform Infra-Red Spectroscopy (FT-IR) was not utilised as much in this field. A recommendation for cultivating periodontal bacteria in a synthetic medium designed to replicate the conditions and composition of saliva in the oral environment is suggested to facilitate the identification of their metabolites. This approach is instrumental in assessing the potential of these metabolites as biomarkers for systemic illnesses.
Collapse
Affiliation(s)
- Jawaher Albahri
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Heather Allison
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Kathryn A Whitehead
- Microbiology at Interfaces, Department of Life Sciences, Manchester Metropolitan University, Chester St, Manchester, M1 5GD, UK.
| | - Howbeer Muhamadali
- Centre for Metabolomics Research, Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, UK.
| |
Collapse
|
3
|
Han JM, Yun I, Yang KM, Kim HS, Kim YY, Jeong W, Hong SS, Hwang I. Ethanol extract from Astilbe chinensis inflorescence suppresses inflammation in macrophages and growth of oral pathogenic bacteria. PLoS One 2024; 19:e0306543. [PMID: 38959234 PMCID: PMC11221678 DOI: 10.1371/journal.pone.0306543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
Chronic oral inflammation and biofilm-mediated infections drive diseases such as dental caries and periodontitis. This study investigated the anti-inflammatory and antibacterial potential of an ethanol extract from Astilbe chinensis inflorescence (GA-13-6) as a prominent candidate for natural complex substances (NCS) with therapeutic potential. In LPS-stimulated RAW 264.7 macrophages, GA-13-6 significantly suppressed proinflammatory mediators, including interleukin-6 (IL-6), tumor necrosis factor (TNF), and nitric oxide (NO), surpassing purified astilbin, a known bioactive compound found in A. chinensis. Furthermore, GA-13-6 downregulated the expression of cyclooxygenase-2 (COX2) and inducible nitric oxide synthase (iNOS), indicating an inhibitory effect on the inflammatory cascade. Remarkably, GA-13-6 exhibited selective antibacterial activity against Streptococcus mutans, Streptococcus sanguinis, and Porphyromonas gingivalis, key players in dental caries and periodontitis, respectively. These findings suggest that complex GA-13-6 holds the potential for the treatment or prevention of periodontal and dental diseases, as well as various other inflammation-related conditions, while averting the induction of antibiotic resistance.
Collapse
Affiliation(s)
- Jong Min Han
- DOCSmedi OralBiome Co. Ltd., Goyang-si, Republic of Korea
| | - Ina Yun
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyung Mi Yang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hye-Sung Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young-Youn Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Wonsik Jeong
- Bio Industry Department, Gyeonggido Business & Science Accelerator (GBSA), Suwon-si, Gyeonggi-do, Republic of Korea
| | - Seong Su Hong
- Bio Industry Department, Gyeonggido Business & Science Accelerator (GBSA), Suwon-si, Gyeonggi-do, Republic of Korea
| | - Inseong Hwang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
4
|
Cardona-Mendoza A, Roa Molina NS, Castillo DM, Lafaurie GI, Gualtero Escobar DF. Human Coronary Artery Endothelial Cell Response to Porphyromonas gingivalis W83 in a Collagen Three-Dimensional Culture Model. Microorganisms 2024; 12:248. [PMID: 38399652 PMCID: PMC10892777 DOI: 10.3390/microorganisms12020248] [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: 12/06/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/25/2024] Open
Abstract
P. gingivalis has been reported to be an endothelial cell inflammatory response inducer that can lead to endothelial dysfunction processes related to atherosclerosis; however, these studies have been carried out in vitro in cell culture models on two-dimensional (2D) plastic surfaces that do not simulate the natural environment where pathology develops. This work aimed to evaluate the pro-inflammatory response of human coronary artery endothelial cells (HCAECs) to P. gingivalis in a 3D cell culture model compared with a 2D cell culture. HCAECs were cultured for 7 days on type I collagen matrices in both cultures and were stimulated at an MOI of 1 or 100 with live P. gingivalis W83 for 24 h. The expression of the genes COX-2, eNOS, and vWF and the levels of the pro-inflammatory cytokines thromboxane A2 (TXA-2) and prostaglandin I2 (PGI2) were evaluated. P. gingivalis W83 in the 2D cell culture increased IL-8 levels at MOI 100 and decreased MCP-1 levels at both MOI 100 and MOI 1. In contrast, the 3D cell culture induced an increased gene expression of COX-2 at both MOIs and reduced MCP-1 levels at MOI 100, whereas the gene expression of eNOS, vWF, and IL-8 and the levels of TXA2 and PGI2 showed no significant changes. These data suggest that in the collagen 3D culture model, P. gingivalis W83 induces a weak endothelial inflammatory response.
Collapse
Affiliation(s)
- Andrés Cardona-Mendoza
- Grupo de Inmunología Celular y Molecular Universidad El Bosque-INMUBO, Vicerrectoría de Investigaciones, Facultad de Odontología, Universidad El Bosque, Bogota 11001, Colombia;
- Unidad de Investigación Básica Oral-UIBO, Vicerrectoría de Investigaciones, Facultad de Odontología, Universidad El Bosque, Bogota 11001, Colombia; (D.M.C.); (G.I.L.)
| | - Nelly Stella Roa Molina
- Centro de Investigaciones Odontológicas (CIO), Facultad de Odontología, Pontificia Universidad Javeriana, Bogota 110231, Colombia;
| | - Diana Marcela Castillo
- Unidad de Investigación Básica Oral-UIBO, Vicerrectoría de Investigaciones, Facultad de Odontología, Universidad El Bosque, Bogota 11001, Colombia; (D.M.C.); (G.I.L.)
| | - Gloria Inés Lafaurie
- Unidad de Investigación Básica Oral-UIBO, Vicerrectoría de Investigaciones, Facultad de Odontología, Universidad El Bosque, Bogota 11001, Colombia; (D.M.C.); (G.I.L.)
| | - Diego Fernando Gualtero Escobar
- Unidad de Investigación Básica Oral-UIBO, Vicerrectoría de Investigaciones, Facultad de Odontología, Universidad El Bosque, Bogota 11001, Colombia; (D.M.C.); (G.I.L.)
| |
Collapse
|
5
|
Kadatane SP, Satariano M, Massey M, Mongan K, Raina R. The Role of Inflammation in CKD. Cells 2023; 12:1581. [PMID: 37371050 DOI: 10.3390/cells12121581] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/02/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic kidney disease (CKD) affects many adults worldwide. Persistent low-grade inflammation is a substantial factor in its development and progression and has correlated with increased mortality and cardiovascular problems. This low-grade inflammation is a product of dysregulation of the normal balance between pro- and anti-inflammatory markers. Various factors such as increased innate immune system activation, reactive oxygen species production, periodontal disease, dysregulation of anti-inflammatory systems and intestinal dysbiosis result in the dysregulation of this balance. Furthermore, this low-grade inflammation has down-effects such as hypertension, renal fibrosis and acceleration of renal function decline. Moreover, low-grade inflammation over time has been linked to malignancy in CKD. As CKD progresses, many patients require dialysis, which has a negative bidirectional relationship with persistent inflammation. Treatment options for inflammation in CKD are vast, including cytokine inhibitors, statins and diets. However, more research is needed to create a standardized management plan. In this review, we will examine the normal physiology of the kidney and its relationship with the immune system. We will then delve into the pathology behind persistent inflammation, the various causes of inflammation, the downstream effects of inflammation, dialysis and potential treatments for inflammation in CKD.
Collapse
Affiliation(s)
| | - Matthew Satariano
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Michael Massey
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Kai Mongan
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Rupesh Raina
- Akron Nephrology Associates/Cleveland Clinic Akron General Medical Center, Akron, OH 44302, USA
- Department of Nephrology, Akron Children's Hospital, Akron, OH 44308, USA
| |
Collapse
|
6
|
Zhong M, Huang J, Wu Z, Chan KG, Wang L, Li J, Lee LH, Law JWF. Potential Roles of Selectins in Periodontal Diseases and Associated Systemic Diseases: Could They Be Targets for Immunotherapy? Int J Mol Sci 2022; 23:14280. [PMID: 36430760 PMCID: PMC9698067 DOI: 10.3390/ijms232214280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Periodontal diseases are predisposing factors to the development of many systemic disorders, which is often initiated via leukocyte infiltration and vascular inflammation. These diseases could significantly affect human health and quality of life. Hence, it is vital to explore effective therapies to prevent disease progression. Periodontitis, which is characterized by gingival bleeding, disruption of the gingival capillary's integrity, and irreversible destruction of the periodontal supporting bone, appears to be caused by overexpression of selectins in periodontal tissues. Selectins (P-, L-, and E-selectins) are vital members of adhesion molecules regulating inflammatory and immune responses. They are mainly located in platelets, leukocytes, and endothelial cells. Furthermore, selectins are involved in the immunopathogenesis of vascular inflammatory diseases, such as cardiovascular disease, diabetes, cancers, and so on, by mediating leukocyte recruitment, platelet activation, and alteration of endothelial barrier permeability. Therefore, selectins could be new immunotherapeutic targets for periodontal disorders and their associated systemic diseases since they play a crucial role in immune regulation and endothelium dysfunction. However, the research on selectins and their association with periodontal and systemic diseases remains limited. This review aims to discuss the critical roles of selectins in periodontitis and associated systemic disorders and highlights the potential of selectins as therapeutic targets.
Collapse
Affiliation(s)
- Mei Zhong
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Jiangyong Huang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Zhe Wu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Lijing Wang
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiang Li
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Jodi Woan-Fei Law
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
7
|
Park DY, Park JY, Lee D, Hwang I, Kim HS. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells 2022; 11:1079. [PMID: 35406643 PMCID: PMC8997512 DOI: 10.3390/cells11071079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
The oral cavity is the gateway for microorganisms into your body where they disseminate not only to the directly connected respiratory and digestive tracts but also to the many remote organs. Oral microbiota, travelling to the end of the intestine and circulating in our bodies through blood vessels, not only affect a gut microbiome profile but also lead to many systemic diseases. By gathering information accumulated from the era of focal infection theory to the age of revolution in microbiome research, we propose a pivotal role of "leaky gum", as an analogy of "leaky gut", to underscore the importance of the oral cavity in systemic health. The oral cavity has unique structures, the gingival sulcus (GS) and the junctional epithelium (JE) below the GS, which are rarely found anywhere else in our body. The JE is attached to the tooth enamel and cementum by hemidesmosome (HD), which is structurally weaker than desmosome and is, thus, vulnerable to microbial infiltration. In the GS, microbial biofilms can build up for life, unlike the biofilms on the skin and intestinal mucosa that fall off by the natural process. Thus, we emphasize that the GS and the JE are the weakest leaky point for microbes to invade the human body, making the leaky gum just as important as, or even more important than, the leaky gut.
Collapse
Affiliation(s)
- Do-Young Park
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Jin Young Park
- Department of Gastrointestinal Endoscopy, Apple Tree Healthcare Center, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| | - Inseong Hwang
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Hye-Sung Kim
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| |
Collapse
|
8
|
Li L, Zhang YL, Liu XY, Meng X, Zhao RQ, Ou LL, Li BZ, Xing T. Periodontitis Exacerbates and Promotes the Progression of Chronic Kidney Disease Through Oral Flora, Cytokines, and Oxidative Stress. Front Microbiol 2021; 12:656372. [PMID: 34211440 PMCID: PMC8238692 DOI: 10.3389/fmicb.2021.656372] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/01/2021] [Indexed: 12/25/2022] Open
Abstract
Periodontitis is a type of systemic immune inflammation that is caused by the complex infection of a variety of microorganisms in the subgingival plaque and the imbalance of the microbial ecological environment in the mouth. Periodontitis and chronic kidney disease (CKD) share many risk factors, such as obesity, smoking, and age. A growing body of data supports a strong correlation between periodontitis and kidney disease. Evidence supports the role of periodontal inflammation and elevated serum inflammatory mediators in renal atherosclerosis, renal deterioration, and end-stage renal disease (ESRD) development. Periodontitis is a risk factor for kidney disease. However, to our knowledge, there are few studies detailing the possible link between periodontitis and CKD. This review summarizes the possible mechanisms underlying periodontitis and CKD. More importantly, it highlights novel and potential pathogenic factors for CKD, including bacteria, pro-inflammatory mediators and oxidative stress. However, most research on the relationship between periodontitis and systemic disease has not determined causality, and these diseases are largely linked by bidirectional associations. Future research will focus on exploring these links to contribute to new treatments for CKD.
Collapse
Affiliation(s)
- Ling Li
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Ya-Li Zhang
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Xing-Yu Liu
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Xiang Meng
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
| | - Rong-Quan Zhao
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Lin-Lin Ou
- School of Stomatology, Anhui Medical University, Hefei, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Tian Xing
- School of Stomatology, Anhui Medical University, Hefei, China
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, China
| |
Collapse
|
9
|
Sargolzaie N, Naghibi N, Khajavi A, Moeintaghavi A, Tashnizi MA, Ghazvini K, Shiezadeh F. Quantitative Detection of Periodontopathogenic Bacteria in Atherosclerotic Plaques from Coronary Arteries by Real-Time PCR. Open Dent J 2020. [DOI: 10.2174/1874210602014010724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Objectives:
Epidemiologic studies have suggested periodontitis as a risk factor for Coronary Artery Diseas (CAD). Detection of periopathogens in atheromatous plaque provides some evidence for the causal relationship between these two conditions. The aim of this study was to determine the presence and quantity of periopathogens in coronary atherosclerotic plaques in patients undergoing Coronary Artery Bypass Graft (CABG) surgery.
Methods:
20 patients who were candidates for endarterectomy were enrolled in this study for the periodontal examination. Subgingival and coronary atherosclerotic plaque samples were then collected. Thereafter, quantitative detection of Aggregatibacter actinomycetemcomitans (A.a), Porphyromonas gingivali (P.g), and all bacteria detected by Real-Time PCR (RT-PCR) were measured. The correlation analysis was also used to evaluate the relationship between quantities of periopathogens in atherosclerotic and subgingival plaque samples.
Results:
A.a was detected in 13 patients (65%) with subgingival plaques and 4 patients (20%) with atherosclerotic plaques. In addition, P.g was found in 15 patients (75%) with subgingival and 10 patients (50%) with atherosclerotic plaques. A.a represented means of 2.7% and 10.04% of detected bacteria in both atherosclerotic and subgingival plaque samples, respectively. The mean of quantity of P.g was 10.85% and 12.87% of the detected bacteria obtained from atherosclerotic and subginigival samples, respectively. Correlation analysis showed a significant correlation between the quantities of A.a in the atherosclerotic and subgingival plaques, but such a significant relationship was not found for P.g.
Conclusion:
This study confirmed the detection of A.a and P.g in atheromatous plaque. The quantitative data suggested that periopathogens comprise a significant proportion of atherosclerotic plaque microbiome, which may consequently contribute to the development of CAD.
Collapse
|
10
|
Repeated Porphyromonas gingivalis W83 exposure leads to release pro-inflammatory cytokynes and angiotensin II in coronary artery endothelial cells. Sci Rep 2019; 9:19379. [PMID: 31852912 PMCID: PMC6920421 DOI: 10.1038/s41598-019-54259-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
The role of Porphyromonas gingivalis (P. gingivalis) or its virulence factors, including lipopolysaccharide (LPS) not only has been related with periodontitis but also with endothelial dysfunction, a key mechanism involved in the genesis of atherosclerosis and hypertension that involving systemic inflammatory markers as angiotensin II (Ang II) and cytokines. This study compares the effect of repeated and unique exposures of P. gingivalis W83 LPS and live bacteria on the production and expression of inflammatory mediators and vasoconstrictor molecules with Ang II. Human coronary artery endothelial cells (HCAEC) were stimulated with purified LPS of P. gingivalis (1.0, 3.5 or 7.0 μg/mL) or serial dilutions of live bacteria (MOI 1: 100 - 1:0,1) at a single or repeated exposure for a time of 24 h. mRNA expression levels of AGTR1, AGTR2, IL-8, IL-1β and MCP-1 were determined by RT-qPCR, and IL-6, MCP-1, IL-8, IL-1β and GM-CSF levels were measured by flow cytometry, ELISA determined Ang II levels. Live bacteria in a single dose increased mRNA levels of AGTR1, and repeated doses increased mRNA levels of IL-8 and IL-1β (p < 0.05). Repeated exposure of live-P. gingivalis induced significant production IL-6, MCP-1 and GM-CSF (p < 0.05). Moreover, these MCP-1, IL-6 and GM-CSF levels were greater than in cells treated with single exposure (p < 0.05), The expression of AGTR1 and production of Ang II induced by live-P. gingivalis W83 showed a vasomotor effect of whole bacteria in HCAEC more than LPS. In conclusion, the findings of this study suggest that repeated exposure of P. gingivalis in HCAEC induces the activation of proinflammatory and vasoconstrictor molecules that lead to endothelial dysfunction being a key mechanism of the onset and progression of arterial hypertension and atherosclerosis.
Collapse
|
11
|
The Distinct Immune-Stimulatory Capacities of Porphyromonas gingivalis Strains 381 and ATCC 33277 Are Determined by the fimB Allele and Gingipain Activity. Infect Immun 2019; 87:IAI.00319-19. [PMID: 31570556 DOI: 10.1128/iai.00319-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/19/2019] [Indexed: 11/20/2022] Open
Abstract
The Porphyromonas gingivalis strain ATCC 33277 (33277) and 381 genomes are nearly identical. However, strain 33277 displays a significantly diminished capacity to stimulate host cell Toll-like receptor 2 (TLR2)-dependent signaling and interleukin-1β (IL-1β) production relative to 381, suggesting that there are strain-specific differences in one or more bacterial immune-modulatory factors. Genomic sequencing identified a single nucleotide polymorphism in the 33277 fimB allele (A→T), creating a premature stop codon in the 33277 fimB open reading frame relative to the 381 fimB allele. Gene exchange experiments established that the 33277 fimB allele reduces the immune-stimulatory capacity of this strain. Transcriptome comparisons revealed that multiple genes related to carboxy-terminal domain (CTD) family proteins, including the gingipains, were upregulated in 33277 relative to 381. A gingipain substrate degradation assay demonstrated that cell surface gingipain activity is higher in 33277, and an isogenic mutant strain deficient for the gingipains exhibited an increased ability to induce TLR2 signaling and IL-1β production. Furthermore, 33277 and 381 mutant strains lacking CTD cell surface proteins were more immune-stimulatory than the parental wild-type strains, consistent with an immune-suppressive role for the gingipains. Our data show that the combination of an intact fimB allele and limited cell surface gingipain activity in P. gingivalis 381 renders this strain more immune-stimulatory. Conversely, a defective fimB allele and high-level cell surface gingipain activity reduce the capacity of P. gingivalis 33277 to stimulate host cell innate immune responses. In summary, genomic and transcriptomic comparisons identified key virulence characteristics that confer divergent host cell innate immune responses to these highly related P. gingivalis strains.
Collapse
|
12
|
Macrophage Migration Inhibitory Factor Levels in Gingival Crevicular Fluid, Saliva, and Serum of Chronic Periodontitis Patients. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7850392. [PMID: 30868074 PMCID: PMC6379857 DOI: 10.1155/2019/7850392] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 02/07/2023]
Abstract
Chronic periodontitis (CP) is an infection that affects the teeth supporting structure. Macrophage migration inhibitory factor (MIF) is an important effector cytokine of the innate immune system. Due to its functional characteristics, MIF may be involved in the immunopathology of CP. The aim of the present study was to evaluate MIF levels in gingival crevicular fluid (GCF), saliva, and serum of CP patients. A cross-sectional study was conducted on 60 subjects divided into two groups: subjects with CP (n= 30) and periodontally healthy subjects without CP (n=30). MIF was quantified in GCF, saliva, and serum of all participants by enzyme-linked immunosorbent assay. MIF concentrations were higher in GCF, saliva, and serum in the group with CP compared with the group without CP and a higher MIF concentration was observed in GCF (p=0.001) and saliva (p=0.009) in the group with CP. MIF intragroup comparisons between fluids demonstrated significant high levels of MIF in saliva compared with GCF and serum in both study groups (p<0.05). A positive correlation was found between clinical signs and MIF concentration in GCF (p<0.05). There is an association between the MIF and the clinical signs of the disease. Therefore, MIF could have an important role in the pathology and progression of CP.
Collapse
|
13
|
Mittal R, Jhaveri VM, Kay SIS, Blackwelder P, Patel K. Pseudomonas aeruginosa Invades Human Aortic Endothelial Cells and Induces Cell Damage in vitro. Cardiovasc Hematol Disord Drug Targets 2018; 19:45-50. [PMID: 29886837 DOI: 10.2174/1871529x18666180611094928] [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: 08/08/2017] [Revised: 12/16/2017] [Accepted: 03/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cardiovascular diseases such as endocarditis are the second most common cause of death worldwide. Infective Endocarditis (IE) is the most severe infection of the heart associated with significant mortality and morbidity. The binding and invasion of Human Aortic Endothelial Cells (HAECs) by pathogenic microbes can play an important role in the pathogenesis of IE. OBJECTIVE Pseudomonas aeruginosa is an emerging pathogen that has been associated with IE. However, it is not known whether P. aeruginosa can bind and interact with HAECs. The aim of this study was to determine whether P. aeruginosa can bind and colonize HAECs. METHODS The invasion of HAECs by P. aeruginosa was assessed by gentamicin protection assay. Cytokine levels were determined by enzyme-linked Immunosorbent Assay (ELISA) kits. Cell damage was determined by Lactate Dehydrogenase (LDH) assay. RESULTS P. aeruginosa can bind and invade HAECs. Infection of HAECs with P. aeruginosa induces TNF-α IL-1β, IL-6 and IL-8 cytokine production leading to the generation of inflammatory milieu that can cause tissue damage as observed in human clinical cases of IE. We also observed that P. aeruginosa induces cell damage in HAECs. CONCLUSION In this study, we demonstrate for first time that P. aeruginosa can invade and survive inside HAECs. This cell culture model can be of immense importance to determine the efficacy of drug targets against IE.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, FL-33136, United States
| | - Vasanti M Jhaveri
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, FL-33136, United States
| | - Sae-In Samantha Kay
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, FL-33136, United States
| | - Patricia Blackwelder
- Center for Advanced Microscopy, University of Miami, Coral Gables, Florida, FT, United States.,RSMAS, University of Miami, Key Biscayne, Florida, FL-33136, United States
| | - Kunal Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, FL-33136, United States
| |
Collapse
|
14
|
Pan C, Liu J, Wang H, Song J, Tan L, Zhao H. Porphyromonas gingivalis can invade periodontal ligament stem cells. BMC Microbiol 2017; 17:38. [PMID: 28212613 PMCID: PMC5316216 DOI: 10.1186/s12866-017-0950-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
Background Porphyromonas gingivalis is strongly associated with the development, progression, severity and recurrence of periodontitis. Periodontal ligament stem cells (PDLSCs) play an important role in the maintenance of periodontal tissue self-renewal and repair. The purpose of this study was to investigate the ability of P. gingivalis to infect PDLSCs using an in vitro monolayer model. Methods We separated and cultured primary PDLSCs using the tissue block with limiting dilution method. The efficiency of P. gingivalis (ATCC 33277) infection of PDLSCs was measured using agar plate culture and quantitative polymerase chain reaction (q-PCR) methods. PDLSCs infected with P. gingivalis were also observed by transmission electron microscopy. Results We assessed stem cell properties including cell morphology, clone formation, growth activity, cell surface antigens and multiple differentiation capacity. The infection rates of P. gingivalis in PDLSC at MOIs of 50, 100, 200, and 500 were 5.83%, 8.12%, 7.77% and 7.53% according to the agar plate culture method. By q-PCR, the efficiencies of P. gingivalis infection of PDLSCs at MOIs of 50, 100, 200, and 500 were 6.74%, 10.56%, 10.36% and 9.78%, respectively. Overall, the infection efficiency based on q-PCR was higher than that according to agar plate culture. Using transmission electron microscopy, we verified that P. gingivalis (ATCC 33277) could infect and invade PDLSCs after 2 h of incubation, and endocytic vacuoles were not found surrounding the internalized bacteria. Conclusions In conclusion, our data demonstrate that P. gingivalis can invade PDLSCs. Electronic supplementary material The online version of this article (doi:10.1186/s12866-017-0950-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chunling Pan
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China.
| | - Junchao Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China
| | - Hongyan Wang
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China
| | - Jia Song
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China
| | - Lisi Tan
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China
| | - Haijiao Zhao
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, 110002, China
| |
Collapse
|
15
|
Porphyromonas gingivalis oral infection promote T helper 17/Treg imbalance in the development of atherosclerosis. J Dent Sci 2016; 12:60-69. [PMID: 30895025 PMCID: PMC6395245 DOI: 10.1016/j.jds.2016.10.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 10/13/2016] [Indexed: 02/08/2023] Open
Abstract
Background/purpose Increasing studies have indicated the involvement of Porphyromonas gingivalis in atherosclerosis. T helper 17 (Th17)/Treg balance is critical during atherosclerosis. However, whether P. gingivalis oral infection is associated with Th17/Treg imbalance is unclear. The aim of the present study was to investigate the effect of P. gingivalis on Th17/Treg balance during atherosclerosis. Materials and methods ApoE–/– and C57BL/6 mice were inoculated orally with P. gingivalis ATCC 33277 for 9 weeks. The alveolar bone loss was assessed by microcomputerized tomography. The area of atherosclerosis plaque was identified by oil red O staining. Plaque stability was analyzed by CD68 and αSMA immunohistochemistry staining and Masson staining. The frequency of Th17 and Treg in spleen was detected by flow cytometry. The mRNA expression of Th17- and Treg-related factors was determined by quantitative polymerase chain reaction. Interleukin (IL)-6, a critical factor in modulating T-cell differentiation, was determined from spleen cells and mouse dendritic cells by enzyme-linked immunosorbent assay. Results Long-term P. gingivalis oral infection induced alveolar bone resorption. In ApoE–/– mice, P. gingivalis enhanced atherosclerotic lesion formation and plaque instability accompanied with a decreased Treg frequency and an increased Th17 cell frequency. In addition, mRNA expression of retinoic acid receptor-related orphan receptor γt and IL-17 was increased, and that of transforming growth factor (TGF) β and IL-10 was decreased in P. gingivalis-infected ApoE–/– mice. Furthermore, secretion of IL-6 was elevated in the spleen of P. gingivalis-infected ApoE–/– mice, as well as in mouse dendritic cells after P. gingivalis infection. Conclusion P. gingivalis oral infection may promote Th17/Treg imbalance by influencing T-cell differentiation during the process of atherosclerosis, with a larger lesion area and decreasing plaque instability.
Collapse
|
16
|
Ho MH, Guo ZM, Chunga J, Goodwin JS, Xie H. Characterization of Innate Immune Responses of Human Endothelial Cells Induced by Porphyromonas gingivalis and Their Derived Outer Membrane Vesicles. Front Cell Infect Microbiol 2016; 6:139. [PMID: 27826542 PMCID: PMC5078693 DOI: 10.3389/fcimb.2016.00139] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/10/2016] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory disease of the blood vessels, is one of the most common causes of morbidity and mortality world-wide. Involvement of Porphyromonas gingivalis in atherosclerosis is supported by observations from epidemiological, clinical, immunological, and molecular studies. Previously we reported that P. gingivalis vesicles have a much higher invasive efficiency than their originating cells. Here, we further compare the role of P. gingivalis cells and their vesicles in expression of chemoattractant proteins including CXCL1, CXCL2, and CXCL8, and adhesive molecules such as E-selectin in human umbilical vein endothelial cells (HUVECs). Both P. gingivalis 33277 cells and vesicles were able to up-regulate expression of these molecules, while the vesicles acted as more potent inducers of the inflammatory response associated with the development of atherosclerosis, consequently resulting in significant monocyte adhesion to a monolayer of HUVECs. Interestingly, we found that elevated expression of CXCL8 and E-selectin in endothelial cells induced by P. gingivalis correlated with the invasive ability of P. gingivalis cells and vesicles. Non-invasive bacterial cells and vesicles had no effect on expression of these genes. This study highlights the potential risk of P. gingivalis cells and vesicles in initiation of atherosclerosis and provides a potential target for the development of novel therapeutics against bacteria-associated atherosclerosis.
Collapse
Affiliation(s)
- Meng-Hsuan Ho
- Oral Biology, School of Dentistry, Meharry Medical CollegeNashville, TN, USA
| | - Zhong-Mao Guo
- Department of Physiology, Meharry Medical CollegeNashville, TN, USA
| | | | - J. Shawn Goodwin
- Department of Biochemistry and Cancer Biology, Meharry Medical CollegeNashville, TN, USA
| | - Hua Xie
- Oral Biology, School of Dentistry, Meharry Medical CollegeNashville, TN, USA
| |
Collapse
|
17
|
Slocum C, Kramer C, Genco CA. Immune dysregulation mediated by the oral microbiome: potential link to chronic inflammation and atherosclerosis. J Intern Med 2016; 280:114-28. [PMID: 26791914 DOI: 10.1111/joim.12476] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cardiovascular disease is an inflammatory disorder characterized by the progressive formation of plaque in coronary arteries, termed atherosclerosis. It is a multifactorial disease that is one of the leading causes of death worldwide. Although a number of risk factors have been associated with disease progression, the underlying inflammatory mechanisms contributing to atherosclerosis remain to be fully delineated. Within the last decade, the potential role for infection in inflammatory plaque progression has received considerable interest. Microbial pathogens associated with periodontal disease have been of particular interest due to the high levels of bacteremia that are observed after routine dental procedures and every day oral activities, such as tooth brushing. Here, we explore the potential mechanisms that may explain how periodontal pathogens either directly or indirectly elicit immune dysregulation and consequently progressive inflammation manifested as atherosclerosis. Periodontal pathogens have been shown to contribute directly to atherosclerosis by disrupting endothelial cell function, one of the earliest indicators of cardiovascular disease. Oral infection is thought to indirectly induce elevated production of inflammatory mediators in the systemic circulation. Recently, a number of studies have been conducted focusing on how disruption of the gut microbiome influences the systemic production of proinflammatory cytokines and consequently exacerbation of inflammatory diseases such as atherosclerosis. It is clear that the immune mechanisms leading to atherosclerotic plaque progression, by oral infection, are complex. Understanding the immune pathways leading to disease progression is essential for the future development of anti-inflammatory therapies for this chronic disease.
Collapse
Affiliation(s)
| | - C Kramer
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| | - C A Genco
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
18
|
Reyes L, Herrera D, Kozarov E, Roldán S, Progulske-Fox A. Periodontal bacterial invasion and infection: contribution to atherosclerotic pathology. J Clin Periodontol 2016; 40 Suppl 14:S30-50. [PMID: 23627333 DOI: 10.1111/jcpe.12079] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2012] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The objective of this review was to perform a systematic evaluation of the literature reporting current scientific evidence for periodontal bacteria as contributors to atherosclerosis. METHODS Literature from epidemiological, clinical and experimental studies concerning periodontal bacteria and atherosclerosis were reviewed. Gathered data were categorized into seven "proofs" of evidence that periodontal bacteria: 1) disseminate from the oral cavity and reach systemic vascular tissues; 2) can be found in the affected tissues; 3) live within the affected site; 4) invade affected cell types in vitro; 5) induce atherosclerosis in animal models of disease; 6) non-invasive mutants of periodontal bacteria cause significantly reduced pathology in vitro and in vivo; and 7) periodontal isolates from human atheromas can cause disease in animal models of infection. RESULTS Substantial evidence for proofs 1 to 6 was found. However, proof 7 has not yet been fulfilled. CONCLUSIONS Despite the lack of evidence that periodontal bacteria obtained from human atheromas can cause atherosclerosis in animal models of infection, attainment of proofs 1 to 6 provides support that periodontal pathogens can contribute to atherosclerosis.
Collapse
Affiliation(s)
- Leticia Reyes
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, FL 32610-0424, USA
| | | | | | | | | |
Collapse
|
19
|
Reyes L, Herrera D, Kozarov E, Roldá S, Progulske-Fox A. Periodontal bacterial invasion and infection: contribution to atherosclerotic pathology. J Periodontol 2016; 84:S30-50. [PMID: 23631583 DOI: 10.1902/jop.2013.1340012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The objective of this review was to perform a systematic evaluation of the literature reporting current scientific evidence for periodontal bacteria as contributors to atherosclerosis. METHODS Literature from epidemiological, clinical and experimental studies concerning periodontal bacteria and atherosclerosis were reviewed. Gathered data were categorized into seven "proofs" of evidence that periodontal bacteria: 1) disseminate from the oral cavity and reach systemic vascular tissues; 2) can be found in the affected tissues; 3) live within the affected site; 4) invade affected cell types in vitro; 5) induce atherosclerosis in animal models of disease; 6) non-invasive mutants of periodontal bacteria cause significantly reduced pathology in vitro and in vivo; and 7) periodontal isolates from human atheromas can cause disease in animal models of infection. RESULTS Substantial evidence for proofs 1 to 6 was found. However, proof 7 has not yet been fulfilled. CONCLUSIONS Despite the lack of evidence that periodontal bacteria obtained from human atheromas can cause atherosclerosis in animal models of infection, attainment proofs 1 to 6 provides support that periodontal pathogens can contribute to atherosclerosis.
Collapse
Affiliation(s)
- Leticia Reyes
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, FL, USA
| | | | | | | | | |
Collapse
|
20
|
Links between atherosclerotic and periodontal disease. Exp Mol Pathol 2016; 100:220-35. [DOI: 10.1016/j.yexmp.2016.01.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/08/2016] [Indexed: 02/06/2023]
|
21
|
Hasturk H, Kantarci A. Activation and resolution of periodontal inflammation and its systemic impact. Periodontol 2000 2015; 69:255-73. [PMID: 26252412 PMCID: PMC4530469 DOI: 10.1111/prd.12105] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2015] [Indexed: 02/06/2023]
Abstract
Inflammation is a highly organized event impacting upon organs, tissues and biological systems. Periodontal diseases are characterized by dysregulation or dysfunction of resolution pathways of inflammation that results in failure to heal and in a dominant chronic, progressive, destructive and predominantly unresolved inflammation. The biological consequences of inflammatory processes may be independent of the etiological agents, such as trauma, microbial organisms and stress. The impact of the inflammatory pathological process depends upon the tissues or organ system affected. Whilst mediators are similar, there is tissue specificity for the inflammatory events. It is plausible that inflammatory processes in one organ could directly lead to pathologies in another organ or tissue. Communication between distant parts of the body and their inflammatory status is also mediated by common signaling mechanisms mediated via cells and soluble mediators. This review focuses on periodontal inflammation, its systemic associations and advances in therapeutic approaches based on mediators acting through orchestration of natural pathways to resolution of inflammation. We also discuss a new treatment concept in which natural pathways of resolution of periodontal inflammation can be used to limit systemic inflammation and promote healing and regeneration.
Collapse
Affiliation(s)
- Hatice Hasturk
- The Forsyth Institute, Department of Applied Oral Sciences, Center for Periodontology, Cambridge, MA 02142, USA. Phone: 617-892-8499; Fax: 617-892-8505
| | - Alpdogan Kantarci
- The Forsyth Institute, Department of Applied Oral Sciences, Center for Periodontology, Cambridge, MA 02142, USA. Phone: 617-892-8530
| |
Collapse
|
22
|
Olsen I, Progulske-Fox A. Invasion of Porphyromonas gingivalis strains into vascular cells and tissue. J Oral Microbiol 2015; 7:28788. [PMID: 26329158 PMCID: PMC4557090 DOI: 10.3402/jom.v7.28788] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/06/2015] [Accepted: 08/07/2015] [Indexed: 01/06/2023] Open
Abstract
Porphyromonas gingivalis is considered a major pathogen in adult periodontitis and is also associated with multiple systemic diseases, for example, cardiovascular diseases. One of its most important virulence factors is invasion of host cells. The invasion process includes attachment, entry/internalization, trafficking, persistence, and exit. The present review discusses these processes related to P. gingivalis in cardiovascular cells and tissue. Although most P. gingivalis strains invade, the invasion capacity of strains and the mechanisms of invasion including intracellular trafficking among them differ. This is consistent with the fact that there are significant differences in the pathogenicity of P. gingivalis strains. P. gingivalis invasion mechanisms are also dependent on types of host cells. Although much is known about the invasion process of P. gingivalis, we still have little knowledge of its exit mechanisms. Nevertheless, it is intriguing that P. gingivalis can remain viable in human cardiovascular cells and atherosclerotic plaque and later exit and re-enter previously uninfected host cells.
Collapse
Affiliation(s)
- Ingar Olsen
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway;
| | - Ann Progulske-Fox
- Department of Oral Biology and Center for Molecular Microbiology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
23
|
Salvatore P, Zullo A, Sommese L, Colicchio R, Picascia A, Schiano C, Mancini FP, Napoli C. Infections and cardiovascular disease: is Bartonella henselae contributing to this matter? J Med Microbiol 2015; 64:799-809. [PMID: 26066633 DOI: 10.1099/jmm.0.000099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease is still the major cause of death worldwide despite the remarkable progress in its prevention and treatment. Endothelial progenitor cells (EPCs) have recently emerged as key players of vascular repair and regenerative medicine applied to cardiovascular disease. A large amount of effort has been put into discovering the factors that could aid or impair the number and function of EPCs, and also into characterizing these cells at the molecular level in order to facilitate their therapeutic applications in vascular disease. Interestingly, the major cardiovascular risk factors have been associated with reduced number and function of EPCs. The bacterial contribution to cardiovascular disease represents a long-standing controversy. The discovery that Bartonella henselae can infect and damage EPCs revitalizes the enduring debate about the microbiological contribution to atherosclerosis, thus allowing the hypothesis that this infection could impair the cardiovascular regenerative potential and increase the risk for cardiovascular disease. In this review, we summarize the rationale suggesting that Bartonella henselae could favour atherogenesis by infecting and damaging EPCs, thus reducing their vascular repair potential. These mechanisms suggest a novel link between communicable and non-communicable human diseases, and put forward the possibility that Bartonella henselae could enhance the susceptibility and worsen the prognosis in cardiovascular disease.
Collapse
Affiliation(s)
- Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.,CEINGE-Advanced Biotechnologies, Naples, Italy
| | - Alberto Zullo
- CEINGE-Advanced Biotechnologies, Naples, Italy.,Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Linda Sommese
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.,Department of Experimental Medicine, Section of Microbiology, Second University of Naples, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Antonietta Picascia
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy.,U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Concetta Schiano
- Foundation SDN, Institute of Diagnostic and Nuclear Development, IRCCS, Naples, Italy
| | | | - Claudio Napoli
- U.O.C. Division of Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU) and Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.,Foundation SDN, Institute of Diagnostic and Nuclear Development, IRCCS, Naples, Italy
| |
Collapse
|
24
|
Distinct lipid a moieties contribute to pathogen-induced site-specific vascular inflammation. PLoS Pathog 2014; 10:e1004215. [PMID: 25010102 PMCID: PMC4092147 DOI: 10.1371/journal.ppat.1004215] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/16/2014] [Indexed: 01/11/2023] Open
Abstract
Several successful pathogens have evolved mechanisms to evade host defense, resulting in the establishment of persistent and chronic infections. One such pathogen, Porphyromonas gingivalis, induces chronic low-grade inflammation associated with local inflammatory bone loss and systemic inflammation manifested as atherosclerosis. P. gingivalis expresses an atypical lipopolysaccharide (LPS) structure containing heterogeneous lipid A species, that exhibit Toll-like receptor-4 (TLR4) agonist or antagonist activity, or are non-activating at TLR4. In this study, we utilized a series of P. gingivalis lipid A mutants to demonstrate that antagonistic lipid A structures enable the pathogen to evade TLR4-mediated bactericidal activity in macrophages resulting in systemic inflammation. Production of antagonistic lipid A was associated with the induction of low levels of TLR4-dependent proinflammatory mediators, failed activation of the inflammasome and increased bacterial survival in macrophages. Oral infection of ApoE−/− mice with the P. gingivalis strain expressing antagonistic lipid A resulted in vascular inflammation, macrophage accumulation and atherosclerosis progression. In contrast, a P. gingivalis strain producing exclusively agonistic lipid A augmented levels of proinflammatory mediators and activated the inflammasome in a caspase-11-dependent manner, resulting in host cell lysis and decreased bacterial survival. ApoE−/− mice infected with this strain exhibited diminished vascular inflammation, macrophage accumulation, and atherosclerosis progression. Notably, the ability of P. gingivalis to induce local inflammatory bone loss was independent of lipid A expression, indicative of distinct mechanisms for induction of local versus systemic inflammation by this pathogen. Collectively, our results point to a pivotal role for activation of the non-canonical inflammasome in P. gingivalis infection and demonstrate that P. gingivalis evades immune detection at TLR4 facilitating chronic inflammation in the vasculature. These studies support the emerging concept that pathogen-mediated chronic inflammatory disorders result from specific pathogen-mediated evasion strategies resulting in low-grade chronic inflammation. Several human pathogens express structurally divergent forms of lipid A, the endotoxic portion of lipopolysaccharide (LPS), as a strategy to evade host innate immune detection and establish persistent infection. Expression of modified lipid A species promotes pathogen evasion of host recognition by Toll-like receptor-4 (TLR4) and the non-canonical inflammasome. The Gram-negative oral anaerobe, Porphyromonas gingivalis, expresses lipid A structures that function as TLR4 agonists or antagonists, or are immunologically inert. It is currently unclear how modulation of P. gingivalis lipid A expression contributes to innate immune recognition, survival, and the ability of the pathogen to induce local and systemic inflammation. In this study, we demonstrate that P. gingivalis expression of antagonist lipid A species results in attenuated production of proinflammatory mediators and evasion of non-canonical inflammasome activation, facilitating bacterial survival in the macrophage. Infection of atherosclerosis-prone ApoE−/− mice with this strain resulted in progression of chronic inflammation in the vasculature. Notably, the ability of P. gingivalis to induce local inflammatory bone loss was independent of lipid A modifications, supporting distinct mechanisms for induction of local versus systemic inflammation. Our work demonstrates that evasion of immune detection at TLR4 contributes to pathogen persistence and facilitates low-grade chronic inflammation.
Collapse
|
25
|
Periodontal pathogens and atherosclerosis: implications of inflammation and oxidative modification of LDL. BIOMED RESEARCH INTERNATIONAL 2014; 2014:595981. [PMID: 24949459 PMCID: PMC4052162 DOI: 10.1155/2014/595981] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/07/2014] [Accepted: 04/28/2014] [Indexed: 12/29/2022]
Abstract
Inflammation is well accepted to play a crucial role in the development of atherosclerotic lesions, and recent studies have demonstrated an association between periodontal disease and cardiovascular disease. Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans, causative agents of destructive chronic inflammation in the periodontium, can accelerate atheroma deposition in animal models. Emerging evidence suggests that vaccination against virulence factors of these pathogens and anti-inflammatory therapy may confer disease resistance. In this review, we focus on the role of inflammatory mechanisms and oxidative modification in the formation and activation of atherosclerotic plaques accelerated by P. gingivalis or A. actinomycetemcomitans in an ApoE-deficient mouse model and high-fat-diet-fed mice. Furthermore, we examine whether mucosal vaccination with a periodontal pathogen or the anti-inflammatory activity of catechins can reduce periodontal pathogen-accelerated atherosclerosis.
Collapse
|
26
|
Mahendra J, Mahendra L, Felix J, Romanos G. Prevelance of periodontopathogenic bacteria in subgingival biofilm and atherosclerotic plaques of patients undergoing coronary revascularization surgery. J Indian Soc Periodontol 2014; 17:719-24. [PMID: 24554879 PMCID: PMC3917199 DOI: 10.4103/0972-124x.124476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 09/24/2013] [Indexed: 12/19/2022] Open
Abstract
Aim: The objective of the present study was to detect the presence of specific periodontopathogenic bacteria in the coronary plaque of patients with coronary artery disease and to find out the significant association between the periodontal status and the presence of pathogenic bacteria in the coronary plaque. Materials and Methods: The study population consisted of 51 patients with chronic generalized periodontitis undergoing coronary artery bypass grafting. Periodontal parameters were recorded and deoxyribonucleic acid was extracted from the subgingival plaque and coronary atherosclerotic plaque samples of the same patients. Polymerase chain reaction was used to amplify the part of 16S ribosomal ribonucleic acid (rRNA) gene to detect the presence of Aggregatibacter actinomycetemcomitans (Aa), Tannerella forsythia (Tf), Porphyromonas gingivali (Pg), Porphyromonas gingivalis (fimA) gene and Treponema denticola (Td). Results: Aa, Tf, Pg, Pg (fimA) gene and Td were detected in 0%, 31.4%, 45.1% 39.2% and 51% of atherosclerotic plaque samples, respectively. Tf was detected in 19.6%, Pg in 39.2%, Pg (fimA) gene in 33.3% and Td in 35.3% of both, subgingival plaque and atherosclerotic plaque samples. Periodontal parameters correlated with the presence of bacteria in coronary plaque. Aa could not be detected in coronary plaque samples. Conclusions: The study confirmed the detection of Red complex bacteria in coronary plaque samples and these bacteria correlated with the severity of periodontal destruction.
Collapse
Affiliation(s)
- Jaideep Mahendra
- Department of Periodontics, Meenakshi Ammal Dental College and Hospital, Chennai, India
| | - Little Mahendra
- Department of Periodontics, Rajah Muthaiah Medical College and Hospital, Annamalai University, Annamalai Nagar, Chidambaram, Tamil Nadu, India
| | - John Felix
- Department of Bio-Statistics, Rajah Muthaiah Medical College and Hospital, Annamalai University, Annamalai Nagar, Chidambaram, Tamil Nadu, India
| | - Georgios Romanos
- Department of Clinical Dentistry, Department of Periodontology, Eastman Institute for Oral Health, University of Rochester, Rochester, USA
| |
Collapse
|
27
|
Yang J, Wu J, Liu Y, Huang J, Lu Z, Xie L, Sun W, Ji Y. Porphyromonas gingivalis infection reduces regulatory T cells in infected atherosclerosis patients. PLoS One 2014; 9:e86599. [PMID: 24466164 PMCID: PMC3900568 DOI: 10.1371/journal.pone.0086599] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 12/11/2013] [Indexed: 12/22/2022] Open
Abstract
Increasing evidence has shown periodontal pathogen Porphyromonas gingivalis (P.gingivalis) infection contributes to atherosclerosis (AS) progression. P.gingivalis fimbriae act as an important virulence factor in AS. Regulatory T cells (Tregs) may play a crucial role in autoimmune response during this process. However, whether P.gingivalis infection is associated with Tregs dysregulation during AS is still unknown and the prevalence of different P.gingivalis FimA genotypes during this process is unclear. Here we analyzed the distribution of Tregs and in P.gingivalis-infected atherosclerotic patients to reveal the relationship between P.gingivalis infection and Tregs reduction/dysfunction and to elucidate their role in periodontitis-AS interaction. FimA genotype was also examined to determine the prevalence of fimbriae. Our results showed that P.gingivalis infection reduced Tregs in atherosclerotic patients compared with non-atherosclerotic patients and health controls. Concentration of TGF-β1, which plays an important role in the development of Tregs, also decreased in P.gingivalis infected patients. Furthermore, type II FimA seems to show higher prevalence than the other five detected types. The population of Tregs further decreased in patients with type II FimA compared with the other types. P.gingivlias FimA genotype II was the dominant type associated with decreased Treg population. These results indicate that P.gingivalis infection may be associated with Tregs dysregulation in AS; type II FimA may be a predominant genotype in this process.
Collapse
Affiliation(s)
- Jie Yang
- Department of Periodontology, Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
- Central Laboratory of Stomatology, Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Juan Wu
- Department of Periodontology, Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yu Liu
- Department of Periodontology, Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Jin Huang
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu Province, China
| | - Zhipin Lu
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu Province, China
| | - Liping Xie
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Weibin Sun
- Department of Periodontology, Hospital of Stomatology, Medical School, Nanjing University, Nanjing, Jiangsu Province, China
- * E-mail: (WS); (YJ)
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing, Jiangsu Province, China
- * E-mail: (WS); (YJ)
| |
Collapse
|
28
|
Beyond Toll-Like Receptors: Porphyromonas gingivalis Induces IL-6, IL-8, and VCAM-1 Expression Through NOD-Mediated NF-κB and ERK Signaling Pathways in Periodontal Fibroblasts. Inflammation 2013; 37:522-33. [DOI: 10.1007/s10753-013-9766-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
Enersen M, Nakano K, Amano A. Porphyromonas gingivalis fimbriae. J Oral Microbiol 2013; 5:20265. [PMID: 23667717 PMCID: PMC3647041 DOI: 10.3402/jom.v5i0.20265] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 04/11/2013] [Accepted: 04/11/2013] [Indexed: 12/22/2022] Open
Abstract
Marginal periodontitis is not a homogeneous disease but is rather influenced by an intricate set of host susceptibility differences as well as diversities in virulence among the harbored organisms. It is likely that clonal heterogeneity of subpopulations with both high and low levels of pathogenicity exists among organisms harbored by individuals with negligible, slight, or even severe periodontal destruction. Therefore, specific virulent clones of periodontal pathogens may cause advanced and/or aggressive periodontitis. Porphyromonas gingivalis is a predominant periodontal pathogen that expresses a number of potential virulence factors involved in the pathogenesis of periodontitis, and accumulated evidence shows that its expression of heterogenic virulence properties is dependent on clonal diversity. Fimbriae are considered to be critical factors that mediate bacterial interactions with and invasion of host tissues, with P. gingivalis shown to express two distinct fimbria-molecules, long and short fimbriae, on the cell surface, both of which seem to be involved in development of periodontitis. Long fimbriae are classified into six types (I to V and Ib) based on the diversity of fimA genes encoding FimA (a subunit of long fimbriae). Studies of clones with type II fimA have revealed their significantly greater adhesive and invasive capabilities as compared to other fimA type clones. Long and short fimbriae induce various cytokine expressions such as IL-1α, IL-β, IL-6, and TNF-α, which result in alveolar bone resorption. Although the clonal diversity of short fimbriae is unclear, distinct short fimbria-molecules have been found in different strains. These fimbriae variations likely influence the development of periodontal disease.
Collapse
Affiliation(s)
- Morten Enersen
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | | | | |
Collapse
|
30
|
Mahendra J, Mahendra L, Felix J, Romanos GE. Genetic analysis of Porphyromonas gingivalis
(fimA), Aggregatibacter actinomycetemcomitans,
and red complex in coronary plaque. ACTA ACUST UNITED AC 2013; 5:201-7. [DOI: 10.1111/jicd.12030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 12/18/2012] [Indexed: 11/28/2022]
Affiliation(s)
- Jaideep Mahendra
- Department of Periodontology; Meenakshi Ammal Dental College; Meenakshi Academy of Higher Education and Research; Chennai India
| | - Little Mahendra
- Department of Periodontics; Annamalai University; Chidambaram India
| | - John Felix
- Department of Community Medicine; Raja Muthaiah Medical College and Hospital; Annamalai University; Chidambaram India
| | | |
Collapse
|
31
|
Chatzidimitriou D, Kirmizis D, Gavriilaki E, Chatzidimitriou M, Malisiovas N. Atherosclerosis and infection: is the jury still not in? Future Microbiol 2013; 7:1217-30. [PMID: 23030426 DOI: 10.2217/fmb.12.87] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory process accounting for increased cardiovascular and cerebrovascular morbidity and mortality. A wealth of recent data has implicated several infectious agents, mainly Chlamydophila pneumoniae, Helicobacter pylori, CMV and periodontal pathogens, in atherosclerosis. Thus, we sought to comprehensively review the available data on the topic, exploring in particular the pathogenetic mechanisms, and discuss anticipated future directions.
Collapse
|
32
|
Rodrigues PH, Reyes L, Chadda AS, Bélanger M, Wallet SM, Akin D, Dunn W, Progulske-Fox A. Porphyromonas gingivalis strain specific interactions with human coronary artery endothelial cells: a comparative study. PLoS One 2012; 7:e52606. [PMID: 23300720 PMCID: PMC3530483 DOI: 10.1371/journal.pone.0052606] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/19/2012] [Indexed: 12/24/2022] Open
Abstract
Both epidemiologic and experimental findings suggest that infection with Porphyromonas gingivalis exacerbates progression of atherosclerosis. As P. gingivalis exhibits significant strain variation, it is reasonable that different strains possess different capabilities and/or mechanisms by which they promote atherosclerosis. Using P. gingivalis strains that have been previously evaluated in the ApoE null atherosclerosis model, we assessed the ability of W83, A7436, 381, and 33277 to adhere, invade, and persist in human coronary artery endothelial (HCAE) cells. W83 and 381 displayed an equivalent ability to adhere to HCAE cells, which was significantly greater than both A7436 and 33277 (P<0.01). W83, 381, and 33277 were more invasive than A7436 (P<0.0001). However, only W83 and A7436 were able to remain viable up to 48 hours in HCAE cell cultures, whereas 381 was cleared by 48 hours and 33277 was cleared by 24 hours. These differences in persistence were in part due to strain specific differences in intracellular trafficking. Both W83 and 381 trafficked through the autophagic pathway, but not A7436 or 33277. Internalized 381 was the only strain that was dependent upon the autophagic pathway for its survival. Finally, we assessed the efficacy of these strains to activate HCAE cells as defined by production of IL-6, IL-8, IL-12p40, MCP-1, RANTES, TNF-α, and soluble adhesion molecules (sICAM-1, sVCAM-1, and sE-selectin). Only moderate inflammation was observed in cells infected with either W83 or A7436, whereas cells infected with 381 exhibited the most profound inflammation, followed by cells infected with 33277. These results demonstrate that virulence mechanisms among different P. gingivalis strains are varied and that pathogenic mechanisms identified for one strain are not necessarily applicable to other strains.
Collapse
Affiliation(s)
- Paulo H. Rodrigues
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Leticia Reyes
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Amandeep S. Chadda
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Myriam Bélanger
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
| | - Shannon M. Wallet
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, Florida, United States of America
| | - Debra Akin
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - William Dunn
- Department of Anatomy and Cell Biology, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Ann Progulske-Fox
- Department of Oral Biology, College of Dentistry and Center for Molecular Microbiology, University of Florida, Gainesville, Florida, United States of America
| |
Collapse
|
33
|
Trypanosoma cruzi SSP4 Amastigote Protein Induces Expression of Immunoregulatory and Immunosuppressive Molecules in Peripheral Blood Mononuclear Cells. J Trop Med 2012; 2012:829139. [PMID: 23209478 PMCID: PMC3503440 DOI: 10.1155/2012/829139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/25/2012] [Accepted: 10/02/2012] [Indexed: 12/20/2022] Open
Abstract
The acute phase of Chagas' disease in mice and human is marked by states of immunosuppression, in which Trypanosoma cruzi replicates extensively and releases immunomodulatory molecules that delay parasite-specific responses mediated by effector T cells. This mechanism of evasion allows the parasite to spread in the host. Parasite molecules that regulate the host immune response during Chagas' disease have not been fully identified, particularly proteins of the amastigote stage. In this work, we evaluated the role of the GPI anchored SSP4 protein of T. cruzi as an immunomodulatory molecule in peripheral blood mononuclear cells (PBMCs). rMBP::SSP4 protein was able to stimulate nitric oxide (NO) production. Likewise, rMBP::SSP4 induced the expression of genes and production of molecules involved in the inflammatory process, such as, cytokines, chemokines, and adhesion molecules (CAMs) as determined by RT-PCR and ELISA. These results suggest that the amastigote SSP4 molecule could play a key role in the immunoregulatory and/or immunosuppressive process observed in the acute phase of infection with T. cruzi.
Collapse
|
34
|
Madrigal AG, Barth K, Papadopoulos G, Genco CA. Pathogen-mediated proteolysis of the cell death regulator RIPK1 and the host defense modulator RIPK2 in human aortic endothelial cells. PLoS Pathog 2012; 8:e1002723. [PMID: 22685397 PMCID: PMC3369954 DOI: 10.1371/journal.ppat.1002723] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 04/13/2012] [Indexed: 01/07/2023] Open
Abstract
Porphyromonas gingivalis is the primary etiologic agent of periodontal disease that is associated with other human chronic inflammatory diseases, including atherosclerosis. The ability of P. gingivalis to invade and persist within human aortic endothelial cells (HAEC) has been postulated to contribute to a low to moderate chronic state of inflammation, although how this is specifically achieved has not been well defined. In this study, we demonstrate that P. gingivalis infection of HAEC resulted in the rapid cleavage of receptor interacting protein 1 (RIPK1), a mediator of tumor necrosis factor (TNF) receptor-1 (TNF-R1)-induced cell activation or death, and RIPK2, a key mediator of both innate immune signaling and adaptive immunity. The cleavage of RIPK1 or RIPK2 was not observed in cells treated with apoptotic stimuli, or cells stimulated with agonists to TNF-R1, nucleotide oligomerization domain receptor 1(NOD1), NOD2, Toll-like receptor 2 (TLR2) or TLR4. P. gingivalis-induced cleavage of RIPK1 and RIPK2 was inhibited in the presence of a lysine-specific gingipain (Kgp) inhibitor. RIPK1 and RIPK2 cleavage was not observed in HAEC treated with an isogenic mutant deficient in the lysine-specific gingipain, confirming a role for Kgp in the cleavage of RIPK1 and RIPK2. Similar proteolysis of poly (ADP-ribose) polymerase (PARP) was observed. We also demonstrated direct proteolysis of RIPK2 by P. gingivalis in a cell-free system which was abrogated in the presence of a Kgp-specific protease inhibitor. Our studies thus reveal an important role for pathogen-mediated modification of cellular kinases as a potential strategy for bacterial persistence within target host cells, which is associated with low-grade chronic inflammation, a hallmark of pathogen-mediated chronic inflammatory disorders.
Collapse
Affiliation(s)
- Andrés G. Madrigal
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Kenneth Barth
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - George Papadopoulos
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Caroline Attardo Genco
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Kozarov E. Bacterial invasion of vascular cell types: vascular infectology and atherogenesis. Future Cardiol 2012; 8:123-38. [PMID: 22185451 DOI: 10.2217/fca.11.75] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
To portray the chronic inflammation in atherosclerosis, leukocytic cell types involved in the immune response to invading pathogens are often the focus. However, atherogenesis is a complex pathological deterioration of the arterial walls, where vascular cell types are participants with regards to deterioration and disease. Since other recent reviews have detailed the role of both the innate and adaptive immune response in atherosclerosis, herein we will summarize the latest developments regarding the association of bacteria with vascular cell types: infections as a risk factor for atherosclerosis; bacterial invasion of vascular cell types; the atherogenic sequelae of bacterial presence such as endothelial activation and blood clotting; and the identification of the species that are able to colonize this niche. The evidence of a polybacterial infectious component of the atheromatous lesions opens the doors for exploration of the new field of vascular infectology and for the study of atherosclerosis microbiome.
Collapse
Affiliation(s)
- Emil Kozarov
- Section of Oral & Diagnostic Sciences, Columbia University Medical Center, 630 West 168 Street, P&S Box 20, New York, NY 10032, USA.
| |
Collapse
|
36
|
Abstract
It is now well accepted that besides the cholesterol associated mechanisms of atherogenesis, inflammation plays a crucial role in all stages of the development of the atherosclerotic lesion. This 'inflammation hypothesis' raises the possibility that through systemic elevations of pro-inflammatory cytokines, periodontal diseases might also contribute to systemic inflammation and, therefore, to atherogenesis. In fact, there is evidence that periodontal diseases are associated with higher systemic levels of high-sensitivity C-reactive protein and a low grade systemic inflammation. This phenomenon has been explained based on mechanisms associated with either the infectious or the inflammatory nature of periodontal diseases. The purposes of this article were to review (1) the evidence suggesting a role for oral bacterial species, particularly periodontal pathogens, in atherogenesis; (2) the potential mechanisms explaining an etiological role for oral bacteria in atherosclerosis; (3) the evidence suggesting that periodontal infections are accompanied by a heightened state of systemic inflammation; (4) the potential sources of systemic inflammatory biomarkers associated with periodontal diseases; and (5) the effects of periodontal therapy on systemic inflammatory biomarkers and cardiovascular risk.
Collapse
Affiliation(s)
- R Teles
- Department of Periodontology, The Forsyth Institute, Cambridge, MA 02142, USA.
| | | |
Collapse
|
37
|
Roh HC, Yoo DY, Ko SH, Kim YJ, Kim JM. Bacteroides fragilis enterotoxin upregulates intercellular adhesion molecule-1 in endothelial cells via an aldose reductase-, MAPK-, and NF-κB-dependent pathway, leading to monocyte adhesion to endothelial cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:1931-41. [PMID: 21724992 DOI: 10.4049/jimmunol.1101226] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Enterotoxigenic Bacteroides fragilis (ETBF) produces a ∼ 20-kDa heat-labile enterotoxin (BFT) that plays an essential role in mucosal inflammation. Although a variety of inflammatory cells is found at ETBF-infected sites, little is known about leukocyte adhesion in response to BFT stimulation. We investigated whether BFT affected the expression of ICAM-1 and monocytic adhesion to endothelial cells (ECs). Stimulation of HUVECs and rat aortic ECs with BFT resulted in the induction of ICAM-1 expression. Upregulation of ICAM-1 was dependent on the activation of IκB kinase (IKK) and NF-κB signaling. In contrast, suppression of AP-1 did not affect ICAM-1 expression in BFT-stimulated cells. Suppression of NF-κB activity in HUVECs significantly reduced monocytic adhesion, indicating that ICAM-1 expression is indispensable for BFT-induced adhesion of monocytes to the endothelium. Inhibition of JNK resulted in a significant attenuation of BFT-induced ICAM-1 expression in ECs. Moreover, inhibition of aldose reductase significantly reduced JNK-dependent IKK/NF-κB activation, ICAM-1 expression, and adhesion of monocytes to HUVECs. These results suggest that a signaling pathway involving aldose reductase, JNK, IKK, and NF-κB is required for ICAM-1 induction in ECs exposed to BFT, and may be involved in the leukocyte-adhesion cascade following infection with ETBF.
Collapse
Affiliation(s)
- Hyun Cheol Roh
- Department of Microbiology, Hanyang University College of Medicine, Seoul 133-791, Korea
| | | | | | | | | |
Collapse
|
38
|
Tuomainen AM, Hyvärinen K, Ehlers PI, Mervaala E, Leinonen M, Saikku P, Kovanen PT, Jauhiainen M, Pussinen PJ. The effect of proatherogenic microbes on macrophage cholesterol homeostasis in apoE-deficient mice. Microb Pathog 2011; 51:217-24. [PMID: 21421042 DOI: 10.1016/j.micpath.2011.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2010] [Accepted: 03/11/2011] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pathogens such as Aggregatibacter actinomycetemcomitans (Aa) and Chlamydia pneumoniae (Cpn) associate with an increased risk for cardiovascular diseases by inducing inflammation. We hypothesized that the pathogens affect the vascular wall by disturbing cholesterol homeostasis and endothelial function. METHODS Aa- and Cpn-infections were induced in apoE-deficient mice by intravenous and intranasal applications, respectively. Cholesterol efflux from mouse peritoneal macrophages to apo(lipoprotein)A-I was assessed. The efflux capacity of mouse sera as acceptors of cholesterol from RAW264.7-macrophages was determined. Additionally, endothelial function was studied by following the relaxation capacity of rat mesenteric arteries after incubation in the conditioned culture media of the peritoneal macrophages isolated from the mice. RESULTS Infection increased serum phospholipid transfer protein (PLTP) and lipopolysaccharide (LPS) activity, as well as serum amyloid A (SAA) and TNF-α concentrations. Peritoneal macrophages of mice with Aa-infection showed increased cholesterol uptake and reduced cholesterol efflux. Sera of Cpn and Cpn + Aa-infected mice had reduced cholesterol efflux capacity from RAW264.7-macrophages. Conditioned macrophage medium from mice with chronic C. pneumoniae infection induced endothelial dysfunction. Additionally, concentrations of serum adhesion molecules, intercellular adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM) in Cpn-groups and E-selectin in Cpn + Aa-group, were elevated. The serum markers of endothelial function correlated positively with SAA. CONCLUSIONS Aa- and Cpn-infections may generate proatherogenic changes in the vascular wall by affecting the macrophage cholesterol homeostasis and endothelial function.
Collapse
Affiliation(s)
- Anita M Tuomainen
- Institute of Dentistry, University of Helsinki, FI-00014 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ramírez JH, Arce RM, Contreras A. Periodontal treatment effects on endothelial function and cardiovascular disease biomarkers in subjects with chronic periodontitis: protocol for a randomized clinical trial. Trials 2011; 12:46. [PMID: 21324167 PMCID: PMC3049125 DOI: 10.1186/1745-6215-12-46] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 02/16/2011] [Indexed: 01/22/2023] Open
Abstract
Background Periodontal disease (PD) is an infectious clinical entity characterized by the destruction of supporting tissues of the teeth as the result of a chronic inflammatory response in a susceptible host. It has been proposed that PD as subclinical infection may contribute to the etiology and to the pathogenesis of several systemic diseases including Atherosclerosis. A number of epidemiological studies link periodontal disease/edentulism as independent risk factor for acute myocardial infarction, peripheral vascular disease, and cerebrovascular disease. Moreover, new randomized controlled clinical trials have shown an improvement on cardiovascular surrogate markers (endothelial function, sICAM, hsPCR level, fibrinogen) after periodontal treatment. Nonetheless, such trials are still limited in terms of external validity, periodontal treatment strategies, CONSORT-based design and results consistency/extrapolation. The current study is designed to evaluate if periodontal treatment with scaling and root planning plus local delivered chlorhexidine improves endothelial function and other biomarkers of cardiovascular disease in subjects with moderate to severe periodontitis. Methods/Design This randomized, single-blind clinical trial will be performed at two health centers and will include two periodontal treatment strategies. After medical/periodontal screening, a baseline endothelium-dependent brachial artery flow-mediated dilatation (FMD) and other systemic surrogate markers will be obtained from all recruited subjects. Patients then will be randomized to receive either supragingival/subgingival plaque cleaning and calculus removal plus chlorhexidine (treatment group) or supragingival plaque removal only (control group). A second and third FMD will be obtained after 24 hours and 12 weeks in both treatment arms. Each group will consist of 49 patients (n = 98) and all patients will be followed-up for secondary outcomes and will be monitored through a coordinating center. The primary outcomes are FMD differences baseline, 24 hours and 3 months after treatment. The secondary outcomes are differences in C-reactive protein (hs-CRP), glucose serum levels, blood lipid profile, and HOMA index. Discussion This RCT is expected to provide more evidence on the effects of different periodontal treatment modalities on FMD values, as well as to correlate such findings with different surrogate markers of systemic inflammation with cardiovascular effects. Trial registration number ClinicalTrials.gov Identifier: NCT00681564.
Collapse
Affiliation(s)
- Jorge H Ramírez
- Periodontal Medicine Research Group, Department of Periodontology, School of Dentistry, Universidad del Valle, Calle 4B 36-00, Cali, Colombia.
| | | | | |
Collapse
|
40
|
Bakthavatchalu V, Meka A, Sathishkumar S, Lopez MC, Bhattacharyya I, Boyce BF, Mans JJ, Lamont RJ, Baker HV, Ebersole JL, Kesavalu L. Tannerella forsythia infection-induced calvarial bone and soft tissue transcriptional profiles. Mol Oral Microbiol 2011; 25:317-30. [PMID: 20883221 DOI: 10.1111/j.2041-1014.2010.00583.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tannerella forsythia is associated with subgingival biofilms in adult periodontitis, although the molecular mechanisms contributing to chronic inflammation and loss of periodontal bone remain unclear. We examined changes in the host transcriptional profiles during a T. forsythia infection using a murine calvarial model of inflammation and bone resorption. Tannerella forsythia was injected into the subcutaneous soft tissue over calvariae of BALB/c mice for 3 days, after which the soft tissues and calvarial bones were excised. RNA was isolated and Murine GeneChip (Affymetrix, Santa Clara, CA) array analysis of transcript profiles showed that 3226 genes were differentially expressed in the infected soft tissues (P < 0.05) and 2586 genes were differentially transcribed in calvarial bones after infection. Quantitative real-time reverse transcription-polymerase chain reaction analysis of transcription levels of selected genes corresponded well with the microarray results. Biological pathways significantly impacted by T. forsythia infection in calvarial bone and soft tissue included leukocyte transendothelial migration, cell adhesion molecules (immune system), extracellular matrix-receptor interaction, adherens junction, and antigen processing and presentation. Histologic examination revealed intense inflammation and increased osteoclasts in calvariae compared with controls. In conclusion, localized T. forsythia infection differentially induces transcription of a broad array of host genes, and the profiles differ between inflamed soft tissues and calvarial bone.
Collapse
Affiliation(s)
- V Bakthavatchalu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Hayashi C, Gudino CV, Gibson FC, Genco CA. Review: Pathogen-induced inflammation at sites distant from oral infection: bacterial persistence and induction of cell-specific innate immune inflammatory pathways. Mol Oral Microbiol 2011; 25:305-16. [PMID: 20883220 DOI: 10.1111/j.2041-1014.2010.00582.x] [Citation(s) in RCA: 184] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A hallmark of infection with the gram-negative pathogen Porphyromonas gingivalis is the induction of a chronic inflammatory response. P. gingivalis induces a local chronic inflammatory response that results in oral inflammatory bone destruction, which manifests as periodontal disease. In addition to chronic inflammation at the initial site of infection, mounting evidence has accumulated supporting a role for P. gingivalis-mediated periodontal disease as a risk factor for several systemic diseases including, diabetes, preterm birth, stroke, and atherosclerotic cardiovascular disease. A growing number of in vitro studies have demonstrated that P. gingivalis infection stimulates cell activation commensurate with expected responses paralleling inflammatory atherosclerotic-type responses. Furthermore, various mouse models have been used to examine the ability of P. gingivalis to stimulate chronic inflammatory plaque accumulation and recent studies have pointed to a pivotal role for innate immune signaling via the Toll-like receptors in the chronic inflammation associated with P. gingivalis infection. In this review we discuss the pathogen and host cell specificity of these responses and discuss possible mechanisms by which this oral pathogen can induce and maintain a chronic state of inflammation at sites distant from oral infection.
Collapse
Affiliation(s)
- C Hayashi
- Department of Medicine, Sections of Infectious Diseases, Boston University School of Medicine, Boston, MA 02118, USA
| | | | | | | |
Collapse
|
42
|
Shaik-Dasthagirisaheb YB, Kantarci A, Gibson FC. Immune response of macrophages from young and aged mice to the oral pathogenic bacterium Porphyromonas gingivalis. IMMUNITY & AGEING 2010; 7:15. [PMID: 21114831 PMCID: PMC3001696 DOI: 10.1186/1742-4933-7-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Accepted: 11/29/2010] [Indexed: 11/12/2022]
Abstract
Periodontal disease is a chronic inflammatory gum disease that in severe cases leads to tooth loss. Porphyromonas gingivalis (Pg) is a bacterium closely associated with generalized forms of periodontal disease. Clinical onset of generalized periodontal disease commonly presents in individuals over the age of 40. Little is known regarding the effect of aging on inflammation associated with periodontal disease. In the present study we examined the immune response of bone marrow derived macrophages (BMM) from young (2-months) and aged (1-year and 2-years) mice to Pg strain 381. Pg induced robust expression of cytokines; tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10, chemokines; neutrophil chemoattractant protein (KC), macrophage colony stimulating factor (MCP)-1, macrophage inflammatory protein (MIP)-1α and regulated upon activation normal T cell expressed and secreted (RANTES), as well as nitric oxide (NO, measured as nitrite), and prostaglandin E2 (PGE2) from BMM of young mice. BMM from the 2-year age group produced significantly less TNF-α, IL-6 and NO in response to Pg as compared with BMM from 2-months and 1-year of age. We did not observe any difference in the levels of IL-1β, IL-10 and PGE2 produced by BMM in response to Pg. BMM from 2-months and 1-year of age produced similar levels of all chemokines measured with the exception of MCP-1, which was reduced in BMM from 1-year of age. BMM from the 2-year group produced significantly less MCP-1 and MIP-1α compared with 2-months and 1-year age groups. No difference in RANTES production was observed between age groups. Employing a Pg attenuated mutant, deficient in major fimbriae (Pg DPG3), we observed reduced ability of the mutant to stimulate inflammatory mediator expression from BMMs as compared to Pg 381, irrespective of age. Taken together these results support senescence as an important facet of the reduced immunological response observed by BMM of aged host to the periodontal pathogen Pg.
Collapse
Affiliation(s)
- Yazdani B Shaik-Dasthagirisaheb
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | |
Collapse
|
43
|
Bakthavatchalu V, Meka A, Sathishkumar S, Lopez MC, Verma RK, Wallet SM, Bhattacharyya I, Boyce BF, Mans JJ, Lamont RJ, Baker HV, Ebersole JL, Kesavalu L. Molecular characterization of Treponema denticola infection-induced bone and soft tissue transcriptional profiles. Mol Oral Microbiol 2010; 25:260-74. [PMID: 20618700 DOI: 10.1111/j.2041-1014.2010.00575.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Treponema denticola is associated with subgingival biofilms in adult periodontitis and with acute necrotizing ulcerative gingivitis. However, the molecular mechanisms by which T. denticola impacts periodontal inflammation and alveolar bone resorption remain unclear. Here, we examined changes in the host transcriptional profiles during a T. denticola infection using a murine calvarial model of inflammation and bone resorption. T. denticola was injected into the subcutaneous soft tissue over the calvaria of BALB/c mice for 3 days, after which the soft tissues and the calvarial bones were excised. RNA was isolated and analysed for transcript profiling using Murine GeneChip arrays. Following T. denticola infection, 2905 and 1234 genes in the infected calvarial bones and soft tissues, respectively, were differentially expressed (P <or= 0.05). Biological pathways significantly impacted by T. denticola infection in calvarial bone and calvarial tissue included leukocyte transendothelial migration, cell adhesion (immune system) molecules, cell cycle, extracellular matrix-receptor interaction, focal adhesion, B-cell receptor signaling and transforming growth factor-beta signaling pathways resulting in proinflammatory, chemotactic effects, and T-cell stimulation. In conclusion, localized T. denticola infection differentially induces transcription of a broad array of host genes, the profiles of which differed between inflamed calvarial bone and soft tissues.
Collapse
Affiliation(s)
- V Bakthavatchalu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhang D, Zheng H, Zhao J, Lin L, Li C, Liu J, Pan Y. Porphorymonas gingivalis induces intracellular adhesion molecule-1 expression in endothelial cells through the nuclear factor-kappaB pathway, but not through the p38 MAPK pathway. J Periodontal Res 2010; 46:31-8. [DOI: 10.1111/j.1600-0765.2010.01305.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
45
|
Kebschull M, Demmer RT, Papapanou PN. "Gum bug, leave my heart alone!"--epidemiologic and mechanistic evidence linking periodontal infections and atherosclerosis. J Dent Res 2010; 89:879-902. [PMID: 20639510 DOI: 10.1177/0022034510375281] [Citation(s) in RCA: 330] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Evidence from epidemiologic studies suggests that periodontal infections are independently associated with subclinical and clinical atherosclerotic vascular disease. Although the strength of the reported associations is modest, the consistency of the data across diverse populations and a variety of exposure and outcome variables suggests that the findings are not spurious or attributable only to the effects of confounders. Analysis of limited data from interventional studies suggests that periodontal treatment generally results in favorable effects on subclinical markers of atherosclerosis, although such analysis also indicates considerable heterogeneity in responses. Experimental mechanistic in vitro and in vivo studies have established the plausibility of a link between periodontal infections and atherogenesis, and have identified biological pathways by which these effects may be mediated. However, the utilized models are mostly mono-infections of host cells by a limited number of 'model' periodontal pathogens, and therefore may not adequately portray human periodontitis as a polymicrobial, biofilm-mediated disease. Future research must identify in vivo pathways in humans that may (i) lead to periodontitis-induced atherogenesis, or (ii) result in treatment-induced reduction of atherosclerosis risk. Data from these studies will be essential for determining whether periodontal interventions have a role in the primary or secondary prevention of atherosclerosis.
Collapse
Affiliation(s)
- M Kebschull
- Division of Periodontics, Section of Oral and Diagnostic Sciences, College of Dental Medicine, 630 W 168th Street, PH-7-E-110, New York, NY 10032, USA
| | | | | |
Collapse
|
46
|
Distribution of Porphyromonas gingivalis fimA genotypes in primary endodontic infections. ACTA ACUST UNITED AC 2010; 109:474-8. [PMID: 20219603 DOI: 10.1016/j.tripleo.2009.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 11/03/2009] [Accepted: 11/04/2009] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Long fimbriae (FimA) are important virulence factors of Porphyromonas gingivalis. Based on the diversity of the fimA gene, this species is classified into 6 genotypes. This study surveyed samples from primary endodontic infections for the presence of these P. gingivalis fimA variants. STUDY DESIGN Genomic DNA isolated from samples taken from 25 root canals of teeth with chronic apical periodontitis and 25 aspirates from acute apical abscess was used as template in polymerase chain reaction (PCR) assays directed toward the detection of the different P. gingivalis fimA genotypes. RESULTS Porphyromonas gingivalis was detected by a 16S rRNA gene-based PCR in 36% of the total number of cases sampled (44% of chronic apical periodontitis and 28% of abscess aspirates). In cases of chronic apical periodontitis, P. gingivalis variant type IV was the most prevalent (24%), followed by types I (20%), II (16%), and III (8%). In acute abscess samples, variant type II was the most prevalent (12%), followed by types III and IV (8% of each) and type I (4%). Combinations of up to 3 different genotypes were detected in a few cases. No single fimA genotype variant or combination thereof was significantly associated with symptoms. Overall, fimA types IV (16%), II (14%), and I (12%) were the most prevalent. CONCLUSIONS Findings demonstrated that different P. gingivalis fimA genotypes can be present in primary endodontic infections.
Collapse
|
47
|
Meka A, Bakthavatchalu V, Sathishkumar S, Lopez MC, Verma RK, Wallet SM, Bhattacharyya I, Boyce BF, Handfield M, Lamont RJ, Baker HV, Ebersole JL, Kesavalu L. Porphyromonas gingivalis infection-induced tissue and bone transcriptional profiles. Mol Oral Microbiol 2010; 25:61-74. [PMID: 20331794 DOI: 10.1111/j.2041-1014.2009.00555.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Porphyromonas gingivalis has been associated with subgingival biofilms in adult periodontitis. However, the molecular mechanisms of its contribution to chronic gingival inflammation and loss of periodontal structural integrity remain unclear. This investigation aimed to examine changes in the host transcriptional profiles during a P. gingivalis infection using a murine calvarial model of inflammation and bone resorption. P. gingivalis FDC 381 was injected into the subcutaneous soft tissue over the calvaria of BALB/c mice for 3 days, after which the soft tissues and calvarial bones were excised. RNA was isolated from infected soft tissues and calvarial bones and was analysed for transcript profiles using Murine GeneChip((R)) arrays to provide a molecular profile of the events that occur following infection of these tissues. After P. gingivalis infection, 6452 and 2341 probe sets in the infected soft tissues and calvarial bone, respectively, were differentially expressed (P </= 0.05). Biological pathways significantly impacted by P. gingivalis infection in tissues and calvarial bone included cell adhesion (immune system) molecules, Toll-like receptors, B-cell receptor signaling, transforming growth factor-beta cytokine family receptor signaling, and major histocompatibility complex class II antigen processing pathways resulting in proinflammatory, chemotactic effects, T-cell stimulation, and downregulation of antiviral and T-cell chemotactic effects. P. gingivalis-induced inflammation activated osteoclasts, leading to local bone resorption. This is the first in vivo evidence that localized P. gingivalis infection differentially induces transcription of a broad array of host genes, the profiles of which differed between inflamed soft tissues and calvarial bone.
Collapse
Affiliation(s)
- A Meka
- Department of Periodontology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sun W, Wu J, Lin L, Huang Y, Chen Q, Ji Y. Porphyromonas gingivalisstimulates the release of nitric oxide by inducing expression of inducible nitric oxide synthases and inhibiting endothelial nitric oxide synthases. J Periodontal Res 2010; 45:381-8. [DOI: 10.1111/j.1600-0765.2009.01249.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
49
|
Friedlander AH, Sung EC, Chung EM, Garrett NR. Radiographic quantification of chronic dental infection and its relationship to the atherosclerotic process in the carotid arteries. ACTA ACUST UNITED AC 2010; 109:615-21. [DOI: 10.1016/j.tripleo.2009.10.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 10/14/2009] [Accepted: 10/19/2009] [Indexed: 01/17/2023]
|
50
|
Pollreisz A, Huang Y, Roth GA, Cheng B, Kebschull M, Papapanou PN, Schmidt AM, Lalla E. Enhanced monocyte migration and pro-inflammatory cytokine production by Porphyromonas gingivalis infection. J Periodontal Res 2009; 45:239-45. [PMID: 19778327 DOI: 10.1111/j.1600-0765.2009.01225.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVE Porphyromonas gingivalis, a major periodontal pathogen, has been reported to be involved in atherogenesis. In order to further understand this pathogen's link with systemic inflammation and vascular disease, we investigated its influence on murine monocytes and macrophages from three different sources. MATERIAL AND METHODS Concanavalin A-elicited peritoneal macrophages, peripheral blood monocyte-derived macrophages and WEHI 274.1 monocytes were infected with either P. gingivalis 381 or its non-invasive fimbriae-deficient mutant, DPG3. RESULTS Infection with P. gingivalis 381 markedly induced monocyte migration and significantly enhanced production of the pro-inflammatory cytokines, tumor necrosis factor-alpha and interleukin-6. Consistent with a role for this pathogen's major fimbriae and/or its invasive capacity, infection with DPG3 had a minimal effect on both monocyte attraction and pro-inflammatory cytokine production. CONCLUSION Since monocyte recruitment and activation are important steps in the development of vascular inflammation and atherosclerosis, these results suggest that P. gingivalis infection may be involved in these processes.
Collapse
Affiliation(s)
- A Pollreisz
- Division of Periodontics, Section of Oral and Diagnostic Sciences, College of Dental Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|