1
|
Araujo TT, Dionizio A, Carvalho TDS, Debortolli ALB, Vertuan M, de Souza BM, Camara JVF, Henrique-Silva F, Chiaratti M, Santos A, Alves L, Ferro M, Magalhães AC, Buzalaf MAR. Acquired pellicle and biofilm engineering with CaneCPI-5: Insights from proteomic and microbiomics analysis. Arch Oral Biol 2024; 166:106025. [PMID: 38943859 DOI: 10.1016/j.archoralbio.2024.106025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 07/01/2024]
Abstract
OBJECTIVE In this in vivo proof-of-concept study, acquired pellicle engineering was implemented to promote alterations in the protein composition of the acquired enamel pellicle (AEP) and the bacterial composition of the dental biofilm after treatment with Sugarcane cystatin (CaneCPI-5). DESIGN After prophylaxis, 10 volunteers rinsed (10 mL, 1 min) with the following solutions: 1) deionized water (H2O- negative control or 2) 0.1 mg/mL CaneCPI-5. The AEP and biofilm were formed along 2 or 3 h, respectively. The AEP was collected with electrode filter papers soaked in 3 % citric acid. After protein extraction, samples were analyzed by quantitative shotgun label-free proteomics. The biofilm microbiome was collected with a dental curette. The DNA was extracted, amplified, and analyzed by 16S-rRNA Next Generation Sequencing (NGS). RESULTS Treatment with CaneCPI-5 increased several proteins with antimicrobial, acid-resistance, affinity for hydroxyapatite, structural and calcium binding properties, such as Cysteine-rich-3 (6-fold-p = 0.03), Cystatin-B (5.5-fold-p < 0.01), Neutrophil-defensin 1 (4.7-fold-p < 0.01), Mucin (3.9-fold-p < 0.01), Immunoglobulin-heavy-constant (3.8-fold-p < 0.01) and Lactotransferrin (2.8-fold-p < 0.01). Microbiome revealed that several commensal bacteria had their abundance increased after rinsing with CaneCPI-5, such as Corynebacterium and Neisseria, while Streptococcus and Prevotella nigrescens were decreased. The results indicate the efficiency of CaneCPI-5 in promoting beneficial changes in the AEP and biofilm, making this phytocystatin a potential target for incorporation into dental products. CONCLUSION Cane demonstrated the capability to alter the protein composition of the acquired enamel pellicle (AEP) and the initial colonizers of the biofilm, enhancing the presence of proteins and bacteria crucial for dental protection.
Collapse
Affiliation(s)
- Tamara Teodoro Araujo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Aline Dionizio
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | | | | - Mariele Vertuan
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - Beatriz Martines de Souza
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | - João Victor Frazão Camara
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, Building 73, 66421 Homburg, Saarland, Germany
| | - Flavio Henrique-Silva
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Marcos Chiaratti
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Angélica Santos
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Lindomar Alves
- Departamento de Genética e Evolução, Universidade Federal de São Carlos, São Carlos 13565-905, Brazil
| | - Milene Ferro
- Department of General and Applied Biology, Paulista State University (UNESP), Rio Claro 13500230, Brazil
| | - Ana Carolina Magalhães
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Bauru, Brazil
| | | |
Collapse
|
2
|
Zhang Z, Zhan F. Type 2 Cystatins and Their Roles in the Regulation of Human Immune Response and Cancer Progression. Cancers (Basel) 2023; 15:5363. [PMID: 38001623 PMCID: PMC10670837 DOI: 10.3390/cancers15225363] [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: 09/21/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Cystatins are a family of intracellular and extracellular protease inhibitors that inhibit cysteine cathepsins-a group of lysosomal cysteine proteases that participate in multiple biological processes, including protein degradation and post-translational cleavage. Cysteine cathepsins are associated with the development of autoimmune diseases, tumor progression, and metastasis. Cystatins are categorized into three subfamilies: type 1, type 2, and type 3. The type 2 cystatin subfamily is the largest, containing 10 members, and consists entirely of small secreted proteins. Although type 2 cystatins have many shared biological roles, each member differs in structure, post-translational modifications (e.g., glycosylation), and expression in different cell types. These distinctions allow the type 2 cystatins to have unique biological functions and properties. This review provides an overview of type 2 cystatins, including their biological similarities and differences, their regulatory effect on human immune responses, and their roles in tumor progression, immune evasion, and metastasis.
Collapse
Affiliation(s)
| | - Fenghuang Zhan
- Myeloma Center, Winthrop P. Rockefeller Cancer Institute, Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
3
|
An active domain SA-2 derived from cystatin-SA, and its antifungal activity. Amino Acids 2023; 55:101-112. [PMID: 36333524 DOI: 10.1007/s00726-022-03207-8] [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: 04/14/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022]
Abstract
Infections induced by fungi, especially the drug-resistant fungi, are difficult clinical problems. Conventional antifungal treatment is effective but due to resistance, treatment failure, and treatment-related toxicity, there is a need for new antifungal drugs. In this study, SA-2 (YYRRLLRVLRRRW) was derived from Cystatin-SA, a saliva protein with a molecular weight of 14 kDa. Meanwhile, the structure-activity of SA-2 and its mutants was also studied. We detected the antimicrobial activity and cytotoxicity of SA-2 and found that SA-2 had a low cytotoxicity toward mammalian cells but a good inhibitory effect on Candida albicans (C. albicans) and Cryptococcus neoformans (C. neoformans), with MIC values of 16-64 μg/mL and 8-32 μg/mL, respectively. Interestingly, SA-2 effectively killed fluconazole-resistant C. neoformans and C. albicans within 12 h. This antifungal activity against fluconazole-resistant fungi was comparable to that of amphotericin B. In addition, the C. neoformans-infected mice model was established to evaluate the anti-infective activity of SA-2 in vivo. Results showed that SA-2 significantly reduced the counts of fungi in lung and brain tissues to protect fluconazole-resistant C. neoformans-infected mice from death without changing mice body weights. Moreover, the dramatically increased pro-inflammatory cytokines TNF-α, IL-6 and IL-1β induced by intranasal infection of C. neoformans could be obviously declined due to the treatment of SA-2, which may be attributed to the elimination of C. neoformans in time in the infected tissue. For the mode of actions underlying SA-2 against C. neoformans, we found that the cationic peptide SA-2 could adhere to the negatively charged fungal cell membrane to increase the surface potential of C. neoformans in a dose-dependent manner, and finally disrupted the integrity of fungal cell membrane, reflecting as a 60% positive rate of propidium iodide uptake of C. neoformans cells after SA-2 (4 × MIC) treatment. Our study indicated that SA-2 has the potential to develop as a new therapeutic agent against infection induced by drug-resistant fungi.
Collapse
|
4
|
Yamamoto K, Yamamoto S. Comparison of proteins with anti-influenza virus effects in parotid and submandibular-sublingual saliva in humans. BMC Oral Health 2022; 22:639. [PMID: 36566172 PMCID: PMC9789508 DOI: 10.1186/s12903-022-02686-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Saliva possesses antiviral activity, with submandibular-sublingual (SMSL) saliva having higher antiviral activity than parotid saliva. Various salivary proteins have inactivating effects on influenza A virus (IAV), but the detailed relationship between antiviral proteins and salivary anti-IAV activities in the parotid and SMSL glands is unknown. Here, to identify salivary proteins with anti-IAV activity, salivary proteins from parotid and SMSL glands were identified, quantified, and compared using liquid chromatography-mass spectrometry. METHODS Twelve healthy male volunteers participated in the study. Parotid and SMSL saliva was collected by suction and collection devices. We assessed anti-IAV activities, protein concentrations, and protein-bound sialic acid concentrations in parotid and SMSL saliva. RESULTS SMSL had significantly higher anti-IAV activity than parotid saliva. SMSL also had higher concentrations of glycoproteins, such as mucin 5B and mucin 7, protein-bound sialic acid, cystatins, and lysozyme C, compared with parotid saliva. Salivary mucin 5B and mucin 7 concentrations significantly positively correlated with the salivary protein-bound sialic acid concentration. Salivary anti-IAV activity significantly positively correlated with protein-bound sialic acid, mucin 5B, mucin 7, cystatin-C, -S, and -SN concentrations. CONCLUSION Salivary mucins, cystatins, and lysozyme C contribute to the high anti-IAV activity of SMSL saliva.
Collapse
Affiliation(s)
- Kenkichi Yamamoto
- grid.419719.30000 0001 0816 944XPersonal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, 131-8501 Tokyo, Japan
| | - Shinji Yamamoto
- grid.419719.30000 0001 0816 944XPersonal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, 131-8501 Tokyo, Japan
| |
Collapse
|
5
|
Nocera AL, Mueller SK, Workman AD, Wu D, McDonnell K, Sadow PM, Amiji MM, Bleier BS. Cystatin SN is a potent upstream initiator of epithelial-derived type 2 inflammation in chronic rhinosinusitis. J Allergy Clin Immunol 2022; 150:872-881. [PMID: 35660375 PMCID: PMC9547833 DOI: 10.1016/j.jaci.2022.04.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Cystatin SN (CST1) and cystatin SA (CST2) are cysteine protease inhibitors that protect against allergen, viral, and bacterial proteases. Cystatins are overexpressed in the setting of allergic rhinitis and chronic rhinosinusitis with nasal polyps (CRSwNP); however, their role in promoting type 2 inflammation remains poorly characterized. OBJECTIVE The purpose of this study was to use integrated poly-omics and a murine exposure model to explore the link between cystatin overexpression in CRSwNP and type 2 inflammation. METHODS In this institutional review board- and institutional animal care and use committee-approved study, we compared tissue, exosome, and mucus CST1 and CST2 between CRSwNP and controls (n = 10 per group) by using matched whole exome sequencing, transcriptomic, proteomic, posttranslational modification, histologic, functional, and bioinformatic analyses. C57/BL6 mice were dosed with 3.9 μg/mL of CST1 or PBS intranasally for 5 to 18 days in the presence or absence of epithelial ABCB1a knockdown. Inflammatory cytokines were quantified by using Quansys multiplex assays or ELISAs. RESULTS Of the 1305 proteins quantified, CST1 and CST2 were among the most overexpressed protease inhibitors in tissue, exosome, and mucus samples; they were localized to the epithelial layer. Multiple posttranslational modifications were identified in the polyp tissue. Exosomal CST1 and CST2 were strongly and significantly correlated with eosinophils and Lund-Mackay scores. Murine type 2 cytokine secretion and TH2 cell infiltration increased in a time-dependent manner following CST1 exposure and was abrogated by epithelial knockdown of ABCB1a, a regulator of epithelial cytokine secretion. CONCLUSION CST1 is a potent upstream initiator of epithelial-derived type 2 inflammation in CRSwNP. Therapeutic strategies targeting CST activity and its associated posttranslational modifications deserve further interrogation.
Collapse
Affiliation(s)
- Angela L Nocera
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass; Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Mass
| | - Sarina K Mueller
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass; Department of Otolaryngology/Head and Neck Surgery, University of Erlangen-Nuremberg, Nuremberg, Germany
| | - Alan D Workman
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Dawei Wu
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Kristen McDonnell
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, Mass
| | - Benjamin S Bleier
- Department of Otolaryngology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Mass.
| |
Collapse
|
6
|
Chen M, Lin W, Gan J, Lu W, Wang M, Wang X, Yi J, Zhao Z. Transcriptomic Mapping of Human Parotid Gland at Single-Cell Resolution. J Dent Res 2022; 101:972-982. [PMID: 35220796 DOI: 10.1177/00220345221076069] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
As the largest salivary gland in oral cavity, the parotid gland plays an important role in initial digesting and lubricating food. The abnormal secretory function of the parotid gland can lead to dental caries and oral mucosal inflammation. In recent years, single-cell RNA sequencing (scRNA-seq) has been used to explore the heterogeneity and diversity of cells in various organs and tissues. However, the transcription profile of the human parotid gland at single-cell resolution has not been reported yet. In this study, we constructed the cell atlas of human parotid gland using the 10× Genomics platform. Characteristic gene analysis identified the biological functions of serous acinar cell populations in secreting digestive enzymes and antibacterial proteins. We revealed the specificity and similarity of the parotid gland compared to other digestive glands through comparative analyses of other published scRNA-seq data sets. We also identified the cell-specific expression of hub genes for Sjögren syndrome in the human parotid gland by integrating the results of genome-wide association studies and bulk RNA-seq, which highlighted the importance of immune cell dysfunction in parotid Sjögren syndrome pathogenesis.
Collapse
Affiliation(s)
- M. Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - W. Lin
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Gan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - W. Lu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - M. Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X. Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J. Yi
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z. Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Yamamoto K, Hiraishi M, Haneoka M, Fujinaka H, Yano Y. Protease inhibitor concentrations in the saliva of individuals experiencing oral dryness. BMC Oral Health 2021; 21:661. [PMID: 34930236 PMCID: PMC8686663 DOI: 10.1186/s12903-021-02024-x] [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: 09/17/2021] [Accepted: 12/12/2021] [Indexed: 11/10/2022] Open
Abstract
Background Oral dryness is a common symptom that may interfere with swallowing, chewing, and taste. The most common reason for oral dryness is hyposalivation. Some individuals experiencing oral dryness do not have hyposalivation, however, and the reverse is also true. Here, we focused on healthy individuals with a lower salivary flow rate and evaluated the relationship between the perception of oral dryness and salivary parameters to clarify the cause underlying the perception of oral dryness. Methods A total of 59 participants were divided into 2 groups with a lower or higher salivary flow rate according to the median salivary flow rate. In participants with a lower salivary flow rate, we assessed salivary bacterial counts, protease activities, protein concentrations, oral parameters, and the subjective perception of oral dryness. Results Protease activities and concentrations of protease inhibitors such as cystatin-D and cystatin-SA in the saliva of participants experiencing oral dryness were significantly higher and lower, respectively, than in those not experiencing oral dryness, even though no difference in the salivary flow rate was detected. Salivary cystatin-D and cystatin-SA concentrations correlated negatively with salivary protease activities. Conclusions The composition of salivary protease inhibitors and increased protease activities affect the subjective perception of oral dryness.
Collapse
Affiliation(s)
- Kenkichi Yamamoto
- Personal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, Tokyo, 131-8501, Japan.
| | - Makiko Hiraishi
- Personal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| | - Mai Haneoka
- Analytical Science Research Laboratories, Kao Corporation, Tochigi, Japan
| | - Hidetake Fujinaka
- Personal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| | - Yoshitaka Yano
- Personal Health Care Products Research Laboratories, Kao Corporation, 2-1-3 Bunka, Sumida-ku, Tokyo, 131-8501, Japan
| |
Collapse
|
8
|
Balan P, Chong YS, Lin Q, Lim TK, Li H, Wong ML, Lopez V, He HG, Seneviratne CJ. Isobaric tags for relative and absolute quantification (iTRAQ)-based quantitative analysis of the salivary proteome during healthy pregnancy and pregnancy gingivitis. J Clin Periodontol 2021; 48:1559-1569. [PMID: 34605060 DOI: 10.1111/jcpe.13559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023]
Abstract
AIM The present study aimed to investigate the salivary proteome profiles of pregnant women with gingivitis (PG) or without gingivitis (HP) and non-pregnant healthy controls (HC) by employing iTRAQ-based proteomics. MATERIALS AND METHODS Saliva samples were collected from 30 Chinese women comprising 10 subjects in each of the three groups (PG, HP, and HC). The samples were subjected to iTRAQ-based proteomics analysis, and ELISA was performed to validate the results. The subsequent observations were validated in a cohort of 48 subjects. RESULTS Pathways associated with neutrophil-mediated immune response and antioxidant defence mechanism were significantly higher in PG than HC. The abundance of salivary cystatins (S, SA, and SN) and antimicrobials were significantly decreased in PG and HP, while cystatin C and D were additionally decreased in PG. Cystatin C was mapped to all the major catabolic pathways and was the most re-wired protein in pregnancy gingivitis. Further validation demonstrated cystatin C to be significantly lower in PG than HC. CONCLUSIONS While the decrease in levels of salivary cystatins and antimicrobial proteins may predispose healthy pregnant women to pregnancy gingivitis, it may cause persistence of inflammation in pregnant women with gingivitis.
Collapse
Affiliation(s)
- Preethi Balan
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center, Singapore.,Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Yap S Chong
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University Hospital, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Teck Kwang Lim
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore
| | - Huihua Li
- Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| | - Mun L Wong
- Discipline of Primary Dental Care and Population Oral Health, Faculty of Dentistry, National University of Singapore, Singapore
| | - Violeta Lopez
- School of Nursing, Hubei University of Medicine, Shiyan City, Hubei Province, China
| | - Hong Gu He
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chaminda Jayampath Seneviratne
- Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Center, Singapore.,Oral Health Academic Clinical Program, Duke NUS Medical School, Singapore
| |
Collapse
|
9
|
Siqueira WL, Canales MP, Crosara KTB, Marin LM, Xiao Y. Proteome difference among the salivary proteins adsorbed onto metallic orthodontic brackets and hydroxyapatite discs. PLoS One 2021; 16:e0254909. [PMID: 34319997 PMCID: PMC8318307 DOI: 10.1371/journal.pone.0254909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 07/06/2021] [Indexed: 11/18/2022] Open
Abstract
The aim of this study was to investigate the atomic composition and the proteome of the salivary proteins adsorbed on the surface of orthodontic metallic bracket. For this, the atomic composition of orthodontic metallic brackets was analyzed with X-ray Photoelectron Spectroscopy (XPS). The acquired bracket pellicle was characterized after brackets were immersed in human whole saliva supernatant for 2 hours at 37°C. Hydroxyapatite (HA) discs were used as a control. Acquired pellicle was harvested from the HA discs (n = 12) and from the metallic brackets (n = 12). Proteomics based on mass spectrometry technology was used for salivary protein identification and characterization. Results showed that most of the proteins adsorbed on the surface of orthodontic metallic brackets and on the HA discs were identified specifically to each group, indicating a small overlapping between the salivary proteins on each study group. A total of 311 proteins present on the HA discs were unique to this group while 253 proteins were unique to metallic brackets, and only 45 proteins were common to the two groups. Even though most proteins were unique to each study group, proteins related to antimicrobial activity, lubrication, and remineralization were present in both groups. These findings demonstrate that the salivary proteins adsorbed on the bracket surface are dependent on the material molecular composition.
Collapse
Affiliation(s)
- Walter Luiz Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
- * E-mail:
| | - Maria Pia Canales
- Schulich Dentistry & Medicine, The University of Western Ontario, London, ON, Canada
| | | | - Lina Maria Marin
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yizhi Xiao
- Schulich Dentistry & Medicine, The University of Western Ontario, London, ON, Canada
| |
Collapse
|
10
|
Bescos R, Brookes ZL, Belfield LA, Fernandez-Sanjurjo M, Casas-Agustench P. Modulation of oral microbiota: A new frontier in exercise supplementation. PHARMANUTRITION 2020. [DOI: 10.1016/j.phanu.2020.100230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
11
|
Panwar H, Rokana N, Aparna SV, Kaur J, Singh A, Singh J, Singh KS, Chaudhary V, Puniya AK. Gastrointestinal stress as innate defence against microbial attack. J Appl Microbiol 2020; 130:1035-1061. [PMID: 32869386 DOI: 10.1111/jam.14836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/09/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022]
Abstract
The human gastrointestinal (GI) tract has been bestowed with the most difficult task of protecting the underlying biological compartments from the resident commensal flora and the potential pathogens in transit through the GI tract. It has a unique environment in which several defence tactics are at play while maintaining homeostasis and health. The GI tract shows myriad number of environmental extremes, which includes pH variations, anaerobic conditions, nutrient limitations, elevated osmolarity etc., which puts a check to colonization and growth of nonfriendly microbial strains. The GI tract acts as a highly selective barrier/platform for ingested food and is the primary playground for balance between the resident and uninvited organisms. This review focuses on antimicrobial defense mechanisms of different sections of human GI tract. In addition, the protective mechanisms used by microbes to combat the human GI defence systems are also discussed. The ability to survive this innate defence mechanism determines the capability of probiotic or pathogen strains to confer health benefits or induce clinical events respectively.
Collapse
Affiliation(s)
- H Panwar
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - N Rokana
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - S V Aparna
- Department of Dairy Microbiology, College of Dairy Science and Technology, Kerala Veterinary and Animal Science University, Mannuthy, Thrissur, India
| | - J Kaur
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - A Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - J Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - K S Singh
- Structure and Function of Proteins, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - V Chaudhary
- Department of Microbiology, Punjab Agriculture University, Ludhiana, Punjab, India
| | - A K Puniya
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, India
| |
Collapse
|
12
|
Sant’Anna MDL, Oliveira LT, Gomes DV, Marques STF, Provance DW, Sorenson MM, Salerno VP. Physical exercise stimulates salivary secretion of cystatins. PLoS One 2019; 14:e0224147. [PMID: 31648256 PMCID: PMC6874361 DOI: 10.1371/journal.pone.0224147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/07/2019] [Indexed: 11/18/2022] Open
Abstract
Physical exercise is known to activate the sympathetic nervous system, which influences the production of saliva from salivary glands. Our examination of saliva collected from highly trained athletes before and after a number of physical competititions showed an increase in the secretion of S-type cystatins and cystatin C as a subacute response to aerobic and anaerobic exercise. The elevation in salivary cystatins was transient and the recovery time course differed from that of amylase and other salivary proteins. An in vitro assay was developed based on a cell line from a human submandibular gland (HSG) that differentiated into acinus-like structures. Treatments with the β-adrenergic agonist isoproterenol caused a shift in the intracellular distribution of S-type cystatins and cystatin C, promoting their accumulation at the outer regions of the acinus prior to release and suggesting the activation of a directional transport involving co-migration of both molecules. In another treatment using non-differentiated HSG cells, it was evident that both expression and secretion of cystatin C increased upon addition of the β-adrenergic agonist, and these effects were essentially eliminated by the antagonist propranolol. The HSG cell line appears to have potential as a model for exploring the mechanism of cystatin secretion, particularly the S-type cystatins that originate primarily in the submandibular glands.
Collapse
Affiliation(s)
- Marcelo de Lima Sant’Anna
- Institute of Medical Biochemistry, Federal University of Rio de Janeiro,
Rio de Janeiro, Brazil
- Department of Physical Activity Biosciences, Federal University of Rio de
Janeiro, Rio de Janeiro, Brazil
- Almirante Sylvio de Carmargo Training Center, Brazilian Navy, Rio de
Janeiro, Brazil
| | | | - Diego Viana Gomes
- Department of Physical Activity Biosciences, Federal University of Rio de
Janeiro, Rio de Janeiro, Brazil
| | | | - D. William Provance
- Center for Technological Development in Health, Oswaldo Cruz Insitute,
Rio de Janeiro, Brazil
| | | | - Verônica Pinto Salerno
- Department of Physical Activity Biosciences, Federal University of Rio de
Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Techatanawat S, Surarit R, Chairatvit K, Roytrakul S, Khovidhunkit W, Thanakun S, Izumi Y, Khovidhunkit SOP. Salivary and serum cystatin SA levels in patients with type 2 diabetes mellitus or diabetic nephropathy. Arch Oral Biol 2019; 104:67-75. [PMID: 31174096 DOI: 10.1016/j.archoralbio.2019.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/07/2019] [Accepted: 05/21/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate putative salivary biomarkers for screening and diagnosis of type 2 diabetes mellitus and diabetic nephropathy. DESIGN Saliva and serum samples were collected from 29 patients with type 2 diabetes, 20 patients with diabetic nephropathy, eight patients with non-diabetic induced nephropathy, and 25 healthy subjects. Initially, pooled unstimulated saliva samples from six sex- and age-matched healthy subjects and six patients with type 2 diabetes were subjected to two-dimensional gel electrophoresis, followed by mass spectrometry. Protein expression of cystatin SA in the saliva of patients with type 2 diabetes was further examined in saliva and serum using enzyme-linked immunosorbent assay (ELISA). RESULTS Two-dimensional gel electrophoresis revealed upregulation of salivary cystatin SA in patients with type 2 diabetes. ELISA showed a weak trend of increasing salivary cystatin SA levels in patients with type 2 diabetes, compared with those levels in healthy subjects. When patients were stratified according to periodontal status, linear regression analyses revealed that salivary cystatin SA levels were associated with Periodontal Screening and Recording (PSR) index (β = 0.297, p < 0.05) when the analysis was adjusted for age, sex, HbA1C, estimated glomerular filtration rate (eGFR), and number of teeth. Serum cystatin SA levels were negatively associated with eGFR (β = -0.534, p < 0.0001) when the analysis was adjusted for age, sex, HbA1C, number of teeth, and PSR index. CONCLUSIONS Salivary cystatin SA was associated with periodontal disease severity; moreover, serum cystatin SA levels could reflect kidney function.
Collapse
Affiliation(s)
- Suteera Techatanawat
- Ph.D. program in Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand; Department of General Dentistry, Faculty of Dentistry, Srinakharinwirot University, Bangkok, Thailand
| | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Kongthawat Chairatvit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Weerapan Khovidhunkit
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supanee Thanakun
- Department of Oral Medicine and Periodontology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Yuichi Izumi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | |
Collapse
|
14
|
The role of natural salivary defences in maintaining a healthy oral microbiota. J Dent 2019; 80 Suppl 1:S3-S12. [DOI: 10.1016/j.jdent.2018.08.010] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/22/2018] [Indexed: 01/19/2023] Open
|
15
|
Sanguansermsri P, Jenkinson HF, Thanasak J, Chairatvit K, Roytrakul S, Kittisenachai S, Puengsurin D, Surarit R. Comparative proteomic study of dog and human saliva. PLoS One 2018; 13:e0208317. [PMID: 30513116 PMCID: PMC6279226 DOI: 10.1371/journal.pone.0208317] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 11/15/2018] [Indexed: 11/18/2022] Open
Abstract
Saliva contains many proteins that have an important role in biological process of the oral cavity and is closely associated with many diseases. Although the dog is a common companion animal, the composition of salivary proteome and its relationship with that of human are unclear. In this study, shotgun proteomics was used to compare the salivary proteomes of 7 Thai village dogs and 7 human subjects. Salivary proteomes revealed 2,532 differentially expressed proteins in dogs and humans, representing various functions including cellular component organization or biogenesis, cellular process, localization, biological regulation, response to stimulus, developmental process, multicellular organismal process, metabolic process, immune system process, apoptosis and biological adhesion. The oral proteomes of dogs and humans were appreciably different. Proteins related to apoptosis processes and biological adhesion were predominated in dog saliva. Drug-target network predictions by STITCH Version 5.0 showed that dog salivary proteins were found to have potential roles in tumorigenesis, anti-inflammation and antimicrobial processes. In addition, proteins related to regeneration and healing processes such as fibroblast growth factor and epidermal growth factor were also up-regulated in dogs. These findings provide new information on dog saliva composition and will be beneficial for the study of dog saliva in diseased and health conditions in the future.
Collapse
Affiliation(s)
- Phutsa Sanguansermsri
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
- Department of Clinical Medicine and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | | | - Jitkamol Thanasak
- Department of Clinical Medicine and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Kongthawat Chairatvit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Sittiruk Roytrakul
- Proteomics Research Laboratory, National Center for Genetic Engineering and Biotechnology, Pathumthani, Thailand
| | - Suthathip Kittisenachai
- Proteomics Research Laboratory, National Center for Genetic Engineering and Biotechnology, Pathumthani, Thailand
| | | | - Rudee Surarit
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
16
|
Grant M, Kilsgård O, Åkerman S, Klinge B, Demmer RT, Malmström J, Jönsson D. The Human Salivary Antimicrobial Peptide Profile according to the Oral Microbiota in Health, Periodontitis and Smoking. J Innate Immun 2018; 11:432-444. [PMID: 30485856 PMCID: PMC6738235 DOI: 10.1159/000494146] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/01/2018] [Indexed: 01/10/2023] Open
Abstract
Antimicrobial peptides (AMPs) are a diverse family of peptides that defend the mucosal surfaces of the oral cavity and other locations. Many AMPs have multiple functions and properties that influence aspects of innate defense and colonization by microorganisms. The human oral cavity is home to the second-most diverse microbiome, and the health of the mouth is influenced by the presence of these bacteria as well as by extrinsic factors such as periodontitis and smoking. This study hypothesized that the AMP profile is different in the presence of extrinsic factors and that this would also be reflected in the bacteria present. The AMP profile was analyzed by quantitative selected-reaction-monitoring mass spectrometry analysis and 40 bacterial species were quantified by DNA-DNA hybridization in saliva donated by 41 individuals. Periodontal status was assessed through dental examination and smoking status through medical charting. Periodontal health (in nonsmokers) was associated with a higher abundance of ribonuclease 7, protachykinin 1, β-defensin 128, lipocalin 1, bactericidal permeability-increasing protein fold-containing family B member 3, and bone-marrow proteoglycan. Nonsmoking periodontal disease was associated with an abundance of neutrophil defensin 1 and cathelicidin. However, 7 AMPs were overabundant in periodontal disease in smokers: adrenomedullin, eosinophil peroxidase, 3 different histones, myeloperoxidase, and neutrophil defensin 1. There were no differentially abundant AMPs in smokers versus nonsmokers with periodontal health. Correlation network inference of healthy nonsmokers, healthy smokers, nonsmoking periodontitis, or smoking periodontitis donors demonstrated very different networks growing in complexity with increasing numbers of stressors. The study highlights the importance of the interaction between the oral cavity and its resident microbiota and how this may be influenced by periodontal disease and smoking.
Collapse
Affiliation(s)
- Melissa Grant
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham and Birmingham Community Healthcare Foundation Trust, Birmingham, United Kingdom
| | - Ola Kilsgård
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sigvard Åkerman
- Swedish Dental Service of Skåne, Lund, Sweden
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Björn Klinge
- Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Solna, Sweden
- Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Ryan T Demmer
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Johan Malmström
- Division of Infection Medicine, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Daniel Jönsson
- Swedish Dental Service of Skåne, Lund, Sweden,
- Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden,
| |
Collapse
|
17
|
Midha A, Janek K, Niewienda A, Henklein P, Guenther S, Serra DO, Schlosser J, Hengge R, Hartmann S. The Intestinal Roundworm Ascaris suum Releases Antimicrobial Factors Which Interfere With Bacterial Growth and Biofilm Formation. Front Cell Infect Microbiol 2018; 8:271. [PMID: 30131945 PMCID: PMC6090379 DOI: 10.3389/fcimb.2018.00271] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022] Open
Abstract
Ascariasis is a widespread soil-transmitted helminth infection caused by the intestinal roundworm Ascaris lumbricoides in humans, and the closely related Ascaris suum in pigs. Progress has been made in understanding interactions between helminths and host immune cells, but less is known concerning the interactions of parasitic nematodes and the host microbiota. As the host microbiota represents the direct environment for intestinal helminths and thus a considerable challenge, we studied nematode products, including excretory-secretory products (ESP) and body fluid (BF), of A. suum to determine their antimicrobial activities. Antimicrobial activities against gram-positive and gram-negative bacterial strains were assessed by the radial diffusion assay, while effects on biofilm formation were assessed using the crystal violet static biofilm and macrocolony assays. In addition, bacterial neutralizing activity was studied by an agglutination assay. ESP from different A. suum life stages (in vitro-hatched L3, lung-stage L3, L4, and adult) as well as BF from adult males were analyzed by mass spectrometry. Several proteins and peptides with known and predicted roles in nematode immune defense were detected in ESP and BF samples, including members of A. suum antibacterial factors (ASABF) and cecropin antimicrobial peptide families, glycosyl hydrolase enzymes such as lysozyme, as well as c-type lectin domain-containing proteins. Native, unconcentrated nematode products from intestine-dwelling L4-stage larvae and adults displayed broad-spectrum antibacterial activity. Additionally, adult A. suum ESP interfered with biofilm formation by Escherichia coli, and caused bacterial agglutination. These results indicate that A. suum uses a variety of factors with broad-spectrum antibacterial activity to affirm itself within its microbe-rich environment in the gut.
Collapse
Affiliation(s)
- Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Katharina Janek
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Shared Facility for Mass Spectrometry, Berlin, Germany
| | - Agathe Niewienda
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Shared Facility for Mass Spectrometry, Berlin, Germany
| | - Petra Henklein
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Berlin, Germany
| | - Sebastian Guenther
- Department of Veterinary Medicine, Institute of Animal Hygiene and Environmental Health, Freie Universität Berlin, Berlin, Germany.,Department of Pharmaceutical Biology, Institute of Pharmacy, Ernst-Moritz-Arndt-Universität Greifswald, Greifswald, Germany
| | - Diego O Serra
- Institute of Biology/Microbiology, Humboldt-Universität-zu-Berlin, Berlin, Germany
| | - Josephine Schlosser
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| | - Regine Hengge
- Institute of Biology/Microbiology, Humboldt-Universität-zu-Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
18
|
Mookherjee N, Piyadasa H, Ryu MH, Rider C, Ezzati P, Spicer V, Carlsten C. Inhaled diesel exhaust alters the allergen-induced bronchial secretome in humans. Eur Respir J 2018; 51:51/1/1701385. [DOI: 10.1183/13993003.01385-2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/30/2017] [Indexed: 12/31/2022]
Abstract
Diesel exhaust (DE) is a paradigm for traffic-related air pollution. Human adaptation to DE is poorly understood and currently based on oversimplified models. DE promotes allergic responses, but protein expression changes mediated by this interaction have not been systematically investigated. The aim of this study was to define the effect of inhaled DE on allergen-induced proteins in the lung.We performed a randomised and blinded controlled human crossover exposure study. Participants inhaled filtered air or DE; thereafter, contralateral lung segments were challenged with allergen or saline. Using label-free quantitative proteomics, we comprehensively defined DE-mediated alteration of allergen-driven secreted proteins (secretome) in bronchoalveolar lavage. We further examined expression of proteins selected from the secretome data in independent validation experiments using Western blots, ELISA and immunohistochemistry.We identified protein changes unique to co-exposure (DE+allergen), undetected with mono-exposures (DE or allergen alone). Validation studies confirmed that specific proteins (e.g.the antimicrobial peptide cystatin-SA) were significantly enhanced with DE+allergen compared to either mono-exposure.This study demonstrates that common environmental co-exposures can uniquely alter protein responses in the lungs, illuminating biology that mono-exposures cannot. This study highlights the value of complex humanin vivomodels in detailing airway responses to inhaled pollution.
Collapse
|
19
|
Hong SW, Seo DG, Baik JE, Cho K, Yun CH, Han SH. Differential profiles of salivary proteins with affinity to Streptococcus mutans lipoteichoic acid in caries-free and caries-positive human subjects. Mol Oral Microbiol 2014; 29:208-18. [PMID: 24848678 DOI: 10.1111/omi.12057] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2014] [Indexed: 12/15/2022]
Abstract
Streptococcus mutans is a representative oral pathogen that causes dental caries and pulpal inflammation. Its lipoteichoic acid (Sm.LTA) is known to be an important cell-wall virulence factor involved in bacterial adhesion and induction of inflammation. Since Sm.LTA-binding proteins (Sm.LTA-BPs) might play an important role in pathogenesis and host immunity, we identified the Sm.LTA-BPs in the saliva of caries-free and caries-positive human subjects using Sm.LTA-conjugated beads and LTQ-Orbitrap hybrid Fourier transform mass spectrometry. Sm.LTA was conjugated to N-hydroxysuccinimidyl-Sepharose(®) 4 Fast Flow beads (Sm.LTA-beads). Sm.LTA retained its biological properties during conjugation, as determined by the expression of nitric oxide and interferon-γ-inducible protein 10 in a murine macrophage cell line and activation of Toll-like receptor 2 (TLR2) in CHO/CD14/TLR2 cells. Sm.LTA-BPs were isolated from pooled saliva prepared from 10 caries-free or caries-positive human subjects each, electrophoresed to see their differential expression in each group, and further identified by high-resolution mass spectrometry. A total of 8 and 12 Sm.LTA-BPs were identified with statistical significance in the pooled saliva from the caries-free and caries-positive human subjects, respectively. Unique Sm.LTA-BPs found in caries-free saliva included histone H4, profilin-1 and neutrophil defensin-1, and those in caries-positive saliva included cystatin-C, cystatin-SN, cystatin-S, cystatin-D, lysozyme C, calmodulin-like protein 3 and β-actin. The Sm.LTA-BPs found in both groups were hemoglobin subunits α and β, prolactin-inducible protein, protein S100-A9, and SPLUNC2. Collectively, we identified Sm.LTA-BPs in the saliva of caries-free and caries-positive subjects, which exhibit differential protein profiles.
Collapse
Affiliation(s)
- S W Hong
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
20
|
Wang Q, Zhang W, Li H, Aprecio R, Wu W, Lin Y, Li Y. Effects of 25-hydroxyvitamin D3 on cathelicidin production and antibacterial function of human oral keratinocytes. Cell Immunol 2013; 283:45-50. [DOI: 10.1016/j.cellimm.2013.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Revised: 05/29/2013] [Accepted: 06/13/2013] [Indexed: 01/25/2023]
|
21
|
Fine DH, Furgang D, McKiernan M, Rubin M. Can salivary activity predict periodontal breakdown in A. actinomycetemcomitans infected adolescents? Arch Oral Biol 2012; 58:611-20. [PMID: 23219180 DOI: 10.1016/j.archoralbio.2012.10.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Revised: 09/25/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE While Aggregatibacter actinomycetemcomitans (Aa) is highly associated with localised aggressive periodontitis (LAP) many Aa-carriers do not develop LAP. This study was designed to determine whether specific salivary factors could distinguish between subjects who have Aa initially and remain healthy (H/AA) as compared to those who develop LAP (LAP/AA). DESIGN H/AA subjects and healthy controls with no Aa (H) were enrolled in a longitudinal cohort study to investigate initiation of bone loss (LAP) over 3 years. After detection of LAP, stored saliva from 10 H, 10 H/AA, and 10 LAP/AA subjects was thawed, processed, and tested for (1) lactoferrin (Lf) concentration and iron levels; (2) agglutination of Aa; (3) killing of Gram-positive bacteria. RESULTS LAP/AA saliva levels of Lf iron were low prior to and after bone loss (3.6+1.7ngFe/μg) (LAP/AA vs. H and H/AA p≤0.01). Saliva from H/AA subjects caused Aa to agglutinate significantly more than H or LAP/AA saliva (p≤0.01). LAP/AA saliva killed Streptococcus mutans, Streptococcus sanguis and Lactobacillus in vitro by >83%. Saliva from H individuals killed these bacteria by <3.3% (LAP/AA vs. H; p≤0.01). H/AA killing was intermediate. CONCLUSION LAP/AA saliva showed: low levels of Lf iron, minimal Aa agglutinating activity, and high killing activity against Gram-positive bacteria. Aa-positive healthy saliva (H/AA) showed: higher levels of Lf iron, maximal Aa agglutinating activity, and moderate killing of Gram-positive bacteria. A salivary activity profile can distinguish between subjects who are Aa-positive and remain healthy from those who develop LAP.
Collapse
Affiliation(s)
- Daniel H Fine
- Department of Oral Biology, New Jersey Dental School, University of Medicine and Dentistry of New Jersey, Newark, NJ 071003, USA.
| | | | | | | |
Collapse
|