1
|
Mishra A, Dou Y, Fletcher HM. Characterization of thioredoxin-thioredoxin reductase system in Filifactor alocis. Mol Oral Microbiol 2025; 40:50-63. [PMID: 39428740 DOI: 10.1111/omi.12486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Filifactor alocis is a newly appreciated member of the periodontal community with a strong periodontal disease correlation. Little is known about the survival mechanisms by which F. alocis copes with oxidative stress and establishes the infection within the local inflammatory microenvironment of the periodontal pocket. The aim of this study is to investigate if F. alocis putative peroxiredoxin/AhpC protein FA768 may constitute an alkyl hydroperoxide reductase system utilizing putative thioredoxin reductase protein FA608, and putative thioredoxin/glutaredoxin homolog FA1411/FA455. METHODS FA768, FA608, FA1411 and FA455 proteins from F. alocis were expressed and purified from Escherichia coli. Insulin and 5,5-dithio-bis-2-nitrobenzoic acid (DTNB) reduction assays were performed to determine if purified FA1411 and FA455 proteins could be a substrate for FA608. The peroxidase activity of FA768 was examined by measuring its ability to reduce hydrogen peroxide (H2O2) with FA608 and FA1411/FA455 provided as the reducing systems. Further, the hydroperoxide substrate specificity of FA768 was analyzed by monitoring the NADPH oxidation in the presence of different peroxides, including H2O2, cumyl hydroperoxide (CHP), and tert-butyl hydroperoxide (t-BHP). RESULTS In this study, we have demonstrated the existence of a functioning thioredoxin-dependent alkyl hydroperoxide system in F. alocis. This system is comprised of a thioredoxin reductase (FA608), a thioredoxin/glutaredoxin homolog (FA1411/FA455), and a typical 2-cysteine peroxiredoxin/AhpC (FA768). FA608, together with FA1411/FA455, can function as a thioredoxin reductase system to reduce insulin, DTNB, and FA768. FA455 is a glutaredoxin-like protein with thioredoxin functions in F. alocis. Both the FA768/FA608/FA1411 and FA768/FA608/FA455 reductase systems were NADPH-dependent and exhibited specificity for broad hydroperoxide substrates H2O2, CHP, and t-BHP. CONCLUSIONS This is the first study of a thioredoxin dependent alkyl hydroperoxide system from a periodontal pathogen. This system is proposed to protect F. alocis against oxidative stress due to the likely absence of a catalase or an additional peroxiredoxin homolog.
Collapse
Affiliation(s)
- Arunima Mishra
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Yuetan Dou
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Hansel M Fletcher
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
2
|
Chen YY, Tan L, Su XL, Chen NX, Liu Q, Feng YZ, Guo Y. NOD2 contributes to Parvimonas micra-induced bone resorption in diabetic rats with experimental periodontitis. Mol Oral Microbiol 2024; 39:446-460. [PMID: 38757737 DOI: 10.1111/omi.12467] [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: 02/15/2024] [Revised: 04/19/2024] [Accepted: 04/24/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) may affect the oral microbial community, exacerbating periodontal inflammation; however, its pathogenic mechanisms remain unclear. As nucleotide-binding oligomerization domain 2 (NOD2) plays a crucial role in the activation during periodontitis (PD), it is hypothesized that changes in the oral microbial community due to diabetes enhance periodontal inflammation through the activation of NOD2. METHODS We collected subgingival plaque from 180 subjects who were categorized into two groups based on the presence or absence of T2DM. The composition of oral microbiota was detected by 16S rRNA high-throughput sequencing. In animal models of PD with or without T2DM, we assessed alveolar bone resorption by micro-computerized tomography and used immunohistochemistry to detect NOD2 expression in alveolar bone. Primary osteoblasts were cultured in osteogenic induction medium with high or normal glucose and treated with inactivated bacteria. After 24 h of inactivated bacteria intervention, the osteogenic differentiation ability was detected by alkaline phosphatase (ALP) staining, and the expressions of NOD2 and interleukin-12 (IL-6) were detected by western blot. RESULTS The relative abundance of Parvimonas and Filifactor in the T2DM group was increased compared to the group without T2DM. In animal models, alveolar bone mass was decreased in PD, particularly in T2DM with PD (DMPD) group, compared to controls. Immunohistochemistry revealed NOD2 in osteoblasts from the alveolar bone in both the PD group and DMPD group, especially in the DMPD group. In vitro, intervention with inactivated Parvimonas significantly reduced ALP secretion of primary osteoblasts in high glucose medium, accompanied by increased expression of NOD2 and IL-6. CONCLUSIONS The results suggest that T2DM leading to PD may be associated with the activation of NOD2 by Parvimonas.
Collapse
MESH Headings
- Animals
- Nod2 Signaling Adaptor Protein/metabolism
- Periodontitis/microbiology
- Periodontitis/metabolism
- Rats
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/microbiology
- Diabetes Mellitus, Type 2/metabolism
- Alveolar Bone Loss/microbiology
- Alveolar Bone Loss/metabolism
- Male
- Osteoblasts/metabolism
- Humans
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/microbiology
- Interleukin-6/metabolism
- Female
- Disease Models, Animal
- Middle Aged
- Osteogenesis
- Rats, Sprague-Dawley
- X-Ray Microtomography
- RNA, Ribosomal, 16S
- Dental Plaque/microbiology
- Adult
- Interleukin-12/metabolism
Collapse
Affiliation(s)
- Ying-Yi Chen
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
- Department of Stomatology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences(Qingdao Central Hospital), Qingdao, China
| | - Li Tan
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Lin Su
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ning-Xin Chen
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Liu
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Zhi Feng
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Guo
- Hunan Provincial Clinical Research Center for Oral Diseases, Hunan Provincial Engineering Research Center of Digital Oral and Maxillofacial Defect Repair, Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Paul A, Wellslager B, Williamson M, Yilmaz Ö. Bacterial Protein Signatures Identified in Porphyromonas gingivalis Containing-Autophagic Vacuoles Reveal Co-Evolution Between Oral Red/Orange Complex Bacteria and Gut Bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.11.602567. [PMID: 39026754 PMCID: PMC11257597 DOI: 10.1101/2024.07.11.602567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Modern oral bacterial species present as a concoction of commensal and opportunistic pathogens originating from their evolution in humans. Due to the intricate colonization mechanisms shared amongst oral and gut bacteria, these bacteria have likely evolved together to establish and adapt in the human oro-digestive tract, resulting in the transfer of genetic information. Our liquid chromatography-with-tandem-mass-spectrometry (LC-MS-MS) analyses have revealed protein signatures, Elongation Factor Tu, RagB/SusD nutrient uptake outer membrane protein and DnaK, specifically from Porphyromonas gingivalis -containing autophagic vacuoles isolated from the infected human primary gingival epithelial cells. Interestingly, our Mass-Spectrometry analysis reported similar proteins from closely related oral bacteria, Tannerella forsythia and Prevotella intermedia . In our phylogenetic study of these key protein signatures, we have established that pathogenic oral bacteria share extensive relatedness to each other and gut resident bacteria. We show that in the virulence factors identified from gut bacteria, Elongation Factor Tu and DnaK, there are several structural similarities and conservations with proteins from oral pathogenic bacteria. There are also major similarities in the RagB/SusD proteins of oral bacteria to prominent gut bacteria. These findings not only highlight the shared virulence mechanisms amongst oral bacterial pathogens/pathobionts but also gut bacteria and elucidate their co-evolutions in the human host.
Collapse
|
4
|
Wellslager B, Roberts J, Chowdhury N, Madan L, Orellana E, Yilmaz Ö. Porphyromonas gingivalis activates Heat-Shock-Protein 27 to drive a LC3C-specific probacterial form of select autophagy that is redox sensitive for intracellular bacterial survival in human gingival mucosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601539. [PMID: 39005460 PMCID: PMC11244920 DOI: 10.1101/2024.07.01.601539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Porphyromonas gingivalis , a major oral pathobiont, evades canonical host pathogen clearance in human primary gingival epithelial cells (GECs) by initiating a non-canonical variant of autophagy consisting of Microtubule-associated protein 1A/1B-light chain 3 (LC3)-rich autophagosomes, which then act as replicative niches. Simultaneously, P. gingivalis inhibits apoptosis and oxidative-stress, including extracellular-ATP (eATP)-mediated reactive-oxygen-species (ROS) production via phosphorylating Heat Shock Protein 27 (HSp27) with the bacterial nucleoside-diphosphate-kinase (Ndk). Here, we have mechanistically identified that P. gingivalis -mediated induction of HSp27 is crucial for the recruitment of the LC3 isoform, LC3C, to drive the formation of live P. gingivalis -containing Beclin1-ATG14-rich autophagosomes that are redox sensitive and non-degrading. HSp27 depletions of both infected GECs and gingiva-mimicking organotypic-culture systems resulted in the collapse of P. gingivalis -mediated autophagosomes, and abolished P. gingivalis -induced LC3C-specific autophagic-flux in a HSp27-dependent manner. Concurrently, HSp27 depletion accompanied by eATP treatment abrogated protracted Beclin 1-ATG14 partnering and decreased live intracellular P. gingivalis levels. These events were only partially restored via treatments with the antioxidant N-acetyl cysteine (NAC), which rescued the cellular redox environment independent of HSp27. Moreover, the temporal phosphorylation of HSp27 by the bacterial Ndk results in HSp27 tightly partnering with LC3C, hindering LC3C canonical cleavage, extending Beclin 1-ATG14 association, and halting canonical maturation. These findings pinpoint how HSp27 pleiotropically serves as a major platform-molecule, redox regulator, and stepwise modulator of LC3C during P. gingivalis -mediated non-canonical autophagy. Thus, our findings can determine specific molecular strategies for interfering with the host-adapted P. gingivalis ' successful mucosal colonization and oral dysbiosis.
Collapse
|
5
|
Sheridan M, Chowdhury N, Wellslager B, Oleinik N, Kassir MF, Lee HG, Engevik M, Peterson Y, Pandruvada S, Szulc ZM, Yilmaz Ö, Ogretmen B. Opportunistic pathogen Porphyromonas gingivalis targets the LC3B-ceramide complex and mediates lethal mitophagy resistance in oral tumors. iScience 2024; 27:109860. [PMID: 38779482 PMCID: PMC11108982 DOI: 10.1016/j.isci.2024.109860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/29/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024] Open
Abstract
Mechanisms by which Porphyromonas gingivalis (P. gingivalis) infection enhances oral tumor growth or resistance to cell death remain elusive. Here, we determined that P. gingivalis infection mediates therapeutic resistance via inhibiting lethal mitophagy in cancer cells and tumors. Mechanistically, P. gingivalis targets the LC3B-ceramide complex by associating with LC3B via bacterial major fimbriae (FimA) protein, preventing ceramide-dependent mitophagy in response to various therapeutic agents. Moreover, ceramide-mediated mitophagy is induced by Annexin A2 (ANXA2)-ceramide association involving the E142 residue of ANXA2. Inhibition of ANXA2-ceramide-LC3B complex formation by wild-type P. gingivalis prevented ceramide-dependent mitophagy. Moreover, a FimA-deletion mutant P. gingivalis variant had no inhibitory effects on ceramide-dependent mitophagy. Further, 16S rRNA sequencing of oral tumors indicated that P. gingivalis infection altered the microbiome of the tumor macroenvironment in response to ceramide analog treatment in mice. Thus, these data provide a mechanism describing the pro-survival roles of P. gingivalis in oral tumors.
Collapse
Affiliation(s)
- Megan Sheridan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Nityananda Chowdhury
- Department of Oral Health Sciences, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Bridgette Wellslager
- Department of Oral Health Sciences, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Natalia Oleinik
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Mohamed Faisal Kassir
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Han G. Lee
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Mindy Engevik
- Department of Regenerative Medicine, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Yuri Peterson
- Department of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Subramanya Pandruvada
- Department of Oral Health Sciences, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Zdzislaw M. Szulc
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Özlem Yilmaz
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Department of Oral Health Sciences, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas Street, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Panta P, Tummakomma P, Purumandla U, Turimella S, Chintalapani S, Muttineni N, Kukkunuru GRT. Detection of Novel Periodontal Pathogens Using Fluorescence In Situ Hybridization: A Clinical Study. WORLD JOURNAL OF DENTISTRY 2024; 15:155-160. [DOI: 10.5005/jp-journals-10015-2369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
7
|
Mishra A, Dou Y, Wang C, Fletcher HM. Filifactor alocis enhances survival of Porphyromonas gingivalis W83 in response to H 2 O 2 -induced stress. Mol Oral Microbiol 2024; 39:12-26. [PMID: 38041478 PMCID: PMC10842171 DOI: 10.1111/omi.12445] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
A dysbiotic microbial community whose members have specific/synergistic functions that are modulated by environmental conditions, can disturb homeostasis in the subgingival space leading to destructive inflammation, plays a role in the progression of periodontitis. Filifactor alocis, a gram-positive, anaerobic bacterium, is a newly recognized microbe that shows a strong correlation with periodontal disease. Our previous observations suggested F. alocis to be more resistant to oxidative stress compared to Porphyromonas gingivalis. The objective of this study is to further determine if F. alocis, because of its increased resistance to oxidative stress, can affect the survival of other 'established' periodontal pathogens under environmental stress conditions typical of the periodontal pocket. Here, we have shown that via their interaction, F. alocis protects P. gingivalis W83 under H2 O2 -induced oxidative stress conditions. Transcriptional profiling of the interaction of F. alocis and P. gingivalis in the presence of H2 O2 -induced stress revealed the modulation of several genes, including those with ABC transporter and other cellular functions. The ABC transporter operon (PG0682-PG0685) of P. gingivalis was not significant to its enhanced survival when cocultured with F. alocis under H2 O2 -induced oxidative stress. In F. alocis, one of the most highly up-regulated operons (FA0894-FA0897) is predicted to encode a putative manganese ABC transporter, which in other bacteria can play an essential role in oxidative stress protection. Collectively, the results may indicate that F. alocis could likely stabilize the microbial community in the inflammatory microenvironment of the periodontal pocket by reducing the oxidative environment. This strategy could be vital to the survival of other pathogens, such as P. gingivalis, and its ability to adapt and persist in the periodontal pocket.
Collapse
Affiliation(s)
- Arunima Mishra
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350, USA
| | - Yuetan Dou
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350, USA
| | - Charles Wang
- Department of Basic Sciences, School of Medicine, Center for Genomics, Loma Linda University, Loma Linda, California 92350, USA
| | - Hansel M Fletcher
- Division of Microbiology & Molecular Genetics, School of Medicine, Loma Linda University, Loma Linda, California 92350, USA
| |
Collapse
|
8
|
Iskander MMZ, Lamont GJ, Tan J, Pisano M, Uriarte SM, Scott DA. Tobacco smoke exacerbates Filifactor alocis pathogenicity. J Clin Periodontol 2023; 50:121-130. [PMID: 36122937 PMCID: PMC9976951 DOI: 10.1111/jcpe.13729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/11/2022] [Accepted: 09/14/2022] [Indexed: 11/30/2022]
Abstract
AIM Filifactor alocis has recently emerged as a periodontal pathobiont that appears to thrive in the oral cavity of smokers. We hypothesized that identification of smoke-responsive F. alocis genes would provide insight into adaptive strategies and that cigarette smoke would enhance F. alocis pathogenesis in vivo. MATERIALS AND METHODS F. alocis was grown in vitro and cigarette smoke extract-responsive genes determined by RNAseq. Mice were exposed, or not, to mainstream 1R6F research cigarette smoke and infected with F. alocis, or not, in an acute ligature model of periodontitis. Key clinical, infectious, and immune data were collected. RESULTS In culture, F. alocis growth was unaffected by smoke conditioning and only a small number of genes were specifically regulated by smoke exposure. Reduced murine mass, differences in F. alocis-cognizant antibody production, and altered immune profiles as well as altered alveolar bone loss were all attributable to smoke exposure and/or F. alocis infection in vivo. CONCLUSIONS F. alocis is well-adapted to tobacco-rich conditions and its pathogenesis is enhanced by tobacco smoke exposure. A smoke-exposed ligature model of periodontitis shows promise as a tool with which to further unravel mechanisms underlying tobacco-enhanced, bacteria-induced disease.
Collapse
Affiliation(s)
- Mina M Z Iskander
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Gwyneth J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Jinlian Tan
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Michele Pisano
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky, USA
| |
Collapse
|
9
|
Karakaya E, Abdul Y, Chowdhury N, Wellslager B, Jamil S, Albayram O, Yilmaz Ö, Ergul A. Porphyromonas gingivalis infection upregulates the endothelin (ET) system in brain microvascular endothelial cells. Can J Physiol Pharmacol 2022; 100:679-688. [PMID: 35442801 PMCID: PMC9583200 DOI: 10.1139/cjpp-2022-0035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelin-1 (ET-1), the most potent vasoconstrictor identified to date, contributes to cerebrovascular dysfunction and brain ET-1 levels were shown to be related to Alzheimer's disease and related dementias (ADRD) progression. ET-1 also contributes to neuroinflammation, especially in infections of the central nervous system. Recent studies causally linked chronic periodontal infection with an opportunistic anaerobic bacterium Porphyromonas gingivalis (Coykendall et al.) Shah & Collins to AD development. Thus, the goal of the study was to determine the impact of P. gingivalis infection on the ET system and cell senescence in brain microvascular endothelial cells. Cells were infected with a multiplicity of infection 50 P. gingivalis with and without extracellular ATP-induced oxidative stress for 24 h. Cell lysates were collected for analysis of endothelin A receptor (ETA)/endothelin B receptor (ETB) receptor as well as senescence markers. ET-1 levels in cell culture media were measured with enzyme-linked immunosorbent assay. P. gingivalis infection increased ET-1 (pg/mL) secretion, as well as the ETA receptor expression, whereas decreased lamin A/C expression compared to control. Tight junction protein claudin-5 was also decreased under these conditions. ETA or ETB receptor blockade during infection did not affect ET-1 secretion or the expression of cell senescence markers. Current findings suggest that P. gingivalis infection may compromise endothelial integrity and activate the ET system.
Collapse
Affiliation(s)
- Eda Karakaya
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina
- Department of Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Yasir Abdul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina
- Department of Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | | | | | - Sarah Jamil
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina
- Department of Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Onder Albayram
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina
- Department of Neurosciences, Medical University of South Carolina
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina
| | - Adviye Ergul
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina
- Department of Neurosciences, Medical University of South Carolina
| |
Collapse
|
10
|
Zhu J, Chu W, Luo J, Yang J, He L, Li J. Dental Materials for Oral Microbiota Dysbiosis: An Update. Front Cell Infect Microbiol 2022; 12:900918. [PMID: 35846759 PMCID: PMC9280126 DOI: 10.3389/fcimb.2022.900918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
The balance or dysbiosis of the microbial community is a major factor in maintaining human health or causing disease. The unique microenvironment of the oral cavity provides optimal conditions for colonization and proliferation of microbiota, regulated through complex biological signaling systems and interactions with the host. Once the oral microbiota is out of balance, microorganisms produce virulence factors and metabolites, which will cause dental caries, periodontal disease, etc. Microbial metabolism and host immune response change the local microenvironment in turn and further promote the excessive proliferation of dominant microbes in dysbiosis. As the product of interdisciplinary development of materials science, stomatology, and biomedical engineering, oral biomaterials are playing an increasingly important role in regulating the balance of the oral microbiome and treating oral diseases. In this perspective, we discuss the mechanisms underlying the pathogenesis of oral microbiota dysbiosis and introduce emerging materials focusing on oral microbiota dysbiosis in recent years, including inorganic materials, organic materials, and some biomolecules. In addition, the limitations of the current study and possible research trends are also summarized. It is hoped that this review can provide reference and enlightenment for subsequent research on effective treatment strategies for diseases related to oral microbiota dysbiosis.
Collapse
Affiliation(s)
- Jieyu Zhu
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenlin Chu
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Jun Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jiaojiao Yang, ; Libang He,
| | - Libang He
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jiaojiao Yang, ; Libang He,
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Park DY, Park JY, Lee D, Hwang I, Kim HS. Leaky Gum: The Revisited Origin of Systemic Diseases. Cells 2022; 11:1079. [PMID: 35406643 PMCID: PMC8997512 DOI: 10.3390/cells11071079] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
The oral cavity is the gateway for microorganisms into your body where they disseminate not only to the directly connected respiratory and digestive tracts but also to the many remote organs. Oral microbiota, travelling to the end of the intestine and circulating in our bodies through blood vessels, not only affect a gut microbiome profile but also lead to many systemic diseases. By gathering information accumulated from the era of focal infection theory to the age of revolution in microbiome research, we propose a pivotal role of "leaky gum", as an analogy of "leaky gut", to underscore the importance of the oral cavity in systemic health. The oral cavity has unique structures, the gingival sulcus (GS) and the junctional epithelium (JE) below the GS, which are rarely found anywhere else in our body. The JE is attached to the tooth enamel and cementum by hemidesmosome (HD), which is structurally weaker than desmosome and is, thus, vulnerable to microbial infiltration. In the GS, microbial biofilms can build up for life, unlike the biofilms on the skin and intestinal mucosa that fall off by the natural process. Thus, we emphasize that the GS and the JE are the weakest leaky point for microbes to invade the human body, making the leaky gum just as important as, or even more important than, the leaky gut.
Collapse
Affiliation(s)
- Do-Young Park
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Jin Young Park
- Department of Gastrointestinal Endoscopy, Apple Tree Healthcare Center, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Dahye Lee
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| | - Inseong Hwang
- DOCSmedi Co., Ltd., 4F, 143, Gangseong-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
| | - Hye-Sung Kim
- Department of Orthodontics, Apple Tree Dental Hospital, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea;
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, 1450, Jungang-ro, Ilsanseo-gu, Goyang-si 10387, Korea
| |
Collapse
|
12
|
Liu H, Tang Y, Zhang S, Liu H, Wang Z, Li Y, Wang X, Ren L, Yang K, Qin L. Anti-infection mechanism of a novel dental implant made of titanium-copper (TiCu) alloy and its mechanism associated with oral microbiology. Bioact Mater 2022; 8:381-395. [PMID: 34541408 PMCID: PMC8429474 DOI: 10.1016/j.bioactmat.2021.05.053] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/19/2021] [Accepted: 05/30/2021] [Indexed: 12/13/2022] Open
Abstract
This work was focused on study of anti-infection ability and its underlying mechanism of a novel dental implant made of titanium-copper (TiCu) alloy. In general, most studies on antibacterial implants have used a single pathogen to test their anti-infection ability using infectious animal models. However, dental implant-associated infections are polymicrobial diseases. We innovatively combine the classic ligature model in dogs with sucrose-rich diets to induce oral infections via the canine native oral bacteria. The anti-infection ability, biocompatibility and underlying mechanism of TiCu implant were systematically investigated in comparison with pure Ti implant via general inspection, hematology, imageology (micro-CT), microbiology (16S rDNA and metagenome), histology, and Cu ion detections. Compared with Ti implant, TiCu implant demonstrated remarkable anti-infection potentials with excellent biocompatibility. Additionally, the underlying anti-infection mechanism of TiCu implant was considered to involve maintaining the oral microbiota homeostasis. It was found that the carbohydrates in the plaques formed on the surface of TiCu implant were metabolized through the tricarboxylic acid cycle (TCA) cycles, which prevented the formation of an acidic microenvironment and inhibited the accumulation of acidogens and pathogens, thereby maintaining the microflora balance between aerobic and anaerobic bacteria.
Collapse
Affiliation(s)
- Hui Liu
- School of Materials Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Yulong Tang
- Department of Stomatology, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, 110016, China
| | - Shuyuan Zhang
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Huan Liu
- School of Materials Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, 230026, China
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Zijian Wang
- Department of Stomatology, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, 110016, China
| | - Yue Li
- Department of Stomatology, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, 110016, China
| | - Xinluan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518057, China
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Ling Ren
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Ke Yang
- Shi-changxu Innovation Center for Advanced Materials, Institute of Metal Research, Chinese Academy of Sciences, 72 Wenhua Road, Shenyang, 110016, China
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518057, China
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
13
|
Wirth R, Pap B, Maróti G, Vályi P, Komlósi L, Barta N, Strang O, Minárovits J, Kovács KL. Toward Personalized Oral Diagnosis: Distinct Microbiome Clusters in Periodontitis Biofilms. Front Cell Infect Microbiol 2022; 11:747814. [PMID: 35004342 PMCID: PMC8727345 DOI: 10.3389/fcimb.2021.747814] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Periodontitis is caused by pathogenic subgingival microbial biofilm development and dysbiotic interactions between host and hosted microbes. A thorough characterization of the subgingival biofilms by deep amplicon sequencing of 121 individual periodontitis pockets of nine patients and whole metagenomic analysis of the saliva microbial community of the same subjects were carried out. Two biofilm sampling methods yielded similar microbial compositions. Taxonomic mapping of all biofilms revealed three distinct microbial clusters. Two clinical diagnostic parameters, probing pocket depth (PPD) and clinical attachment level (CAL), correlated with the cluster mapping. The dysbiotic microbiomes were less diverse than the apparently healthy ones of the same subjects. The most abundant periodontal pathogens were also present in the saliva, although in different representations. The single abundant species Tannerella forsythia was found in the diseased pockets in about 16–17-fold in excess relative to the clinically healthy sulcus, making it suitable as an indicator of periodontitis biofilms. The discrete microbial communities indicate strong selection by the host immune system and allow the design of targeted antibiotic treatment selective against the main periodontal pathogen(s) in the individual patients.
Collapse
Affiliation(s)
- Roland Wirth
- Department of Biotechnology, University of Szeged, Szeged, Hungary
| | - Bernadett Pap
- Biological Research Center, Institute of Plant Biology, Szeged, Hungary
| | - Gergely Maróti
- Biological Research Center, Institute of Plant Biology, Szeged, Hungary
| | - Péter Vályi
- Department of Periodontology, University of Szeged, Szeged, Hungary
| | - Laura Komlósi
- Department of Oral Surgery, University of Szeged, Szeged, Hungary
| | - Nikolett Barta
- Department of Biotechnology, University of Szeged, Szeged, Hungary
| | - Orsolya Strang
- Department of Biotechnology, University of Szeged, Szeged, Hungary.,Department of Oral Biology and Experimental Dental Research, University of Szeged, Szeged, Hungary
| | - János Minárovits
- Department of Oral Biology and Experimental Dental Research, University of Szeged, Szeged, Hungary
| | - Kornél L Kovács
- Department of Biotechnology, University of Szeged, Szeged, Hungary.,Department of Oral Biology and Experimental Dental Research, University of Szeged, Szeged, Hungary
| |
Collapse
|
14
|
Xu F, Pushalkar S, Lin Z, Thomas SC, Persaud JK, Sierra MA, Vardhan M, Vasconcelos R, Akapo A, Guo Y, Gordon T, Corby PM, Kamer AR, Li X, Saxena D. Electronic cigarette use enriches periodontal pathogens. Mol Oral Microbiol 2022; 37:63-76. [PMID: 34997976 DOI: 10.1111/omi.12361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/28/2022]
Abstract
The effect of electronic cigarette (e-cigarette) smoking, especially its long-term impact on oral health, is poorly understood. Here, we conducted a longitudinal clinical study with two study visits, 6 months apart, to investigate the effect of e-cigarette use on the bacterial community structure in the saliva of 101 periodontitis patients. Our data demonstrated that e-cigarette use altered the oral microbiome in periodontitis patients, enriching members of the Filifactor, Treponema, and Fusobacterium taxa. For patients at the same periodontal disease stage, cigarette smokers and e-cigarette smokers shared more similarities in their oral bacterial composition. E-cigarette smoking may have a similar potential as cigarette smoking at altering the bacterial composition of saliva over time, leading to an increase in the relative abundance of periodontal disease-associated pathogens such as Porphyromonas gingivalis and Fusobacterium nucleatum. The correlation analysis showed that certain genera, such as Dialister, Selenomonas, and Leptotrichia in the e-cigarette smoking group, were positively correlated with the levels of proinflammatory cytokines, including IFN-γ, IL-1β, and TNF-α. E-cigarette use was also associated with elevated levels of proinflammatory cytokines such as IFN-γ and TNF-α, which contribute to oral microbiome dysbiosis and advanced disease state. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Fangxi Xu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Smruti Pushalkar
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Ziyan Lin
- Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Scott C Thomas
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Julia Kishanie Persaud
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Maria A Sierra
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Mridula Vardhan
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Rebeca Vasconcelos
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Adenike Akapo
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Yuqi Guo
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Terry Gordon
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, United States
| | - Patricia M Corby
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Angela R Kamer
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York, NY, United States
| | - Xin Li
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| | - Deepak Saxena
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, United States
| |
Collapse
|
15
|
Microbiota in Periodontitis: Advances in the Omic Era. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1373:19-43. [DOI: 10.1007/978-3-030-96881-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
16
|
Abstract
Inflammasomes are multiprotein complexes that assemble in host cells upon recognition of infection or danger via pattern recognition receptors and/or danger recognition receptors. The assembly of inflammasomes results in the activation of caspase-1 and is followed by the enzymatic maturation and secretion of inflammatory cytokines like interleukin 1β (IL-1β) and IL-18.In the oral cavity, gingival epithelial cells (GECs) line the mucosa and have a barrier role for invading pathogens. In these cells, the NLRP3 inflammasome is the best studied and has been shown to play a role in the inflammatory immune response against a variety of oral pathogens. As such, in order to study gingivitis and other oral pathologic inflammatory conditions, studying the activation of inflammasomes is important. The simplest way to detect inflammasome activation is to detect the activated caspase-1, as well as the secretion of mature IL-1β and IL-18, via ELISA, Western blot, and immunofluorescence techniques.Here we describe the detection of NLRP3 inflammasome activation by the oral pathogen Porphyromonas gingivalis in human GECs via these three methods and mention other methods and techniques that we have successfully used together with these in our quest to understand the host-pathogen interaction.
Collapse
Affiliation(s)
- Kalina R Atanasova
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Özlem Yilmaz
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, SC, USA.
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
17
|
Porphyromonas gingivalis infection promotes mitochondrial dysfunction through Drp1-dependent mitochondrial fission in endothelial cells. Int J Oral Sci 2021; 13:28. [PMID: 34475379 PMCID: PMC8413291 DOI: 10.1038/s41368-021-00134-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/24/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023] Open
Abstract
Porphyromonas gingivalis (P. gingivalis), a key pathogen in periodontitis, has been shown to accelerate the progression of atherosclerosis (AS). However, the definite mechanisms remain elusive. Emerging evidence supports an association between mitochondrial dysfunction and AS. In our study, the impact of P. gingivalis on mitochondrial dysfunction and the potential mechanism were investigated. The mitochondrial morphology of EA.hy926 cells infected with P. gingivalis was assessed by transmission electron microscopy, mitochondrial staining, and quantitative analysis of the mitochondrial network. Fluorescence staining and flow cytometry analysis were performed to determine mitochondrial reactive oxygen species (mtROS) and mitochondrial membrane potential (MMP) levels. Cellular ATP production was examined by a luminescence assay kit. The expression of key fusion and fission proteins was evaluated by western blot and immunofluorescence. Mdivi-1, a specific Drp1 inhibitor, was used to elucidate the role of Drp1 in mitochondrial dysfunction. Our findings showed that P. gingivalis infection induced mitochondrial fragmentation, increased the mtROS levels, and decreased the MMP and ATP concentration in vascular endothelial cells. We observed upregulation of Drp1 (Ser616) phosphorylation and translocation of Drp1 to mitochondria. Mdivi-1 blocked the mitochondrial fragmentation and dysfunction induced by P. gingivalis. Collectively, these results revealed that P. gingivalis infection promoted mitochondrial fragmentation and dysfunction, which was dependent on Drp1. Mitochondrial dysfunction may represent the mechanism by which P. gingivalis exacerbates atherosclerotic lesions.
Collapse
|
18
|
Patel S, Howard D, Chowdhury N, Derieux C, Wellslager B, Yilmaz Ö, French L. Characterization of Human Genes Modulated by Porphyromonas gingivalis Highlights the Ribosome, Hypothalamus, and Cholinergic Neurons. Front Immunol 2021; 12:646259. [PMID: 34194426 PMCID: PMC8236716 DOI: 10.3389/fimmu.2021.646259] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/20/2021] [Indexed: 12/12/2022] Open
Abstract
Porphyromonas gingivalis, a bacterium associated with periodontal disease, is a suspected cause of Alzheimer's disease. This bacterium is reliant on gingipain proteases, which cleave host proteins after arginine and lysine residues. To characterize gingipain susceptibility, we performed enrichment analyses of arginine and lysine proportion proteome-wide. Genes differentially expressed in brain samples with detected P. gingivalis reads were also examined. Genes from these analyses were tested for functional enrichment and specific neuroanatomical expression patterns. Proteins in the SRP-dependent cotranslational protein targeting to membrane pathway were enriched for these residues and previously associated with periodontal and Alzheimer's disease. These ribosomal genes are up-regulated in prefrontal cortex samples with detected P. gingivalis sequences. Other differentially expressed genes have been previously associated with dementia (ITM2B, MAPT, ZNF267, and DHX37). For an anatomical perspective, we characterized the expression of the P. gingivalis associated genes in the mouse and human brain. This analysis highlighted the hypothalamus, cholinergic neurons, and the basal forebrain. Our results suggest markers of neural P. gingivalis infection and link the cholinergic and gingipain hypotheses of Alzheimer's disease.
Collapse
Affiliation(s)
- Sejal Patel
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Derek Howard
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Nityananda Chowdhury
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Casey Derieux
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Bridgette Wellslager
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, United States
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Leon French
- Krembil Centre for Neuroinformatics, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Institute for Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
19
|
Ebersole JL, Nagarajan R, Kirakodu S, Gonzalez OA. Transcriptomic phases of periodontitis lesions using the nonhuman primate model. Sci Rep 2021; 11:9282. [PMID: 33927312 PMCID: PMC8085193 DOI: 10.1038/s41598-021-88803-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/09/2021] [Indexed: 11/09/2022] Open
Abstract
We used a nonhuman primate model of ligature-induced periodontitis to identify patterns of gingival transcriptomic after changes demarcating phases of periodontitis lesions (initiation, progression, resolution). A total of 18 adult Macaca mulatta (12-22 years) had ligatures placed (premolar, 1st molar teeth) in all 4 quadrants. Gingival tissue samples were obtained (baseline, 2 weeks, 1 and 3 months during periodontitis and at 5 months resolution). Gene expression was analyzed by microarray [Rhesus Gene 1.0 ST Array (Affymetrix)]. Compared to baseline, a large array of genes were significantly altered at initiation (n = 6049), early progression (n = 4893), and late progression (n = 5078) of disease, with the preponderance being up-regulated. Additionally, 1918 genes were altered in expression with disease resolution, skewed towards down-regulation. Assessment of the genes demonstrated specific profiles of epithelial, bone/connective tissue, apoptosis/autophagy, metabolism, regulatory, immune, and inflammatory responses that were related to health, stages of disease, and tissues with resolved lesions. Unique transcriptomic profiles occured during the kinetics of the periodontitis lesion exacerbation and remission. We delineated phase specific gene expression profiles of the disease lesion. Detection of these gene products in gingival crevicular fluid samples from human disease may contribute to a better understanding of the biological dynamics of the disease to improve patient management.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, B221, University of Nevada Las Vegas, 1001 Shadow Lane, Las Vegas, NV, 89106, USA.
- Center for Oral Health Research College of Dentistry, University of Kentucky, Lexington, KY, USA.
| | | | - Sreenatha Kirakodu
- Center for Oral Health Research College of Dentistry, University of Kentucky, Lexington, KY, USA
| | - Octavio A Gonzalez
- Center for Oral Health Research College of Dentistry, University of Kentucky, Lexington, KY, USA
- Division of Periodontology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|