1
|
Ma L, Cao Z. Periodontopathogen-Related Cell Autophagy-A Double-Edged Sword. Inflammation 2025; 48:1-14. [PMID: 38762837 DOI: 10.1007/s10753-024-02049-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
The periodontium is a highly organized ecosystem, and the imbalance between oral microorganisms and host defense leads to periodontal diseases. The periodontal pathogens, mainly Gram-negative anaerobic bacteria, colonize the periodontal niches or enter the blood circulation, resulting in periodontal tissue destruction and distal organ damage. This phenomenon links periodontitis with various systemic conditions, including cardiovascular diseases, malignant tumors, steatohepatitis, and Alzheimer's disease. Autophagy is an evolutionarily conserved cellular self-degradation process essential for eliminating internalized pathogens. Nowadays, increasing studies have been carried out in cells derived from periodontal tissues, immune system, and distant organs to investigate the relationship between periodontal pathogen infection and autophagy-related activities. On one hand, as a vital part of innate and adaptive immunity, autophagy actively participates in host resistance to periodontal bacterial infection. On the other, certain periodontal pathogens exploit autophagic vesicles or pathways to evade immune surveillance, therefore achieving survival within host cells. This review provides an overview of the autophagy process and focuses on periodontopathogen-related autophagy and their involvements in cells of different tissue origins, so as to comprehensively understand the role of autophagy in the occurrence and development of periodontal diseases and various periodontitis-associated systemic illnesses.
Collapse
Affiliation(s)
- Li Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
2
|
Cao M, Wang S, Zhou S, Yan M, Zou Y, Cui Y, Lou X, Gao Y, Chen Y, Han Z, Qian Y, Chen J, Li X. Development of monoclonal antibodies against P. gingivalis Mfa1 and their protective capacity in an experimental periodontitis model. mSphere 2025; 10:e0072124. [PMID: 39699191 PMCID: PMC11774036 DOI: 10.1128/msphere.00721-24] [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: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Porphyromonas gingivalis (P. gingivalis), a gram-negative, black-pigmented anaerobe, is a major etiological agent and a leading cause of periodontitis. Fimbriae protein Mfa1 is a key virulence factor of P. gingivalis and plays a crucial role in bacterial adhesion, colonization, biofilm formation, and persistent inflammation, making it a promising therapeutic target. However, the role of anti-Mfa1 antibodies and the underlying protective mechanisms remain largely unexplored. Here, we developed and characterized the monoclonal antibodies (mAbs) targeting the Mfa1 protein of P. gingivalis. Function analysis showed that anti-Mfa1 mAbs mediated bacterial agglutination and inhibited P. gingivalis adhesion to saliva-coated hydroxyapatite and host cells. Notably, anti-Mfa1 mAbs significantly reduced bacterial burden and alveolar bone loss in a P. gingivalis-induced experimental periodontitis model. These results show that anti-Mfa1 mAbs can be beneficial in alleviating P. gingivalis infections, and provide important insights for the development of adequate adjuvant treatment regimens for Mfa1-targeted therapeutics. IMPORTANCE Fimbriae (pili) play an important role in bacterial adhesion, invasion of host cells and tissues, and formation of biofilms. Studies have shown that two types of fimbriae of Porphyromonas gingivalis, FimA and Mfa1, are important for colonization and infection through their binding to host tissues and other bacteria. While anti-FimA antibodies have been shown to improve periodontitis, the effect of anti-Mfa1 antibodies on P. gingivalis infection and periodontitis was previously unknown. In this study, we report for the first time that anti-Mfa1 monoclonal antibodies can reduce P. gingivalis infection and improve periodontitis. These findings suggest that Mfa1 represents a promising therapeutic target, and the development of anti-Mfa1 mAbs holds a potential as essential diagnostic and adjunctive therapeutic tools for managing P. gingivalis-related diseases.
Collapse
Affiliation(s)
- Mingya Cao
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Siyu Wang
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Shengke Zhou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Min Yan
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yu Zou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yuan Cui
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Xinyu Lou
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yichang Gao
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Ying Chen
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Zijing Han
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Yi Qian
- The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| | - Jingying Chen
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| | - Xia Li
- Joint National Laboratory for Antibody Drug Engineering, School of Medicine, Henan University, Kaifeng, China
| |
Collapse
|
3
|
Shawkatova I, Durmanova V, Javor J. Alzheimer's Disease and Porphyromonas gingivalis: Exploring the Links. Life (Basel) 2025; 15:96. [PMID: 39860036 PMCID: PMC11766648 DOI: 10.3390/life15010096] [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: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Recent research highlights compelling links between oral health, particularly periodontitis, and systemic diseases, including Alzheimer's disease (AD). Although the biological mechanisms underlying these associations remain unclear, the role of periodontal pathogens, particularly Porphyromonas gingivalis, has garnered significant attention. P. gingivalis, a major driver of periodontitis, is recognized for its potential systemic effects and its putative role in AD pathogenesis. This review examines evidence connecting P. gingivalis to hallmark AD features, such as amyloid β accumulation, tau hyperphosphorylation, neuroinflammation, and other neuropathological features consistent with AD. Virulence factors, such as gingipains and lipopolysaccharides, were shown to be implicated in blood-brain barrier disruption, neuroinflammation, and neuronal damage. P. gingivalis-derived outer membrane vesicles may serve to disseminate virulence factors to brain tissues. Indirect mechanisms, including systemic inflammation triggered by chronic periodontal infections, are also supposed to exacerbate neurodegenerative processes. While the exact pathways remain uncertain, studies detecting P. gingivalis virulence factors and its other components in AD-affected brains support their possible role in disease pathogenesis. This review underscores the need for further investigation into P. gingivalis-mediated mechanisms and their interplay with host responses. Understanding these interactions could provide critical insights into novel strategies for reducing AD risk through periodontal disease management.
Collapse
Affiliation(s)
- Ivana Shawkatova
- Institute of Immunology, Faculty of Medicine, Comenius University in Bratislava, Odborarske nam. 14, 811 08 Bratislava, Slovakia; (V.D.); (J.J.)
| | | | | |
Collapse
|
4
|
Fan X, Qu PY, Luan KF, Sun CY, Ren HP, Sun XH, Lan J. A cleaved adhesin DNA vaccine targeting dendritic cell against Porphyromonas gingivalis-induced periodontal disease. Mol Oral Microbiol 2024; 39:433-445. [PMID: 38696249 DOI: 10.1111/omi.12465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/28/2024] [Accepted: 04/09/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Arg-gingipain A (RgpA) is the primary virulence factor of Porphyromonas gingivalis and contains hemagglutinin adhesin (HA), which helps bacteria adhere to cells and proteins. Hemagglutinin's functional domains include cleaved adhesin (CA), which acts as a hemagglutination and hemoglobin-binding actor. Here, we confirmed that the HA and CA genes are immunogenic, and using adjuvant chemokine to target dendritic cells (DCs) enhanced protective autoimmunity against P. gingivalis-induced periodontal disease. METHODS C57 mice were immunized prophylactically with pVAX1-CA, pVAX1-HA, pVAX1, and phosphate-buffered saline (PBS) through intramuscular injection every 2 weeks for a total of three administrations before P. gingivalis-induced periodontitis. The DCs were analyzed using flow cytometry and ribonucleic acid sequencing (RNA-seq) transcriptomic assays following transfection with CA lentivirus. The efficacy of the co-delivered molecular adjuvant CA DNA vaccine was evaluated in vivo using flow cytometry, immunofluorescence techniques, and micro-computed tomography. RESULTS After the immunization, both the pVAX1-CA and pVAX1-HA groups exhibited significantly elevated P. gingivalis-specific IgG and IgG1, as well as a reduction in bone loss around periodontitis-affected teeth, compared to the pVAX1 and PBS groups (p < 0.05). The expression of CA promoted the secretion of HLA, CD86, CD83, and DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) in DCs. Furthermore, the RNA-seq analysis revealed a significant increase in the chemokine (C-C motif) ligand 19 (p < 0.05). A notable elevation in the quantities of DCs co-labeled with CD11c and major histocompatibility complex class II, along with an increase in interferon-gamma (IFN-γ) cells, was observed in the inguinal lymph nodes of mice subjected to CCL19-CA immunization. This outcome effectively illustrated the preservation of peri-implant bone mass in rats afflicted with P. gingivalis-induced peri-implantitis (p < 0.05). CONCLUSIONS The co-administration of a CCL19-conjugated CA DNA vaccine holds promise as an innovative and targeted immunization strategy against P. gingivalis-induced periodontitis and peri-implantitis.
Collapse
Affiliation(s)
- Xin Fan
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Peng-Yu Qu
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Ke-Feng Luan
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Chen-Yu Sun
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Hui-Ping Ren
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Xue-Hui Sun
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Jing Lan
- School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| |
Collapse
|
5
|
Meghil MM, Cutler CW. Influence of Vitamin D on Periodontal Inflammation: A Review. Pathogens 2023; 12:1180. [PMID: 37764988 PMCID: PMC10537363 DOI: 10.3390/pathogens12091180] [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: 08/09/2023] [Revised: 09/09/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
The active form of vitamin D is the hormonally active 1,25(OH)2D3 (Vit D) vitamin, which plays an important role in bone biology and host immunity. The vitamin D receptor (VDR) is a nuclear ligand-dependent transcription factor expressed by many cells. Ligation of VDR by VitD regulates a wide plethora of genes and physiologic functions through the formation of the complex Vit D-VDR signaling cascade. The influence of Vit D-VDR signaling in host immune response to microbial infection has been of interest to many researchers. This is particularly important in oral health and diseases, as oral mucosa is exposed to a complex microbiota, with certain species capable of causing disruption to immune homeostasis. In this review, we focus on the immune modulatory roles of Vit D in the bone degenerative oral disease, periodontitis.
Collapse
Affiliation(s)
- Mohamed M. Meghil
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Christopher W. Cutler
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
6
|
Ricciardi RM, Cipollone A, D'Ardes D, Di Giacomo D, Pignatelli P, Cipollone F, Curia MC, Magni P, Bucci M. Risk Factors and Immunoinflammatory Mechanisms Leading to Atherosclerosis: Focus on the Role of Oral Microbiota Dysbiosis. Microorganisms 2023; 11:1479. [PMID: 37374981 DOI: 10.3390/microorganisms11061479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/25/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Cardiovascular diseases (CVD), including myocardial infarction and stroke, are currently the leading cause of morbidity, disability and mortality worldwide. Recently, researchers have focused their attention on the alterations of the gut and oral microbiota, investigating the possible role of their dysbiosis in the pathogenesis and/or progression of CVD. In this regard, it has been shown that endothelial dysfunction, a major feature of CVD, can also be induced by chronic periodontal infection, due to a systemic pro-inflammatory condition, as suggested by increased plasma levels of acute phase proteins, IL-6 and fibrinogen. Moreover, proatherogenic dysfunctions can also be promoted by direct bacterial invasion of the endothelium. This review reports the current evidence about the possible role of oral microbiota dysbiosis and the related immunoinflammatory components in the pathophysiology of atherosclerosis and associated CVD. It is concluded that integration of oral microbiota sampling into clinical practice may result in a more accurate assessment of CV risk in patients and even modify their prognosis.
Collapse
Affiliation(s)
- Riccardo Mattia Ricciardi
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| | - Alessia Cipollone
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| | - Damiano D'Ardes
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| | - Davide Di Giacomo
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| | - Pamela Pignatelli
- COMDINAV DUE, Nave Cavour, Italian Navy, Stazione Navale Mar Grande-Viale Jonio, 74122 Taranto, Italy
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| | - Maria Cristina Curia
- Department of Medical, Oral and Biotechnological Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
| | - Paolo Magni
- Epidemiology and Preventive Pharmacology Service (SEFAP), Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, 20133 Milan, Italy
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| | - Marco Bucci
- Department of Medicine and Aging Sciences, Università degli Studi "Gabriele d'Annunzio" di Chieti-Pescara, 66100 Chieti, Italy
- Regional Center for the Study of Atherosclerosis, Hypertension and Dyslipidemia, "SS Annunziata" Hospital-ASL, 66100 Chieti, Italy
| |
Collapse
|
7
|
Moreno CM, Boeree E, Freitas CMT, Weber KS. Immunomodulatory role of oral microbiota in inflammatory diseases and allergic conditions. FRONTIERS IN ALLERGY 2023; 4:1067483. [PMID: 36873050 PMCID: PMC9981797 DOI: 10.3389/falgy.2023.1067483] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023] Open
Abstract
In recent years, the interplay between oral microbiota and systemic disease has gained attention as poor oral health is associated with several pathologies. The oral microbiota plays a role in the maintenance of overall health, and its dysbiosis influences chronic inflammation and the pathogenesis of gum diseases. Periodontitis has also been associated with other diseases and health complications such as cancer, neurogenerative and autoimmune disorders, chronic kidney disease, cardiovascular diseases, rheumatic arthritis, respiratory health, and adverse pregnancy outcomes. The host microbiota can influence immune cell development and immune responses, and recent evidence suggests that changes in oral microbiota composition may also contribute to sensitization and the development of allergic reactions, including asthma and peanut allergies. Conversely, there is also evidence that allergic reactions within the gut may contribute to alterations in oral microbiota composition. Here we review the current evidence of the role of the oral microbiota in inflammatory diseases and health complications, as well as its future relevance in improving health and ameliorating allergic disease.
Collapse
Affiliation(s)
- Carlos M Moreno
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Ellie Boeree
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Claudia M Tellez Freitas
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
8
|
Shahoumi LA, Saleh MHA, Meghil MM. Virulence Factors of the Periodontal Pathogens: Tools to Evade the Host Immune Response and Promote Carcinogenesis. Microorganisms 2023; 11:115. [PMID: 36677408 PMCID: PMC9860638 DOI: 10.3390/microorganisms11010115] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Periodontitis is the most common chronic, inflammatory oral disease that affects more than half of the population in the United States. The disease leads to destruction of the tooth-supporting tissue called periodontium, which ultimately results in tooth loss if uncured. The interaction between the periodontal microbiota and the host immune cells result in the induction of a non-protective host immune response that triggers host tissue destruction. Certain pathogens have been implicated periodontal disease formation that is triggered by a plethora of virulence factors. There is a collective evidence on the impact of periodontal disease progression on systemic health. Of particular interest, the role of the virulence factors of the periodontal pathogens in facilitating the evasion of the host immune cells and promotion of carcinogenesis has been the focus of many researchers. The aim of this review is to examine the influence of the periodontal pathogens Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans), Porphyromonas gingivalis (P. gingivalis), and Fusobacterium nucleatum (F. nucleatum) in the modulation of the intracellular signaling pathways of the host cells in order to evade the host immune response and interfere with normal host cell death and the role of their virulence factors in this regard.
Collapse
Affiliation(s)
- Linah A. Shahoumi
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Muhammad H. A. Saleh
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Mohamed M. Meghil
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia at Augusta University, Augusta, GA 30912, USA
- Department of Periodontics, The Dental College of Georgia at Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Li C, Yu R, Ding Y. Association between Porphyromonas Gingivalis and systemic diseases: Focus on T cells-mediated adaptive immunity. Front Cell Infect Microbiol 2022; 12:1026457. [PMID: 36467726 PMCID: PMC9712990 DOI: 10.3389/fcimb.2022.1026457] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/25/2022] [Indexed: 12/01/2023] Open
Abstract
The association between periodontal disease and systemic disease has become a research hotspot. Porphyromonas gingivalis (P. gingivalis), a crucial periodontal pathogen, affects the development of systemic diseases. The pathogenicity of P. gingivalis is largely linked to interference with the host's immunity. This review aims to discover the role of P. gingivalis in the modulation of the host's adaptive immune system through a large number of virulence factors and the manipulation of cellular immunological responses (mainly mediated by T cells). These factors may affect the cause of large numbers of systemic diseases, such as atherosclerosis, hypertension, adverse pregnancy outcomes, inflammatory bowel disease, diabetes mellitus, non-alcoholic fatty liver disease, rheumatoid arthritis, and Alzheimer's disease. The point of view of adaptive immunity may provide a new idea for treating periodontitis and related systemic diseases.
Collapse
Affiliation(s)
- Cheng Li
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ran Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yumei Ding
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
10
|
Mazumdar V, Joshi K, Nandi BR, Namani S, Gupta VK, Radhakrishnan G. Host F-Box Protein 22 Enhances the Uptake of Brucella by Macrophages and Drives a Sustained Release of Proinflammatory Cytokines through Degradation of the Anti-Inflammatory Effector Proteins of Brucella. Infect Immun 2022; 90:e0006022. [PMID: 35420446 PMCID: PMC9119127 DOI: 10.1128/iai.00060-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 11/20/2022] Open
Abstract
Brucella species are intracellular bacterial pathogens, causing the worldwide zoonotic disease brucellosis. Brucella invades professional and nonprofessional phagocytic cells, followed by resisting intracellular killing and establishing a replication permissive niche. Brucella also modulates the innate and adaptive immune responses of the host for its chronic persistence. The complex intracellular cycle of Brucella depends in a major way on multiple host factors, but limited information is available on host and bacterial proteins that play an essential role in the invasion, intracellular replication, and modulation of host immune responses. By employing a small interfering RNA (siRNA) screening, we identified a role for the host protein FBXO22 in the Brucella-macrophage interaction. FBXO22 is the key element in the SCF E3 ubiquitination complex, where it determines the substrate specificity for ubiquitination and degradation of various host proteins. Downregulation of FBXO22 by siRNA or the CRISPR-Cas9 system resulted in diminished uptake of Brucella into macrophages, which was dependent on NF-κB-mediated regulation of phagocytic receptors. FBXO22 expression was upregulated in Brucella-infected macrophages, which resulted in induction of phagocytic receptors and enhanced production of proinflammatory cytokines through NF-κB. Furthermore, we found that FBXO22 recruits the effector proteins of Brucella, including the anti-inflammatory proteins TcpB and OMP25, for degradation through the SCF complex. We did not observe any role for another F-box-containing protein of the SCF complex, β-TrCP, in the Brucella-macrophage interaction. Our findings unravel novel functions of FBXO22 in host-pathogen interaction and its contribution to pathogenesis of infectious diseases.
Collapse
Affiliation(s)
- Varadendra Mazumdar
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Kiranmai Joshi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Binita Roy Nandi
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
- Regional Centre for Biotechnology (RCB), Faridabad, Haryana, India
| | - Swapna Namani
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| | - Vivek Kumar Gupta
- ICAR-Indian Veterinary Research Institute (ICAR-IVRI), Izatnagar, Bareilly, India
| | - Girish Radhakrishnan
- Laboratory of Immunology and Microbial Pathogenesis, National Institute of Animal Biotechnology (NIAB), Hyderabad, Telangana, India
| |
Collapse
|
11
|
Almarghlani A, Settem RP, Croft AJ, Metcalfe S, Giangreco M, Kay JG. Interleukin-34 Permits Porphyromonas gingivalis Survival and NF-κB p65 Inhibition in Macrophages. Mol Oral Microbiol 2022; 37:109-121. [PMID: 35576119 DOI: 10.1111/omi.12366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/29/2022]
Abstract
Interleukin-34 (IL-34) is a cytokine that supports the viability and differentiation of macrophages. An important cytokine for the development of epidermal immunity, IL-34 is present and plays a role in the immunity of the oral environment. IL-34 has been linked to inflammatory periodontal diseases, which involve innate phagocytes, including macrophages. Whether IL-34 can alter the ability of macrophages to effectively interact with oral microbes is currently unclear. Using macrophages derived from human blood monocytes with either the canonical cytokine colony-stimulating factor (CSF)1 or IL-34, we compared the ability of the macrophages to phagocytose, kill, and respond through the production of cytokines to the periodontal keystone pathogen Porphyromonas gingivalis. While macrophages derived from both cytokines were able to engulf the bacterium equally, IL-34 derived macrophages were much less capable of killing internalized P. gingivalis. Of the macrophage cell surface receptors known to interact with P. gingivalis, DC-SIGN was found to have the largest variation between IL-34 and CSF1-derived macrophages. We also found that upon interaction with P. gingivalis, IL-34 derived macrophages produced significantly less of the neutrophil chemotactic factor IL-8 than macrophages derived in the presence of CSF1. Mechanistically, we identified that levels of IL-8 corresponded with P. gingivalis survival and dephosphorylation of the major transcription factor NF-κB p65. Overall, we found that macrophages differentiated in the presence of IL-34, a dominant cytokine in the oral gingiva, have a reduced ability to kill the keystone pathogen P. gingivalis and may be susceptible to specific bacteria-mediated cytokine modification. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ammar Almarghlani
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA.,Current Address: Department of Periodontics, Faculty of Dentistry, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Rajendra P Settem
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Andrew J Croft
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Sarah Metcalfe
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Matthew Giangreco
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| | - Jason G Kay
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14214, USA
| |
Collapse
|
12
|
A Tale of Two Fimbriae: How Invasion of Dendritic Cells by Porphyromonas gingivalis Disrupts DC Maturation and Depolarizes the T-Cell-Mediated Immune Response. Pathogens 2022; 11:pathogens11030328. [PMID: 35335652 PMCID: PMC8954744 DOI: 10.3390/pathogens11030328] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 12/29/2022] Open
Abstract
Porphyromonas gingivalis (P. gingivalis) is a unique pathogen implicated in severe forms of periodontitis (PD), a disease that affects around 50% of the US population. P. gingivalis is equipped with a plethora of virulence factors that it uses to exploit its environment and survive. These include distinct fimbrial adhesins that enable it to bind to other microbes, colonize inflamed tissues, acquire nutrients, and invade cells of the stroma and immune system. Most notable for this review is its ability to invade dendritic cells (DCs), which bridge the innate and adaptive immune systems. This invasion process is tightly linked to the bridging functions of resultant DCs, in that it can disable (or stimulate) the maturation function of DCs and cytokines that are secreted. Maturation molecules (e.g., MHCII, CD80/CD86, CD40) and inflammatory cytokines (e.g., IL-1b, TNFa, IL-6) are essential signals for antigen presentation and for proliferation of effector T-cells such as Th17 cells. In this regard, the ability of P. gingivalis to coordinately regulate its expression of major (fimA) and minor (mfa-1) fimbriae under different environmental influences becomes highly relevant. This review will, therefore, focus on the immunoregulatory role of P. gingivalis fimbriae in the invasion of DCs, intracellular signaling, and functional outcomes such as alveolar bone loss and immune senescence.
Collapse
|
13
|
El-Awady AR, Elashiry M, Morandini AC, Meghil MM, Cutler CW. Dendritic cells a critical link to alveolar bone loss and systemic disease risk in periodontitis: Immunotherapeutic implications. Periodontol 2000 2022; 89:41-50. [PMID: 35244951 DOI: 10.1111/prd.12428] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Extensive research in humans and animal models has begun to unravel the complex mechanisms that drive the immunopathogenesis of periodontitis. Neutrophils mount an early and rapid response to the subgingival oral microbiome, producing destructive enzymes to kill microbes. Chemokines and cytokines are released that attract macrophages, dendritic cells, and T cells to the site. Dendritic cells, the focus of this review, are professional antigen-presenting cells on the front line of immune surveillance. Dendritic cells consist of multiple subsets that reside in the epithelium, connective tissues, and major organs. Our work in humans and mice established that myeloid dendritic cells are mobilized in periodontitis. This occurs in lymphoid and nonlymphoid oral tissues, in the bloodstream, and in response to Porphyromonas gingivalis. Moreover, the dendritic cells mature in situ in gingival lamina propria, forming immune conjugates with cluster of differentiation (CD) 4+ T cells, called oral lymphoid foci. At such foci, the decisions are made as to whether to promote bone destructive T helper 17 or bone-sparing regulatory T cell responses. Interestingly, dendritic cells lack potent enzymes and reactive oxygen species needed to kill and degrade endocytosed microbes. The keystone pathogen P. gingivalis exploits this vulnerability by invading dendritic cells in the tissues and peripheral blood using its distinct fimbrial adhesins. This promotes pathogen dissemination and inflammatory disease at distant sites, such as atherosclerotic plaques. Interestingly, our recent studies indicate that such P. gingivalis-infected dendritic cells release nanosized extracellular vesicles called exosomes, in higher numbers than uninfected dendritic cells do. Secreted exosomes and inflammasome-related cytokines are a key feature of the senescence-associated secretory phenotype. Exosomes communicate in paracrine with neighboring stromal cells and immune cells to promote and amplify cellular senescence. We have shown that dendritic cell-derived exosomes can be custom tailored to target and reprogram specific immune cells responsible for inflammatory bone loss in mice. The long-term goal of these immunotherapeutic approaches, ongoing in our laboratory and others, is to promote human health and longevity.
Collapse
Affiliation(s)
- Ahmed R El-Awady
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mahmoud Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Ana C Morandini
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, Georgia, USA.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
14
|
Elebyary O, Barbour A, Fine N, Tenenbaum HC, Glogauer M. The Crossroads of Periodontitis and Oral Squamous Cell Carcinoma: Immune Implications and Tumor Promoting Capacities. FRONTIERS IN ORAL HEALTH 2022; 1:584705. [PMID: 35047982 PMCID: PMC8757853 DOI: 10.3389/froh.2020.584705] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022] Open
Abstract
Periodontitis (PD) is increasingly considered to interact with and promote a number of inflammatory diseases, including cancer. In the case of oral squamous cell carcinoma (OSCC) the local inflammatory response associated with PD is capable of triggering altered cellular events that can promote cancer cell invasion and proliferation of existing primary oral carcinomas as well as supporting the seeding of metastatic tumor cells into the gingival tissue giving rise to secondary tumors. Both the immune and stromal components of the periodontium exhibit phenotypic alterations and functional differences during PD that result in a microenvironment that favors cancer progression. The inflammatory milieu in PD is ideal for cancer cell seeding, migration, proliferation and immune escape. Understanding the interactions governing this attenuated anti-tumor immune response is vital to unveil unexplored preventive or therapeutic possibilities. Here we review the many commonalities between the oral-inflammatory microenvironment in PD and oral-inflammatory responses that are associated with OSCC progression, and how these conditions can act to promote and sustain the hallmarks of cancer.
Collapse
Affiliation(s)
- Omnia Elebyary
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | | | - Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - Howard C Tenenbaum
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada.,Department of Dentistry, Centre for Advanced Dental Research and Care, Mount Sinai Hospital, Toronto, ON, Canada.,Department of Dental Oncology, Maxillofacial and Ocular Prosthetics, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
15
|
Elashiry M, Elsayed R, Cutler CW. Exogenous and Endogenous Dendritic Cell-Derived Exosomes: Lessons Learned for Immunotherapy and Disease Pathogenesis. Cells 2021; 11:cells11010115. [PMID: 35011677 PMCID: PMC8750541 DOI: 10.3390/cells11010115] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
Immune therapeutic exosomes, derived exogenously from dendritic cells (DCs), the 'directors' of the immune response, are receiving favorable safety and tolerance profiles in phase I and II clinical trials for a growing number of inflammatory and neoplastic diseases. DC-derived exosomes (EXO), the focus of this review, can be custom tailored with immunoregulatory or immunostimulatory molecules for specific immune cell targeting. Moreover, the relative stability, small size and rapid uptake of EXO by recipient immune cells offer intriguing options for therapeutic purposes. This necessitates an in-depth understanding of mechanisms of EXO biogenesis, uptake and routing by recipient immune cells, as well as their in vivo biodistribution. Against this backdrop is recognition of endogenous exosomes, secreted by all cells, the molecular content of which is reflective of the metabolic state of these cells. In this regard, exosome biogenesis and secretion is regulated by cell stressors of chronic inflammation and tumorigenesis, including dysbiotic microbes, reactive oxygen species and DNA damage. Such cell stressors can promote premature senescence in young cells through the senescence associated secretory phenotype (SASP). Pathological exosomes of the SASP amplify inflammatory signaling in stressed cells in an autocrine fashion or promote inflammatory signaling to normal neighboring cells in paracrine, without the requirement of cell-to-cell contact. In summary, we review relevant lessons learned from the use of exogenous DC exosomes for immune therapy, as well as the pathogenic potential of endogenous DC exosomes.
Collapse
|
16
|
Hasegawa Y, Nagano K. Porphyromonas gingivalis FimA and Mfa1 fimbriae: Current insights on localization, function, biogenesis, and genotype. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:190-200. [PMID: 34691295 PMCID: PMC8512630 DOI: 10.1016/j.jdsr.2021.09.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/16/2022] Open
Abstract
In general, the periodontal pathogen Porphyromonas gingivalis expresses distinct FimA and Mfa1 fimbriae. Each of these consists of five FimA–E and five Mfa1–5 proteins encoded by the fim and mfa gene clusters, respectively. The main shaft portion comprises FimA and Mfa1, whereas FimB and Mfa2 are localized on the basal portion and function as anchors and elongation terminators. FimC–E and Mfa3–5 participate in the assembly of an accessory protein complex on the tips of each fimbria. Hence, they serve as ligands for the receptors on host cells and other oral bacterial species. The crystal structures of FimA and Mfa1 fimbrial proteins were recently elucidated and new insights into the localization, function, and biogenesis of these proteins have been reported. Several studies indicated a correlation between P. gingivalis pathogenicity and the fimA genotype but not the mfa1 genotype. We recently revealed polymorphisms of all genes in the fim and mfa gene clusters. Intriguingly, mfa5 occurred in numerous different forms and underwent duplication. Detailed structural and functional knowledge of the fimbrial proteins in the context of the entire filament could facilitate the development of innovative therapeutic strategies for structure-based drug design.
Collapse
Affiliation(s)
- Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Keiji Nagano
- Division of Microbiology, Department of Oral Biology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, Japan
| |
Collapse
|
17
|
Zenobia C, Herpoldt KL, Freire M. Is the oral microbiome a source to enhance mucosal immunity against infectious diseases? NPJ Vaccines 2021; 6:80. [PMID: 34078913 PMCID: PMC8172910 DOI: 10.1038/s41541-021-00341-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/24/2021] [Indexed: 12/14/2022] Open
Abstract
Mucosal tissues act as a barrier throughout the oral, nasopharyngeal, lung, and intestinal systems, offering first-line protection against potential pathogens. Conventionally, vaccines are applied parenterally to induce serotype-dependent humoral response but fail to drive adequate mucosal immune protection for viral infections such as influenza, HIV, and coronaviruses. Oral mucosa, however, provides a vast immune repertoire against specific microbial pathogens and yet is shaped by an ever-present microbiome community that has co-evolved with the host over thousands of years. Adjuvants targeting mucosal T-cells abundant in oral tissues can promote soluble-IgA (sIgA)-specific protection to confer increased vaccine efficacy. Th17 cells, for example, are at the center of cell-mediated immunity and evidence demonstrates that protection against heterologous pathogen serotypes is achieved with components from the oral microbiome. At the point of entry where pathogens are first encountered, typically the oral or nasal cavity, the mucosal surfaces are layered with bacterial cohabitants that continually shape the host immune profile. Constituents of the oral microbiome including their lipids, outer membrane vesicles, and specific proteins, have been found to modulate the Th17 response in the oral mucosa, playing important roles in vaccine and adjuvant designs. Currently, there are no approved adjuvants for the induction of Th17 protection, and it is critical that this research is included in the preparedness for the current and future pandemics. Here, we discuss the potential of oral commensals, and molecules derived thereof, to induce Th17 activity and provide safer and more predictable options in adjuvant engineering to prevent emerging infectious diseases.
Collapse
Affiliation(s)
| | | | - Marcelo Freire
- Departments of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, USA.
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
18
|
Elsayed R, Elashiry M, Liu Y, El-Awady A, Hamrick M, Cutler CW. Porphyromonas gingivalis Provokes Exosome Secretion and Paracrine Immune Senescence in Bystander Dendritic Cells. Front Cell Infect Microbiol 2021; 11:669989. [PMID: 34141629 PMCID: PMC8204290 DOI: 10.3389/fcimb.2021.669989] [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: 02/19/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Periodontitis is a disease of ageing or inflammaging, and is comorbid with other more severe age-related chronic diseases. With advanced age comes an increase in accumulation of senescent cells that release soluble and insoluble pro-inflammatory factors collectively termed the senescence associated secretory phenotype (SASP). In the present report, we examined whether immune cells typical of those at the oral mucosa-microbe interface, are vulnerable to cellular senescence (CS) and the role of dysbiotic oral pathogen Porphyromonas gingivalis. Bone marrow-derived dendritic cells (DCs) from young (yDCs) and old (oDCs) mice were co-cultured in vitro with CS inducer doxorubicin or P.gingivalis (Pg), plus or minus senolytic agent rapamycin. CS profiling revealed elevated CS mediators SA-β-Gal, p16 INK4A, p53, and p21Waf1/Clip1 in oDCs, or yDCs in response to doxorubicin or P. gingivalis, reversible with rapamycin. Functional studies indicate impaired maturation function of oDCs, and yDC exposed to P. gingivalis; moreover, OVA-driven proliferation of CD4+ T cells from young OTII transgenic mice was impaired by oDCs or yDCs+Pg. The SASP of DCs, consisting of secreted exosomes and inflammasome-related cytokines was further analyzed. Exosomes of DCs cocultured with P. gingivalis (PgDCexo) were purified, quantitated and characterized. Though typical in terms of size, shape and phenotype, PgDCexo were 2-fold greater in number than control DCs, with several important distinctions. Namely, PgDCexo were enriched in age-related miRNAs, and miRNAs reported to disrupt immune homeostasis through negative regulation of apoptosis and autophagy functions. We further show that PgDCexo were enriched in P. gingivalis fimbrial adhesin protein mfa1 and in inflammasome related cytokines IL-1β, TNFα and IL-6. Functionally PgDCexo were readily endocytosed by recipient yDCs, amplifying functional impairment in maturation and ability to promote Ova-driven proliferation of OTII CD4+ T cells from young mice. In conclusion P. gingivalis induces premature (autocrine) senescence in DCs by direct cellular invasion and greatly amplifies senescence, in paracrine, of bystander DCs by secretion of inflammatory exosomes. The implications of this pathological pathway for periodontal disease in vivo is under investigation in mouse models.
Collapse
Affiliation(s)
- Ranya Elsayed
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Mahmoud Elashiry
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA, United States
| | - Ahmed El-Awady
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Mark Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta, GA, United States
| | - Christopher W. Cutler
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, GA, United States,*Correspondence: Christopher W. Cutler,
| |
Collapse
|
19
|
Hajishengallis G, Lamont RJ. Polymicrobial communities in periodontal disease: Their quasi-organismal nature and dialogue with the host. Periodontol 2000 2021; 86:210-230. [PMID: 33690950 DOI: 10.1111/prd.12371] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/05/2020] [Accepted: 03/28/2020] [Indexed: 12/11/2022]
Abstract
In health, indigenous polymicrobial communities at mucosal surfaces maintain an ecological balance via both inter-microbial and host-microbial interactions that promote their own and the host's fitness, while preventing invasion by exogenous pathogens. However, genetic and acquired destabilizing factors (including immune deficiencies, immunoregulatory defects, smoking, diet, obesity, diabetes and other systemic diseases, and aging) may disrupt this homeostatic balance, leading to selective outgrowth of species with the potential for destructive inflammation. This process, known as dysbiosis, underlies the development of periodontitis in susceptible hosts. The pathogenic process is not linear but involves a positive-feedback loop between dysbiosis and the host inflammatory response. The dysbiotic community is essentially a quasi-organismal entity, where constituent organisms communicate via sophisticated physical and chemical signals and display functional specialization (eg, accessory pathogens, keystone pathogens, pathobionts), which enables polymicrobial synergy and dictates the community's pathogenic potential or nososymbiocity. In this review, we discuss early and recent studies in support of the polymicrobial synergy and dysbiosis model of periodontal disease pathogenesis. According to this concept, disease is not caused by individual "causative pathogens" but rather by reciprocally reinforced interactions between physically and metabolically integrated polymicrobial communities and a dysregulated host inflammatory response.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
20
|
Microbial Lipid A Remodeling Controls Cross-Presentation Efficiency and CD8 T Cell Priming by Modulating Dendritic Cell Function. Infect Immun 2021; 89:IAI.00335-20. [PMID: 33257533 DOI: 10.1128/iai.00335-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 11/10/2020] [Indexed: 12/18/2022] Open
Abstract
The majority of Gram-negative bacteria elicit a potent immune response via recognition of lipid A expressed on the outer bacterial membrane by the host immune receptor Toll-like receptor 4 (TLR4). However, some Gram-negative bacteria evade detection by TLR4 or alter the outcome of TLR4 signaling by modification of lipid A species. Although the role of lipid A modifications on host innate immunity has been examined in some detail, it is currently unclear how lipid A remodeling influences host adaptive immunity. One prototypic Gram-negative bacterium that modifies its lipid A structure is Porphyromonas gingivalis, an anaerobic pathobiont that colonizes the human periodontium and induces chronic low-grade inflammation that is associated with periodontal disease as well as a number of systemic inflammatory disorders. P. gingivalis produces dephosphorylated and deacylated lipid A structures displaying altered activities at TLR4. Here, we explored the functional role of P. gingivalis lipid A modifications on TLR4-dependent innate and adaptive immune responses in mouse bone marrow-derived dendritic cells (BMDCs). We discovered that lipid A 4'-phosphate removal is required for P. gingivalis to evade BMDC-dependent proinflammatory cytokine responses and markedly limits the bacterium's capacity to induce beta interferon (IFN-β) production. In addition, lipid A 4'-phosphatase activity prevents canonical bacterium-induced delay in antigen degradation, which leads to inefficient antigen cross-presentation and a failure to cross-prime CD8 T cells specific for a P. gingivalis-associated antigen. We propose that lipid A modifications produced by this bacterium alter host TLR4-dependent adaptive immunity to establish chronic infections associated with a number of systemic inflammatory disorders.
Collapse
|
21
|
Enterococcus faecalis Induces Differentiation of Immune-Aberrant Dendritic Cells from Murine Bone Marrow-Derived Stem Cells. Infect Immun 2020; 88:IAI.00338-20. [PMID: 32839187 DOI: 10.1128/iai.00338-20] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/19/2020] [Indexed: 12/13/2022] Open
Abstract
Enterococcus faecalis, long implicated in serious systemic infections and failure of root canal treatment, is a persistent inhabitant of oral periapical lesions. Dendritic cells (DCs) and other innate immune cells patrol the oral mucosa for infecting microbes. Dendritic cells are efficient at capturing microbes when immature, whereupon they can transform into potent antigen-presenting cells upon full maturation. Autophagy, a sophisticated intracellular process first described for elimination of damaged organelles, regulates DC maturation and other important immune functions of DCs. The present study examined how E. faecalis influences the differentiation of murine bone marrow-derived stem cells (BMSCs) into functional DCs in the presence of the cytokines granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). Although the viability and differentiation of DCs were not affected by E. faecalis, expression of the autophagy-related proteins ATG7, Beclin1, and LC3bI/II were significantly suppressed in an mTOR-dependent manner. Ultrastructurally, E. faecalis was identified in single-membrane vacuoles, some of which were in the process of binary fission. Bacterium-containing autophagosomes were absent within the cytoplasm. Accessory molecules (major histocompatibility complex class II [MHC-II], CD80, and CD86) and anti-inflammatory cytokine (transforming growth factor β1 [TGF-β1]) were suppressed in E. faecalis-induced DCs, while IL-1β, tumor necrosis factor alpha (TNF-α), and IL-12 levels were upregulated. When pulsed with ovalbumin (OVA), the E. faecalis-induced DCs showed reduction in CD4+ OVA-specific OT-II T cell proliferation. It is concluded that E. faecalis promotes the differentiation of bone marrow stem cells into CD11c-positive DCs with aberrant immune functions while retaining the capability of proinflammatory cytokine induction.
Collapse
|
22
|
Möller B, Kollert F, Sculean A, Villiger PM. Infectious Triggers in Periodontitis and the Gut in Rheumatoid Arthritis (RA): A Complex Story About Association and Causality. Front Immunol 2020; 11:1108. [PMID: 32582191 PMCID: PMC7283532 DOI: 10.3389/fimmu.2020.01108] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic immune mediated inflammatory disease of unknown origin, which is predominantly affecting the joints. Antibodies against citrullinated peptides are a rather specific immunological hallmark of this heterogeneous entity. Furthermore, certain sequences of the third hypervariable region of human leukocyte antigen (HLA)-DR class II major histocompatibility (MHC) molecules, the so called "shared epitope" sequences, appear to promote autoantibody positive types of RA. However, MHC-II molecule and other genetic associations with RA could not be linked to immune responses against specific citrullinated peptides, nor do genetic factors fully explain the origin of RA. Consequently, non-genetic factors must play an important role in the complex interaction of endogenous and exogenous disease factors. Tobacco smoking was the first environmental factor that was associated with onset and severity of RA. Notably, smoking is also an established risk factor for oral diseases. Furthermore, smoking is associated with extra-articular RA manifestations such as interstitial lung disease in anatomical proximity to the airway mucosa, but also with subcutaneous rheumatoid nodules. In the mouth, Porphyromonas gingivalis is a periodontal pathogen with unique citrullinating capacity of foreign microbial antigens as well as candidate RA autoantigens. Although the original hypothesis that this single pathogen is causative for RA remained unproven, epidemiological as well as experimental evidence linking periodontitis (PD) with RA is rapidly accumulating. Other periopathogens such as Aggregatibacter actinomycetemcomitans and Prevotella intermedia were also proposed to play a specific immunodominant role in context of RA. However, demonstration of T cell reactivity against citrullinated, MHC-II presented autoantigens from RA synovium coinciding with immunity against Prevotella copri (Pc.), a gut microbe attracted attention to another mucosal site, the intestine. Pc. was accumulated in the feces of clinically healthy subjects with citrulline directed immune responses and was correlated with RA onset. In conclusion, we retrieved more than one line of evidence for mucosal sites and different microbial taxa to be potentially involved in the development of RA. This review gives an overview of infectious agents and mucosal pathologies, and discusses the current evidence for causality between different exogenous or mucosal factors and systemic inflammation in RA.
Collapse
Affiliation(s)
- Burkhard Möller
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Florian Kollert
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Peter M Villiger
- Department for Rheumatology, Immunology and Allergology, Inselspital-University Hospital of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Meghil MM, Cutler CW. Oral Microbes and Mucosal Dendritic Cells, "Spark and Flame" of Local and Distant Inflammatory Diseases. Int J Mol Sci 2020; 21:E1643. [PMID: 32121251 PMCID: PMC7084622 DOI: 10.3390/ijms21051643] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/20/2022] Open
Abstract
Mucosal health and disease is mediated by a complex interplay between the microbiota ("spark") and the inflammatory response ("flame"). Pathobionts, a specific class of microbes, exemplified by the oral microbe Porphyromonas gingivalis, live mostly "under the radar" in their human hosts, in a cooperative relationship with the indigenous microbiota. Dendritic cells (DCs), mucosal immune sentinels, often remain undisturbed by such microbes and do not alert adaptive immunity to danger. At a certain tipping point of inflammation, an "awakening" of pathobionts occurs, wherein their active growth and virulence are stimulated, leading to a dysbiosis. Pathobiont becomes pathogen, and commensal becomes accessory pathogen. The local inflammatory outcome is the Th17-mediated degenerative bone disease, periodontitis (PD). In systemic circulation of PD subjects, inflammatory DCs expand, carrying an oral microbiome and promoting Treg and Th17 responses. At distant peripheral sites, comorbid diseases including atherosclerosis, Alzheimer's disease, macular degeneration, chronic kidney disease, and others are reportedly induced. This review will review the immunobiology of DCs, examine the complex interplay of microbes and DCs in the pathogenesis of PD and its comorbid inflammatory diseases, and discuss the role of apoptosis and autophagy in this regard. Overall, the pathophysiological mechanisms of DC-mediated chronic inflammation and tissue destruction will be summarized.
Collapse
Affiliation(s)
| | - Christopher W. Cutler
- Department of Periodontics, The Dental College of Georgia at Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
24
|
Abstract
The etiopathogenesis of severe periodontitis includes herpesvirus-bacteria coinfection. This article evaluates the pathogenicity of herpesviruses (cytomegalovirus and Epstein-Barr virus) and periodontopathic bacteria (Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis) and coinfection of these infectious agents in the initiation and progression of periodontitis. Cytomegalovirus and A. actinomycetemcomitans/P. gingivalis exercise synergistic pathogenicity in the development of localized ("aggressive") juvenile periodontitis. Cytomegalovirus and Epstein-Barr virus are associated with P. gingivalis in adult types of periodontitis. Periodontal herpesviruses that enter the general circulation may also contribute to disease development in various organ systems. A 2-way interaction is likely to occur between periodontal herpesviruses and periodontopathic bacteria, with herpesviruses promoting bacterial upgrowth, and bacterial factors reactivating latent herpesviruses. Bacterial-induced gingivitis may facilitate herpesvirus colonization of the periodontium, and herpesvirus infections may impede the antibacterial host defense and alter periodontal cells to predispose for bacterial adherence and invasion. Herpesvirus-bacteria synergistic interactions, are likely to comprise an important pathogenic determinant of aggressive periodontitis. However, mechanistic investigations into the molecular and cellular interaction between periodontal herpesviruses and bacteria are still scarce. Herpesvirus-bacteria coinfection studies may yield significant new discoveries of pathogenic determinants, and drug and vaccine targets to minimize or prevent periodontitis and periodontitis-related systemic diseases.
Collapse
Affiliation(s)
- Casey Chen
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Pinghui Feng
- Section of Infection and Immunity, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Jørgen Slots
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
25
|
Meghil MM, Tawfik OK, Elashiry M, Rajendran M, Arce RM, Fulton DJ, Schoenlein PV, Cutler CW. Disruption of Immune Homeostasis in Human Dendritic Cells via Regulation of Autophagy and Apoptosis by Porphyromonas gingivalis. Front Immunol 2019; 10:2286. [PMID: 31608069 PMCID: PMC6769118 DOI: 10.3389/fimmu.2019.02286] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
As fundamental processes of immune homeostasis, autophagy, and apoptosis must be maintained to mitigate risk of chronic inflammation and autoimmune diseases. Periodontitis is a chronic inflammatory disease characterized by oral microbial dysbiosis, and dysregulation of dendritic cell (DC) and T cell responses. The aim of this study was to elucidate the underlying mechanisms by which the oral microbe Porphyromonas gingivalis (P. gingivalis) manipulates dendritic cell signaling to perturb both autophagy and apoptosis. Using a combination of Western blotting, flow cytometry, qRT-PCR and immunofluorescence analysis, we show a pivotal role for the minor (Mfa1) fimbriae of P. gingivalis in nuclear/cytoplasmic shuttling of Akt and FOXO1 in human monocyte-derived DCs. Mfa1-induced Akt nuclear localization and activation ultimately induced mTOR. Activation of the Akt/mTOR axis downregulated intracellular LC3II, also known as Atg8, required for autophagosome formation and maturation. Use of allosteric panAkt inhibitor MK2206 and mTOR inhibitor rapamycin confirmed the role of Akt/mTOR signaling in autophagy inhibition by P. gingivalis in DCs. Interestingly, this pathway was also linked to induction of the anti-apoptotic protein Bcl2, decreased caspase-3 cleavage and decreased expression of pro-apoptotic proteins Bax and Bim, thus promoting longevity of host DCs. Addition of ABT-199 peptide to disrupt the interaction of antiapoptotic Bcl2 and its proapoptotic partners BAK/BAX restored apoptotic death to P. gingivalis-infected DC cells. In summary, we have identified the underlying mechanism by which P. gingivalis promotes its own survival and that of its host DCs.
Collapse
Affiliation(s)
- Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Omnia K Tawfik
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Mahmoud Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States.,Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Mythilypriya Rajendran
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - Roger M Arce
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| | - David J Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Patricia V Schoenlein
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
26
|
Rabelo MDS, El-Awady A, Moura Foz A, Hisse Gomes G, Rajendran M, Meghil MM, Lowry S, Romito GA, Cutler CW, Susin C. Influence of T2DM and prediabetes on blood DC subsets and function in subjects with periodontitis. Oral Dis 2019; 25:2020-2029. [PMID: 31541516 DOI: 10.1111/odi.13200] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 09/06/2019] [Accepted: 09/13/2019] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To compare the myeloid and plasmacytoid DC counts and maturation status among subjects with/without generalized periodontitis (GP) and type 2 diabetes mellitus (T2DM). METHODS The frequency and maturation status of myeloid and plasmacytoid blood DCs were analyzed by flow cytometry in four groups of 15 subjects: healthy controls, T2DM with generalized CP (T2DM + GP), prediabetes with GP (PD + GP), and normoglycemics with GP (NG + GP). RT-PCR was used to determine levels of Porphyromonas gingivalis in the oral biofilms and within panDCs. The role of exogenous glucose effects on differentiation and apoptosis of healthy human MoDCs was explored in vitro. RESULTS Relative to controls and to NG + GP, T2DM + GP showed significantly lower CD1c + and CD303 + DC counts, while CD141 + DCs were lower in T2DM + GP relative to controls. Blood DC maturation required for mobilization and immune responsiveness was not observed. A statistically significant trend was observed for P. gingivalis levels in the biofilms of groups as follows: controls <NG+GP < PD+GP < T2DM+GP. Moreover, significantly higher P. gingivalis levels were observed in blood DCs of NG + GP than controls, whereas no differences were observed between controls and PD + GP/T2DM + GP. In vitro differentiation of MoDCs was significantly decreased, and apoptosis was increased by physiologically relevant glucose levels. CONCLUSION Type 2 diabetes mellitus appears to inhibit important DC immune homeostatic functions, including expansion and bacterial scavenging, which might be mediated by hyperglycemia.
Collapse
Affiliation(s)
- Mariana de Sousa Rabelo
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil.,Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Ahmed El-Awady
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Adriana Moura Foz
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Giovane Hisse Gomes
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Mythilpriya Rajendran
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Mohamed M Meghil
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Scott Lowry
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Giuseppe Alexandre Romito
- Division of Periodontics, Department of Stomatology, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Christopher W Cutler
- Department of Periodontics, The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Cristiano Susin
- Department of Periodontology, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
27
|
Rajendran M, Looney S, Singh N, Elashiry M, Meghil MM, El-Awady AR, Tawfik O, Susin C, Arce RM, Cutler CW. Systemic Antibiotic Therapy Reduces Circulating Inflammatory Dendritic Cells and Treg-Th17 Plasticity in Periodontitis. THE JOURNAL OF IMMUNOLOGY 2019; 202:2690-2699. [PMID: 30944162 DOI: 10.4049/jimmunol.1900046] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/05/2019] [Indexed: 01/08/2023]
Abstract
Periodontitis (PD) is a common dysbiotic inflammatory disease that leads to local bone deterioration and tooth loss. PD patients experience low-grade bacteremias with oral microbes implicated in the risk of heart disease, cancer, and kidney failure. Although Th17 effectors are vital to fighting infection, functional imbalance of Th17 effectors and regulatory T cells (Tregs) promote inflammatory diseases. In this study, we investigated, in a small pilot randomized clinical trial, whether expansion of inflammatory blood myeloid dendritic cells (DCs) and conversion of Tregs to Th17 cells could be modulated with antibiotics (AB) as part of initial therapy in PD patients. PD patients were randomly assigned to either 7 d of peroral metronidazole/amoxicillin AB treatment or no AB, along with standard care debridement and chlorhexidine mouthwash. 16s ribosomal RNA analysis of keystone pathogen Porphyromonas gingivalis and its consortium members Fusobacterium nucleatum and Streptococcus gordonii confirmed the presence of all three species in the reservoirs (subgingival pockets and blood DCs) of PD patients before treatment. Of the three species, P. gingivalis was reduced in both reservoirs 4-6 wk after therapy. Further, the frequency of CD1C+CCR6+ myeloid DCs and IL-1R1 expression on IL-17A+FOXP3+CD4+ T cells in PD patients were reduced to healthy control levels. The latter led to decreased IL-1β-stimulated Treg plasticity in PD patients and improvement in clinical measures of PD. Overall, we identified an important, albeit short-term, beneficial role of AB therapy in reducing inflammatory DCs and Treg-Th17 plasticity in humans with PD.
Collapse
Affiliation(s)
- Mythilypriya Rajendran
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Stephen Looney
- Department of Biostatistics and Epidemiology, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Nagendra Singh
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912.,Cancer Research Center, Augusta University, Augusta, GA 30912
| | - Mahmoud Elashiry
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Ahmed R El-Awady
- Department of Research, Immunology Program, Children's Cancer Hospital, Cairo 57357, Egypt
| | - Omnia Tawfik
- Department of Oral Medicine and Periodontology, Cairo University, Cairo 12613, Egypt; and
| | - Cristiano Susin
- Department of Periodontology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Roger M Arce
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA 30912
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia at Augusta University, Augusta, GA 30912;
| |
Collapse
|
28
|
Polymicrobial synergy within oral biofilm promotes invasion of dendritic cells and survival of consortia members. NPJ Biofilms Microbiomes 2019; 5:11. [PMID: 32179736 PMCID: PMC6423025 DOI: 10.1038/s41522-019-0084-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Years of human microbiome research have confirmed that microbes rarely live or function alone, favoring diverse communities. Yet most experimental host-pathogen studies employ single species models of infection. Here, the influence of three-species oral microbial consortium on growth, virulence, invasion and persistence in dendritic cells (DCs) was examined experimentally in human monocyte-derived dendritic cells (DCs) and in patients with periodontitis (PD). Cooperative biofilm formation by Streptococcus gordonii, Fusobacterium nucleatum and Porphyromonas gingivalis was documented in vitro using growth models and scanning electron microscopy. Analysis of growth rates by species-specific 16s rRNA probes revealed distinct, early advantages to consortium growth for S. gordonii and F. nucleatum with P. gingivalis, while P. gingivalis upregulated its short mfa1 fimbriae, leading to increased invasion of DCs. F. nucleatum was only taken up by DCs when in consortium with P. gingivalis. Mature consortium regressed DC maturation upon uptake, as determined by flow cytometry. Analysis of dental plaques of PD and healthy subjects by 16s rRNA confirmed oral colonization with consortium members, but DC hematogenous spread was limited to P. gingivalis and F. nucleatum. Expression of P. gingivalis mfa1 fimbriae was increased in dental plaques and hematogenous DCs of PD patients. P. gingivalis in the consortium correlated with an adverse clinical response in the gingiva of PD subjects. In conclusion, we have identified polymicrobial synergy in a three-species oral consortium that may have negative consequences for the host, including microbial dissemination and adverse peripheral inflammatory responses.
Collapse
|
29
|
Peptide-Based Inhibitors of Fimbrial Biogenesis in Porphyromonas gingivalis. Infect Immun 2019; 87:IAI.00750-18. [PMID: 30642895 DOI: 10.1128/iai.00750-18] [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: 10/01/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Periodontitis is a progressive inflammatory disease that affects roughly half of American adults. Colonization of the oral cavity by the Gram-negative bacterial pathogen Porphyromonas gingivalis is a key event in the initiation and development of periodontal disease. Adhesive surface structures termed fimbriae (pili) mediate interactions of P. gingivalis with other bacteria and with host cells throughout the course of disease. The P. gingivalis fimbriae are assembled via a novel mechanism that involves proteolytic processing of lipidated precursor subunits and their subsequent polymerization on the bacterial surface. Given their extracellular assembly mechanism and central roles in pathogenesis, the P. gingivalis fimbriae are attractive targets for anti-infective therapeutics to prevent or treat periodontal disease. Here we confirm that conserved sequences in the N and C termini of the Mfa1 fimbrial subunit protein perform critical roles in subunit polymerization. We show that treatment of P. gingivalis with peptides corresponding to the conserved C-terminal region inhibits the extracellular assembly of Mfa fimbriae on the bacterial surface. We also show that peptide treatment interferes with the function of Mfa fimbriae by reducing P. gingivalis adhesion to Streptococcus gordonii in a dual-species biofilm model. Finally, we show that treatment of bacteria with similar peptides inhibits extracellular polymerization of the Fim fimbriae, which are also expressed by P. gingivalis These results support a donor strand-based assembly mechanism for the P. gingivalis fimbriae and demonstrate the feasibility of using extracellular peptides to disrupt the biogenesis and function of these critical periodontal disease virulence factors.
Collapse
|
30
|
Arjunan P, Meghil MM, Pi W, Xu J, Lang L, El-Awady A, Sullivan W, Rajendran M, Rabelo MS, Wang T, Tawfik OK, Kunde-Ramamoorthy G, Singh N, Muthusamy T, Susin C, Teng Y, Arce RM, Cutler CW. Oral Pathobiont Activates Anti-Apoptotic Pathway, Promoting both Immune Suppression and Oncogenic Cell Proliferation. Sci Rep 2018; 8:16607. [PMID: 30413788 PMCID: PMC6226501 DOI: 10.1038/s41598-018-35126-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/30/2018] [Indexed: 12/25/2022] Open
Abstract
Chronic periodontitis (CP) is a microbial dysbiotic disease linked to increased risk of oral squamous cell carcinomas (OSCCs). To address the underlying mechanisms, mouse and human cell infection models and human biopsy samples were employed. We show that the ‘keystone’ pathogen Porphyromonas gingivalis, disrupts immune surveillance by generating myeloid-derived dendritic suppressor cells (MDDSCs) from monocytes. MDDSCs inhibit CTLs and induce FOXP3 + Tregs through an anti-apoptotic pathway. This pathway, involving pAKT1, pFOXO1, FOXP3, IDO1 and BIM, is activated in humans with CP and in mice orally infected with Mfa1 expressing P. gingivalis strains. Mechanistically, activation of this pathway, demonstrating FOXP3 as a direct FOXO1-target gene, was demonstrated by ChIP-assay in human CP gingiva. Expression of oncogenic but not tumor suppressor markers is consistent with tumor cell proliferation demonstrated in OSCC-P. gingivalis cocultures. Importantly, FimA + P. gingivalis strain MFI invades OSCCs, inducing inflammatory/angiogenic/oncogenic proteins stimulating OSCCs proliferation through CXCR4. Inhibition of CXCR4 abolished Pg-MFI-induced OSCCs proliferation and reduced expression of oncogenic proteins SDF-1/CXCR4, plus pAKT1-pFOXO1. Conclusively, P. gingivalis, through Mfa1 and FimA fimbriae, promotes immunosuppression and oncogenic cell proliferation, respectively, through a two-hit receptor-ligand process involving DC-SIGN+hi/CXCR4+hi, activating a pAKT+hipFOXO1+hiBIM−lowFOXP3+hi and IDO+hi- driven pathway, likely to impact the prognosis of oral cancers in patients with periodontitis.
Collapse
Affiliation(s)
- Pachiappan Arjunan
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.
| | - Mohamed M Meghil
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.,Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Wenhu Pi
- Department of Radiation Oncology, Indiana University, Indianapolis, Indiana, United States of America
| | - Jinxian Xu
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Liwei Lang
- Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Ahmed El-Awady
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - William Sullivan
- Department of Energy, Joint Genome Institute, California, United States of America
| | - Mythilypriya Rajendran
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Mariana Sousa Rabelo
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.,Department of Periodontics, University of São Paulo, Sao Paulo, Brazil
| | - Tong Wang
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Omnia K Tawfik
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | | | - Nagendra Singh
- Department of Biochemistry & Molecular Biology, Cancer Research Center, Augusta University, Augusta, Georgia, United States of America
| | - Thangaraju Muthusamy
- Department of Biochemistry & Molecular Biology, Cancer Research Center, Augusta University, Augusta, Georgia, United States of America
| | - Cristiano Susin
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, Georgia, United States of America
| | - Roger M Arce
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Christopher W Cutler
- Department of Periodontics, Dental College of Georgia, Augusta University, Augusta, Georgia, United States of America.
| |
Collapse
|
31
|
Lee JY, Miller DP, Wu L, Casella CR, Hasegawa Y, Lamont RJ. Maturation of the Mfa1 Fimbriae in the Oral Pathogen Porphyromonas gingivalis. Front Cell Infect Microbiol 2018; 8:137. [PMID: 29868494 PMCID: PMC5954841 DOI: 10.3389/fcimb.2018.00137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
The Mfa1 fimbriae of the periodontal pathogen Porphyromonas gingivalis are involved in adhesion, including binding to synergistic species in oral biofilms. Mfa1 fimbriae are comprised of 5 proteins: the structural component Mfa1, the anchor Mfa2, and Mfa3-5 which constitute the fimbrial tip complex. Interactions among the Mfa proteins and the polymerization mechanism for Mfa1 are poorly understood. Here we show that Mfa3 can bind to Mfa1, 2, 4, and 5 in vitro, and may function as an adaptor protein interlinking other fimbrial subunits. Polymerization of Mfa1 is independent of Mfa3-5 and requires proteolytic processing mediated by the RgpA/B arginine gingipains of P. gingivalis. Both the N- and C- terminal regions of Mfa1 are necessary for polymerization; however, potential β-strand disrupting amino acid substitutions in these regions do not impair Mfa1 polymerization. In contrast, substitution of hydrophobic amino acids with charged residues in either the N- or C- terminal domains yielded Mfa1 proteins that failed to polymerize. Collectively, these results indicate that Mfa3 serves as an adaptor protein between Mfa1 and other accessory fimbrial proteins. Mfa1 fimbrial polymerization is dependent on hydrophobicity in both the N- and C-terminal regions, indicative of an assembly mechanism involving the terminal regions forming a hydrophobic binding interface between Mfa1 subunits.
Collapse
Affiliation(s)
- Jae Y Lee
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, United States
| | - Daniel P Miller
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, United States
| | - Leng Wu
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, United States
| | - Carolyn R Casella
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| | - Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, United States
| |
Collapse
|
32
|
Fleetwood AJ, Lee MKS, Singleton W, Achuthan A, Lee MC, O'Brien-Simpson NM, Cook AD, Murphy AJ, Dashper SG, Reynolds EC, Hamilton JA. Metabolic Remodeling, Inflammasome Activation, and Pyroptosis in Macrophages Stimulated by Porphyromonas gingivalis and Its Outer Membrane Vesicles. Front Cell Infect Microbiol 2017; 7:351. [PMID: 28824884 PMCID: PMC5543041 DOI: 10.3389/fcimb.2017.00351] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/21/2017] [Indexed: 12/19/2022] Open
Abstract
Porphyromonas gingivalis is one of the bacterial species most closely associated with periodontitis and can shed large numbers of outer membrane vesicles (OMVs), which are increasingly thought to play a significant role in bacterial virulence and pathogenicity. Macrophages are amongst the first immune cells to respond to bacteria and their products, so we sought to directly compare the response of macrophages to P. gingivalis or its purified OMVs. Macrophages stimulated with OMVs produced large amounts of TNFα, IL-12p70, IL-6, IL-10, IFNβ, and nitric oxide compared to cells infected with P. gingivalis, which produced very low levels of these mediators. Both P. gingivalis and OMVs induced a shift in macrophage metabolism from oxidative phosphorylation (OXPHOS) to glycolysis, which was supported by enhanced lactate release, decreased mitochondrial oxygen consumption with reduced spare respiratory capacity, as well as increased mitochondrial reactive oxygen species (ROS) production. Corresponding to this metabolic shift, gene expression analysis of macrophages infected with P. gingivalis or stimulated with OMVs revealed a broad transcriptional upregulation of genes critical to glycolysis and a downregulation of genes associated with the TCA cycle. Upon examination of inflammasome signaling and pyroptosis it was found that P. gingivalis did not activate the inflammasome in macrophages as the mature forms of caspase-1, IL-1β, and IL-18 were not detected and there was no extracellular release of lactate dehydrogenase (LDH) or 7-AAD staining. In comparison, macrophages stimulated with OMVs potently activated caspase-1, produced large amounts of IL-1β, IL-18, released LDH, and were positive for 7-AAD indicative of pyroptotic cell death. These data directly quantitate the distinct effects of P. gingivalis and its OMVs on macrophage inflammatory phenotype, mitochondrial function, inflammasome activation, and pyroptotic cell death that may have potential implications for their roles in chronic periodontitis.
Collapse
Affiliation(s)
- Andrew J Fleetwood
- Department of Medicine, University of Melbourne, Royal Melbourne HospitalParkville, VIC, Australia
| | - Man K S Lee
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes InstituteMelbourne, VIC, Australia
| | - William Singleton
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, University of MelbourneVIC, Australia
| | - Adrian Achuthan
- Department of Medicine, University of Melbourne, Royal Melbourne HospitalParkville, VIC, Australia
| | - Ming-Chin Lee
- Department of Medicine, University of Melbourne, Royal Melbourne HospitalParkville, VIC, Australia
| | - Neil M O'Brien-Simpson
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, University of MelbourneVIC, Australia
| | - Andrew D Cook
- Department of Medicine, University of Melbourne, Royal Melbourne HospitalParkville, VIC, Australia
| | - Andrew J Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes InstituteMelbourne, VIC, Australia
| | - Stuart G Dashper
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, University of MelbourneVIC, Australia
| | - Eric C Reynolds
- Oral Health Cooperative Research Centre, Melbourne Dental School, Bio21 Institute, University of MelbourneVIC, Australia
| | - John A Hamilton
- Department of Medicine, University of Melbourne, Royal Melbourne HospitalParkville, VIC, Australia
| |
Collapse
|
33
|
Ebersole JL, Dawson D, Emecen-Huja P, Nagarajan R, Howard K, Grady ME, Thompson K, Peyyala R, Al-Attar A, Lethbridge K, Kirakodu S, Gonzalez OA. The periodontal war: microbes and immunity. Periodontol 2000 2017; 75:52-115. [DOI: 10.1111/prd.12222] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Glowczyk I, Wong A, Potempa B, Babyak O, Lech M, Lamont RJ, Potempa J, Koziel J. Inactive Gingipains from P. gingivalis Selectively Skews T Cells toward a Th17 Phenotype in an IL-6 Dependent Manner. Front Cell Infect Microbiol 2017; 7:140. [PMID: 28497028 PMCID: PMC5406403 DOI: 10.3389/fcimb.2017.00140] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/05/2017] [Indexed: 01/05/2023] Open
Abstract
Gingipain cysteine proteases are considered key virulence factors of Porphyromonas gingivalis. They significantly influence antibacterial and homeostatic functions of macrophages, neutrophils, the complement system, and cytokine networks. Recent data indicate the role of P. gingivalis in T cell differentiation; however, the involvement of gingipains in this process remains elusive. Therefore, the aim of this study was to investigate the contribution of danger signals triggered by the gingipains on the generation of Th17 cells, which play a key role in protection against bacterial diseases but may cause chronic inflammation and bone resorption. To this end we compared the effects of the wild-type strain of P. gingivalis (W83) with its isogenic mutant devoid of gingipain activity (ΔKΔRAB), and bacterial cells pretreated with a highly-specific inhibitor of gingipains activity (KYTs). Antigen presenting cells (APCs), both professional (dendritic cells), and non-professional (gingival keratinocytes), exposed to viable bacteria expressed high amounts of cytokines (IL-6, IL-21, IL-23). These cytokines are reported to either stimulate or balance the Th17-dependent immune response. Surprisingly, cells infected with P. gingivalis devoid of gingipain activity showed increased levels of all tested cytokines compared to bacteria with fully active enzymes. The effect was dependent on both the reduction of cytokine proteolysis and the lack of cross-talk with other bacterial virulence factors, including LPS and fimbriae that induce de novo synthesis of cytokines. The profile of lymphocyte T differentiation from naive T cells showed enhanced generation of Th17 in response to bacteria with inactive gingipains. Moreover, we found that gingipain-dependent induction of Th17 cells was highly specific, since other T cell-subsets remained unchanged. Finally, inhibition of IL-6 signaling in dendritic cells led to a significant depletion of the Th17 population. Cumulatively, this study revealed a previously undisclosed role of gingipain activity in the process of Th17 differentiation reliant on blocking signaling through IL-6. Since inactivation of gingipains accelerates the skewing of T cells toward Th17 cells, which are detrimental in periodontitis, IL-6 signaling may serve as an attractive target for treatment of the disease.
Collapse
Affiliation(s)
- Izabela Glowczyk
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Alicia Wong
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Barbara Potempa
- Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of LouisvilleLouisville, KY, USA
| | - Olena Babyak
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland
| | - Maciej Lech
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland.,Department of Nephrology, Klinikum der Ludwig-Maximilians-Universität München, Medizinische Klinik und Poliklinik IVMunich, Germany
| | - Richard J Lamont
- Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of LouisvilleLouisville, KY, USA
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland.,Center for Oral Health and Systemic Disease, University of Louisville School of Dentistry, University of LouisvilleLouisville, KY, USA
| | - Joanna Koziel
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian UniversityKrakow, Poland
| |
Collapse
|
35
|
Tyagi RK, Miles B, Parmar R, Garg NK, Dalai SK, Baban B, Cutler CW. Human IDO-competent, long-lived immunoregulatory dendritic cells induced by intracellular pathogen, and their fate in humanized mice. Sci Rep 2017; 7:41083. [PMID: 28198424 PMCID: PMC5309771 DOI: 10.1038/srep41083] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 12/08/2016] [Indexed: 02/06/2023] Open
Abstract
Targeting of myeloid-dendritic cell receptor DC-SIGN by numerous chronic infectious agents, including Porphyromonas gingivalis, is shown to drive-differentiation of monocytes into dysfunctional mDCs. These mDCs exhibit alterations of their fine-tuned homeostatic function and contribute to dysregulated immune-responses. Here, we utilize P. gingivalis mutant strains to show that pathogen-differentiated mDCs from primary human-monocytes display anti-apoptotic profile, exhibited by elevated phosphorylated-Foxo1, phosphorylated-Akt1, and decreased Bim-expression. This results in an overall inhibition of DC-apoptosis. Direct stimulation of complex component CD40 on DCs leads to activation of Akt1, suggesting CD40 involvement in anti-apoptotic effects observed. Further, these DCs drove dampened CD8+ T-cell and Th1/Th17 effector-responses while inducing CD25+Foxp3+CD127- Tregs. In vitro Treg induction was mediated by DC expression of indoleamine 2,3-dioxygenase, and was confirmed in IDO-KO mouse model. Pathogen-infected &CMFDA-labeled MoDCs long-lasting survival was confirmed in a huMoDC reconstituted humanized mice. In conclusion, our data implicate PDDCs as an important target for resolution of chronic infection.
Collapse
MESH Headings
- Animals
- Apoptosis
- Bcl-2-Like Protein 11/metabolism
- CD40 Antigens/metabolism
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation
- Cytokines/metabolism
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/microbiology
- Forkhead Box Protein O1/metabolism
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/cytology
- Monocytes/metabolism
- Porphyromonas gingivalis/genetics
- Porphyromonas gingivalis/pathogenicity
- Proto-Oncogene Proteins c-akt/metabolism
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/cytology
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Rajeev K. Tyagi
- Department of Periodontics, College of Dental Medicine, Georgia Regents University, Augusta, GA 30912, USA
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad 382481, Gujarat, India
| | - Brodie Miles
- Division of Infectious Diseases, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA
| | - Rajesh Parmar
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad 382481, Gujarat, India
| | - Neeraj K. Garg
- Drug Delivery Research Group, University Institute of Pharmaceutical Sciences, UGC center of Advanced Studies, Panjab University, Chandigarh, India
| | - Sarat K. Dalai
- Institute of Science, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad 382481, Gujarat, India
| | - Babak Baban
- Department of Oral Biology, Georgia Regents University, Augusta, GA 30912, USA
| | - Christopher W. Cutler
- Department of Periodontics, College of Dental Medicine, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
36
|
El-Awady AR, Arce RM, Cutler CW. Dendritic cells: microbial clearance via autophagy and potential immunobiological consequences for periodontal disease. Periodontol 2000 2017; 69:160-80. [PMID: 26252408 PMCID: PMC4530502 DOI: 10.1111/prd.12096] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2015] [Indexed: 12/15/2022]
Abstract
Dendritic cells are potent antigen‐capture and antigen‐presenting cells that play a key role in the initiation and regulation of the adaptive immune response. This process of immune homeostasis, as maintained by dendritic cells, is susceptible to dysregulation by certain pathogens during chronic infections. Such dysregulation may lead to disease perpetuation with potentially severe systemic consequences. Here we discuss in detail how intracellular pathogens exploit dendritic cells and escape degradation by altering or evading autophagy. This novel mechanism explains, in part, the chronic, persistent nature observed in several immuno‐inflammatory diseases, including periodontal disease. We also propose a hypothetical model of the plausible role of autophagy in the context of periodontal disease. Promotion of autophagy may open new therapeutic strategies in the search of a ‘cure’ for periodontal disease in humans.
Collapse
|
37
|
Horie T, Inomata M, Into T, Hasegawa Y, Kitai N, Yoshimura F, Murakami Y. Identification of OmpA-Like Protein of Tannerella forsythia as an O-Linked Glycoprotein and Its Binding Capability to Lectins. PLoS One 2016; 11:e0163974. [PMID: 27711121 PMCID: PMC5053532 DOI: 10.1371/journal.pone.0163974] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/16/2016] [Indexed: 11/22/2022] Open
Abstract
Bacterial glycoproteins are associated with physiological and pathogenic functions of bacteria. It remains unclear whether bacterial glycoproteins can bind to specific classes of lectins expressed on host cells. Tannerella forsythia is a gram-negative oral anaerobe that contributes to the development of periodontitis. In this study, we aimed to find lectin-binding glycoproteins in T. forsythia. We performed affinity chromatography of wheat germ agglutinin, which binds to N-acetylglucosamine (GlcNAc) and sialic acid (Sia), and identified OmpA-like protein as the glycoprotein that has the highest affinity. Mass spectrometry revealed that OmpA-like protein contains O-type N-acetylhexosamine and hexose. Fluorometry quantitatively showed that OmpA-like protein contains Sia. OmpA-like protein was found to bind to lectins including E-selectin, P-selectin, L-selectin, Siglec-5, Siglec-9, Siglec-10, and DC-SIGN. The binding of OmpA-like protein to these lectins, except for the Siglecs, depends on the presence of calcium. N-acetylneuraminic acid (NeuAc), which is the most abundant Sia, inhibited the binding of OmpA-like protein to all of these lectins, whereas GlcNAc and mannose only inhibited the binding to DC-SIGN. We further found that T. forsythia adhered to human oral epithelial cells, which express E-selectin and P-selectin, and that this adhesion was inhibited by addition of NeuAc. Moreover, adhesion of an OmpA-like protein-deficient T. forsythia strain to the cells was reduced compared to that of the wild-type strain. Our findings indicate that OmpA-like protein of T. forsythia contains O-linked sugar chains that can mediate interactions with specific lectins. This interaction is suggested to facilitate adhesion of T. forsythia to the surface of host cells.
Collapse
Affiliation(s)
- Toshi Horie
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, Japan
- Department of Orthodontics, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Megumi Inomata
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, Japan
- * E-mail:
| | - Takeshi Into
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Yoshiaki Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - Noriyuki Kitai
- Department of Orthodontics, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Fuminobu Yoshimura
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - Yukitaka Murakami
- Department of Oral Microbiology, Division of Oral Infections and Health Sciences, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| |
Collapse
|
38
|
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
|
39
|
Hajishengallis G, Krauss JL, Jotwani R, Lambris JD. Differential capacity for complement receptor-mediated immune evasion by Porphyromonas gingivalis depending on the type of innate leukocyte. Mol Oral Microbiol 2016; 32:154-165. [PMID: 27081768 DOI: 10.1111/omi.12161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2016] [Indexed: 01/02/2023]
Abstract
The complement system plays a central role in immunity and inflammation, although certain pathogens can exploit complement to undermine protective immunity. In this context, the periodontal keystone pathogen Porphyromonas gingivalis was previously shown by our group to evade killing by neutrophils or macrophages through exploitation of complement C5a receptor 1 (C5aR1) and complement receptor 3 (CR3). Here, we examined whether P. gingivalis uses complement receptors to also subvert killing by dendritic cells. In line with earlier independent studies, intracellular viable P. gingivalis bacteria could be recovered from mouse bone-marrow-derived dendritic cells (BMDC) or human monocyte-derived dendritic cells (MDDC) exposed to the pathogen. However, in the presence of C5a, the intracellular survival of P. gingivalis was significantly decreased in a C5aR1-dependent way. Further work using wild-type and receptor-knockout BMDC showed that, in the presence of C3a, the C3a receptor (C3aR) similarly enhanced the intracellular killing of P. gingivalis. In contrast, C5aR2, an alternative receptor for C5a (G protein-coupled receptor 77), was associated with increased intracellular P. gingivalis viable counts, consistent with the notion that C5aR2 functions as a negative regulator of C5aR1 activity. Moreover, P. gingivalis failed to use CR3 as a phagocytic receptor in BMDC, in contrast to our earlier findings in macrophages where CR3-mediated uptake promotes P. gingivalis survival. Collectively, these data show that complement receptors mediate cell-type-specific effects on how innate leukocytes handle P. gingivalis, which appears to exploit complement to preferentially evade those cells (neutrophils and macrophages) that are most often encountered in its predominant niche, the periodontal pocket.
Collapse
Affiliation(s)
- G Hajishengallis
- Department of Microbiology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - J L Krauss
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - R Jotwani
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - J D Lambris
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
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
|
41
|
Zenobia C, Hajishengallis G. Porphyromonas gingivalis virulence factors involved in subversion of leukocytes and microbial dysbiosis. Virulence 2016; 6:236-43. [PMID: 25654623 PMCID: PMC4601496 DOI: 10.1080/21505594.2014.999567] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The oral bacterium Porphyromonas gingivalis has special nutrient requirements due to its asaccharolytic nature subsisting on small peptides cleaved from host proteins. Using proteases and other virulence factors, P. gingivalis thrives as a component of a polymicrobial community in nutritionally favorable inflammatory environments. In this regard, P. gingivalis has a number of strategies that subvert the host immune response in ways that promote its colonization and facilitate the outgrowth of the surrounding microbial community. The focus of this review is to discuss at the molecular level how P. gingivalis subverts leukocytes to create a favorable environment for a select community of bacteria that, in turn, adversely affects the periodontal tissues.
Collapse
Affiliation(s)
- Camille Zenobia
- a Department of Microbiology; University of Pennsylvania School of Dental Medicine ; Philadelphia , PA , USA
| | | |
Collapse
|
42
|
Xiao W, Dong G, Pacios S, Alnammary M, Barger LA, Wang Y, Wu Y, Graves DT. FOXO1 deletion reduces dendritic cell function and enhances susceptibility to periodontitis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1085-93. [PMID: 25794707 DOI: 10.1016/j.ajpath.2014.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 12/03/2014] [Accepted: 12/30/2014] [Indexed: 02/07/2023]
Abstract
The host response plays both protective and destructive roles in periodontitis. FOXO1 is a transcription factor that is activated in dendritic cells (DCs), but its function in vivo has not been examined. We investigated the role of FOXO1 in activating DCs in experimental (CD11c.Cre(+).FOXO1(L/L)) compared with matched control mice (CD11c.Cre(-).FOXO1(L/L)) in response to oral pathogens. Lineage-specific FOXO1 deletion reduced the recruitment of DCs to oral mucosal epithelium by approximately 40%. FOXO1 was needed for expression of genes that regulate migration, including integrins αν and β3 and matrix metalloproteinase-2. Ablation of FOXO1 in DCs significantly decreased IL-12 produced by DCs in mucosal surfaces. Moreover, FOXO1 deletion reduced migration of DCs to lymph nodes, reduced capacity of DCs to induce formation of plasma cells, and reduced production of bacteria-specific antibody. The decrease in DC function in the experimental mice led to increased susceptibility to periodontitis through a mechanism that involved a compensatory increase in osteoclastogenic factors, IL-1β, IL-17, and RANKL. Thus, we reveal a critical role for FOXO1 in DC recruitment to oral mucosal epithelium and activation of adaptive immunity induced by oral inoculation of bacteria.
Collapse
Affiliation(s)
- Wenmei Xiao
- Department of Periodontology, School and Hospital of Stomatology, Peking University, Beijing, China; Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Guangyu Dong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sandra Pacios
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maher Alnammary
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laura A Barger
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yu Wang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Implantology, School of Stomatology, Jilin University, Changchun, China
| | - Yingying Wu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
43
|
Arjunan P, El-Awady A, Dannebaum RO, Kunde-Ramamoorthy G, Cutler CW. High-throughput sequencing reveals key genes and immune homeostatic pathways activated in myeloid dendritic cells by Porphyromonas gingivalis 381 and its fimbrial mutants. Mol Oral Microbiol 2015; 31:78-93. [PMID: 26466817 DOI: 10.1111/omi.12131] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2015] [Indexed: 12/14/2022]
Abstract
The human microbiome consists of highly diverse microbial communities that colonize our skin and mucosal surfaces, aiding in maintenance of immune homeostasis. The keystone pathogen Porphyromonas gingivalis induces a dysbiosis and disrupts immune homeostasis through as yet unclear mechanisms. The fimbrial adhesins of P. gingivalis facilitate biofilm formation, invasion of and dissemination by blood dendritic cells; hence, fimbriae may be key factors in disruption of immune homeostasis. In this study we employed RNA-sequencing transcriptome profiling to identify differentially expressed genes (DEGs) in human monocyte-derived dendritic cells (MoDCs) in response to in vitro infection/exposure by Pg381 or its isogenic mutant strains that solely express minor-Mfa1 fimbriae (DPG3), major-FimA fimbriae (MFI) or are deficient in both fimbriae (MFB) relative to uninfected control. Our results yielded a total of 479 DEGs that were at least two-fold upregulated and downregulated in MoDCs significantly (P ≤ 0.05) by all four strains and certain DEGs that were strain-specific. Interestingly, the gene ontology biological and functional analysis shows that the upregulated genes in DPG3-induced MoDCs were more significant than other strains and associated with inflammation, immune response, anti-apoptosis, cell proliferation, and other homeostatic functions. Both transcriptome and quantitative polymerase chain reaction results show that DPG3, which solely expresses Mfa1, increased ZNF366, CD209, LOX1, IDO1, IL-10, CCL2, SOCS3, STAT3 and FOXO1 gene expression. In conclusion, we have identified key DC-mediated immune homeostatic pathways that could contribute to dysbiosis in periodontal infection with P. gingivalis.
Collapse
Affiliation(s)
- P Arjunan
- Department of Periodontics, Georgia Regents University, Augusta, GA, USA
| | - A El-Awady
- Department of Periodontics, Georgia Regents University, Augusta, GA, USA
| | - R O Dannebaum
- Department of Energy, Joint Genome Institute, Walnut Creek, CA, USA
| | - G Kunde-Ramamoorthy
- Department of Energy, Joint Genome Institute, Walnut Creek, CA, USA.,Department of Biochemistry, National University of Singapore, Singapore
| | - C W Cutler
- Department of Periodontics, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
44
|
Nagano K, Hasegawa Y, Yoshida Y, Yoshimura F. A Major Fimbrilin Variant of Mfa1 Fimbriae in Porphyromonas gingivalis. J Dent Res 2015; 94:1143-8. [PMID: 26001707 DOI: 10.1177/0022034515588275] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The periodontal pathogen Porphyromonas gingivalis is known to express 2 distinct types of fimbriae: FimA and Mfa1 fimbriae. However, we previously reported that fimbria-like structures were found in a P. gingivalis strain in which neither FimA nor Mfa1 fimbriae were detected. In this study, we identified a major protein in the bacterial lysates of the strain, which has been reported as the 53-kDa major outer membrane protein of P. gingivalis (53K protein) and subsequently reported as a major fimbrilin of a novel-type fimbria. Sequencing of the chromosomal DNA of the strain showed that the 53k gene (encoding the 53K protein) was located at a locus corresponding to the mfa1 gene (encoding the Mfa1 protein, which is a major fimbrilin of Mfa1 fimbriae) of the ATCC 33277 type strain. However, the 53K and Mfa1 proteins showed a low amino acid sequence homology and different antigenicity. The 53K protein was detected in 34 of 84 (41%) P. gingivalis strains, while the Mfa1 protein was detected in 44% of the strains. No strain expressed both 53K and Mfa1 proteins. Additionally, fimbriae were normally expressed in mutants in which the 53k and mfa1 genes were interchanged. These results indicate that the 53K protein is another major fimbrilin of Mfa1 fimbriae in P. gingivalis.
Collapse
Affiliation(s)
- K Nagano
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - Y Hasegawa
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - Y Yoshida
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| | - F Yoshimura
- Department of Microbiology, School of Dentistry, Aichi Gakuin University, Nagoya, Aichi, Japan
| |
Collapse
|
45
|
Abstract
Periodontitis is a dysbiotic inflammatory disease with an adverse impact on systemic health. Recent studies have provided insights into the emergence and persistence of dysbiotic oral microbial communities that can mediate inflammatory pathology at local as well as distant sites. This Review discusses the mechanisms of microbial immune subversion that tip the balance from homeostasis to disease in oral or extra-oral sites.
Collapse
|
46
|
El-Awady AR, Miles B, Scisci E, Kurago ZB, Palani CD, Arce RM, Waller JL, Genco CA, Slocum C, Manning M, Schoenlein PV, Cutler CW. Porphyromonas gingivalis evasion of autophagy and intracellular killing by human myeloid dendritic cells involves DC-SIGN-TLR2 crosstalk. PLoS Pathog 2015; 10:e1004647. [PMID: 25679217 PMCID: PMC4352937 DOI: 10.1371/journal.ppat.1004647] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/30/2014] [Indexed: 11/18/2022] Open
Abstract
Signaling via pattern recognition receptors (PRRs) expressed on professional antigen presenting cells, such as dendritic cells (DCs), is crucial to the fate of engulfed microbes. Among the many PRRs expressed by DCs are Toll-like receptors (TLRs) and C-type lectins such as DC-SIGN. DC-SIGN is targeted by several major human pathogens for immune-evasion, although its role in intracellular routing of pathogens to autophagosomes is poorly understood. Here we examined the role of DC-SIGN and TLRs in evasion of autophagy and survival of Porphyromonas gingivalis in human monocyte-derived DCs (MoDCs). We employed a panel of P. gingivalis isogenic fimbriae deficient strains with defined defects in Mfa-1 fimbriae, a DC-SIGN ligand, and FimA fimbriae, a TLR2 agonist. Our results show that DC-SIGN dependent uptake of Mfa1+P. gingivalis strains by MoDCs resulted in lower intracellular killing and higher intracellular content of P. gingivalis. Moreover, Mfa1+P. gingivalis was mostly contained within single membrane vesicles, where it survived intracellularly. Survival was decreased by activation of TLR2 and/or autophagy. Mfa1+P. gingivalis strain did not induce significant levels of Rab5, LC3-II, and LAMP1. In contrast, P. gingivalis uptake through a DC-SIGN independent manner was associated with early endosomal routing through Rab5, increased LC3-II and LAMP-1, as well as the formation of double membrane intracellular phagophores, a characteristic feature of autophagy. These results suggest that selective engagement of DC-SIGN by Mfa-1+P. gingivalis promotes evasion of antibacterial autophagy and lysosome fusion, resulting in intracellular persistence in myeloid DCs; however TLR2 activation can overcome autophagy evasion and pathogen persistence in DCs. Among the most successful of human microbes are intracellular pathogens. By entering the intracellular milieu, these pathogens are protected from harsh environmental factors in the host, including the humoral and cellular immune responses. Porphyromonas gingivalis is an opportunistic pathogen that colonizes the oral mucosa and accesses the bloodstream and distant sites such as the blood vessel walls, brain, placenta and other organs. Still unclear is how P. gingivalis traverses from oral mucosa to these distant sites. Dendritic cells are highly migratory antigen presenting cells that “patrol” the blood, skin, mucosa and all the major organ systems. Capture of microbes by dendritic cells activates a tightly regulated series of events, including directed migration towards the secondary lymphoid organs, where processed antigens are ostensibly presented to T cells. Autophagy is now recognized as an integral component of microbial clearance, antigen processing and presentation by dendritic cells. We report here that P. gingivalis is able to subvert autophagic destruction within dendritic cells. This occurs through its glycoprotein fimbriae, called Mfa-1, which targets the C-type lectin DC-SIGN on dendritic cells. The other major fimbriae on P. gingivalis, FimA, targets TLR2, which promotes autophagic destruction of P. gingivalis. We conclude that DC-SIGN-TLR2 crosstalk determines the intracellular fate of this pathogen within dendritic cells, and may have profound implications for the treatment of many chronic diseases involving low-grade infections.
Collapse
Affiliation(s)
- Ahmed R. El-Awady
- Department of Periodontics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Brodie Miles
- Department of Medicine, University of Colorado, Aurora, Colorado, United States of America
| | - Elizabeth Scisci
- School of Dental Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Zoya B. Kurago
- Department of Oral Health and Diagnostic Sciences, Georgia Regents University, Augusta, Georgia, United States of America
| | - Chithra D. Palani
- Department of Oral Health and Diagnostic Sciences, Georgia Regents University, Augusta, Georgia, United States of America
| | - Roger M. Arce
- Department of Periodontics, Georgia Regents University, Augusta, Georgia, United States of America
| | - Jennifer L. Waller
- Department of Biostatistics and Epidemiology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Caroline A. Genco
- Department of Medicine, Section of Infectious Diseases, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Connie Slocum
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Matthew Manning
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - Patricia V. Schoenlein
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - Christopher W. Cutler
- Department of Periodontics, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
47
|
Miles B, Abdel-Ghaffar KA, Gamal AY, Baban B, Cutler CW. Blood dendritic cells: "canary in the coal mine" to predict chronic inflammatory disease? Front Microbiol 2014; 5:6. [PMID: 24478766 PMCID: PMC3902297 DOI: 10.3389/fmicb.2014.00006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/07/2014] [Indexed: 12/19/2022] Open
Abstract
The majority of risk factors for chronic inflammatory diseases are unknown. This makes personalized medicine for assessment, prognosis, and choice of therapy very difficult. It is becoming increasingly clear, however, that low-grade subclinical infections may be an underlying cause of many chronic inflammatory diseases and thus may contribute to secondary outcomes (e.g., cancer). Many diseases are now categorized as inflammatory-mediated diseases that stem from a dysregulation in host immunity. There is a growing need to study the links between low-grade infections, the immune responses they elicit, and how this impacts overall health. One such link explored in detail here is the extreme sensitivity of myeloid dendritic cells (mDCs) in peripheral blood to chronic low-grade infections and the role that these mDCs play in arbitrating the resulting immune responses. We find that emerging evidence supports a role for pathogen-induced mDCs in chronic inflammation leading to increased risk of secondary clinical disease. The mDCs that are elevated in the blood as a result of low-grade bacteremia often do not trigger a productive immune response, but can disseminate the pathogen throughout the host. This aberrant trafficking of mDCs can accelerate systemic inflammatory disease progression. Conversely, restoration of dendritic cell homeostasis may aid in pathogen elimination and minimize dissemination. Thus it would seem prudent when assessing chronic inflammatory disease risk to consider blood mDC numbers, and the microbial content (microbiome) and activation state of these mDCs. These may provide important clues (“the canary in the coal mine”) of high inflammatory disease risk. This will facilitate development of novel immunotherapies to eliminate such smoldering infections in atherosclerosis, cancer, rheumatoid arthritis, and pre-eclampsia.
Collapse
Affiliation(s)
- Brodie Miles
- Department of Periodontics, College of Dental Medicine, Georgia Regents University Augusta, GA, USA
| | | | | | - Babak Baban
- Department of Oral Biology, Georgia Regents University Augusta, GA, USA
| | - Christopher W Cutler
- Department of Periodontics, College of Dental Medicine, Georgia Regents University Augusta, GA, USA
| |
Collapse
|
48
|
Barth K, Remick DG, Genco CA. Disruption of immune regulation by microbial pathogens and resulting chronic inflammation. J Cell Physiol 2013; 228:1413-22. [PMID: 23255141 DOI: 10.1002/jcp.24299] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 11/27/2012] [Indexed: 12/23/2022]
Abstract
Activation of the immune response is a tightly regulated, coordinated effort that functions to control and eradicate exogenous microorganisms, while also responding to endogenous ligands. Determining the proper balance of inflammation is essential, as chronic inflammation leads to a wide array of host pathologies. Bacterial pathogens can instigate chronic inflammation via an extensive repertoire of evolved evasion strategies that perturb immune regulation. In this review, we discuss two model pathogens, Mycobacterium tuberculosis and Porphyromonas gingivalis, which efficiently escape various aspects of the immune system within professional and non-professional immune cell types to establish chronic inflammation.
Collapse
Affiliation(s)
- Kenneth Barth
- Department of Medicine Section of Infectious Disease, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
49
|
Secondary lymphoid organ homing phenotype of human myeloid dendritic cells disrupted by an intracellular oral pathogen. Infect Immun 2013; 82:101-11. [PMID: 24126519 DOI: 10.1128/iai.01157-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Several intracellular pathogens, including a key etiological agent of chronic periodontitis, Porphyromonas gingivalis, infect blood myeloid dendritic cells (mDCs). This infection results in pathogen dissemination to distant inflammatory sites (i.e., pathogen trafficking). The alteration in chemokine-chemokine receptor expression that contributes to this pathogen trafficking function, particularly toward sites of neovascularization in humans, is unclear. To investigate this, we utilized human monocyte-derived DCs (MoDCs) and primary endothelial cells in vitro, combined with ex vivo-isolated blood mDCs and serum from chronic periodontitis subjects and healthy controls. Our results, using conditional fimbria mutants of P. gingivalis, show that P. gingivalis infection of MoDCs induces an angiogenic migratory profile. This profile is enhanced by expression of DC-SIGN on MoDCs and minor mfa-1 fimbriae on P. gingivalis and is evidenced by robust upregulation of CXCR4, but not secondary lymphoid organ (SLO)-homing CCR7. This disruption of SLO-homing capacity in response to respective chemokines closely matches surface expression of CXCR4 and CCR7 and is consistent with directed MoDC migration through an endothelial monolayer. Ex vivo-isolated mDCs from the blood of chronic periodontitis subjects, but not healthy controls, expressed a similar migratory profile; moreover, sera from chronic periodontitis subjects expressed elevated levels of CXCL12. Overall, we conclude that P. gingivalis actively "commandeers" DCs by reprogramming the chemokine receptor profile, thus disrupting SLO homing, while driving migration toward inflammatory vascular sites.
Collapse
|
50
|
Myneni SR, Settem RP, Sharma A. Bacteria take control of tolls and T cells to destruct jaw bone. Immunol Invest 2013; 42:519-31. [DOI: 10.3109/08820139.2013.822761] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|