1
|
Remori V, Airoldi M, Alberio T, Fasano M, Azzi L. Prediction of Oral Cancer Biomarkers by Salivary Proteomics Data. Int J Mol Sci 2024; 25:11120. [PMID: 39456901 PMCID: PMC11508456 DOI: 10.3390/ijms252011120] [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: 09/12/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Oral cancer, representing 2-4% of all cancer cases, predominantly consists of Oral Squamous Cell Carcinoma (OSCC), which makes up 90% of oral malignancies. Early detection of OSCC is crucial, and identifying specific proteins in saliva as biomarkers could greatly improve early diagnosis. Here, we proposed a strategy to pinpoint candidate biomarkers. Starting from a list of salivary proteins detected in 10 OSCC patients and 20 healthy controls, we combined a univariate approach and a multivariate approach to select candidates. To reduce the number of proteins selected, a Protein-Protein Interaction network was built to consider only connected proteins. Then, an over-representation analysis (ORA) determined the enriched pathways. The network from 172 differentially abundant proteins highlighted 50 physically connected proteins, selecting relevant candidates for targeted experimental validations. Notably, proteins like Heat shock 70 kDa protein 1A/1B, Pyruvate kinase PKM, and Phosphoglycerate kinase 1 were suggested to be differentially regulated in OSCC patients, with implications for oral carcinogenesis and tumor growth. Additionally, the ORA revealed enrichment in immune system, complement, and coagulation pathways, all known to play roles in tumorigenesis and cancer progression. The employed method has successfully identified potential biomarkers for early diagnosis of OSCC using an accessible body fluid.
Collapse
Affiliation(s)
- Veronica Remori
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy; (V.R.); (T.A.); (M.F.)
| | - Manuel Airoldi
- Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy;
| | - Tiziana Alberio
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy; (V.R.); (T.A.); (M.F.)
| | - Mauro Fasano
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy; (V.R.); (T.A.); (M.F.)
| | - Lorenzo Azzi
- Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy;
| |
Collapse
|
2
|
Seaman WT, Keener O, Nanayakkara D, Mollan KR, Premkumar L, Cuadra EC, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. Sci Rep 2024; 14:21788. [PMID: 39294156 PMCID: PMC11411107 DOI: 10.1038/s41598-024-67504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 07/11/2024] [Indexed: 09/20/2024] Open
Abstract
Oral fluids provide ready detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to evaluate relationships between oral virus, oral and systemic anti-SARS-CoV-2-specific antibodies, and symptoms. Oral fluids (saliva/throat wash (saliva/TW)) and serum were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+ human participants (n = 45). SARS-CoV-2 RT-qPCR and N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR for subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA and ELISA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. At time of enrollment (baseline, BL), LFA-detected N-antigen in 86% of TW and was immunoblot-confirmed. However, only 3/17 were saliva/TW qPCR+ . Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three anti-spike sero-negative participants suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At enrollment, symptomatic participants demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (63%/54%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral and serum IgG correlated 100% with NP+ PCR status. Cough and fatigue severity (p = 0.010 and 0.018 respectively), and presence of weakness, nausea, and composite upper respiratory symptoms (p = 0.037, 0.005, and 0.017, respectively) were negatively associated with saliva IgM but not TW or serum IgM. Throat wash IgM levels were higher in women compared to men, although the association did not reach statistical significance (median: 290 (female) versus 0.697, p = 0.056). Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms and early oral IgM responses during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Olive Keener
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- North Carolina School of Math and Science, Durham, NC, USA
| | - Dinelka Nanayakkara
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katie R Mollan
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Edwing Centeno Cuadra
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Corbin D Jones
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Audrey Pettifor
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natalie M Bowman
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA
| | | | - Jennifer Webster-Cyriaque
- National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
- Division of Oral and Craniofacial Health Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- UNC School of Medicine, University of North Carolina at Chapel Hill, 111 Mason Farm Rd, Medical Biomolecular Research Building, Room 2341b, Chapel Hill, NC, 27599, USA.
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Ebersole JL, Hasturk H, Huber M, Gellibolian R, Markaryan A, Zhang XD, Miller CS. Realizing the clinical utility of saliva for monitoring oral diseases. Periodontol 2000 2024; 95:203-219. [PMID: 39010260 DOI: 10.1111/prd.12581] [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: 03/18/2024] [Revised: 05/01/2024] [Accepted: 05/16/2024] [Indexed: 07/17/2024]
Abstract
In the era of personalized/precision health care, additional effort is being expended to understand the biology and molecular mechanisms of disease processes. How these mechanisms are affected by individual genetics, environmental exposures, and behavioral choices will encompass an expanding role in the future of optimally preventing and treating diseases. Considering saliva as an important biological fluid for analysis to inform oral disease detection/description continues to expand. This review provides an overview of saliva as a diagnostic fluid and the features of various biomarkers that have been reported. We emphasize the use of salivary biomarkers in periodontitis and transport the reader through extant literature, gaps in knowledge, and a structured approach toward validating and determine the utility of biomarkers in periodontitis. A summation of the findings support the likelihood that a panel of biomarkers including both host molecules and specific microorganisms will be required to most effectively identify risk for early transition to disease, ongoing disease activity, progression, and likelihood of response to standard periodontal therapy. The goals would be to develop predictive algorithms that serve as adjunctive diagnostic tools which provide the clinician and patient important information for making informed clinical decisions.
Collapse
Affiliation(s)
- Jeffrey L Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Hatice Hasturk
- Immunology and Inflammation, Center for Clinical and Translational Research, The ADA Forsyth Institute, Cambridge, Massachusetts, USA
| | - Michaell Huber
- Department of Comprehensive Dentistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | | - Xiaohua D Zhang
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Craig S Miller
- Department of Oral Health Practice, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
4
|
Rudzinska-Radecka M, Bańcerowski B, Marczyński R, Mukherjee D, Sikora T, Morawska K, Mielczarek A, Moździerski M, Hajduk B, Kotowicz B. Evaluation of Salivary Biomarkers and Spirometry for Diagnosing COPD in Non-Smokers and Smokers of Polish Origin. Biomedicines 2024; 12:1206. [PMID: 38927413 PMCID: PMC11200520 DOI: 10.3390/biomedicines12061206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory condition with global implications. Accurate and timely diagnosis is critical; however, traditional diagnostic methods (based on spirometry) show limitations, prompting the search for predictive biomarkers and modern diagnostic techniques. This study explored the validation of COPD-related biomarkers (C-reactive protein, procalcitonin, neutrophil elastase, and alpha-1 antitrypsin) in saliva. A diverse cohort, including healthy non-smokers, healthy smokers, and COPD patients of Polish origin, underwent spirometry and marker analysis. The data correlated with clinical factors, revealing noteworthy relations. Firstly, salivary biomarker levels were compared with serum concentrations, demonstrating notable positive or negative correlations, depending on the factor. Further analysis within healthy individuals revealed associations between biomarker levels, spirometry, and clinical characteristics such as age, sex, and BMI. Next, COPD patients exhibited an enhanced concentration of biomarkers compared to healthy groups. Finally, the study introduced a breathing assessment survey, unveiling significant associations between self-perceived breathing and spirometric and tested parameters. Outcomes emphasized the relevance of subjective experiences in COPD research. In conclusion, this research underscored the potential of salivary biomarkers as diagnostic tools for COPD, offering a non-invasive and accessible alternative to traditional methods. The findings paved the way for improved modern diagnostic approaches.
Collapse
Affiliation(s)
- Magdalena Rudzinska-Radecka
- Recumed Ltd., 05-092 Łomianki, Poland
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warszawa, Poland
| | | | | | - Debjita Mukherjee
- Institute of Physical Chemistry, Polish Academy of Sciences, 01-224 Warszawa, Poland
| | - Tomasz Sikora
- Military Institute of Chemistry and Radiometry, 00-910 Warsaw, Poland (K.M.)
| | - Karolina Morawska
- Military Institute of Chemistry and Radiometry, 00-910 Warsaw, Poland (K.M.)
| | - Agnieszka Mielczarek
- Department of Conservative Dentistry and Endodontics, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | | | - Bogdan Hajduk
- TS Out-Patients Clinic for Cardiovascular and Pulmonary Diseases, 01-460 Warsaw, Poland;
| | - Beata Kotowicz
- Cancer Biomarker and Cytokines Laboratory Unit, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| |
Collapse
|
5
|
Moreno E, Ciordia S, Fátima SM, Jiménez D, Martínez-Sanz J, Vizcarra P, Ron R, Sánchez-Conde M, Bargiela R, Sanchez-Carrillo S, Moreno S, Corrales F, Ferrer M, Serrano-Villar S. Proteomic snapshot of saliva samples predicts new pathways implicated in SARS-CoV-2 pathogenesis. Clin Proteomics 2024; 21:37. [PMID: 38778280 PMCID: PMC11112864 DOI: 10.1186/s12014-024-09482-9] [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: 10/13/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Information on the microbiome's human pathways and active members that can affect SARS-CoV-2 susceptibility and pathogenesis in the salivary proteome is very scarce. Here, we studied a unique collection of samples harvested from April to June 2020 from unvaccinated patients. METHODS We compared 10 infected and hospitalized patients with severe (n = 5) and moderate (n = 5) coronavirus disease (COVID-19) with 10 uninfected individuals, including non-COVID-19 but susceptible individuals (n = 5) and non-COVID-19 and nonsusceptible healthcare workers with repeated high-risk exposures (n = 5). RESULTS By performing high-throughput proteomic profiling in saliva samples, we detected 226 unique differentially expressed (DE) human proteins between groups (q-value ≤ 0.05) out of 3376 unambiguously identified proteins (false discovery rate ≤ 1%). Major differences were observed between the non-COVID-19 and nonsusceptible groups. Bioinformatics analysis of DE proteins revealed human proteomic signatures related to inflammatory responses, central cellular processes, and antiviral activity associated with the saliva of SARS-CoV-2-infected patients (p-value ≤ 0.0004). Discriminatory biomarker signatures from human saliva include cystatins, protective molecules present in the oral cavity, calprotectins, involved in cell cycle progression, and histones, related to nucleosome functions. The expression levels of two human proteins related to protein transport in the cytoplasm, DYNC1 (p-value, 0.0021) and MAPRE1 (p-value, 0.047), correlated with angiotensin-converting enzyme 2 (ACE2) plasma activity. Finally, the proteomes of microorganisms present in the saliva samples showed 4 main microbial functional features related to ribosome functioning that were overrepresented in the infected group. CONCLUSION Our study explores potential candidates involved in pathways implicated in SARS-CoV-2 susceptibility, although further studies in larger cohorts will be necessary.
Collapse
Affiliation(s)
- Elena Moreno
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Sergio Ciordia
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Santos Milhano Fátima
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Daniel Jiménez
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
| | - Javier Martínez-Sanz
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Pilar Vizcarra
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Raquel Ron
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Matilde Sánchez-Conde
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Rafael Bargiela
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, LL57 2UW, UK
| | - Sergio Sanchez-Carrillo
- Instituto de Catalisis y Petroleoquimica (ICP), CSIC, 28049, Madrid, Spain
- Centro de Biologia Molecular Severo Ochoa (CBM), CSIC-UAM, 28049, Madrid, Spain
| | - Santiago Moreno
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Facultad de Medicina, Universidad de Alcalá de Henares, 28801, Alcalá de Henares, Madrid, Spain
| | - Fernando Corrales
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Manuel Ferrer
- Instituto de Catalisis y Petroleoquimica (ICP), CSIC, 28049, Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
6
|
Kainat R, Ahmed I, Alolaywi AM, Waheed H, Sultan ZK, Moin SF. Assessment of Salivary MMP-8 and IL-1β for the Diagnosis of Periodontal Diseases in Pakistani Population. Eur J Dent 2024; 18:672-679. [PMID: 38086426 PMCID: PMC11132764 DOI: 10.1055/s-0043-1772779] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
OBJECTIVE Clinical methods use the subjective diagnosis of periodontal diseases by visual observation that could result in differences and variability of diagnosis. The addition of specific markers could aid in the accurate diagnosis of the local population. The objective of the study was to target two of the major proteins for possible significance in such an approach. MATERIALS AND METHODS Unstimulated saliva samples were collected from 60 participants aged between 18 and 70 years. Three groups each with twenty participants were recruited into periodontitis, gingivitis, and healthy control. STATISTICAL ANALYSIS The samples were analyzed using human enzyme-linked immunosorbent assay kits for matrix metalloproteinase-8 (MMP-8) and interleukin-1β (IL-1β). RESULTS SPSS version 20 was used to analyze the result. Posthoc analysis by Tukey's test revealed that MMP-8 levels were higher in gingivitis and periodontitis groups as compared with healthy controls. The test also revealed that IL-1β levels were higher in the periodontitis group compared with the healthy control and gingivitis group. Additionally, one-way analysis of variance analysis showed a significant effect on probing depth in gingivitis and periodontitis patients. The mean age of periodontitis group was significantly higher than other groups. CONCLUSION Salivary biomarkers may provide useful diagnostic information and could be utilized as tests for periodontal disease screening, prognosis, and prediction.
Collapse
Affiliation(s)
- Rida Kainat
- Department of Biochemistry, Baqai Medical University, Karachi, Pakistan
| | - Iftikhar Ahmed
- Department of Biochemistry, Baqai Medical University, Karachi, Pakistan
| | | | - Humera Waheed
- Dow College of Biotechnology, Dow University of Health Sciences, Karachi, Pakistan
| | - Zohaib Khurshid Sultan
- Department of Prosthodontics and Dental Implantology, College of Dentistry, King Faisal University, Al-Ahsa, Saudia Arabia
- Department of Anatomy, Faculty of Dentistry, Center of Excellence for Regenerative Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Syed Faraz Moin
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi, Pakistan
| |
Collapse
|
7
|
Gürsoy UK, Özdemir Kabalak M, Gürsoy M. Advances in periodontal biomarkers. Adv Clin Chem 2024; 120:145-168. [PMID: 38762240 DOI: 10.1016/bs.acc.2024.03.003] [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] [Indexed: 05/20/2024]
Abstract
Due to technologic advancements, periodontology has witnessed a boost in biomarker research over the past three decades. Indeed, with the aid of omics, our understanding of the healthy periodontium, pathogenesis of periodontal diseases, and healing after periodontal treatment has improved significantly. Yet, the traditional methods, periodontal probing and radiographies, remain the most common methods to diagnose periodontal disease and monitor treatment. Although these approaches can produce reliable diagnostic outcomes, they generally detect disease only after significant tissue degradation thus making treatment outcome highly uncertain. Accordingly, laboratories worldwide have collaborated with clinicians to design accurate, rapid and cost-effective biomarkers for periodontal disease diagnosis. Despite these efforts, biomarkers that can be widely used in early disease diagnosis and for treatment outcome prediction are far from daily use. The aim of this chapter is to give a general overview on periodontal health and diseases, and review recent advancements in periodontal biomarker research. A second aim will discuss the strengths and limitations of translating periodontal biomarker research to clinical practice. Genetic biomarkers of periodontitis are not discussed as the available confirmatory data is scarce.
Collapse
Affiliation(s)
| | | | - Mervi Gürsoy
- Periodontology, Institute of Dentistry, University of Turku, Turku, Finland; Oral Health Care, Welfare Division, City of Turku, Turku, Finland
| |
Collapse
|
8
|
Jiao LL, Dong HL, Liu MM, Wu PL, Cao Y, Zhang Y, Gao FG, Zhu HY. The potential roles of salivary biomarkers in neurodegenerative diseases. Neurobiol Dis 2024; 193:106442. [PMID: 38382884 DOI: 10.1016/j.nbd.2024.106442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
Current research efforts on neurodegenerative diseases are focused on identifying novel and reliable biomarkers for early diagnosis and insight into disease progression. Salivary analysis is gaining increasing interest as a promising source of biomarkers and matrices for measuring neurodegenerative diseases. Saliva collection offers multiple advantages over the currently detected biofluids as it is easily accessible, non-invasive, and repeatable, allowing early diagnosis and timely treatment of the diseases. Here, we review the existing findings on salivary biomarkers and address the potential value in diagnosing neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Amyotrophic lateral sclerosis. Based on the available research, β-amyloid, tau protein, α-synuclein, DJ-1, Huntington protein in saliva profiles display reliability and validity as the biomarkers of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ling-Ling Jiao
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China
| | - Hui-Lin Dong
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Meng-Meng Liu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Peng-Lin Wu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Yi Cao
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Yuan Zhang
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China
| | - Fu-Gao Gao
- Xuzhou Cigarette Factory, China Tobacco Jiangsu Industrial Co Ltd, Xuzhou 221005, China.
| | - Huai-Yuan Zhu
- China Tobacco Jiangsu Industrial Co Ltd, Nanjing 210019, China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, China.
| |
Collapse
|
9
|
Ferrari E, Gallo M, Spisni A, Antonelli R, Meleti M, Pertinhez TA. Human Serum and Salivary Metabolomes: Diversity and Closeness. Int J Mol Sci 2023; 24:16603. [PMID: 38068926 PMCID: PMC10706786 DOI: 10.3390/ijms242316603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Saliva, which contains molecular information that may reflect an individual's health status, has become a valuable tool for discovering biomarkers of oral and general diseases. Due to the high vascularization of the salivary glands, there is a molecular exchange between blood and saliva. However, the composition of saliva is complex and influenced by multiple factors. This study aimed to investigate the possible relationships between the salivary and serum metabolomes to gain a comprehensive view of the metabolic phenotype under physiological conditions. Using 1H-NMR spectroscopy, we obtained the serum metabolite profiles of 20 healthy young individuals and compared them with the metabolomes of parotid, submandibular/sublingual, and whole-saliva samples collected concurrently from the same individuals using multivariate and univariate statistical analysis. Our results show that serum is more concentrated and less variable for most of the shared metabolites than the three saliva types. While we found moderate to strong correlations between serum and saliva concentrations of specific metabolites, saliva is not simply an ultrafiltrate of blood. The intense oral metabolism prevents very strong correlations between serum and salivary concentrations. This study contributes to a better understanding of salivary metabolic composition, which is crucial for utilizing saliva in laboratory diagnostics.
Collapse
Affiliation(s)
- Elena Ferrari
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.F.); (A.S.); (T.A.P.)
| | - Mariana Gallo
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.F.); (A.S.); (T.A.P.)
| | - Alberto Spisni
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.F.); (A.S.); (T.A.P.)
| | - Rita Antonelli
- Centro Universitario Odontoiatria, University of Parma, 43126 Parma, Italy; (R.A.); (M.M.)
| | - Marco Meleti
- Centro Universitario Odontoiatria, University of Parma, 43126 Parma, Italy; (R.A.); (M.M.)
| | - Thelma A. Pertinhez
- Laboratory of Biochemistry and Metabolomics, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (E.F.); (A.S.); (T.A.P.)
| |
Collapse
|
10
|
Diaz PM, Leehans A, Ravishankar P, Daily A. Multiomic Approaches for Cancer Biomarker Discovery in Liquid Biopsies: Advances and Challenges. Biomark Insights 2023; 18:11772719231204508. [PMID: 37846373 PMCID: PMC10576933 DOI: 10.1177/11772719231204508] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
Cancer is a complex and heterogeneous disease that poses a significant threat to global health. Early diagnosis and treatment are critical for improving patient outcomes, and the use of liquid biopsies has emerged as a promising approach for cancer detection and monitoring. Traditionally, cancer diagnosis has relied on invasive tissue biopsies, the collection of which can prove challenging for patients and the results of which may not always provide accurate results due to tumor heterogeneity. Liquid biopsies have gained increasing attention as they provide a non-invasive and accessible source of cancer biomarkers, which can be used to diagnose cancer, monitor treatment response, and detect relapse. The integration of -omics technologies, such as proteomics, genomics, and metabolomics, has further enhanced the capabilities of liquid biopsies by introducing precision oncology and enabling the tailoring of treatment for individual patients based on their unique tumor biology. In this review, we will discuss the challenges and advances in the field of cancer liquid biopsies and the integration of -omics technologies for different types of liquid biopsies, including blood, tear, urine, sweat, saliva, and cerebrospinal fluid.
Collapse
Affiliation(s)
- Paola Monterroso Diaz
- Namida Lab Inc., Fayetteville, AR, USA
- University of Arkansas, Department of Biomedical Engineering, Fayetteville, AR, USA
| | | | | | | |
Collapse
|
11
|
Baker JL. Illuminating the oral microbiome and its host interactions: recent advancements in omics and bioinformatics technologies in the context of oral microbiome research. FEMS Microbiol Rev 2023; 47:fuad051. [PMID: 37667515 PMCID: PMC10503653 DOI: 10.1093/femsre/fuad051] [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: 01/31/2023] [Revised: 08/02/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
The oral microbiota has an enormous impact on human health, with oral dysbiosis now linked to many oral and systemic diseases. Recent advancements in sequencing, mass spectrometry, bioinformatics, computational biology, and machine learning are revolutionizing oral microbiome research, enabling analysis at an unprecedented scale and level of resolution using omics approaches. This review contains a comprehensive perspective of the current state-of-the-art tools available to perform genomics, metagenomics, phylogenomics, pangenomics, transcriptomics, proteomics, metabolomics, lipidomics, and multi-omics analysis on (all) microbiomes, and then provides examples of how the techniques have been applied to research of the oral microbiome, specifically. Key findings of these studies and remaining challenges for the field are highlighted. Although the methods discussed here are placed in the context of their contributions to oral microbiome research specifically, they are pertinent to the study of any microbiome, and the intended audience of this includes researchers would simply like to get an introduction to microbial omics and/or an update on the latest omics methods. Continued research of the oral microbiota using omics approaches is crucial and will lead to dramatic improvements in human health, longevity, and quality of life.
Collapse
Affiliation(s)
- Jonathon L Baker
- Department of Oral Rehabilitation & Biosciences, School of Dentistry, Oregon Health & Science University, 3181 Sam Jackson Park Road, Portland, OR 97202, United States
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA 92037, United States
- Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA 92093, United States
| |
Collapse
|
12
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. RESEARCH SQUARE 2023:rs.3.rs-3154698. [PMID: 37645853 PMCID: PMC10462189 DOI: 10.21203/rs.3.rs-3154698/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
Affiliation(s)
- William T Seaman
- National Institute of Dental and Craniofacial Research, National Institutes of Health
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Messana I, Manconi B, Cabras T, Boroumand M, Sanna MT, Iavarone F, Olianas A, Desiderio C, Rossetti DV, Vincenzoni F, Contini C, Guadalupi G, Fiorita A, Faa G, Castagnola M. The Post-Translational Modifications of Human Salivary Peptides and Proteins Evidenced by Top-Down Platforms. Int J Mol Sci 2023; 24:12776. [PMID: 37628956 PMCID: PMC10454625 DOI: 10.3390/ijms241612776] [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: 06/19/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
In this review, we extensively describe the main post-translational modifications that give rise to the multiple proteoforms characterized to date in the human salivary proteome and their potential role. Most of the data reported were obtained by our group in over twenty-five years of research carried out on human saliva mainly by applying a top-down strategy. In the beginning, we describe the products generated by proteolytic cleavages, which can occur before and after secretion. In this section, the most relevant families of salivary proteins are also described. Next, we report the current information concerning the human salivary phospho-proteome and the limited news available on sulfo-proteomes. Three sections are dedicated to the description of glycation and enzymatic glycosylation. Citrullination and N- and C-terminal post-translational modifications (PTMs) and miscellaneous other modifications are described in the last two sections. Results highlighting the variation in the level of some proteoforms in local or systemic pathologies are also reviewed throughout the sections of the manuscript to underline the impact and relevance of this information for the development of new diagnostic biomarkers useful in clinical practice.
Collapse
Affiliation(s)
- Irene Messana
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Barbara Manconi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Tiziana Cabras
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | | | - Maria Teresa Sanna
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Federica Iavarone
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Alessandra Olianas
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Claudia Desiderio
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Diana Valeria Rossetti
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, Consiglio Nazionale delle Ricerche, 00168 Rome, Italy; (I.M.); (C.D.); (D.V.R.)
| | - Federica Vincenzoni
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.I.); (F.V.)
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
| | - Cristina Contini
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Giulia Guadalupi
- Department of Life and Environmental Sciences, University of Cagliari, 09124 Cagliari, Italy; (B.M.); (M.T.S.); (A.O.); (C.C.); (G.G.)
| | - Antonella Fiorita
- Fondazione Policlinico Universitario A. Gemelli Fondazione IRCCS, 00168 Rome, Italy;
- Dipartimento di Scienze dell’Invecchiamento, Neurologiche, Ortopediche e della Testa e del Collo, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gavino Faa
- Unit of Pathology, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Massimo Castagnola
- Proteomics Laboratory, European Center for Brain Research, (IRCCS) Santa Lucia Foundation, 00168 Rome, Italy;
| |
Collapse
|
14
|
Seaman WT, Keener O, Mei W, Mollan KR, Jones CD, Pettifor A, Bowman NM, Wang F, Webster-Cyriaque J. Oral SARS-CoV-2 host responses predict the early COVID-19 disease course. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.06.23286853. [PMID: 37609199 PMCID: PMC10441495 DOI: 10.1101/2023.03.06.23286853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Objectives Oral fluids provide ready detection of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host responses. This study sought to determine relationships between oral virus, oral anti-SARS-CoV-2-specific antibodies, and symptoms. Methods Saliva/throat wash (saliva/TW) were collected from asymptomatic and symptomatic, nasopharyngeal (NP) SARS-CoV-2 RT-qPCR+, subjects (n=47). SARS-CoV-2 RT-qPCR, N-antigen detection by immunoblot and lateral flow assay (LFA) were performed. RT-qPCR targeting viral subgenomic RNA (sgRNA) was sequence confirmed. SARS-CoV-2-anti-S protein RBD LFA assessed IgM and IgG responses. Structural analysis identified host salivary molecules analogous to SARS-CoV-2-N-antigen. Statistical analyses were performed. Results At baseline, LFA-detected N-antigen was immunoblot-confirmed in 82% of TW. However, only 3/17 were saliva/TW qPCR+. Sixty percent of saliva and 83% of TW demonstrated persistent N-antigen at 4 weeks. N-antigen LFA signal in three negative subjects suggested potential cross-detection of 4 structurally analogous salivary RNA binding proteins (alignment 19-29aa, RMSD 1-1.5 Angstroms). At entry, symptomatic subjects demonstrated replication-associated sgRNA junctions, were IgG+ (94%/100% in saliva/TW), and IgM+ (75%/63%). At 4 weeks, SARS-CoV-2 IgG (100%/83%) and IgM (80%/67%) persisted. Oral IgG correlated 100% with NP+PCR status. Cough and fatigue severity (p=0.0008 and 0.016), and presence of nausea, weakness, and composite upper respiratory symptoms (p=0.005, 0.037 and 0.017) were negatively associated with oral IgM. Female oral IgM levels were higher than male (p=0.056). Conclusion Important to transmission and disease course, oral viral replication and persistence showed clear relationships with select symptoms, early Ig responses, and gender during early infection. N-antigen cross-reactivity may reflect mimicry of structurally analogous host proteins.
Collapse
|
15
|
Jang H, Choudhury S, Yu Y, Sievers BL, Gelbart T, Singh H, Rawlings SA, Proal A, Tan GS, Qian Y, Smith D, Freire M. Persistent immune and clotting dysfunction detected in saliva and blood plasma after COVID-19. Heliyon 2023; 9:e17958. [PMID: 37483779 PMCID: PMC10362241 DOI: 10.1016/j.heliyon.2023.e17958] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/21/2023] [Accepted: 07/03/2023] [Indexed: 07/25/2023] Open
Abstract
A growing number of studies indicate that coronavirus disease 2019 (COVID-19) is associated with inflammatory sequelae, but molecular signatures governing the normal versus pathologic convalescence process have not been well-delineated. Here, we characterized global immune and proteome responses in matched plasma and saliva samples obtained from COVID-19 patients collected between 20 and 90 days after initial clinical symptoms resolved. Convalescent subjects showed robust total IgA and IgG responses and positive antibody correlations in saliva and plasma samples. Shotgun proteomics revealed persistent inflammatory patterns in convalescent samples including dysfunction of salivary innate immune cells, such as neutrophil markers (e.g., myeloperoxidase), and clotting factors in plasma (e.g., fibrinogen), with positive correlations to acute COVID-19 disease severity. Saliva samples were characterized by higher concentrations of IgA, and proteomics showed altered myeloid-derived pathways that correlated positively with SARS-CoV-2 IgA levels. Beyond plasma, our study positions saliva as a viable fluid to monitor normal and aberrant immune responses including vascular, inflammatory, and coagulation-related sequelae.
Collapse
Affiliation(s)
- Hyesun Jang
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | | | - Yanbao Yu
- Department of Chemistry & Biochemistry, University of Delaware, Newark, DE, USA, 19716
| | - Benjamin L Sievers
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Terri Gelbart
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Harinder Singh
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Stephen A Rawlings
- MMP Adult Infectious Disease, Maine Medical Center, South Portland, ME, 04106, USA
| | - Amy Proal
- PolyBio Research Foundation. Mercer Island, WA, USA
| | - Gene S Tan
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Yu Qian
- Informatics, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
| | - Davey Smith
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marcelo Freire
- Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, and Rockville, MD, USA
- Division of Infectious Diseases and Global Public Health Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
16
|
Cofré-Leiva C, Camargo-Ayala PA, Vergara-Pérez A, Hernández-Olivos R, Sanhueza S, Nova-Lamperti E, Zúñiga-Hernández J, Rivera C. Salivary proteins offer insights into keratinocyte death during aphthous stomatitis. A case-crossover study. BMC Oral Health 2023; 23:279. [PMID: 37170213 PMCID: PMC10176878 DOI: 10.1186/s12903-023-02955-7] [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: 01/18/2023] [Accepted: 04/09/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND The death of oral keratinocytes is a crucial step in the emergence of recurrent aphthous stomatitis (RAS, also known as aphthae or aphthous ulcers). Since there are no experimental models available to research aphthous ulcers, little is understood about this process. We hypothesize that saliva can be a data bank of information that offers insights on epithelial damage. METHODS In this case-crossover study, we assessed the salivary proteome of patients with RAS (n = 36) in the presence and absence of ulcers using discovery proteomics and bioinformatics. Additionally, we contrasted these patterns with those of healthy individuals (n = 31) who had no prior aphthous ulceration. RESULTS Salivary proteome showed that during the ulcerative phase, controlled cell death was downregulated. Due to its ability to distinguish between individuals with and without ulcers, the ATF6B protein raises the possibility that endoplasmic reticulum (ER) stress is responsible for the damage that leads to the death of oral keratinocytes. The high abundance of TRAP1 and ERN1 matches with this biological discovery. The type of death is immunogenic, according to the functional data found in a cell death database. CONCLUSION We identified a cellular process that can lead to the death of oral keratinocytes in the etiopathogenesis process of RAS. Future studies should be conducted to identify what is responsible for the increase in ER stress signaling that would lead to an anti-cell death response.
Collapse
Affiliation(s)
- Camila Cofré-Leiva
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
| | - Paola Andrea Camargo-Ayala
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
| | - Angela Vergara-Pérez
- Laboratorio de Histopatología Oral y Maxilofacial, Departamento de Estomatología, Facultad de Odontología, Universidad de Talca, Avenida Lircay S/N, Campus Norte Universidad de Talca, Talca, Oficina, N°4, Chile
| | - Romina Hernández-Olivos
- Laboratorio de Histopatología Oral y Maxilofacial, Departamento de Estomatología, Facultad de Odontología, Universidad de Talca, Avenida Lircay S/N, Campus Norte Universidad de Talca, Talca, Oficina, N°4, Chile
| | - Sergio Sanhueza
- Laboratorio de Inmunología Translacional, Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Estefanía Nova-Lamperti
- Laboratorio de Inmunología Translacional, Departamento de Bioquímica Clínica e Inmunología, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Jessica Zúñiga-Hernández
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Talca, Chile
| | - César Rivera
- Laboratorio de Histopatología Oral y Maxilofacial, Departamento de Estomatología, Facultad de Odontología, Universidad de Talca, Avenida Lircay S/N, Campus Norte Universidad de Talca, Talca, Oficina, N°4, Chile.
| |
Collapse
|
17
|
Ogura K, Endo M, Hase T, Negami H, Tsuchiya K, Nishiuchi T, Suzuki T, Ogai K, Sanada H, Okamoto S, Sugama J. Potential biomarker proteins for aspiration pneumonia detected by shotgun proteomics using buccal mucosa samples: a cross-sectional case-control study. Clin Proteomics 2023; 20:9. [PMID: 36894881 PMCID: PMC9996945 DOI: 10.1186/s12014-023-09398-w] [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: 06/15/2022] [Accepted: 02/21/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Aspiration pneumonia (AP), which is a major cause of death in the elderly, does present with typical symptoms in the early stages of onset, thus it is difficult to detect and treat at an early stage. In this study, we identified biomarkers that are useful for the detection of AP and focused on salivary proteins, which may be collected non-invasively. Because expectorating saliva is often difficult for elderly people, we collected salivary proteins from the buccal mucosa. METHODS We collected samples from the buccal mucosa of six patients with AP and six control patients (no AP) in an acute-care hospital. Following protein precipitation using trichloroacetic acid and washing with acetone, the samples were analyzed by liquid chromatography and tandem mass spectrometry (LC-MS/MS). We also determined the levels of cytokines and chemokines in non-precipitated samples from buccal mucosa. RESULTS Comparative quantitative analysis of LC-MS/MS spectra revealed 55 highly (P values < 0.10) abundant proteins with high FDR confidence (q values < 0.01) and high coverage (> 50%) in the AP group compared with the control group. Among the 55 proteins, the protein abundances of four proteins (protein S100-A7A, eukaryotic translation initiation factor 1, Serpin B4, and peptidoglycan recognition protein 1) in the AP group showed a negative correlation with the time post-onset; these proteins are promising AP biomarker candidates. In addition, the abundance of C-reactive protein (CRP) in oral samples was highly correlated with serum CRP levels, suggesting that oral CRP levels may be used as a surrogate to predict serum CRP in AP patients. A multiplex cytokine/chemokine assay revealed that MCP-1 tended to be low, indicating unresponsiveness of MCP-1 and its downstream immune pathways in AP. CONCLUSION Our findings suggest that oral salivary proteins, which are obtained non-invasively, can be utilized for the detection of AP.
Collapse
Affiliation(s)
- Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan
| | - Maho Endo
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan.,Nursing Department, Fujita Health University Hospital, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 4701192, Japan
| | - Takashi Hase
- Department of Oral and Maxillofacial Surgery, Noto General Hospital, 6-4 Fujibashi, Nanao, Ishikawa, 9260816, Japan
| | - Hitomi Negami
- Department of Oral and Maxillofacial Surgery, Noto General Hospital, 6-4 Fujibashi, Nanao, Ishikawa, 9260816, Japan
| | - Kohsuke Tsuchiya
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University. Kakuma-Cho, Kanazawa, Ishikawa, 9201164, Japan
| | - Takumi Nishiuchi
- Division of Functional Genomics, Advanced Science Research Center, Kanazawa University, Kanazawa, Ishikawa, 9200934, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University. Kakuma-Cho, Kanazawa, Ishikawa, 9201164, Japan
| | - Kazuhiro Ogai
- Institute of Medical, Pharmaceutical and Health Sciences, AI Hospital/Macro Signal Dynamics Research and Development Center, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan
| | - Hiromi Sanada
- Ishikawa Prefectural Nursing University, 1-1 Gakuendai, Kahoku, Ishikawa, 929-1210, Japan
| | - Shigefumi Okamoto
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan. .,Department of Clinical Laboratory Sciences, Faculty of Health Sciences, Institute of Medical, Pharmaceutical, and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa, Ishikawa, 9200942, Japan.
| | - Junko Sugama
- Research Center for Implementation Nursing Science Initiative, Innovation Promotion Division, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 4701192, Japan
| |
Collapse
|
18
|
Miranda LFB, Lima CV, Pagin R, Costa RC, Pereira MMA, de Avila ED, Bertolini M, Retamal-Valdes B, Shibli JA, Feres M, Barão VAR, Souza JGS. Effect of Processing Methods of Human Saliva on the Proteomic Profile and Protein-Mediated Biological Processes. J Proteome Res 2023; 22:857-870. [PMID: 36779809 DOI: 10.1021/acs.jproteome.2c00652] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
The use of saliva as a protein source prior to microbiological and biological assays requires previous processing. However, the effect of these processing methods on the proteomic profile of saliva has not been tested. Stimulated human saliva was collected from eight healthy volunteers. Non-processed saliva was compared with 0.22 μm filtered, 0.45 μm filtered, and pasteurized saliva, by liquid chromatography-mass spectrometry. Data are available via ProteomeXchange with identifier PXD039248. The effect of processed saliva on microbial adhesion was tested using bacterial and fungus species and in biological cell behavior using HaCaT immortalized human keratinocytes. Two hundred and seventy-eight proteins were identified in non-processed saliva, of which 54 proteins (≈19%) were exclusive. Saliva processing reduced identified proteins to 222 (≈80%) for the 0.22 μm group, 219 (≈79%) for the 0.45 μm group, and 201 (≈72%) for the pasteurized saliva, compared to non-processed saliva. The proteomic profile showed similar molecular functions and biological processes. The different saliva processing methods did not alter microbial adhesion (ANOVA, p > 0.05). Interestingly, pasteurized saliva reduced keratinocyte cell viability. Saliva processing methods tested reduced the proteomic profile diversity of saliva but maintained similar molecular functions and biological processes, not interfering with microbial adhesion and cell viability, except for pasteurization, which reduced cell viability.
Collapse
Affiliation(s)
- Luis Fernando B Miranda
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sa̅o Paulo 13414-903, Brazil
| | - Carolina V Lima
- Department of Restorative Dentistry, Federal University of Paraná (UFPR), Curitiba, Paraná 80210-170, Brazil
| | - Rafaela Pagin
- Department of Periodontology, Dental Research Division, Guarulhos University (UnG), Guarulhos, Sa̅o Paulo 07023-070, Brazil
| | - Raphael C Costa
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sa̅o Paulo 13414-903, Brazil
| | - Marta Maria A Pereira
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo 14801-385, Brazil
| | - Erica D de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araraquara, São Paulo State University (UNESP), Araraquara, São Paulo 14801-385, Brazil.,Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, São Paulo 16066-840, Brazil
| | - Martinna Bertolini
- Department of Periodontics and Preventive Dentistry, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Belén Retamal-Valdes
- Department of Periodontology, Dental Research Division, Guarulhos University (UnG), Guarulhos, Sa̅o Paulo 07023-070, Brazil
| | - Jamil A Shibli
- Department of Periodontology, Dental Research Division, Guarulhos University (UnG), Guarulhos, Sa̅o Paulo 07023-070, Brazil
| | - Magda Feres
- Department of Periodontology, Dental Research Division, Guarulhos University (UnG), Guarulhos, Sa̅o Paulo 07023-070, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, Sa̅o Paulo 13414-903, Brazil
| | - João Gabriel S Souza
- Department of Periodontology, Dental Research Division, Guarulhos University (UnG), Guarulhos, Sa̅o Paulo 07023-070, Brazil.,Dental Science School (Faculdade de Ciências Odontológicas - FCO), Montes Claros, Minas Gerais 39401-303, Brazil
| |
Collapse
|
19
|
Morio KA, Sternowski RH, Brogden KA. Induction of Endogenous Antimicrobial Peptides to Prevent or Treat Oral Infection and Inflammation. Antibiotics (Basel) 2023; 12:antibiotics12020361. [PMID: 36830272 PMCID: PMC9952314 DOI: 10.3390/antibiotics12020361] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Antibiotics are often used to treat oral infections. Unfortunately, excessive antibiotic use can adversely alter oral microbiomes and promote the development of antibiotic-resistant microorganisms, which can be difficult to treat. An alternate approach could be to induce the local transcription and expression of endogenous oral antimicrobial peptides (AMPs). To assess the feasibility and benefits of this approach, we conducted literature searches to identify (i) the AMPs expressed in the oral cavity; (ii) the methods used to induce endogenous AMP expression; and (iii) the roles that expressed AMPs may have in regulating oral inflammation, immunity, healing, and pain. Search results identified human neutrophil peptides (HNP), human beta defensins (HBD), and cathelicidin AMP (CAMP) gene product LL-37 as prominent AMPs expressed by oral cells and tissues. HNP, HBD, and LL-37 expression can be induced by micronutrients (trace elements, elements, and vitamins), nutrients, macronutrients (mono-, di-, and polysaccharides, amino acids, pyropeptides, proteins, and fatty acids), proinflammatory agonists, thyroid hormones, and exposure to ultraviolet (UV) irradiation, red light, or near infrared radiation (NIR). Localized AMP expression can help reduce infection, inflammation, and pain and help oral tissues heal. The use of a specific inducer depends upon the overall objective. Inducing the expression of AMPs through beneficial foods would be suitable for long-term health protection. Additionally, the specialized metabolites or concentrated extracts that are utilized as dosage forms would maintain the oral and intestinal microbiome composition and control oral and intestinal infections. Inducing AMP expression using irradiation methodologies would be applicable to a specific oral treatment area in addition to controlling local infections while regulating inflammatory and healing processes.
Collapse
Affiliation(s)
| | | | - Kim A. Brogden
- College of Dentistry, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
20
|
Arora A, Kaur D, Patiyal S, Kaur D, Tomer R, Raghava GPS. SalivaDB-a comprehensive database for salivary biomarkers in humans. Database (Oxford) 2023; 2023:7030099. [PMID: 36747479 PMCID: PMC9902669 DOI: 10.1093/database/baad002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/01/2022] [Accepted: 01/25/2023] [Indexed: 02/08/2023]
Abstract
Saliva as a non-invasive diagnostic fluid has immense potential as a tool for early diagnosis and prognosis of patients. The information about salivary biomarkers is broadly scattered across various resources and research papers. It is important to bring together all the information on salivary biomarkers to a single platform. This will accelerate research and development in non-invasive diagnosis and prognosis of complex diseases. We collected widespread information on five types of salivary biomarkers-proteins, metabolites, microbes, micro-ribonucleic acid (miRNA) and genes found in humans. This information was collected from different resources that include PubMed, the Human Metabolome Database and SalivaTecDB. Our database SalivaDB contains a total of 15 821 entries for 201 different diseases and 48 disease categories. These entries can be classified into five categories based on the type of biomolecules; 6067, 3987, 2909, 2272 and 586 entries belong to proteins, metabolites, microbes, miRNAs and genes, respectively. The information maintained in this database includes analysis methods, associated diseases, biomarker type, regulation status, exosomal origin, fold change and sequence. The entries are linked to relevant biological databases to provide users with comprehensive information. We developed a web-based interface that provides a wide range of options like browse, keyword search and advanced search. In addition, a similarity search module has been integrated which allows users to perform a similarity search using Basic Local Alignment Search Tool and Smith-Waterman algorithm against biomarker sequences in SalivaDB. We created a web-based database-SalivaDB, which provides information about salivary biomarkers found in humans. A wide range of web-based facilities have been integrated to provide services to the scientific community. https://webs.iiitd.edu.in/raghava/salivadb/.
Collapse
Affiliation(s)
- Akanksha Arora
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India
| | - Dashleen Kaur
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India
| | - Sumeet Patiyal
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India
| | - Dilraj Kaur
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India
| | - Ritu Tomer
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India
| | | |
Collapse
|
21
|
Han P, Li X, Wei W, Ivanovski S. Saliva Diagnosis Using Small Extracellular Vesicles and Salivaomics. Methods Mol Biol 2023; 2588:25-39. [PMID: 36418680 DOI: 10.1007/978-1-0716-2780-8_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Saliva is a complex oral biofluid composed of various biological molecules secreted by major and minor salivary glands, as well as by-products of host oral cells, oral bacteria, small extracellular vesicles (sEVs), and gingival crevicular fluid. Recently, salivary small extracellular vesicles and salivary multi-omics (microbiome, transcriptome, DNA methylome and proteome) are emerging as potential diagnostic tools for oral diseases, including the highly prevalent periodontitis. Here, we describe the methodologies for how to isolate salivary sEVs using the size exclusion chromatography method, and how to perform salivaomics, which may guide future dental research.
Collapse
Affiliation(s)
- Pingping Han
- School of Dentistry, The University of Queensland, Brisbane, Australia.,School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Brisbane, Australia
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China.,Medical Research Institute, Wuhan University, Wuhan, China.,Department of Neurosurgery and Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Wei Wei
- Department of Neurosurgery, Zhongnan Hospital, Wuhan University, Wuhan, China.,Department of Neurosurgery and Brain Research Center, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Australia. .,School of Dentistry, Centre for Orofacial Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Brisbane, Australia.
| |
Collapse
|
22
|
Maruyama Y, Seki T, Ando S, Tanabe H, Mori H. Analysis of IGHA1 and other salivary proteins post half marathon in female participants. PeerJ 2023; 11:e15075. [PMID: 37193030 PMCID: PMC10183162 DOI: 10.7717/peerj.15075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/24/2023] [Indexed: 05/18/2023] Open
Abstract
Background High-intensity exercise (HIE), such as that in marathons and triathlons, suppresses transient local and systemic immunity. Serum and salivary immunoglobulin heavy constant alpha 1 (IGHA1) are major markers of immunosuppression by HIE. Although much is known about the systemic immunosuppressive response, little is known about its local response in the oral cavity, lungs, bronchial tubes, and skin. The oral cavity allows bacteria or viruses to enter the body. Saliva covers the epidermis of the oral cavity and plays an important role in the local stress response by preventing infection. In this study, we examined the properties of saliva secreted during the local stress response for half-marathon (HM) induced IGHA1 protein expression using quantitative proteomics. Methods The Exercise Group (ExG) (19 healthy female university students) participated in a HM race. The Non-Exercise Group (NExG) (16 healthy female university students) did not participate in the ExG. The ExG saliva samples were collected 1 h pre and 2 h and 4 h post-HM. The NExG saliva samples were collected at the same time intervals. The saliva volume, protein concentration, and relative IGHA1 expression were analyzed. In addition, 1 h pre and 2 h post- HM saliva samples were analyzed by iTRAQ. The identified factors in iTRAQ were analyzed for the ExG and the NExG using western blotting. Results We identified kallikrein 1 (KLK1), immunoglobulin kappa chain (IgK), and cystatin S (CST4) as suppression factors, as well as IGHA1, which has been reported to be an immunological stress marker. IGHA1 (p = 0.003), KLK1 (p = 0.011), IGK (p = 0.002), and CST4 (p = 0.003) were suppressed 2 h post-HM compared with their levels pre HM, and IGHA1 (p < 0.001), KLK1 (p = 0.004), and CST4 (p = 0.006) were suppressed 4 h post-HM. There was also a positive correlation between IGHA1, IGK, and CST4 levels at 2 and 4 h post-HM. In addition, KLK1 and IGK levels at 2 h post-HM were positively correlated. Conclusion Our study demonstrated that the salivary proteome is regulated, and antimicrobial proteins are suppressed post-HM. These results suggest that oral immunity was transiently suppressed post-HM. The positive correlation of each protein at 2 and 4 h post-HM suggests that the suppressed state was similarly regulated up to 4 h after a HM. The proteins identified in this study may have applications as stress markers for recreational runners and individuals who perform moderate to HIE on a regular basis.
Collapse
Affiliation(s)
- Yosuke Maruyama
- Faculty of Health and Welfare Science Department of Nutritional Sciences, Nayoro City University, Nayoro, Hokkaido, Japan
| | - Tomoaki Seki
- National Institute of Fitness and Sports Kanoya, Kanoya, Kagoshima, Japan
| | - Seiichi Ando
- Clinical Nutrition and Internal Medicine, Kobe Women’s University, Kobe, Hyogo, Japan
| | - Hiroki Tanabe
- Faculty of Health and Welfare Science Department of Nutritional Sciences, Nayoro City University, Nayoro, Hokkaido, Japan
| | - Hitoshi Mori
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Aichi, Japan
| |
Collapse
|
23
|
Vach K, Al-Ahmad A, Anderson A, Woelber JP, Karygianni L, Wittmer A, Hellwig E. Examining the Composition of the Oral Microbiota as a Tool to Identify Responders to Dietary Changes. Nutrients 2022; 14:5389. [PMID: 36558547 PMCID: PMC9780922 DOI: 10.3390/nu14245389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The role of diet and nutrition in the prevention of oral diseases has recently gained increasing attention. Understanding the influence of diet on oral microbiota is essential for developing meaningful prevention approaches to oral diseases, and the identification of typical and atypical responders may contribute to this. METHODS We used data from an experimental clinical study in which 11 participants were exposed to different dietary regimens in five consecutive phases. To analyse the influence of additional nutritional components, we examined changes in bacterial concentrations measured by culture techniques compared to a run-in phase. A measure of correspondence between the mean and individual patterns of the bacterial composition is introduced. RESULTS The distance measures introduced showed clear differences between the subjects. In our data, two typical and three atypical responders appear to have been identified. CONCLUSIONS The proposed method is suitable to identify typical and atypical responders, even in small datasets. We recommend routinely performing such analyses.
Collapse
Affiliation(s)
- Kirstin Vach
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, Stefan-Meier-Str. 26, D-79104 Freiburg, Germany
| | - Ali Al-Ahmad
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany
| | - Annette Anderson
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany
| | - Johan Peter Woelber
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany
| | - Lamprini Karygianni
- Clinic for Conservative and Preventive Dentistry, Center of Dental Medicine, University of Zurich, Plattenstrasse 11, CH-8032 Zurich, Switzerland
| | - Annette Wittmer
- Department of Medical Microbiology and Hygiene, Institute of Medical Microbiology and Hygiene, Faculty of Medicine and Medical Center, University of Freiburg, Hermann-Herder-Straße 11, D-79104 Freiburg, Germany
| | - Elmar Hellwig
- Department of Operative Dentistry and Periodontology, Center for Dental Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Hugstetter Straße 55, D-79106 Freiburg, Germany
| |
Collapse
|
24
|
António M, Lima T, Vitorino R, Daniel-da-Silva AL. Interaction of Colloidal Gold Nanoparticles with Urine and Saliva Biofluids: An Exploratory Study. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:4434. [PMID: 36558287 PMCID: PMC9785464 DOI: 10.3390/nano12244434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
The use of gold nanoparticles for drug delivery, photothermal or photodynamic therapy, and biosensing enhances the demand for knowledge about the protein corona formed on the surface of nanoparticles. In this study, gold nanospheres (AuNSs), gold nanorods (AuNRs), and gold nanoflowers (AuNFs) were incubated with saliva or urine. After the interaction, the surface of gold nanoparticles was investigated using UV-VIS spectroscopy, zeta potential, and dynamic light scattering. The shifting of the localized surface plasmon resonance (LSPR) band, the increase in hydrodynamic diameter, and the changes in the surface charge of nanoparticles indicated the presence of biomolecules on the surface of AuNSs, AuNRs, and AuNFs. The incubation of AuNFs with saliva led to nanoparticle aggregation and minimal protein adsorption. AuNSs and AuNRs incubated in saliva were analyzed through liquid chromatography with tandem mass spectrometry (LC-MS/MS) to identify the 96 proteins adsorbed on the surface of the gold nanoparticles. Among the 20 most abundant proteins identified, 14 proteins were common in both AuNSs and AuNRs. We hypothesize that the adsorption of these proteins was due to their high sulfur content, allowing for their interaction with gold nanoparticles via the Au-S bond. The presence of distinct proteins on the surface of AuNSs or AuNRs was also investigated and possibly related to the competition between proteins present on the external layers of corona and gold nanoparticle morphology.
Collapse
Affiliation(s)
- Maria António
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Tânia Lima
- iBiMED-Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute of Porto (GEBC CI-IPOP) & Porto Comprehensive Cancer Center (P.CCC), 4200-072 Porto, Portugal
| | - Rui Vitorino
- iBiMED-Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- UnIC@RISE, Department of Surgery and Phycology, Cardiovascular R&D Center, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ana L. Daniel-da-Silva
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
25
|
Intraglandular mesenchymal stem cell treatment induces changes in the salivary proteome of irradiated patients. COMMUNICATIONS MEDICINE 2022; 2:160. [PMID: 36496530 PMCID: PMC9735277 DOI: 10.1038/s43856-022-00223-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hyposalivation and xerostomia (dry mouth), are the leading site-effects to treatment of head and neck cancer. Currently, there are no effective therapies to alleviate radiation-induced hyposalivation. Adipose tissue-derived mesenchymal stem/stromal cells (AT-MSCs) have shown potential for restoring salivary gland function. However, the mode of action is unknown. The purpose of the present study was therefore to characterize the effect of AT-MSC therapy on the salivary proteome in previously irradiated head and neck cancer patients. METHODS Whole saliva was collected from patients with radiation-induced salivary gland hypofunction (n = 8) at baseline, and 120 days after AT-MSC treatment, and from healthy controls (n = 10). The salivary proteome was characterized with mass spectrometry based proteomics, and data was compared within the AT-MSC group (baseline versus day 120) and between AT-MSC group and healthy controls. Significance levels between groups were determined by using double-sided t-test, and visualized by means of principal component analysis, volcano plots and cluster analysis. RESULTS Here we show that 140 human proteins are significantly differentially expressed in saliva from patients with radiation-induced hypofunction versus healthy controls. AT-MSC treatment induce a significant impact on the salivary proteome, as 99 proteins are differentially expressed at baseline vs. 120 days after treatment. However, AT-MSC treatment does not restore healthy conditions, as 212 proteins are significantly differentially expressed in saliva 120 days after AT-MSCs treatment, as compared to healthy controls. CONCLUSION The results indicate an increase in proteins related to tissue regeneration in AT-MSCs treated patients. Our study demonstrates the impact of AT-MSCs on the salivary proteome, thereby providing insight into the potential mode of action of this novel treatment approach.
Collapse
|
26
|
Salivary Biomarkers in Oral Squamous Cell Carcinoma: A Proteomic Overview. Proteomes 2022; 10:proteomes10040037. [PMID: 36412636 PMCID: PMC9680331 DOI: 10.3390/proteomes10040037] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is one of the most frequent cancers worldwide. Endoscopic methods may be useful in the evaluation of oral injuries even though the diagnostic gold standard is a biopsy. Targeted screenings could be considered the best way to prevent the occurrence of oral cancer. Aimed to elucidate the potential identification of specific biomarkers of OSCC, the use of saliva is convenient and noninvasive. Many studies reported more than a hundred putative saliva biomarkers for OSCC, and proteogenomic approaches were fundamental to disclosing this issue. METHODS Relevant literature published in the last few years was systematically searched on PubMed and we focused on articles about the use and study of salivary biomarkers in the diagnostics of head and neck cancer (n = 110). Thereafter, we performed a selection focusing on diagnosis with salivary proteomics in OSCC (n = 8). RESULTS Saliva proteomics can be a source of biomarkers for OSCC. We reviewed literature of biomarker proteins in saliva that could also be evaluated as probable targets for non-invasive screening of oral neoplasm such as cytokines, matrix metalloproteinases, and acute-phase response proteins. CONCLUSIONS The measurement of salivary biomarkers is a highly hopeful technique for the diagnosis of OSCC. Proteogenomic approaches could permit an accurate and early diagnosis of OSCC. This review seeks to generate an up-to-date view on translational OSCC issues by raising awareness of researchers, physicians, and surgeons. Renewed clinical studies, which will validate the sensitivity and specificity of salivary biomarkers, are necessary to translate these results into possible strategies for early diagnosis of OSCC, thus improving patient outcomes.
Collapse
|
27
|
António M, Lima T, Vitorino R, Daniel-da-Silva AL. Label-free dynamic light scattering assay for C-reactive protein detection using magnetic nanoparticles. Anal Chim Acta 2022; 1222:340169. [DOI: 10.1016/j.aca.2022.340169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022]
|
28
|
Chibly AM, Aure MH, Patel VN, Hoffman MP. Salivary gland function, development, and regeneration. Physiol Rev 2022; 102:1495-1552. [PMID: 35343828 PMCID: PMC9126227 DOI: 10.1152/physrev.00015.2021] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/27/2021] [Accepted: 03/17/2022] [Indexed: 02/08/2023] Open
Abstract
Salivary glands produce and secrete saliva, which is essential for maintaining oral health and overall health. Understanding both the unique structure and physiological function of salivary glands, as well as how they are affected by disease and injury, will direct the development of therapy to repair and regenerate them. Significant recent advances, particularly in the OMICS field, increase our understanding of how salivary glands develop at the cellular, molecular, and genetic levels: the signaling pathways involved, the dynamics of progenitor cell lineages in development, homeostasis, and regeneration, and the role of the extracellular matrix microenvironment. These provide a template for cell and gene therapies as well as bioengineering approaches to repair or regenerate salivary function.
Collapse
Affiliation(s)
- Alejandro M Chibly
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Marit H Aure
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Vaishali N Patel
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| | - Matthew P Hoffman
- Matrix and Morphogenesis Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
29
|
Dayon L, Cominetti O, Affolter M. Proteomics of Human Biological Fluids for Biomarker Discoveries: Technical Advances and Recent Applications. Expert Rev Proteomics 2022; 19:131-151. [PMID: 35466824 DOI: 10.1080/14789450.2022.2070477] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Biological fluids are routine samples for diagnostic testing and monitoring. Blood samples are typically measured because of their moderate collection invasiveness and high information content on health and disease. Several body fluids, such as cerebrospinal fluid (CSF), are also studied and suited to specific pathologies. Over the last two decades proteomics has quested to identify protein biomarkers but with limited success. Recent technologies and refined pipelines have accelerated the profiling of human biological fluids. AREAS COVERED We review proteomic technologies for the identification of biomarkers. Those are based on antibodies/aptamers arrays or mass spectrometry (MS), but new ones are emerging. Advances in scalability and throughput have allowed to better design studies and cope with the limited sample size that had until now prevailed due to technological constraints. With these enablers, plasma/serum, CSF, saliva, tears, urine, and milk proteomes have been further profiled; we provide a non-exhaustive picture of some recent highlights (mainly covering literature from last five years in the Scopus database) using MS-based proteomics. EXPERT OPINION While proteomics has been in the shadow of genomics for years, proteomic tools and methodologies have reached a certain maturity. They are better suited to discover innovative and robust biofluid biomarkers.
Collapse
Affiliation(s)
- Loïc Dayon
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland.,Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ornella Cominetti
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland
| | - Michael Affolter
- Proteomics, Nestlé Institute of Food Safety & Analytical Sciences, Nestlé Research, CH-1015 Lausanne, Switzerland
| |
Collapse
|