1
|
Saravanan C, S M N Mydin RB, Mohamed Sheriff NR, Kaur G, Singh Dhaliwal S, Musa MY. Salivaomics in head and neck cancer. Clin Chim Acta 2025; 565:119952. [PMID: 39216814 DOI: 10.1016/j.cca.2024.119952] [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: 06/27/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Salivaomics is a promising method for the early detection and monitoring of head and neck cancer (HNC). By analyzing salivary proteomics, RNA, and DNA, it identifies biomarkers that distinguish HNC patients from healthy individuals. Saliva's non-invasive, easily collectible nature and affordability make it an advantageous screening tool. Multiomics approaches, which explore genetic mutations, gene expression patterns, protein profiles, and metabolite levels, provide a comprehensive molecular perspective that enhances clinical applicability. The approaches enhance the precision of diagnoses, enable the development and application of targeted therapies, and contribute to the overall advancement of personalized medicine. Despite its potential, larger-scale studies are essential for validating biomarkers, and assessing sensitivity, accuracy, and specificity in detecting HNC. This review highlights salivaomics' potential as a non-invasive, accessible biological sample for early disease detection in HNC and underscores the value of multiomics in advancing this research. Salivaomics offers significant insights into the underlying mechanisms of HNC, enabling the discovery of robust, non-invasive biomarkers for improved disease management.
Collapse
Affiliation(s)
- Chandrarohini Saravanan
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM), 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Rabiatul Basria S M N Mydin
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia (USM), 13200 Kepala Batas, Pulau Pinang, Malaysia.
| | - Nur Rizikin Mohamed Sheriff
- School of Distance Education, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia; Division of Research & Innovation, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia
| | - Satvinder Singh Dhaliwal
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, 11800 Minden, Pulau Pinang, Malaysia; Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Bentley, Australia; Duke-NUS Medical School, National University of Singapore, Queenstown, Singapore; Singapore University of Social Sciences, 463 Clementi Road, Clementi 599494, Singapore
| | - Muhamad Yusri Musa
- Department of Clinical Medicine, Universiti Sains Malaysia, 13200 Kepala Batas, Pulau Pinang, Malaysia; Pusat Perubatan, Universiti Sains Malaysia, 13200 Bertam, Kepala Batas, Pulau Pinang 84001, Malaysia
| |
Collapse
|
2
|
Davenport C, Arevalo-Rodriguez I, Mateos-Haro M, Berhane S, Dinnes J, Spijker R, Buitrago-Garcia D, Ciapponi A, Takwoingi Y, Deeks JJ, Emperador D, Leeflang MMG, Van den Bruel A. The effect of sample site and collection procedure on identification of SARS-CoV-2 infection. Cochrane Database Syst Rev 2024; 12:CD014780. [PMID: 39679851 DOI: 10.1002/14651858.cd014780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
BACKGROUND Sample collection is a key driver of accuracy in the diagnosis of SARS-CoV-2 infection. Viral load may vary at different anatomical sampling sites and accuracy may be compromised by difficulties obtaining specimens and the expertise of the person taking the sample. It is important to optimise sampling accuracy within cost, safety and accessibility constraints. OBJECTIVES To compare the sensitivity of different sampling collection sites and methods for the detection of current SARS-CoV-2 infection with any molecular or antigen-based test. SEARCH METHODS Electronic searches of the Cochrane COVID-19 Study Register and the COVID-19 Living Evidence Database from the University of Bern (which includes daily updates from PubMed and Embase and preprints from medRxiv and bioRxiv) were undertaken on 22 February 2022. We included independent evaluations from national reference laboratories, FIND and the Diagnostics Global Health website. We did not apply language restrictions. SELECTION CRITERIA We included studies of symptomatic or asymptomatic people with suspected SARS-CoV-2 infection undergoing testing. We included studies of any design that compared results from different sample types (anatomical location, operator, collection device) collected from the same participant within a 24-hour period. DATA COLLECTION AND ANALYSIS Within a sample pair, we defined a reference sample and an index sample collected from the same participant within the same clinical encounter (within 24 hours). Where the sample comparison was different anatomical sites, the reference standard was defined as a nasopharyngeal or combined naso/oropharyngeal sample collected into the same sample container and the index sample as the alternative anatomical site. Where the sample comparison was concerned with differences in the sample collection method from the same site, we defined the reference sample as that closest to standard practice for that sample type. Where the sample pair comparison was concerned with differences in personnel collecting the sample, the more skilled or experienced operator was considered the reference sample. Two review authors independently assessed the risk of bias and applicability concerns using the QUADAS-2 and QUADAS-C checklists, tailored to this review. We present estimates of the difference in the sensitivity (reference sample (%) minus index sample sensitivity (%)) in a pair and as an average across studies for each index sampling method using forest plots and tables. We examined heterogeneity between studies according to population (age, symptom status) and index sample (time post-symptom onset, operator expertise, use of transport medium) characteristics. MAIN RESULTS This review includes 106 studies reporting 154 evaluations and 60,523 sample pair comparisons, of which 11,045 had SARS-CoV-2 infection. Ninety evaluations were of saliva samples, 37 nasal, seven oropharyngeal, six gargle, six oral and four combined nasal/oropharyngeal samples. Four evaluations were of the effect of operator expertise on the accuracy of three different sample types. The majority of included evaluations (146) used molecular tests, of which 140 used RT-PCR (reverse transcription polymerase chain reaction). Eight evaluations were of nasal samples used with Ag-RDTs (rapid antigen tests). The majority of studies were conducted in Europe (35/106, 33%) or the USA (27%) and conducted in dedicated COVID-19 testing clinics or in ambulatory hospital settings (53%). Targeted screening or contact tracing accounted for only 4% of evaluations. Where reported, the majority of evaluations were of adults (91/154, 59%), 28 (18%) were in mixed populations with only seven (4%) in children. The median prevalence of confirmed SARS-CoV-2 was 23% (interquartile (IQR) 13%-40%). Risk of bias and applicability assessment were hampered by poor reporting in 77% and 65% of included studies, respectively. Risk of bias was low across all domains in only 3% of evaluations due to inappropriate inclusion or exclusion criteria, unclear recruitment, lack of blinding, nonrandomised sampling order or differences in testing kit within a sample pair. Sixty-eight percent of evaluation cohorts were judged as being at high or unclear applicability concern either due to inflation of the prevalence of SARS-CoV-2 infection in study populations by selectively including individuals with confirmed PCR-positive samples or because there was insufficient detail to allow replication of sample collection. When used with RT-PCR • There was no evidence of a difference in sensitivity between gargle and nasopharyngeal samples (on average -1 percentage points, 95% CI -5 to +2, based on 6 evaluations, 2138 sample pairs, of which 389 had SARS-CoV-2). • There was no evidence of a difference in sensitivity between saliva collection from the deep throat and nasopharyngeal samples (on average +10 percentage points, 95% CI -1 to +21, based on 2192 sample pairs, of which 730 had SARS-CoV-2). • There was evidence that saliva collection using spitting, drooling or salivating was on average -12 percentage points less sensitive (95% CI -16 to -8, based on 27,253 sample pairs, of which 4636 had SARS-CoV-2) compared to nasopharyngeal samples. We did not find any evidence of a difference in the sensitivity of saliva collected using spitting, drooling or salivating (sensitivity difference: range from -13 percentage points (spit) to -21 percentage points (salivate)). • Nasal samples (anterior and mid-turbinate collection combined) were, on average, 12 percentage points less sensitive compared to nasopharyngeal samples (95% CI -17 to -7), based on 9291 sample pairs, of which 1485 had SARS-CoV-2. We did not find any evidence of a difference in sensitivity between nasal samples collected from the mid-turbinates (3942 sample pairs) or from the anterior nares (8272 sample pairs). • There was evidence that oropharyngeal samples were, on average, 17 percentage points less sensitive than nasopharyngeal samples (95% CI -29 to -5), based on seven evaluations, 2522 sample pairs, of which 511 had SARS-CoV-2. A much smaller volume of evidence was available for combined nasal/oropharyngeal samples and oral samples. Age, symptom status and use of transport media do not appear to affect the sensitivity of saliva samples and nasal samples. When used with Ag-RDTs • There was no evidence of a difference in sensitivity between nasal samples compared to nasopharyngeal samples (sensitivity, on average, 0 percentage points -0.2 to +0.2, based on 3688 sample pairs, of which 535 had SARS-CoV-2). AUTHORS' CONCLUSIONS When used with RT-PCR, there is no evidence for a difference in sensitivity of self-collected gargle or deep-throat saliva samples compared to nasopharyngeal samples collected by healthcare workers when used with RT-PCR. Use of these alternative, self-collected sample types has the potential to reduce cost and discomfort and improve the safety of sampling by reducing risk of transmission from aerosol spread which occurs as a result of coughing and gagging during the nasopharyngeal or oropharyngeal sample collection procedure. This may, in turn, improve access to and uptake of testing. Other types of saliva, nasal, oral and oropharyngeal samples are, on average, less sensitive compared to healthcare worker-collected nasopharyngeal samples, and it is unlikely that sensitivities of this magnitude would be acceptable for confirmation of SARS-CoV-2 infection with RT-PCR. When used with Ag-RDTs, there is no evidence of a difference in sensitivity between nasal samples and healthcare worker-collected nasopharyngeal samples for detecting SARS-CoV-2. The implications of this for self-testing are unclear as evaluations did not report whether nasal samples were self-collected or collected by healthcare workers. Further research is needed in asymptomatic individuals, children and in Ag-RDTs, and to investigate the effect of operator expertise on accuracy. Quality assessment of the evidence base underpinning these conclusions was restricted by poor reporting. There is a need for further high-quality studies, adhering to reporting standards for test accuracy studies.
Collapse
Affiliation(s)
- Clare Davenport
- Department of Applied Health Science, School of Health Sciences, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Ingrid Arevalo-Rodriguez
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS). CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Miriam Mateos-Haro
- Clinical Biostatistics Unit, Hospital Universitario Ramón y Cajal (IRYCIS). CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Clinical Medicine and Public Health Programme, Universidad de Granada, Granada, Spain
| | - Sarah Berhane
- Department of Applied Health Science, School of Health Sciences, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jacqueline Dinnes
- Department of Applied Health Science, School of Health Sciences, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - René Spijker
- Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam Public Health, Amsterdam, Netherlands
- Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Diana Buitrago-Garcia
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
- Hospital Universitario Mayor - Méderi. Universidad del Rosario, Bogotá, Colombia
| | - Agustín Ciapponi
- Argentine Cochrane Centre, Institute for Clinical Effectiveness and Health Policy (IECS-CONICET), Buenos Aires, Argentina
| | - Yemisi Takwoingi
- Department of Applied Health Science, School of Health Sciences, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | - Jonathan J Deeks
- Department of Applied Health Science, School of Health Sciences, University of Birmingham, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre, University Hospitals Birmingham NHS Foundation Trust and University of Birmingham, Birmingham, UK
| | | | - Mariska M G Leeflang
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
- Amsterdam, Netherlands
| | - Ann Van den Bruel
- Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Popa PȘ, Popa-Cazacu EC, Zaharescu A, Popa GV, Matei MN. Minimizing Oxidative Stress in Oral Surgery: A Comparative Study of Laser-Assisted and Conventional Third Molar Extractions. Dent J (Basel) 2024; 12:402. [PMID: 39727459 DOI: 10.3390/dj12120402] [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: 11/05/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: This study aims to compare the effects of conventional surgical techniques and laser-assisted methods on salivary oxidative stress biomarkers following third molar extraction, in order to evaluate the potential benefits of laser surgery in reducing oxidative stress and promoting faster recovery. Methods: A total of 154 patients, aged 16-30, undergoing third molar extractions were included in the study. Patients were divided into two groups: conventional surgery (n = 75) and laser-assisted surgery (n = 79). Saliva samples were collected at baseline, and 24, 48, 72, and 168 h postoperatively. The levels of total antioxidant capacity (TAC), malondialdehyde (MDA), and 8-hydroxy-2'-deoxyguanosine (8-OHdG) were measured as indicators of oxidative stress. Results: Initial biomarker levels were similar across all participants. Postoperative oxidative stress increased in both groups, with significantly higher levels in the conventional surgery group at 48 and 72 h. Salivary biomarkers of oxidative stress were significantly lower in the laser group at 48 and 72 h post-surgery (p < 0.05), indicating a faster recovery. By 168 h, biomarker levels in the laser group had nearly returned to baseline, whereas levels in the conventional group remained slightly elevated. Conclusions: Laser-assisted surgery significantly reduces oxidative stress and promotes faster recovery when compared with conventional methods, as evidenced by the more rapid normalization of salivary biomarkers. These findings suggest that laser techniques may offer superior clinical outcomes in third molar extractions.
Collapse
Affiliation(s)
- Paul Șerban Popa
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 47 Domnească Str., 800181 Galați, Romania
| | | | - Anamaria Zaharescu
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 47 Domnească Str., 800181 Galați, Romania
| | - Gabriel Valeriu Popa
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 47 Domnească Str., 800181 Galați, Romania
| | - Mădălina Nicoleta Matei
- Faculty of Medicine and Pharmacy, "Dunărea de Jos" University of Galați, 47 Domnească Str., 800181 Galați, Romania
| |
Collapse
|
4
|
Prasad M, Sekar R, Priya MDL, Varma SR, Karobari MI. A new perspective on diagnostic strategies concerning the potential of saliva-based miRNA signatures in oral cancer. Diagn Pathol 2024; 19:147. [PMID: 39548527 PMCID: PMC11568613 DOI: 10.1186/s13000-024-01575-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
Oral cancer, the most prevalent cancer worldwide, is far more likely to occur after the age of forty-five, according to the World Health Organization. Although many biomarkers have been discovered over the years using non-invasive saliva samples, biopsies, and human blood, these biomarkers have not been incorporated into standard clinical practice. Investigating the function of microRNAs (miRNAs) in the diagnosis, aetiology, prognosis, and treatment of oral cancer has drawn more attention in recent years. Though salivary microRNA can act as a window into the molecular environment of the tumour, there are challenges due to the heterogeneity of oral squamous cell carcinoma (OSCC), diversity in sample collection, processing techniques, and storage conditions. The up and downregulation of miRNAs has been found to have a profound role in OSCC as it regulates tumour stages by targeting many genes. As a result, the regulatory functions of miRNAs in OSCC underscore their significance in the field of cancer biology. Salivary miRNAs are useful diagnostic and prognostic indicators because their abnormal expression profiles shed light on tumour behaviour and patient prognosis. In addition to their diagnostic and prognostic value, miRNAs hold promise as therapeutic targets for oral cancer intervention. The current review sheds light on the challenges and potentials of microRNA studies that could lead to a better understanding of oral cancer prognosis, diagnosis, and therapeutic intervention. Furthermore, the clinical translation of OSCC biomarkers requires cooperation between investigators, physicians, regulatory bodies, and business partners. There is much potential for improving early identification, tracking therapy response, and forecasting outcomes in OSCC patients by including saliva-based miRNAs as biomarkers.
Collapse
Affiliation(s)
- Monisha Prasad
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, Tamil Nadu, 602105, India
| | - Ramya Sekar
- Department of Oral and Maxillofacial Pathology & Oral Microbiology, Meenakshi Ammal Dental College and Hospital, MAHER, Alapakkam Main Road, Maduravoyal, Chennai, Tamil Nadu, 600095, India
| | | | - Sudhir Rama Varma
- Department of Clinical Sciences, College of Dentistry, Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman University, Ajman - 346, Ajman, UAE
| | - Mohmed Isaqali Karobari
- Department of Conservative Dentistry and Endodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 600077, India.
- Department of Restorative Dentistry & Endodontics, Faculty of Dentistry, University of Puthisastra, Phnom Penh, 12211, Cambodia.
| |
Collapse
|
5
|
Suragimath G, Patil S, Suragimath DG, Sr A. Salivaomics: A Revolutionary Non-invasive Approach for Oral Cancer Detection. Cureus 2024; 16:e74381. [PMID: 39723315 PMCID: PMC11669377 DOI: 10.7759/cureus.74381] [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: 10/13/2024] [Accepted: 11/24/2024] [Indexed: 12/28/2024] Open
Abstract
Salivaomics has emerged as a ground-breaking field in the detection and management of oral cancer (OC), offering a non-invasive, efficient, and patient-friendly alternative to traditional diagnostic methods. This innovative approach leverages the comprehensive molecular insights provided by genomics, transcriptomics, proteomics, metabolomics, and microbiomics. The potential of salivaomics lies in its ability to enable early detection, predict malignant transformation, and monitor treatment outcomes and disease recurrence. Advancing salivary diagnostics necessitates the standardization of saliva collection and processing protocols, identification and validation of robust biomarkers, and development of cutting-edge detection technologies. A single biomarker is unlikely to fulfill all diagnostic requirements; thus, research should focus on developing a panel of biomolecules to enhance diagnostic accuracy and management of OC. Salivaomics stands at the forefront of non-invasive diagnostic methods, with the promise to revolutionize early detection and management of OC. Future research directions should emphasize the integration of multi-omics data for superior biomarker discovery, the development of portable and cost-effective point-of-care devices, and the fostering of interdisciplinary collaborations to drive innovation. Overcoming these challenges will facilitate the translation of salivaomics into routine clinical practice, significantly improving early diagnosis, treatment, and prognosis of OC. This review provides a comprehensive overview of salivaomics, detailing the use of saliva as a diagnostic fluid. It covers saliva collection, preparation, transportation, storage methods, and various analytical techniques. Additionally, the review discusses the current challenges and future directions of this transformative technology, emphasizing its potential to enhance clinical outcomes in OC.
Collapse
Affiliation(s)
- Girish Suragimath
- Periodontology, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Satish Patil
- Microbiology, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Disha G Suragimath
- General Medicine, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Ashwinirani Sr
- Oral Medicine and Radiology, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| |
Collapse
|
6
|
Li Y, Ou Y, Fan K, Liu G. Salivary diagnostics: opportunities and challenges. Theranostics 2024; 14:6969-6990. [PMID: 39629130 PMCID: PMC11610148 DOI: 10.7150/thno.100600] [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: 07/07/2024] [Accepted: 09/29/2024] [Indexed: 12/06/2024] Open
Abstract
Saliva contains a diverse array of biomarkers indicative of various diseases. Saliva testing has been a major advancement towards non-invasive point-of-care diagnosis with clinical significance. However, there are challenges associated with salivary diagnosis from sample treatment and standardization. This review highlights the biomarkers in saliva and their role in identifying relevant diseases. It provides an overview and discussion about the current practice of saliva collection and processing, and advancements in saliva detection systems from in vitro methods to wearable oral devices. The review also addresses challenges in saliva diagnostics and proposes solutions, aiming to offer a comprehensive understanding and practical guidance for improving saliva-based detection in clinical diagnosis. Saliva diagnosis provides a rapid, effective, and safe alternative to traditional blood and urine tests for screening large populations and enhancing infectious disease diagnosis and surveillance. It meets the needs of various fields such as disease management, drug screening, and personalized healthcare with advances in saliva detection systems offering high sensitivity, fast response times, portability, and automation. Standardization of saliva collection, treatment, biomarker discovery, and detection between different laboratories needs to be implemented to obtain reliable salivary diagnosis in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Guozhen Liu
- Integrated Devices and Intelligent Diagnosis (ID2) Laboratory, CUHK(SZ)-Boyalife Joint Laboratory for Regenerative Medicine Engineering, Biomedical Engineering Programme, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| |
Collapse
|
7
|
Fadli NA, Abdul Rahman M, Karsani SA, Ramli R. Oral and Gingival Crevicular Fluid Biomarkers for Jawbone Turnover Diseases: A Scoping Review. Diagnostics (Basel) 2024; 14:2184. [PMID: 39410587 PMCID: PMC11475764 DOI: 10.3390/diagnostics14192184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Gingival crevicular fluid (GCF) and oral fluid have emerged as promising diagnostic tools for detecting biomarkers. This review aimed to evaluate the existing literature on using oral fluids as a source of biomarkers for bone turnover diseases affecting the jawbone. A comprehensive search strategy was executed between August 2014 and August 2024 across five major databases (Web of Science, EBSCOhost Dentistry & Oral Sciences Source, Cochrane Library, Scopus, and PubMed) and grey literature sources. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) was applied. The screening was facilitated using Rayyan at rayyan.ai and Endnote X20 software tools, culminating in the evaluation of 14,965 citations from databases and 34 from grey literature. Following rigorous scrutiny, 37 articles were selected for inclusion in this review, encompassing diseases such as periodontitis, medication-related osteonecrosis of the jaw (MRONJ), and osteoporosis. The quality of the included observational studies was assessed using the Revised Risk of Bias Assessment Tool for Non-Randomized Studies (RoBANS 2). Interleukin-1 beta (IL-1β), sclerostin, osteoprotegerin (OPG), and interleukin-34 (IL-34) emerged as significant biomarkers in GCF, and they were mainly from periodontitis and osteoporosis. Osteocalcin (OC), IL-1β, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), OPG, and matrix metalloproteinase-9 (MMP-9) were significant in oral fluid or saliva, and they were from periodontitis, MRONJ, and osteoporosis. These findings underscore the potential use of oral fluids, which are regarded as non-invasive tools for biomarker identification in bone turnover. Many biomarkers overlap, and it is important to identify other specific biomarkers to enable accurate diagnosis of these conditions.
Collapse
Affiliation(s)
- Nurfatima Azzahra Fadli
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Mariati Abdul Rahman
- Department of Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Saiful Anuar Karsani
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Roszalina Ramli
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia
| |
Collapse
|
8
|
Karambelkar MV, Varma S, Suragimath G, Zope SA, Mashalkar VS, Kale AV. Comparative Evaluation of Salivary Cathelicidin and 8-Isoprostane Levels Among Smokeless Tobacco Users and Non-users and Their Correlation With Periodontal Health and Disease: A Cross-Sectional Study. Cureus 2024; 16:e67646. [PMID: 39314598 PMCID: PMC11417437 DOI: 10.7759/cureus.67646] [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: 07/01/2024] [Accepted: 08/23/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction Periodontal diseases arise from host-microbial interactions influenced by tobacco products. Salivary antimicrobial peptides such as salivary cathelicidin and prostaglandins such as 8-isoprostane are part of the inflammatory cascade affecting periodontal disease pathogenesis. Methodology A total of 93 patients, 31 in each group that is healthy, periodontitis, and periodontitis with smokeless tobacco habit patients, were enrolled. The case history was recorded, and clinical examination was performed using periodontal parameter analysis of oral hygiene index simplified (OHIS), Russell's index, periodontal pocket depth (PPD), and clinical attachment level (CAL). The saliva samples were collected and subjected to an enzyme-linked immunosorbent assay (ELISA) to evaluate cathelicidin and 8-isoprostane. The results were analysed and compared statistically. Results The OHIS, Russell's index, pocket probing depth, and CAL were high in patients with periodontitis and tobacco habit (p<0.001). The cathelicidin levels were the highest in patients with periodontitis and the tobacco habit (1.6 g/mL). The level of 8-isoprostane was the highest in patients with periodontitis with tobacco habit (1.8 pg/mL). Smokeless tobacco users showed higher levels of cathelicidin and 8-isoprostane in periodontitis with tobacco than in the healthy group. Conclusion Increased cathelicidin and 8-isoprostane levels in smokeless tobacco users with periodontitis suggest risk biomarkers for tobacco-influenced periodontitis.
Collapse
Affiliation(s)
- Mugdha V Karambelkar
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Siddhartha Varma
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Girish Suragimath
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Sameer A Zope
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Vaishali S Mashalkar
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Apurva V Kale
- Periodontology, School of Dental Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| |
Collapse
|
9
|
Hasan NWM, Baharin B, Mohd N, Rahman MA, Hassan N. Comparative effects of e-cigarette smoking on periodontal status, salivary pH, and cotinine levels. BMC Oral Health 2024; 24:861. [PMID: 39069628 PMCID: PMC11285603 DOI: 10.1186/s12903-024-04650-7] [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: 09/10/2023] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND The nicotine in e-cigarette liquid can negatively impact periodontal tissues by altering the salivary pH and elevating cotinine levels. Thus, the study aimed to determine the periodontal parameters, salivary pH, and cotinine levels among cigarette, e-cigarette, and never-smokers. METHODS A total of 144 participants were recruited (48 cigarette smokers, 48 e-cigarette smokers, and 48 never-smokers). Clinical periodontal parameters, including plaque index (PI), gingival index (GI), periodontal probing pocket depth (PPD), and clinical attachment loss (CAL) were recorded, excluding third molars. The level of unstimulated whole salivary pH was measured using a portable pH meter and the levels of salivary cotinine were measured using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS Data were analysed statistically using analysis of variance. Mean scores of PPD, percentage of pocket depth ≥ 4 mm, and CAL (p < 0.05) were significantly higher among cigarette smokers than those in e-cigarette and never-smokers, while GI (p < 0.05) were significantly higher among e-cigarette smokers. The unstimulated salivary pH was more acidic among cigarette smokers (p < 0.05) and e-cigarette smokers (p < 0.05) than in never-smokers. The cotinine levels were higher among cigarette smokers (p < 0.05) and e-cigarette smokers (p < 0.05) than in never-smokers. CONCLUSIONS Clinical periodontal parameters were poorer in cigarette smokers than in e-cigarette smokers and never-smokers. Meanwhile, cigarette and e-cigarette smokers have more acidic salivary pH and higher cotinine levels than in never-smokers.
Collapse
Affiliation(s)
- Nurul Wahida Mohd Hasan
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
- Unit of Periodontics, School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Badiah Baharin
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia.
| | - Nurulhuda Mohd
- Department of Restorative Dentistry, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Mariati Abdul Rahman
- Department of Craniofacial Diagnostics and Biosciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur, 50300, Malaysia
| | - Nooraryana Hassan
- World Health Organization Framework Convention On Tobacco Control (WHO FCTC) and Tobacco Control Unit, Disease Control Division, Ministry of Health, Putrajaya, Malaysia
| |
Collapse
|
10
|
He B, Cao Y, Zhuang Z, Deng Q, Qiu Y, Pan L, Zheng X, Shi B, Lin L, Chen F. The potential value of oral microbial signatures for prediction of oral squamous cell carcinoma based on machine learning algorithms. Head Neck 2024; 46:1660-1670. [PMID: 38695435 DOI: 10.1002/hed.27795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 03/30/2024] [Accepted: 04/22/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE This study aimed to explore the potential predictive value of oral microbial signatures for oral squamous cell carcinoma (OSCC) risk based on machine learning algorithms. METHODS The oral microbiome signatures were assessed in the unstimulated saliva samples of 80 OSCC patients and 179 healthy individuals using 16S rRNA gene sequencing. Four different machine learning classifiers were used to develop prediction models. RESULTS Compared with control participants, OSCC patients had a higher microbial dysbiosis index (MDI, p < 0.001). Among four machine learning classifiers, random forest (RF) provided the best predictive performance, followed by the support vector machines, artificial neural networks and naive Bayes. After controlling the potential confounders using propensity score matching, the optimal RF model was further developed incorporating a minimal set of 20 bacteria genera, exhibiting better predictive performance than the MDI (AUC: 0.992 vs. 0.775, p < 0.001). CONCLUSIONS The novel MDI and RF model developed in this study based on oral microbiome signatures may serve as noninvasive tools for predicting OSCC risk.
Collapse
Affiliation(s)
- Baochang He
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yujie Cao
- Department of Stomatology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Zhaocheng Zhuang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Qingrong Deng
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yu Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lizhen Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaoyan Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lisong Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
- Clinical Research Unit, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
11
|
Nagarajan M, Dayasekaran V, Jayavel K, Jayaraj M, Chellaswamy S, Ranganathan K, Kaliamoorthy S, Chellapandi S, Baskaran R, Elumalai A. Exploring Genetic Link of Residual Ridge Resorption in Completely Edentulous Individuals: A Prospective Case-Control Clinical Study. Cureus 2024; 16:e64039. [PMID: 39114253 PMCID: PMC11303737 DOI: 10.7759/cureus.64039] [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: 05/29/2024] [Accepted: 07/06/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Residual ridge resorption presents obstacles in prosthodontic treatment, affecting denture stability and the success of dental implants. Genetic elements, specifically the single nucleotide polymorphism (SNP) 1772C>T variant within the hypoxia-inducible factor 1 subunit alpha (HIF-1α) gene, are hypothesized to contribute to residual ridge resorption progression. Nevertheless, its impact remains insufficiently investigated, especially within the context of South Indian populations. We sought to investigate the connection between SNP 1772C>T and residual ridge resorption (RRR) among fully edentulous individuals, considering demographic factors, genotyping methodologies, and statistical evaluations. METHODS In a prospective case-control study, we recruited 100 completely edentulous participants from South India. Participants were categorized based on alveolar ridge height. Saliva samples were non-invasively collected for DNA extraction, and polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis was employed to determine genotype distribution using the HphI restriction enzyme. The statistical evaluations comprised the utilization of chi-square and Fisher's exact tests. RESULTS We observed no significant variations in genotype distributions between the case and control cohorts (CT: p=0.24; CC: p=0.65; TT: p=0.30). The heterozygous genotype CT was prevalent in both groups. CONCLUSIONS Although we did not observe significant associations between SNP 1772C>T and RRR, our findings imply a genetic predisposition to residual ridge resorption that warrants further exploration. Variations in genetic susceptibility across ethnicities and the influence of other genetic variants on residual ridge resorption require additional investigation. This study lays the groundwork for personalized prosthodontic care by highlighting the potential of genetic analysis in routine dental practice to improve treatment strategies.
Collapse
Affiliation(s)
- Mahendirakumar Nagarajan
- Department of Prosthodontics and Crown and Bridge, Government Dental College and Hospital, Cuddalore, IND
| | - Vijitha Dayasekaran
- Department of Prosthodontics and Crown and Bridge, Government Dental College and Hospital, Cuddalore, IND
| | - Kavitha Jayavel
- Department of Periodontics and Implantology, Government Dental College and Hospital, Cuddalore, IND
| | - Merlin Jayaraj
- Department of Oral and Maxillofacial Pathology, Chettinad Dental College and Research Institute, Chennai, IND
| | - Sreeja Chellaswamy
- Department of Oral and Maxillofacial Pathology, Chettinad Dental College and Research Institute, Chennai, IND
| | - Krishnaraj Ranganathan
- Department of Prosthodontics and Crown and Bridge, Government Dental College and Hospital, Cuddalore, IND
| | - Sriram Kaliamoorthy
- Department of Dentistry, Vinayaka Mission's Medical College and Hospital, Vinayaka Mission's Research Foundation (DU), Karaikal, IND
| | - Sugirtha Chellapandi
- Department of Periodontics, Chettinad Dental College and Research Institute, Chennai, IND
| | - Radhika Baskaran
- Department of Periodontics, Chettinad Dental College and Research Institute, Chennai, IND
| | - Agila Elumalai
- Department of Periodontics, Chettinad Dental College and Research Institute, Chennai, IND
| |
Collapse
|
12
|
Mateos-Olivares M, Pastor-Idoate S, Martín-Vallejo J, García-Vazquez C, Pastor JC, Usategui-Martín R, Sobas EM. Stress and sleep deprivation-related biomarkers in saliva in patients with retinitis pigmentosa. PLoS One 2024; 19:e0304261. [PMID: 38870197 PMCID: PMC11175419 DOI: 10.1371/journal.pone.0304261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/08/2024] [Indexed: 06/15/2024] Open
Abstract
PURPOSE Patients with Retinitis Pigmentosa (RP) commonly experience sleep-related issues and are susceptible to stress. Moreover, variatiaons in their vision are often linked to anxiety, stress and drowsiness, indicating that stress and sleep deprivation lead to a decline in vision, and vision improves when both are mitigated. The objective of this study was to investigate the utility of salivary biomarkers as biochemical indicators of anxiety and sleep deprivation in RP patients. METHODS Seventy-eight RP patients and 34 healthy controls were included in this observational study. Anxiety and sleep-quality questionnaires, a complete ophthalmological exam for severity grading and, the collection of salivary samples from participants were assessed for participants. The activity of biomarkers was estimated by ELISA, and statistical analysis was performed to determine associations between the parameters. Associations between underlying psychological factors, grade of disease severity, and biomarkers activity were also examined. RESULTS Fifty-two (67%) of patients had a severe RP, and 26 (33%) had a mild-moderate grade. Fifty-eight (58,9%) patients reported severe levels of anxiety and 18 (23.,1%) a high level. Forty-six (59%) patients obtained pathological values in sleep-quality questionaries and 43 (55.1%) in sleepiness. Patients with RP exhibited significant differences in testosterone, cortisol, sTNFαRII, sIgA and melatonin as compared to controls and patients with a mild-moderate and advanced stage of disease showed greater differences. In covariate analysis, patients with a severe anxiety level also showed greater differences in mean salivary cortisol, sTNFαRII and melatonin and male patients showed lower IgA levels than female. CONCLUSIONS The present findings suggest that salivary biomarkers could be suitable non-invasive biochemical markers for the objective assessment of sleep deprivation and anxiety in RP patients. Further research is needed to characterize the effects of untreated negative psychological states and sleep deprivation on increased variability of vision and disease progression, if any.
Collapse
Affiliation(s)
- Milagros Mateos-Olivares
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Department of Ophthalmology, Clinical University Hospital of Valladolid, Valladolid, Spain
| | - Salvador Pastor-Idoate
- Department of Ophthalmology, Clinical University Hospital of Valladolid, Valladolid, Spain
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Networks of Cooperative Research oriented to Health Results (RICORS), Carlos III Health Institute, Madrid, Spain
| | - Javier Martín-Vallejo
- Department of Statistics, Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca, Spain
| | | | - José Carlos Pastor
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Networks of Cooperative Research oriented to Health Results (RICORS), Carlos III Health Institute, Madrid, Spain
| | - Ricardo Usategui-Martín
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Networks of Cooperative Research oriented to Health Results (RICORS), Carlos III Health Institute, Madrid, Spain
- Faculty of Medicine, Department of Cell Biology, Genetics, Histology and Pharmacology, University of Valladolid, Valladolid, Spain
| | - Eva María Sobas
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, Valladolid, Spain
- Networks of Cooperative Research oriented to Health Results (RICORS), Carlos III Health Institute, Madrid, Spain
- Faculty of Nursing, University of Valladolid, Valladolid, Spain
| |
Collapse
|
13
|
Madi M, Abdelsalam M, Elakel A, Zakaria O, AlGhamdi M, Alqahtani M, AlMuhaish L, Farooqi F, Alamri TA, Alhafid IA, Alzahrani IM, Alam AH, Alhashmi MT, Alasseri IA, AlQuorain AA, AlQuorain AA. Salivary interleukin-17A and interleukin-18 levels in patients with celiac disease and periodontitis. PeerJ 2024; 12:e17374. [PMID: 38756445 PMCID: PMC11097963 DOI: 10.7717/peerj.17374] [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: 10/27/2023] [Accepted: 04/19/2024] [Indexed: 05/18/2024] Open
Abstract
Background An increased level of interleukin-17A and interleukin-18 in the serum and intestinal mucosa of celiac disease patients reflecting the severity of villous atrophy and inflammation was documented. Thus, the objective of this study was to evaluate the concentrations of salivary-17A, interleukin-1 beta, and interleukin-18 in patients with celiac disease who are on a gluten-free diet, both with and without periodontitis, and to compare these levels with those in healthy individuals. Methods The study involved 23 participants with serologically confirmed celiac disease (CD) and 23 control subjects. The CD patients had been following a gluten-free diet (GFD) for a minimum of 1 year and had no other autoimmune disorders. The research involved collecting demographic data, conducting periodontal examinations, gathering unstimulated whole saliva, and performing enzyme-linked immunosorbent assays to measure salivary interleukin-17A, interleukin-1 beta, and interleukin-18 levels. Spearman's correlation analysis was utilized to explore the relationships between CD markers in patients on a GFD and their periodontal clinical findings. Results The periodontal findings indicated significantly lower values in celiac disease patients adhering to a gluten-free diet compared to control subjects (p = 0.001). No significant differences were found in salivary IL-17A, IL-18, and IL-1B levels between celiac disease patients and control subjects. Nevertheless, the levels of all interleukins were elevated in periodontitis patients in both the celiac and control groups. The IL-1 Beta level was significantly higher in periodontitis patients compared to non-periodontitis patients in the control group (p = 0.035). Significant negative correlations were observed between serum IgA levels and plaque index (r = -0.460, p = 0.010), as well as gingival index (r = -0.396, p = 0.030) in CD patients on a gluten-free diet. Conclusion Celiac disease patients on gluten-free diet exhibited better periodontal health compared to control subjects. However, increased levels of salivary IL-17A, IL-18 and IL-1B levels were associated with periodontitis. Additionally, serum IgA level was significantly inversely associated with periodontitis clinical manifestations and with salivary inflammatory mediators in CD patients on GFD.
Collapse
Affiliation(s)
- Marwa Madi
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Maha Abdelsalam
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Ahmed Elakel
- Department of Preventive Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Osama Zakaria
- Department of Biomedical Dental Sciences, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Maher AlGhamdi
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Mohammed Alqahtani
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Luba AlMuhaish
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Faraz Farooqi
- Department of Dental Education, College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Turki A. Alamri
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Ibrahim A. Alhafid
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Ibrahim M. Alzahrani
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Adel H. Alam
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Majed T. Alhashmi
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Ibrahim A. Alasseri
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Ahmad A. AlQuorain
- College of medicine, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Abdulaziz A. AlQuorain
- Department of Internal Medicine, Gastroenterology Division, King Fahad University Hospital, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| |
Collapse
|
14
|
Rocha VAD, Cruz-Machado SDS, Silva IA, Fernandes PACM, Markus RP, Bueno M. Identification of Inflammatory Mediators in Saliva Samples From Hospitalized Newborns: Potential Biomarkers? Clin Nurs Res 2024; 33:207-219. [PMID: 38506123 DOI: 10.1177/10547738241238249] [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: 03/21/2024]
Abstract
Saliva measurements serve as a noninvasive tool for clinically monitoring newborns (NB) and children, a vulnerable population with promising potential for both research and clinical practice. Saliva acts as a repository for various inflammatory biomarkers involved in diverse biological functions. Particularly for children, it offers numerous advantages when compared to plasma and urine sampling. Nevertheless, there is a significant knowledge gap regarding detectable levels of cytokines in the saliva of newborns and children, as well as studies aiming to assess the relationship of this content with physiological and pathological processes. OBJECTIVES To characterize the levels of 11 inflammatory mediators (IFNg, IL1b, IL2, IL4, IL6, IL8, IL10, IL12, IL17, TNF, and VEGF) in saliva samples from NB on the first and second day of hospitalization in the Neonatal Intensive Care Unit (NICU). METHOD Exploratory study, descriptive, nested within a primary clinical, observational, and prospective study, conducted in the NICU of a public hospital in São Paulo, Brazil. Demographic data and vital signs were recorded in the clinical records of 90 NB, and five saliva samples from 5 NB were collected between the first and second day of life (D1-D2) at approximately 8-hr intervals (8-9 am, 4-5 pm, and 11-12 pm). Saliva samples were used for the measurement of 11 cytokines (IFNg, IL1b, IL2, IL4, IL6, IL8, IL10, IL12, IL17, TNF, and VEGF). RESULTS Five NBs participated in this exploratory study, and the vital signs showed variability from the first (D1) to the second day (D2) of hospitalization, variability similar to that of the total population of the primary study. The presence and levels of the 11 cytokines were detected in the saliva samples, as well as a statistical correlation between 10 cytokines (IFNg, IL1b, IL2, IL4, IL6, IL10, IL12, IL17, TNF, and VEGF) and vital signs. CONCLUSIONS The novelty of measuring inflammatory mediators in saliva samples from hospitalized NBs in the NICU is highlighted, providing support and new perspectives for the development of clinical and experimental research and an opportunity for developing and implementing new salivary biomarkers in different population segments.
Collapse
Affiliation(s)
- Vanderlei Amadeu da Rocha
- Universidade de São Paulo, Hospital Universitário, Unidade de Terapia Intensiva Pediátrica e Neonatal, São Paulo, SP, Brasil
| | | | - Isília Aparecida Silva
- Escola de Enfermagem, Departamento de Enfermagem Materno-Infantil e Psiquiatrica, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Regina Pekelmann Markus
- Universidade de São Paulo, Instituto de Biociências, Laboratório de Cronofarmacologia, São Paulo, SP, Brasil
| | | |
Collapse
|
15
|
Myochin H, Ohshima N, Izumi T, Hisajima T, Chaleckis R, Mori M. Capillary electrophoresis using triple layer modified capillary facilitating salivary ion analyses: Application to search for potential stress markers induced by cold pressure test. J Chromatogr A 2024; 1720:464769. [PMID: 38442499 DOI: 10.1016/j.chroma.2024.464769] [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: 11/29/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024]
Abstract
In this study, we introduce a novel approach for the analysis of salivary ions using capillary electrophoresis (CE) with a triple-layer coated capillary. The capillary is sequentially coated with cationic silylating reagents, poly(vinylsulfonate), and polybrene to form a custom designed surface that effectively inhibits adsorption of protein matrix on the capillary inner wall and allows for reproducible ion analysis. For the CE with capacitively coupled contactless conductivity detection, we used suitable background electrolytes (BGEs) for salivary ion analysis. Anions were separated using a mixture of 2-(N-morpholino)ethanesulfonic acid and l-arginine, and cations were separated using that with 18-crown-6. This setup enabled rapid separation, within 4 min, together with sensitive detection. We quantified nine common anions and five cations typically found in saliva samples using this CE method, both before and after a cold pressure test (CPT, a standard stress test). The CE system demonstrated consistent ion separation across 30 consecutive measurements without requiring capillary replacement. Notably, the salivary ion balance remained predominantly anion-rich, regardless of the CPT. Cold water exposure induced greater variation in the total anion concentration than in the total cation concentration. Further analysis using multiple regression analysis revealed strong relationships between nitrate and nitrite, formate and phosphate, and potassium and nitrate, before and after the CPT. Notably, potassium and nitrate ions exhibited variations in response to stress. These results provided a method for assessing salivary ion composition and insights into the potential of salivary ions as biomarkers for stress.
Collapse
Affiliation(s)
- Hironori Myochin
- Department of Chemistry and Life Science, Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan
| | - Noriyasu Ohshima
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan
| | - Takashi Izumi
- Department of Biochemistry, Graduate School of Medicine, Gunma University, Maebashi, Gunma 371-8511, Japan; Faculty of Health Care, Teikyo Heisei University, 2-51-4, Higashiikebukuro, Toshima-ku, Tokyo 170-8445, Japan
| | - Tatsuya Hisajima
- Faculty of Health Care, Teikyo Heisei University, 2-51-4, Higashiikebukuro, Toshima-ku, Tokyo 170-8445, Japan
| | - Romanas Chaleckis
- Graduate School of Medical Sciences, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Masanobu Mori
- Department of Chemistry and Life Science, Faculty of Science and Technology, Kochi University, 2-5-1, Akebono-cho, Kochi 780-8520, Japan.
| |
Collapse
|
16
|
Mortazavi H, Yousefi-Koma AA, Yousefi-Koma H. Extensive comparison of salivary collection, transportation, preparation, and storage methods: a systematic review. BMC Oral Health 2024; 24:168. [PMID: 38308289 PMCID: PMC10837873 DOI: 10.1186/s12903-024-03902-w] [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: 09/08/2023] [Accepted: 01/16/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Human saliva as a bodily fluid-similar to blood-is utilized for diagnostic purposes. Unlike blood sampling, collecting saliva is non-invasive, inexpensive, and readily accessible. There are no previously published systematic reviews regarding different collection, transportation, preparation, and storage methods for human saliva. DESIGN This study has been prepared and organized according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) 2020 guidelines. This systematic review has been registered at PROSPERO (Registration ID: CRD42023415384). The study question according to the PICO format was as followed: Comparison of the performance (C) of different saliva sampling, handling, transportation, and storage techniques and methods (I) assessed for analyzing stimulated or unstimulated human saliva (P and O). An electronic search was executed in Scopus, Google Scholar, and PubMed. RESULTS Twenty-three descriptive human clinical studies published between 1995 and 2022 were included. Eight categories of salivary features and biomarkers were investigated (i.e., salivary flow rate, total saliva quantity, total protein, cortisol, testosterone, DNA quality and quantity, pH and buffering pH). Twenty-two saliva sampling methods/devices were utilized. Passive drooling, Salivette®, and spitting were the most utilized methods. Sampling times with optimum capabilities for cortisol, iodine, and oral cancer metabolites are suggested to be 7:30 AM to 9:00 AM, 10:30 AM to 11:00 AM, and 14:00 PM to 20:00 PM, respectively. There were 6 storage methods. Centrifuging samples and storing them at -70 °C to -80 °C was the most utilized storage method. For DNA quantity and quality, analyzing samples immediately after collection without centrifuging or storage, outperformed centrifuging samples and storing them at -70 °C to -80 °C. Non-coated Salivette® was the most successful method/device for analyzing salivary flow rate. CONCLUSION It is highly suggested that scientists take aid from the reported categorized outcomes, and design their study questions based on the current voids for each method/device.
Collapse
Affiliation(s)
- Hamed Mortazavi
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Shahid Chamran Highway, Tehran, 1983963113, Iran
| | - Amir-Ali Yousefi-Koma
- School of Dentistry, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd, Evin, Shahid Chamran Highway, Tehran, 1983963113, Iran.
- Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
17
|
Welsh JA, Goberdhan DCI, O'Driscoll L, Buzas EI, Blenkiron C, Bussolati B, Cai H, Di Vizio D, Driedonks TAP, Erdbrügger U, Falcon‐Perez JM, Fu Q, Hill AF, Lenassi M, Lim SK, Mahoney MG, Mohanty S, Möller A, Nieuwland R, Ochiya T, Sahoo S, Torrecilhas AC, Zheng L, Zijlstra A, Abuelreich S, Bagabas R, Bergese P, Bridges EM, Brucale M, Burger D, Carney RP, Cocucci E, Colombo F, Crescitelli R, Hanser E, Harris AL, Haughey NJ, Hendrix A, Ivanov AR, Jovanovic‐Talisman T, Kruh‐Garcia NA, Ku'ulei‐Lyn Faustino V, Kyburz D, Lässer C, Lennon KM, Lötvall J, Maddox AL, Martens‐Uzunova ES, Mizenko RR, Newman LA, Ridolfi A, Rohde E, Rojalin T, Rowland A, Saftics A, Sandau US, Saugstad JA, Shekari F, Swift S, Ter‐Ovanesyan D, Tosar JP, Useckaite Z, Valle F, Varga Z, van der Pol E, van Herwijnen MJC, Wauben MHM, Wehman AM, Williams S, Zendrini A, Zimmerman AJ, MISEV Consortium, Théry C, Witwer KW. Minimal information for studies of extracellular vesicles (MISEV2023): From basic to advanced approaches. J Extracell Vesicles 2024; 13:e12404. [PMID: 38326288 PMCID: PMC10850029 DOI: 10.1002/jev2.12404] [Citation(s) in RCA: 602] [Impact Index Per Article: 602.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024] Open
Abstract
Extracellular vesicles (EVs), through their complex cargo, can reflect the state of their cell of origin and change the functions and phenotypes of other cells. These features indicate strong biomarker and therapeutic potential and have generated broad interest, as evidenced by the steady year-on-year increase in the numbers of scientific publications about EVs. Important advances have been made in EV metrology and in understanding and applying EV biology. However, hurdles remain to realising the potential of EVs in domains ranging from basic biology to clinical applications due to challenges in EV nomenclature, separation from non-vesicular extracellular particles, characterisation and functional studies. To address the challenges and opportunities in this rapidly evolving field, the International Society for Extracellular Vesicles (ISEV) updates its 'Minimal Information for Studies of Extracellular Vesicles', which was first published in 2014 and then in 2018 as MISEV2014 and MISEV2018, respectively. The goal of the current document, MISEV2023, is to provide researchers with an updated snapshot of available approaches and their advantages and limitations for production, separation and characterisation of EVs from multiple sources, including cell culture, body fluids and solid tissues. In addition to presenting the latest state of the art in basic principles of EV research, this document also covers advanced techniques and approaches that are currently expanding the boundaries of the field. MISEV2023 also includes new sections on EV release and uptake and a brief discussion of in vivo approaches to study EVs. Compiling feedback from ISEV expert task forces and more than 1000 researchers, this document conveys the current state of EV research to facilitate robust scientific discoveries and move the field forward even more rapidly.
Collapse
Affiliation(s)
- Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of PathologyNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Deborah C. I. Goberdhan
- Nuffield Department of Women's and Reproductive HealthUniversity of Oxford, Women's Centre, John Radcliffe HospitalOxfordUK
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublinIreland
- Trinity Biomedical Sciences InstituteTrinity College DublinDublinIreland
- Trinity St. James's Cancer InstituteTrinity College DublinDublinIreland
| | - Edit I. Buzas
- Department of Genetics, Cell‐ and ImmunobiologySemmelweis UniversityBudapestHungary
- HCEMM‐SU Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
- HUN‐REN‐SU Translational Extracellular Vesicle Research GroupSemmelweis UniversityBudapestHungary
| | - Cherie Blenkiron
- Faculty of Medical and Health SciencesThe University of AucklandAucklandNew Zealand
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TurinTurinItaly
| | | | - Dolores Di Vizio
- Department of Surgery, Division of Cancer Biology and TherapeuticsCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Tom A. P. Driedonks
- Department CDL ResearchUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Uta Erdbrügger
- University of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Juan M. Falcon‐Perez
- Exosomes Laboratory, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- Metabolomics Platform, Center for Cooperative Research in BiosciencesBasque Research and Technology AllianceDerioSpain
- IKERBASQUE, Basque Foundation for ScienceBilbaoSpain
| | - Qing‐Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Extracellular Vesicle Research and Clinical Translational CenterThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityMelbourneAustralia
| | - Metka Lenassi
- Faculty of MedicineUniversity of LjubljanaLjubljanaSlovenia
| | - Sai Kiang Lim
- Institute of Molecular and Cell Biology (IMCB)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
- Paracrine Therapeutics Pte. Ltd.SingaporeSingapore
- Department of Surgery, YLL School of MedicineNational University SingaporeSingaporeSingapore
| | - Mỹ G. Mahoney
- Thomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sujata Mohanty
- Stem Cell FacilityAll India Institute of Medical SciencesNew DelhiIndia
| | - Andreas Möller
- Chinese University of Hong KongHong KongHong Kong S.A.R.
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Susmita Sahoo
- Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ana C. Torrecilhas
- Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Departamento de Ciências Farmacêuticas, Instituto de Ciências Ambientais, Químicas e FarmacêuticasUniversidade Federal de São Paulo (UNIFESP) Campus DiademaDiademaBrazil
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Andries Zijlstra
- Department of PathologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- GenentechSouth San FranciscoCaliforniaUSA
| | - Sarah Abuelreich
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Reem Bagabas
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Paolo Bergese
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
- National Center for Gene Therapy and Drugs based on RNA TechnologyPaduaItaly
| | - Esther M. Bridges
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordUK
| | - Marco Brucale
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Dylan Burger
- Kidney Research CentreOttawa Hopsital Research InstituteOttawaCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaCanada
- School of Pharmaceutical SciencesUniversity of OttawaOttawaCanada
| | - Randy P. Carney
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Emanuele Cocucci
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
- Comprehensive Cancer CenterThe Ohio State UniversityColumbusOhioUSA
| | - Federico Colombo
- Division of Pharmaceutics and Pharmacology, College of PharmacyThe Ohio State UniversityColumbusOhioUSA
| | - Rossella Crescitelli
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational Medicine, Institute of Clinical SciencesSahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Edveena Hanser
- Department of BiomedicineUniversity Hospital BaselBaselSwitzerland
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | | | - Norman J. Haughey
- Departments of Neurology and PsychiatryJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and RepairGhent UniversityGhentBelgium
- Cancer Research Institute GhentGhentBelgium
| | - Alexander R. Ivanov
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | - Tijana Jovanovic‐Talisman
- Department of Cancer Biology and Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Nicole A. Kruh‐Garcia
- Bio‐pharmaceutical Manufacturing and Academic Resource Center (BioMARC)Infectious Disease Research Center, Colorado State UniversityFort CollinsColoradoUSA
| | - Vroniqa Ku'ulei‐Lyn Faustino
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Diego Kyburz
- Department of BiomedicineUniversity of BaselBaselSwitzerland
- Department of RheumatologyUniversity Hospital BaselBaselSwitzerland
| | - Cecilia Lässer
- Krefting Research Centre, Department of Internal Medicine and Clinical NutritionInstitute of Medicine at Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Kathleen M. Lennon
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Jan Lötvall
- Krefting Research Centre, Institute of Medicine at Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Adam L. Maddox
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Elena S. Martens‐Uzunova
- Erasmus MC Cancer InstituteUniversity Medical Center Rotterdam, Department of UrologyRotterdamThe Netherlands
| | - Rachel R. Mizenko
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Lauren A. Newman
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andrea Ridolfi
- Department of Physics and Astronomy, and LaserLaB AmsterdamVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Eva Rohde
- Department of Transfusion Medicine, University HospitalSalzburger Landeskliniken GmbH of Paracelsus Medical UniversitySalzburgAustria
- GMP Unit, Paracelsus Medical UniversitySalzburgAustria
- Transfer Centre for Extracellular Vesicle Theralytic Technologies, EV‐TTSalzburgAustria
| | - Tatu Rojalin
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCaliforniaUSA
- Expansion Therapeutics, Structural Biology and BiophysicsJupiterFloridaUSA
| | - Andrew Rowland
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Andras Saftics
- Department of Molecular Medicine, Beckman Research InstituteCity of Hope Comprehensive Cancer CenterDuarteCaliforniaUSA
| | - Ursula S. Sandau
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative MedicineOregon Health & Science UniversityPortlandOregonUSA
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Celer DiagnosticsTorontoCanada
| | - Simon Swift
- Waipapa Taumata Rau University of AucklandAucklandNew Zealand
| | - Dmitry Ter‐Ovanesyan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMassachusettsUSA
| | - Juan P. Tosar
- Universidad de la RepúblicaMontevideoUruguay
- Institut Pasteur de MontevideoMontevideoUruguay
| | - Zivile Useckaite
- College of Medicine and Public HealthFlinders UniversityAdelaideAustralia
| | - Francesco Valle
- Consiglio Nazionale delle Ricerche ‐ Istituto per lo Studio dei Materiali NanostrutturatiBolognaItaly
- Consorzio Interuniversitario per lo Sviluppo dei Sistemi a Grande InterfaseFlorenceItaly
| | - Zoltan Varga
- Biological Nanochemistry Research GroupInstitute of Materials and Environmental Chemistry, Research Centre for Natural SciencesBudapestHungary
- Department of Biophysics and Radiation BiologySemmelweis UniversityBudapestHungary
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, Location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, location AMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Martijn J. C. van Herwijnen
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Marca H. M. Wauben
- Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | | - Andrea Zendrini
- Department of Molecular and Translational MedicineUniversity of BresciaBresciaItaly
- Center for Colloid and Surface Science (CSGI)FlorenceItaly
| | - Alan J. Zimmerman
- Barnett Institute of Chemical and Biological Analysis, Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMassachusettsUSA
| | | | - Clotilde Théry
- Institut Curie, INSERM U932PSL UniversityParisFrance
- CurieCoreTech Extracellular Vesicles, Institut CurieParisFrance
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- EV Core Facility “EXCEL”, Institute for Basic Biomedical SciencesJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
18
|
Blümer M, Heeren J, Mirkovic B, Latzel M, Gordon C, Crowhen D, Meis M, Wagener K, Schulte M. The Impact of Hearing Aids on Listening Effort and Listening-Related Fatigue - Investigations in a Virtual Realistic Listening Environment. Trends Hear 2024; 28:23312165241265199. [PMID: 39095047 PMCID: PMC11378347 DOI: 10.1177/23312165241265199] [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: 08/04/2024] Open
Abstract
Participation in complex listening situations such as group conversations in noisy environments sets high demands on the auditory system and on cognitive processing. Reports of hearing-impaired people indicate that strenuous listening situations occurring throughout the day lead to feelings of fatigue at the end of the day. The aim of the present study was to develop a suitable test sequence to evoke and measure listening effort (LE) and listening-related fatigue (LRF), and, to evaluate the influence of hearing aid use on both dimensions in mild to moderately hearing-impaired participants. The chosen approach aims to reconstruct a representative acoustic day (Time Compressed Acoustic Day [TCAD]) by means of an eight-part hearing-test sequence with a total duration of approximately 2½ h. For this purpose, the hearing test sequence combined four different listening tasks with five different acoustic scenarios and was presented to the 20 test subjects using virtual acoustics in an open field measurement in aided and unaided conditions. Besides subjective ratings of LE and LRF, behavioral measures (response accuracy, reaction times), and an attention test (d2-R) were performed prior to and after the TCAD. Furthermore, stress hormones were evaluated by taking salivary samples. Subjective ratings of LRF increased throughout the test sequence. This effect was observed to be higher when testing unaided. In three of the eight listening tests, the aided condition led to significantly faster reaction times/response accuracies than in the unaided condition. In the d2-R test, an interaction in processing speed between time (pre- vs. post-TCAD) and provision (unaided vs. aided) was found suggesting an influence of hearing aid provision on LRF. A comparison of the averaged subjective ratings at the beginning and end of the TCAD shows a significant increase in LRF for both conditions. At the end of the TCAD, subjective fatigue was significantly lower when wearing hearing aids. The analysis of stress hormones did not reveal significant effects.
Collapse
Affiliation(s)
- M Blümer
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - J Heeren
- Hörzentrum Oldenburg gGmbH, Oldenburg, Germany
- Cluster of Excellence Hearing4All, Oldenburg, Germany
| | - B Mirkovic
- Department of Psychology, University of Oldenburg School VI-Medicine and Health Sciences, Oldenburg, Germany
| | - M Latzel
- Sonova Holding AG, Stäfa, Switzerland
| | - C Gordon
- Sonova New Zealand, Auckland, New Zealand
| | - D Crowhen
- Sonova New Zealand, Auckland, New Zealand
| | - M Meis
- Hörzentrum Oldenburg gGmbH, Oldenburg, Germany
- Cluster of Excellence Hearing4All, Oldenburg, Germany
| | - K Wagener
- Hörzentrum Oldenburg gGmbH, Oldenburg, Germany
- Cluster of Excellence Hearing4All, Oldenburg, Germany
| | - M Schulte
- Hörzentrum Oldenburg gGmbH, Oldenburg, Germany
- Cluster of Excellence Hearing4All, Oldenburg, Germany
| |
Collapse
|
19
|
Pai J, Joshi RK, Bhaskar S, Patil S, Bv S, R S, Iyengar AR, Agarwal N, Shorie M. Revisiting the cortisol reference ranges in humans: the role of demographics. Endocrine 2023; 82:414-418. [PMID: 37501014 DOI: 10.1007/s12020-023-03456-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
The current study explores the effect of demographics on serum cortisol expression in a study group of 52 individuals to improve the current serum reference ranges to produce personalized expression profiles consequently increasing clinical confidence in the diagnosis. The serum cortisol concentration was inspected against demographical data like age, sex, and body mass index and showed an association with age and sex. The serum cortisol values also indicated a positive association with chronic illnesses however this finding requires a more focused study for establishment. Additionally, saliva samples are also collected from the same study group at the same time through drool and an absorbent sponge and correlated with serum values to draw an alternative route of serological testing. Salivary cortisol from drool showed a linear correlation with Pearson's correlation coefficients of 0.71 and 0.72 with serum cortisol and with saliva samples collected using a porous sponge respectively. Overall, the study shows the role of demographics in shaping the reference ranges for cortisol, suggesting a path for developing personalized diagnostics. The study also highlights the efficacy of saliva as an alternative to serum cortisol to facilitate cortisol measurement for efficient stress management.
Collapse
Affiliation(s)
- Jayeeta Pai
- Cortiqa Health Private Limited, Bangalore, India.
| | | | | | - Seema Patil
- DA Pandu Memorial RV Dental College, Bangalore, India
| | - Subhash Bv
- DA Pandu Memorial RV Dental College, Bangalore, India
| | - Shriyanka R
- DA Pandu Memorial RV Dental College, Bangalore, India
| | | | | | | |
Collapse
|
20
|
Garg S, Sachdeva A, Peeters M, McClements J. Point-of-Care Prostate Specific Antigen Testing: Examining Translational Progress toward Clinical Implementation. ACS Sens 2023; 8:3643-3658. [PMID: 37830899 PMCID: PMC10616866 DOI: 10.1021/acssensors.3c01402] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Prostate cancer (PCa) is the second most common male cancer and is attributable to over 375,000 deaths annually. Prostate specific antigen (PSA) is a key biomarker for PCa and therefore measuring patient PSA levels is an important aspect of the diagnostic pathway. Automated immunoassays are currently utilized for PSA analysis, but they require a laboratory setting with specialized equipment and trained personnel. This results in high diagnostic costs, extended therapeutic turnaround times, and restrictions on testing capabilities in resource-limited settings. Consequently, there is a strong drive to develop point-of-care (PoC) PSA tests that can offer accurate, low-cost, and rapid results at the time and place of the patient. However, many emerging PoC tests experience a trade-off between accuracy, affordability, and accessibility which distinctly limits their translational potential. This review comprehensively assesses the translational advantages and limitations of emerging laboratory-level and commercial PoC tests for PSA determination. Electrochemical and optical PSA sensors from 2013 to 2023 are systematically examined. Furthermore, we suggest how the translational potential of emerging tests can be optimized to achieve clinical implementation and thus improve PCa diagnosis globally.
Collapse
Affiliation(s)
- Saweta Garg
- Merz
Court, School of Engineering, Newcastle
University, Claremont Road, NE1 7RU Newcastle upon Tyne, U.K.
- Department
of Chemical Engineering and Analytical Science, School of Engineering, University of Manchester, Manchester M20 4BX, U.K.
| | - Ashwin Sachdeva
- Division
of Cancer Sciences, University of Manchester, Wilmslow Road, Manchester M20 4BX, U.K.
- Department
of Urology, The Christie NHS Foundation
Trust, Manchester M20 4BX, U.K.
| | - Marloes Peeters
- Merz
Court, School of Engineering, Newcastle
University, Claremont Road, NE1 7RU Newcastle upon Tyne, U.K.
| | - Jake McClements
- Merz
Court, School of Engineering, Newcastle
University, Claremont Road, NE1 7RU Newcastle upon Tyne, U.K.
| |
Collapse
|
21
|
Liu S, Hou Y, Li Z, Yang C, Liu G. μPADs on Centrifugal Microfluidic Discs for Rapid Sample-to-Answer Salivary Diagnostics. ACS Sens 2023; 8:3520-3529. [PMID: 37669403 DOI: 10.1021/acssensors.3c01093] [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: 09/07/2023]
Abstract
A fully integrated device for salivary detection with a sample-in-answer-out fashion is critical for noninvasive point-of-care testing (POCT), especially for the screening of contagious disease infection. Microfluidic paper-based analytical devices (μPADs) have demonstrated their huge potential in POCT due to their low cost and easy adaptation with other components. This study developed a generic POCT platform by integrating a centrifugal microfluidic disc with μPADs to realize sample-to-answer salivary diagnostics. Specifically, a custom centrifugal microfluidic disc integrated with μPADs is fabricated, which demonstrated a high efficiency in saliva treatment. To demonstrate the capability of the integrated device for salivary analysis, the SARS-CoV-2 Nucleocapsid (N) protein, a reliable biomarker for SARS-CoV-2 acute infection, is used as the model analyte. By the chemical treatment of the μPAD surface, and by optimizing the protein immobilization conditions, the on-disc μPADs were able to detect the SARS-CoV-2 N protein down to 10 pg mL-1 with a dynamic range of 10-1000 pg mL-1 and an assay time of 8 min. The integrated device was successfully used for the quantification of the N protein of pseudovirus in saliva with high specificity and demonstrated a comparable performance to the commercial paper lateral flow assay test strips.
Collapse
Affiliation(s)
- Shixian Liu
- CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yuting Hou
- CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Zirui Li
- CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Chenyu Yang
- CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Guozhen Liu
- CUHK(SZ)-Boyalife Joint Laboratory of Regenerative Medicine Engineering, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
22
|
Futane A, Senthil M, S J, Srinivasan A, R K, Narayanamurthy V. Sweat analysis for urea sensing: trends and challenges. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4405-4426. [PMID: 37646163 DOI: 10.1039/d3ay01089a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
With increasing population there is a rise in pathological diseases that the healthcare facilities are grappling with. Sweat-based wearable technologies for continuous monitoring have overcome the demerits associated with sweat sampling and sensing. Hence, sweat as an alternative biofluid holds great promise for the quantification of a host of biomarkers and understanding the functioning of the body, thereby deducing ailments quickly and economically. This comprehensive review accounts for recent advances in sweat-based LOCs (Lab-On-Chips), which are a likely alternative to the existing blood-urea sample testing that is invasive and time-consuming. The present review is focused on the advancements in sweat-based Lab-On-Chips (LOCs) as an alternative to invasive and time-consuming blood-urea sample testing. In addition, different sweat collection methods (direct skin, near skin and microfluidic) and their mechanism for urea sensing are explained in detail. The mechanism of urea in biofluids in protein metabolism, balancing nitrogen levels and a crucial factor of kidney function is described. In the end, research and technological advancements are explained to address current challenges and enable its widespread implementation.
Collapse
Affiliation(s)
- Abhishek Futane
- Fakulti Kejuruteraan Elektronik dan Kejuruteraan Komputer, Universiti Teknikal Malaysia Melaka, Hang Tuah Jaya, 76100 Durian Tunggal, Melaka, Malaysia
| | - Mallika Senthil
- Department of Biomedical Engineering, Rajalakshmi Engineering, College, Chennai, India 602105
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jayashree S
- Department of Biomedical Engineering, Rajalakshmi Engineering, College, Chennai, India 602105
| | - Arthi Srinivasan
- Faculty of Chemical and Process Engineering Technology, University Malaysia Pahang (UMP), Lebuhraya Tun Razak, 26300 Gambang, Kunatan, Pahang, Malaysia
| | - Kalpana R
- Department of Biomedical Engineering, Rajalakshmi Engineering, College, Chennai, India 602105
| | - Vigneswaran Narayanamurthy
- Advance Sensors and Embedded Systems (ASECs), Centre for Telecommunication Research & Innovation, Fakulti Teknologi Kejuruteraan Elektrik Dan Elektronik, Universiti Teknikal Malaysia Melaka, Hang Tuah Jaya, Durian Tunggal, Melaka 76100, Malaysia
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India.
| |
Collapse
|
23
|
Bido AT, Ember KJI, Trudel D, Durand M, Leblond F, Brolo AG. Detection of SARS-CoV-2 in saliva by a low-cost LSPR-based sensor. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:3955-3966. [PMID: 37530390 DOI: 10.1039/d3ay00853c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
The SARS-CoV-2 pandemic started more than 3 years ago, but the containment of the spread is still a challenge. Screening is imperative for informed decision making by government authorities to contain the spread of the virus locally. The access to screening tests is disproportional, due to the lack of access to reagents, equipment, finances or because of supply chain disruptions. Low and middle-income countries have especially suffered with the lack of these resources. Here, we propose a low cost and easily constructed biosensor device based on localized surface plasmon resonance, or LSPR, for the screening of SARS-CoV-2. The biosensor device, dubbed "sensor" for simplicity, was constructed in two modalities: (1) viral detection in saliva and (2) antibody against COVID in saliva. Saliva collected from 18 patients were tested in triplicates. Both sensors successfully classified all COVID positive patients (among hospitalized and non-hospitalized). From the COVID negative patients 7/8 patients were correctly classified. For both sensors, sensitivity was determined as 100% (95% CI 79.5-100) and specificity as 87.5% (95% CI 80.5-100). The reagents and equipment used for the construction and deployment of this sensor are ubiquitous and low-cost. This sensor technology can then add to the potential solution for challenges related to screening tests in underserved communities.
Collapse
Affiliation(s)
- Ariadne Tuckmantel Bido
- Department of Chemistry, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia, V8P 5C2, Canada.
| | - Katherine J I Ember
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada
- Division of Neurology, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Dominique Trudel
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada
- Division of Neurology, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Madeleine Durand
- CHUM Research Center, Internal Medicine Service of the Centre Hospitalier de l'Univsersité de Montréal (CHUM), Canada
| | - Frederic Leblond
- Department of Engineering Physics, Polytechnique Montréal, Montreal, QC H3C 3A7, Canada
- Division of Neurology, Centre de recherche du Centre Hospitalier de l'Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Alexandre G Brolo
- Department of Chemistry, University of Victoria, 3800 Finnerty Road, Victoria, British Columbia, V8P 5C2, Canada.
- Centre for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
24
|
Basilicata M, Pieri M, Marrone G, Nicolai E, Di Lauro M, Paolino V, Tomassetti F, Vivarini I, Bollero P, Bernardini S, Noce A. Saliva as Biomarker for Oral and Chronic Degenerative Non-Communicable Diseases. Metabolites 2023; 13:889. [PMID: 37623833 PMCID: PMC10456419 DOI: 10.3390/metabo13080889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
Saliva is a very complex fluid and it is essential to maintain several physiological processes and functions, including oral health, taste, digestion and immunological defenses. Saliva composition and the oral microbiome can be influenced by several factors, like diet and smoking habits, and their alteration can represent an important access point for pathogens and, thus, for systemic illness onset. In this review, we explore the potentiality of saliva as a new tool for the early detection of some pathological conditions, such as oral diseases, chronic degenerative non-communicable diseases, among these chronic kidney disease (CKD). We also examined the possible correlation between oral and systemic diseases and oral and gut microbiota dysbiosis. In particular, we deeply analyzed the relationship between oral diseases and CKD. In this context, some salivary parameters can represent a new device to detect either oral or systemic pathologies. Moreover, the positive modulation of oral and gut microbiota induced by prebiotics, postbiotics, or symbiotics could represent a new possible adjuvant therapy in the clinical management of oral diseases and CKD.
Collapse
Affiliation(s)
- Michele Basilicata
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Giulia Marrone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Nicolai
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Manuela Di Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Vincenza Paolino
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Flaminia Tomassetti
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Vivarini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Patrizio Bollero
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Noce
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- UOSD Nephrology and Dialysis, Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
25
|
Sonpanao P, Janebodin K, Namvichaisirikul N, Thongjit S, Jitprasertwong P. The Prevalence of Xerostomia in Older Thai Individuals with Type II Diabetes Mellitus and Its Association with Type of Toothpaste and Oral Functions: A Cross-Sectional Study Using Questionnaires. Geriatrics (Basel) 2023; 8:76. [PMID: 37489324 PMCID: PMC10366830 DOI: 10.3390/geriatrics8040076] [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: 04/25/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023] Open
Abstract
AIM To investigate the prevalence of xerostomia in older people with diabetes mellitus and its impacts on oral functions, as well as to determine potential risk factors for xerostomia. METHODS An analytical cross-sectional study was conducted on 623 older type 2 diabetes mellitus (T2DM) Thai people using valid structural questionnaires. Patients were interviewed, and data were recorded. Xerostomia was assessed using subjective symptom questionnaires. Risk factors for xerostomia were analyzed using bivariate and multiple logistic regression analyses. RESULTS Among the study participants, 38.4% of the older T2DM people had xerostomia, which is associated with sex, age, type of toothpaste, years of diabetes, hemoglobin A1c level, other systemic diseases, medication, smoking, alcohol consumption, and denture wearing. It was significant that xerostomia was associated with toothpaste containing spicy herbal extracts (OR: 9.32 [3.46 to 15.25]), while toothpaste containing artificial sweeteners tended to lower the risk of xerostomia. In addition, older T2DM adults with xerostomia had greater impaired oral functions, which include difficulties in speaking (OR: 3.31 [1.11 to 9.80]), tasting (OR: 5.12 [3.26 to 8.06]), swallowing (OR: 3.59 [2.32 to 5.53]), and chewing (OR: 3.34 [1.15 to 5.82]). CONCLUSIONS Xerostomia is prevalent in older Thai people with T2DM. The results suggest that toothpaste containing spicy herbal extracts might increase the risk of xerostomia, resulting in various oral function problems. Therefore, greater awareness of xerostomia in this group should be raised to monitor dental health, and professionals should work in parallel with other aspects of oral health promotion.
Collapse
Affiliation(s)
- Panitan Sonpanao
- Translational Medicine (International Program), Institute of Medicine, Suranaree University of Technology (SUT), Nakhon Ratchasima 30000, Thailand
- School of Geriatric Oral Health, Institute of Dentistry, Suranaree University of Technology (SUT), Nakhon Ratchasima 30000, Thailand
| | - Kajohnkiart Janebodin
- Department of Anatomy, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Niwatchai Namvichaisirikul
- Department of Family Medicine and Community Medicine, Institute of Medicine, Suranaree University of Technology (SUT), Nakhon Ratchasima 30000, Thailand
| | - Supattarayan Thongjit
- School of Family and Community Oral Health, Institute of Dentistry, Suranaree University of Technology (SUT), Nakhon Ratchasima 30000, Thailand
| | - Paiboon Jitprasertwong
- School of Geriatric Oral Health, Institute of Dentistry, Suranaree University of Technology (SUT), Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
26
|
Bryant EM, Richardson R, Graham BM. The relationship between salivary Fibroblast Growth Factor-2 and cortisol reactivity and psychological outcomes prior to and during the COVID-19 pandemic. JOURNAL OF AFFECTIVE DISORDERS REPORTS 2023; 13:100606. [PMID: 37304226 PMCID: PMC10246939 DOI: 10.1016/j.jadr.2023.100606] [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: 01/09/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/13/2023] Open
Abstract
Background Fibroblast growth factor-2 (FGF2) is a biomarker that is associated with depression, anxiety and stress in rodents. In humans, we have previously demonstrated that salivary FGF2 increased following stress in a similar pattern to cortisol, and FGF2 (but not cortisol) reactivity predicted repetitive negative thinking, a transdiagnostic risk factor for mental illness. The current study assessed the relationship between FGF2, cortisol, and mental health before and during the COVID-19 pandemic. Methods We employed a longitudinal correlational design using a convenience sample. We assessed whether FGF2 and cortisol reactivity following the Trier Social Stress Task (TSST) were associated with DASS-21 depression, anxiety and stress, measured at the time of the TSST in 2019-20 (n = 87; time 1), and then again in May 2020 during the first wave of COVID-19 in Sydney (n = 34 of the original sample; time 2). Results FGF2 reactivity (but not absolute FGF2 levels) at time 1 predicted depression, anxiety, and stress across timepoints. Cortisol reactivity at time 1 was associated with stress over timepoints, and absolute cortisol levels were associated with depression across timepoints. Limitations The sample was comprised of mostly healthy participants from a student population, and there was high attrition between timepoints. The outcomes need to be replicated in larger, more diverse, samples. Conclusions FGF2 and cortisol may be uniquely predictive of mental health outcomes in healthy samples, potentially allowing for early identification of at-risk individuals.
Collapse
Affiliation(s)
- Emma M Bryant
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| | - Rick Richardson
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| | - Bronwyn M Graham
- University of New South Wales, School of Psychology, Sydney, NSW 2052, Australia
| |
Collapse
|
27
|
Ganie MW, Nainggolan IRA, Bestari R, Hazidar AH, Hasibuan M, Siregar J, Ichwan M, Kusumawati RL, Lubis IND. Use of saliva as an alternative diagnostic method for diagnosis of COVID-19. IJID REGIONS 2023:S2772-7076(23)00038-3. [PMID: 37363193 PMCID: PMC10157387 DOI: 10.1016/j.ijregi.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 06/28/2023]
Abstract
Background Mass population testing has been recommended to contain the spread of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. However, the use of nasopharyngeal swab specimens has caused many logistic challenges. This study investigated the sensitivity and specificity of saliva as a non-invasively-obtained specimen for molecular detection of SARS-CoV-2 RNA. Methods In total, 153 patients with confirmed coronavirus disease 2019 (COVID-19) who had been admitted to the regional referral hospital or who self-isolated at home were included in this study. Nasopharyngeal swab specimens and saliva samples were collected on the same day, and were tested for SARS-CoV-2 infection using reverse transcriptase polymerase chain reaction. Results The sensitivity and specificity of saliva samples were 81.5% and 76.4%, respectively, in cases that had been confirmed as COVID-19 using nasopharyngeal swab samples. Positive predictive values and negative predictive values were 92.3% and 54.1%, respectively. The highest detection rates were found among samples collected 4-7 days since symptom onset. Conclusion Saliva samples showed comparable performance to nasopharyngeal swab specimens for the diagnosis of COVID-19 in adults. The performance of saliva as a diagnostic specimen for COVID-19 testing is particularly significant during the first week of symptoms.
Collapse
Affiliation(s)
| | | | - Ramadhan Bestari
- Faculty of Medicine, Universitas Islam Sumatera Utara, Medan, Indonesia
| | - Al Hamidy Hazidar
- Faculty of Computer Science and information Technology, Universitas Muhammadiyah Sumatera Utara, Medan, Indonesia
- Institute IR 4.0, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Mirzan Hasibuan
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Jelita Siregar
- Department of Clinical Pathology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Muhammad Ichwan
- Department of Pharmacology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - R Lia Kusumawati
- Department of Microbiology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Inke Nadia Diniyanti Lubis
- Department of Paediatrics, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Menzies School of Health Research, Darwin, Australia
| |
Collapse
|
28
|
Abdulaziz Alsufyani A. Post-COVID-19 effect on biochemical parameters in children: Should we take heed? Saudi J Biol Sci 2023; 30:103649. [PMID: 37069947 PMCID: PMC10079315 DOI: 10.1016/j.sjbs.2023.103649] [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: 03/11/2023] [Revised: 03/16/2023] [Accepted: 03/31/2023] [Indexed: 04/09/2023] Open
Abstract
The aim of this research is to analyze the potential impact of the COVID-19 infection on the serum biochemical concentration of children 6 months after recovery from the infection. The study included 72 children with a median age of 11 years. The case group consisted of 37 children who had contracted COVID-19 6 months prior to the analysis. They reported no other pre- or post-covid chronic or systemic diseases. The control group consisted of 35 children who had no prior record of COVID-19 infection. The analysis showed a substantial variation (P = 0.026) in the mean urea values (mmol/L) between the case group (4.513 ± 0.839) and the control group (5.425 ± 1.173). However, both groups' urea levels were within the normal range of their age group. No statistical differences were found analyzing the variations between the two groups in the levels of LDH, AST, ALT, BiliT, GGT, AlbBCG2, CRP, CK, AlKP, UA, Phos, Crea2, Gluc, Ca, Na, K, Cl, TP, TC, TG, and HDL (P > 0.05). The DMFT score was substantially greater (P < 0.002) in the infected team (5.38 ± 2.841) in comparison to the non-infected group (2.6 ± 2.257). The study indicates that COVID-19 infection does not leave biochemical alterations among children who did not have pre-existing conditions. The biochemical analysis suggests that children recover better than adults from COVID-19. Furthermore, it calls for investigating non-lethal COVID-19 infection as a tool to discover underlying conditions. The DMFT score shows a correlation between COVID-19 infection and caries. However, the nature of the correlation is yet to be investigated.
Collapse
Affiliation(s)
- Amal Abdulaziz Alsufyani
- College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
- Ministry of the National Guard - Health Affairs, Jeddah, Saudi Arabia
| |
Collapse
|
29
|
Pundir M, De Rosa MC, Lobanova L, Abdulmawjood S, Chen X, Papagerakis S, Papagerakis P. Structural properties and binding mechanism of DNA aptamers sensing saliva melatonin for diagnosis and monitoring of circadian clock and sleep disorders. Anal Chim Acta 2023; 1251:340971. [PMID: 36925277 DOI: 10.1016/j.aca.2023.340971] [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: 11/14/2022] [Revised: 01/25/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Circadian desynchrony with the external light-dark cycle influences the rhythmic secretion of melatonin which is among the first signs of circadian rhythm sleep disorders. An accurate dim light melatonin onset (established indicator of circadian rhythm sleep disorders) measurement requires lengthy assays, and antibody affinities alterations, especially in patients with circadian rhythm disorders whose melatonin salivary levels vary significantly, making antibodies detection mostly inadequate. In contrast, aptamers with their numerous advantages (e.g., target selectivity, structural flexibility in tuning binding affinities, small size, etc.) can become preferable biorecognition molecules for salivary melatonin detection with high sensitivity and specificity. This study thoroughly characterizes the structural property and binding mechanism of a single-stranded DNA aptamer full sequence (MLT-C-1) and its truncated versions (MLT-A-2, MLT-A-4) to decipher its optimal characteristics for saliva melatonin detection. We use circular dichroism spectroscopy to determine aptamers' conformational changes under different ionic strengths and showed that aptamers display a hairpin loop structure where few base pairs in the stem play a significant role in melatonin binding and formation of aptamer stabilized structure. Through microscale thermophoresis, aptamers demonstrated a high binding affinity in saliva samples (MLT-C-1F Kd = 12.5 ± 1.7 nM; MLT-A-4F Kd = 11.2 ± 1.6 nM; MLT-A-2F Kd = 2.4 ± 2.8 nM; limit-of-detection achieved in pM, highest sensitivity attained for MLT-A-2F aptamer with the lowest detection limit of 1.35 pM). Our data suggest that aptamers are promising as biorecognition molecules and provide the baseline parameters for the development of an aptamer-based point-of-care diagnostic system for melatonin detection and accurate profiling of its fluctuations in saliva.
Collapse
Affiliation(s)
- Meenakshi Pundir
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada; Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada; Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada
| | - Maria C De Rosa
- Department of Chemistry, Faculty of Science, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, K1S 5B6, Canada.
| | - Liubov Lobanova
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada
| | - Shahad Abdulmawjood
- Department of Chemistry, Faculty of Science, Carleton University, 1125 Colonel by Drive, Ottawa, Ontario, K1S 5B6, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada; Department of Mechanical Engineering, School of Engineering, University of Saskatchewan, 57 Campus Dr, S7K 5A9, Saskatoon, Canada.
| | - Silvana Papagerakis
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada; Laboratory of Oral, Head and Neck Cancer - Personalized Diagnostics and Therapeutics, Department of Surgery, College of Medicine, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada; Department of Otolaryngology-Head and Neck Surgery, School of Medicine, University of Michigan, 1500 E Medical Center Dr, Ann Arbor, MI, 48109, United States.
| | - Petros Papagerakis
- Laboratory of Precision Oral Health and Chronobiology, College of Dentistry, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, S7N 5E4, Canada; Division of Biomedical Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, S7K 5A9, Canada.
| |
Collapse
|
30
|
Caixeta DC, Carneiro MG, Rodrigues R, Alves DCT, Goulart LR, Cunha TM, Espindola FS, Vitorino R, Sabino-Silva R. Salivary ATR-FTIR Spectroscopy Coupled with Support Vector Machine Classification for Screening of Type 2 Diabetes Mellitus. Diagnostics (Basel) 2023; 13:diagnostics13081396. [PMID: 37189497 DOI: 10.3390/diagnostics13081396] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/04/2023] [Accepted: 02/08/2023] [Indexed: 05/17/2023] Open
Abstract
The blood diagnosis of diabetes mellitus (DM) is highly accurate; however, it is an invasive, high-cost, and painful procedure. In this context, the combination of ATR-FTIR spectroscopy and machine learning techniques in other biological samples has been used as an alternative tool to develop a non-invasive, fast, inexpensive, and label-free diagnostic or screening platform for several diseases, including DM. In this study, we used the ATR-FTIR tool associated with linear discriminant analysis (LDA) and a support vector machine (SVM) classifier in order to identify changes in salivary components to be used as alternative biomarkers for the diagnosis of type 2 DM. The band area values of 2962 cm-1, 1641 cm-1, and 1073 cm-1 were higher in type 2 diabetic patients than in non-diabetic subjects. The best classification of salivary infrared spectra was by SVM, showing a sensitivity of 93.3% (42/45), specificity of 74% (17/23), and accuracy of 87% between non-diabetic subjects and uncontrolled type 2 DM patients. The SHAP features of infrared spectra indicate the main salivary vibrational modes of lipids and proteins that are responsible for discriminating DM patients. In summary, these data highlight the potential of ATR-FTIR platforms coupled with machine learning as a reagent-free, non-invasive, and highly sensitive tool for screening and monitoring diabetic patients.
Collapse
Affiliation(s)
- Douglas Carvalho Caixeta
- Innovation Center in Salivary Diagnostic and Nanotheranostics, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| | | | - Ricardo Rodrigues
- Innovation Center in Salivary Diagnostic and Nanotheranostics, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| | - Deborah Cristina Teixeira Alves
- Innovation Center in Salivary Diagnostic and Nanotheranostics, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| | - Luís Ricardo Goulart
- Institute of Biotechnology, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| | - Thúlio Marquez Cunha
- School of Medicine, Federal University of Uberlandia (UFU), Uberlandia 38408-100, Minas Gerais, Brazil
| | - Foued Salmen Espindola
- Institute of Biotechnology, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| | - Rui Vitorino
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Robinson Sabino-Silva
- Innovation Center in Salivary Diagnostic and Nanotheranostics, Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlandia 38408-100, Minas Gerais, Brazil
| |
Collapse
|
31
|
Thiha A, Ibrahim F, Joseph K, Petrović B, Kojić S, Dahlan NA, Jamaluddin NF, Qureshi S, Stojanović GM. A novel microfluidic compact disc to investigate electrochemical property changes between artificial and real salivary samples mixed with mouthwashes using electrical impedance analysis. PLoS One 2023; 18:e0280381. [PMID: 36795661 PMCID: PMC9934320 DOI: 10.1371/journal.pone.0280381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/26/2022] [Indexed: 02/17/2023] Open
Abstract
Diagnosing oral diseases at an early stage may lead to better preventive treatments, thus reducing treatment burden and costs. This paper introduces a systematic design of a microfluidic compact disc (CD) consisting of six unique chambers that run simultaneously from sample loading, holding, mixing and analysis. In this study, the electrochemical property changes between real saliva and artificial saliva mixed with three different types of mouthwashes (i.e. chlorhexidine-, fluoride- and essential oil (Listerine)-based mouthwashes) were investigated using electrical impedance analysis. Given the diversity and complexity of patient's salivary samples, we investigated the electrochemical impedance property of healthy real saliva mixed with different types of mouthwashes to understand the different electrochemical property which could be a foundation for diagnosis and monitoring of oral diseases. On the other hand, electrochemical impedance property of artificial saliva, a commonly used moisturizing agent and lubricant for the treatment of xerostomia or dry mouth syndrome was also studied. The findings indicate that artificial saliva and fluoride-based mouthwash showed higher conductance values compared to real saliva and two other different types of mouthwashes. The ability of our new microfluidic CD platform to perform multiplex processes and detection of electrochemical property of different types of saliva and mouthwashes is a fundamental concept for future research on salivary theranostics using point-of-care microfluidic CD platform.
Collapse
Affiliation(s)
- Aung Thiha
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Biomedical Engineering, Centre for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Fatimah Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Biomedical Engineering, Centre for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Centre for Printable Electronics, Universiti Malaya, Kuala Lumpur, Malaysia
- Microwave Research Institute, Universiti Teknologi MARA, Shah Alam, Selangor, Malaysia
- Malaysian Research Institute on Ageing, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- * E-mail: (FI); (BP)
| | - Karunan Joseph
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Biomedical Engineering, Centre for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Bojan Petrović
- Department of Dental Medicine, Faculty of Medicine, University of Novi Sad, Novi Sad, Serbia
- * E-mail: (FI); (BP)
| | - Sanja Kojić
- Faculty of Technical Science, University of Novi Sad, Novi Sad, Serbia
| | - Nuraina Anisa Dahlan
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Biomedical Engineering, Centre for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Nurul Fauzani Jamaluddin
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Department of Biomedical Engineering, Centre for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Saima Qureshi
- Malaysian Research Institute on Ageing, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | |
Collapse
|
32
|
Almukainzi M. Saliva Sampling in Therapeutic Drug Monitoring and Physiologically Based Pharmacokinetic Modeling: Review. Drug Res (Stuttg) 2023; 73:65-69. [PMID: 36368679 DOI: 10.1055/a-1956-9313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Therapeutic drug monitoring investigations based on saliva samples can be utilized as an alternative to blood sampling for many advantages. Moreover, the development of physiologically based pharmacokinetic (PBPK) modeling tools can further help to estimate drug exposure from saliva. This review discusses the use of saliva samples and illustrates the applications and examples of PBPK modeling systems for estimating drug exposure from saliva.
Collapse
Affiliation(s)
- May Almukainzi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Abdullameer MA, Abdulkareem AA. Diagnostic potential of salivary interleukin-17, RANKL, and OPG to differentiate between periodontal health and disease and discriminate stable and unstable periodontitis: A case-control study. Health Sci Rep 2023; 6:e1103. [PMID: 36778772 PMCID: PMC9900720 DOI: 10.1002/hsr2.1103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/20/2023] [Accepted: 01/28/2023] [Indexed: 02/08/2023] Open
Abstract
Background and Aims Limitations of the conventional diagnostic techniques urged researchers to seek novel methods to predict, diagnose, and monitor periodontal disease. Use of the biomarkers available in oral fluids could be a revolutionary surrogate for the manual probing/diagnostic radiograph. Several salivary biomarkers have the potential to accurately discriminate periodontal health and disease. This study aimed to determine the diagnostic sensitivity and specificity of salivary interleukin (IL)-17, receptor activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG), RANKL/OPG for differentiating (1) periodontal health from disease and (2) stable and unstable periodontitis. Methods Participants with periodontitis (n = 50) and gingivitis (n = 25), both diseases represented the cases, and subjects with healthy periodontium (n = 15) as a control were recruited for this study. Periodontitis cases were further equally subdivided into stable and unstable. Whole unstimulated salivary sample were collected from all participants. Periodontal parameters including bleeding on probing, probing pocket depth, clinical attachment loss, and number of missing teeth were recorded. The protein levels of salivary IL-17, RANKL, and OPG were determined by using enzyme-linked immunosorbent assays technique. Results Salivary IL-17, OPG, RANKL, and RANKL/OPG showed high sensitivity and specificity to differentiate periodontal health from gingivitis and periodontitis. Similar pattern was observed in discriminating stable and unstable periodontitis. Salivary IL-17 and RANKL showed a good accuracy to differentiate gingivitis from periodontitis. However, OPG and RANKL/OPG did not exhibit enough sensitivity and specificity to differentiate the latter conditions. Conclusion Salivary IL-17, RANKL, OPG, and RANKL/OPG system are potential candidates for differentiating periodontal health and disease and discriminate stable and unstable periodontitis.
Collapse
Affiliation(s)
- Marwa A. Abdullameer
- Department of HealthMinistry of HealthBaghdadIraq
- College of DentistryUniversity of BaghdadBaghdadIraq
| | | |
Collapse
|
34
|
Huang Z, Yang X, Huang Y, Tang Z, Chen Y, Liu H, Huang M, Qing L, Li L, Wang Q, Jie Z, Jin X, Jia B. Saliva - a new opportunity for fluid biopsy. Clin Chem Lab Med 2023; 61:4-32. [PMID: 36285724 DOI: 10.1515/cclm-2022-0793] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/29/2022] [Indexed: 12/15/2022]
Abstract
Saliva is a complex biological fluid with a variety of biomolecules, such as DNA, RNA, proteins, metabolites and microbiota, which can be used for the screening and diagnosis of many diseases. In addition, saliva has the characteristics of simple collection, non-invasive and convenient storage, which gives it the potential to replace blood as a new main body of fluid biopsy, and it is an excellent biological diagnostic fluid. This review integrates recent studies and summarizes the research contents of salivaomics and the research progress of saliva in early diagnosis of oral and systemic diseases. This review aims to explore the value and prospect of saliva diagnosis in clinical application.
Collapse
Affiliation(s)
- Zhijie Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiaoxia Yang
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yisheng Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Zhengming Tang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Yuanxin Chen
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Hongyu Liu
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Mingshu Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Ling Qing
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Li Li
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Qin Wang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Zhuye Jie
- BGI Genomics, BGI-Shenzhen, Shenzhen, P.R. China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen, P.R. China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xin Jin
- BGI Genomics, BGI-Shenzhen, Shenzhen, P.R. China
- School of Medicine, South China University of Technology, Guangzhou, P.R. China
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, P.R. China
| |
Collapse
|
35
|
Brooks PJ, Malkin EZ, De Michino S, Bratman SV. Isolation of salivary cell-free DNA for cancer detection. PLoS One 2023; 18:e0285214. [PMID: 37130100 PMCID: PMC10153704 DOI: 10.1371/journal.pone.0285214] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023] Open
Abstract
Saliva is an emerging source of disease biomarkers, particularly for cancers of the head and neck. Although analysis of cell-free DNA (cfDNA) in saliva holds promise as a liquid biopsy for cancer detection, currently there are no standardized methodologies for the collection and isolation of saliva for the purposes of studying DNA. Here, we evaluated various saliva collection receptacles and DNA purification techniques, comparing DNA quantity, fragment size, source, and stability. Then, using our optimized techniques, we tested the ability to detect human papillomavirus (HPV) DNA- a bona fide cancer biomarker in a subset of head and neck cancers- from patient saliva samples. For saliva collection, we found that the Oragene OG-600 receptacle yielded the highest concentration of total salivary DNA as well as short fragments <300 bp corresponding to mononucleosomal cell-free DNA. Moreover, these short fragments were stabilized beyond 48 hours after collection in contrast to other saliva collection receptacles. For DNA purification from saliva, the QIAamp Circulating Nucleic Acid kit yielded the highest concentration of mononucleosome-sized DNA fragments. Freeze-thaw of saliva samples did not affect DNA yield or fragment size distribution. Salivary DNA isolated from the OG-600 receptacle was found to be composed of both single and double-stranded DNA, including mitochondrial and microbial sources. While levels of nuclear DNA were consistent over time, levels of mitochondrial and microbial DNA were more variable and increased 48 hours after collection. Finally, we found that HPV DNA was stable in OG-600 receptacles, was reliably detected within the saliva of patients with HPV-positive head and neck cancer, and was abundant among mononucleosome-sized cell-free DNA fragments. Our studies have defined optimal techniques for isolating DNA from saliva that will contribute to future applications in liquid biopsy-based cancer detection.
Collapse
Affiliation(s)
- Patricia J Brooks
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Ethan Z Malkin
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Steven De Michino
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Scott V Bratman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Accelerating the Laboratory Testing Capacity through Saliva Pooling Prior to Direct RT-qPCR for SARS-CoV-2 Detection. Diagnostics (Basel) 2022; 12:diagnostics12123160. [PMID: 36553167 PMCID: PMC9777453 DOI: 10.3390/diagnostics12123160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The testing capacity of the laboratory is paramount for better control of the pandemic caused by SARS-CoV-2. The pooling method is promising to increase testing capacity, and the use of direct NAAT-based detection of SARS-CoV-2 on a non-invasive specimen such as saliva will ultimately accelerate the testing capacity. This study aims to validate the pooling-of-four method to quadruple the testing capacity using RNA-extraction-free saliva specimens. In addition, we intend to investigate the preferable stage of pooling, including pre- or post-heating. The compatibility of this approach was also tested on five commercial kits. Saliva specimens stored at -80 °C for several months were proven viable and were used for various tests in this study. Our findings revealed that pooling-of-four specimens had an overall agreement rate of 98.18% with their individual testing. Moreover, we proved that the pooling procedure could be conducted either pre- or post-heating, with no discordance and no significant difference in Ct values generated. When compared to other commercial detection kits, it demonstrated an overall agreement greater than 85%, which exhibits broad compatibility and ensures easy adaptability in clinical settings. This method has been proven reliable and increases the testing capacity up to fourfold.
Collapse
|
37
|
Materón EM, Gómez FR, Almeida MB, Shimizu FM, Wong A, Teodoro KBR, Silva FSR, Lima MJA, Angelim MKSC, Melendez ME, Porras N, Vieira PM, Correa DS, Carrilho E, Oliveira O, Azevedo RB, Goncalves D. Colorimetric Detection of SARS-CoV-2 Using Plasmonic Biosensors and Smartphones. ACS APPLIED MATERIALS & INTERFACES 2022; 14:54527-54538. [PMID: 36454041 PMCID: PMC9728479 DOI: 10.1021/acsami.2c15407] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/08/2022] [Indexed: 05/27/2023]
Abstract
Low-cost, instrument-free colorimetric tests were developed to detect SARS-CoV-2 using plasmonic biosensors with Au nanoparticles functionalized with polyclonal antibodies (f-AuNPs). Intense color changes were noted with the naked eye owing to plasmon coupling when f-AuNPs form clusters on the virus, with high sensitivity and a detection limit of 0.28 PFU mL-1 (PFU stands for plaque-forming units) in human saliva. Plasmon coupling was corroborated with computer simulations using the finite-difference time-domain (FDTD) method. The strategies based on preparing plasmonic biosensors with f-AuNPs are robust to permit SARS-CoV-2 detection via dynamic light scattering and UV-vis spectroscopy without interference from other viruses, such as influenza and dengue viruses. The diagnosis was made with a smartphone app after processing the images collected from the smartphone camera, measuring the concentration of SARS-CoV-2. Both image processing and machine learning algorithms were found to provide COVID-19 diagnosis with 100% accuracy for saliva samples. In subsidiary experiments, we observed that the biosensor could be used to detect the virus in river waters without pretreatment. With fast responses and requiring small sample amounts (only 20 μL), these colorimetric tests can be deployed in any location within the point-of-care diagnosis paradigm for epidemiological control.
Collapse
Affiliation(s)
- Elsa M. Materón
- São Carlos Institute of Physics,
University of São Paulo, P.O Box 369,
13560-970São Carlos, SP, Brazil
- São Carlos Institute of Chemistry,
University of São Paulo, 13566-590São Carlos,
SP, Brazil
| | - Faustino R. Gómez
- São Carlos Institute of Physics,
University of São Paulo, P.O Box 369,
13560-970São Carlos, SP, Brazil
| | - Mariana B. Almeida
- São Carlos Institute of Chemistry,
University of São Paulo, 13566-590São Carlos,
SP, Brazil
- National Institute of Science and
Technology in Bioanalytics - INCTBio, 13083-970Campinas, SP,
Brazil
| | - Flavio M. Shimizu
- Department of Applied Physics, “Gleb
Wataghin” Institute of Physics (IFGW), University of Campinas
(UNICAMP), 13083-859Campinas, SP, Brazil
| | - Ademar Wong
- Department of Chemistry, Federal
University of São Carlos (UFSCar), 13560-970São Carlos,
São Paulo, Brazil
| | - Kelcilene B. R. Teodoro
- Nanotechnology National Laboratory for Agriculture,
Embrapa Instrumentation, 13560-970São Carlos, SP,
Brazil
| | - Filipe S. R. Silva
- São Carlos Institute of Chemistry,
University of São Paulo, 13566-590São Carlos,
SP, Brazil
| | - Manoel J. A. Lima
- São Carlos Institute of Chemistry,
University of São Paulo, 13566-590São Carlos,
SP, Brazil
| | - Monara Kaelle S. C. Angelim
- Department of Genetics Evolution, Microbiology, and
Immunology, Institute of Biology, University of Campinas,
13083-970Campinas, SP, Brazil
| | - Matias E. Melendez
- Molecular Carcinogenesis Program,
National Cancer Institute, 20231-050Rio de Janeiro, RJ,
Brazil
| | - Nelson Porras
- Physics Department, del Valle
University, AA 25360Cali, Colombia
| | - Pedro M. Vieira
- Department of Genetics Evolution, Microbiology, and
Immunology, Institute of Biology, University of Campinas,
13083-970Campinas, SP, Brazil
| | - Daniel S. Correa
- Nanotechnology National Laboratory for Agriculture,
Embrapa Instrumentation, 13560-970São Carlos, SP,
Brazil
| | - Emanuel Carrilho
- São Carlos Institute of Chemistry,
University of São Paulo, 13566-590São Carlos,
SP, Brazil
- National Institute of Science and
Technology in Bioanalytics - INCTBio, 13083-970Campinas, SP,
Brazil
| | - Osvaldo
N. Oliveira
- São Carlos Institute of Physics,
University of São Paulo, P.O Box 369,
13560-970São Carlos, SP, Brazil
| | - Ricardo B. Azevedo
- Laboratory of Nanobiotechnology, Department of Genetics
and Morphology, Institute of Biological Sciences, University of
Brasilia, 70910-900Brasilia, DF, Brazil
| | - Débora Goncalves
- São Carlos Institute of Physics,
University of São Paulo, P.O Box 369,
13560-970São Carlos, SP, Brazil
| |
Collapse
|
38
|
Balasubramaniam A, Arumugham MI. Salivary oxidative stress level among tobacco chewers and smokers: A comparative Study. J Adv Pharm Technol Res 2022; 13:S21-S25. [PMID: 36643152 PMCID: PMC9836113 DOI: 10.4103/japtr.japtr_116_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/11/2022] [Accepted: 06/12/2022] [Indexed: 01/17/2023] Open
Abstract
Tobacco contains various toxic contents which produce oxygen-free radicals that damage oral tissues. Since saliva encounters tobacco, it has antioxidant defense system and also can serve as a biomarker for oral diseases. Thus, the present study aims to evaluate salivary oxidative stress levels among smokers and chewers. Unstimulated saliva from 240 males who visited tobacco cessation clinic for the first time was collected. Standard protocol was followed to collect saliva and assess salivary antioxidant levels from 80 participants with the habit of smoking, smokeless, and both (smoking and smokeless) tobacco users. The collected data were statistically analyzed. The mean salivary superoxide dismutase and malonyldialdehyde levels were significantly high for participants with both habits (P < 0.000). There was a significant high reduction of glutathione peroxidase and catalase in participants with both habits (P < 0.000). Both smoking and smokeless tobacco modify salivary antioxidant activity. The estimation of salivary oxidative stress can serve as a diagnostic and prognostic biomarker for oral tissue damage and dysplasia. Furthermore, they can function as early biomarkers in preventing dysplastic changes in the oral cavity.
Collapse
Affiliation(s)
- Arthi Balasubramaniam
- Department of Public Health Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India,Address for correspondence: Dr. Arthi Balasubramaniam, Department of Public Health Dentistry, Saveetha Dental College and Hospitals, 162, Poonamalee High Road, Chennai - 600 077, Tamil Nadu, India. E-mail:
| | - Meignana Indiran Arumugham
- Department of Public Health Dentistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
39
|
Cui Y, Zhang H, Wang S, Lu J, He J, Liu L, Liu W. Stimulated Parotid Saliva Is a Better Method for Depression Prediction. Biomedicines 2022; 10:2220. [PMID: 36140321 PMCID: PMC9496557 DOI: 10.3390/biomedicines10092220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Saliva cortisol is considered to be a biomarker of depression prediction. However, saliva collection methods can affect the saliva cortisol level. OBJECTIVE This study aims to determine the ideal saliva collection method and explore the application value of saliva cortisol in depression prediction. METHODS 30 depressed patients and 30 healthy controls were instructed to collect saliva samples in the morning with six collection methods. Simultaneous venous blood was collected. Enzyme-linked immunosorbent assay was used to determine the cortisol level. The 24-observerrated Hamilton depression rating scale (HAMD-24) was used to assess the severity of depression. RESULTS The significant differences in saliva cortisol levels depend on the saliva collection methods. The level of unstimulated whole saliva cortisol was most correlated with blood (r = 0.91). The stimulated parotid saliva cortisol can better predict depression. The area under the curve was 0.89. In addition, the saliva cortisol level of the depression patients was significantly higher than the healthy controls. The correlation between the cortisol level and the HAMD-24 score was highly significant. The higher the saliva cortisol level, the higher the HAMD-24 score. CONCLUSIONS All the above findings point to an exciting opportunity for non-invasive monitoring of cortisol through saliva.
Collapse
Affiliation(s)
- Yangyang Cui
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Hankun Zhang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Song Wang
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Junzhe Lu
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Jinmei He
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Lanlan Liu
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| | - Weiqiang Liu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
- Biomechanics and Biotechnology Lab, Research Institute of Tsinghua University in Shenzhen, Shenzhen 518057, China
| |
Collapse
|
40
|
The New Era of Salivaomics in Dentistry: Frontiers and Facts in the Early Diagnosis and Prevention of Oral Diseases and Cancer. Metabolites 2022; 12:metabo12070638. [PMID: 35888762 PMCID: PMC9319392 DOI: 10.3390/metabo12070638] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/27/2022] [Accepted: 07/06/2022] [Indexed: 12/22/2022] Open
Abstract
Nowadays, with the development of new and highly sensitive, blood is not the only medium of choice for the diagnosis of several diseases and pathological conditions. Saliva is now considered a safe and non-invasive sample to study oral and systemic diseases, showing great diagnostic potential. According to several recent studies, saliva has emerged as an emerging biofluid for the early diagnosis of several diseases, indicated as a mirror of oral and systemic health and a valuable source of clinically relevant information. Indeed, several studies have observed that saliva is useful for detecting and diagnosing malignant tumours, human immunodeficiency virus, heart disease, and autoimmune diseases. The growing realisation that saliva is an inexhaustible source of information has led to the coining of the term ‘Salivaomics’, which includes five “omics” in connection with the main constituents of saliva: genome and epigenome, transcriptomics, metabolomics, lipidomics, proteomics and microbiota. All those may be changed by disease state, so they offer significant advantages in the early diagnosis and prognosis of oral diseases. The aim of the present review isto update and highlight the new frontiers of salivaomics in diagnosing and managing oral disorders, such as periodontitis, premalignant disorders, and oral squamous cell carcinoma (OSCC).
Collapse
|
41
|
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: 37] [Impact Index Per Article: 12.3] [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
|
42
|
Pundir M, Papagerakis S, De Rosa MC, Chronis N, Kurabayashi K, Abdulmawjood S, Prince MEP, Lobanova L, Chen X, Papagerakis P. Emerging biotechnologies for evaluating disruption of stress, sleep, and circadian rhythm mechanism using aptamer-based detection of salivary biomarkers. Biotechnol Adv 2022; 59:107961. [PMID: 35427723 DOI: 10.1016/j.biotechadv.2022.107961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/30/2022] [Accepted: 04/09/2022] [Indexed: 12/26/2022]
|
43
|
Michailidou E, Poulopoulos A, Tzimagiorgis G. Salivary diagnostics of the novel coronavirus SARS-CoV-2 (COVID-19). Oral Dis 2022; 28 Suppl 1:867-877. [PMID: 33211392 PMCID: PMC7753835 DOI: 10.1111/odi.13729] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Laboratory testing for the SARS-CoV-2 virus and the consequent respiratory coronavirus disease 2019 (COVID-19) is categorized into methods that detect the viral presence and methods that detect antibodies produced in the host as a response to infection. Methods that detect viral presence into the host excretions measure current infection by SARS-CoV-2, whereas the detection of human antibodies exploited against SARS-CoV-2 evaluates the past exposure to the virus. OBJECTIVE This review provides a comprehensive overview for the use of saliva as a specimen for the detection of SARS-CoV-2, the methods for the salivary diagnostics utilized till very recently, and the arisen considerations for the diagnosis of COVID-19 disease. CONCLUSION The major advantage of using saliva as a specimen for the detection of SARS-CoV-2 is that saliva collection is a non-invasive method which produces no discomfort to the patient and permits the patients to utilize home self-sampling techniques in order to protect health providers from the exposure to the pathogen. There is an urgent need to increase the active research for the detection of SARS-CoV-2 in the saliva because the non-invasive salivary diagnostics may provide a reliable and cost-effective method suitable for the fast and early detection of COVID-19 infection.
Collapse
Affiliation(s)
- Evangelia Michailidou
- Department of Oral Medicine and Maxillofacial PathologySchool of DentistryAristotle UniversityThessalonikiGreece
| | - Athanasios Poulopoulos
- Department of Oral Medicine and Maxillofacial PathologySchool of DentistryAristotle UniversityThessalonikiGreece
| | - Georgios Tzimagiorgis
- Laboratory of Biological ChemistryMedical SchoolAristotle University of ThessalonikiThessalonikiGreece
| |
Collapse
|
44
|
Costa MM, Martin H, Estellon B, Dupé FX, Saby F, Benoit N, Tissot-Dupont H, Million M, Pradines B, Granjeaud S, Almeras L. Exploratory Study on Application of MALDI-TOF-MS to Detect SARS-CoV-2 Infection in Human Saliva. J Clin Med 2022; 11:295. [PMID: 35053990 PMCID: PMC8781148 DOI: 10.3390/jcm11020295] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/16/2021] [Accepted: 12/31/2021] [Indexed: 12/24/2022] Open
Abstract
SARS-CoV-2 has caused a large outbreak since its emergence in December 2019. COVID-19 diagnosis became a priority so as to isolate and treat infected individuals in order to break the contamination chain. Currently, the reference test for COVID-19 diagnosis is the molecular detection (RT-qPCR) of the virus from nasopharyngeal swab (NPS) samples. Although this sensitive and specific test remains the gold standard, it has several limitations, such as the invasive collection method, the relative high cost and the duration of the test. Moreover, the material shortage to perform tests due to the discrepancy between the high demand for tests and the production capacities puts additional constraints on RT-qPCR. Here, we propose a PCR-free method for diagnosing SARS-CoV-2 based on matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF MS) profiling and machine learning (ML) models from salivary samples. Kinetic saliva samples were collected at enrollment and ten and thirty days later (D0, D10 and D30), to assess the classification performance of the ML models compared to the molecular tests performed on NPS specimens. Spectra were generated using an optimized protocol of saliva collection and successive quality control steps were developed to ensure the reliability of spectra. A total of 360 averaged spectra were included in the study. At D0, the comparison of MS spectra from SARS-CoV-2 positive patients (n = 105) with healthy healthcare controls (n = 51) revealed nine peaks that significantly distinguished the two groups. Among the five ML models tested, support vector machine with linear kernel (SVM-LK) provided the best performance on the training dataset (accuracy = 85.2%, sensitivity = 85.1%, specificity = 85.3%, F1-Score = 85.1%). The application of the SVM-LK model on independent datasets confirmed its performances with 88.9% and 80.8% of correct classification for samples collected at D0 and D30, respectively. Conversely, at D10, the proportion of correct classification had fallen to 64.3%. The analysis of saliva samples by MALDI-TOF MS and ML appears as an interesting supplementary tool for COVID-19 diagnosis, despite the mitigated results obtained for convalescent patients (D10).
Collapse
Affiliation(s)
- Monique Melo Costa
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
| | - Hugo Martin
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
| | - Bertrand Estellon
- Laboratoire d’Informatique et Systèmes, Aix-Marseille University, CNRS, University de Toulon, 13013 Marseille, France; (B.E.); (F.-X.D.)
| | - François-Xavier Dupé
- Laboratoire d’Informatique et Systèmes, Aix-Marseille University, CNRS, University de Toulon, 13013 Marseille, France; (B.E.); (F.-X.D.)
| | - Florian Saby
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
| | - Nicolas Benoit
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Hervé Tissot-Dupont
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
- Aix-Marseille University, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Matthieu Million
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
- Aix-Marseille University, IRD, AP-HM, MEPHI, 13005 Marseille, France
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
- Centre National de Référence du Paludisme, 13005 Marseille, France
| | - Samuel Granjeaud
- CRCM Integrative Bioinformatics Platform, Centre de Recherche en Cancérologie de Marseille, INSERM, U1068, Institut Paoli-Calmettes, CNRS, UMR7258, Aix-Marseille Université UM 105, 13009 Marseille, France;
| | - Lionel Almeras
- Unité Parasitologie et Entomologie, Département Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, 91220 Marseille, France; (M.M.C.); (H.M.); (F.S.); (N.B.); (B.P.)
- Aix-Marseille University, IRD, SSA, AP-HM, VITROME, 13005 Marseille, France
- IHU Méditerranée Infection, 13005 Marseille, France; (H.T.-D.); (M.M.)
| |
Collapse
|
45
|
Cui Y, Yang M, Zhu J, Zhang H, Duan Z, Wang S, Liao Z, Liu W. Developments in diagnostic applications of saliva in Human Organ Diseases. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100115] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
46
|
Abstract
The use of saliva as a diagnostic biofluid has been increasing in recent years, thanks to the identification and validation of new biomarkers and improvements in test accuracy, sensitivity, and precision that enable the development of new noninvasive and cost-effective devices. However, the lack of standardized methods for sample collection, treatment, and storage contribute to the overall variability and lack of reproducibility across analytical evaluations. Furthermore, the instability of salivary biomarkers after sample collection hinders their translation into commercially available technologies for noninvasive monitoring of saliva in home settings. The present review aims to highlight the status of research on the challenges of collecting and using diagnostic salivary samples, emphasizing the methodologies used to preserve relevant proteins, hormones, genomic, and transcriptomic biomarkers during sample handling and analysis.
Collapse
Affiliation(s)
- Luciana d'Amone
- Silklab, Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Giusy Matzeu
- Silklab, Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Fiorenzo G Omenetto
- Silklab, Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States.,Department of Electrical and Computer Engineering, Tufts University, Medford, Massachusetts 02155, United States.,Department of Physics, Tufts University, Medford, Massachusetts 02155, United States.,Laboratory for Living Devices, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
47
|
Potential of Salivary Biomarkers in Autism Research: A Systematic Review. Int J Mol Sci 2021; 22:ijms221910873. [PMID: 34639213 PMCID: PMC8509590 DOI: 10.3390/ijms221910873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/27/2021] [Accepted: 10/05/2021] [Indexed: 12/14/2022] Open
Abstract
The diagnostic process for autism spectrum disorders (ASD) is based on a behavioral analysis of the suspected individual. Despite intensive research, no specific and valid biomarker has been identified for ASD, but saliva, with its advantages such as non-invasive collection, could serve as a suitable alternative to other body fluids. As a source of nucleic acid of both human and microbial origin, protein and non-protein molecules, saliva offers a complex view on the current state of the organism. Additionally, the use of salivary markers seems to be less complicated not only for ASD screening but also for revealing the etiopathogenesis of ASD, since enrolling neurotypical counterparts willing to participate in studies may be more feasible. The aim of the presented review is to provide an overview of the current research performed on saliva in relation to ASD, mutual complementing, and discrepancies that result in difficulties applying the observed markers in clinical practice. We emphasize the methodological limitations of saliva collection and processing as well as the lack of information regarding ASD diagnosis, which is critically discussed.
Collapse
|
48
|
Shakeeb N, Varkey P, Ajit A. Human Saliva as a Diagnostic Specimen for Early Detection of Inflammatory Biomarkers by Real-Time RT-PCR. Inflammation 2021; 44:1713-1723. [PMID: 34031776 PMCID: PMC8143742 DOI: 10.1007/s10753-021-01484-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 01/04/2023]
Abstract
Nowadays human saliva is more frequently studied as a non-invasive, stress-free, and preferable diagnostic material than blood. Supporting evidences acknowledge saliva as a mirror that reflects the body's physical state. Numerous studies have also demonstrated the presence and use of RNA derived from saliva in the early diagnosis of disease by real-time reverse transcriptase polymerase chain reaction (RT-PCR). Assessing the host inflammatory response in patients and its resolution at an early stage can serve as a prognostic and predictive method in determining therapeutic response or disease progression. In this context, the potential of saliva as a specimen to diagnose early inflammatory biomarkers using RT-PCR seems fascinating and useful. Here, we review inflammatory biomarkers within the saliva, focusing on early detection of these biomarkers using RT-PCR and the factors influencing the quality of saliva specimen.
Collapse
Affiliation(s)
- Nourin Shakeeb
- Research and Development, Zum Heilen Diagnostic and Therapeutics Pvt. Ltd, Office No. 12/1543-C, SB Center, 2nd Floor, Museum Road, Thrissur, Kerala 680020 India
| | - Prashanth Varkey
- Jubilee Centre for Medical Research, Jubilee Mission Medical College & Research Institute, P.B.No.737, Thrissur, Kerala 680005 India
| | - Amita Ajit
- Research and Development, Zum Heilen Diagnostic and Therapeutics Pvt. Ltd, Office No. 12/1543-C, SB Center, 2nd Floor, Museum Road, Thrissur, Kerala 680020 India
| |
Collapse
|
49
|
Duś-Ilnicka I, Krala E, Cholewińska P, Radwan-Oczko M. The Use of Saliva as a Biosample in the Light of COVID-19. Diagnostics (Basel) 2021; 11:1769. [PMID: 34679467 PMCID: PMC8534561 DOI: 10.3390/diagnostics11101769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022] Open
Abstract
Saliva is easy to collect and a biofluid that is readily available without the need for special equipment for its collection. The collection process, which is non-invasive and inexpensive, leads to obtaining a biomaterial that can serve as a source of information for molecular diagnostics of diseases in general medicine, genetics and dentistry. Unfortunately, many of the salivary methodologies are lacking important parameters to provide for not only the safety of the operator, but also the quality and reproducibility of the research. Since the COVID-19 pandemic, salivary diagnostics demonstrate a great potential for research of SARS-CoV 2. In this review, good practice for unstimulated saliva collection and patient preparation was provided, based on the latest literature and available guidelines. Schemes for saliva collection procedures were presented following an extended literature search. Descriptions of salivary probes/cups, techniques of saliva collection, and the use of specific buffering solutions for the stability of collected samples for SARS-CoV-2 detection were also evaluated.
Collapse
Affiliation(s)
- Irena Duś-Ilnicka
- Oral Pathology Department, Faculty of Dentistry, Wroclaw Medical University, ul. Krakowska 26, 50-425 Wroclaw, Poland; (E.K.); (M.R.-O.)
| | - Elżbieta Krala
- Oral Pathology Department, Faculty of Dentistry, Wroclaw Medical University, ul. Krakowska 26, 50-425 Wroclaw, Poland; (E.K.); (M.R.-O.)
| | - Paulina Cholewińska
- Institute of Animal Breeding, Faculty of Biology and Animal Breeding, Wroclaw University of Enviromental and Life Sciences, ul. Chełmońskiego 38C, 51-630 Wroclaw, Poland;
| | - Małgorzata Radwan-Oczko
- Oral Pathology Department, Faculty of Dentistry, Wroclaw Medical University, ul. Krakowska 26, 50-425 Wroclaw, Poland; (E.K.); (M.R.-O.)
| |
Collapse
|
50
|
Faheem S, Maqsood S, Hasan A, Imtiaz F, Shaikh F, Farooqui WA. Associations of early childhood caries with salivary beta defensin-3 and childhood anemia: a case-control study. BMC Oral Health 2021; 21:445. [PMID: 34521396 PMCID: PMC8442316 DOI: 10.1186/s12903-021-01810-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Background Human beta defensin-3 (HβD-3) is an antimicrobial peptide present in saliva that protects tooth surfaces from microbial attack. These peptides are part of innate immunity so levels may be affected by different systemic diseases like anemia. Therefore, anemia may predispose an affected child to an increased risk of dental caries. The objectives of this study were to determine the association of early childhood caries (ECC) with HβD-3 levels and observe the association of HβD-3 levels with childhood anemia. Methods A total of 80 children admitted in a pediatric medical ward, age 48–71 months, of either sex were included in the study. The included children were categorized as cases (children with ECC n = 40) and controls (children without ECC n = 40). Children were further segregated into the anemic and non-anemic sub-groups based on the hospital record of hemoglobin level. The salivary concentration of HβD-3 was measured by Enzyme-Linked Immuno-sorbent assay (ELISA). IBM SPSS version 20 software was used for statistical analysis. Two sample t-test and one-way ANOVA were used to compare mean values while spearman was used for correlations at p < 0.05. Results The mean Salivary HβD-3 level in cases (8.87 ± 4.30) was significantly higher (p = 0.042) as compared to controls (7.23 ± 2.57). Salivary HβD-3 level in patients with caries and without anemia was highest (10.80 ± 4.50) whereas salivary HβD-3 level in the presence of caries and anemia was lowest (6.94 ± 3.13) amongst all groups. This difference was statistically significant (p = 0.001). Salivary HβD-3 level was found to be moderately correlated with cases (p = 0.002). An inverse correlation was found between salivary HβD-3 level and anemia (r = -0.479, p = 0.002). Conclusion Anemia may affect the innate immunity of children, and may result in a decreased level of salivary HβD3, thus increasing vulnerability to decay.
Collapse
Affiliation(s)
- Sanam Faheem
- Department of Oral Biology, Dow Dental College, Dow University of Health Sciences, Karachi, Pakistan.
| | - Shahida Maqsood
- Department of Oral Biology, Dow Dental College, Dow University of Health Sciences, Karachi, Pakistan
| | - Arshad Hasan
- Department of Operative Dentistry, Dow Dental College, Dow University of Health Sciences, Karachi, Pakistan
| | - Fouzia Imtiaz
- Department of Biochemistry Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Faheem Shaikh
- London Dental Clinics & Dental Implants, 41-C Badar Commercial Street 10, Phase 5, Badar Commercial DHA, Karachi, Pakistan
| | - Waqas Ahmed Farooqui
- Department of Research, School of Public Health, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|